1
|
Cristóbal-Cañadas D, Parrón-Carrillo R, Parrón-Carreño T. Exosomes in Breast Milk: Their Impact on the Intestinal Microbiota of the Newborn and Therapeutic Perspectives for High-Risk Neonates. Int J Mol Sci 2025; 26:3421. [PMID: 40244312 PMCID: PMC11989396 DOI: 10.3390/ijms26073421] [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: 03/01/2025] [Revised: 04/03/2025] [Accepted: 04/04/2025] [Indexed: 04/18/2025] Open
Abstract
Breast milk exosomes are essential for the nutrition and immune development of the newborn. These 30-150 nm extracellular vesicles contain microRNAs (miRNAs), mesessenger RNAS (mRNA)s, proteins and lipids that facilitate cellular communication and modulate the neonatal immune system. In this article, we analyse the impact of breast milk exosomes on the intestinal microbiota of the newborn, especially in high-risk neonates such as preterm infants or neonates at risk of necrotising enterocolitis (NEC). Exosomes promote the colonisation of beneficial bacteria such as Bifidobacterium and Lactobacillus and strengthen the intestinal barrier. They also regulate the immune response, balancing defence against pathogens and tolerance to non-pathogenic antigens. This effect is key for high-risk infants, who benefit from their anti-inflammatory and preventive properties against complications such as NEC. Research points to their potential therapeutic uses in neonatal care, opening up new opportunities to improve the health of vulnerable newborns through the protective effects of breast milk exosomes.
Collapse
Affiliation(s)
| | - Rocio Parrón-Carrillo
- Department of Psychology, Faculty of Health Sciences, University of Almeria, 04120 Almería, Spain;
| | - Tesifón Parrón-Carreño
- Department of Nursing, Physiotherapy and Medicine, University of Almería, 04120 Almería, Spain
| |
Collapse
|
2
|
Cendron F, Franzoi M, De Marchi M, Rosani U, Penasa M. Characterization of microRNA in cow milk and colostrum. J Dairy Sci 2025; 108:2981-2994. [PMID: 39647631 DOI: 10.3168/jds.2024-25145] [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: 05/10/2024] [Accepted: 11/10/2024] [Indexed: 12/10/2024]
Abstract
MicroRNAs (miRNAs) are short RNA molecules, typically 21 to 25 nucleotides long, synthesized within eukaryotic cells. They play a crucial role in coordinating complex gene expression regulatory networks. The miRNAs are involved in post-transcriptional regulation, either by degrading target mRNA or suppressing their transcription, thereby influencing protein translation. Additionally, characteristics of farm production systems exert a noticeable influence on miRNA profiles, shaping the functional properties of bovine milk. In this study, milk and colostrum samples of Holstein cows (Bos taurus) were collected for RNA extraction and sequencing. Sequencing data underwent quality controls, and short noncoding RNA reads were analyzed to identify and compare miRNA profiles. The sequencing runs yielded 73,567,661 and 44,283,978 reads for milk and colostrum samples, respectively. Following trimming, 8,335,860 and 7,778,212 reads for milk and colostrum samples were retained. A total of 4.3% milk and 37.8% colostrum reads were identified as miRNAs. Overall, 157 miRNAs were common to milk and colostrum, 2 were unique to milk, and 90 were unique to colostrum. After applying a minimal cutoff of 100 reads per miRNA, 146 miRNAs were considered as expressed. In milk, bta-miR-101 was unique; in colostrum, bta-miR-18a, bta-miR-262-3p, bta-miR-130a, and bta-miR-224 were unique; and 141 miRNAs were common to colostrum and milk. Chromosome distribution of these miRNAs varied, with higher representation on autosome 19 and sexual chromosome X. Overall, colostrum contained a greater number of miRNAs, although specific miRNAs such as miR-11-988b, miR-140, and miR-146b were more abundant in milk. Conversely, miRNAs such as miR-7a-5p, let-7e, and miR-16a were more prevalent in colostrum. Validation through quantitative PCR confirmed the expression levels of selected miRNAs from RNA sequencing, establishing concordance between the methods. Analysis of gene regulation by specific miRNAs identified key miRNAs (e.g., miR-223, miR-181a, and miR-155) involved in regulating various genes. Distinct miRNA profiles in milk and colostrum suggest various functionalities, potentially influencing calf development and immune responses. The study unveils a comprehensive map of miRNAs in milk and colostrum and highlights their potential roles in regulating biological processes, as they play an essential role in calf development, disease resistance, and metabolism.
Collapse
Affiliation(s)
- F Cendron
- Department of Agronomy, Food, Natural Resources, Animals and Environment (DAFNAE), University of Padova, 35020 Legnaro (PD), Italy
| | - M Franzoi
- Department of Agronomy, Food, Natural Resources, Animals and Environment (DAFNAE), University of Padova, 35020 Legnaro (PD), Italy
| | - M De Marchi
- Department of Agronomy, Food, Natural Resources, Animals and Environment (DAFNAE), University of Padova, 35020 Legnaro (PD), Italy
| | - U Rosani
- Department of Biology (DiBio), University of Padova, 35131 Padova, Italy.
| | - M Penasa
- Department of Agronomy, Food, Natural Resources, Animals and Environment (DAFNAE), University of Padova, 35020 Legnaro (PD), Italy
| |
Collapse
|
3
|
Arenas G, Barrera MJ, Contreras-Duarte S. The Impact of Maternal Chronic Inflammatory Conditions on Breast Milk Composition: Possible Influence on Offspring Metabolic Programming. Nutrients 2025; 17:387. [PMID: 39940245 PMCID: PMC11820913 DOI: 10.3390/nu17030387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/09/2025] [Accepted: 01/20/2025] [Indexed: 02/14/2025] Open
Abstract
Breastfeeding is the best way to provide newborns with crucial nutrients and produce a unique bond between mother and child. Breast milk is rich in nutritious and non-nutritive bioactive components, such as immune cells, cytokines, chemokines, immunoglobulins, hormones, fatty acids, and other constituents. Maternal effects during gestation and lactation can alter these components, influencing offspring outcomes. Chronic inflammatory maternal conditions, such as obesity, diabetes, and hypertension, impact breast milk composition. Breast milk from obese mothers exhibits changes in fat content, cytokine levels, and hormonal concentrations, potentially affecting infant growth and health. Similarly, diabetes alters the composition of breast milk, impacting immune factors and metabolic markers. Other pro-inflammatory conditions, such as dyslipidemia and metabolic syndrome, have been barely studied. Thus, maternal obesity, diabetes, and altered tension parameters have been described as modifying the composition of breast milk in its macronutrients and other important biomolecules, likely affecting the offspring's weight. This review emphasizes the impact of chronic inflammatory conditions on breast milk composition and its potential implications for offspring development through the revision of full-access original articles.
Collapse
Affiliation(s)
- Gabriela Arenas
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago 7510602, Chile;
| | - María José Barrera
- Facultad de Odontología y Ciencias de la Rehabilitación, Universidad San Sebastián, Santiago 7510157, Chile;
| | - Susana Contreras-Duarte
- Facultad de Ciencias para el Cuidado de la Salud, Universidad San Sebastián, Santiago 8420524, Chile
| |
Collapse
|
4
|
Aljani B, Lindner A, Weigelt M, Zhao M, Sharma V, Bonifacio E, Jones P, Eugster A. Small RNA-Seq and real time rt-qPCR reveal islet miRNA released under stress conditions. Islets 2024; 16:2392343. [PMID: 39154325 PMCID: PMC11332650 DOI: 10.1080/19382014.2024.2392343] [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: 04/29/2024] [Revised: 08/09/2024] [Accepted: 08/09/2024] [Indexed: 08/19/2024] Open
Abstract
Replacement of beta cells through transplantation is a potential therapeutic approach for individuals with pancreas removal or poorly controllable type 1 diabetes. However, stress and death of beta cells pose significant challenges. Circulating miRNA has emerged as potential biomarkers reflecting early beta cell stress and death, allowing for timely intervention. The aim of this study was to identify miRNAs as potential biomarkers for beta cell health. Literature review combined with small RNA sequencing was employed to select islet-enriched miRNA. The release of those miRNA was assessed by RT-qPCR in vivo, using a streptozotocin induced diabetes mouse model and in vitro, through mouse and human islets exposed to varying degrees of hypoxic and cytokine stressors. Utilizing the streptozotocin induced model, we identified 18 miRNAs out of 39 candidate islet-enriched miRNA to be released upon islet stress in vivo. In vitro analysis of culture supernatants from cytokine and/or hypoxia stressed islets identified the release of 45 miRNAs from mouse and 8 miRNAs from human islets. Investigation into the biological pathways targeted by the cytokine- and/or hypoxia-induced miRNA suggested the involvement of MAPK and PI3K-Akt signaling pathways in both mouse and human islets. We have identified miRNAs associated with beta cell health and stress. The findings allowed us to propose a panel of 47 islet-related human miRNA that is potentially valuable for application in clinical contexts of beta cell transplantation and presymptomatic early-stage type 1 diabetes.
Collapse
Affiliation(s)
- Bssam Aljani
- Faculty of Medicine, Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Annett Lindner
- Faculty of Medicine, Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Marc Weigelt
- Faculty of Medicine, Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Min Zhao
- German Center for Environmental Health, Institute of Diabetes Research, Helmholtz Munich, Munich, Germany
| | - Virag Sharma
- Faculty of Medicine, Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Ezio Bonifacio
- Faculty of Medicine, Center for Regenerative Therapies Dresden, Dresden, Germany
- Faculty of Medicine, German Center for Diabetes Research (DZD), Paul Langerhans Institute Dresden of Helmholtz Centre Munich at University Clinic Carl Gustav Carus of TU Dresden, Dresden, Germany
| | - Peter Jones
- Department of Diabetes, School of Cardiovascular and Metabolic Medicine & Sciences, King’s College London, London, UK
| | - Anne Eugster
- Faculty of Medicine, Center for Regenerative Therapies Dresden, Dresden, Germany
| |
Collapse
|
5
|
Yang W, Lin P, Gao R, Fang Z, Wang Z, Ma Z, Shi J, Yu W. Cell-derived biomimetic drug delivery system for inflammatory bowel disease therapy. Mater Today Bio 2024; 29:101332. [PMID: 39606424 PMCID: PMC11600033 DOI: 10.1016/j.mtbio.2024.101332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 10/24/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic recurrent disease with an increasing incidence year by year. At present, no safe and effective treatment for IBD exists. Thus, there is an urgent need to create new therapeutic options that have decreased adverse effects and positive clinical efficacy. A range of nanomaterials have fueled the advancement of nanomedicine in recent years, which is establishing more appealing and prospective treatment approaches for IBD. However, traditional synthetic nanomaterials still have some problems in the IBD drug delivery process, such as weak targeting ability of vectors, difficulty escaping immune surveillance, and poor biosecurity. Natural sources of biological nanomaterials have been identified to solve the above problems. A drug delivery system based on bionic technology is expected to achieve a new breakthrough in the targeted therapy of IBD by nanotechnology due to its organic integration of low immunogenicity and natural targeting of biological materials and the controllability and versatility of synthetic nanocarrier design. We begin this review by outlining the fundamental traits of both inflammatory and healthy intestinal microenvironments. Subsequently, we review the latest application of a cell-derived bionic drug delivery system in IBD therapy. Finally, we discuss the development prospects of this delivery system and challenges to its clinical translation. Biomimetic nanotherapy is believed to offer a new strategy for the treatment of IBD.
Collapse
Affiliation(s)
- Wenjing Yang
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
| | - Peihong Lin
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
| | - Rui Gao
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
| | - Zhengyu Fang
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
| | - Zhouru Wang
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
| | - Zhen Ma
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
| | - Jing Shi
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
| | - Wenying Yu
- School of Pharmacy, Hangzhou Medical College, Hangzhou, 310013, China
- Key Laboratory of Neuropsychiatric Drug Research of Zhejiang Province, Hangzhou Medical College, Hangzhou, 310013, China
| |
Collapse
|
6
|
Wijenayake S, Eisha S, Purohit MK, McGowan PO. Milk derived extracellular vesicle uptake in human microglia regulates the DNA methylation machinery : Short title: milk-derived extracellular vesicles and the epigenetic machinery. Sci Rep 2024; 14:28630. [PMID: 39562680 PMCID: PMC11576889 DOI: 10.1038/s41598-024-79724-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 11/12/2024] [Indexed: 11/21/2024] Open
Abstract
Mammalian milk contains milk-derived extracellular vesicles (MEVs), a group of biological nanovesicles that transport macromolecules. Their ability to cross the blood brain barrier and the presence of cargo capable of modifying gene function have led to the hypothesis that MEVs may play a role in brain function and development. Here, we investigated the uptake of MEVs by human microglia cells in vitro and explored the functional outcomes of MEV uptake. We examined the expression of the miR-148/152 family, highly abundant MEV microRNAs, that directly suppress the translation of DNA methyltransferase (DNMT) enzymes crucial for catalyzing DNA methylation modifications. We also measured phenotypic and inflammatory gene expression in baseline homeostatic and IFN-γ primed microglia to determine if MEVs induce anti-inflammatory effects. We found that MEVs are taken up and localize in baseline and primed microglia. In baseline microglia, MEV supplementation reduced miR-148a-5P levels, increased DNMT1 transcript, protein abundance, and enzymatic activity, compared to cells that did not receive MEVs. In primed microglia, MEV supplementation decreased miR-148a-5P levels and increased DNMT1 protein abundance, but DNMT1 transcript and enzymatic levels remained unchanged. Contrary to predictions, MEV supplementation failed to attenuate pro-inflammatory IL1β expression in primed microglia. This study provides the first evidence of MEV uptake by a brain macrophage, suggesting a potential role in regulating epigenetic machinery and neuroimmune modulation.
Collapse
Affiliation(s)
- Sanoji Wijenayake
- Department of Biology, The University of Winnipeg, Winnipeg, Manitoba, Canada.
- Department of Biological Sciences and Center for Environmental Epigenetics and Development, Scarborough Campus, University of Toronto, Toronto, ON, Canada.
| | - Shafinaz Eisha
- Department of Biological Sciences and Center for Environmental Epigenetics and Development, Scarborough Campus, University of Toronto, Toronto, ON, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Mansi Kamlesh Purohit
- Department of Biological Sciences and Center for Environmental Epigenetics and Development, Scarborough Campus, University of Toronto, Toronto, ON, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Patrick Owen McGowan
- Department of Biological Sciences and Center for Environmental Epigenetics and Development, Scarborough Campus, University of Toronto, Toronto, ON, Canada.
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada.
- Department of Psychology, University of Toronto, Toronto, ON, Canada.
