1
|
Huang R, Zhou G, Cai J, Cao C, Zhu Z, Wu Q, Zhang F, Ding Y. Maternal consumption of urbanized diet compromises early-life health in association with gut microbiota. Gut Microbes 2025; 17:2483783. [PMID: 40176259 PMCID: PMC11988223 DOI: 10.1080/19490976.2025.2483783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Revised: 03/09/2025] [Accepted: 03/18/2025] [Indexed: 04/04/2025] Open
Abstract
Urbanization has significantly transformed dietary habits worldwide, contributing to a globally increased burden of non-communicable diseases and altered gut microbiota landscape. However, it is often overlooked that the adverse effects of these dietary changes can be transmitted from the mother to offspring during early developmental stages, subsequently influencing the predisposition to various diseases later in life. This review aims to delineate the detrimental effects of maternal urban-lifestyle diet (urbanized diet) on early-life health and gut microbiota assembly, provide mechanistic insights on how urbanized diet mediates mother-to-offspring transfer of bioactive substances in both intrauterine and extrauterine and thus affects fetal and neonatal development. Moreover, we also further propose a framework for developing microbiome-targeted precision nutrition and diet strategies specifically for pregnant and lactating women. The establishment of such knowledge can help develop proactive preventive measures from the beginning of life, ultimately reducing the long-term risk of disease and improving public health outcomes.
Collapse
Affiliation(s)
- Rong Huang
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Guicheng Zhou
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Jie Cai
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Cha Cao
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Zhenjun Zhu
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Qingping Wu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou, China
| | - Fen Zhang
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Yu Ding
- Department of Food Science and Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| |
Collapse
|
2
|
Du H, Li K, Guo W, Na M, Zhang J, Zhang J, Na R. Physiological and Microbial Community Dynamics in Does During Mid-Gestation to Lactation and Their Impact on the Growth, Immune Function, and Microbiome Transmission of Offspring Kids. Animals (Basel) 2025; 15:954. [PMID: 40218348 PMCID: PMC11987885 DOI: 10.3390/ani15070954] [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: 02/13/2025] [Revised: 03/17/2025] [Accepted: 03/20/2025] [Indexed: 04/14/2025] Open
Abstract
This study investigated changes in physiological processes and rumen microbial communities in does from mid-gestation to lactation and identified potential associations between these physiological changes and the rumen microbiome. Additionally, we studied the transmission mechanisms of microorganisms between the dam and offspring. Our study demonstrates significant changes in maternal physiological metabolism, immune status, and rumen microbiota from mid-pregnancy through lactation. We identified potential associations between these physiological changes and the rumen microbiome. Moreover, the findings highlight that alterations in maternal physiological metabolism and immune status significantly influence the growth and immune development of offspring kids. Additionally, we observed that the maternal microbiota serves as a key source of gastrointestinal microbial communities in young animals, with early colonization of maternally derived microbes in the offspring's gastrointestinal tract playing a role in shaping their immune system development. The results for primary outcomes are as follows: The serum levels of estrogen and progesterone in pregnant does were greater than those observed during lactation, while the concentration of growth hormone, triiodothyronine, and glucose exhibited an upward trend during lactation. During late gestation, the serum IL-10 concentration in does decreased, while the TNF-α concentration increased. Additionally, on day 140 of gestation, does showed a significant decrease in IgG, total protein, and globulin levels. From mid-gestation to lactation, the abundance of dominant phyla and genera, including Firmicutes, Bacteroidetes, Patescibacteria, Bacteroidales_RF16_group, Clostridia_UCG-014, RF39, and Eubacterium_ventriosum_group, in the rumen of does underwent significant changes. LEfSe analysis identified a series of marker microorganisms in the rumen of does at different physiological stages. A correlation was observed between these dominant bacteria and the serum physiological indicators of the does. Notably, rumen volatile fatty acids also exhibited a correlation with serum physiological indicators. In addition, serum physiological indicators of does were significantly correlated with the growth and immune indicators of their kids. Microbiological origin analysis revealed that the gastrointestinal microbiome of kids primarily originated from the rumen, birth canal, and milk of does. Further analysis identified a correlation between the kids' serum immunometric indicators and certain gastrointestinal microorganisms. In particular, the jejunum microbiota of 28-day-old lactating kids, including Alysiella, Neisseria, and Muribaculaceae, showed a significant positive correlation with serum IL-6 and IL-10 levels. Meanwhile, these genera were dominant in the saliva and milk of does, suggesting a direct microbial transfer from dam to offspring. These microbial communities may play a significant role in modulating the metabolism and immune responses of the offspring, thereby influencing their immune system development.
Collapse
Affiliation(s)
- Haidong Du
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (H.D.); (W.G.); (M.N.); (J.Z.)
| | - Kenan Li
- Grassland Research Institute of Chinese Academy of Agricultural Sciences, Hohhot 010010, China;
| | - Wenliang Guo
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (H.D.); (W.G.); (M.N.); (J.Z.)
| | - Meila Na
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (H.D.); (W.G.); (M.N.); (J.Z.)
| | - Jing Zhang
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (H.D.); (W.G.); (M.N.); (J.Z.)
| | - Jing Zhang
- Intellectual Property Protection Center of Inner Mongolia Autonomous Region, Hohhot 010050, China;
| | - Renhua Na
- College of Animal Science, Inner Mongolia Agricultural University, Hohhot 010018, China; (H.D.); (W.G.); (M.N.); (J.Z.)
| |
Collapse
|
3
|
Armistead B, Peters MQ, Houck J, Carlson M, Balle C, Mulugeta N, Gray CM, Jaspan HB, Harrington WE. Exposure to Human Immunodeficiency Virus Is Associated With Altered Composition of Maternal Microchimeric T Cells in Infants. J Infect Dis 2025; 231:435-439. [PMID: 39435850 PMCID: PMC12054726 DOI: 10.1093/infdis/jiae521] [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: 04/18/2024] [Revised: 08/01/2024] [Accepted: 10/18/2024] [Indexed: 10/23/2024] Open
Abstract
Human immunodeficiency virus-exposed but uninfected infants (iHEU) display altered immunity and are at increased risk of infection. We previously reported that iHEU have decreased maternal microchimerism (MMc)-maternal cells transferred to the offspring in utero/during breastfeeding. We quantified MMc in T-cell subpopulations in iHEU and HIV-unexposed infants (iHU) to determine whether a selective deficiency in MMc contributes to altered cellular immunity. Across all infants, MMc levels were highest in CD8+ T cells; however, the level of CD8+ T-cell MMc was lower in iHEU versus iHU. In limited functional studies, we did not identify cytomegalovirus-specific MMc during infant primary infection.
Collapse
Affiliation(s)
| | | | - John Houck
- Center for Global Infectious Disease Research
| | - Marc Carlson
- Research Scientific Computing, Enterprise Analytics, Seattle Children's Research Institute, Washington
| | - Christina Balle
- Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, South Africa
| | - Nolawit Mulugeta
- Center for Global Infectious Disease Research
- Department of Global Health, University of Washington, Seattle
| | - Clive M Gray
- Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, South Africa
- Division of Immunology, Biomedical Research Institute, Stellenbosch University, Cape Town, South Africa
| | - Heather B Jaspan
- Center for Global Infectious Disease Research
- Institute of Infectious Disease and Molecular Medicine, Division of Immunology, Department of Pathology, University of Cape Town, South Africa
- Department of Global Health, University of Washington, Seattle
- Department of Pediatrics, University of Washington, Seattle
| | - Whitney E Harrington
- Center for Global Infectious Disease Research
- Department of Global Health, University of Washington, Seattle
- Department of Pediatrics, University of Washington, Seattle
| |
Collapse
|
4
|
Balla J, Rathore AP, St John AL. Mechanisms and risk factors for perinatal allergic disease. Curr Opin Immunol 2024; 91:102505. [PMID: 39566249 DOI: 10.1016/j.coi.2024.102505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/19/2024] [Accepted: 10/26/2024] [Indexed: 11/22/2024]
Abstract
Allergies are among the top causes of chronic disease in children. Their pathogenesis classically involves T helper 2 (Th2)-type inflammation driven by IgE-mediated allergen sensing. Triggers influencing allergic disease occur early in life, including before birth. The immature fetal immune system and mucosal barriers undergo periods of plasticity that are open to longitudinal programming by maternal influence. Evidence supports the importance of the maternal immune system in shaping perinatal immunity, as the transfer of cytokines, antibodies, and cells promotes offspring protection from pathogens. However, the same components may lead to allergic predisposition. Maternal-fetal interactions are further modified by epigenetic, metabolic, dietary, and microbiome-mediated effects. Here, we review how diverse maternal exposures and mediators signal across the placenta and through nursing perinatally to promote future tolerance or enhance reactivity against allergens. Improved understanding of the mechanisms predisposing for allergic disease in early life can guide the development of new therapeutics and preventative lifestyle modifications.
