1
|
Sánchez MB, Michel Lara MC, Neira FJ, Rodríguez-Camejo C, Ríos JM, Viruel LB, Moreno-Sosa MT, Pietrobon EO, Soaje M, Jahn GA, Hernández A, Valdez SR, Mackern-Oberti JP. Hyperthyroidism keeps immunoglobulin levels but reduces milk fat and CD11b/c + cells on early lactation. Mol Cell Endocrinol 2024; 594:112370. [PMID: 39276963 DOI: 10.1016/j.mce.2024.112370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/17/2024]
Abstract
Thyroid hormones influence mammary gland differentiation and lactation by binding to thyroid hormone receptors. Hyperthyroidism disrupts pregnancy and lactation, affecting offspring growth and milk production. Despite maternal milk is a vital source of bioactive compounds and nutrients for newborns, it is unclear whether hyperthyroidism alters its composition, mainly immune factors. Therefore, our work aimed to evaluate the influence of hyperthyroidism on milk quality and immunological parameters during early lactation. Twelve-week-old female Wistar rats received daily injections of 0,25 mg/kg T4 (HyperT, n = 20) or vehicle (control, n = 19) starting 8 days before mating and continuing throughout pregnancy. Rats were euthanized on day 2 of lactation for analyzing the impact of hyperthyroidism on mammary gland, serum and milk samples. HyperT pups exhibited reduced weight, length and head circumference with altered serum hormones, glucose and albumin levels. HyperT mammary gland analysis revealed structural changes, including decreased alveolar area, adipose tissue, increased connective tissue and reduced epithelial elongation, accompanied by decreased TRβ1 RNA expression. HyperT milk displayed lower caloric value and fat concentration. HyperT animals exhibited altered milk immune cell counts, displaying increased numbers of CD45+ and CD3+ cells and decreased CD11b/c+ cells without changes on milk and serum IgA, IgG and IgG2a levels. In summary, we have demonstrated that hyperthyroidism affects mammary gland morphology, disrupts pup development and alters biochemical and immunological parameters. Our findings highlight the impact of maternal hyperthyroidism on offspring early development and milk immune composition, underscoring the importance of thyroid function in maternal and neonatal immune health.
Collapse
Affiliation(s)
- María Belén Sánchez
- Instituto de Medicina y Biología Experimental de Cuyo CONICET, Universidad Nacional de Cuyo, Mendoza, Argentina; Facultad de Ciencias Veterinarias y Ambientales, Universidad Juan Agustín Maza, Mendoza, Argentina
| | - María Cecilia Michel Lara
- Instituto de Medicina y Biología Experimental de Cuyo CONICET, Universidad Nacional de Cuyo, Mendoza, Argentina; Facultad de Farmacia y Bioquímica, Universidad Juan Agustín Maza, Mendoza, Argentina
| | - Flavia Judith Neira
- Instituto de Medicina y Biología Experimental de Cuyo CONICET, Universidad Nacional de Cuyo, Mendoza, Argentina; Facultad de Kinesiología y Fisioterapia, Universidad Juan Agustín Maza, Mendoza, Argentina
| | - Claudio Rodríguez-Camejo
- Laboratorio de Inmunología, Facultad de Ciencias/Facultad de Química, Universidad de la República, Instituto de Higiene, Montevideo, Uruguay
| | - Juan Manuel Ríos
- Instituto de Medicina y Biología Experimental de Cuyo CONICET, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Luciana Belén Viruel
- Instituto de Medicina y Biología Experimental de Cuyo CONICET, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - María Tamara Moreno-Sosa
- Instituto de Histología y Embriología de Mendoza, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Elisa Olivia Pietrobon
- Instituto de Medicina y Biología Experimental de Cuyo CONICET, Universidad Nacional de Cuyo, Mendoza, Argentina; Cátedra de Histología y Embriología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Marta Soaje
- Instituto de Medicina y Biología Experimental de Cuyo CONICET, Universidad Nacional de Cuyo, Mendoza, Argentina; Instituto de Fisiología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Graciela Alma Jahn
- Instituto de Medicina y Biología Experimental de Cuyo CONICET, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Ana Hernández
- Laboratorio de Inmunología, Facultad de Ciencias/Facultad de Química, Universidad de la República, Instituto de Higiene, Montevideo, Uruguay
| | - Susana Ruth Valdez
- Instituto de Medicina y Biología Experimental de Cuyo CONICET, Universidad Nacional de Cuyo, Mendoza, Argentina; Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Juan Pablo Mackern-Oberti
- Instituto de Medicina y Biología Experimental de Cuyo CONICET, Universidad Nacional de Cuyo, Mendoza, Argentina; Instituto de Fisiología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina.
| |
Collapse
|
2
|
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
|
3
|
Wander K, Fujita M, Mattison S, Gauck M, Duris M, Kiwelu I, Mmbaga BT. Maternal and infant predictors of proinflammatory milk immune activity in Kilimanjaro, Tanzania. Am J Hum Biol 2024; 36:e24061. [PMID: 38429916 DOI: 10.1002/ajhb.24061] [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: 08/29/2022] [Revised: 11/13/2023] [Accepted: 02/24/2024] [Indexed: 03/03/2024] Open
Abstract
OBJECTIVES The immune system of milk (ISOM) creates a mother-infant immune axis that plays an important role in protecting infants against infectious disease (ID). Tradeoffs in the immune system suggest the potential for both protection and harm, so we conceive of two dimensions via which the ISOM impacts infants: promotion of protective activity and control of activity directed at benign targets. High variability in ISOM activity across mother-infant dyads suggests investment the ISOM may have evolved to be sensitive to maternal and/or infant characteristics. We assessed predictors of appropriate and misdirected proinflammatory ISOM activity in an environment of high ID risk, testing predictions drawn from life history theory and other evolutionary perspectives. METHODS We characterized milk in vitro interleukin-6 (IL-6) responses to Salmonella enterica (a target of protective immune activity; N = 96) and Escherichia coli (a benign target; N = 85) among mother-infant dyads in rural Kilimanjaro, Tanzania. We used ordered logistic regression and mixture models to evaluate maternal and infant characteristics as predictors of IL-6 responses. RESULTS In all models, IL-6 responses to S. enterica increased with maternal age and decreased with gravidity. In mixture models, IL-6 responses to E. coli declined with maternal age and increased with gravidity. No other considered variables were consistently associated with IL-6 responses. CONCLUSIONS The ISOM's capacities for appropriate proinflammatory activity and control of misdirected proinflammatory activity increases with maternal age and decreases with gravidity. These findings are consistent with the hypothesis that the mother-infant immune axis has evolved to respond to maternal life history characteristics.
Collapse
Affiliation(s)
- Katherine Wander
- Department of Anthropology, Binghamton University (SUNY), Binghamton, New York, USA
| | - Masako Fujita
- Department of Anthropology, Michigan State University, East Lansing, Michigan, USA
| | - Siobhán Mattison
- Department of Anthropology, University of New Mexico, Albuquerque, New Mexico, USA
- National Science Foundation, Alexandria, Virginia, USA
| | - Megan Gauck
- Department of Anthropology, Binghamton University (SUNY), Binghamton, New York, USA
| | - Margaret Duris
- Department of Anthropology, Binghamton University (SUNY), Binghamton, New York, USA
| | - Ireen Kiwelu
- Kilimanjaro Clinical Research Institute, Kilimanjaro, Tanzania
- Kilimanjaro Christian Medical Centre, Kilimanjaro, Tanzania
- Kilimanjaro Christian Medical University College, Kilimanjaro, Tanzania
| | - Blandina T Mmbaga
- Kilimanjaro Clinical Research Institute, Kilimanjaro, Tanzania
- Kilimanjaro Christian Medical Centre, Kilimanjaro, Tanzania
- Kilimanjaro Christian Medical University College, Kilimanjaro, Tanzania
| |
Collapse
|
4
|
Anyim R, Li S, Armstrong D, Spathis R, Wander K. Associations between milk cortisol and activity of the immune system of milk. Am J Hum Biol 2023; 35:e23960. [PMID: 37485918 DOI: 10.1002/ajhb.23960] [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: 01/11/2023] [Revised: 06/02/2023] [Accepted: 06/30/2023] [Indexed: 07/25/2023] Open
Abstract
OBJECTIVES Both the immune system of human milk and milk cortisol have complex short- and long-term effects on child health and development. As understanding continues to grow of the independent effects of each of these components of milk, it is also important to investigate their intersection, including how milk cortisol affects the immune system of milk. We began this important endeavor through secondary analyses of archived milk specimens. METHODS Participants were 31 lactating mothers from upstate New York. We estimated milk cortisol concentrations via enzyme immunoassay. We assessed milk proinflammatory cytokine (interleukin-6, IL-6) responses to pathogenic (Salmonella) and commensal (Escherichia, Lactobacillus, Bifidobacterium) bacteria via in vitro stimulation. We estimated ordered logistic regression models to assess associations between milk cortisol and IL-6 responses to bacteria. RESULTS Milk cortisol ranged from 0.098 to 1.007 μg/dL. Milk cortisol was positively associated with IL-6 responses to S. enterica (B: 4.035; 95% CI: 0.674, 7.395) and B. breve (B: 3.675; 95% CI: 0.426, 6.924); this association persisted after controlling for child age. Results were less clear for associations between milk cortisol and IL-6 responses to L. acidophilus (B: 2.318; 95% CI: -1.224, 5.859) and E. coli (B: 2.366; 95% CI: -0.960, 5.692). CONCLUSIONS Complex interactions between cortisol and the immune system extend to milk. Milk cortisol was positively associated with proinflammatory responses to some bacteria in vitro. This may suggest that milk cortisol is causally upstream of protective immune activity.