- Department of Physiology, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
7
|
Chiba T, Takaguri A, Mikuma T, Kimura T, Maeda T. Smoking-induced suppression of β-casein in milk is associated with an increase in miR-210-5p expression in mammary epithelia. Biochem Biophys Rep 2024; 39:101773. [PMID: 39044768 PMCID: PMC11263956 DOI: 10.1016/j.bbrep.2024.101773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/29/2024] [Accepted: 07/01/2024] [Indexed: 07/25/2024] Open
Abstract
Smoking during lactation harmfully affects the amount and constituents of breast milk. Infants who consume breast milk containing miR-210-5p may have a higher risk of brain-related diseases. We investigated whether smoking during lactation decreases β-casein concentrations in milk and whether miR-210-5p expression is involved in smoking-induced β-casein suppression. During lactation, maternal CD1 mice were exposed to cigarette smoke (1.7 mg of tar and 14 mg of nicotine) in a smoke chamber for 1 h twice/day for five consecutive days. Control mice were placed in an air-filled chamber equivalent in size to the smoke chamber, with maternal separation times identical to those of the smoked mice. Maternal exposure to smoke during lactation significantly decreased β-casein expression in the mammary epithelia of smoked mice compared to that of the control mice. Signal transducer and activator transcription 5 (STAT5) and phosphorylated STAT5 (pSTAT5) are transcription factors involved in β-casein expression. In the mammary epithelia of smoked mice, the pSTAT5 and STAT5 levels were significantly lower, and miR-210-5p expression was significantly higher than that of the control mice. The β-casein, pSTAT5, and STAT5 protein levels of miR-210-5p mimic-transfected human mammary epithelial MCF-12A cells were significantly lower than those of control siRNA-transfected cells. These results indicate that smoke exposure led to an increase in miR-210-5p expression in mammary epithelium and a decrease in pSTAT5 and β-casein protein levels through the inhibition of STAT5 expression. Moreover, nicotine treatment decreased β-casein protein levels and increased miR-210-5p expression in non-malignant human mammary epithelial MCF-12A cells in a concentration-dependent manner, demonstrating that nicotine significantly affects the β-casein and miR-210-5p levels of breast milk. These results highlight the adverse effects of smoking on breast milk, providing essential information for healthcare professionals and general citizens.
Collapse
Affiliation(s)
- Takeshi Chiba
- Department of Pharmacy, Juntendo University Hospital, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-8431, Japan
- Laboratory of Clinical Pharmacology, Faculty of Pharmacy, Juntendo University, 6-8-1 Hinode, Urayasu-shi, Chiba, 279-0013, Japan
| | - Akira Takaguri
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 15-4-1, Maeda 7-jo, Teine-ku, Sapporo-shi, Hokkaido, 006-8585, Japan
- Creation Research Institute of Life Science in KITA-no-DAICHI, Hokkaido University of Science, 15-4-1, Maeda 7-jo, Teine-ku, Sapporo-shi, Hokkaido, 006-8585, Japan
| | - Toshiyasu Mikuma
- Department of Physical and Analytical Sciences, Nihon Pharmaceutical University, 10281 Komuro, Ina-machi, Kitaadachi-gun, Saitama, 362-0806, Japan
| | - Toshimi Kimura
- Department of Pharmacy, Juntendo University Hospital, 3-1-3 Hongo, Bunkyo-ku, Tokyo, 113-8431, Japan
- Laboratory of Clinical Pharmacology, Faculty of Pharmacy, Juntendo University, 6-8-1 Hinode, Urayasu-shi, Chiba, 279-0013, Japan
| | - Tomoji Maeda
- Department of Clinical Pharmacology and Pharmaceutics, Nihon Pharmaceutical University, 10281 Komuro, Ina-machi, Kitaadachi-gun, Saitama-shi, 362-0806, Japan
| |
Collapse
|
8
|
Lanctôt S, Blouin R, Thibault C, Lacasse P. Effect of milk stasis on mammary gland involution and the microRNA profile. J Dairy Sci 2024; 107:7435-7445. [PMID: 38788842 DOI: 10.3168/jds.2023-24603] [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: 12/22/2023] [Accepted: 04/02/2024] [Indexed: 05/26/2024]
Abstract
The presence of an autocrine factor in milk that can trigger mammary gland involution was proposed more than 50 yr ago. To provide evidence for the existence of one or more autocrine factors, 10 multiparous cows in late lactation were quarter-milked for 7 d. Following this baseline period, the right front quarter of each cow was left unmilked, and the other quarters were milked for 7 d. Before the last milking of that period, milk (mammary secretions) was collected aseptically from both front quarters. After that milking, 250 mL of the collected samples were infused in the cows' respective rear quarters. No quarters were milked for the following 7 d (milk stasis period), and quarter milking was then resumed in all quarters for the last 7 d of the experiment (remilking period). Quarter milk samples were collected during the baseline period, before the milk stasis period, and during the remilking period. These samples were used for measuring milk components and the concentration of involution markers (SCC, BSA, and lactoferrin). Samples of mammary secretions were collected manually from the quarters during the milk stasis period for involution marker determination. We extracted RNA from samples collected from front quarters before the last milking before the milk stasis period for microRNA (miRNA) determination. As anticipated, the longer milk stasis period implemented for the right front quarter resulted in a more advanced involution than in the left front quarter, based on the concentration of involution markers in the mammary secretions, lower milk production recovery, and changes in milk composition during the remilking period. All 3 involution marker concentrations in the mammary secretions increased in both rear quarters, but were greater in the right quarter secretions than in the left quarter secretions. Resuming milking reinitiated milk production in all quarters, but milk production recovery in the right rear quarters was less robust than that in the left rear quarters (54.3 ± 1.4% vs. 61.6 ± 1.4%, respectively). Milk from the quarters infused with mammary secretions (right rear) had a lower lactose content, but a higher milk protein content and higher SCC than the quarters infused with milk. We detected a total of 359 miRNAs, 76 of which were differentially expressed in milk and mammary secretions. Expression of bta-miR-221 and bta-miR-223 was upregulated in mammary secretions 34- and 40-fold, respectively. The results of the present experiment support the contention that milk stasis leads to the accumulation of one or more factors that trigger involution. The results also indicate that milk stasis leads to changes in the miRNA profile of the milk, but whether such changes are a cause or a consequence of the involution process remains to be established.
Collapse
Affiliation(s)
- S Lanctôt
- Département de Biologie, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| | - R Blouin
- Département de Biologie, Université de Sherbrooke, Sherbrooke, QC J1K 2R1, Canada
| | - C Thibault
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, Sherbrooke, QC J1M 0C8, Canada
| | - P Lacasse
- Sherbrooke Research and Development Centre, Agriculture and Agri-Food Canada, Sherbrooke, QC J1M 0C8, Canada.
| |
Collapse
|
9
|
Melnik BC, Weiskirchen R, Stremmel W, John SM, Schmitz G. Risk of Fat Mass- and Obesity-Associated Gene-Dependent Obesogenic Programming by Formula Feeding Compared to Breastfeeding. Nutrients 2024; 16:2451. [PMID: 39125332 PMCID: PMC11314333 DOI: 10.3390/nu16152451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/21/2024] [Accepted: 07/25/2024] [Indexed: 08/12/2024] Open
Abstract
It is the purpose of this review to compare differences in postnatal epigenetic programming at the level of DNA and RNA methylation and later obesity risk between infants receiving artificial formula feeding (FF) in contrast to natural breastfeeding (BF). FF bears the risk of aberrant epigenetic programming at the level of DNA methylation and enhances the expression of the RNA demethylase fat mass- and obesity-associated gene (FTO), pointing to further deviations in the RNA methylome. Based on a literature search through Web of Science, Google Scholar, and PubMed databases concerning the dietary and epigenetic factors influencing FTO gene and FTO protein expression and FTO activity, FTO's impact on postnatal adipogenic programming was investigated. Accumulated translational evidence underscores that total protein intake as well as tryptophan, kynurenine, branched-chain amino acids, milk exosomal miRNAs, NADP, and NADPH are crucial regulators modifying FTO gene expression and FTO activity. Increased FTO-mTORC1-S6K1 signaling may epigenetically suppress the WNT/β-catenin pathway, enhancing adipocyte precursor cell proliferation and adipogenesis. Formula-induced FTO-dependent alterations of the N6-methyladenosine (m6A) RNA methylome may represent novel unfavorable molecular events in the postnatal development of adipogenesis and obesity, necessitating further investigations. BF provides physiological epigenetic DNA and RNA regulation, a compelling reason to rely on BF.
Collapse
Affiliation(s)
- Bodo C. Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, D-49076 Osnabrück, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, D-52074 Aachen, Germany;
| | - Wolfgang Stremmel
- Praxis for Internal Medicine, Beethovenstrasse 2, D-76530 Baden-Baden, Germany;
| | - Swen Malte John
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, D-49076 Osnabrück, Germany
- Institute for Interdisciplinary Dermatological Prevention and Rehabilitation (iDerm), University of Osnabrück, D-49076 Osnabrück, Germany;
| | - Gerd Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital of Regensburg, D-93053 Regensburg, Germany;
| |
Collapse
|
10
|
Papakonstantinou E, Dragoumani K, Chrousos GP, Vlachakis D. Exosomal Epigenetics. EMBNET.JOURNAL 2024; 29:e1049. [PMID: 38845750 PMCID: PMC11155272 DOI: 10.14806/ej.29.0.1049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
Epigenetics is the study of heritable changes in gene expression that occur without changes to the underlying DNA sequence. Epigenetic modifications can include DNA methylation, histone modifications, and non-coding RNAs, among others. These modifications can influence the expression of genes by altering the way DNA is packaged and accessed by transcriptional machinery, thereby affecting cellular function and behavior. Epigenetic modifications can be influenced by a variety of factors, including environmental exposures, lifestyle factors, and aging, whilst abnormal epigenetic modifications have been implicated in a range of diseases, including cancer, neurodegenerative disorders, and cardiovascular disease. The study of epigenetics has the potential to provide new insights into the mechanisms of disease and could lead to the development of new diagnostic and therapeutic strategies. Exosomes can transfer epigenetic information to recipient cells, thereby influencing various physiological and pathological processes, and the identification of specific epigenetic modifications that are associated with a particular disease could lead to the development of targeted therapies that restore normal gene expression patterns. In recent years, the emerging role of exosomal epigenetics in human breast milk, highlighting its significance in infant nutrition and immune development. Milk exosomes are shown to carry epigenetic regulators, including miRNAs and long non-coding RNAs, which can modulate gene expression in recipient cells. These epigenetic modifications mediated by milk exosomal RNAs have implications for the development of the gastrointestinal tract, immune system, and metabolic processes in infants.
Collapse
Affiliation(s)
- Eleni Papakonstantinou
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
- University Research Institute of Maternal and Child Health & Precision Medicine, and UNESCO Chair on Adolescent Health Care, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Konstantina Dragoumani
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
| | - George P Chrousos
- University Research Institute of Maternal and Child Health & Precision Medicine, and UNESCO Chair on Adolescent Health Care, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Dimitrios Vlachakis
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
- University Research Institute of Maternal and Child Health & Precision Medicine, and UNESCO Chair on Adolescent Health Care, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
- School of Informatics, Faculty of Natural & Mathematical Sciences, King's College London, London, U.K
| |
Collapse
|
11
|
Papakonstantinou E, Dragoumani K, Mitsis T, Chrousos GP, Vlachakis D. Milk exosomes and a new way of communication between mother and child. EMBNET.JOURNAL 2024; 29:e1050. [PMID: 38845751 PMCID: PMC11155261 DOI: 10.14806/ej.29.0.1050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
Extracellular vesicles are a heterogeneous group of lipid-bound vesicles released by cells into the extracellular space. EVs are an important mediator of intercellular communications and carry a wide variety of molecules that exert a biological function, such as lipids, nucleic acids, proteins, ions, and adenosine triphosphate (ATP). Extracellular vesicles are classified into microvesicles, exosomes, and apoptotic bodies depending on their biogenesis and size. Exosomes are spherical lipid-bilayer vesicles with a diameter of about 40 to 100 nm. Exosomes originate from intracellular endosomal compartments, while microvesicles originated directly from a cell's plasma membrane and apoptotic bodies originate from cells undergoing apoptosis and are released via outward blebbing and fragmentation of the plasma membrane. Specifically, exosomes have garnered great attention since they display great potential as both biomarkers and carriers of therapeutic molecules.
Collapse
Affiliation(s)
- Eleni Papakonstantinou
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
- University Research Institute of Maternal and Child Health & Precision Medicine, and UNESCO Chair on Adolescent Health Care, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Konstantina Dragoumani
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
| | - Thanasis Mitsis
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
| | - George P Chrousos
- University Research Institute of Maternal and Child Health & Precision Medicine, and UNESCO Chair on Adolescent Health Care, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Dimitrios Vlachakis
- Laboratory of Genetics, Department of Biotechnology, School of Applied Biology and Biotechnology, Agricultural University of Athens, Athens, Greece
- University Research Institute of Maternal and Child Health & Precision Medicine, and UNESCO Chair on Adolescent Health Care, National and Kapodistrian University of Athens, "Aghia Sophia" Children's Hospital, Athens, Greece
- School of Informatics, Faculty of Natural & Mathematical Sciences, King's College London, London, U.K
| |
Collapse
|
12
|
Xu YR, Zhao J, Huang HY, Lin YCD, Lee TY, Huang HD, Yang Y, Wang YF. Recent insights into breast milk microRNA: their role as functional regulators. Front Nutr 2024; 11:1366435. [PMID: 38689935 PMCID: PMC11058965 DOI: 10.3389/fnut.2024.1366435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/02/2024] [Indexed: 05/02/2024] Open
Abstract
Breast milk (BM) is a primary biofluid that plays a crucial role in infant development and the regulation of the immune system. As a class of rich biomolecules in BM, microRNAs (miRNAs) are regarded as active factors contributing to infant growth and development. Surprisingly, these molecules exhibit resilience in harsh conditions, providing an opportunity for infants to absorb them. In addition, many studies have shown that miRNAs in breast milk, when absorbed into the gastrointestinal system, can act as a class of functional regulators to effectively regulate gene expression. Understanding the absorption pattern of BM miRNA may facilitate the creation of formula with a more optimal miRNA balance and pave the way for novel drug delivery techniques. In this review, we initially present evidence of BM miRNA absorption. Subsequently, we compile studies that integrate both in vivo and in vitro findings to illustrate the bioavailability and biodistribution of BM miRNAs post-absorption. In addition, we evaluate the strengths and weaknesses of previous studies and discuss potential variables contributing to discrepancies in their outcomes. This literature review indicates that miRNAs can be absorbed and act as regulatory agents.
