1
|
Ham YM, Kang Y, Kang SJ, Lee S, Lee J, Rhee WJ. Advanced Enrichment and Separation of Extracellular Vesicles through the Super Absorbent Polymer Nanosieves. ACS APPLIED MATERIALS & INTERFACES 2024; 16:65863-65876. [PMID: 39560656 DOI: 10.1021/acsami.4c14542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2024]
Abstract
Extracellular vesicles (EVs) are promising therapeutic biomaterials capable of transferring their cargo molecules and external drugs to other cells in vivo and contain various biomarkers that can be used in liquid biopsies. The clinical application of EVs requires an efficient EV enrichment system for the large-scale production or high-throughput isolation of EVs from liquid samples, such as culture media, plant juices, and body fluids. However, current EV enrichment methods, such as ultrafiltration and ultracentrifugation, have limited applicability owing to their associated costs, inefficiency, scalability, and centrifugation time. Herein, we describe the development of a nanosieve based on a superabsorbent polymer for selective EV enrichment. The nanosieve absorbs small molecules while expelling large molecules, such as EVs, through the nanosized channels. We successfully concentrated EVs from clinical samples, such as serum and plasma, with superior cost and time efficiencies. The nanosieves did not interact with the EVs during enrichment, allowing the retention of their therapeutic functions. In addition, the nanosieve surface was specifically engineered to provide multifunctionality to effectively promote EV capture from bulk solutions. Overall, our nanosieve-based EV enrichment method is effective, time- and cost-saving, versatile, scalable, and modulable, and is an excellent option for EV production.
Collapse
Affiliation(s)
- Yoo Min Ham
- Department of Bioengineering and Nano-Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Yubin Kang
- Department of Bioengineering and Nano-Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Su Jin Kang
- Department of Bioengineering and Nano-Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Soobin Lee
- Department of Bioengineering and Nano-Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Jiyoon Lee
- Department of Bioengineering and Nano-Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Won Jong Rhee
- Department of Bioengineering and Nano-Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
- Division of Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
- Research Center for Bio Materials & Process Development, Incheon National University, Incheon 22012, Republic of Korea
| |
Collapse
|
2
|
Gaylord A, Holzhausen EA, Chalifour B, Patterson WB, Tung PW, Baccarelli AA, Goran MI, Alderete TL, Kupsco A. tRNA-derived RNAs in human milk extracellular vesicles and associations with breastfeeding variables and maternal diet. Epigenomics 2024; 16:1429-1441. [PMID: 39580634 PMCID: PMC11622811 DOI: 10.1080/17501911.2024.2430943] [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: 07/10/2024] [Accepted: 11/14/2024] [Indexed: 11/26/2024] Open
Abstract
AIMS To describe tDRs in human milk EVs and their associations with maternal body mass index, age, dietary indices, breastfeeding frequency, season and time of milk collection in a Latina population. MATERIALS & METHODS We sequenced small RNAs from EVs from 109 mature human milk samples collected at 1 month after delivery in the Southern California Mother's Milk Study. We grouped tDRs using hierarchical clustering and clusters were compared across tDR characteristics. We analyzed associations of tDRs with intrinsic maternal variables (body mass index, age), maternal nutrition (caloric intake, Healthy Eating Index, Dietary Inflammatory Index), and variables related to feeding and milk collection (breastfeeding frequency, season and time of milk collection) using negative binomial models. RESULTS We identified 338 tDRs expressed in 90% or more of milk EV samples, of which 113 were identified in all samples. tDR-1:26-Gly-CCC-1-M4 accounted for most reads (79%). Pathway analysis revealed a wide array of biological processes and disease mechanisms across the four tDR clusters. tDRs were associated with season of collection, time of collection, breastfeeding frequency, and the dietary inflammatory index. CONCLUSIONS tDRs are abundant in milk EVs and may be sensitive to maternal diet, seasonality, time of day, and breastfeeding frequency.