Collapse
Affiliation(s)
- Rachael Anyim
- Department of Anthropology, Binghamton University, Binghamton, New York, USA
| | - Shanita Li
- Department of Anthropology, Binghamton University, Binghamton, New York, USA
| | - Daniel Armstrong
- Department of Anthropology, Binghamton University, Binghamton, New York, USA
| | - Rita Spathis
- Department of Anthropology, Binghamton University, Binghamton, New York, USA
| | - Katherine Wander
- Department of Anthropology, Binghamton University, Binghamton, New York, USA
| |
Collapse
|
5
|
Wander K, Fujita M, Mattison S, Gauck M, Duris M, Kiwelu I, Mmbaga BT. Does the immune system of milk increase activity for infants experiencing infectious disease episodes in Kilimanjaro, Tanzania? Am J Hum Biol 2023; 35:e23897. [PMID: 36951242 DOI: 10.1002/ajhb.23897] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 02/27/2023] [Accepted: 03/08/2023] [Indexed: 03/24/2023] Open
Abstract
INTRODUCTION Multiple studies have reported that milk immune content increases for infants experiencing infectious disease (ID) episodes, suggesting that the immune system of milk (ISOM) offers enhanced protection when needed to combat ID. METHODS To test the hypothesis that ISOM content and/or activity increases during an infant's ID episode, we characterized milk secretory immunoglobulin A (sIgA; a major ISOM constituent) and in vitro interleukin-6 (IL-6) responses to Salmonella enterica and Escherichia coli, as system-level biomarkers of ISOM activity, in a prospective study among 96 mother-infant dyads in Kilimanjaro, Tanzania. RESULTS After control for covariates, no milk immune variables (sIgA, Coef: 0.03; 95% CI -0.25, 0.32; in vitro IL-6 response to S. enterica, Coef: 0.23; 95% CI: -0.67, 1.13; IL-6 response to E. coli, Coef: -0.11; 95% CI: -0.98, 0.77) were associated with prevalent ID (diagnosed at the initial participation visit). Among infants experiencing an incident ID (diagnosed subsequent to the initial participation), milk immune content and responses were not substantially higher or lower than the initial visit (sIgA, N: 61; p: 0.788; IL-6 response to S. enterica, N: 56; p: 0.896; IL-6 response to E. coli, N: 36; p: 0.683); this was unchanged by exclusion of infants with ID at the time of initial participation. CONCLUSION These findings are not consistent with the hypothesis that milk delivers enhanced immune protection when infants experience ID. In environments with a high burden of ID, dynamism may be less valuable to maternal reproductive success than stability in the ISOM.
Collapse
Affiliation(s)
- Katherine Wander
- Department of Anthropology, Binghamton University (SUNY), Binghamton, New York, USA
| | - Masako Fujita
- Department of Anthropology, Michigan State University, East Lansing, Michigan, USA
| | - Siobhán Mattison
- Department of Anthropology, University of New Mexico, Albuquerque, New Mexico, USA
| | - Megan Gauck
- Department of Anthropology, Binghamton University (SUNY), Binghamton, New York, USA
| | - Margaret Duris
- Department of Anthropology, Binghamton University (SUNY), Binghamton, New York, USA
| | - Ireen Kiwelu
- Kilimanjaro Clinical Research Institute, Moshi, Tanzania
- Kilimanjaro Christian Medical Centre, Moshi, Tanzania
- Kilimanjaro Christian Medical University College, Moshi, Tanzania
| | - Blandina T Mmbaga
- Kilimanjaro Clinical Research Institute, Moshi, Tanzania
- Kilimanjaro Christian Medical Centre, Moshi, Tanzania
- Kilimanjaro Christian Medical University College, Moshi, Tanzania
| |
Collapse
|
6
|
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: 7] [Impact Index Per Article: 7.0] [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
|
7
|
Chandler TL, Newman A, Cha JE, Sipka AS, Mann S. Leukocytes, microRNA, and complement activity in raw, heat-treated, and frozen colostrum and their dynamics as colostrum transitions to mature milk in dairy cows. J Dairy Sci 2023:S0022-0302(23)00220-5. [PMID: 37164855 DOI: 10.3168/jds.2022-22876] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 01/06/2023] [Indexed: 05/12/2023]
Abstract
The objectives of this study were to evaluate the abundance and viability of leukocytes, the abundance of microRNA, and the activity of the complement pathway in (1) colostrum following heat-treatment or freezing, and (2) colostrum, transition milk, and mature milk. In experiment 1, composite colostrum samples were harvested from individual cows (n = 14) on a commercial dairy farm in NY and split into 3 aliquots using single-use colostrum bags. One aliquot was immediately cooled on ice following harvest (RAW) and stored at 4°C overnight, one was heat-treated for 60 min at 60°C (HT) before being cooled on ice and stored at 4°C overnight, and one was frozen at -20°C overnight (FR). The following morning, all samples were warmed to 40°C before further processing. In experiment 2, cows were sampled in a longitudinal study where composite samples were collected from colostrum (first milking, n = 23), transition milk (3 to 4 d postpartum, n = 13), and mature milk (6 to 7 d postpartum, n = 13). In both experiments colostrum was harvested from the first milking within 8 h of calving and samples were processed within 14 h of collection. Colostral leukocytes were isolated before viability was determined by trypan blue exclusion and manual differential cell counts were performed. Extracellular vesicles were isolated from whey by ultracentrifugation to isolate and quantify microRNA. Activity of the alternative complement pathway was determined in casein-depleted whey by semi-solid phase hemolysis assay. Somatic cell counts were determined for all raw samples. Macrophages and neutrophils made up the greatest proportion of leukocytes in colostrum followed by lymphocytes. Lymphocyte proportion increased as colostrum transitioned to mature milk, but overall somatic cell numbers declined concurrently. Viable cells were not isolated from HT or FR samples. Abundance of microRNA isolated from transition and mature milk was decreased compared with colostrum, did not differ between HT and RAW, but was increased in FR compared with RAW. Alternative complement pathway activity was decreased in HT, but not FR compared with RAW, and was not measurable in transition or mature milk. Postharvest heat-treatment and freezing of colostrum eliminated viable colostral leukocytes and affected microRNA abundance and complement activity. Leukocyte proportions, microRNA abundance, and complement activity changed as colostrum transitioned to mature milk. Although there were clear changes in the colostral components under study in relation to treatment and transition to mature milk, the biological significance of the described treatment effects and temporal changes were not investigated here.
Collapse
Affiliation(s)
- T L Chandler
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853
| | - A Newman
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853
| | - J E Cha
- Division of Nutritional Sciences, College of Human Ecology, Cornell University, Ithaca, NY 14853
| | - A S Sipka
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853
| | - S Mann
- Department of Population Medicine and Diagnostic Sciences, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853.
| |
Collapse
|
8
|
Cowardin CA, Syed S, Iqbal N, Jamil Z, Sadiq K, Iqbal J, Ali SA, Moore SR. Environmental enteric dysfunction: gut and microbiota adaptation in pregnancy and infancy. Nat Rev Gastroenterol Hepatol 2023; 20:223-237. [PMID: 36526906 PMCID: PMC10065936 DOI: 10.1038/s41575-022-00714-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/16/2022] [Indexed: 03/31/2023]
Abstract
Environmental enteric dysfunction (EED) is a subclinical syndrome of intestinal inflammation, malabsorption and barrier disruption that is highly prevalent in low- and middle-income countries in which poverty, food insecurity and frequent exposure to enteric pathogens impair growth, immunity and neurodevelopment in children. In this Review, we discuss advances in our understanding of EED, intestinal adaptation and the gut microbiome over the 'first 1,000 days' of life, spanning pregnancy and early childhood. Data on maternal EED are emerging, and they mirror earlier findings of increased risks for preterm birth and fetal growth restriction in mothers with either active inflammatory bowel disease or coeliac disease. The intense metabolic demands of pregnancy and lactation drive gut adaptation, including dramatic changes in the composition, function and mother-to-child transmission of the gut microbiota. We urgently need to elucidate the mechanisms by which EED undermines these critical processes so that we can improve global strategies to prevent and reverse intergenerational cycles of undernutrition.