Collapse
Affiliation(s)
- Yi-Ran Xu
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Jinglu Zhao
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Hsi-Yuan Huang
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Yang-Chi-Dung Lin
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Tzong-Yi Lee
- Institute of Bioinformatics and Systems Biology and Center for Intelligent Drug Systems and Smart Bio-devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Hsien-Da Huang
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| | - Yi Yang
- Department of Nephrology, Center for Regeneration and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
- Zhejiang-Denmark Joint Laboratory of Regeneration and Aging Medicine, Yiwu, China
| | - Yong-Fei Wang
- Warshel Institute for Computational Biology, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, China
| |
Collapse
|
13
|
Naik NC, Holzhausen EA, Chalifour BN, Coffman MM, Lurmann F, Goran MI, Bode L, Alderete TL. Air pollution exposure may impact the composition of human milk oligosaccharides. Sci Rep 2024; 14:6730. [PMID: 38509153 PMCID: PMC10954706 DOI: 10.1038/s41598-024-57158-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 03/14/2024] [Indexed: 03/22/2024] Open
Abstract
Human milk oligosaccharides (HMOs) impact neonate immunity and health outcomes. However, the environmental factors influencing HMO composition remain understudied. This study examined the associations between ambient air pollutant (AAP) exposure and HMOs at 1-month postpartum. Human milk samples were collected at 1-month postpartum (n = 185). AAP (PM2.5, PM10, NO2) exposure included the 9-month pregnancy period through 1-month postpartum. Associations between AAP with (1) HMO diversity, (2) the sum of sialylated and fucosylated HMOs, (3) 6 a priori HMOs linked with infant health, and (4) all HMOs were examined using multivariable linear regression and principal component analysis (PCA). Exposure to AAP was associated with lower HMO diversity. PM2.5 and PM10 exposure was positively associated with the HMO 3-fucosyllactose (3FL); PM2.5 exposure was positively associated with the sum of total HMOs, sum of fucosylated HMOs, and the HMO 2'-fucosyllactose (2'FL). PCA indicated the PM2.5, PM10, and NO2 exposures were associated with HMO profiles. Individual models indicated that AAP exposure was associated with five additional HMOs (LNFP I, LNFP II, DFLNT, LNH). This is the first study to demonstrate associations between AAP and breast milk HMOs. Future longitudinal studies will help determine the long-term impact of AAP on human milk composition.
Collapse
Affiliation(s)
- Noopur C Naik
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
- Cleveland Clinic Lerner College of Medicine at Case Western Reserve University College of Medicine, Cleveland, OH, USA
| | | | - Bridget N Chalifour
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Maria M Coffman
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | | | - Michael I Goran
- Department of Pediatrics, Children's Hospital of Los Angeles, Los Angeles, CA, USA
| | - Lars Bode
- Department of Pediatrics, Larson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MOMI CORE), Human Milk Institute (HMI), University of California, San Diego, La Jolla, CA, USA
| | - Tanya L Alderete
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA.
| |
Collapse
|
14
|
Liu K, Chen Z, Hu W, He B, Xu D, Guo Y, Wang H. Intrauterine developmental origin, programming mechanism, and prevention strategy of fetal-originated hypercholesterolemia. Obes Rev 2024; 25:e13672. [PMID: 38069529 DOI: 10.1111/obr.13672] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 10/30/2023] [Accepted: 11/05/2023] [Indexed: 02/28/2024]
Abstract
There is increasing evidence that hypercholesterolemia has an intrauterine developmental origin. However, the pathogenesis of fetal-originated is still lacking in a theoretical system, which makes its clinical early prevention and treatment difficult. It has been found that an adverse environment during pregnancy (e.g., xenobiotic exposure) may lead to changes in fetal blood cholesterol levels through changing maternal cholesterol metabolic function and/or placental cholesterol transport function and may also directly affect the liver cholesterol metabolic function of the offspring in utero and continue after birth. Adverse environmental conditions during pregnancy may also raise maternal glucocorticoid levels and promote the placental glucocorticoid barrier opening, leading to fetal overexposure to maternal glucocorticoids. Intrauterine high-glucocorticoid exposure can alter the liver cholesterol metabolism of offspring, resulting in an increased susceptibility to hypercholesterolemia after birth. Abnormal epigenetic modifications are involved in the intrauterine programming mechanism of fetal-originated hypercholesterolemia. Some interventions targeted at pregnant mothers or offspring in early life have been proposed to effectively prevent and treat the development of fetal-originated hypercholesterolemia. In this paper, the recent research progress on fetal-originated hypercholesterolemia was reviewed, with emphasis on intrauterine maternal glucocorticoid programming mechanisms, in order to provide a theoretical basis for its early clinical warning, prevention, and treatment.
Collapse
Affiliation(s)
- Kexin Liu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, China
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Ze Chen
- Department of Cardiology, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Wen Hu
- Department of Pharmacy, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Bo He
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| | - Dan Xu
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| | - Yu Guo
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| | - Hui Wang
- Department of Pharmacology, Wuhan University School of Basic Medical Sciences, Wuhan, China
- Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan, China
| |
Collapse
|
15
|
Mahala S, Kumar A, Pandey HO, Saxena S, Khanna S, Kumar M, Kumar D, De UK, Pandey AK, Dutt T. Milk exosomal microRNA profiling identified miR-375 and miR-199-5p for regulation of immune response during subclinical mastitis of crossbred cattle. Mol Biol Rep 2024; 51:59. [PMID: 38165514 DOI: 10.1007/s11033-023-09070-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 11/10/2023] [Indexed: 01/03/2024]
Abstract
BACKGROUND The dairy industry has experienced significant economic losses as a result of mastitis, an inflammatory disease of cows, including both subclinical and clinical cases. Milk exosome microRNAs have gained attention due to their stable and selective wrapping nature, offering potential for the prognosis and diagnosis of bovine mastitis, the most common pathological condition of the mammary gland. METHODS AND RESULTS In the present investigation, the microRNA profile of milk exosomes was explored using high-throughput small RNA sequencing data in sub-clinical mastitic and healthy crossbred Vrindavani cattle. In both groups, 349 microRNAs were identified, with 238 (68.19%) microRNAs co-expressed; however, 35 and 76 distinct microRNAs were found in subclinical mastitic and healthy cattle, respectively. Differential expression analysis revealed 11 microRNAs upregulated, and 18 microRNAs were downregulated in sub-clinical mastitic cattle. The functional annotation of the target genes of differentially expressed known and novel microRNAs including bta-miR-375, bta-miR-199-5p and bta-miR-12030 reveals their involvement in the regulation of immune response and inflammatory mechanisms and could be involved in development of mastitis. CONCLUSIONS The analysis of milk exosomal miRNAs cargos hold great promise as an approach to study the underlying molecular mechanisms associated with mastitis in high milk producing dairy cattle. Concurrently, the significantly downregulated miR-375 may upregulate key target genes, including CTLA4, IHH, IRF1, and IL7R. These genes are negative regulators of immune response pathways, which could be associated with impaired inflammatory mechanisms in mammary cells. According to the findings, bta-miR-375 could be a promising biomarker for the development of mastitis in dairy cattle.
Collapse
Affiliation(s)
- Sudarshan Mahala
- Animal Genetics Division, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, 243122, India
| | - Amit Kumar
- Animal Genetics Division, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, 243122, India.
| | - Hari Om Pandey
- Livestock Production and Management Section, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, 243122, India
| | - Shikha Saxena
- Animal Genetics Division, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, 243122, India
| | - Shivani Khanna
- Animal Genetics Division, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, 243122, India
| | - Manoj Kumar
- Animal Genetics Division, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, 243122, India
| | - Deepak Kumar
- Veterinary Biotechnology Division, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, 243122, India
| | - Ujjwal Kumar De
- Medicine Division, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, 243122, India
| | - Ashwni Kumar Pandey
- Animal Genetics Division, Indian Council of Agricultural Research (ICAR)-Indian Veterinary Research Institute, Bareilly, Uttar Pradesh, 243122, India
| | - Triveni Dutt
- Livestock Production and Management Section, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, 243122, India
| |
Collapse
|
16
|
Moholdt T, Stanford KI. Exercised breastmilk: a kick-start to prevent childhood obesity? Trends Endocrinol Metab 2024; 35:23-30. [PMID: 37735048 PMCID: PMC11005327 DOI: 10.1016/j.tem.2023.08.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/23/2023]
Abstract
Exercise has systemic health benefits through effects on multiple tissues, with intertissue communication. Recent studies indicate that exercise may improve breastmilk composition and thereby reduce the intergenerational transmission of obesity. Even if breastmilk is considered optimal infant nutrition, there is evidence for variations in its composition between mothers who are normal weight, those with obesity, and those who are physically active. Nutrition early in life is important for later-life susceptibility to obesity and other metabolic diseases, and maternal exercise may provide protection against the development of metabolic disease. Here we summarize recent research on the influence of maternal obesity on breastmilk composition and discuss the potential role of exercise-induced adaptations to breastmilk as a kick-start to prevent childhood obesity.
Collapse
Affiliation(s)
- Trine Moholdt
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology, Trondheim, Norway; Department of Gynaecology and Obstetrics, St. Olav's Hospital, Trondheim, Norway.
| | - Kristin I Stanford
- Dorothy M. Davis Heart and Lung Research Institute, Department of Physiology and Cell Biology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| |
Collapse
|
17
|
Vahkal B, Altosaar I, Tremblay E, Gagné D, Hüttman N, Minic Z, Côté M, Blais A, Beaulieu J, Ferretti E. Gestational age at birth influences protein and RNA content in human milk extracellular vesicles. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e128. [PMID: 38938674 PMCID: PMC11080785 DOI: 10.1002/jex2.128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 10/26/2023] [Accepted: 11/02/2023] [Indexed: 06/29/2024]
Abstract
Human milk extracellular vesicles (HM EVs) are proposed to protect against disease development in infants. This protection could in part be facilitated by the bioactive EV cargo of proteins and RNA. Notably, mothers birth infants of different gestational ages with unique needs, wherein the EV cargo of HM may diverge. We collected HM from lactating mothers within two weeks of a term or preterm birth. Following purification of EVs, proteins and mRNA were extracted for proteomics and sequencing analyses, respectively. Over 2000 protein groups were identified, and over 8000 genes were quantified. The total number of proteins and mRNA did not differ significantly between the two conditions, while functional bioinformatics of differentially expressed cargo indicated enrichment in immunoregulatory cargo for preterm HM EVs. In term HM EVs, significantly upregulated cargo was enriched in metabolism-related functions. Based on gene expression signatures from HM-contained single cell sequencing data, we proposed that a larger portion of preterm HM EVs are secreted by immune cells, whereas term HM EVs contain more signatures of lactocyte epithelial cells. Proposed differences in EV cargo could indicate variation in mother's milk based on infants' gestational age and provide basis for further functional characterisation.
Collapse
Affiliation(s)
- Brett Vahkal
- Department of Biochemistry, Microbiology and ImmunologyUniversity of OttawaOttawaCanada
- uOttawa Centre for Infection, Immunity, and InflammationOttawaCanada
- Ottawa Institute of Systems BiologyOttawaCanada
| | - Illimar Altosaar
- Department of Biochemistry, Microbiology and ImmunologyUniversity of OttawaOttawaCanada
| | - Eric Tremblay
- Department of Immunology and Cell BiologyUniversité de SherbrookeSherbrookeCanada
| | - David Gagné
- Department of Immunology and Cell BiologyUniversité de SherbrookeSherbrookeCanada
| | - Nico Hüttman
- Faculty of Science, John L. Holmes Mass Spectrometry FacilityUniversity of OttawaOttawaCanada
| | - Zoran Minic
- Faculty of Science, John L. Holmes Mass Spectrometry FacilityUniversity of OttawaOttawaCanada
| | - Marceline Côté
- Department of Biochemistry, Microbiology and ImmunologyUniversity of OttawaOttawaCanada
- uOttawa Centre for Infection, Immunity, and InflammationOttawaCanada
- Ottawa Institute of Systems BiologyOttawaCanada
| | - Alexandre Blais
- Department of Biochemistry, Microbiology and ImmunologyUniversity of OttawaOttawaCanada
- uOttawa Centre for Infection, Immunity, and InflammationOttawaCanada
- Ottawa Institute of Systems BiologyOttawaCanada
- Brain and Mind InstituteUniversity of OttawaOttawaCanada
- Éric Poulin Centre for Neuromuscular DiseaseOttawaCanada
| | | | - Emanuela Ferretti
- Department of Pediatrics, Division of NeonatologyChildren's Hospital of Eastern OntarioOttawaCanada
| |
Collapse
|
18
|
Yeruva L, Mulakala BK, Rajasundaram D, Gonzalez S, Cabrera-Rubio R, Martínez-Costa C, Collado MC. Human milk miRNAs associate to maternal dietary nutrients, milk microbiota, infant gut microbiota and growth. Clin Nutr 2023; 42:2528-2539. [PMID: 37931372 DOI: 10.1016/j.clnu.2023.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 10/06/2023] [Accepted: 10/10/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND Maternal diet influences the milk composition, yet little information is available on the impact of maternal diet on milk miRNAs expression. Further, the association of human milk miRNAs to maternal diet and milk microbiota is not explored. In addition, the role of milk miRNAs on the infant gut microbiota, infant growth and development has not been investigated. METHODS Milk samples were collected from 60 healthy lactating women at ≤15d post-partum, HTG transcriptome assay was performed to examine milk miRNA profile. Maternal clinical and dietary clusters information were available and infant anthropometric measures were followed up to one year of age. Milk and infant microbiota were analyzed by 16S rRNA gene sequencing and integrative multi-omics data analysis was performed to identify potential association between microRNA, maternal dietary nutrients and microbiota. RESULTS Discriminant analysis revealed that the milk miRNAs were clustered into groups according to the maternal protein source. Interestingly, 31 miRNAs were differentially expressed (P adj < 0.05) between maternal dietary clusters (Cluster 1: enriched in plant protein and fibers and Cluster 2: enriched in animal protein), with 30 miRNAs downregulated in the plant protein group relative to animal protein group. Pathway analysis revealed that the top enriched pathways (P adj < 0.01) were involved in cell growth and proliferation processes. Furthermore, significant features contributing to the clustering were associated with maternal dietary nutrients and milk microbiota (r > 0.70). Further, miR-378 and 320 family miRNAs involved in adipogenesis were positively correlated to the infant BMI-z-scores, weight, and weight for length-z-scores at 6 months of age. CONCLUSIONS Maternal dietary source impacts the milk miRNA expression profile. Further, miRNAs were associated with maternal dietary nutrients, milk microbiota and to the infant gut microbiota and infant growth and development. CLINICAL TRIAL The study is registered in ClinicalTrials.gov. The identification number is NCT03552939.