Collapse
Affiliation(s)
- Jozef Balla
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, 169857 Singapore
| | - Abhay Ps Rathore
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, 169857 Singapore; Department of Pathology, Duke University Medical Center, Durham, North Carolina 27705, USA
| | - Ashley L St John
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, 169857 Singapore; Department of Pathology, Duke University Medical Center, Durham, North Carolina 27705, USA; Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; SingHealth Duke-NUS Global Health Institute, Singapore.
| |
Collapse
|
5
|
Saager ES, van Stigt AH, Lerkvaleekul B, Lutter L, Hellinga AH, van der Wal MM, Bont LJ, Leusen JH, van’t Land B, van Wijk F. Human breastmilk memory T cells throughout lactation manifest activated tissue-oriented profile with prominent regulation. JCI Insight 2024; 9:e181788. [PMID: 39435660 PMCID: PMC11530127 DOI: 10.1172/jci.insight.181788] [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: 10/23/2024] Open
Abstract
Breastfeeding provides important immunological benefits to the neonate, but how the different immunoactive components in breastmilk contribute to immunity remains poorly understood. Here, we characterized human breastmilk T cells using single-cell RNA-Seq and flow cytometry. Breastmilk contained predominantly memory T cells, with expression of immune signaling genes, high proliferation, and an effector Th1/cytotoxic profile with high cytokine production capacities. Elevated activation was balanced by an enriched Treg population and immune regulatory markers in conventional memory T cells. Gene and surface expression of tissue-residency markers indicate that breastmilk T cells represented tissue-adapted rather than circulatory T cells. In addition, breastmilk T cells had a broad homing profile and higher activation markers in these migratory subsets. The partly overlapping transcriptome profile between breastmilk and breast tissue T cells, particularly cytotoxic T cells, might support a role in local immune defense in the mammary gland. However, unique features of breastmilk, such as Tregs, might imply an additional role in neonatal immune support. We found some correlations between the breastmilk T cell profile and clinical parameters, most notably with maternal and household factors. Together, our data suggest that breastmilk contains an adapted T cell population that exerts their function in specific tissue sites.
Collapse
Affiliation(s)
- Elise S. Saager
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Arthur H. van Stigt
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Butstabong Lerkvaleekul
- Division of Rheumatology, Department of Pediatrics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Lisanne Lutter
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht, Netherlands
- Department of Pathology, Amsterdam University Medical Centre, Amsterdam, Netherlands
| | - Anneke H. Hellinga
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht, Netherlands
| | - M. Marlot van der Wal
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Louis J. Bont
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht, Netherlands
- Department of Paediatric Immunology and Infectious Diseases, Wilhelmina Children’s Hospital/University Medical Center Utrecht, Utrecht, Netherlands
- ReSViNET foundation, Zeist, Netherlands
| | - Jeanette H.W. Leusen
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Belinda van’t Land
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht, Netherlands
- CoE Immunology, Danone Global Research & Innovation Center, Utrecht, Netherlands
| | - Femke van Wijk
- Center for Translational Immunology, University Medical Centre Utrecht, Utrecht, Netherlands
| | | |
Collapse
|
6
|
Cherayil BJ, Jain N. From Womb to World: Exploring the Immunological Connections between Mother and Child. Immunohorizons 2024; 8:552-562. [PMID: 39172025 PMCID: PMC11374749 DOI: 10.4049/immunohorizons.2400032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 07/23/2024] [Indexed: 08/23/2024] Open
Abstract
Mother and child are immunologically interconnected by mechanisms that we are only beginning to understand. During pregnancy, multiple molecular and cellular factors of maternal origin are transferred across the placenta and influence the development and function of the fetal and newborn immune system. Altered maternal immune states arising from pregnancy-associated infections or immunizations have the potential to program offspring immune function in ways that may have long-term health consequences. In this study, we review current literature on the impact of prenatal infection and vaccination on the developing immune system, highlight knowledge gaps, and look to the horizon to envision maternal interventions that could benefit both the mother and her child.
Collapse
Affiliation(s)
- Bobby J. Cherayil
- Mucosal Immunology and Biology Research Center, Mass General for Children, Charlestown, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
| | - Nitya Jain
- Mucosal Immunology and Biology Research Center, Mass General for Children, Charlestown, MA
- Department of Pediatrics, Harvard Medical School, Boston, MA
- Center for Computational and Integrative Biology, Mass General Brigham, Boston, MA
| |
Collapse
|
7
|
Malinská N, Grobárová V, Knížková K, Černý J. Maternal-Fetal Microchimerism: Impacts on Offspring's Immune Development and Transgenerational Immune Memory Transfer. Physiol Res 2024; 73:315-332. [PMID: 39027950 PMCID: PMC11299782 DOI: 10.33549/physiolres.935296] [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: 12/06/2023] [Accepted: 02/06/2024] [Indexed: 07/27/2024] Open
Abstract
Maternal-fetal microchimerism is a fascinating phenomenon in which maternal cells migrate to the tissues of the offspring during both pregnancy and breastfeeding. These cells primarily consist of leukocytes and stem cells. Remarkably, these maternal cells possess functional potential in the offspring and play a significant role in shaping their immune system development. T lymphocytes, a cell population mainly found in various tissues of the offspring, have been identified as the major cell type derived from maternal microchimerism. These T lymphocytes not only exert effector functions but also influence the development of the offspring's T lymphocytes in the thymus and the maturation of B lymphocytes in the lymph nodes. Furthermore, the migration of maternal leukocytes also facilitates the transfer of immune memory across generations. Maternal microchimerism has also been observed to address immunodeficiencies in the offspring. This review article focuses on investigating the impact of maternal cells transported within maternal microchimerism on the immune system development of the offspring, as well as elucidating the effector functions of maternal cells that migrate through the placenta and breast milk to reach the offspring.
Collapse
Affiliation(s)
- N Malinská
- Laboratory of Cell Immunology, Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic.
| | | | | | | |
Collapse
|
8
|
Noel SC, Madranges JF, Gothié JDM, Ewald J, Milnerwood AJ, Kennedy TE, Scott ME. Maternal gastrointestinal nematode infection alters hippocampal neuroimmunity, promotes synaptic plasticity, and improves resistance to direct infection in offspring. Sci Rep 2024; 14:10773. [PMID: 38730262 PMCID: PMC11087533 DOI: 10.1038/s41598-024-60865-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
The developing brain is vulnerable to maternal bacterial and viral infections which induce strong inflammatory responses in the mother that are mimicked in the offspring brain, resulting in irreversible neurodevelopmental defects, and associated cognitive and behavioural impairments. In contrast, infection during pregnancy and lactation with the immunoregulatory murine intestinal nematode, Heligmosomoides bakeri, upregulates expression of genes associated with long-term potentiation (LTP) of synaptic networks in the brain of neonatal uninfected offspring, and enhances spatial memory in uninfected juvenile offspring. As the hippocampus is involved in spatial navigation and sensitive to immune events during development, here we assessed hippocampal gene expression, LTP, and neuroimmunity in 3-week-old uninfected offspring born to H. bakeri infected mothers. Further, as maternal immunity shapes the developing immune system, we assessed the impact of maternal H. bakeri infection on the ability of offspring to resist direct infection. In response to maternal infection, we found an enhanced propensity to induce LTP at Schaffer collateral synapses, consistent with RNA-seq data indicating accelerated development of glutamatergic synapses in uninfected offspring, relative to those from uninfected mothers. Hippocampal RNA-seq analysis of offspring of infected mothers revealed increased expression of genes associated with neurogenesis, gliogenesis, and myelination. Furthermore, maternal infection improved resistance to direct infection of H. bakeri in offspring, correlated with transfer of parasite-specific IgG1 to their serum. Hippocampal immunohistochemistry and gene expression suggest Th2/Treg biased neuroimmunity in offspring, recapitulating peripheral immunoregulation of H. bakeri infected mothers. These findings indicate maternal H. bakeri infection during pregnancy and lactation alters peripheral and neural immunity in uninfected offspring, in a manner that accelerates neural maturation to promote hippocampal LTP, and upregulates the expression of genes associated with neurogenesis, gliogenesis, and myelination.
Collapse
Affiliation(s)
- Sophia C Noel
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, 3801 University Street, Montreal, QC, H3A 2B4, Canada.
- Institute of Parasitology, McGill University (Macdonald Campus), 21,111 Lakeshore Road, Sainte-Anne de Bellevue, QC, H9X 3V9, Canada.
| | - Jeanne F Madranges
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Jean-David M Gothié
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Jessica Ewald
- Institute of Parasitology, McGill University (Macdonald Campus), 21,111 Lakeshore Road, Sainte-Anne de Bellevue, QC, H9X 3V9, Canada
| | - Austen J Milnerwood
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Timothy E Kennedy
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital, 3801 University Street, Montreal, QC, H3A 2B4, Canada
| | - Marilyn E Scott
- Institute of Parasitology, McGill University (Macdonald Campus), 21,111 Lakeshore Road, Sainte-Anne de Bellevue, QC, H9X 3V9, Canada.
| |
Collapse
|
9
|
Fernandes KA, Lim AI. Maternal-driven immune education in offspring. Immunol Rev 2024; 323:288-302. [PMID: 38445769 DOI: 10.1111/imr.13315] [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: 03/07/2024]
Abstract
Maternal environmental exposures, particularly during gestation and lactation, significantly influence the immunological development and long-term immunity of offspring. Mammalian immune systems develop through crucial inputs from the environment, beginning in utero and continuing after birth. These critical developmental windows are essential for proper immune system development and, once closed, may not be reopened. This review focuses on the mechanisms by which maternal exposures, particularly to pathogens, diet, and microbiota, impact offspring immunity. Mechanisms driving maternal-offspring immune crosstalk include transfer of maternal antibodies, changes in the maternal microbiome and microbiota-derived metabolites, and transfer of immune cells and cytokines via the placenta and breastfeeding. We further discuss the role of transient maternal infections, which are common during pregnancy, in providing tissue-specific immune education to offspring. We propose a "maternal-driven immune education" hypothesis, which suggests that offspring can use maternal encounters that occur during a critical developmental window to develop optimal immune fitness against infection and inflammation.