Collapse
Affiliation(s)
- Carrie A Cowardin
- Division of Paediatric Gastroenterology, Hepatology and Nutrition, Department of Paediatrics, Child Health Research Center, University of Virginia, Charlottesville, VA, USA
| | - Sana Syed
- Division of Paediatric Gastroenterology, Hepatology and Nutrition, Department of Paediatrics, Child Health Research Center, University of Virginia, Charlottesville, VA, USA
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Najeeha Iqbal
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Zehra Jamil
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Kamran Sadiq
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Junaid Iqbal
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Syed Asad Ali
- Department of Paediatrics and Child Health, Aga Khan University, Karachi, Pakistan
| | - Sean R Moore
- Division of Paediatric Gastroenterology, Hepatology and Nutrition, Department of Paediatrics, Child Health Research Center, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
9
|
Benslama Y, Dennouni-Medjati N, Dali-Sahi M, Meziane FZ, Harek Y. Childhood type 1 diabetes mellitus and risk factor of interactions between dietary cow's milk intake and HLA-DR3/DR4 genotype. J Biomol Struct Dyn 2022; 40:10931-10939. [PMID: 34282715 DOI: 10.1080/07391102.2021.1953599] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Short-term breastfeeding and early exposure to dairy products into infant diets, may be critical factors for development of type 1 diabetes. In this study, we investigate whether cow's milk proteins are risk factors for type 1 diabetes in genetically susceptible individuals (HLA DR3/DR4) by using statistical analysis and in silico approach. In order to verify the potential risk of the early introduction of cow's milk, we conducted this study to validate the veracity of this hypothesis in our population. We included 121 subjects, 55 type 1 diabetics and 74 controls from the region of Tlemcen (Algeria). Thus, the in silico approach was performed to determine the molecular mimicry region between Bovine serum albumin and beta-lactoglobulin with self-Islet antigen 2 and glutamate decarboxylase 65 by determining their sequences and their 3D structures. The risk factors associated with type 1 diabetes in a genetically predisposed individual (HLA DR3/DR4) retained by the logistic model are: type 1 and type 2 diabetes inheritance, the early introduction of cow's milk before 6 months and breastfeeding less than 9 months. Besides, the epitopes of cow's milk proteins have the capacity to bind to predisposing HLA class II molecules (HLA DR3/DR4) and induce an immune reaction by the secretion of Interleukin 4 (Th2) and Interferon (Th1) which lead to the destruction of pancreatic beta cells. The early introduction of cow's milk proteins in susceptible individuals is considered as risk factors for the pathogenesis of T1DM. The in silico approach confirm that BSA and BLG share sequence and structure homology with IA2 and GAD65.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Yasmine Benslama
- Laboratory of Analytical Chemistry and Electrochemistry, Abou Bekr Belkaid University of Tlemcen, Tlemcen, Algeria
| | - Nouria Dennouni-Medjati
- Laboratory of Analytical Chemistry and Electrochemistry, Abou Bekr Belkaid University of Tlemcen, Tlemcen, Algeria
| | - Majda Dali-Sahi
- Laboratory of Analytical Chemistry and Electrochemistry, Abou Bekr Belkaid University of Tlemcen, Tlemcen, Algeria
| | - Fatima Zahra Meziane
- Laboratory of Analytical Chemistry and Electrochemistry, Abou Bekr Belkaid University of Tlemcen, Tlemcen, Algeria
| | - Yahia Harek
- Laboratory of Analytical Chemistry and Electrochemistry, Abou Bekr Belkaid University of Tlemcen, Tlemcen, Algeria
| |
Collapse
|
10
|
Breastfeeding by chikungunya virus-infected dams confers resistance to challenge in the offspring. Transl Res 2022; 255:109-118. [PMID: 36526155 DOI: 10.1016/j.trsl.2022.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022]
Abstract
Vertical transmission of Chikungunya virus (CHIKV) has been reported in humans, but the transmission routes have not been completely understood, and experimental animal models are needed to enable detailed investigation of the transmission and pathogenesis of congenital infections. The intertwining of immune response and virus components at the gestation/breastfeeding interfaces between mother and fetus/newborn may have effects during the offspring development. An experimental model of CHIKV was established by infecting pregnant BALB/c female mice that enabled confirmation that dams inoculated up to the 10th gestational day transmit CHIKV transplacentally to approximately 8.4% of the fetuses, resulting in severe teratogenic effects. CHIKV neutralizing antibodies were detected in sera from adult mice born to healthy females and breastfed by CHIKV-infected dams, while no neutralization was detected in sera from animals born to CHIKV-infected dams. Moreover, adult mice born to healthy dams and cross-fostered for breastfeeding by CHIKV-infected dams were resistant to challenge with CHIKV on the 90th day after birth. The animals also had reduced viral loads in brain and spleen as compared to controls. There was expression of fluorescent CHIKV non-structural protein, and detection of viral RNA by RT-PCR in breast tissue from infected dams. CHIKV RNA and proteins were also detected in breast milk retrieved from the stomachs of recently fed newborns. The experimental results were also complemented by the finding of CHIKV RNA in 6% of colostrum samples from healthy lactating women in a CHIKV-endemic area. Breastfeeding induces immune protection to challenge with CHIKV in mice.
Collapse
|
11
|
Castellan FS, Irie N. Postnatal depletion of maternal cells biases T lymphocytes and natural killer cells' profiles toward early activation in the spleen. Biol Open 2022; 11:bio059334. [PMID: 36349799 PMCID: PMC9672855 DOI: 10.1242/bio.059334] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 10/11/2022] [Indexed: 08/27/2023] Open
Abstract
The maternal cells transferred into the fetus during gestation persist long after birth in the progeny. These maternal cells have been hypothesized to promote the maturation of the fetal immune system in utero but there are still significant gaps in our knowledge of their potential roles after birth. To provide insights into these maternal cells' postnatal functional roles, we set up a transgenic mouse model to specifically eliminate maternal cells in the neonates by diphtheria toxin injection and confirmed significant depletion in the spleens. We then performed immunophenotyping of the spleens of two-week-old pups by mass cytometry to pinpoint the immune profile differences driven by the depletion of maternal cells in early postnatal life. We observed a heightened expression of markers related to activation and maturation in some natural killer and T cell populations. We hypothesize these results to indicate a potential postnatal regulation of lymphocytic responses by maternal cells. Together, our findings highlight an immunological influence of maternal microchimeric cells postnatally, possibly protecting against adverse hypersensitivity reactions of the neonate at a crucial time of new encounters with self and environmental antigens.
Collapse
Affiliation(s)
- Flore S. Castellan
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Naoki Irie
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| |
Collapse
|
12
|
Lengi AJ, Stewart JW, Makris M, Rhoads ML, Corl BA. Heat Stress Increases Mammary Epithelial Cells and Reduces Viable Immune Cells in Milk of Dairy Cows. Animals (Basel) 2022; 12:2810. [PMID: 36290196 PMCID: PMC9597744 DOI: 10.3390/ani12202810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/10/2022] [Accepted: 10/14/2022] [Indexed: 11/17/2022] Open
Abstract
Somatic cells normally found in milk are generally either immune cells such as lymphocytes, monocytes and granulocytes, or mammary epithelial cells. The number and composition of somatic cells in milk can be influenced by a variety of factors, including infection and temperature-humidity index. The objective of this study was to determine the specific effects of heat stress on the cellular composition of the somatic cell population in milk. We used flow cytometry to ascertain the concentration and viability of mammary epithelial cells, T cells, monocyte/macrophage, and granulocytes in milk from cows maintained under heat stressed conditions compared to thermoneutral conditions. We found a significant 10% increase in the natural log concentration of epithelial cells in the milk of heat stressed cows compared to thermoneutral cows (9.3 vs. 8.4 ln(cells/mL, p = 0.02)). We also found a 12% decrease in the log concentration of live CD45+ cells (p = 0.04), and a 17% decrease in the log concentration of live CD45+ granulocytes (p = 0.04). No changes were found in CD3+CD45+ cells or CD14+CD45+ cells, however, we noted an unusual population of CD14+CD45- cells that showed significant increases of 10% (p = 0.03) and 12% (p = 0.01) in the log concentration of total and dead cells, respectively, under heat stressed conditions. These results suggest that heat stress influences the relative populations and viability of some somatic cells populations in milk. Increased losses of secretory epithelial cells into milk could have implications for milk production, and fewer viable immune cells could negatively impact the immunocompetence of dairy cows under heat stress.
Collapse
Affiliation(s)
- Andrea J. Lengi
- Department of Dairy Science, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - Jacob W. Stewart
- Department of Animal and Poultry Sciences, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - Melissa Makris
- Flow Cytometry Laboratory, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA 24061, USA
| | - Michelle L. Rhoads
- Department of Animal and Poultry Sciences, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| | - Benjamin A. Corl
- Department of Dairy Science, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
13
|
Doerfler R, Melamed JR, Whitehead KA. The Effect of Infant Gastric Digestion on Human Maternal Milk Cells. Mol Nutr Food Res 2022; 66:e2200090. [PMID: 35984112 PMCID: PMC9532377 DOI: 10.1002/mnfr.202200090] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 06/24/2022] [Indexed: 12/30/2022]
Abstract
SCOPE Human breast milk contains a variety of cell types that have potential roles in infant immunity and development. One challenge associates with defining the purpose(s) of milk cells in the infant is a poor understanding of the effect of digestion on cell fate. METHODS AND RESULTS This study first demonstrates that milk cell death occurs after gastric digestion in mice. Then flow cytometry and RT-PCR are used to understand the mechanism of human milk cell death and quantify live cell types before and after simulated gastric digestion. This study finds that digestion in simulated gastric fluid for 30 min reduces cell viability from 72% to 27%, with most cell death is caused by the acidic pH. The primary mechanism of cell death is caspase-mediated apoptosis. The non-cellular components of milk offer only mild protection against cell death from stomach acid. CONCLUSIONS Gastric digestion does not select for a specific resilient cell population to survive-most cell types die in equal proportions in the gastric environment. Taken together, these results provide a foundation with which to understand the fate of human breast milk cells in the infant's intestine and beyond.