Collapse
Affiliation(s)
- Laxmi Yeruva
- Microbiome and Metabolism Research Unit, USDA-ARS, SEA, Little Rock, AR, USA; Arkansas Children's Nutrition Center, Little Rock, AR, USA.
| | - Bharat Kumar Mulakala
- Microbiome and Metabolism Research Unit, USDA-ARS, SEA, Little Rock, AR, USA; Arkansas Children's Nutrition Center, Little Rock, AR, USA; Texas A&M AgriLife Institute for Advancing Health Through Agriculture, TX, USA
| | | | - Sonia Gonzalez
- Department of Functional Biology, Faculty of Medicine, University of Oviedo, Oviedo, Spain; Diet, Microbiota and Health Group, Instituto de Investigación Sanitaria del Principado de Asturias (DIMISA, ISPA), Oviedo, Spain
| | - Raul Cabrera-Rubio
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Valencia, Spain
| | | | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-National Research Council (IATA-CSIC), Valencia, Spain.
| |
Collapse
|
19
|
Słyk-Gulewska P, Kondracka A, Kwaśniewska A. MicroRNA as a new bioactive component in breast milk. Noncoding RNA Res 2023; 8:520-526. [PMID: 37520770 PMCID: PMC10371784 DOI: 10.1016/j.ncrna.2023.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 06/24/2023] [Accepted: 06/25/2023] [Indexed: 08/01/2023] Open
Abstract
Breast milk is a complex and multifaceted fluid that plays a critical role in the development of infants. It is composed of water, carbohydrates, fats, proteins, vitamins, and minerals, as well as numerous bioactive compounds such as hormones, oligosaccharides, and immune proteins. Additionally, breast milk contains microRNAs, which have been found to regulate gene expression and impact various aspects of infant development. This text provides an overview of the components of human breast milk and their importance in infant development, with a focus on microRNAs. MicroRNAs are short RNA sequences that regulate gene expression posttranscriptionally, and they play an important role in shaping the mechanisms of immunity, protecting against oxidative stress, and promoting thermogenesis. The composition of breast milk can vary in the same mother between different feedings, as it changes in response to various factors such as the infant's age, feeding frequency and duration, time of day, and maternal health status. Despite the variations in breast milk composition, it still provides complete nutrition for the infant. The unique microRNA profiles in breast milk and how they are affected by various factors can have significant implications for disease prevention and treatment. Further research is needed to better understand the functions of individual microRNA molecules and their potential therapeutic applications.
Collapse
|
20
|
Freiría-Martínez L, Iglesias-Martínez-Almeida M, Rodríguez-Jamardo C, Rivera-Baltanás T, Comís-Tuche M, Rodrígues-Amorím D, Fernández-Palleiro P, Blanco-Formoso M, Diz-Chaves Y, González-Freiria N, Suárez-Albo M, Martín-Forero-Maestre M, Durán Fernández-Feijoo C, Fernández-Lorenzo JR, Concheiro Guisán A, Olivares JM, Spuch C. Human Breast Milk microRNAs, Potential Players in the Regulation of Nervous System. Nutrients 2023; 15:3284. [PMID: 37513702 PMCID: PMC10384760 DOI: 10.3390/nu15143284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/12/2023] [Accepted: 07/15/2023] [Indexed: 07/30/2023] Open
Abstract
Human milk is the biological fluid with the highest exosome amount and is rich in microRNAs (miRNAs). These are key regulators of gene expression networks in both normal physiologic and disease contexts, miRNAs can influence many biological processes and have also shown promise as biomarkers for disease. One of the key aspects in the regeneration of the nervous system is that there are practically no molecules that can be used as potential drugs. In the first weeks of lactation, we know that human breast milk must contain the mechanisms to transmit molecular and biological information for brain development. For this reason, our objective is to identify new modulators of the nervous system that can be used to investigate neurodevelopmental functions based on miRNAs. To do this, we collected human breast milk samples according to the time of delivery and milk states: mature milk and colostrum at term; moderate and very preterm mature milk and colostrum; and late preterm mature milk. We extracted exosomes and miRNAs and realized the miRNA functional assays and target prediction. Our results demonstrate that miRNAs are abundant in human milk and likely play significant roles in neurodevelopment and normal function. We found 132 different miRNAs were identified across all samples. Sixty-nine miRNAs had significant differential expression after paired group comparison. These miRNAs are implicated in gene regulation of dopaminergic/glutamatergic synapses and neurotransmitter secretion and are related to the biological process that regulates neuron projection morphogenesis and synaptic vesicle transport. We observed differences according to the delivery time and with less clarity according to the milk type. Our data demonstrate that miRNAs are abundant in human milk and likely play significant roles in neurodevelopment and normal function.
Collapse
Affiliation(s)
- Luis Freiría-Martínez
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO (Servizo Galego de Saúde-Universidade de Vigo), 36312 Vigo, Spain
- Department of Functional Biology and Health Sciences, Campus Lagoas Marcosende, Universidade de Vigo, 36310 Vigo, Spain
| | - Marta Iglesias-Martínez-Almeida
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO (Servizo Galego de Saúde-Universidade de Vigo), 36312 Vigo, Spain
- Department of Functional Biology and Health Sciences, Campus Lagoas Marcosende, Universidade de Vigo, 36310 Vigo, Spain
| | - Cynthia Rodríguez-Jamardo
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO (Servizo Galego de Saúde-Universidade de Vigo), 36312 Vigo, Spain
- Department of Functional Biology and Health Sciences, Campus Lagoas Marcosende, Universidade de Vigo, 36310 Vigo, Spain
| | - Tania Rivera-Baltanás
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO (Servizo Galego de Saúde-Universidade de Vigo), 36312 Vigo, Spain
| | - María Comís-Tuche
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO (Servizo Galego de Saúde-Universidade de Vigo), 36312 Vigo, Spain
- Department of Functional Biology and Health Sciences, Campus Lagoas Marcosende, Universidade de Vigo, 36310 Vigo, Spain
| | - Daniela Rodrígues-Amorím
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO (Servizo Galego de Saúde-Universidade de Vigo), 36312 Vigo, Spain
- Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Patricia Fernández-Palleiro
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO (Servizo Galego de Saúde-Universidade de Vigo), 36312 Vigo, Spain
| | - María Blanco-Formoso
- Department of Physical Chemistry, Singular Center for Biomedical Research (CINBIO), Universidade de Vigo, 36310 Vigo, Spain
| | - Yolanda Diz-Chaves
- Laboratory of Endocrinology, Singular Center for Biomedical Research (CINBIO), Universidade de Vigo, 36310 Vigo, Spain
| | | | - María Suárez-Albo
- Neonatal Intensive Care Unit, Alvaro Cunqueiro Hospital, 36312 Vigo, Spain
| | | | | | | | | | - Jose Manuel Olivares
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO (Servizo Galego de Saúde-Universidade de Vigo), 36312 Vigo, Spain
- CIBERSAM (Network Biomedical Research Center on Mental Health), 28029 Madrid, Spain
| | - Carlos Spuch
- Translational Neuroscience Group, Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS-UVIGO (Servizo Galego de Saúde-Universidade de Vigo), 36312 Vigo, Spain
- CIBERSAM (Network Biomedical Research Center on Mental Health), 28029 Madrid, Spain
| |
Collapse
|
21
|
Bakshi S, Paswan VK, Yadav SP, Bhinchhar BK, Kharkwal S, Rose H, Kanetkar P, Kumar V, Al-Zamani ZAS, Bunkar DS. A comprehensive review on infant formula: nutritional and functional constituents, recent trends in processing and its impact on infants' gut microbiota. Front Nutr 2023; 10:1194679. [PMID: 37415910 PMCID: PMC10320619 DOI: 10.3389/fnut.2023.1194679] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/30/2023] [Indexed: 07/08/2023] Open
Abstract
Human milk is considered the most valuable form of nutrition for infants for their growth, development and function. So far, there are still some cases where feeding human milk is not feasible. As a result, the market for infant formula is widely increasing, and formula feeding become an alternative or substitute for breastfeeding. The nutritional value of the formula can be improved by adding functional bioactive compounds like probiotics, prebiotics, human milk oligosaccharides, vitamins, minerals, taurine, inositol, osteopontin, lactoferrin, gangliosides, carnitine etc. For processing of infant formula, diverse thermal and non-thermal technologies have been employed. Infant formula can be either in powdered form, which requires reconstitution with water or in ready-to-feed liquid form, among which powder form is readily available, shelf-stable and vastly marketed. Infants' gut microbiota is a complex ecosystem and the nutrient composition of infant formula is recognized to have a lasting effect on it. Likewise, the gut microbiota establishment closely parallels with host immune development and growth. Therefore, it must be contemplated as an important factor for consideration while developing formulas. In this review, we have focused on the formulation and manufacturing of safe and nutritious infant formula equivalent to human milk or aligning with the infant's needs and its ultimate impact on infants' gut microbiota.
Collapse
Affiliation(s)
- Shiva Bakshi
- Department of Dairy Science and Food Technology, Institute of Agricultural Sciences, Banaras Hindu University, Varanasi, India
| | - Vinod Kumar Paswan
- Department of Dairy Science and Food Technology, Institute of Agricultural Sciences, Banaras Hindu University, Varanasi, India
| | - Satya Prakash Yadav
- Department of Dairy Science and Food Technology, Institute of Agricultural Sciences, Banaras Hindu University, Varanasi, India
| | - Basant Kumar Bhinchhar
- Department of Livestock Production Management, Sri Karan Narendra Agriculture University, Jobner, India
| | - Sheela Kharkwal
- Department of Agriculture Economics, Sri Karan Narendra Agriculture University, Jobner, India
| | - Hency Rose
- Division of Dairy Technology, ICAR—National Dairy Research Institute, Karnal, India
| | - Prajasattak Kanetkar
- Department of Dairy Science and Food Technology, Institute of Agricultural Sciences, Banaras Hindu University, Varanasi, India
| | - Vishal Kumar
- Department of Dairy Science and Food Technology, Institute of Agricultural Sciences, Banaras Hindu University, Varanasi, India
| | - Zakarya Ali Saleh Al-Zamani
- Department of Dairy Science and Food Technology, Institute of Agricultural Sciences, Banaras Hindu University, Varanasi, India
- Department of Food Technology and Science, Faculty of Agriculture and Veterinary Medicine, Ibb University, Ibb, Yemen
| | - Durga Shankar Bunkar
- Department of Dairy Science and Food Technology, Institute of Agricultural Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
22
|
Torrez Lamberti MF, Parker LA, Gonzalez CF, Lorca GL. Pasteurization of human milk affects the miRNA cargo of EVs decreasing its immunomodulatory activity. Sci Rep 2023; 13:10057. [PMID: 37344543 DOI: 10.1038/s41598-023-37310-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Accepted: 06/20/2023] [Indexed: 06/23/2023] Open
Abstract
In this report, we evaluated the effect of the pasteurization (P) process of mother's own milk (MOM) on the miRNA content of extracellular vesicles (EVs) and its impact on innate immune responses. Differences in size or particle number were not observed upon pasteurization of MOM (PMOM). However, significant differences were observed in the EV membrane marker CD63 and miRNA profiles. miRNA sequencing identified 33 differentially enriched miRNAs between MOMEV and PMOMEV. These changes correlated with significant decreases in the ability of PMOMEV to modulate IL-8 secretion in intestinal Caco2 cells where only MOMEV were able to decrease IL-8 secretion in presence of TNFα. While EVs from MOMEV and PMOMEV were both able to induce a tolerogenic M2-like phenotype in THP-1 macrophages, a significant decrease in the transcript levels of IL-10 and RNA sensing genes was observed with PMOMEV. Together, our data indicates that pasteurization of MOM impacts the integrity and functionality of MOMEV, decreasing its EVs-mediated immunomodulatory activity. This data provides biomarkers that may be utilized during the optimization of milk processing to preserve its bioactivity.
Collapse
Affiliation(s)
- Monica F Torrez Lamberti
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, USA
| | | | - Claudio F Gonzalez
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, USA
| | - Graciela L Lorca
- Department of Microbiology and Cell Science, Genetics Institute, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, USA.
| |
Collapse
|
23
|
Ryoo E. Recent advances in epigenetic mechanisms, diagnosis, and treatment of pediatric gastrointestinal allergic disorders. Clin Exp Pediatr 2023; 66:250-251. [PMID: 37211329 PMCID: PMC10248319 DOI: 10.3345/cep.2023.00136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 04/10/2023] [Accepted: 04/20/2023] [Indexed: 05/23/2023] Open
Affiliation(s)
- Eell Ryoo
- Department of Pediatrics, Gachon University Gil Medical Center, Incheon, Korea
| |
Collapse
|
24
|
Du Y, Gao Y, Hu M, Hou J, Yang L, Wang X, Du W, Liu J, Xu Q. Colonization and development of the gut microbiome in calves. J Anim Sci Biotechnol 2023; 14:46. [PMID: 37031166 PMCID: PMC10082981 DOI: 10.1186/s40104-023-00856-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 02/16/2023] [Indexed: 04/10/2023] Open
Abstract
Colonization and development of the gut microbiome are crucial for the growth and health of calves. In this review, we summarized the colonization, beneficial nutrition, immune function of gut microbiota, function of the gut barrier, and the evolution of core microbiota in the gut of calves of different ages. Homeostasis of gut microbiome is beneficial for nutritional and immune system development of calves. Disruption of the gut microbiome leads to digestive diseases in calves, such as diarrhea and intestinal inflammation. Microbiota already exists in the gut of calf fetuses, and the colonization of microbiota continues to change dynamically under the influence of various factors, which include probiotics, diet, age, and genotype. Colonization depends on the interaction between the gut microbiota and the immune system of calves. The abundance and diversity of these commensal microbiota stabilize and play a critical role in the health of calves.
Collapse
Affiliation(s)
- Yufeng Du
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Ya Gao
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Mingyang Hu
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jinxiu Hou
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Linhai Yang
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xianghuang Wang
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Wenjuan Du
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Jianxin Liu
- MoE Key Laboratory of Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Qingbiao Xu
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
- MoE Key Laboratory of Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
25
|
Neville MC, Demerath EW, Hahn-Holbrook J, Hovey RC, Martin-Carli J, McGuire MA, Newton ER, Rasmussen KM, Rudolph MC, Raiten DJ. Parental factors that impact the ecology of human mammary development, milk secretion, and milk composition-a report from "Breastmilk Ecology: Genesis of Infant Nutrition (BEGIN)" Working Group 1. Am J Clin Nutr 2023; 117 Suppl 1:S11-S27. [PMID: 37173058 PMCID: PMC10232333 DOI: 10.1016/j.ajcnut.2022.11.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 11/22/2022] [Accepted: 11/29/2022] [Indexed: 05/15/2023] Open
Abstract
The goal of Working Group 1 in the Breastmilk Ecology: Genesis of Infant Nutrition (BEGIN) Project was to outline factors influencing biological processes governing human milk secretion and to evaluate our current knowledge of these processes. Many factors regulate mammary gland development in utero, during puberty, in pregnancy, through secretory activation, and at weaning. These factors include breast anatomy, breast vasculature, diet, and the lactating parent's hormonal milieu including estrogen, progesterone, placental lactogen, cortisol, prolactin, and growth hormone. We examine the effects of time of day and postpartum interval on milk secretion, along with the role and mechanisms of lactating parent-infant interactions on milk secretion and bonding, with particular attention to the actions of oxytocin on the mammary gland and the pleasure systems in the brain. We then consider the potential effects of clinical conditions including infection, pre-eclampsia, preterm birth, cardiovascular health, inflammatory states, mastitis, and particularly, gestational diabetes and obesity. Although we know a great deal about the transporter systems by which zinc and calcium pass from the blood stream into milk, the interactions and cellular localization of transporters that carry substrates such as glucose, amino acids, copper, and the many other trace metals present in human milk across plasma and intracellular membranes require more research. We pose the question of how cultured mammary alveolar cells and animal models can help answer lingering questions about the mechanisms and regulation of human milk secretion. We raise questions about the role of the lactating parent and the infant microbiome and the immune system during breast development, secretion of immune molecules into milk, and protection of the breast from pathogens. Finally, we consider the effect of medications, recreational and illicit drugs, pesticides, and endocrine-disrupting chemicals on milk secretion and composition, emphasizing that this area needs much more research attention.