Collapse
Affiliation(s)
- Abigail Gaylord
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
| | | | - Bridget Chalifour
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - William B. Patterson
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Pei Wen Tung
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
| | - Andrea A. Baccarelli
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Michael I. Goran
- Department of Pediatrics, Children’s Hospital Los Angeles, Los Angeles, CA, USA
| | - Tanya L. Alderete
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Allison Kupsco
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, USA
| |
Collapse
|
3
|
Cainelli E, Vedovelli L, Bisiacchi P. The mother-child interface: A neurobiological metamorphosis. Neuroscience 2024; 561:92-106. [PMID: 39427701 DOI: 10.1016/j.neuroscience.2024.10.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/30/2024] [Accepted: 10/15/2024] [Indexed: 10/22/2024]
Abstract
From the start of pregnancy, mother and child induce reciprocal neurobiological changes in the brain that will prove critical for neurodevelopment and survival of both. Molecular communication between mother and fetus is constantly active and persists even after the fetus starts to synthesize its hormones in late gestation. Intriguingly, some mother and fetus exchange cells remain in the other's brain and body with long-lasting effects and memories that do not follow the laws of classical genetics but involve complex epigenetic mechanisms. After childbirth, mother and child go through a transitional phase, a sort of limbo in which both will have a peculiar functioning profile, which is adaptive for contingencies but also renders them vulnerable. The interplay between these two "limbo" states allows for an easier transition to the subsequent phases of development. In this review, we will trace mother's and child's path from pregnancy to the months following birth and, in particular, unravel i) the key features of pregnancy and brain development and the reciprocal influences; ii) how a transitory pattern of functioning characterize mother and child, moving them toward more flexible and evolved forms; and iii) how mother and fetus act during childbirth to promote neuroprotection, pain reduction, and neurophysiological changes. Therefore, this review covers a wide range of topics, integrating neuroanatomical, neurological, biochemical, neurophysiological, and psychological studies in a meaningful way, trying to integrate them in a holistic view of the mother-child interface that is usually neglected.
Collapse
Affiliation(s)
- Elisa Cainelli
- Department of General Psychology, University of Padova, 35131 Padova, Italy.
| | - Luca Vedovelli
- Unit of Biostatistics, Epidemiology, and Public Health, Department of Cardiac, Thoracic, Vascular and Public Health Sciences, University of Padova, 35131 Padova, Italy.
| | - Patrizia Bisiacchi
- Department of General Psychology, University of Padova, 35131 Padova, Italy; Padova Neuroscience Center, PNC, 35131 Padova, Italy.
| |
Collapse
|
4
|
Holzhausen EA, Patterson WB, Wong BH, Kim S, Kupsco A, Howe CG, Bode L, Goran MI, Alderete TL. Associations between human milk EV-miRNAs and oligosaccharide concentrations in human milk. Front Immunol 2024; 15:1463463. [PMID: 39635519 PMCID: PMC11614774 DOI: 10.3389/fimmu.2024.1463463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 10/17/2024] [Indexed: 12/07/2024] Open
Abstract
Introduction Human milk contains human milk oligosaccharides (HMOs) and microRNAs (miRNAs), which are key bioactive components. HMOs are indigestible carbohydrates that impact infant growth and development. miRNAs are small, non-coding RNAs that regulate post-transcriptional gene expression. miRNAs are abundant in human milk and can be contained in extracellular vesicles (EVs). There is evidence that miRNAs are synthesized in the mammary epithelium and may influence mammary gland development and milk synthesis. However, the relationships between miRNAs and HMOs have yet to be fully characterized. Methods This study examined the associations between 210 human milk EV-miRNAs and 19 HMOs in a cohort of 98 Latina mothers. HMO measures included summary measures and concentrations of 19 HMOs. Relationships between EV-miRNAs and HMOs were examined using principal components analysis and associations between individual EV-miRNAs and HMOs were assessed. Results Overall patterns of EV-miRNA levels, summarized using principal components, were associated with HMO summary measures and concentrations. Levels of individual EV-miRNAs were associated with HMO summary measures and individual concentrations of 2'FL, 3FL, 3'SL, 6'SL, FLNH, LNFP I, and LNH. Discussion Results from this study suggest that human milk EV-miRNAs are associated with the concentration of HMOs, which may have important effects on infant growth and development.
Collapse
Affiliation(s)
- Elizabeth A. Holzhausen
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - William B. Patterson
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Benjamin H. Wong
- University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Sewan Kim
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Allison Kupsco
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, United States
| | - Caitlin G. Howe
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - 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, San Diego, CA, United States
| | - Michael I. Goran
- Department of Pediatrics, Children’s Hospital Los Angeles, Los Angeles, CA, United States
| | - Tanya L. Alderete
- Department of Environmental Health and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| |
Collapse
|
5
|
Çelik E, Cemali Ö, Şahin TÖ, Deveci G, Biçer NÇ, Hirfanoğlu İM, Ağagündüz D, Budán F. Human Breast Milk Exosomes: Affecting Factors, Their Possible Health Outcomes, and Future Directions in Dietetics. Nutrients 2024; 16:3519. [PMID: 39458514 PMCID: PMC11510026 DOI: 10.3390/nu16203519] [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: 09/21/2024] [Revised: 10/12/2024] [Accepted: 10/14/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Human breast milk is a complex biological fluid containing multifaceted biological compounds that boost immune and metabolic system development that support the short- and long-term health of newborns. Recent literature suggests that human breast milk is a substantial source of nutrients, bioactive molecules, and exosomes. Objectives: This review examines the factors influencing exosomes noted in human milk and the impacts of exosomes on infant health. Furthermore, it discusses potential future prospects for exosome research in dietetics. Methods: Through a narrative review of the existing literature, we focused on exosomes in breast milk, exosome components and their potential impact on exosome health. Results: Exosomes are single-membrane extracellular vesicles of endosomal origin, with an approximate radius of 20-200 nm. They are natural messengers that cells secrete to transport a wide range of diverse cargoes, including deoxyribonucleic acid, ribonucleic acid, proteins, and lipids between various cells. Some studies have reported that the components noted in exosomes in human breast milk could be transferred to the infant and cause epigenetic changes. Thus, it can affect gene expression and cellular event regulation in several tissues. Conclusions: In this manner, exosomes are associated with several pathways, including the immune system, oxidative stress, and cell cycle, and they can affect the short- and long-term health of infants. However, there is still much to learn about the functions, effectiveness, and certain impacts on the health of human breast milk exosomes.