Collapse
Affiliation(s)
| | - Ai Ing Lim
- Department of Molecular Biology, Princeton University, Princeton, New Jersey, USA
| |
Collapse
|
10
|
Armistead B, Peters MQ, Houck J, Carlson M, Balle C, Mulugeta N, Gray CM, Jaspan HB, Harrington WE. Exposure to HIV alters the composition of maternal microchimeric T cells in infants. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.01.583002. [PMID: 38496450 PMCID: PMC10942331 DOI: 10.1101/2024.03.01.583002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Infants exposed to HIV but uninfected (iHEU) display altered cellular immunity and are at increased risk of infection through poorly understood mechanisms. We previously reported that iHEU have lower levels of maternal microchimerism (MMc), maternal cells transferred to the offspring in utero/during breastfeeding. We evaluated MMc levels in T cell subsets in iHEU and HIV unexposed infants (iHU) to determine whether a selective deficiency in MMc may contribute to altered cellular immunity. Across all infants, MMc levels were highest in CD8+ T cells; however, the level of MMc in the CD8 T cell subset was significantly lower in iHEU compared to iHU.
Collapse
Affiliation(s)
- Blair Armistead
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - M Quinn Peters
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - John Houck
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Marc Carlson
- Research Scientific Computing, Enterprise Analytics, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Christina Balle
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Nolawit Mulugeta
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Clive M Gray
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Division of Immunology, Biomedical Research Institute, Stellenbosch University, Cape Town, South Africa
| | - Heather B Jaspan
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Whitney E Harrington
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
11
|
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
|
12
|
Pačes J, Grobárová V, Zadražil Z, Knížková K, Malinská N, Tušková L, Boes M, Černý J. MHC II-EGFP Knock-in Mouse Model. Curr Protoc 2023; 3:e925. [PMID: 37934124 DOI: 10.1002/cpz1.925] [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: 11/08/2023]
Abstract
The MHC II-EGFP knock-in mouse model enables us to visualize and track MHC-II-expressing cells in vivo by expressing enhanced green fluorescent protein (EGFP) fused to the MHC class II molecule under the MHC II beta chain promoter. Using this model, we can easily identify MHC-II-expressing cells, including dendritic cells, B cells, macrophages, and ILC3s, which play a key role as antigen-presenting cells (APCs) for CD4+ T cells. In addition, we can also precisely identify and analyze APC-containing tissues and organs. Even after fixation, EGFP retains its fluorescence, so this model is suitable for immunofluorescence studies, facilitating an unbiased characterization of the histological context, especially with techniques such as light-sheet fluorescence microscopy. Furthermore, the MHC II-EGFP knock-in mouse model is valuable for studying the molecular mechanisms of MHC II gene regulation and expression by making it possible to correlate MHC II expression (MHC II-EGFP) with surface fraction through antibody detection, thereby shedding light on the intricate regulation of MHC II expression. Overall, this model is an essential asset for quantitative and systems immunological research, providing insights into immune cell dynamics and localization, with a tool for precise cell identification and with the ability to study MHC II gene regulation, thus furthering the understanding of immune responses and underlying mechanisms © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Characterization of antigen-specific MHC II loading compartment tubulation toward the immunological synapse Basic Protocol 2: Characterization of overall versus surface MHC II expression Basic Protocol 3: Identification and preparation of the lymphoid organs Basic Protocol 4: Quantification of APC content in lymphoid organs by fluorescence stereomicroscopy Basic Protocol 5: Quantification and measurement of intestinal lymphoid tissue by light-sheet fluorescence stereomicroscopy Basic Protocol 6: Visualization of corneal APCs Basic Protocol 7: Quantification of MHC II+ cells in maternal milk by flow cytometry Support Protocol 1: Cell surface staining and flow cytometry analysis of spleen mononuclear cells.
Collapse
Affiliation(s)
- Jan Pačes
- Laboratory of Cell Immunology, Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Valéria Grobárová
- Laboratory of Cell Immunology, Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Zdeněk Zadražil
- Laboratory of Cell Immunology, Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Karolina Knížková
- Laboratory of Cell Immunology, Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Nikola Malinská
- Laboratory of Cell Immunology, Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Liliana Tušková
- Laboratory of Cell Immunology, Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| | - Marianne Boes
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Jan Černý
- Laboratory of Cell Immunology, Department of Cell Biology, Faculty of Science, Charles University, Prague, Czech Republic
| |
Collapse
|
13
|
Bermejo-Haro MY, Camacho-Pacheco RT, Brito-Pérez Y, Mancilla-Herrera I. The hormonal physiology of immune components in breast milk and their impact on the infant immune response. Mol Cell Endocrinol 2023:111956. [PMID: 37236499 DOI: 10.1016/j.mce.2023.111956] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023]
Abstract
During pregnancy, the maternal body undergoes a considerable transformation regarding the anatomy, metabolism, and immune profile that, after delivery, allows for protection and nourishment of the offspring via lactation. Pregnancy hormones are responsible for the development and functionality of the mammary gland for breast milk production, but little is known about how hormones control its immune properties. Breast milk composition is highly dynamic, adapting to the nutritional and immunological needs that the infant requires in the first months of life and is responsible for the main immune modeling of breastfed newborns. Therefore, alterations in the mechanisms that control the endocrinology of mammary gland adaptation for lactation could disturb the properties of breast milk that prepare the neonatal immune system to respond to the first immunologic challenges. In modern life, humans are chronically exposed to endocrine disruptors (EDs), which alter the endocrine physiology of mammals, affecting the composition of breast milk and hence the neonatal immune response. In this review, we provide a landscape of the possible role of hormones in the control of passive immunity transferred by breast milk and the possible effect of maternal exposure to EDs on lactation, as well as their impacts on the development of neonatal immunity.
Collapse
Affiliation(s)
- Mextli Y Bermejo-Haro
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico; Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico; Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, 11340, Mexico
| | - Rodrigo T Camacho-Pacheco
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico; Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico; Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, 11340, Mexico
| | - Yesenia Brito-Pérez
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico; Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico; Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, 11340, Mexico
| | - Ismael Mancilla-Herrera
- Infectology and Immunology Department, National Institute of Perinatology (INPer), Mexico City, Mexico.
| |
Collapse
|
14
|
Shook LL, Edlow AG. Safety and Efficacy of Coronavirus Disease 2019 (COVID-19) mRNA Vaccines During Lactation. Obstet Gynecol 2023; 141:483-491. [PMID: 36649326 PMCID: PMC9975040 DOI: 10.1097/aog.0000000000005093] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 12/08/2022] [Indexed: 01/18/2023]
Abstract
In this review, we summarize the data on the safety and side-effect profile of coronavirus disease 2019 (COVID-19) vaccines during lactation to date, review what is known about mRNA vaccine components in breast milk, and discuss the efficacy of COVID-19 vaccines in providing immune protection for the breastfeeding infant. The Centers for Disease Control and Prevention and the American College of Obstetricians and Gynecologists recommend that lactating individuals receive COVID-19 mRNA vaccines and stay up to date on booster doses, including the bivalent COVID-19 booster. The lack of serious side effects in mothers or infants across numerous large studies and registries of COVID-19 vaccination in pregnancy and lactation is reassuring. Although small quantities of mRNA may be transiently detectable in breast milk after maternal vaccination, there are no data demonstrating that vaccine mRNA can survive the infant gastrointestinal tract and no evidence that breast milk from lactating individuals who have received a COVID-19 mRNA vaccine can cause harm to breastfeeding infants. In contrast, numerous studies demonstrate that the breast milk of vaccinated individuals contains severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-specific functional antibodies and T cells, which benefit the breastfeeding infant's developing immune system. Transfer of SARS-CoV-2-specific antibodies from mother to infant is highest when vaccination occurs during pregnancy compared with lactation, because the breastfeeding infant receives both long-lasting antibodies through the placenta and breast-milk antibodies through breast milk. With clear data demonstrating efficacy and safety and no data demonstrating harm to mother or infant after COVID-19 vaccine administration during lactation, any recommendations to avoid vaccination while breastfeeding or to withhold breast milk from the infant for any period of time after vaccination are not supported by available evidence.