Collapse
Affiliation(s)
- Margaret C Neville
- Department of Obstetrics and Gynecology, University of Colorado, Aurora, CO, USA.
| | - Ellen W Demerath
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN, United States
| | - Jennifer Hahn-Holbrook
- Department of Psychological Sciences, University of California Merced, Merced, CA, United States
| | - Russell C Hovey
- Department of Animal Science, University of California Davis, Davis, CA, United States
| | - Jayne Martin-Carli
- Department of Pediatrics, University of Colorado, Aurora, CO, United States
| | - Mark A McGuire
- Idaho Agricultural Experiment Station, University of Idaho, Moscow, ID, United States
| | - Edward R Newton
- Department of Obstetrics and Gynecology, Brody School of Medicine, East Carolina University, Greenville, NC, United States
| | - Kathleen M Rasmussen
- Nancy Schlegel Meinig Professor of Maternal and Child Nutrition, Division of Nutritional Sciences, Cornell University, Ithaca, NY, United States
| | - Michael C Rudolph
- The University of Oklahoma Health Science Center, Oklahoma City, OK, United States
| | - Daniel J Raiten
- Pediatric Growth and Nutrition Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
26
|
Ahlberg E, Al-Kaabawi A, Thune R, Simpson MR, Pedersen SA, Cione E, Jenmalm MC, Tingö L. Breast milk microRNAs: Potential players in oral tolerance development. Front Immunol 2023; 14:1154211. [PMID: 36999032 PMCID: PMC10045994 DOI: 10.3389/fimmu.2023.1154211] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 02/28/2023] [Indexed: 03/16/2023] Open
Abstract
Breast milk is an essential source of nutrition and hydration for the infant. In addition, this highly complex biological fluid contains numerous immunologically active factors such as microorganisms, immunoglobulins, cytokines and microRNAs (miRNAs). Here, we set out to predict the function of the top 10 expressed miRNAs in human breast milk, focusing on their relevance in oral tolerance development and allergy prevention in the infant. The top expressed miRNAs in human breast milk were identified on basis of previous peer-reviewed studies gathered from a recent systematic review and an updated literature search. The miRNAs with the highest expression levels in each study were used to identify the 10 most common miRNAs or miRNA families across studies and these were selected for subsequent target prediction. The predictions were performed using TargetScan in combination with the Database for Annotation, Visualization and Integrated Discovery. The ten top expressed miRNAs were: let-7-5p family, miR-148a-3p, miR-30-5p family, miR-200a-3p + miR-141-3p, miR-22-3p, miR-181-5p family, miR-146b-5p, miR-378a-3p, miR-29-3p family, miR-200b/c-3p and miR-429-3p. The target prediction identified 3,588 potential target genes and 127 Kyoto Encyclopedia of Genes and Genomes pathways; several connected to the immune system, including TGF-b and T cell receptor signaling and T-helper cell differentiation. This review highlights the role of breast milk miRNAs and their potential contribution to infant immune maturation. Indeed, breast milk miRNAs seem to be involved in several pathways that influence oral tolerance development.
Collapse
Affiliation(s)
- Emelie Ahlberg
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Ahmed Al-Kaabawi
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Rebecka Thune
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Melanie Rae Simpson
- Department of Public Health and Nursing, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Sindre Andre Pedersen
- Library Section for Research Support, Data and Analysis, NTNU University Library, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Erika Cione
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Cosenza, Italy
| | - Maria Christina Jenmalm
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Lina Tingö
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
- Nutrition-Gut-Brain Interactions Research Centre, School of Medical Sciences, Örebro University, Örebro, Sweden
- Food and Health Programme, Örebro University, Örebro, Sweden
- *Correspondence: Lina Tingö,
| |
Collapse
|
27
|
Hue DT, Petrovski K, Chen T, Williams JL, Bottema CDK. Analysis of immune-related microRNAs in cows and newborn calves. J Dairy Sci 2023; 106:2866-2878. [PMID: 36870833 DOI: 10.3168/jds.2022-22398] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 11/13/2022] [Indexed: 03/06/2023]
Abstract
Bovine colostrum contains a high concentration of immune-related microRNAs (miRNAs) that are packaged in exosomes and are very stable. In this study, 5 immune-related miRNAs (miR-142-5p, miR-150, miR-155, miR-181a, and miR-223) were quantified in dam blood, colostrum, and calf blood using reverse transcription quantitative PCR. Their levels in calf blood after colostrum ingestion were investigated to assess whether miRNAs are transferred from the dam to newborn calves. Three groups of Holstein-Friesian bull calves were bottle-fed 2 L of colostrum or milk from different sources twice per day. The group A calves received colostrum from their own dam and the group B calves were fed foster dam colostrum. Each pair of group A and group B calves were fed identical colostrum from the same milking of the corresponding group A dam for 3 d and then bulk tank milk for 7 d after birth. Group C calves were fed only 2L of "pooled colostrum" from multiple dams d 0 to 4 postpartum, and then fed bulk tank milk thereafter for 7 d after birth. The groups were fed colostrum from different sources and different amounts to assess possible miRNA absorption from the colostrum. All miRNAs were at the highest level in colostrum at d 0 and then decreased rapidly after d 1. The level of miR-150 had the largest decrease from 489 × 106 copies/µL (d 0) to 78 × 106 copies/µL (d 1). MicroRNA-223 and miR-155 were the most abundant in both colostrum and milk. Dam colostrum had significantly higher levels of miR-142-5p, miR-155, and miR-181a than the bulk tank milk. However, only the miR-155 concentration was significantly higher in the dam colostrum than in the pooled colostrum. The concentrations of miRNAs in the colostrum were less than in the cow blood (100- to 1,000-fold less). There was no significant correlation between the level of miRNAs in the dam blood and their colostrum, suggesting that miRNA is synthesized locally by the mammary gland rather than being transferred from the blood. MicroRNA-223 had the highest level in both calf and cow blood compared with the other 4 immune-related miRNAs. Calves were born with high levels of immune-related miRNAs in their blood, and there were no significant differences in miRNA levels between the 3 calf groups at birth or after they were fed different colostrum. This suggests that these miRNAs were not transferred from the colostrum to the newborn calves.
Collapse
Affiliation(s)
- Do T Hue
- Davies Livestock Research Centre, School of Animal and Veterinary Sciences, University of Adelaide, Roseworthy Campus, SA 5371, Australia; Faculty of Animal Science, Vietnam National University of Agriculture, Trau Quy, Gia Lam, Hanoi, 12406 Vietnam.
| | - Kiro Petrovski
- Davies Livestock Research Centre, School of Animal and Veterinary Sciences, University of Adelaide, Roseworthy Campus, SA 5371, Australia
| | - Tong Chen
- Davies Livestock Research Centre, School of Animal and Veterinary Sciences, University of Adelaide, Roseworthy Campus, SA 5371, Australia
| | - John L Williams
- Davies Livestock Research Centre, School of Animal and Veterinary Sciences, University of Adelaide, Roseworthy Campus, SA 5371, Australia; Dipartimento di Scienze Animali, della Nutrizione e degli Alimenti, Università Cattolica del Sacro Cuore, 29122 Piacenza, Italy
| | - Cynthia D K Bottema
- Davies Livestock Research Centre, School of Animal and Veterinary Sciences, University of Adelaide, Roseworthy Campus, SA 5371, Australia
| |
Collapse
|
28
|
Abstract
Human breast milk is the optimal nutrition for all infants and is comprised of many bioactive and immunomodulatory components. The components in human milk, such as probiotics, human milk oligosaccharides (HMOs), extracellular vesicles, peptides, immunoglobulins, growth factors, cytokines, and vitamins, play a critical role in guiding neonatal development beyond somatic growth. In this review, we will describe the bioactive factors in human milk and discuss how these factors shape neonatal immunity, the intestinal microbiome, intestinal development, and more from the inside out.
Collapse
Affiliation(s)
- Sarah F Andres
- Department of Pediatrics, Pediatric GI Division, School of Medicine, Oregon Health and Science University, Portland, OR 97229, United States
| | - Brian Scottoline
- Division of Neonatology, Department of Pediatrics, Oregon Health & Science University, Portland, OR 97239, United States
| | - Misty Good
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, University of North Carolina at Chapel Hill, 101 Manning Drive, Campus Box 7596, Chapel Hill, NC 27599, United States.
| |
Collapse
|
29
|
Kim JH, Kim KU, Min H, Lee ES, Lim IS, Song J, Kang I, Yi DY. Changes in microRNAs during Storage and Processing of Breast Milk. Metabolites 2023; 13:139. [PMID: 36837760 PMCID: PMC9963775 DOI: 10.3390/metabo13020139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/31/2022] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Human breast milk (HBM) is the ideal source of nutrients for infants and is rich in microRNA (miRNA). In recent years, expressed breast milk feeding rather than direct breastfeeding has become increasingly prevalent for various reasons. Expressed HBM requires storage and processing, which can cause various changes in the ingredients. We investigated how the miRNAs in HBM change due to processes often used in real life. HBM samples collected from 10 participants were each divided into seven groups according to the storage temperature, thawing method, and storage period. In addition, we analyzed the miRNA changes in each group. The number of microRNAs that showed significant expression was not large compared to the thousands of miRNAs contained in breast milk. Therefore, it is difficult to suggest that the various storage and thawing processes have a great influence on the overall expression of miRNA. However, a short-term refrigeration storage method revealed little change in nutrients compared to other storage and thawing methods. Taking all factors into consideration, short-term refrigeration is recommended to minimize changes in the composition or function of breast milk.
Collapse
Affiliation(s)
- Jun Hwan Kim
- Department of Pediatrics, Chung-Ang University Hospital, Seoul 06973, Republic of Korea
- College of Medicine, Chung-Ang University, Seoul 06972, Republic of Korea
| | - Ki-Uk Kim
- College of Pharmacy, Chung-Ang University, Seoul 06972, Republic of Korea
| | - Hyeyoung Min
- College of Pharmacy, Chung-Ang University, Seoul 06972, Republic of Korea
| | - Eun Sun Lee
- Department of Pediatrics, Chung-Ang University Hospital, Seoul 06973, Republic of Korea
| | - In Seok Lim
- Department of Pediatrics, Chung-Ang University Hospital, Seoul 06973, Republic of Korea
- College of Medicine, Chung-Ang University, Seoul 06972, Republic of Korea
| | - Jeonglyn Song
- Chung-Ang University Industry Academic Cooperation Foundation, Seoul 06972, Republic of Korea
| | - Insoo Kang
- Departments of Internal Medicine, Section of Rheumatology, Allergy & Immunology, Yale University School of Medicine, S525C TAC, 300 Cedar Street, New Haven, CT 06520, USA
| | - Dae Yong Yi
- Department of Pediatrics, Chung-Ang University Hospital, Seoul 06973, Republic of Korea
- College of Medicine, Chung-Ang University, Seoul 06972, Republic of Korea
| |
Collapse
|
30
|
Ukraintsev SE, Samal TN. The Appearance and Establishment of Breastfeeding Amongst Animals: From Echidna and Platypus to Mammals and Human. CURRENT PEDIATRICS 2022. [DOI: 10.15690/vsp.v21i6.2492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The paper provides review of the scientific literature dedicated to the evolutionary aspects of breastfeeding in the animal kingdom. Differences in breast milk composition amongst different mammals along with changes in breast milk composition during evolution provided. Special attention is paid to oligosaccharides — unique components of the breast milk of Homo sapiens.
Collapse
|
31
|
Hicks SD, Beheshti R, Chandran D, Warren K, Confair A. Infant consumption of microRNA miR-375 in human milk lipids is associated with protection from atopy. Am J Clin Nutr 2022; 116:1654-1662. [PMID: 36166840 DOI: 10.1093/ajcn/nqac266] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 09/19/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Human milk is thought to reduce infant atopy risk. The biologic mechanism for this protective effect is not fully understood. OBJECTIVES We tested the hypothesis that infant consumption of 4 microRNAs (miR-146b-5p, miR-148b-3p, miR-21-5p, and miR-375-3p) in human milk would be associated with reduced atopy risk. METHODS The Breast Milk Influence of the Microtranscriptome Profile on Atopy in Children over Time (IMPACT) study involved a cohort of mother-infant dyads who planned to breastfeed beyond 4 mo. Infant consumption of the 4 human milk microRNAs (miRNAs) in the first 6 mo was calculated as the product of milk miRNA concentration and the number of human milk feeds, across 3 lactation stages: early milk (0-4 wk), transitional milk (4-16 wk), and mature milk (16-24 wk). The primary outcome was infant atopy in the first year, defined as atopic dermatitis (AD), food allergies, or wheezing. The final analysis included 432 human milk samples and 7824 wk of longitudinal health data from 163 dyads. RESULTS Seventy-three infants developed atopy. Forty-one were diagnosed with AD (25%), 33 developed food allergy (20%), and 10 had wheezing (6%). Eleven developed >1 condition (7%). Infants who did not develop atopy consumed higher concentrations of miR-375-3p (d = 0.18, P = 0.022, adj P = 0.044) and miR-148b-3p (d = 0.23, P = 0.007, adj P = 0.028). The consumption of miR-375-3p (X2 = 5.7, P = 0.017, OR: 0.92, 95% CI: 0.86, 0.99) was associated with reduced atopy risk. Concentrations of miR-375-3p increased throughout lactation (r = 0.46, F = 132.3, P = 8.4 × 10-34) and were inversely associated with maternal body mass (r = -0.11, t = -2.1, P = 0.032). CONCLUSIONS This study provides evidence that infant consumption of miR-375-3p may reduce atopy risk.