Collapse
Affiliation(s)
- Elif Çelik
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Süleyman Demirel University, Isparta 32260, Türkiye;
| | - Özge Cemali
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Trakya University, Edirne 22030, Türkiye;
| | - Teslime Özge Şahin
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Afyonkarahisar Health Sciences University, Afyonkarahisar 03030, Türkiye;
| | - Gülsüm Deveci
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Çankırı Karatekin University, Çankırı 18100, Türkiye;
| | - Nihan Çakır Biçer
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Acıbadem Mehmet Ali Aydınlar University, Istanbul 34752, Türkiye;
| | | | - Duygu Ağagündüz
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, Ankara 06490, Türkiye
| | - Ferenc Budán
- Institute of Physiology, Medical School, University of Pécs, H-7624 Pécs, Hungary
| |
Collapse
|
6
|
Lu D, Zhang X, Ye H, Wang J, Han D. Milk-Derived Extracellular Vesicles Carrying ssc-let-7 c Alleviate Early Intestinal Inflammation and Regulate Macrophage Polarization via Targeting the PTEN-Mediated PI3K/Akt Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:22092-22104. [PMID: 39188059 DOI: 10.1021/acs.jafc.4c03155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/28/2024]
Abstract
Milk-derived extracellular vesicles (mEVs) are beneficial to the health of infants. However, the effect of mEVs on early intestinal inflammation is not well established. Herein, weaned colitic mice were used to explore the potential effects and underlying mechanisms of porcine mEVs (pmEVs) on intestinal inflammation during early life. We found that pmEVs administration attenuated early life intestinal inflammation and promoted colonic barrier integrity in mice. The anti-inflammatory effect of pmEVs was achieved by shifting a proinflammatory macrophage (M1) toward an anti-inflammatory macrophage (M2). Moreover, pmEVs can be absorbed by macrophages and reduce proinflammatory polarization (stimulated by LPS) in vitro. Noteworthily, ssc-let-7c was found to be highly expressed in pmEVs that can regulate the polarization of macrophages by targeting the tensin homologue deleted on chromosome ten (PTEN), thereby activating the PI3K/Akt pathway. Collectively, our findings revealed a crucial role of mEVs in early intestinal immunity and its underlying mechanism.
Collapse
Affiliation(s)
- Dongdong Lu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
- Adaptation Physiology Group, Department of Animal Science, Wageningen University& Research, Wageningen 6700 AH, The Netherlands
| | - Xiangyu Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Hao Ye
- Adaptation Physiology Group, Department of Animal Science, Wageningen University& Research, Wageningen 6700 AH, The Netherlands
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| | - Dandan Han
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China
| |
Collapse
|
7
|
Albiach-Delgado A, Moreno-Casillas JL, Ten-Doménech I, Cascant-Vilaplana MM, Moreno-Giménez A, Gómez-Ferrer M, Sepúlveda P, Kuligowski J, Quintás G. Oxylipin profile of human milk and human milk-derived extracellular vesicles. Anal Chim Acta 2024; 1313:342759. [PMID: 38862207 DOI: 10.1016/j.aca.2024.342759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/13/2024] [Accepted: 05/20/2024] [Indexed: 06/13/2024]
Abstract
BACKGROUND Small Extracellular Vesicles (sEVs) are nano-sized vesicles that are present in all biofluids including human milk (HM) playing a crucial role in cell-to-cell communication and the stimulation of the neonatal immune system. Oxylipins, which are bioactive lipids formed from polyunsaturated fatty acids, have gained considerable attention due to their potential role in mitigating disease progression and modulating the inflammatory status of breastfed infants. This study aims at an in-depth characterization of the oxylipin profiles of HM and, for the first time, of HM-derived sEVs (HMEVs) employing an ad-hoc developed and validated ultra-performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS) method. RESULTS The UPLC-MS/MS method covered a panel of 13 oxylipins for quantitation and 93 oxylipins for semi-quantitation. In 200 μL of HM and HMEV isolates of 15 individuals, 42 out of 106 oxylipins were detected in either HM or HMEVs, with 38 oxylipins being detected in both matrices. Oxylipins presented distinct profiles in HM and HMEVs, suggesting specific mechanisms responsible for the encapsulation of target molecules in HMEVs. Ten and eight oxylipins were quantified with ranges between 0.03 - 73 nM and 0.30 pM-0.07 nM in HM and HMEVs, respectively. The most abundant oxylipins found in HMEVs were docosahexaenoic acid derivatives (17-HDHA and 14-HDHA) with known anti-inflammatory properties, and linoleic acid derivatives (9-10-DiHOME and 12,13-DiHOME) in HM samples. SIGNIFICANCE AND NOVELTY This is the first time a selective, relative enrichment of anti-inflammatory oxylipins in HMEVs has been described. Future studies will focus on the anti-inflammatory and pro-healing capacity of oxylipins encapsulated in HMEVs, with potential clinical applications in the field of preterm infant care, specifically the prevention of severe intestinal complications including necrotizing enterocolitis.