Collapse
Affiliation(s)
- Lydia L. Shook
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA
| | - Andrea G. Edlow
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Massachusetts General Hospital and Harvard Medical School, Boston, MA
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
15
|
Svanes C, Holloway JW, Krauss-Etschmann S. Preconception origins of asthma, allergies and lung function: The influence of previous generations on the respiratory health of our children. J Intern Med 2023; 293:531-549. [PMID: 36861185 DOI: 10.1111/joim.13611] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/03/2023]
Abstract
Emerging research suggests that exposures occurring years before conception are important determinants of the health of future offspring and subsequent generations. Environmental exposures of both the father and mother, or exposure to disease processes such as obesity or infections, may influence germline cells and thereby cause a cascade of health outcomes in multiple subsequent generations. There is now increasing evidence that respiratory health is influenced by parental exposures that occur long before conception. The strongest evidence relates adolescent tobacco smoking and overweight in future fathers to increased asthma and lower lung function in their offspring, supported by evidence on parental preconception occupational exposures and air pollution. Although this literature is still sparse, the epidemiological analyses reveal strong effects that are consistent across studies with different designs and methodologies. The results are strengthened by mechanistic research from animal models and (scarce) human studies that have identified molecular mechanisms that can explain the epidemiological findings, suggesting transfer of epigenetic signals through germline cells, with susceptibility windows in utero (both male and female line) and prepuberty (male line). The concept that our lifestyles and behaviours may influence the health of our future children represents a new paradigm. This raises concerns for future health in decades to come with respect to harmful exposures but may also open for radical rethinking of preventive strategies that may improve health in multiple generations, reverse the imprint of our parents and forefathers, and underpin strategies that can break the vicious circle of propagation of health inequalities across generations.
Collapse
Affiliation(s)
- Cecilie Svanes
- Centre for International Health, Department of Global Public Health and Primary Care, University of Bergen, Bergen, Norway.,Department of Occupational Medicine, Haukeland University Hospital, Bergen, Norway
| | - John W Holloway
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Susanne Krauss-Etschmann
- Division of Early Life Origins of Chronic Lung Diseases, Research Center Borstel, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Borstel, Germany.,Institute of Experimental Medicine, Christian-Albrechts-Universität zu Kiel, Kiel, Germany
| |
Collapse
|
16
|
Armistead B, Jiang Y, Carlson M, Ford ES, Jani S, Houck J, Wu X, Jing L, Pecor T, Kachikis A, Yeung W, Nguyen T, Coig R, Minkah N, Larsen SE, Coler RN, Koelle DM, Harrington WE. Spike-specific T cells are enriched in breastmilk following SARS-CoV-2 mRNA vaccination. Mucosal Immunol 2023; 16:39-49. [PMID: 36642379 PMCID: PMC9836998 DOI: 10.1016/j.mucimm.2023.01.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/20/2022] [Accepted: 01/07/2023] [Indexed: 01/15/2023]
Abstract
Human breastmilk is rich in T cells; however, their specificity and function are largely unknown. We compared the phenotype, diversity, and antigen specificity of T cells in breastmilk and peripheral blood of lactating individuals who received SARS-CoV-2 messenger RNA (mRNA) vaccination. Relative to blood, breastmilk contained higher frequencies of T effector and central memory populations that expressed mucosal-homing markers. T cell receptor sequence overlap was limited between blood and breastmilk. Overabundant breastmilk clones were observed in all individuals, were diverse, and contained complementarity-determining regions in three sequences with known epitope specificity, including to SARS-CoV-2 spike. SARS-CoV-2 spike-specific T cell receptors were more frequent in breastmilk compared to blood and expanded in breastmilk following a 3rd mRNA vaccine dose. Our observations indicate that the lactating breast contains a distinct T cell population that can be modulated by maternal vaccination with potential implications for passive infant protection.
Collapse
Affiliation(s)
- Blair Armistead
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Yonghou Jiang
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Marc Carlson
- Research Scientific Computing, Enterprise Analytics, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Emily S Ford
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA; Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Saumya Jani
- Department of Medicine, University of Washington, Seattle, Washington, USA; Department of Laboratory Medicine & Pathology, University of Washington, Seattle, Washington, USA
| | - John Houck
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Xia Wu
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Lichen Jing
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Tiffany Pecor
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Alisa Kachikis
- Department of Obstetrics & Gynecology, University of Washington, Seattle, Washington, USA
| | - Winnie Yeung
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Tina Nguyen
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Rene Coig
- Department of Laboratory Medicine & Pathology, University of Washington, Seattle, Washington, USA
| | - Nana Minkah
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA; Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Sasha E Larsen
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Rhea N Coler
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA; Department of Pediatrics, University of Washington, Seattle, Washington, USA; Department of Global Health, University of Washington, Seattle, Washington, USA
| | - David M Koelle
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA; Department of Medicine, University of Washington, Seattle, Washington, USA; Department of Laboratory Medicine & Pathology, University of Washington, Seattle, Washington, USA; Department of Global Health, University of Washington, Seattle, Washington, USA; Benaroya Research Institute, Seattle, Washington, USA
| | - Whitney E Harrington
- Center for Global Infectious Disease Research, Seattle Children's Research Institute, Seattle, Washington, USA; Department of Pediatrics, University of Washington, Seattle, Washington, USA; Department of Global Health, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
17
|
Antibiotic Treatment during Gestation Enhances Susceptibility to Mycobacterium tuberculosis in Offspring. Microbiol Spectr 2022; 10:e0249122. [PMID: 36314979 PMCID: PMC9769670 DOI: 10.1128/spectrum.02491-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Whether antibiotic treatment during gestation impacts T cell immunity to vaccination in offspring is unexplored. Dams treated with polymyxin B (PMB) during gestation (Mg) displayed altered microbial communities prior to delivery compared to control dams (Mc). Differences in microbiota were also evident in pups born to polymyxin B-treated dams (Pg) compared to control pups (Pc). When pups were immunized with Bacille Calmette-Guerin (BCG), we observed no difference in TB10.4-specific T cells between Pc and Pg 4 weeks postimmunization. Significantly fewer splenic CD4 T cells from BCG-vaccinated Pg produced interleukin-2 (IL-2) upon stimulation, suggesting a possible functional deficiency. There was no difference in purified protein derivative (PPD)-specific IgG between Pc and Pg at this time point. However, when infected with Mycobacterium tuberculosis, Pg displayed significantly higher bacterial burden in the lung than Pc. Our results show that maternal PMB treatment during gestation may not impact splenic antigen-specific T cell responses following BCG vaccination but alters susceptibility to M. tuberculosis in offspring. IMPORTANCE The composition of the pioneer microbiota that colonize the infant gut are determined by the mother. Polymyxin B-induced changes in the maternal microbiota during pregnancy impact the offspring gut microbiota but not vaccine-specific CD4 T cell response. However, when infected with Mycobacterium tuberculosis, offspring born to mothers with an altered gut microbiota are susceptible to infection compared to those born to mothers not exposed to antibiotics.
Collapse
|
18
|
Armistead B, Jiang Y, Carlson M, Ford ES, Jani S, Houck J, Wu X, Jing L, Pecor T, Kachikis A, Yeung W, Nguyen T, Minkah N, Larsen SE, Coler RN, Koelle DM, Harrington WE. Spike-specific T cells are enriched in breastmilk following SARS-CoV-2 mRNA vaccination. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2022:2021.12.03.21267036. [PMID: 36203549 PMCID: PMC9536058 DOI: 10.1101/2021.12.03.21267036] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Human breastmilk is rich in T cells; however, their specificity and function are largely unknown. We compared the phenotype, diversity, and antigen specificity of T cells in the breastmilk and peripheral blood of lactating individuals who received SARS-CoV-2 mRNA vaccination. Relative to blood, breastmilk contained higher frequencies of T effector and central memory populations that expressed mucosal-homing markers. T cell receptor (TCR) sequence overlap was limited between blood and breastmilk. Overabundan t breastmilk clones were observed in all individuals, were diverse, and contained CDR3 sequences with known epitope specificity including to SARS-CoV-2 Spike. Spike-specific TCRs were more frequent in breastmilk compared to blood and expanded in breastmilk following a third mRNA vaccine dose. Our observations indicate that the lactating breast contains a distinct T cell population that can be modulated by maternal vaccination with potential implications for infant passive protection. One-Sentence Summary The breastmilk T cell repertoire is distinct and enriched for SARS-CoV-2 Spike-specificity after maternal mRNA vaccination.