Collapse
Affiliation(s)
- Rose Doerfler
- Department of Chemical EngineeringCarnegie Mellon UniversityPittsburghPA15213USA
| | - Jilian R. Melamed
- Department of Chemical EngineeringCarnegie Mellon UniversityPittsburghPA15213USA
| | - Kathryn A. Whitehead
- Department of Chemical EngineeringCarnegie Mellon UniversityPittsburghPA15213USA,Department of Biomedical EngineeringCarnegie Mellon UniversityPittsburghPA15213USA
| |
Collapse
|
14
|
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
|
15
|
Wander K, Fujita M, Mattison SM, Duris M, Gauck M, Hopt T, Lacy K, Foligno A, Ulloa R, Dodge C, Mowo F, Kiwelu I, Mmbaga BT. Tradeoffs in milk immunity affect infant infectious disease risk. Evol Med Public Health 2022; 10:295-304. [PMID: 35769951 PMCID: PMC9233416 DOI: 10.1093/emph/eoac020] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 05/10/2022] [Indexed: 11/22/2022] Open
Abstract
Background and objectives The human immune system has evolved to balance protection against infection with control of immune-mediated damage and tolerance of commensal microbes. Such tradeoffs between protection and harm almost certainly extend to the immune system of milk. Methodology Among breastfeeding mother-infant dyads in Kilimanjaro, Tanzania, we characterized in vitro proinflammatory milk immune responses to Salmonella enterica (an infectious agent) and Escherichia coli (a benign target) as the increase in interleukin-6 after 24 h of incubation with each bacterium. We characterized incident infectious diseases among infants through passive monitoring. We used Cox proportional hazards models to describe associations between milk immune activity and infant infectious disease. Results Among infants, risk for respiratory infections declined with increasing milk in vitro proinflammatory response to S. enterica (hazard ratio [HR]: 0.68; 95% confidence interval [CI]: 0.54, 0.86; P: 0.001), while risk for gastrointestinal infections increased with increasing milk in vitro proinflammatory response to E. coli (HR: 1.44; 95% CI: 1.05, 1.99; P: 0.022). Milk proinflammatory responses to S. enterica and E. coli were positively correlated (Spearman's rho: 0.60; P: 0.000). Conclusions and implications These findings demonstrate a tradeoff in milk immune activity: the benefits of appropriate proinflammatory activity come at the hazard of misdirected proinflammatory activity. This tradeoff is likely to affect infant health in complex ways, depending on prevailing infectious disease conditions. How mother-infant dyads optimize proinflammatory milk immune activity should be a central question in future ecological-evolutionary studies of the immune system of milk.
Collapse
Affiliation(s)
- Katherine Wander
- Department of Anthropology, Binghamton University (SUNY), Binghamton, NY, USA
| | - Masako Fujita
- Department of Anthropology, Michigan State University, East Lansing, MI, USA
| | - Siobhan M Mattison
- Department of Anthropology, University of New Mexico, Albuquerque, NM, USA
| | - Margaret Duris
- Department of Anthropology, Binghamton University (SUNY), Binghamton, NY, USA
| | - Megan Gauck
- Department of Anthropology, Binghamton University (SUNY), Binghamton, NY, USA
| | - Tessa Hopt
- Department of Anthropology, Binghamton University (SUNY), Binghamton, NY, USA
| | - Katherine Lacy
- Department of Anthropology, Binghamton University (SUNY), Binghamton, NY, USA
| | - Angela Foligno
- Department of Anthropology, Binghamton University (SUNY), Binghamton, NY, USA
| | - Rebecca Ulloa
- Department of Anthropology, Binghamton University (SUNY), Binghamton, NY, USA
| | - Connor Dodge
- Department of Anthropology, Binghamton University (SUNY), Binghamton, NY, USA
| | - Frida Mowo
- Kilimanjaro Christian Medical Centre, Moshi, Kilimanjaro, Tanzania
- Kilimanjaro Christian Medical University College, Moshi, Kilimanjaro, Tanzania
| | - Ireen Kiwelu
- Kilimanjaro Christian Medical Centre, Moshi, Kilimanjaro, Tanzania
- Kilimanjaro Christian Medical University College, Moshi, Kilimanjaro, Tanzania
- Kilimanjaro Clinical Research Institute, Moshi, Kilimanjaro, Tanzania
| | - Blandina T Mmbaga
- Kilimanjaro Christian Medical Centre, Moshi, Kilimanjaro, Tanzania
- Kilimanjaro Christian Medical University College, Moshi, Kilimanjaro, Tanzania
- Kilimanjaro Clinical Research Institute, Moshi, Kilimanjaro, Tanzania
- Duke Global Health Institute, Duke University, Durham, NC, USA
| |
Collapse
|
16
|
Gonçalves J, Juliano AM, Charepe N, Alenquer M, Athayde D, Ferreira F, Archer M, Amorim MJ, Serrano F, Soares H. Secretory IgA and T cells targeting SARS-CoV-2 spike protein are transferred to the breastmilk upon mRNA vaccination. Cell Rep Med 2021; 2:100468. [PMID: 34873588 PMCID: PMC8636305 DOI: 10.1016/j.xcrm.2021.100468] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 10/09/2021] [Accepted: 11/15/2021] [Indexed: 12/24/2022]
Abstract
In view of the scarcity of data to guide decision making, we evaluated how BNT162b2 and mRNA-1273 vaccines affect the immune response in lactating women and the protective profile of breastmilk. Compared with controls, lactating women had a higher frequency of circulating RBD memory B cells and higher anti-RBD antibody titers but similar neutralizing capacity. We show that upon vaccination, immune transfer to breastmilk occurs through a combination of anti-spike secretory IgA (SIgA) antibodies and spike-reactive T cells. Although we found that the concentration of anti-spike IgA in breastmilk might not be sufficient to directly neutralize SARS-CoV-2, our data suggest that cumulative transfer of IgA might provide the infant with effective neutralization capacity. Our findings put forward the possibility that breastmilk might convey both immediate (through anti-spike SIgA) and long-lived (via spike-reactive T cells) immune protection to the infant. Further studies are needed to address this possibility and to determine the functional profile of spike T cells.
Collapse
Affiliation(s)
- Juliana Gonçalves
- Human Immunobiology and Pathogenesis Group, CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisbon, Portugal
- iNOVA4Health, Lisbon, Portugal
| | - A. Margarida Juliano
- Human Immunobiology and Pathogenesis Group, CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisbon, Portugal
- iNOVA4Health, Lisbon, Portugal
| | - Nádia Charepe
- Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
- CHRC, CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisbon, Portugal
| | - Marta Alenquer
- Cell Biology of Viral Infection Lab, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Diogo Athayde
- Membrane Protein Crystallography Laboratory, Instituto de Tecnologia Química e Biológica, ITQB-NOVA, Oeiras, Portugal
| | - Filipe Ferreira
- Cell Biology of Viral Infection Lab, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Margarida Archer
- Membrane Protein Crystallography Laboratory, Instituto de Tecnologia Química e Biológica, ITQB-NOVA, Oeiras, Portugal
| | - Maria João Amorim
- Cell Biology of Viral Infection Lab, Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Fátima Serrano
- Centro Hospitalar Universitário Lisboa Central, Lisbon, Portugal
- CHRC, CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisbon, Portugal
| | - Helena Soares
- Human Immunobiology and Pathogenesis Group, CEDOC, NOVA Medical School | Faculdade de Ciências Médicas, NOVA University of Lisbon, Lisbon, Portugal
- iNOVA4Health, Lisbon, Portugal
| |
Collapse
|
17
|
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: 4] [Impact Index Per Article: 1.3] [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
|
18
|
Wander K, Fujita M, Spathis R, Li S, Armstrong D, Pechera J, Anyim R, Manganello M, Torres C, Herbin C, Cradin B, Margolin F. In Vitro Stimulation of Whole Milk Specimens: A Field-Friendly Method to Assess Milk Immune Activity. J Hum Lact 2021; 37:736-745. [PMID: 33788640 DOI: 10.1177/0890334421999628] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND The immune system of milk protects against infections and guides immune system development. A system-level understanding of milk immune activity is critical for research into infant infectious disease risk and lifelong health. RESEARCH AIM To describe a protocol to characterize immune activity in human milk via in vitro stimulation for use in population-based (rather than clinical) research. METHODS This study proceeded in two phases, each with a cross-sectional design. Human milk specimens were incubated for 24 hr at 37 °C in mammalian cell culture medium with stimuli (e.g., Salmonella enterica) in a CO2-enriched environment. Immune responses to stimuli were characterized as the change in cytokine: [stimulated]/[baseline]. Predictors of cytokine responses were evaluated with generalized linear models. RESULTS Patterns were detectable across mother-child dyads: Interleukin-6 responses to stimuli were generally positively associated with child age and with maternal autoimmune disease. CONCLUSIONS Our method allows characterization of pro-inflammatory milk immune activity in vitro in population-based (rather than clinical) research settings. In vitro activity has a system-level interpretation and is likely to be of broad utility in global health research in settings with high infectious disease risk, where understanding the immune system of milk is critical to understanding maternal and child health.