Collapse
Affiliation(s)
- Abel Albiach-Delgado
- Neonatal Research Group, Health Research Institute Hospital La Fe (IIS La Fe), Avda Fernando Abril Martorell 106, 46026, Valencia, Spain; Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Developmental Origin Network (RICORS-SAMID) (RD21/0012/0015), Instituto de Salud Carlos III, Madrid, Spain; Servicio de Análisis de Vesículas Extracelulares (SAVE), Health Research Institute Hospital La Fe (IIS La Fe), Avda Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Jose L Moreno-Casillas
- Neonatal Research Group, Health Research Institute Hospital La Fe (IIS La Fe), Avda Fernando Abril Martorell 106, 46026, Valencia, Spain; Servicio de Análisis de Vesículas Extracelulares (SAVE), Health Research Institute Hospital La Fe (IIS La Fe), Avda Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Isabel Ten-Doménech
- Neonatal Research Group, Health Research Institute Hospital La Fe (IIS La Fe), Avda Fernando Abril Martorell 106, 46026, Valencia, Spain; Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Developmental Origin Network (RICORS-SAMID) (RD21/0012/0015), Instituto de Salud Carlos III, Madrid, Spain; Servicio de Análisis de Vesículas Extracelulares (SAVE), Health Research Institute Hospital La Fe (IIS La Fe), Avda Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Mari Merce Cascant-Vilaplana
- Neonatal Research Group, Health Research Institute Hospital La Fe (IIS La Fe), Avda Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Alba Moreno-Giménez
- Neonatal Research Group, Health Research Institute Hospital La Fe (IIS La Fe), Avda Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Marta Gómez-Ferrer
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avda Fernando Abril Martorell 106, 46026, Valencia, Spain
| | - Pilar Sepúlveda
- Regenerative Medicine and Heart Transplantation Unit, Health Research Institute Hospital La Fe (IIS La Fe), Avda Fernando Abril Martorell 106, 46026, Valencia, Spain; Centro de Investigación Biomédica en Red Enfermedades Cardiovasculares (CIBERCV), Carlos III Institute of Health, Madrid, Spain; Cardiology Service, University & Polytechnic Hospital La Fe, Avenida Fernando Abril Martorell 106, 46026, Valencia, Spain; Department of Pathology, University of Valencia, Avenida Blasco Ibáñez 15, 46010, Valencia, Spain.
| | - Julia Kuligowski
- Neonatal Research Group, Health Research Institute Hospital La Fe (IIS La Fe), Avda Fernando Abril Martorell 106, 46026, Valencia, Spain; Primary Care Interventions to Prevent Maternal and Child Chronic Diseases of Perinatal and Developmental Origin Network (RICORS-SAMID) (RD21/0012/0015), Instituto de Salud Carlos III, Madrid, Spain; Servicio de Análisis de Vesículas Extracelulares (SAVE), Health Research Institute Hospital La Fe (IIS La Fe), Avda Fernando Abril Martorell 106, 46026, Valencia, Spain.