Collapse
Affiliation(s)
- Blair Armistead
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA, USA
| | - Yonghou Jiang
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA, USA
| | - Marc Carlson
- Research Scientific Computing, Enterprise Analytics, Seattle Children’s Research Institute; Seattle, WA, USA
| | - Emily S Ford
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center; Seattle, WA, USA
- Department of Medicine, University of Washington; Seattle, WA, USA
| | - Saumya Jani
- Department of Medicine, University of Washington; Seattle, WA, USA
- Department of Laboratory Medicine & Pathology, University of Washington; Seattle, WA, USA
| | - John Houck
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA, USA
| | - Xia Wu
- Department of Medicine, University of Washington; Seattle, WA, USA
| | - Lichen Jing
- Department of Medicine, University of Washington; Seattle, WA, USA
| | - Tiffany Pecor
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA, USA
| | - Alisa Kachikis
- Department of Obstetrics & Gynecology, University of Washington; Seattle, WA, USA
| | - Winnie Yeung
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA, USA
| | - Tina Nguyen
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA, USA
| | - Nana Minkah
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA, USA
- Department of Pediatrics, University of Washington; Seattle, WA, USA
| | - Sasha E Larsen
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA, USA
| | - Rhea N Coler
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA, USA
- Department of Global Health, University of Washington; Seattle, WA, USA
- Department of Pediatrics, University of Washington; Seattle, WA, USA
| | - David M Koelle
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center; Seattle, WA, USA
- Department of Medicine, University of Washington; Seattle, WA, USA
- Department of Laboratory Medicine & Pathology, University of Washington; Seattle, WA, USA
- Department of Global Health, University of Washington; Seattle, WA, USA
- Benaroya Research Institute; Seattle, WA, USA
| | - Whitney E Harrington
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute; Seattle, WA, USA
- Department of Global Health, University of Washington; Seattle, WA, USA
- Department of Pediatrics, University of Washington; Seattle, WA, USA
| |
Collapse
|
19
|
Narayanaswamy V, Pentecost BT, Telfer JC, Burnside AS, Schneider SS, Alfandari D, Baker RL, Saiju A, Nodiff S, Arcaro KF. Durable antibody and effector memory T cell responses in breastmilk from women with SARS-CoV-2. Front Immunol 2022; 13:985226. [PMID: 36172379 PMCID: PMC9512087 DOI: 10.3389/fimmu.2022.985226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Accepted: 08/24/2022] [Indexed: 11/24/2022] Open
Abstract
Background Given that only 25% of pregnant women elect to receive a COVID-19 vaccine, maternal SARS-CoV-2 infection remains an important route of conferring protective passive immunity to breastfed infants of mothers who are not vaccinated. Methods We enrolled 30 lactating participants between December 2020 and March 2021 who had a positive PCR-test and their first COVID-19 symptoms within the previous 21 days. Participants were asked to provide serial bilateral milk samples at 12 timepoints (~ every 3 days) over a period of 35 days. A second set of samples was collected at least four months after the beginning of the first set. Participants also were asked to provide their dried blood spots and infant stool samples. All samples were tested for receptor-binding domain (RBD)-specific immunoglobulin (Ig)A, IgG, and IgM. Milk samples were assessed for neutralizing ability against the spike protein and four SARS-CoV-2 variants: D614G, Alpha (B.1.1.7), Beta (B.1.351), and Gamma (P.1). Permeability of the breast epithelium was assessed by measuring the sodium to potassium ions (Na:K) in milk. Using flow cytometry, memory CD4 and CD8 T cells (CD45RO+ and CCR7+/-) and mucosal-homing CD4 and CD8 T cells (CD103+) were determined in cells from milk expressed at 35 days and at least 4 months after their first milk donation. Results Milk antibodies from SARS-CoV-2 positive participants neutralized the spike complex. Milk from 73, 90, and 53% of participants had binding reactivities to RBD-specific IgA, IgG, and IgM, respectively. In contrast to blood spots, which showed increased levels of IgG, but not IgA or IgM, the COVID-19 response in milk was associated with a robust IgA response. Twenty-seven percent of participants had increased breast-epithelium permeability, as indicated by Na:K ≥ 0.6. The percentage of CD45RO+CCR7- effector-memory T cells in the day ≥120 milk samples was significantly higher than day 35 samples (P< 0.05). Conclusions Antibodies in milk from participants with recent SARS-CoV-2 infection and those who recovered can neutralize the spike complex. For the first time we show that breastmilk T cells are enriched for mucosal memory T cells, further emphasizing the passive protection against SARS-CoV-2 conferred to infants via breastmilk.
Collapse
Affiliation(s)
- Vignesh Narayanaswamy
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Brian T. Pentecost
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Janice C. Telfer
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Amy S. Burnside
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Sallie S. Schneider
- Pioneer Valley Life Sciences Institute, Baystate Medical Center, Springfield, MA, United States
| | - Dominique Alfandari
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Ryan L. Baker
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Aman Saiju
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Sam Nodiff
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| | - Kathleen F. Arcaro
- Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, United States
| |
Collapse
|
20
|
Stunting as a Risk Factor of Soil-Transmitted Helminthiasis in Children: A Literature Review. Interdiscip Perspect Infect Dis 2022; 2022:8929025. [PMID: 35967932 PMCID: PMC9365611 DOI: 10.1155/2022/8929025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 07/08/2022] [Accepted: 07/09/2022] [Indexed: 11/18/2022] Open
Abstract
As a high-burden neglected tropical disease, soil-transmitted helminth (STH) infections remain a major problem in the world, especially among children under five years of age. Since young children are at high risk of being infected, STH infection can have a long-term negative impact on their life, including impaired growth and development. Stunting, a form of malnutrition in young children, has been long assumed as one of the risk factors in acquiring the STH infections. However, the studies on STH infection in children under five with stunting have been lacking, resulting in poor identification of the risk. Accordingly, we collected and reviewed existing related research articles to provide an overview of STH infection in a susceptible population of stunted children under five years of age in terms of prevalence and risk factors. There were 17 studies included in this review related to infection with Ascaris lumbricoides, Trichuris trichiura, hookworm, and Strongyloides stercoralis from various countries. The prevalence of STH infection in stunted children ranged from 12.5% to 56.5%. Increased inflammatory markers and intestinal microbiota dysbiosis might have increased the intensity of STH infection in stunted children that caused impairment in the immune system. While the age from 2 to 5 years along with poor hygiene and sanitation has shown to be the most common risk factors of STH infections in stunted children; currently there are no studies that show direct results of stunting as a risk factor for STH infection. While stunting itself may affect the pathogenesis of STH infection, further research on stunting as a risk factor for STH infection is encouraged.
Collapse
|
21
|
Balle C, Armistead B, Kiravu A, Song X, Happel AU, Hoffmann AA, Kanaan SB, Nelson JL, Gray CM, Jaspan HB, Harrington WE. Factors influencing maternal microchimerism throughout infancy and its impact on infant T cell immunity. J Clin Invest 2022; 132:e148826. [PMID: 35550376 PMCID: PMC9246390 DOI: 10.1172/jci148826] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/10/2022] [Indexed: 11/17/2022] Open
Abstract
Determinants of the acquisition and maintenance of maternal microchimerism (MMc) during infancy and the impact of MMc on infant immune responses are unknown. We examined factors that influence MMc detection and level across infancy and the effect of MMc on T cell responses to bacillus Calmette-Guérin (BCG) vaccination in a cohort of HIV-exposed, uninfected and HIV-unexposed infants in South Africa. MMc was measured in whole blood from 58 infants using a panel of quantitative PCR assays at day 1, and 7, 15, and 36 weeks of life. Infants received BCG at birth, and selected whole blood samples from infancy were stimulated in vitro with BCG and assessed for polyfunctional CD4+ T cell responses. MMc was present in most infants across infancy, with levels ranging from 0 to 1,193/100,000 genomic equivalents and was positively impacted by absence of maternal HIV, maternal and infant HLA compatibility, infant female sex, and exclusive breastfeeding. Initiation of maternal antiretroviral therapy prior to pregnancy partially restored MMc level in HIV-exposed, uninfected infants. Birth MMc was associated with an improved polyfunctional CD4+ T cell response to BCG. These data emphasize that both maternal and infant factors influence the level of MMc, which may subsequently affect infant T cell responses.
Collapse
Affiliation(s)
- Christina Balle
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Blair Armistead
- Seattle Children’s Research Institute, Seattle, Washington, USA
| | - Agano Kiravu
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Xiaochang Song
- Seattle Children’s Research Institute, Seattle, Washington, USA
- University of Washington School of Medicine, Seattle, Washington, USA
| | - Anna-Ursula Happel
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Angela A. Hoffmann
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Sami B. Kanaan
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - J. Lee Nelson
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Clive M. Gray
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Heather B. Jaspan
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Seattle Children’s Research Institute, Seattle, Washington, USA
- Department of Pediatrics and
- Department of Global Health, University of Washington, Seattle, Washington, USA
| | - Whitney E. Harrington
- Seattle Children’s Research Institute, Seattle, Washington, USA
- Department of Pediatrics and
| |
Collapse
|
22
|
Langel SN, Blasi M, Permar SR. Maternal immune protection against infectious diseases. Cell Host Microbe 2022; 30:660-674. [PMID: 35550669 DOI: 10.1016/j.chom.2022.04.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The maternal immune system protects developing offspring against pathogens before birth via transplacental transfer and after birth through secreted milk. This transferred maternal immunity influences each generation's susceptibility to infections and responsiveness to immunization. Thus, boosting immunity in the maternal-neonatal dyad is a potentially valuable public health strategy. Additionally, at critical times during fetal and postnatal development, environmental factors and immune stimuli influence immune development. These "windows of opportunity" offer a chance to identify both risk and protective factors that promote long-term health and limit disease. Here, we review pre- and postpartum maternal immune factors that protect against infectious agents in offspring and how they may shape the infant's immune landscape over time. Additionally, we discuss the influence of maternal immunity on the responsiveness to immunization in early life. Lastly, when maternal factors are insufficient to prevent neonatal infectious diseases, we discuss pre- and postnatal therapeutic strategies for the maternal-neonatal dyad.