Collapse
Affiliation(s)
- Steven D Hicks
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Ramin Beheshti
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Desirae Chandran
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Kaitlyn Warren
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Alexandra Confair
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| |
Collapse
|
32
|
Chiba T, Takaguri A, Kooka A, Kowatari K, Yoshizawa M, Fukushi Y, Hongo F, Sato H, Fujisawa M, Wada S, Maeda T. Suppression of milk-derived miR-148a caused by stress plays a role in the decrease in intestinal ZO-1 expression in infants. Clin Nutr 2022; 41:2691-2698. [PMID: 36343560 DOI: 10.1016/j.clnu.2022.10.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 09/17/2022] [Accepted: 10/09/2022] [Indexed: 01/27/2023]
Abstract
BACKGROUND & AIMS Milk-derived miR-148a-3p (miR-148a), which is abundant in breast milk, has been shown to be associated with the development of infants' intestines. Although it is well known that stress during lactation changes milk constituents in terms of lipid and protein, no studies have examined the influence of stress on miR-148a expression in breast milk. The objective of this study is to investigate the relationship between stress and miR-148a expression in milk, and to evaluate whether the changes in milk-derived miR-148a expression-caused by the mother's exposure to stress-influence intestinal ZO-1 expression in infants. METHODS The participants of this study were healthy Japanese women who were nursing. Psychological stress evaluation of the subjects was conducted using a short form of the Profile of Mood State Second Edition-Adult (POMS-2). Additionally, miR-148a expressions in restraint stressed nursing mice were investigated using quantitative real-time PCR. The levels of a tight junction protein zonula occludens-1 (ZO-1) and DNA methyltransferase 1 (DNMT1), which is a direct target of miR-148a, in ileum in neonatal mice breastfed by stressed nursing mice were investigated using Western blot. Furthermore, to investigate the influence of miR-148a on ZO-1 expression within the intestine, the levels of ZO-1 and DNMT1 in human intestinal epithelial Caco-2 cells with lentivirus-mediated miR-148a overexpression were evaluated. RESULTS A significantly negative correlation was observed between relative miR-148a expression in breast milk and the total mood disturbance T-score. Each T-score on negative mood subscales of anger-hostility, confusion-bewilderment, depression-dejection, fatigue-inertia, and tension-anxiety was significantly negatively correlated with relative miR-148a expression in breast milk: a positive mood subscale vigor-activity T-score was significantly positively correlated with relative miR-148a expression in breast milk. A positive mood friendliness T-score, estimated separately from other scores, was significantly positively correlated with relative miR-148a expression in breast milk. Additionally, the relative expression of miR-148a in the milk obtained from stressed mice was significantly lower than that of control mice. The relative level of ZO-1 in ileum of neonatal mice nursed by stressed mice was significantly lower than that of neonatal mice nursed by control mice. Additionally, the relative level of DNMT1 in ileum of neonatal mice nursed by stressed mice was significantly higher than that of neonatal mice nursed by control mice. Furthermore, the relative level of ZO-1 in miR-148a-overexpressed Caco-2 cells was significantly higher than that in control cells. The relative level of DNMT1 in miR-148a-overexpressed Caco-2 cells was significantly lower than that in control cells. CONCLUSIONS Mothers' exposure to stress during lactation may cause miR-148a expression in breast milk. Additionally, stressed-induced suppression of miR-148a expression in breast milk may cause a decrease in intestinal ZO-1 level via the increase in DNMT1 in infants' intestines. These observations are beneficial information for breastfeeding mothers and their families and perinatal medical professionals. Our findings encourage monitoring maternal psychological stress during lactation to promote breastfeeding and adequate infant nutrition.
Collapse
Affiliation(s)
- Takeshi Chiba
- Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 15-4-1, Maeda 7-jo, Teine-ku, Sapporo-shi, Hokkaido, Japan; Creation Research Institute of Life Science in KITA-no-DAICHI, Hokkaido University of Science, 15-4-1, Maeda 7-jo, Teine-ku, Sapporo-shi, Hokkaido, Japan.
| | - Akira Takaguri
- Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 15-4-1, Maeda 7-jo, Teine-ku, Sapporo-shi, Hokkaido, Japan; Creation Research Institute of Life Science in KITA-no-DAICHI, Hokkaido University of Science, 15-4-1, Maeda 7-jo, Teine-ku, Sapporo-shi, Hokkaido, Japan
| | - Aya Kooka
- Department of Pharmacy, Teine Keijinkai Hospital, 12-1-10, Maeda 1-jo, Teine-ku, Sapporo-shi, Hokkaido, Japan
| | - Kiyoko Kowatari
- Department of Nursing, Teine Keijinkai Hospital, 12-1-10, Maeda 1-jo, Teine-ku, Sapporo-shi, Hokkaido, Japan
| | - Megumi Yoshizawa
- Department of Nursing, Teine Keijinkai Hospital, 12-1-10, Maeda 1-jo, Teine-ku, Sapporo-shi, Hokkaido, Japan
| | - Yoshiyuki Fukushi
- Department of Obstetrics and Gynecology, Teine Keijinkai Hospital, 12-1-10, Maeda 1-jo, Teine-ku, Sapporo-shi, Hokkaido, Japan
| | - Fuminori Hongo
- Department of Pharmacy, Teine Keijinkai Hospital, 12-1-10, Maeda 1-jo, Teine-ku, Sapporo-shi, Hokkaido, Japan
| | - Hideki Sato
- Faculty of Pharmaceutical Sciences, Hokkaido University of Science, 15-4-1, Maeda 7-jo, Teine-ku, Sapporo-shi, Hokkaido, Japan
| | - Miho Fujisawa
- Center for Liberal Arts and Sciences, Iwate Medical University, 1-1-1, Idaidori, Yahaba-cho, Shiwagun, Iwate, Japan
| | - Shinichiro Wada
- Department of Obstetrics and Gynecology, Teine Keijinkai Hospital, 12-1-10, Maeda 1-jo, Teine-ku, Sapporo-shi, Hokkaido, Japan
| | - Tomoji Maeda
- Department of Clinical Pharmacology and Pharmaceutics, Nihon Pharmaceutical University, Kitaadachi-gun, Saitama-shi, Japan
| |
Collapse
|
33
|
Eisha S, Joarder I, Wijenayake S, McGowan PO. Non-nutritive bioactive components in maternal milk and offspring development: a scoping review. J Dev Orig Health Dis 2022; 13:665-673. [PMID: 35387707 DOI: 10.1017/s2040174422000149] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Lactation is a critical time in mammalian development, where maternal factors shape offspring outcomes. In this scoping review, we discuss current literature concerning maternal factors that influence lactation biology and highlight important associations between changes in milk composition and offspring outcomes. Specifically, we explore maternal nutritional, psychosocial, and environmental exposures that influence non-nutritive bioactive components in milk and their links to offspring growth, development, metabolic, and behavioral outcomes. A comprehensive literature search was conducted using the Preferred Reporting Items for Systematic Reviews and Meta-Analysis Extension for Scoping Reviews (PRISMA-ScR) guidelines. Predetermined eligibility criteria were used to analyze 3,275 papers, and the final review included 40 primary research articles. Outcomes of this review identify maternal obesity to be a leading maternal factor influencing the non-nutritive bioactive composition of milk with notable links to offspring outcomes. Offspring growth and development are the most common modes of programming associated with changes in non-nutritive milk composition due to maternal factors in early life. In addition to discussing studies investigating these key associations, we also identify knowledge gaps in the current literature and suggest opportunities and considerations for future studies.
Collapse
Affiliation(s)
- Shafinaz Eisha
- Department of Biological Sciences, Center for Environmental Epigenetics and Development, University of Toronto Scarborough, Toronto, ON, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Ishraq Joarder
- Department of Biological Sciences, Center for Environmental Epigenetics and Development, University of Toronto Scarborough, Toronto, ON, Canada
| | - Sanoji Wijenayake
- Department of Biological Sciences, Center for Environmental Epigenetics and Development, University of Toronto Scarborough, Toronto, ON, Canada
- Department of Biology, Richardson College for the Environment and Science Complex, The University of Winnipeg, Winnipeg, MB, Canada
| | - Patrick O McGowan
- Department of Biological Sciences, Center for Environmental Epigenetics and Development, University of Toronto Scarborough, Toronto, ON, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
- Department of Psychology, University of Toronto, Toronto, ON, Canada
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
34
|
Mecocci S, Trabalza-Marinucci M, Cappelli K. Extracellular Vesicles from Animal Milk: Great Potentialities and Critical Issues. Animals (Basel) 2022; 12:ani12233231. [PMID: 36496752 PMCID: PMC9740508 DOI: 10.3390/ani12233231] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 10/25/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
Other than representing the main source of nutrition for newborn mammals, milk delivers a sophisticated signaling system from mother to child that promotes postnatal health. The bioactive components transferred through the milk intake are important for the development of the newborn immune system and include oligosaccharides, lactoferrin, lysozyme, α-La, and immunoglobulins. In the last 15 years, a pivotal role in this mother-to-child exchange has been attributed to extracellular vesicles (EVs). EVs are micro- and nanosized structures enclosed in a phospholipidic double-layer membrane that are produced by all cell types and released in the extracellular environment, reaching both close and distant cells. EVs mediate the intercellular cross-talk from the producing to the receiving cell through the transfer of molecules contained within them such as proteins, antigens, lipids, metabolites, RNAs, and DNA fragments. The complex cargo can induce a wide range of functional modulations in the recipient cell (i.e., anti-inflammatory, immunomodulating, angiogenetic, and pro-regenerative modulations) depending on the type of producing cells and the stimuli that these cells receive. EVs can be recovered from every biological fluid, including blood, urine, bronchoalveolar lavage fluid, saliva, bile, and milk, which is one of the most promising scalable vesicle sources. This review aimed to present the state-of-the-art of animal-milk-derived EV (mEV) studies due to the exponential growth of this field. A focus on the beneficial potentialities for human health and the issues of studying vesicles from milk, particularly for the analytical methodologies applied, is reported.
Collapse
|
35
|
Greenwood K, Engel R, Grace S. Osteopathic intervention for infants with breastfeeding difficulty: A retrospective case series. INT J OSTEOPATH MED 2022. [DOI: 10.1016/j.ijosm.2022.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
|
36
|
Fan Y, Qin M, Zhu J, Chen X, Luo J, Chen T, Sun J, Zhang Y, Xi Q. MicroRNA sensing and regulating microbiota-host crosstalk via diet motivation. Crit Rev Food Sci Nutr 2022; 64:4116-4133. [PMID: 36287029 DOI: 10.1080/10408398.2022.2139220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Accumulating evidence has demonstrated that diet-derived gut microbiota participates in the regulation of host metabolism and becomes the foundation for precision-based nutritional interventions and the biomarker for potential individual dietary recommendations. However, the specific mechanism of the gut microbiota-host crosstalk remains unclear. Recent studies have identified that noncoding RNAs, as important elements in the regulation of the initiation and termination of gene expression, mediate microbiota-host communication. Besides, the cross-kingdom regulation of non-host derived microRNAs also influence microbiota-host crosstalk via diet motivation. Hence, understanding the relationship between gut microbiota, miRNAs, and host metabolism is indispensable to revealing individual differences in dietary motivation and providing targeted recommendations and strategies. In this review, we first present an overview of the interaction between diet, host genetics, and gut microbiota and collected some latest research associated with microRNAs modulated gut microbiota and intestinal homeostasis. Then, specifically described the possible molecular mechanisms of microRNAs in sensing and regulating gut microbiota-host crosstalk. Lastly, summarized the prospect of microRNAs as biomarkers in disease diagnosis, and the disadvantages of microRNAs in regulating gut microbiota-host crosstalk. We speculated that microRNAs could become potential novel circulating biomarkers for personalized dietary strategies to achieve precise nutrition in future clinical research implications.
Collapse
Affiliation(s)
- Yaotian Fan
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Mengran Qin
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jiahao Zhu
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Xingping Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
- Key Laboratory of Animal Nutrition in Jiangxi Province, Jiangxi Agricultural University, Nanchang, China
| | - Junyi Luo
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Ting Chen
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Jiajie Sun
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Yongliang Zhang
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Qianyun Xi
- Guangdong Provincial Key Laboratory of Animal Nutrition Control, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
37
|
MicroRNAs of Milk in Cells, Plasma, and Lipid Fractions of Human Milk, and Abzymes Catalyzing Their Hydrolysis. Int J Mol Sci 2022; 23:ijms232012070. [PMID: 36292926 PMCID: PMC9603112 DOI: 10.3390/ijms232012070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 10/04/2022] [Accepted: 10/08/2022] [Indexed: 11/17/2022] Open
Abstract
Human milk provides neonates with various components that ensure newborns’ growth, including protection from bacterial and viral infections. In neonates, the biological functions of many breast milk components can be very different compared with their functions in the body fluids of healthy adults. Catalytic antibodies (abzymes) that hydrolyze peptides, proteins, DNAs, RNAs, and oligosaccharides were detected, not only in the blood sera of autoimmune patients, but also in human milk. Non-coding microRNAs (18−25 nucleotides) are intra- and extracellular molecules of different human fluids. MiRNAs possess many different biological functions, including the regulation of several hundred genes. Five of them, miR-148a-3p, miR-200c-3p, miR-378a-3p, miR-146b-5p, and let-7f-5p, were previously found in milk in high concentrations. Here, we determined relative numbers of miRNA copies in 1 mg of analyzed cells, lipid fractions, and plasmas of human milk samples. The relative amount of microRNA decreases in the following order: cells ≈ lipid fraction > plasma. IgGs and sIgAs were isolated from milk plasma, and their activities in the hydrolysis of five microRNAs was compared. In general, sIgAs demonstrated higher miRNA-hydrolyzing activities than IgGs antibodies. The hydrolysis of five microRNAs by sIgAs and IgGs was site-specific. The relative activity of each microRNA hydrolysis was very dependent on the milk preparation. The correlation coefficients between the contents of five RNAs in milk plasma, and the relative activities of sIgAs compared to IgGs in hydrolyses, strongly depended on individual microRNA, and changed from −0.01 to 0.80. Thus, it was shown that milk contains specific antibodies (abzymes) that hydrolyze microRNAs specific for human milk.
Collapse
|
38
|
Bottin JH, Eussen SRBM, Igbinijesu AJ, Mank M, Koyembi JCJ, Nyasenu YT, Ngaya G, Mad-Bondo D, Kongoma JB, Stahl B, Sansonetti PJ, Bourdet-Sicard R, Moya-Alvarez V. Food Insecurity and Maternal Diet Influence Human Milk Composition between the Infant's Birth and 6 Months after Birth in Central-Africa. Nutrients 2022; 14:4015. [PMID: 36235668 PMCID: PMC9573613 DOI: 10.3390/nu14194015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/19/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
Although the World Health Organization (WHO) and UNICEF recommend that infants should be exclusively breastfed for the first 6 months of life, evidence is scarce on how the mother’s undernourishment status at delivery and maternal dietary factors influence human milk (HM) composition during the first 6 months of life in regions with high food insecurity. The maternal undernourishment status at delivery, maternal diet, and HM nutrients were assessed among 46 women and their 48 vaginally born infants in Bangui at 1, 4, 11, 18, and 25 weeks after birth through 24-h recalls and food consumption questionnaires from December 2017 to June 2019 in the context of the "Mother-to-Infant TransmIssion of microbiota in Central-Africa" (MITICA) study. High food insecurity indexes during the follow-up were significantly associated with them having lower levels of many of the human milk oligosaccharides (HMOs) that were measured and with lower levels of retinol (aß-coef = −0.2, p value = 0.04), fatty acids (aß-coef = −7.2, p value = 0.03), and amino acids (aß-coef = −2121.0, p value < 0.001). On the contrary, women from food-insecure households displayed significantly higher levels of lactose in their HM (aß-coef = 3.3, p value = 0.02). In parallel, the consumption of meat, poultry, and fish was associated with higher HM levels of many of the HMOs that were measured, total amino acids (aß-coef = 5484.4, p value < 0.001), and with lower HM levels of lactose (aß-coef = −15.6, p value = 0.01). Food insecurity and maternal diet had a meaningful effect on HM composition with a possible impact being an infant undernourishment risk. Our results plead for consistent actions on food security as an effective manner to influence the nutritional content of HM and thereby, potentially improve infant survival and healthy growth.