| | - Guillermo Quintás
- Health and Biomedicine, Leitat Technological Center, Avda Fernando Abril Martorell 106, 46026, Valencia, Spain
| |
Collapse
|
8
|
Neettiyath A, Chung K, Liu W, Lee LP. Nanoplasmonic sensors for extracellular vesicles and bacterial membrane vesicles. NANO CONVERGENCE 2024; 11:23. [PMID: 38918255 PMCID: PMC11199476 DOI: 10.1186/s40580-024-00431-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 06/03/2024] [Indexed: 06/27/2024]
Abstract
Extracellular vesicles (EVs) are promising tools for the early diagnosis of diseases, and bacterial membrane vesicles (MVs) are especially important in health and environment monitoring. However, detecting EVs or bacterial MVs presents significant challenges for the clinical translation of EV-based diagnostics. In this Review, we provide a comprehensive discussion on the basics of nanoplasmonic sensing and emphasize recent developments in nanoplasmonics-based optical sensors to effectively identify EVs or bacterial MVs. We explore various nanoplasmonic sensors tailored for EV or bacterial MV detection, emphasizing the application of localized surface plasmon resonance through gold nanoparticles and their multimers. Additionally, we highlight advanced EV detection techniques based on surface plasmon polaritons using plasmonic thin film and nanopatterned structures. Furthermore, we evaluate the improved detection capability of surface-enhanced Raman spectroscopy in identifying and classifying these vesicles, aided by plasmonic nanostructures. Nanoplasmonic sensing techniques have remarkable precision and sensitivity, making them a potential tool for accurate EV detection in clinical applications, facilitating point-of-care molecular diagnostics. Finally, we summarize the challenges associated with nanoplasmonic EV or bacterial MV sensors and offer insights into potential future directions for this evolving field.
Collapse
Affiliation(s)
- Aparna Neettiyath
- Renal Division and Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
- Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Kyungwha Chung
- Renal Division and Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
- Harvard Medical School, Harvard University, Boston, MA 02115, USA
- Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, Suwon 16419, Korea
| | - Wenpeng Liu
- Renal Division and Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
- Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Luke P Lee
- Renal Division and Division of Engineering in Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA.
- Harvard Medical School, Harvard University, Boston, MA 02115, USA.
- Department of Bioengineering, University of California, Berkeley, CA 94720, USA.
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, CA 94720, USA.
- Institute of Quantum Biophysics, Department of Biophysics, Sungkyunkwan University, Suwon 16419, Korea.
- Department of Chemistry and Nano Science, Ewha Womans University, Seoul 03760, Korea.
| |
Collapse
|
9
|
Ma L, Huo Y, Tang Q, Wang X, Wang W, Wu D, Li Y, Chen L, Wang S, Zhu Y, Wang W, Liu Y, Xu N, Chen L, Yu G, Chen J. Human Breast Milk Exosomal miRNAs are Influenced by Premature Delivery and Affect Neurodevelopment. Mol Nutr Food Res 2024; 68:e2300113. [PMID: 38644336 DOI: 10.1002/mnfr.202300113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 02/27/2024] [Indexed: 04/23/2024]
Abstract
SCOPE This study investigates the exosomal microRNA (miRNA) profiles of term and preterm breast milk, including the most abundant and differentially expressed (DE) miRNAs, and their impact on neurodevelopment in infants. METHODS AND RESULTS Mature milk is collected from the mothers of term and preterm infants. Using high-throughput sequencing and subsequent data analysis, exosomal miRNA profiles of term and preterm human breast milk (HBM) are acquired and it is found that the let-7 and miR-148 families are the most abundant miRNAs. Additionally, 23 upregulated and 15 downregulated miRNAs are identified. MiR-3168 is the most upregulated miRNA in preterm HBM exosome, exhibiting targeting activity toward multiple genes involved in the SMAD and MAPK signaling pathways and playing a crucial role in early neurodevelopment. Additionally, the effects of miR-3168 on neurodevelopment is confirmed and it is determined that it is an essential factor in the differentiation of neural stem cells (NSCs). CONCLUSION This study demonstrates that miRNA expression in breast milk exosomes can be influenced by preterm delivery, thereby potentially impacting neurodevelopment in preterm infants.