Collapse
Affiliation(s)
| | - Masako Fujita
- 3078 Michigan State University, East Lansing, MI, USA
| | - Rita Spathis
- 14787 Binghamton University (SUNY), Binghamton, NY, USA
| | - Shanita Li
- 14787 Binghamton University (SUNY), Binghamton, NY, USA
| | | | - Jane Pechera
- 14787 Binghamton University (SUNY), Binghamton, NY, USA
| | - Rachael Anyim
- 14787 Binghamton University (SUNY), Binghamton, NY, USA
| | | | - Cruz Torres
- 14787 Binghamton University (SUNY), Binghamton, NY, USA
| | | | - Brooke Cradin
- 14787 Binghamton University (SUNY), Binghamton, NY, USA
| | | |
Collapse
|
19
|
Dawod B, Marshall JS, Azad MB. Breastfeeding and the developmental origins of mucosal immunity: how human milk shapes the innate and adaptive mucosal immune systems. Curr Opin Gastroenterol 2021; 37:547-556. [PMID: 34634003 PMCID: PMC11451935 DOI: 10.1097/mog.0000000000000778] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW Breastfeeding provides passive immunity while the neonatal immune system matures, and may also protect against chronic immune-mediated conditions long after weaning. This review summarizes current knowledge and new discoveries about human milk and mucosal immunity. RECENT FINDINGS New data suggest that certain microbes in maternal milk may seed and shape the infant gut microbiota, which play a key role in regulating gut barrier integrity and training the developing immune system. Human milk oligosaccharides, best known for their prebiotic functions, have now been shown to directly modulate gene expression in mast and goblet cells in the gastrointestinal tract. Epidemiologic data show a reduced risk of peanut sensitization among infants breastfed by peanut-consuming mothers, suggesting a role for milk-borne food antigens in tolerance development. Cross-fostering experiments in mice suggest the soluble Toll-like receptor 2, found in human milk, may be critical in this process. Finally, interest in human milk antibodies surged during the pandemic with the identification of neutralizing severe acute respiratory syndrome coronavirus 2 antibodies in maternal milk following both natural infection and vaccination. SUMMARY Human milk provides critical immune protection and stimulation to breastfed infants. Understanding the underlying mechanisms could identify new therapeutic targets and strategies for disease prevention across the lifespan.
Collapse
Affiliation(s)
- Bassel Dawod
- Department of Pathology
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia
| | - Jean S. Marshall
- Department of Pathology
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia
| | - Meghan B. Azad
- Manitoba Interdisciplinary Lactation Centre (MILC), Children's Hospital Research Institute of Manitoba
- Department of Pediatrics and Child Health
- Department of Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
20
|
Myles IA, Datta SK. Frontline Science: Breast milk confers passive cellular immunity via CD8-dependent mechanisms. J Leukoc Biol 2021; 109:709-715. [PMID: 32881103 DOI: 10.1002/jlb.3hi0820-406rr] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 08/08/2020] [Accepted: 08/18/2020] [Indexed: 12/14/2022] Open
Abstract
Most modern research into the immune effects of breast milk has focused on the impacts of immunoglobulin or oligosaccharide content. However, immediately prior to parturition, the cell populations of breast milk become selectively enriched for CD8+ T cells of an effector memory subtype. Despite this observation that the cellular content of breast milk contains a distinct leukocyte population when compared to peripheral blood, the physiologic role of these CD8+ effector memory cells is unknown. Research encompassing animal models and humans has demonstrated that leukocytes are capable of transferring antigen-specific immunity even when lysed, dialyzed to enrich for fractions less than 10 kDa, and orally administered. Our previous work built upon these reports to elucidate several aspects of this dialyzable leukocyte extract (DLE) activity: only DLE from T effector memory CD8+ cells was capable of transferring antigen-specific immunity; the DLE activity was TCRβ dependent; dendritic cells (DCs) were the cellular target of DLE; and DLE enhanced immune activity in epithelial challenge models via induction of IL-6 from DCs. Herein, we reveal that breast milk dialysate activates similar cytokine and genetic pathways as DLE taken from peripheral blood and murine spleens through TCRβ- and CD8-dependent mechanisms. These findings suggest that the CD8+ memory T cells enriched in breast milk, even after potential lysis in the infant gut, may represent a mechanism for passive transfer of cellular immunity from mother to child.
Collapse
Affiliation(s)
- Ian A Myles
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Sandip K Datta
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
21
|
Ellis J. Passive transfer of colostral leukocytes: A benefit/risk analysis. THE CANADIAN VETERINARY JOURNAL = LA REVUE VETERINAIRE CANADIENNE 2021; 62:233-239. [PMID: 33692577 PMCID: PMC7877691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Passive immunity is a requirement for survival of conventionally reared domestic animals. Maternal antibody is recognized as the primary effector mechanism of protection of the neonate. There is interest, especially in bovine medicine, in the maternal leukocytes that are also transferred in colostrum. These leukocytes are thought to somehow aid in the development of the neonatal immune system and participate in cellular immune reactions in the neonate, although the means by which this is accomplished is unknown. The purpose of this article is to review the immunology of this maternal/neonatal interaction, and on that basis, evaluate the practical benefits and risks of assuring passive transfer of colostral leukocytes.
Collapse
Affiliation(s)
- John Ellis
- Department of Veterinary Microbiology, Western College of Veterinary Medicine, University of Saskatchewan, 52 Campus Drive, Saskatoon, Saskatchewan S7N 5B4
| |
Collapse
|
22
|
Laouar A. Maternal Leukocytes and Infant Immune Programming during Breastfeeding. Trends Immunol 2020; 41:225-239. [PMID: 32057705 DOI: 10.1016/j.it.2020.01.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 01/10/2020] [Accepted: 01/12/2020] [Indexed: 12/11/2022]
Abstract
The fetal immune system develops in a rather sterile environment relative to the outside world and, therefore, lacks antigenic education. Soon after birth, the newborn is exposed to the hostile environment of pathogens. Recently, animal- and limited human-based studies have indicated that help from the mother, upon transfer of leukocytes and their products via breast milk feeding, greatly assists the newborn's immune system. Here, I discuss the newest advances on how milk leukocytes impact early life immunity, with an emphasis on the development of the infant T cell repertoire and early immune responses in the periphery and gut-associated lymphoid tissue. A deeper understanding of these novel mechanistic insights may inform potential translational approaches to improving immunity in infants.
Collapse
Affiliation(s)
- Amale Laouar
- Surgery Department and the Child Health Institute of New Jersey, Robert Wood Johnson Medical School-Rutgers University, 89 French Street, New Brunswick, NJ 08901, USA.
| |
Collapse
|
23
|
Nochi T, Suzuki S, Ito S, Morita S, Furukawa M, Fuchimoto D, Sasahara Y, Usami K, Niimi K, Itano O, Kitago M, Matsuda S, Matsuo A, Suyama Y, Sakai Y, Wu G, Bazer FW, Watanabe K, Onishi A, Aso H. Elucidation of the Effects of a Current X-SCID Therapy on Intestinal Lymphoid Organogenesis Using an In Vivo Animal Model. Cell Mol Gastroenterol Hepatol 2020; 10:83-100. [PMID: 32017983 PMCID: PMC7210612 DOI: 10.1016/j.jcmgh.2020.01.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 01/26/2020] [Accepted: 01/27/2020] [Indexed: 12/26/2022]
Abstract
BACKGROUND & AIMS Organ-level research using an animal model lacking Il2rg, the gene responsible for X-linked severe combined immunodeficiency (X-SCID), is clinically unavailable and would be a powerful tool to gain deeper insights into the symptoms of patients with X-SCID. METHODS We used an X-SCID animal model, which was first established in our group by the deletion of Il2rg gene in pigs, to understand the clinical signs from multiple perspectives based on pathology, immunology, microbiology, and nutrition. We also treated the X-SCID pigs with bone marrow transplantation (BMT) for mimicking a current therapeutic treatment for patients with X-SCID and investigated the effect at the organ-level. Moreover, the results were confirmed using serum and fecal samples collected from patients with X-SCID. RESULTS We demonstrated that X-SCID pigs completely lacked Peyer's patches (PPs) and IgA production in the small intestine, but possessed some dysfunctional intestinal T and B cells. Another novel discovery was that X-SCID pigs developed a heterogeneous intestinal microflora and possessed abnormal plasma metabolites, indicating that X-SCID could be an immune disorder that affects various in vivo functions. Importantly, the organogenesis of PPs in X-SCID pigs was not promoted by BMT. Although a few isolated lymphoid follicles developed in the small intestine of BMT-treated X-SCID pigs, there was no evidence that they contributed to IgA production and microflora formation. Consistently, most patients with X-SCID who received BMT possessed abnormal intestinal immune and microbial environments regardless of the presence of sufficient serum IgG. CONCLUSIONS These results indicate that the current BMT therapies for patients with X-SCID may be insufficient to induce the organogenesis of intestinal lymphoid tissues that are associated with numerous functions in vivo.