Collapse
Affiliation(s)
| | | | | | - Marko Mank
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands or
| | | | - Yawo Tufa Nyasenu
- Laboratoire d’Analyses Médicales, Institut Pasteur de Bangui, Bangui BP923, Central African Republic
- Laboratoire de Biologie Moléculaire et d’Immunologie, Université de Lomé, Lomé P.O. Box 1396, Togo
| | - Gilles Ngaya
- Laboratoire d’Analyses Médicales, Institut Pasteur de Bangui, Bangui BP923, Central African Republic
| | - Daniel Mad-Bondo
- Direction du Service de Santé de la Gendarmerie, Sis Camp Henri IZAMO, Bangui BP790, Central African Republic
| | - Jean-Bertrand Kongoma
- Direction du Service de Santé de la Gendarmerie, Sis Camp Henri IZAMO, Bangui BP790, Central African Republic
| | - Bernd Stahl
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands or
- Department of Chemical Biology & Drug Discovery, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CG Utrecht, The Netherlands
| | - Philippe J. Sansonetti
- Chaire de Microbiologie et Maladies Infectieuses, Collège de France, 75005 Paris, France
- Unité de Pathogénie Microbienne Moléculaire, INSERM U1202, Department of Cell Biology and Infection, Institut Pasteur, 75015 Paris, France
| | | | - Violeta Moya-Alvarez
- Unité de Pathogénie Microbienne Moléculaire, INSERM U1202, Department of Cell Biology and Infection, Institut Pasteur, 75015 Paris, France
- Epidemiology of Emergent Diseases Unit, Global Health Department, Institut Pasteur, 75015 Paris, France
| |
Collapse
|
39
|
Suvekbala V, Ramachandran H, Veluchamy A, Mascarenhas MAB, Ramprasath T, Nair MKC, Garikipati VNS, Gundamaraju R, Subbiah R. The Promising Epigenetic Regulators for Refractory Epilepsy: An Adventurous Road Ahead. Neuromolecular Med 2022:10.1007/s12017-022-08723-0. [DOI: 10.1007/s12017-022-08723-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 07/13/2022] [Indexed: 10/14/2022]
|
40
|
Norouzi M, Bakhtiarizadeh MR, Salehi A. Investigation of the transability of dietary small non-coding RNAs to animals. Front Genet 2022; 13:933709. [PMID: 36134021 PMCID: PMC9483711 DOI: 10.3389/fgene.2022.933709] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 07/28/2022] [Indexed: 11/13/2022] Open
Abstract
Our daily diet not only provides essential nutrients needed for survival and growth but also supplies bioactive ingredients to promote health and prevent disease. Recent studies have shown that exogenous microRNAs (miRNAs), xenomiRs, may enter the consumer’s body through dietary intake and regulate gene expression. This fascinating phenomenon suggests that xenomiRs can act as a new class of bioactive substances associated with mammalian systems. In contrast, several studies have failed to detect xenomiRs in consumers and reported that the observed diet-derived miRNAs in the previous studies can be related to the false positive effects of experiments. This discrepancy can be attributed to the potential artifacts related to the process of experiments, small sample size, and inefficient bioinformatics pipeline. Since this hypothesis is not generally accepted yet, more studies are required. Here, a stringent and reliable bioinformatics pipeline was used to analyze 133 miRNA sequencing data from seven different studies to investigate this phenomenon. Generally, our results do not support the transfer of diet-derived miRNAs into the animal/human tissues in every situation. Briefly, xenomiRs were absent from most samples, and also, their expressions were very low in the samples where they were present, which is unlikely to be sufficient to regulate cell transcripts. Furthermore, this study showed that the possibility of miRNAs being absorbed through animals’ diets and thus influencing gene expression during specific periods of biological development is not inconceivable. In this context, our results were in agreement with the theory of the transfer of small RNAs under certain conditions and periods as xenomiRs were found in colostrum which may modulate infants’ immune systems via post-transcriptional regulation. These findings provide evidence for the selective absorption of diet-derived small RNAs, which need to be investigated in future studies to shed light on the mechanisms underlying the transference of diet-derived miRNAs.
Collapse
Affiliation(s)
- Milad Norouzi
- Department of Animal and Poultry Science, College of Aburaihan, University of Tehran, Tehran, Iran
| | | | - Abdolreza Salehi
- Department of Animal and Poultry Science, College of Aburaihan, University of Tehran, Tehran, Iran
| |
Collapse
|
41
|
Tovarnytska A. Breastfeeding impact on patent ductus arteriosus closure in preterm newborns. CHILD`S HEALTH 2022; 17:184-191. [DOI: 10.22141/2224-0551.17.4.2022.1515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Abstract
Background. Patent ductus arteriosus (PDA) is a serious problem in clinical neonatology and pediatrics. Today treatment of PDA remains debatable because of high risk of complications with medical and invasive treatment. The purpose of the study was to determine the dependence of PDA closure on the type of feeding the newborn. Materials and methods. We have made a retrospective analysis of 300 case histories of children in neonatal units. We selected two comparison groups: the main group consisted of breastfed babies born at 37 weeks gestation (n = 102); controls — preterm infants who received artificial feeding since birth (n = 198). Results. At discharge from the hospital, 44 (14.7 ± 2.0 %) children had PDA (p < 0.05). The percentage of infants on artificial feeding with PDA was 2.3 times higher compared to breastfed babies: 18.2 ± 2.7 % versus 7.8 ± 2.7 % (p = 0.017 by χ2 test). Respiratory distress syndrome was more common in the control group as well: 72.7 ± 3.2 % versus 46.1 ± 4.9 % (p < 0.001). Additionally, the group of formula-fed children more often had severe complications of respiratory distress syndrome: respiratory failure type III — 25.8 ± 3.1 % versus12.7 ± 3.3 % (р = 0.009); development of bronchopulmonary dysplasia — 13.1 ± 2.4 % versus 2.9 ± 1.7 % (p = 0.005). The need for invasive mechanical ventilation in the intensive care unit was observed much less often in breastfed children. Moreover, the level of respiratory disorders (type II respiratory failure) in both groups differed insignificantly (p = 0.742): 24.5 ± 4.3 % in the main group and 26.3 ± 3.1 % in controls. Conclusions. Breastfeeding from the first days of life contributes to the closure of the ductus arteriosus and the favorable course of respiratory distress syndrome in premature babies.
Collapse
|
42
|
Li DF, Yang MF, Xu J, Xu HM, Zhu MZ, Liang YJ, Zhang Y, Tian CM, Nie YQ, Shi RY, Wang LS, Yao J. Extracellular Vesicles: The Next Generation Theranostic Nanomedicine for Inflammatory Bowel Disease. Int J Nanomedicine 2022; 17:3893-3911. [PMID: 36092245 PMCID: PMC9462519 DOI: 10.2147/ijn.s370784] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 08/25/2022] [Indexed: 12/02/2022] Open
Abstract
The recent rapid development in the field of extracellular vesicles (EVs) based nanotechnology has provided unprecedented opportunities for nanomedicine platforms. As natural nanocarriers, EVs such as exosomes, exosome-like nanoparticles and outer membrane vesicles (OMVs), have unique structure/composition/morphology characteristics, and show excellent physical and chemical/biochemical properties, making them a new generation of theranostic nanomedicine. Here, we reviewed the characteristics of EVs from the perspective of their formation and biological function in inflammatory bowel disease (IBD). Moreover, EVs can crucially participate in the interaction and communication of intestinal epithelial cells (IECs)-immune cells-gut microbiota to regulate immune response, intestinal inflammation and intestinal homeostasis. Interestingly, based on current representative examples in the field of exosomes and exosome-like nanoparticles for IBD treatment, it is shown that plant, milk, and cells-derived exosomes and exosome-like nanoparticles can exert a therapeutic effect through their components, such as proteins, nucleic acid, and lipids. Moreover, several drug loading methods and target modification of exosomes are used to improve their therapeutic capability. We also discussed the application of exosomes and exosome-like nanoparticles in the treatment of IBD. In this review, we aim to better and more clearly clarify the underlying mechanisms of the EVs in the pathogenesis of IBD, and provide directions of exosomes and exosome-like nanoparticles mediated for IBD treatment.
Collapse
Affiliation(s)
- De-feng Li
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, People’s Republic of China
| | - Mei-feng Yang
- Department of Hematology, Yantian District People’s Hospital, Shenzhen, People’s Republic of China
| | - Jing Xu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital (School of Medicine of South China University of Technology), Guangzhou, People’s Republic of China
| | - Hao-ming Xu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital (School of Medicine of South China University of Technology), Guangzhou, People’s Republic of China
| | - Min-zheng Zhu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital (School of Medicine of South China University of Technology), Guangzhou, People’s Republic of China
| | - Yu-jie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen, People’s Republic of China
| | - Yuan Zhang
- Department of Medical Administration, Huizhou Institute of Occupational Diseases Control and Prevention, Huizhou, People’s Republic of China
| | - Cheng-mei Tian
- Department of Emergency, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, People’s Republic of China
| | - Yu-qiang Nie
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital (School of Medicine of South China University of Technology), Guangzhou, People’s Republic of China
| | - Rui-yue Shi
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, People’s Republic of China
| | - Li-sheng Wang
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, People’s Republic of China
| | - Jun Yao
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, People’s Republic of China
| |
Collapse
|
43
|
Sajjadi E, Gaudioso G, Terrasi A, Boggio F, Venetis K, Ivanova M, Bertolasi L, Lopez G, Runza L, Premoli A, Lorenzini D, Guerini-Rocco E, Ferrero S, Vaira V, Fusco N. Osteoclast-like stromal giant cells in breast cancer likely belong to the spectrum of immunosuppressive tumor-associated macrophages. Front Mol Biosci 2022; 9:894247. [PMID: 36090031 PMCID: PMC9462457 DOI: 10.3389/fmolb.2022.894247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 07/28/2022] [Indexed: 12/11/2022] Open
Abstract
Background: Breast cancer with osteoclast-like stromal giant cells (OSGC) is an exceedingly rare morphological pattern of invasive breast carcinoma. The tumor immune microenvironment (TIME) of these tumors is populated by OSGC, which resemble osteoclasts and show a histiocytic-like immunophenotype. Their role in breast cancer is unknown. The osteoclast maturation in the bone is regulated by the expression of cytokines that are also present in the TIME of tumors and in breast cancer tumor-associated macrophages (TAMs). TAMs-mediated anti-tumor immune pathways are regulated by miRNAs akin to osteoclast homeostasis. Here, we sought to characterize the different cellular compartments of breast cancers with OSGC and investigate the similarities of OSGC with tumor and TIME in terms of morphology, protein, and miRNA expression, specifically emphasizing on monocytic signatures. Methods and Results: Six breast cancers with OSGC were included. Tumor-infiltrating lymphocytes (TILs) and TAMs were separately quantified. The different cellular populations (i.e., normal epithelium, cancer cells, and OSGC) were isolated from tissue sections by laser-assisted microdissection. After RNA purification, 752 miRNAs were analyzed using a TaqMan Advanced miRNA Low-Density Array for all samples. Differentially expressed miRNAs were identified by computing the fold change (log2Ratio) using the Kolmogorov-Smirnov test and p values were corrected for multiple comparisons using the false discovery rate (FDR) approach. As a similarity analysis among samples, we used the Pearson test. The association between pairs of variables was investigated using Fisher exact test. Classical and non-classical monocyte miRNA signatures were finally applied. All OSGC displayed CD68 expression, TILs (range, 45–85%) and high TAMs (range, 35–75%). Regarding the global miRNAs profile, OSGC was more similar to cancer cells than to non-neoplastic ones. Shared deregulation of miR-143-3p, miR-195-5p, miR-181a-5p, and miR-181b-5p was observed between OSGC and cancer cells. The monocyte-associated miR-29a-3p and miR-21-3p were dysregulated in OSGCs compared with non-neoplastic or breast cancer tissues. Conclusion: Breast cancers with OSGC have an activated TIME. Shared epigenetic events occur during the ontogenesis of breast cancer cells and OSGC but the innumophenotype and miRNA profiles of the different cellular compartmens suggest that OSGC likely belong to the spectrum of M2 TAMs.