Collapse
Affiliation(s)
- Ling Ma
- Department of Child Health Care, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Yanyan Huo
- Department of Child Health Care, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Qingyuan Tang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiulian Wang
- Department of Child Health Care, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Weiqin Wang
- Department of Child Health Care, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Dan Wu
- Department of Child Health Care, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Yicheng Li
- Department of Child Health Care, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Lingyan Chen
- Department of Child Health Care, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China
- Department of Occupational Therapy Science, Nagasaki University Graduate School of Biomedical Science, 1-7-1 Sakamoto, Nagasaki, 852-8520, Japan
| | - Shasha Wang
- Department of Child Health Care, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China
| | - Yiwen Zhu
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wenli Wang
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yuan Liu
- Department of Food Science & Technology, School of Agriculture & Biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Nanjie Xu
- Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Li Chen
- Department of Orthodontics, Shanghai Stomatological Hospital & School of Stomatology, Fudan University, Shanghai, 200001, China
| | - Guangjun Yu
- School of Public Health, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jinjin Chen
- Department of Child Health Care, Shanghai Children's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200062, China
| |
Collapse
|
10
|
Lu D, Liu Y, Kang L, Zhang X, Hu J, Ye H, Huang B, Wu Y, Zhao J, Dai Z, Wang J, Han D. Maternal fiber-rich diet promotes early-life intestinal development in offspring through milk-derived extracellular vesicles carrying miR-146a-5p. J Nanobiotechnology 2024; 22:65. [PMID: 38365722 PMCID: PMC10870446 DOI: 10.1186/s12951-024-02344-4] [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: 12/04/2023] [Accepted: 02/12/2024] [Indexed: 02/18/2024] Open
Abstract
BACKGROUNDS The intestinal development in early life is profoundly influenced by multiple biological components of breast milk, in which milk-derived extracellular vesicles (mEVs) contain a large amount of vertically transmitted signal from the mother. However, little is known about how maternal fiber-rich diet regulates offspring intestinal development by influencing the mEVs. RESULTS In this study, we found that maternal resistant starch (RS) consumption during late gestation and lactation improved the growth and intestinal health of offspring. The mEVs in breast milk are the primary factor driving these beneficial effects, especially enhancing intestinal cell proliferation and migration. To be specific, administration of mEVs after maternal RS intake enhanced intestinal cell proliferation and migration in vivo (performed in mice model and indicated by intestinal histological observation, EdU assay, and the quantification of cyclin proteins) and in vitro (indicated by CCK8, MTT, EdU, and wound healing experiments). Noteworthily, miR-146a-5p was found to be highly expressed in the mEVs from maternal RS group, which also promotes intestinal cell proliferation in cells and mice models. Mechanically, miR-146a-5p target to silence the expression of ubiquitin ligase 3 gene NEDD4L, thereby inhibiting DVL2 ubiquitination, activating the Wnt pathway, and promoting intestinal development. CONCLUSION These findings demonstrated the beneficial role of mEVs in the connection between maternal fiber rich diet and offspring intestinal growth. In addition, we identified a novel miRNA-146a-5p-NEDD4L-β-catenin/Wnt signaling axis in regulating early intestinal development. This work provided a new perspective for studying the influence of maternal diet on offspring development.
Collapse
Affiliation(s)
- Dongdong Lu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
- Adaptation Physiology Group, Wageningen University & Research, Wageningen, 6700 AH, The Netherlands
| | - Yisi Liu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Luyuan Kang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Xiangyu Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jie Hu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Hao Ye
- Adaptation Physiology Group, Wageningen University & Research, Wageningen, 6700 AH, The Netherlands
| | - Bingxu Huang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Yujun Wu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Jinbiao Zhao
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Zhaolai Dai
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Junjun Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China
| | - Dandan Han
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
11
|
Yung C, Zhang Y, Kuhn M, Armstrong RJ, Olyaei A, Aloia M, Scottoline B, Andres SF. Neonatal enteroids absorb extracellular vesicles from human milk-fed infant digestive fluid. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.03.556067. [PMID: 38187651 PMCID: PMC10769189 DOI: 10.1101/2023.09.03.556067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Human milk contains extracellular vesicles (HMEVs). Pre-clinical models suggest that HMEVs may enhance intestinal function and limit inflammation; however, it is unknown if HMEVs or their cargo survive neonatal human digestion. This limits the ability to leverage HMEV cargo as additives to infant nutrition or as therapeutics. This study aimed to develop an EV isolation pipeline from small volumes of human milk and neonatal intestinal contents after milk feeding (digesta) to address the hypothesis that HMEVs survive in vivo neonatal digestion to be taken up intestinal epithelial cells (IECs). Digesta was collected from nasoduodenal sampling tubes or ostomies. EVs were isolated from raw and pasteurized human milk and digesta by density-gradient ultracentrifugation following two-step skimming, acid precipitation of caseins, and multi-step filtration. EVs were validated by electron microscopy, western blotting, nanoparticle tracking analysis, resistive pulse sensing, and super-resolution microscopy. EV uptake was tested in human neonatal enteroids. HMEVs and digesta EVs (dEVs) show typical EV morphology and are enriched in CD81 and CD9, but depleted of β-casein and lactalbumin. HMEV and some dEV fractions contain mammary gland-derived protein BTN1A1. Neonatal human enteroids rapidly take up dEVs in part via clathrin-mediated endocytosis. Our data suggest that EVs can be isolated from digestive fluid and that these dEVs can be absorbed by IECs.