Collapse
Affiliation(s)
- Stephanie N Langel
- Department of Surgery, Duke Center for Human Systems Immunology, Durham, NC, USA
| | - Maria Blasi
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA; Department of Medicine, Division of Infectious Diseases, Duke University Medical Center, Durham, NC, USA
| | - Sallie R Permar
- Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
23
|
López-Cervantes JP, Lønnebotn M, Jogi NO, Calciano L, Kuiper IN, Darby MG, Dharmage SC, Gómez-Real F, Hammer B, Bertelsen RJ, Johannessen A, Würtz AML, Mørkve Knudsen T, Koplin J, Pape K, Skulstad SM, Timm S, Tjalvin G, Krauss-Etschmann S, Accordini S, Schlünssen V, Kirkeleit J, Svanes C. The Exposome Approach in Allergies and Lung Diseases: Is It Time to Define a Preconception Exposome? INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:12684. [PMID: 34886409 PMCID: PMC8657011 DOI: 10.3390/ijerph182312684] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 12/30/2022]
Abstract
Emerging research suggests environmental exposures before conception may adversely affect allergies and lung diseases in future generations. Most studies are limited as they have focused on single exposures, not considering that these diseases have a multifactorial origin in which environmental and lifestyle factors are likely to interact. Traditional exposure assessment methods fail to capture the interactions among environmental exposures and their impact on fundamental biological processes, as well as individual and temporal factors. A valid estimation of exposure preconception is difficult since the human reproductive cycle spans decades and the access to germ cells is limited. The exposome is defined as the cumulative measure of external exposures on an organism (external exposome), and the associated biological responses (endogenous exposome) throughout the lifespan, from conception and onwards. An exposome approach implies a targeted or agnostic analysis of the concurrent and temporal multiple exposures, and may, together with recent technological advances, improve the assessment of the environmental contributors to health and disease. This review describes the current knowledge on preconception environmental exposures as related to respiratory health outcomes in offspring. We discuss the usefulness and feasibility of using an exposome approach in this research, advocating for the preconception exposure window to become included in the exposome concept.
Collapse
Affiliation(s)
- Juan Pablo López-Cervantes
- Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, 5020 Bergen, Norway; (M.L.); (A.J.); (G.T.); (J.K.); (C.S.)
- Department of Occupational Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (N.O.J.); (T.M.K.); (S.M.S.)
| | - Marianne Lønnebotn
- Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, 5020 Bergen, Norway; (M.L.); (A.J.); (G.T.); (J.K.); (C.S.)
- Department of Occupational Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (N.O.J.); (T.M.K.); (S.M.S.)
| | - Nils Oskar Jogi
- Department of Occupational Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (N.O.J.); (T.M.K.); (S.M.S.)
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (F.G.-R.); (R.J.B.)
| | - Lucia Calciano
- Unit of Epidemiology and Medical Statistics, Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy; (L.C.); (S.A.)
| | | | - Matthew G. Darby
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town 7925, South Africa;
| | - Shyamali C. Dharmage
- School of Population and Global Health, University of Melbourne, Melbourne, VIC 3010, Australia; (S.C.D.); (J.K.)
| | - Francisco Gómez-Real
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (F.G.-R.); (R.J.B.)
- Department of Obstetrics and Gynecology, Haukeland University Hospital, 5053 Bergen, Norway
| | - Barbara Hammer
- Department of Pulmonology, Medical University of Vienna, 1090 Vienna, Austria;
| | | | - Ane Johannessen
- Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, 5020 Bergen, Norway; (M.L.); (A.J.); (G.T.); (J.K.); (C.S.)
| | - Anne Mette Lund Würtz
- Danish Ramazzini Centre, Department of Public Health—Work, Environment and Health, Aarhus University, 8000 Aarhus, Denmark; (A.M.L.W.); (K.P.); (V.S.)
| | - Toril Mørkve Knudsen
- Department of Occupational Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (N.O.J.); (T.M.K.); (S.M.S.)
- Department of Clinical Science, University of Bergen, 5021 Bergen, Norway; (F.G.-R.); (R.J.B.)
| | - Jennifer Koplin
- School of Population and Global Health, University of Melbourne, Melbourne, VIC 3010, Australia; (S.C.D.); (J.K.)
- Murdoch Children’s Research Institute, Melbourne, VIC 3052, Australia
| | - Kathrine Pape
- Danish Ramazzini Centre, Department of Public Health—Work, Environment and Health, Aarhus University, 8000 Aarhus, Denmark; (A.M.L.W.); (K.P.); (V.S.)
| | - Svein Magne Skulstad
- Department of Occupational Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (N.O.J.); (T.M.K.); (S.M.S.)
| | - Signe Timm
- Department of Regional Health Research, University of Southern Denmark, 5230 Odense, Denmark;
- Research Unit, Kolding Hospital, University Hospital of Southern Denmark, 6000 Kolding, Denmark
| | - Gro Tjalvin
- Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, 5020 Bergen, Norway; (M.L.); (A.J.); (G.T.); (J.K.); (C.S.)
- Department of Occupational Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (N.O.J.); (T.M.K.); (S.M.S.)
| | | | - Simone Accordini
- Unit of Epidemiology and Medical Statistics, Department of Diagnostics and Public Health, University of Verona, 37134 Verona, Italy; (L.C.); (S.A.)
| | - Vivi Schlünssen
- Danish Ramazzini Centre, Department of Public Health—Work, Environment and Health, Aarhus University, 8000 Aarhus, Denmark; (A.M.L.W.); (K.P.); (V.S.)
- National Research Centre for the Working Environment, 2100 Copenhagen, Denmark
| | - Jorunn Kirkeleit
- Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, 5020 Bergen, Norway; (M.L.); (A.J.); (G.T.); (J.K.); (C.S.)
- Department of Occupational Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (N.O.J.); (T.M.K.); (S.M.S.)
| | - Cecilie Svanes
- Center for International Health, Department of Global Public Health and Primary Care, University of Bergen, 5020 Bergen, Norway; (M.L.); (A.J.); (G.T.); (J.K.); (C.S.)
- Department of Occupational Medicine, Haukeland University Hospital, 5021 Bergen, Norway; (N.O.J.); (T.M.K.); (S.M.S.)
| |
Collapse
|
24
|
Taylor M, Pillaye J, Horsnell WGC. Inherent maternal type 2 immunity: Consequences for maternal and offspring health. Semin Immunol 2021; 53:101527. [PMID: 34838445 DOI: 10.1016/j.smim.2021.101527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/10/2021] [Accepted: 11/10/2021] [Indexed: 02/08/2023]
Abstract
An inherent elevation in type 2 immunity is a feature of maternal and offspring immune systems. This has diverse implications for maternal and offspring biology including influencing success of pregnancy, offspring immune development and maternal and offspring ability to control infection and diseases such as allergies. In this review we provide a broad insight into how this immunological feature of pregnancy and early life impacts both maternal and offspring biology. We also suggest how understanding of this axis of immune influence is and may be utilised to improve maternal and offspring health.
Collapse
Affiliation(s)
- Matthew Taylor
- Institute of Immunology and Infection Research, Ashworth Laboratories, The Kings Buildings, University of Edinburgh, West Mains Road, Edinburgh, EH9 3JT, UK.
| | - Jamie Pillaye
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - William Gordon Charles Horsnell
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK; Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa), Institute of Infectious Disease and Molecular Medicine (IDM), Department of Pathology, Division of Immunology, Faculty of Health Science, University of Cape Town, Cape Town, 7925, South Africa.
| |
Collapse
|
25
|
El Ahdab N, Haque M, Madogwe E, Koski KG, Scott ME. Maternal nematode infection upregulates expression of Th2/Treg and diapedesis related genes in the neonatal brain. Sci Rep 2021; 11:22082. [PMID: 34764345 PMCID: PMC8585879 DOI: 10.1038/s41598-021-01510-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 10/26/2021] [Indexed: 11/28/2022] Open
Abstract
Intestinal nematode infections common during pregnancy have recently been shown to have impacts that extend to their uninfected offspring including altered brain gene expression. If maternal immune signals reach the neonatal brain, they might alter neuroimmune development. We explored expression of genes associated with four distinct types of T cells (Th1, Th2, Th17, Treg) and with leukocyte transendothelial migration and endocytosis transport across the blood–brain barrier (BBB) in the postnatal brain of offspring of nematode-infected mice, through secondary analysis of a whole brain gene expression database. Th1/Th17 expression was lowered by maternal infection as evidenced by down-regulated expression of IL1β, Th1 receptors and related proteins, and of IL22 and several Th17 genes associated with immunopathology. In contrast, Th2/Treg related pathways were upregulated as shown by higher expression of IL4 and TGF-β family genes. Maternal infection also upregulated expression of pathways and integrin genes involved in transport of leukocytes in between endothelial cells but downregulated endosome vesicle formation related genes that are necessary for endocytosis of immunoglobulins across the BBB. Taken together, pup brain gene expression indicates that maternal nematode infection enhanced movement of leukocytes across the neonatal BBB and promoted a Th2/Treg environment that presumably minimizes the proinflammatory Th1 response in the pup brain.