Collapse
Affiliation(s)
- Tomonori Nochi
- International Education and Research Center for Food and Agricultural Immunology, Tohoku University Graduate School of Agricultural Science, Miyagi, Japan,International Research and Development Center for Mucosal Vaccine, Institute of Medical Science, University of Tokyo, Tokyo, Japan,Correspondence Address correspondence to: Tomonori Nochi, International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, 468-1 Aoba, Aramaki, Aoba-ku, Sendai, Miyagi 980-8572, Japan. fax: +81-22-757-4315.
| | - Shunichi Suzuki
- Division of Animal Science, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Japan
| | - Shun Ito
- International Education and Research Center for Food and Agricultural Immunology, Tohoku University Graduate School of Agricultural Science, Miyagi, Japan
| | - Shotaro Morita
- International Education and Research Center for Food and Agricultural Immunology, Tohoku University Graduate School of Agricultural Science, Miyagi, Japan
| | - Mutsumi Furukawa
- International Education and Research Center for Food and Agricultural Immunology, Tohoku University Graduate School of Agricultural Science, Miyagi, Japan
| | - Daiichiro Fuchimoto
- Division of Animal Science, Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Japan
| | - Yoji Sasahara
- Department of Pediatrics, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Katsuki Usami
- International Education and Research Center for Food and Agricultural Immunology, Tohoku University Graduate School of Agricultural Science, Miyagi, Japan
| | - Kanae Niimi
- International Education and Research Center for Food and Agricultural Immunology, Tohoku University Graduate School of Agricultural Science, Miyagi, Japan
| | - Osamu Itano
- Department of Hepato-Biliary-Pancreatic and Gastrointestinal Surgery, International University of Health and Welfare School of Medicine, Chiba, Japan
| | - Minoru Kitago
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Sachiko Matsuda
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Ayumi Matsuo
- International Education and Research Center for Food and Agricultural Immunology, Tohoku University Graduate School of Agricultural Science, Miyagi, Japan
| | - Yoshihisa Suyama
- International Education and Research Center for Food and Agricultural Immunology, Tohoku University Graduate School of Agricultural Science, Miyagi, Japan
| | - Yoshifumi Sakai
- International Education and Research Center for Food and Agricultural Immunology, Tohoku University Graduate School of Agricultural Science, Miyagi, Japan
| | - Guoyao Wu
- Department of Animal Science, Texas A&M University, College Station, Texas
| | - Fuller W. Bazer
- Department of Animal Science, Texas A&M University, College Station, Texas
| | - Kouichi Watanabe
- International Education and Research Center for Food and Agricultural Immunology, Tohoku University Graduate School of Agricultural Science, Miyagi, Japan
| | - Akira Onishi
- Department of Animal Science and Resources, Nihon University College of Bioresource Sciences, Kanagawa, Japan
| | - Hisashi Aso
- International Education and Research Center for Food and Agricultural Immunology, Tohoku University Graduate School of Agricultural Science, Miyagi, Japan
| |
Collapse
|
24
|
Yahaya T, Shemishere U. Association between Bioactive Molecules in Breast Milk and Type 1 Diabetes Mellitus. Sultan Qaboos Univ Med J 2020; 20:e5-e12. [PMID: 32190364 PMCID: PMC7065699 DOI: 10.18295/squmj.2020.20.01.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 08/23/2019] [Accepted: 11/03/2019] [Indexed: 02/05/2023] Open
Abstract
The association between breastfeeding and type 1 diabetes mellitus (T1DM) is controversial. However, several recent studies have established a link between these two factors, necessitating a need to review this subject to raise public awareness. Current research indicates that breast milk contains a variety of bioactive substances including immunoglobulins, oligosaccharides, insulin, lactoferrin, lysozyme, cytokines, epidermal growth factors, leukocytes, nucleotides, beneficial bacteria and vitamins. Such substances strengthen the breastfeeding infant's immune system, both directly, by increasing gut microbiota diversity and attacking harmful bacteria and pro-inflammatory molecules, and indirectly, by increasing thymus performance. Accordingly, a lack of or inadequate breastfeeding may predispose infants to several autoimmune disorders, including T1DM. Nursing mothers and caregivers are therefore advised to follow optimal breastfeeding practices prior to introducing complementary foods.
Collapse
Affiliation(s)
- Tajudeen Yahaya
- Department of Biology, Federal University Birnin Kebbi, Birnin Kebbi, Nigeria
| | - Ufuoma Shemishere
- Department of Biochemistry & Molecular Biology, Federal University Birnin Kebbi, Birnin Kebbi, Nigeria
| |
Collapse
|
25
|
Fujimura T, Lum SZC, Nagata Y, Kawamoto S, Oyoshi MK. Influences of Maternal Factors Over Offspring Allergies and the Application for Food Allergy. Front Immunol 2019; 10:1933. [PMID: 31507589 PMCID: PMC6716146 DOI: 10.3389/fimmu.2019.01933] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 07/30/2019] [Indexed: 12/19/2022] Open
Abstract
The prevalence of food allergy has been steadily rising worldwide with the highest incidence noted among younger children, and increasingly recognized as a growing public concern. The first known ingestion of foods often causes allergic reaction, suggesting that sensitization of offspring with food allergens may occur during pregnancy and/or through breastfeeding. This creates a milieu that shapes the neonatal immune responses to these allergens. However, the effects of maternal allergen exposure and maternal sensitization with allergens on development of allergies in offspring remain controversial. This review discusses recent advances from human data in our understanding of how maternal factors, namely, food allergens, allergen-specific immunoglobulins, cytokines, genetics, and environmental factors transferred during pregnancy or breastfeeding influence offspring allergies and how such effects may be applicable to food allergy. Based on information obtained from mouse models of asthma and food allergy, the review also dissects the mechanisms by which maternal factors, including the impact of immune complexes, transforming growth factor-β, vitamin A, and regulatory T-cell responses, contribute to the induction of neonatal tolerance vs. development of allergic responses to maternally transferred allergens.
Collapse
Affiliation(s)
- Takashi Fujimura
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States.,Hiroshima Research Center for Healthy Aging (HiHA), Graduate School of Advanced Sciences of Matter, Hiroshima University, Higashi-Hiroshima, Japan
| | | | - Yuka Nagata
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States.,Division of Gastrointestinal Pathophysiology, Institute of Natural Medicine, University of Toyama, Toyama, Japan
| | - Seiji Kawamoto
- Hiroshima Research Center for Healthy Aging (HiHA), Graduate School of Advanced Sciences of Matter, Hiroshima University, Higashi-Hiroshima, Japan
| | - Michiko K Oyoshi
- Division of Immunology, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
26
|
Cosgrove C, Dellacecca ER, van den Berg JH, Haanen JB, Nishimura MI, Le Poole IC, Bergmans HEN. Transgenerational transfer of gene-modified T cells. J Immunother Cancer 2019; 7:186. [PMID: 31307533 PMCID: PMC6631543 DOI: 10.1186/s40425-019-0657-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 06/27/2019] [Indexed: 01/05/2023] Open
Abstract
Tumor immunotherapy using gene-modified T cells has already met with considerable success in the treatment of metastatic melanoma and B cell lymphoma. With improving patient prognoses, new questions arise. In particular, the long-term consequences of treatment among individuals of childbearing age could now be considered. Former patients can carry a cohort of transgenic memory T cells long after treatment has ceased and the effector T cell population has contracted. When patients become parents well after treatment is completed, expectant mothers may still pass transgenic T cells to their unborn children. Consequences should be more measurable if the mother also breastfeeds the baby. Maternal T cells may shape immune responses in the child, can tolerize the child to maternal antigens, and might cause either beneficial or adverse effects in the offspring. The hypothesis put forth is that transgenic T cells transferred from mother to child during and after pregnancy might have consequences that have not been adequately considered to date. Depending on the targeted antigen and the MHC eventually required to present it, such transfer may be beneficial, uneventful or even damaging. Such potential consequences are addressed in this paper. The transgenic T cells might form a pocket of memory T cells in secondary lymphoid organs of the child, expand upon antigen stimulation, and react. However, simple measures might be devised to avoid any reason for concern. These considerations provide ample incentive to probe transgenerational transfer of transgenic T cells.
Collapse
Affiliation(s)
- Cormac Cosgrove
- Department of Dermatology/ Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Emilia R Dellacecca
- Department of Dermatology/ Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Joost H van den Berg
- Division of Molecular Oncology & Immunology/ Netherlands Cancer Institute, Amsterdam, Netherlands
| | - John B Haanen
- Division of Molecular Oncology & Immunology/ Netherlands Cancer Institute, Amsterdam, Netherlands
| | | | - I Caroline Le Poole
- Departments of Dermatology, Microbiology and Immunology, Northwestern University at Chicago, Lurie Comprehensive Cancer Center, Rm 3-121, 303 East Superior Street, Chicago, IL, 60611, USA.
| | - Hans E N Bergmans
- National Institute for Health and the Environment (RIVM), Bilthoven, Netherlands
| |
Collapse
|
27
|
Darby MG, Chetty A, Mrjden D, Rolot M, Smith K, Mackowiak C, Sedda D, Nyangahu D, Jaspan H, Toellner KM, Waisman A, Quesniaux V, Ryffel B, Cunningham AF, Dewals BG, Brombacher F, Horsnell WGC. Pre-conception maternal helminth infection transfers via nursing long-lasting cellular immunity against helminths to offspring. SCIENCE ADVANCES 2019; 5:eaav3058. [PMID: 31236458 PMCID: PMC6587632 DOI: 10.1126/sciadv.aav3058] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 04/24/2019] [Indexed: 06/01/2023]
Abstract
Maternal immune transfer is the most significant source of protection from early-life infection, but whether maternal transfer of immunity by nursing permanently alters offspring immunity is poorly understood. Here, we identify maternal immune imprinting of offspring nursed by mothers who had a pre-conception helminth infection. Nursing of pups by helminth-exposed mothers transferred protective cellular immunity to these offspring against helminth infection. Enhanced control of infection was not dependent on maternal antibody. Protection associated with systemic development of protective type 2 immunity in T helper 2 (TH2) impaired IL-4Rα-/- offspring. This maternally acquired immunity was maintained into maturity and required transfer (via nursing) to the offspring of maternally derived TH2-competent CD4 T cells. Our data therefore reveal that maternal exposure to a globally prevalent source of infection before pregnancy provides long-term nursing-acquired immune benefits to offspring mediated by maternally derived pathogen-experienced lymphocytes.