Collapse
|
12
|
Muse ME, Armstrong DA, Hoen AG, Gilbert-Diamond D, Gui J, Palys TJ, Kolling FW, Christensen BC, Karagas MR, Howe CG. Maternal-Infant Factors in Relation to Extracellular Vesicle and Particle miRNA in Prenatal Plasma and in Postpartum Human Milk. Int J Mol Sci 2024; 25:1538. [PMID: 38338815 PMCID: PMC10855220 DOI: 10.3390/ijms25031538] [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: 11/22/2023] [Revised: 01/19/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
MicroRNAs (miRNA) in extracellular vesicles and particles (EVPs) in maternal circulation during pregnancy and in human milk postpartum are hypothesized to facilitate maternal-offspring communication via epigenetic regulation. However, factors influencing maternal EVP miRNA profiles during these two critical developmental windows remain largely unknown. In a pilot study of 54 mother-child dyads in the New Hampshire Birth Cohort Study, we profiled 798 EVP miRNAs, using the NanoString nCounter platform, in paired maternal second-trimester plasma and mature (6-week) milk samples. In adjusted models, total EVP miRNA counts were lower for plasma samples collected in the afternoon compared with the morning (p = 0.024). Infant age at sample collection was inversely associated with total miRNA counts in human milk EVPs (p = 0.040). Milk EVP miRNA counts were also lower among participants who were multiparous after delivery (p = 0.047), had a pre-pregnancy BMI > 25 kg/m2 (p = 0.037), or delivered their baby via cesarean section (p = 0.021). In post hoc analyses, we also identified 22 specific EVP miRNA that were lower among participants who delivered their baby via cesarean section (Q < 0.05). Target genes of delivery mode-associated miRNAs were over-represented in pathways related to satiety signaling in infants (e.g., CCKR signaling) and mammary gland development and lactation (e.g., FGF signaling, EGF receptor signaling). In conclusion, we identified several key factors that may influence maternal EVP miRNA composition during two critical developmental windows, which should be considered in future studies investigating EVP miRNA roles in maternal and child health.
Collapse
Affiliation(s)
- Meghan E. Muse
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, 1 Medical Center Dr, Lebanon, NH 03755, USA (M.R.K.); (C.G.H.)
| | - David A. Armstrong
- Research Service, V.A. Medical Center, Hartford, VT 05009, USA
- Department of Dermatology, Dartmouth Health, Lebanon, NH 03756, USA
| | - Anne G. Hoen
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, 1 Medical Center Dr, Lebanon, NH 03755, USA (M.R.K.); (C.G.H.)
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Diane Gilbert-Diamond
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, 1 Medical Center Dr, Lebanon, NH 03755, USA (M.R.K.); (C.G.H.)
| | - Jiang Gui
- Department of Biomedical Data Science, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Thomas J. Palys
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, 1 Medical Center Dr, Lebanon, NH 03755, USA (M.R.K.); (C.G.H.)
| | - Frederick W. Kolling
- Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Brock C. Christensen
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, 1 Medical Center Dr, Lebanon, NH 03755, USA (M.R.K.); (C.G.H.)
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, USA
| | - Margaret R. Karagas
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, 1 Medical Center Dr, Lebanon, NH 03755, USA (M.R.K.); (C.G.H.)
| | - Caitlin G. Howe
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, 1 Medical Center Dr, Lebanon, NH 03755, USA (M.R.K.); (C.G.H.)
| |
Collapse
|
13
|
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
|
14
|
Froń A, Orczyk-Pawiłowicz M. Understanding the Immunological Quality of Breast Milk in Maternal Overweight and Obesity. Nutrients 2023; 15:5016. [PMID: 38140275 PMCID: PMC10746120 DOI: 10.3390/nu15245016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 11/30/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
Maternal obesity, affecting many pregnant women globally, not only poses immediate health risks but also modulates breast milk composition. Obesity is linked to inflammation and oxidative stress, impacting breast milk's immune properties. This paper explores the intricate relationship between maternal metabolic disorders, such as obesity, and breast milk's immunological components. We conducted a thorough search for original and review articles published until 17 October 2023 in the PUBMED/Scopus database. This search included several terms related to human breast milk, immunological properties, and obesity. Articles were selected with the consensus of all authors. Maternal metabolic disorders have discernible effects on the composition of immune-related components in breast milk, such as immunoglobulins, lactoferrin, leptin, ghrelin, adiponectin, C-reactive protein, growth factors, extracellular vesicles, and lymphocytes. These changes in breast milk composition can significantly impact the newborn's immune system, with potential long-term health implications beyond the immediate postnatal period. Maternal metabolic health is a critical factor in shaping the health trajectory of the neonate through breastfeeding, although the full advantages of breastfeeding for children of mothers with obesity remain uncertain. Ongoing research aims to understand and unravel these links.