Collapse
Affiliation(s)
- Nawal El Ahdab
- Institute of Parasitology, McGill University (Macdonald Campus), 21,111 Lakeshore Road, Ste-Anne de Bellevue, QC, H9X 3V9, Canada
| | - Manjurul Haque
- Institute of Parasitology, McGill University (Macdonald Campus), 21,111 Lakeshore Road, Ste-Anne de Bellevue, QC, H9X 3V9, Canada
| | - Ejimedo Madogwe
- Department of Animal Science, McGill University (Macdonald Campus), 21,111 Lakeshore Road, Ste-Anne de Bellevue, QC, H9X 3V9, Canada
| | - Kristine G Koski
- School of Human Nutrition, McGill University (Macdonald Campus), 21,111 Lakeshore Road, Ste-Anne de Bellevue, QC, H9X 3V9, Canada
| | - Marilyn E Scott
- Institute of Parasitology, McGill University (Macdonald Campus), 21,111 Lakeshore Road, Ste-Anne de Bellevue, QC, H9X 3V9, Canada.
| |
Collapse
|
26
|
Lacorcia M, Bhattacharjee S, Laubhahn K, Alhamdan F, Ram M, Muschaweckh A, Potaczek DP, Kosinska A, Garn H, Protzer U, Renz H, Prazeres da Costa C. Fetomaternal immune cross talk modifies T-cell priming through sustained changes to DC function. J Allergy Clin Immunol 2021; 148:843-857.e6. [PMID: 33684437 DOI: 10.1016/j.jaci.2021.02.031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 02/18/2021] [Accepted: 02/26/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND Prenatal exposure to infections can modify immune development. These environmental disturbances during early life potentially alter the incidence of inflammatory disorders as well as priming of immune responses. Infection with the helminth Schistosoma mansoni is widely studied for its ability to alter immune responsiveness and is associated with variations in coinfection, allergy, and vaccine efficacy in endemic populations. OBJECTIVE Exposure to maternal schistosomiasis during early life, even without transmission of infection, can result in priming effects on offspring immune responses to bystander antigenic challenges as related to allergic responsiveness and vaccination, with this article seeking to further clarify the effects and underlying immunologic imprinting. METHODS Here, we have combined a model of chronic maternal schistosomiasis infection with a thorough analysis of subsequent offspring immune responses to allergy and vaccination models, including viral challenge and steady-state changes to immune cell compartments. RESULTS We have demonstrated that maternal schistosomiasis alters CD4+ responses during allergic sensitization and challenge in a skewed IL-4/B-cell-dominant response to antigenic challenge associated with limited inflammatory response. Beyond that, we have uncovered previously unidentified alterations to CD8+ T-cell responses during immunization that are dependent on vaccine formulation and have functional impact on the efficacy of vaccination against viral infection in a murine hepatitis B virus model. CONCLUSION In addition to steady-state modifications to CD4+ T-cell polarization and B-cell priming, we have traced these modified CD8+ responses to an altered dendritic cell phenotype sustained into adulthood, providing evidence for complex priming effects imparted by infection via fetomaternal cross talk.
Collapse
Affiliation(s)
- Matthew Lacorcia
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, Munich, Germany
| | - Sonakshi Bhattacharjee
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, Munich, Germany
| | - Kristina Laubhahn
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, Munich, Germany; Pediatric Allergology, Department of Pediatrics, Dr von Hauner Children's Hospital, University Hospital, Ludwig Maximilian University Munich, Munich, Germany; German Center for Lung Research, Ludwig Maximilian University Munich, Munich, Germany
| | - Fahd Alhamdan
- Biochemical Pharmacological Center, Translational Inflammation Division & Core Facility for Single Cell Multiomics, Philipps University Marburg, Marburg, Germany
| | - Marija Ram
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, Munich, Germany
| | - Andreas Muschaweckh
- Department of Neurology, University Hospital rechts der Isar, Technical University of Munich, Munich, Germany
| | - Daniel P Potaczek
- Biochemical Pharmacological Center, Translational Inflammation Division & Core Facility for Single Cell Multiomics, Philipps University Marburg, Marburg, Germany
| | - Anna Kosinska
- Institute for Virology Technical University of Munich, Munich, Germany
| | - Holger Garn
- Biochemical Pharmacological Center, Translational Inflammation Division & Core Facility for Single Cell Multiomics, Philipps University Marburg, Marburg, Germany
| | - Ulrike Protzer
- Institute for Virology Technical University of Munich, Munich, Germany
| | - Harald Renz
- Biochemical Pharmacological Center, Translational Inflammation Division & Core Facility for Single Cell Multiomics, Philipps University Marburg, Marburg, Germany
| | - Clarissa Prazeres da Costa
- Institute for Medical Microbiology, Immunology and Hygiene, Technical University of Munich, Munich, Germany.
| |
Collapse
|
27
|
Lim AI, McFadden T, Link VM, Han SJ, Karlsson RM, Stacy A, Farley TK, Lima-Junior DS, Harrison OJ, Desai JV, Lionakis MS, Shih HY, Cameron HA, Belkaid Y. Prenatal maternal infection promotes tissue-specific immunity and inflammation in offspring. Science 2021; 373:373/6558/eabf3002. [PMID: 34446580 DOI: 10.1126/science.abf3002] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 07/07/2021] [Indexed: 12/18/2022]
Abstract
The immune system has evolved in the face of microbial exposure. How maternal infection experienced at distinct developmental stages shapes the offspring immune system remains poorly understood. Here, we show that during pregnancy, maternally restricted infection can have permanent and tissue-specific impacts on offspring immunity. Mechanistically, maternal interleukin-6 produced in response to infection can directly impose epigenetic changes on fetal intestinal epithelial stem cells, leading to long-lasting impacts on intestinal immune homeostasis. As a result, offspring of previously infected dams develop enhanced protective immunity to gut infection and increased inflammation in the context of colitis. Thus, maternal infection can be coopted by the fetus to promote long-term, tissue-specific fitness, a phenomenon that may come at the cost of predisposition to inflammatory disorders.
Collapse
Affiliation(s)
- Ai Ing Lim
- Metaorganism Immunity Section, Laboratory of Immune System Biology and Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Taryn McFadden
- Metaorganism Immunity Section, Laboratory of Immune System Biology and Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Verena M Link
- Metaorganism Immunity Section, Laboratory of Immune System Biology and Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Seong-Ji Han
- Metaorganism Immunity Section, Laboratory of Immune System Biology and Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Rose-Marie Karlsson
- Section on Neuroplasticity, Mood and Anxiety Disorders Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Apollo Stacy
- Metaorganism Immunity Section, Laboratory of Immune System Biology and Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.,Postdoctoral Research Associate Training Program, National Institute of General Medical Sciences, Bethesda, MD 20892, USA
| | - Taylor K Farley
- Metaorganism Immunity Section, Laboratory of Immune System Biology and Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.,Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7FY, UK
| | - Djalma S Lima-Junior
- Metaorganism Immunity Section, Laboratory of Immune System Biology and Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Oliver J Harrison
- Metaorganism Immunity Section, Laboratory of Immune System Biology and Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jigar V Desai
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michail S Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Han-Yu Shih
- Neuro-Immune Regulome Unit, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Heather A Cameron
- Metaorganism Immunity Section, Laboratory of Immune System Biology and Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.,Section on Neuroplasticity, Mood and Anxiety Disorders Program, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Immune System Biology and Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA. .,NIAID Microbiome Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
28
|
Wood H, Acharjee A, Pearce H, Quraishi MN, Powell R, Rossiter A, Beggs A, Ewer A, Moss P, Toldi G. Breastfeeding promotes early neonatal regulatory T-cell expansion and immune tolerance of non-inherited maternal antigens. Allergy 2021; 76:2447-2460. [PMID: 33432577 DOI: 10.1111/all.14736] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 12/01/2020] [Accepted: 12/15/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND Breastfeeding is associated with long-term health benefits, such as a lower incidence of childhood infections, asthma, obesity and autoimmune disorders. However, little is known regarding how the maternal and neonatal immune systems interact after parturition when the neonate receives nutrition from maternal breast milk. METHODS We undertook a comparative analysis of immune repertoire and function at birth and 3 weeks of age in a cohort of 38 term neonates born by caesarean section grouped according to feeding method (breast milk versus formula). We used flow cytometry to study the immune phenotype in neonatal and maternal blood samples and mixed lymphocyte reactions to establish the proliferation response of neonatal versus maternal lymphocytes and vice versa. The microbiome of neonatal stool samples was also investigated using 16S rRNA sequencing. RESULTS We show that the proportion of regulatory T cells (Tregs) increases in this period and is nearly twofold higher in exclusively breastfed neonates compared with those who received formula milk only. Moreover, breastfed neonates show a specific and Treg-dependent reduction in proliferative T-cell responses to non-inherited maternal antigens (NIMA), associated with a reduction in inflammatory cytokine production. We also observed the enrichment of short chain fatty acid producing taxa (Veillonella and Gemella) in stool samples of exclusively breastfed neonates. CONCLUSIONS These data indicate that exposure of the neonate to maternal cells through breastfeeding acts to drive the maturation of Tregs and 'tolerizes' the neonate towards NIMA.
Collapse
Affiliation(s)
- Hannah Wood
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
- Department of Neonatology, Birmingham Women's and Children's NHS FT, Birmingham, UK
| | - Animesh Acharjee
- Centre for Computational Biology, Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
- Institute of Translational Medicine, University Hospitals Birmingham NHS FT, Birmingham, UK
- NIHR Surgical Reconstruction and Microbiology Research Centre, University Hospitals Birmingham NHS FT, Birmingham, UK
| | - Hayden Pearce
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | | | - Richard Powell
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Amanda Rossiter
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - Andrew Beggs
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Andrew Ewer
- Department of Neonatology, Birmingham Women's and Children's NHS FT, Birmingham, UK
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, UK
| | - Paul Moss
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Gergely Toldi
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
- Department of Neonatology, Birmingham Women's and Children's NHS FT, Birmingham, UK
| |
Collapse
|
29
|
Houston S. Siamon Gordon: A half-century fascination with macrophages. J Exp Med 2021; 217:151889. [PMID: 32579151 PMCID: PMC7336303 DOI: 10.1084/jem.20191069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Siamon Gordon is a Glaxo Wellcome Professor Emeritus of Cellular Pathology at the University of Oxford and a fellow of the Royal Society. Throughout his career, Siamon has focused on macrophages, and his work led to the identification of the pan-macrophage marker F4/80 and the description of a role for Dectin-1 in the innate recognition of β-glucans. I caught up with Siamon to discuss his career path and his thoughts on macrophages.