Collapse
Affiliation(s)
- Matthew G. Darby
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town 7925, South Africa
| | - Alisha Chetty
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town 7925, South Africa
| | - Dunja Mrjden
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town 7925, South Africa
| | - Marion Rolot
- Fundamental and Applied Research in Animals and Health (FARAH), Immunology-Vaccinology, Faculty of Veterinary Medicine (B43b), University of Liège, Liège, Belgium
| | - Katherine Smith
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town 7925, South Africa
- Institute of Infection and Immunity, University of Cardiff, Cardiff, UK
| | - Claire Mackowiak
- Laboratory of Molecular and Experimental Immunology and Neuro-genetics, UMR 7355, CNRS-University of Orleans and Le Studium Institute for Advanced Studies, Rue Dupanloup, 45000 Orléans, France
| | - Delphine Sedda
- Laboratory of Molecular and Experimental Immunology and Neuro-genetics, UMR 7355, CNRS-University of Orleans and Le Studium Institute for Advanced Studies, Rue Dupanloup, 45000 Orléans, France
| | - Donald Nyangahu
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town 7925, South Africa
| | - Heather Jaspan
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town 7925, South Africa
- Seattle Children’s Research Institute and Departments of Paediatrics and Global Health, University of Washington, Seattle, WA, USA
| | - Kai-Michael Toellner
- Institute of Immunology and Immunotherapy and School of Immunity and Infection, University of Birmingham, B15 2TT Birmingham, UK
| | - Ari Waisman
- Institute for Molecular Medicine, University of Mainz, Mainz, Germany
| | - Valerie Quesniaux
- Laboratory of Molecular and Experimental Immunology and Neuro-genetics, UMR 7355, CNRS-University of Orleans and Le Studium Institute for Advanced Studies, Rue Dupanloup, 45000 Orléans, France
| | - Bernhard Ryffel
- Laboratory of Molecular and Experimental Immunology and Neuro-genetics, UMR 7355, CNRS-University of Orleans and Le Studium Institute for Advanced Studies, Rue Dupanloup, 45000 Orléans, France
| | - Adam F. Cunningham
- Institute of Immunology and Immunotherapy and School of Immunity and Infection, University of Birmingham, B15 2TT Birmingham, UK
- Institute of Microbiology and Infection, University of Birmingham, B15 2TT Birmingham, UK
| | - Benjamin G. Dewals
- Fundamental and Applied Research in Animals and Health (FARAH), Immunology-Vaccinology, Faculty of Veterinary Medicine (B43b), University of Liège, Liège, Belgium
| | - Frank Brombacher
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town 7925, South Africa
- International Centre for Genetic Engineering and Biotechnology, Cape Town 7925, South Africa
- South African Medical Research Council, Cape Town, South Africa
| | - William G. C. Horsnell
- Institute of Infectious Disease and Molecular Medicine and Division of Immunology, University of Cape Town, Cape Town 7925, South Africa
- Laboratory of Molecular and Experimental Immunology and Neuro-genetics, UMR 7355, CNRS-University of Orleans and Le Studium Institute for Advanced Studies, Rue Dupanloup, 45000 Orléans, France
- Institute of Microbiology and Infection, University of Birmingham, B15 2TT Birmingham, UK
| |
Collapse
|
28
|
Gerlinskaya LA, Varlachev AV, Krotov GI, Kontsevaya GV, Moshkin MP. Mother-fetus immunogenetic dialogue as a factor of progeny immune system development. Vavilovskii Zhurnal Genet Selektsii 2019. [DOI: 10.18699/vj18.444] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Despite the advances in medicine, about 4 million children under the age of 6 months die annually around the world due to infection, which is 450 deaths per hour (UNISEF, 2009). The degree of development of the immune system of children born in time is determined by many factors, including the immunogenetic similarity or difference of mother and fetus organisms, which, in turn, is due to the genotypes of mating pairs, as well as the selection of surrogate mothers duringin vitrofertilization. From our review of the literature, it follows that immunogenetic interactions of mother and fetus organisms, which occur at all stages of pre- and postnatal development, have a signifcant effect on the resistance of offspring to infections and allergens. Before implantation, the mother’s immune responses are formed under the influence of semen fluid antigens, leukocytes and cytokines, as well as under the influence of the genes of the major histocompatibility complex, which are expressed in embryos at the stage of two cells. After implantation, transplacental transfer of immunoglobulins and immunocompetent cells becomes of immunomodulating importance. It is important to emphasize that, although substances with a high molecular weight usually do not pass through the placenta, this rule does not apply to immunoglobulin G (IgG), which, with a molecular weight of about 160 kDa, overcomes the transplacental barrier due to binding to the fetal Fc receptor. The level of IgG in newborns usually correlates with the level of maternal antibodies. During the period of natural feeding, the immune protection of newborns is provided by the mechanisms of innate immunity and the factors of humoral immunity of mothers. It has been shown that immunoglobulins from the milk of many animal species are transferred through the neonatal intestinal epithelium to the blood. Since breast milk contains large amounts of various immunoactive components, including proteins, cytokines, hormones, immunoglobulins, exosomes containing micro-RNA, and viable immune cells, the immunomodulating effects of breast milk persist even after elimination of maternal immunoglobulins from the blood of the offspring, up to maturation. Analysis of a large body of experimental data shows that the study of mechanisms of “motherfetus” and “mother-newborn” interactions are the basis of a knowledge base needed to fnd means of life-long directed modulation of the descendants’ immune status.
Collapse
|
29
|
Dewals BG, Layland LE, Prazeres da Costa C, Horsnell WG. Maternal helminth infections and the shaping of offspring immunity. Parasite Immunol 2018; 41:e12599. [PMID: 30372527 DOI: 10.1111/pim.12599] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Revised: 09/21/2018] [Accepted: 10/23/2018] [Indexed: 12/23/2022]
Abstract
Helminth infections leave a long-lasting immunological footprint on their hosts. Clinical studies have provided first evidence that maternal helminth infections can result in an altered immune profile in their offspring which can potentially shape how they respond to conditions throughout life. This can relate to changes in offspring induction of immune responses against other diseases. However, whether these changes result in actual changes in offspring ability to control disease is unclear. Our understanding of which immune mechanisms are altered and how they are changed is limited. In this review, we highlight what we know from human and mouse studies about this important context of helminth exposure. Moreover, we discuss how mechanisms such as antibody transfer, antigen exposure, maternal cell uptake, chimerism and epigenetics are all likely to be functional contributors to the striking changes that are seen in offspring born or nursed by helminth exposed mothers.
Collapse
Affiliation(s)
- Benjamin G Dewals
- Immunology-Vaccinology, Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine-FARAH, University of Liège, Liège, Belgium
| | - Laura E Layland
- Institute of Medical Microbiology, Immunology and Parasitology (IMMIP), University Hospital of Bonn, Bonn, Germany.,German Centre for Infection Research (DZIF), Partner Site, Bonn-Cologne, Bonn, Germany
| | - Clarissa Prazeres da Costa
- Institute of Medical Microbiology, Immunology and Hygiene, Technische Universität München, Munich, Germany
| | - William G Horsnell
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa.,Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK.,Laboratory of Molecular and Experimental Immunology and Neurogenetics, UMR 7355, CNRS-University of Orleans and Le Studium Institute for Advanced Studies, Orléans, France
| |
Collapse
|
30
|
Aydın MŞ, Yiğit EN, Vatandaşlar E, Erdoğan E, Öztürk G. Transfer and Integration of Breast Milk Stem Cells to the Brain of Suckling Pups. Sci Rep 2018; 8:14289. [PMID: 30250150 PMCID: PMC6155265 DOI: 10.1038/s41598-018-32715-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 09/12/2018] [Indexed: 01/19/2023] Open
Abstract
Beside its unique nutritional content breast milk also contains live cells from the mother. Fate of these cells in the offspring has not been adequately described. In this study, we aimed to detect and identify maternal cells in the suckling’s blood and the brain. Green fluorescent protein expressing transgenic female mice (GFP+) were used as foster mothers to breastfeed wildtype newborn pups. One week and two months after the birth, blood samples and brains of the sucklings were analyzed to detect presence of GFP+ cells by fluorescence activated cell sorting, polymerase chain reaction and immunohistochemistry on the brain sections and optically cleared brains. The tests confirmed that maternal cells were detectable in the blood and the brain of the pups and that they differentiated into both neuronal and glial cell types in the brain. This phenomenon represents breastfeeding – induced microchimerism in the brain with functional implications remain to be understood.