Collapse
Affiliation(s)
- Elham Sajjadi
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Gabriella Gaudioso
- Division of Pathology, Fondazione IRCCS Ca’ Granda—Ospedale Maggiore Policlinico, Milan, Italy
| | - Andrea Terrasi
- Division of Molecular Biology, Biomedical Center, Faculty of Medicine, LMU Munich, Planegg-Martinsried, Munich, Germany
| | - Francesca Boggio
- Division of Pathology, Fondazione IRCCS Ca’ Granda—Ospedale Maggiore Policlinico, Milan, Italy
| | - Konstantinos Venetis
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Mariia Ivanova
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Letizia Bertolasi
- Division of Pathology, Fondazione IRCCS Ca’ Granda—Ospedale Maggiore Policlinico, Milan, Italy
| | - Gianluca Lopez
- Division of Pathology, Fondazione IRCCS Ca’ Granda—Ospedale Maggiore Policlinico, Milan, Italy
| | - Letterio Runza
- Division of Pathology, Fondazione IRCCS Ca’ Granda—Ospedale Maggiore Policlinico, Milan, Italy
| | - Alice Premoli
- Division of Pathology, Fondazione IRCCS Ca’ Granda—Ospedale Maggiore Policlinico, Milan, Italy
| | - Daniele Lorenzini
- Division of Pathology, Fondazione IRCCS Ca’ Granda—Ospedale Maggiore Policlinico, Milan, Italy
| | - Elena Guerini-Rocco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Stefano Ferrero
- Division of Pathology, Fondazione IRCCS Ca’ Granda—Ospedale Maggiore Policlinico, Milan, Italy
- Department of Biomedical, Surgical, and Dental Sciences, University of Milan, Milan, Italy
| | - Valentina Vaira
- Division of Pathology, Fondazione IRCCS Ca’ Granda—Ospedale Maggiore Policlinico, Milan, Italy
- Department of Pathophysiology and Transplantation, University of Milan, Milan, Italy
| | - Nicola Fusco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
- *Correspondence: Nicola Fusco,
| |
Collapse
|
44
|
Miura H, Jimbo I, Oda M, Noguchi M, Kawasaki K, Osada-Oka M, Tsukahara T, Inoue R. Effect of Porcine Colostral Exosomes on T Cells in the Peripheral Blood of Suckling Piglets. Animals (Basel) 2022; 12:ani12172172. [PMID: 36077893 PMCID: PMC9455021 DOI: 10.3390/ani12172172] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 08/19/2022] [Accepted: 08/20/2022] [Indexed: 12/17/2022] Open
Abstract
Simple Summary Exosomes in porcine colostrum have gained attention as the possible key compounds involved in the growth and/or development of suckling piglets. In this study, peripheral blood mononuclear cells (PBMCs) from suckling piglets were cultured with or without milk-derived exosomes (control) in vitro. Porcine colostral exosomes increased the proportion of cytotoxic T (Tc) cells, while this phenomenon was not observed in PBMC whose endocytosis was inhibited. Moreover, exosome-treated PBMCs had a higher cytokine IL-2 concentration in the culture supernatant than the control. The present study demonstrated that porcine colostral exosomes could increase the Tc cell proportion in the peripheral blood of a suckling piglet, with the underlying mechanism believed to be the stimulation of IL-2 production in PBMCs via endocytosis. Abstract Growing evidence indicates that porcine colostral exosomes may contribute to the healthy development of piglets. Here, we evaluated in vitro the effect of porcine milk-derived exosomes, in particular colostral exosomes, on T cells in the peripheral blood of suckling piglets. A total of seven sows and thirteen suckling piglets were used. Peripheral blood mononuclear cells (PBMCs) from suckling piglets were cultured with or without milk-derived exosomes (control). Using flow cytometry, the proportion of each T cell subset in cultured PBMCs was analyzed three days post-incubation. PBMCs cultured with porcine colostral exosomes had a higher proportion of CD3+CD4−CD8+ T cells (cytotoxic T cells; Tc) than the control. However, exosomes induced no increase in the Tc cell population in PBMC whose endocytosis was inhibited. We further measured the concentrations of cytokines in the culture supernatant. Exosome-treated PBMCs had a higher cytokine IL-2 concentration than the control. The present study demonstrated that porcine colostral exosomes could increase the Tc cell proportion in the peripheral blood of suckling piglets, with the underlying mechanism believed to be the stimulation of IL-2 production in PBMCs via endocytosis. Moreover, our results suggested that porcine colostral exosomes were involved in the development of cellular immunity in suckling piglets.
Collapse
Affiliation(s)
- Hiroto Miura
- Laboratory of Animal Science, Setsunan University, Hirakata, Osaka 573-0101, Japan
| | - Itsuki Jimbo
- Laboratory of Animal Science, Kyoto Prefectural University, Kyoto, Kyoto 606-8522, Japan
| | - Machi Oda
- Laboratory of Animal Science, Setsunan University, Hirakata, Osaka 573-0101, Japan
| | - Michiko Noguchi
- Laboratory of Theriogenology, Azabu University, Sagamihara, Kanagawa 252-5201, Japan
| | - Kiyonori Kawasaki
- Graduate School of Agriculture, Kagawa University, Kita, Kagawa 761-0795, Japan
| | - Mayuko Osada-Oka
- Food Hygiene and Environmental Health, Faculty of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto, Kyoto 606-8522, Japan
| | | | - Ryo Inoue
- Laboratory of Animal Science, Setsunan University, Hirakata, Osaka 573-0101, Japan
- Laboratory of Animal Science, Kyoto Prefectural University, Kyoto, Kyoto 606-8522, Japan
- Correspondence: ; Tel.: +81-(0)-728965469
| |
Collapse
|
45
|
Zheng Y, Correa-Silva S, Palmeira P, Carneiro-Sampaio M. Maternal vaccination as an additional approach to improve the protection of the nursling: Anti-infective properties of breast milk. Clinics (Sao Paulo) 2022; 77:100093. [PMID: 35963149 PMCID: PMC9382412 DOI: 10.1016/j.clinsp.2022.100093] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/08/2022] [Accepted: 07/14/2022] [Indexed: 11/30/2022] Open
Abstract
Human milk constitutes a secretion with unique functions of both nourishing the nursling and providing protection against enteric and respiratory infections, mainly due to its content of secretory IgA antibodies but also due to the presence of a plethora of bioactive factors. Specific IgA antibodies are produced locally by plasma cells derived from B lymphocytes that migrate from other mucosae to the mammary gland during lactation, particularly from the gastrointestinal and respiratory tracts. Therefore, here, the authors will provide a comprehensive review of the content and functions of different nutritional and bioactive anti-infectious components from breast milk, such as oligosaccharides, lactoferrin, haptocorrin, α-lactalbumin, k-casein, lysozyme, lactoperoxidase, mucin, fatty acids, defensins, cytokines and chemokines, hormones and growth factors, complement proteins, leukocytes and nucleic acids, including microRNAs, among many others, and the induction of antibody responses in breast milk after maternal vaccination with several licensed vaccines, including the anti-SARS-CoV-2 vaccine preparations used worldwide. Currently, in the midst of the pandemic, maternal vaccination has re-emerged as a crucial source of passive immunity to the neonate through the placenta and breastfeeding, considering that maternal vaccination can induce specific antibodies if performed during pregnancy and after delivery. There have been some reports in the literature about milk IgA antibodies induced by bacterial antigens or inactivated virus vaccines, such as anti-diphtheria-tetanus-pertussis, anti-influenza viruses, anti-pneumococcal and meningococcal polysaccharide preparations. Regarding anti-SARS-CoV-2 vaccines, most studies demonstrate elevated levels of specific IgA and IgG antibodies in milk with virus-neutralizing ability after maternal vaccination, which represents an additional approach to improve the protection of the nursling during the entire breastfeeding period.
Collapse
Affiliation(s)
- Yingying Zheng
- Department of Pediatrics, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil.
| | - Simone Correa-Silva
- Department of Pediatrics, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil; Universidade Paulista, UNIP, São Paulo, SP, Brazil.
| | - Patricia Palmeira
- Laboratory of Medical Investigation (LIM-36), Department of Pediatrics, Hospital das Clínicas HCFMUSP, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil.
| | - Magda Carneiro-Sampaio
- Department of Pediatrics, Faculdade de Medicina, Universidade de São Paulo, São Paulo, SP, Brazil.
| |
Collapse
|
46
|
Gregg B, Ellsworth L, Pavela G, Shah K, Berger PK, Isganaitis E, VanOmen S, Demerath EW, Fields DA. Bioactive compounds in mothers milk affecting offspring outcomes: A narrative review. Pediatr Obes 2022; 17:e12892. [PMID: 35060344 PMCID: PMC9177518 DOI: 10.1111/ijpo.12892] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 12/07/2021] [Accepted: 01/03/2022] [Indexed: 01/01/2023]
Abstract
BACKGROUND Compared to the exhaustive study of transgenerational programming of obesity and diabetes through exposures in the prenatal period, postnatal programming mechanisms are understudied, including the potential role of breast milk composition linking maternal metabolic status (body mass index and diabetes) and offspring growth, metabolic health and future disease risk. METHODS This narrative review will principally focus on four emergent bioactive compounds [microRNA's (miRNA), lipokines/signalling lipids, small molecules/metabolites and fructose] that, until recently were not known to exist in breast milk. The objective of this narrative review is to integrate evidence across multiple fields of study that demonstrate the importance of these compositional elements of breast milk during lactation and the subsequent effect of breast milk components on the health of the infant. RESULTS Current knowledge on the presence of miRNA's, lipokines/signalling lipids, small molecules/metabolites and fructose in breast milk and their associations with infant outcomes is compelling, but far from resolved. Two themes emerge: (1) maternal metabolic phenotypes are associated with these bioactives and (2) though existing in milk at low concentrations, they are also associated with offspring growth and body composition. CONCLUSION Breast milk research is gaining momentum though we must remain focused on understanding how non-nutritive bioactive components are affected by the maternal phenotype, how they subsequently impact infant outcomes. Though early, there is evidence to suggest fructose is associated with fat mass in the 1st months of life whereas 12,13 diHOME (brown fat activator) and betaine are negatively associated with early adiposity and growth.
Collapse
Affiliation(s)
- Brigid Gregg
- Department of Pediatrics, Division of Pediatric Endocrinology, University of Michigan, Ann Arbor, MI, USA
| | - Lindsay Ellsworth
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Gregory Pavela
- Department of Health Behavior, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kruti Shah
- Department of Pediatrics, Section of Endocrinology and Diabetes, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Paige K. Berger
- Department of Pediatrics, The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | - Elvira Isganaitis
- Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA (USA)
| | - Sheri VanOmen
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Ellen W. Demerath
- Division of Epidemiology and Community Health, University of Minnesota, Minneapolis, MN, USA
| | - David A. Fields
- Department of Pediatrics, Section of Endocrinology and Diabetes, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA,Correspondence to: Address: University of Oklahoma Health Sciences Center, 1200 Children's Avenue Suite 4500, Oklahoma City, OK73104, USA
| |
Collapse
|
47
|
Li W, Li W, Wang X, Zhang H, Wang L, Gao T. Comparison of miRNA profiles in milk-derived extracellular vesicles and bovine mammary glands. Int Dairy J 2022. [DOI: 10.1016/j.idairyj.2022.105444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
48
|
Dietary Improvement during Lactation Normalizes miR-26a, miR-222 and miR-484 Levels in the Mammary Gland, but Not in Milk, of Diet-Induced Obese Rats. Biomedicines 2022; 10:biomedicines10061292. [PMID: 35740314 PMCID: PMC9219892 DOI: 10.3390/biomedicines10061292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 05/24/2022] [Accepted: 05/24/2022] [Indexed: 02/01/2023] Open
Abstract
We aimed to evaluate in rats whether the levels of specific miRNA are altered in the mammary gland (MG) and milk of diet-induced obese dams, and whether improving maternal nutrition during lactation attenuates such alterations. Dams fed with a standard diet (SD) (control group), with a Western diet (WD) prior to and during gestation and lactation (WD group), or with WD prior to and during gestation but moved to SD during lactation (Rev group) were followed. The WD group showed higher miR-26a, miR-222 and miR-484 levels than the controls in the MG, but the miRNA profile in Rev animals was not different from those of the controls. The WD group also displayed higher miR-125a levels than the Rev group. Dams of the WD group, but not the Rev group, displayed lower mRNA expression levels of Rb1 (miR-26a’s target) and Elovl6 (miR-125a’s target) than the controls in the MG. The WD group also presented lower expression of Insig1 (miR-26a’s target) and Cxcr4 (miR-222’s target) than the Rev group. However, both WD and Rev animals displayed lower expression of Vegfa (miR-484’s target) than the controls. WD animals also showed greater miR-26a, miR-125a and miR-222 levels in the milk than the controls, but no differences were found between the WD and Rev groups. Thus, implementation of a healthy diet during lactation normalizes the expression levels of specific miRNAs and some target genes in the MG of diet-induced obese dams but not in milk.
Collapse
|
49
|
Selma-Royo M, Calvo-Lerma J, Bäuerl C, Esteban-Torres M, Cabrera-Rubio R, Collado MC. Human milk microbiota: what did we learn in the last 20 years? MICROBIOME RESEARCH REPORTS 2022; 1:19. [PMID: 38046359 PMCID: PMC10688795 DOI: 10.20517/mrr.2022.05] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/06/2022] [Accepted: 04/29/2022] [Indexed: 12/05/2023]
Abstract
Human milk (HM) is the gold standard for infant nutrition during the first months of life. Beyond its nutritional components, its complex bioactive composition includes microorganisms, their metabolites, and oligosaccharides, which also contribute to gut colonization and immune system maturation. There is growing evidence of the beneficial effects of bacteria present in HM. However, current research presents limited data on the presence and functions of other organisms. The potential biological impacts on maternal and infant health outcomes, the factors contributing to milk microbes' variations, and the potential functions in the infant's gut remain unclear. This review provides a global overview of milk microbiota, what the actual knowledge is, and what the gaps and challenges are for the next years.
Collapse
Affiliation(s)
| | | | | | | | | | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia 46980, Spain
| |
Collapse
|
50
|
Hatmal MM, Al-Hatamleh MAI, Olaimat AN, Alshaer W, Hasan H, Albakri KA, Alkhafaji E, Issa NN, Al-Holy MA, Abderrahman SM, Abdallah AM, Mohamud R. Immunomodulatory Properties of Human Breast Milk: MicroRNA Contents and Potential Epigenetic Effects. Biomedicines 2022; 10:1219. [PMID: 35740242 PMCID: PMC9219990 DOI: 10.3390/biomedicines10061219] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 05/15/2022] [Accepted: 05/17/2022] [Indexed: 02/07/2023] Open
Abstract
Infants who are exclusively breastfed in the first six months of age receive adequate nutrients, achieving optimal immune protection and growth. In addition to the known nutritional components of human breast milk (HBM), i.e., water, carbohydrates, fats and proteins, it is also a rich source of microRNAs, which impact epigenetic mechanisms. This comprehensive work presents an up-to-date overview of the immunomodulatory constituents of HBM, highlighting its content of circulating microRNAs. The epigenetic effects of HBM are discussed, especially those regulated by miRNAs. HBM contains more than 1400 microRNAs. The majority of these microRNAs originate from the lactating gland and are based on the remodeling of cells in the gland during breastfeeding. These miRNAs can affect epigenetic patterns by several mechanisms, including DNA methylation, histone modifications and RNA regulation, which could ultimately result in alterations in gene expressions. Therefore, the unique microRNA profile of HBM, including exosomal microRNAs, is implicated in the regulation of the genes responsible for a variety of immunological and physiological functions, such as FTO, INS, IGF1, NRF2, GLUT1 and FOXP3 genes. Hence, studying the HBM miRNA composition is important for improving the nutritional approaches for pregnancy and infant's early life and preventing diseases that could occur in the future. Interestingly, the composition of miRNAs in HBM is affected by multiple factors, including diet, environmental and genetic factors.
Collapse
Affiliation(s)
- Ma’mon M. Hatmal
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Mohammad A. I. Al-Hatamleh
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu 16150, Malaysia;
| | - Amin N. Olaimat
- Department of Clinical Nutrition and Dietetics, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan; (A.N.O.); (M.A.A.-H.)
| | - Walhan Alshaer
- Cell Therapy Center (CTC), The University of Jordan, Amman 11942, Jordan;
| | - Hanan Hasan
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan;
| | - Khaled A. Albakri
- Faculty of Medicine, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Enas Alkhafaji
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, The University of Jordan, Amman 11942, Jordan;
| | - Nada N. Issa
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Murad A. Al-Holy
- Department of Clinical Nutrition and Dietetics, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan; (A.N.O.); (M.A.A.-H.)
| | - Salim M. Abderrahman
- Department of Biology and Biotechnology, Faculty of Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Atiyeh M. Abdallah
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar;
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu 16150, Malaysia;
| |
Collapse
|