Collapse
|
30
|
Maternal schistosomiasis impairs offspring Interleukin-4 production and B cell expansion. PLoS Pathog 2021; 17:e1009260. [PMID: 33524040 PMCID: PMC7877777 DOI: 10.1371/journal.ppat.1009260] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 02/11/2021] [Accepted: 12/28/2020] [Indexed: 12/13/2022] Open
Abstract
Epidemiological studies have identified a correlation between maternal helminth infections and reduced immunity to some early childhood vaccinations, but the cellular basis for this is poorly understood. Here, we investigated the effects of maternal Schistosoma mansoni infection on steady-state offspring immunity, as well as immunity induced by a commercial tetanus/diphtheria vaccine using a dual IL-4 reporter mouse model of maternal schistosomiasis. We demonstrate that offspring born to S. mansoni infected mothers have reduced circulating plasma cells and peripheral lymph node follicular dendritic cells at steady state. These reductions correlate with reduced production of IL-4 by iNKT cells, the cellular source of IL-4 in the peripheral lymph node during early life. These defects in follicular dendritic cells and IL-4 production were maintained long-term with reduced secretion of IL-4 in the germinal center and reduced generation of TFH, memory B, and memory T cells in response to immunization with tetanus/diphtheria. Using single-cell RNASeq following tetanus/diphtheria immunization of offspring, we identified a defect in cell-cycle and cell-proliferation pathways in addition to a reduction in Ebf-1, a key B-cell transcription factor, in the majority of follicular B cells. These reductions are dependent on the presence of egg antigens in the mother, as offspring born to single-sex infected mothers do not have these transcriptional defects. These data indicate that maternal schistosomiasis leads to long-term defects in antigen-induced cellular immunity, and for the first time provide key mechanistic insight into the factors regulating reduced immunity in offspring born to S. mansoni infected mothers. Maternal helminth infections are a global public health concern and correlate with altered infant immune responses to some childhood immunizations, but a mechanistic understanding of how maternal helminth infection alters the cellular immune responses of offspring is lacking. Here we establish a model of maternal Schistosoma mansoni infection in dual IL-4 reporter mice. We find that offspring born to mothers infected with S. mansoni have impaired production of IL-4 during homeostasis, and following immunization with a Tetanus-Diphtheria vaccine. We identified that iNKT cells are the dominant source of IL-4 during early life homeostasis, and that diminished IL-4 production was associated with both reduced B cell and follicular dendritic cell responses. These defects were maintained long-term, affecting memory B and T cell responses. Single-cell RNASeq analysis of immunized offspring identified egg antigen-dependent reductions in B-cell cell cycle and proliferation-related genes. These data reveal that maternal infection leads to long-lasting defects in the cellular responses to heterologous antigens and provide vital insight into the influence of maternal infection on offspring immunity.
Collapse
|
31
|
Chetty A, Omondi MA, Butters C, Smith KA, Katawa G, Ritter M, Layland L, Horsnell W. Impact of Helminth Infections on Female Reproductive Health and Associated Diseases. Front Immunol 2020; 11:577516. [PMID: 33329545 PMCID: PMC7719634 DOI: 10.3389/fimmu.2020.577516] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 10/27/2020] [Indexed: 12/25/2022] Open
Abstract
A growing body of knowledge exists on the influence of helminth infections on allergies and unrelated infections in the lung and gastrointestinal (GI) mucosa. However, the bystander effects of helminth infections on the female genital mucosa and reproductive health is understudied but important considering the high prevalence of helminth exposure and sexually transmitted infections in low- and middle-income countries (LMICs). In this review, we explore current knowledge about the direct and systemic effects of helminth infections on unrelated diseases. We summarize host disease-controlling immunity of important sexually transmitted infections and introduce the limited knowledge of how helminths infections directly cause pathology to female reproductive tract (FRT), alter susceptibility to sexually transmitted infections and reproduction. We also review work by others on type 2 immunity in the FRT and hypothesize how these insights may guide future work to help understand how helminths alter FRT health.
Collapse
Affiliation(s)
- Alisha Chetty
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town, South Africa
| | - Millicent A Omondi
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town, South Africa
| | - Claire Butters
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town, South Africa
| | - Katherine Ann Smith
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town, South Africa.,School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Gnatoulma Katawa
- Ecole Supérieure des Techniques Biologiques et Alimentaires, Université de Lomé, Lomé, Togo
| | - Manuel Ritter
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), Bonn, Germany
| | - Laura Layland
- Institute for Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital Bonn (UKB), Bonn, Germany
| | - William Horsnell
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town, South Africa.,Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
| |
Collapse
|
32
|
Nyangahu DD, Darby M, Havyarimana E, Brown BP, Horsnell W, Jaspan HB. Preconception helminth infection alters offspring microbiota and immune subsets in a mouse model. Parasite Immunol 2020; 42:e12721. [PMID: 32277499 PMCID: PMC7423732 DOI: 10.1111/pim.12721] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 03/12/2020] [Accepted: 03/31/2020] [Indexed: 12/17/2022]
Abstract
Both maternal microbiota and helminth infection may alter offspring immunity but the relationship between these is underexplored. We hypothesized that maternal helminth exposure prior to pregnancy has lasting consequences on offspring intestinal microbiota and consequent immunity. Female BALB/c adult mice were infected with 500L3 Nippostrongylus brasiliensis (N brasiliensis). Infection was cleared by ivermectin treatment, and mice were mated 3 weeks post-infection (NbM). Control mice were not infected but were exposed to ivermectin (NvM). We analysed maternal gut microbiota during pregnancy, breastmilk microbiota and offspring faecal microbiota and immunity 2 weeks after delivery. During pregnancy, NbM (Mothers previously infected with Nippostrongylus brasiliensis) displayed significantly altered stool bacterial communities (R2 = .242; P = .001), with increased abundance of Enterococcaceae versus NvM (Naive mothers). Similarly, we observed a profound impact on breastmilk microbiota in NbM vs NvM. Moreover, NbM pups showed significantly altered gut microbial communities at 14 days of age versus those born to NvM with increased relative abundance of Coriobacteriaceae and Micrococcaceae. These changes were associated with alterations in pup immunity including increased frequencies and numbers of activated CD4 T cells (CD4 + CD44hi) in NbM offspring spleens. Taken together, we show that preconception helminth infections impact offspring immunity possibly through alteration of maternal and offspring microbiota.
Collapse
Affiliation(s)
- Donald D Nyangahu
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, RSA
- Departments of Pediatrics and Global Health, University of Washington, Seattle, WA
| | - Matthew Darby
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, RSA
| | - Enock Havyarimana
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, RSA
| | - Bryan P Brown
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle WA, USA
| | - William Horsnell
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, RSA
- Institute of Microbiology and Infection, University of Birmingham, B15 2TT Birmingham, UK
| | - Heather B. Jaspan
- Division of Immunology, Department of Pathology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, RSA
- Departments of Pediatrics and Global Health, University of Washington, Seattle, WA
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle WA, USA
| |
Collapse
|
33
|
Sakwinska O, Bosco N. Host Microbe Interactions in the Lactating Mammary Gland. Front Microbiol 2019; 10:1863. [PMID: 31456777 PMCID: PMC6701204 DOI: 10.3389/fmicb.2019.01863] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 07/29/2019] [Indexed: 12/13/2022] Open
Abstract
The bacteria present in human milk constitute the human milk microbiome (hMM). Both the older culture-based work and the more recent studies using molecular detection of bacterial DNA have reached similar conclusions: the hMM mostly consists of commensal staphylococci such as Staphylococcus epidermidis, and streptococci. The prevalence of other bacterial groups such lactobacilli varies widely, while the abundance and prevalence of bifidobacteria is generally low. Recently, the hMM became accepted as a part of a physiologically normal state with suggested potential health benefits. Most research on the hMM has focused on its composition and potential effect on the breastfed infant. A major role as a microbiome inoculum for the infant gut has been proposed, but remains to be clearly demonstrated. Herein, we also discuss the emerging connection between the hMM and mammary gland physiology and lactation. Similarities between the mammary gland and mucosal interfaces are considerable, and in particular mucosal-like immune attributes of mammary gland. The potential role of hMM-host interactions in the mammary gland in maternal health is explored with a primary focus on lactational mastitis.
Collapse
Affiliation(s)
- Olga Sakwinska
- Nestlé Research, Nestlé Institute of Health Sciences, Lausanne, Switzerland
| | - Nabil Bosco
- Nestlé Research, Nestlé Institute of Health Sciences, Lausanne, Switzerland
- Nestlé Research Singapore Hub, Singapore, Singapore
| |
Collapse
|