Collapse
Affiliation(s)
- Mehmet Şerif Aydın
- Regenerative and Restorative Medicine Research Center, Istanbul Medipol University, Istanbul, 34810, Turkey
| | - Esra Nur Yiğit
- Regenerative and Restorative Medicine Research Center, Istanbul Medipol University, Istanbul, 34810, Turkey
| | - Emre Vatandaşlar
- Regenerative and Restorative Medicine Research Center, Istanbul Medipol University, Istanbul, 34810, Turkey
| | - Ender Erdoğan
- Department of Histology and Embryology, Faculty of Medicine, Selcuk University, Konya, 42030, Turkey
| | - Gürkan Öztürk
- Regenerative and Restorative Medicine Research Center, Istanbul Medipol University, Istanbul, 34810, Turkey. .,Department of Physiology, International School of Medicine, Istanbul Medipol University, Istanbul, 34810, Turkey.
| |
Collapse
|
31
|
Engmann O. Dairy cows - an opportunity in the research field of non-genetic inheritance? ENVIRONMENTAL EPIGENETICS 2018; 4:dvy014. [PMID: 30034822 PMCID: PMC6049035 DOI: 10.1093/eep/dvy014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/03/2018] [Revised: 03/04/2018] [Accepted: 04/25/2018] [Indexed: 05/04/2023]
Abstract
More than 1 billion cattle are raised annually for meat and milk production. Dairy cows are repeatedly impregnated and separated from their calves, usually within the first 24 h after birth. Here, I suggest that dairy cows undergo a procedure comparable to the 'Maternal separation combined with unpredictable maternal stress' paradigm (MSUS), which is used to study the non-genetic inheritance (NGI) of phenotypes in rodents. I discuss what research on dairy cows may bring to the research field of NGI. The resulting research findings are likely to have benefits to our understanding of MSUS, NGI and consumer safety.
Collapse
Affiliation(s)
- Olivia Engmann
- Brain Research Institute, Faculty of Medicine, University of Zurich, Winterthurer Strasse 190, 8057 Zurich, Switzerland
| |
Collapse
|
32
|
Abstract
Breast feeding has been associated with improved infant outcomes in multiple aspects, including immune outcomes such as infections and potentially atopy and autoimmunity. However associations do not necessarily implicate cause and effect and at this point, exactly how breast feeding and components of breast milk may modulate the infant's immune compartment remains unclear, especially in humans. Some lines of evidence suggest that breastfeeding affects the development of the infant's thymus, a critical organ for T cell development. This may be a direct effect mediated by breast milk components or alternatively, a secondary effect from the impact of breast feeding on the infant's gut microbiome. Here we discuss the potential mechanisms and impact of this association between breast feeding and thymic development.
Collapse
Affiliation(s)
- Peter S Hsu
- Allergy and Immunology, Kids Research, The Children's Hospital at Westmead, Sydney, NSW, Australia.,Discipline of Child and Adolescent Health, The University of Sydney, Sydney, NSW, Australia
| | - Ralph Nanan
- Charles Perkins Centre Nepean, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
33
|
Ghosh MK, Muller HK, Walker AM. Lactation-Based Maternal Educational Immunity Crosses MHC Class I Barriers and Can Impart Th1 Immunity to Th2-Biased Recipients. THE JOURNAL OF IMMUNOLOGY 2017; 199:1729-1736. [PMID: 28747348 DOI: 10.4049/jimmunol.1601375] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 06/23/2017] [Indexed: 11/19/2022]
Abstract
We have previously demonstrated lactational transfer of T cell-based immunity from dam to foster pup. In the short term, a significant part of transferred immunity is passive cellular immunity. However, as time progresses, this is replaced by what we have described as maternal educational immunity such that by young adulthood, all immune cells responding to a foster dam immunogen are the product of the foster pup's thymus. To reduce confounding factors, this original demonstration used congenic/syngeneic dam and foster pup pairs. In this study, we investigated lactational transfer of immunity to Mycobacterium tuberculosis in MHC class I-mismatched animals, as well as from Th1-biased dams to Th2-biased foster pups. Using immunized C57BL/6J dams, lactational transfer to nonimmunized BALB/cJ foster pups resulted in much greater immunity than direct immunization in 5-wk-old pups (ex vivo assay of pup splenocytes). At this age, 82% of immunogen-responding cells in the pup spleen were produced through maternal educational immunity. FVB/NJ nonimmunized foster recipients had a greater number of maternal cells in the spleen and thymus but a much larger percentage was Foxp3+, resulting in equivalent immunity to direct immunization. Depletion of maternal Foxp3+ cells from pup splenocytes illustrated a substantial role for lactationally transferred dam regulatory T cells in suppression of the ex vivo response in FVB/NJ, but not BALB/cJ, recipients. We conclude that lactational transfer of immunity can cross MHC class I barriers and that Th1 immunity can be imparted to Th2-biased offspring; in some instances, it can be greater than that achieved by direct immunization.
Collapse
Affiliation(s)
- Mrinal K Ghosh
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA 92521; and
| | - H Konrad Muller
- School of Medicine, University of Tasmania, Hobart 7000, Tasmania, Australia
| | - Ameae M Walker
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA 92521; and
| |
Collapse
|
34
|
Dill R, Walker AM. Role of Prolactin in Promotion of Immune Cell Migration into the Mammary Gland. J Mammary Gland Biol Neoplasia 2017; 22:13-26. [PMID: 27900586 PMCID: PMC5313375 DOI: 10.1007/s10911-016-9369-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Accepted: 11/21/2016] [Indexed: 01/13/2023] Open
Abstract
Immune cells in the mammary gland play a number of important roles, including protection against infection during lactation and, after passing into milk, modulation of offspring immunity. However, little is known about the mechanism of recruitment of immune cells to the lactating gland in the absence of infection. Given the importance of prolactin to other aspects of lactation, we hypothesized it would also play a role in immune cell recruitment. Prolactin treatment of adult female mice for a period equivalent to pregnancy and the first week of lactation increased immune cell flux through the mammary gland, as reflected in the number of immune cells in mammary gland-draining, but not other lymph nodes. Conditioned medium from luminal mammary epithelial HC11 cell cultures was chemo-attractive to CD4+ and CD8+ T cells, CD4+ and CD8+ memory T cells, B cells, macrophages, monocytes, eosinophils, and neutrophils. Prolactin did not act as a direct chemo-attractant, but through effects on luminal mammary epithelial cells, increased the chemo-attractant properties of conditioned medium. Macrophages and neutrophils constitute the largest proportion of cells in milk from healthy glands. Depletion of CCL2 and CXCL1 from conditioned medium reduced chemo-attraction of monocytes and neutrophils, and prolactin increased expression of these two chemokines in mammary epithelial cells. We conclude that prolactin is an important player in the recruitment of immune cells to the mammary gland both through its activities to increase epithelial cell number as well as production of chemo-attractants on a per cell basis.
Collapse
Affiliation(s)
- Riva Dill
- Division of Biomedical Sciences, School of Medicine, University of California, 900 University Ave. 1260 Webber Hall, Riverside, CA, 92521, USA.
| | - Ameae M Walker
- Division of Biomedical Sciences, School of Medicine, University of California, 900 University Ave. 1260 Webber Hall, Riverside, CA, 92521, USA
| |
Collapse
|
35
|
Maternal house dust mite exposure during pregnancy enhances severity of house dust mite-induced asthma in murine offspring. J Allergy Clin Immunol 2017; 140:1404-1415.e9. [PMID: 28192144 DOI: 10.1016/j.jaci.2016.12.972] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 12/06/2016] [Accepted: 12/12/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND Atopic status of the mother and maternal exposure to environmental factors are associated with increased asthma risk. Moreover, animal models demonstrate that exposure to allergens in strongly sensitized mothers influences offspring asthma development, suggesting that in utero exposures can influence offspring asthma. However, it is unclear whether maternal exposure to common human allergens such as house dust mite (HDM), in the absence of additional adjuvants, influences offspring asthma development. OBJECTIVE We sought to determine whether maternal HDM exposure influences asthma development in offspring. METHODS Pregnant female mice were exposed to PBS or HDM during pregnancy. Using offspring of PBS- or HDM-exposed mothers, the magnitude of HDM or Aspergillus fumigatus (AF) extract-induced airway hyperresponsiveness (AHR), airway inflammation, immunoglobulin production, TH2-associated cytokine synthesis, and pulmonary dendritic cell activity was assessed. RESULTS Compared with offspring of PBS-exposed mothers, offspring of HDM-exposed mothers demonstrate increased AHR, airway inflammation, TH2 cytokine production, and immunoglobulin levels and a modest decrease in the phagocytic capacity of pulmonary macrophage populations following HDM exposure. Increased sensitivity to AF-induced airway disease was not observed. Offspring of HDM-exposed B-cell-deficient mothers also demonstrated increased HDM-induced AHR, suggesting that transfer of maternal immunoglobulins is not required. CONCLUSIONS Our data demonstrate that maternal exposure to HDM during pregnancy increases asthma sensitivity in offspring in an HDM-specific manner, suggesting that vertical transmission of maternal immune responses may be involved. These findings have important implications for regulation of asthma risk, and suggest that exposure to HDM in the developed world may have underappreciated influences on the overall prevalence of allergic asthma.
Collapse
|