Collapse
Affiliation(s)
- Anita Froń
- Division of Chemistry and Immunochemistry, Department of Biochemistry and Immunochemistry, Wroclaw Medical University, M. Skłodowskiej-Curie 48/50, 50-369 Wroclaw, Poland
| | - Magdalena Orczyk-Pawiłowicz
- Division of Chemistry and Immunochemistry, Department of Biochemistry and Immunochemistry, Wroclaw Medical University, M. Skłodowskiej-Curie 48/50, 50-369 Wroclaw, Poland
| |
Collapse
|
15
|
Kaeffer B. Human Breast Milk miRNAs: Their Diversity and Potential for Preventive Strategies in Nutritional Therapy. Int J Mol Sci 2023; 24:16106. [PMID: 38003296 PMCID: PMC10671413 DOI: 10.3390/ijms242216106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/02/2023] [Accepted: 11/06/2023] [Indexed: 11/26/2023] Open
Abstract
The endogenous miRNAs of breast milk are the products of more than 1000 nonprotein-coding genes, giving rise to mature small regulatory molecules of 19-25 nucleotides. They are incorporated in macromolecular complexes, loaded on Argonaute proteins, sequestrated in exosomes and lipid complexes, or present in exfoliated cells of epithelial, endothelial, or immune origins. Their expression is dependent on the stage of lactation; however, their detection depends on progress in RNA sequencing and the reappraisal of the definition of small RNAs. Some miRNAs from plants are detected in breast milk, opening the possibility of the stimulation of immune cells from the allergy repertoire. Each miRNA harbors a seeding sequence, which targets mRNAs, gene promoters, or long noncoding RNAs. Their activities depend on their bioavailability. Efficient doses of miRNAs are estimated to be roughly 100 molecules in the cytoplasm of target cells from in vitro and in vivo experiments. Each miRNA is included in networks of stimulation/inhibition/sequestration, driving the expression of cellular phenotypes. Three types of stress applied during lactation to manipulate miRNA supply were explored using rodent offspring: a foster mother, a cafeteria diet, and early weaning. This review presents the main mature miRNAs described from current mothers' cohorts and their bioavailability in experimental models as well as studies assessing the potential of miR-26 or miR-320 miRNA families to alter offspring phenotypes.
Collapse
Affiliation(s)
- Bertrand Kaeffer
- Nantes Université, INRAE, UMR 1280, PhAN, F-44000 Nantes, France
| |
Collapse
|
16
|
Holzhausen EA, Kupsco A, Chalifour BN, Patterson WB, Schmidt KA, Mokhtari P, Lurmann F, Baccarelli AA, Goran MI, Alderete TL. Human milk EV-miRNAs: a novel biomarker for air pollution exposure during pregnancy. ENVIRONMENTAL RESEARCH, HEALTH : ERH 2023; 1:035002. [PMID: 37692372 PMCID: PMC10486183 DOI: 10.1088/2752-5309/ace075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 05/23/2023] [Accepted: 06/21/2023] [Indexed: 09/12/2023]
Abstract
Exposure to ambient and near-roadway air pollution during pregnancy has been linked with several adverse health outcomes for pregnant women and their babies. Emerging research indicates that microRNA (miRNA) expression can be altered by exposure to air pollutants in a variety of tissues. Additionally, miRNAs from breast tissue and circulating miRNAs have previously been proposed as a biomarker for breast cancer diagnosis and prognosis. Therefore, this study sought to evaluate the associations between pregnancy exposures to ambient (PM10, PM2.5, NO2, O3) and near-roadway air pollution (total NOx, freeway NOx, non-freeway NOx) with breast milk extracellular vesicle miRNA (EV-miRNA), measured at 1-month postpartum, in a cohort of 108 Latina women living in Southern California. We found that PM10 exposure during pregnancy was positively associated with hsa-miR-200c-3p, hsa-miR-200b-3p, and hsa-let-7c-5p, and was negatively associated with hsa-miR-378d. We also found that pregnancy PM2.5 exposure was positively associated with hsa-miR-200c-3p and hsa-miR-200b-3p. First and second trimester exposure to PM10 and PM2.5 was associated with several EV-miRNAs with putative messenger RNA targets related to cancer. This study provides preliminary evidence that air pollution exposure during pregnancy is associated with human milk EV-miRNA expression.
Collapse
Affiliation(s)
- Elizabeth A Holzhausen
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO,
United States of America
| | - Allison Kupsco
- Department of Environmental Health Sciences, Columbia University Mailman School of Public
Health, New York, NY, United States of America
| | - Bridget N Chalifour
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO,
United States of America
| | - William B Patterson
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO,
United States of America
| | - Kelsey A Schmidt
- Department of Pediatrics, Children’s Hospital Los Angeles, Los Angeles, CA, United
States of America
| | - Pari Mokhtari
- Department of Pediatrics, Children’s Hospital Los Angeles, Los Angeles, CA, United
States of America
| | | | - Andrea A Baccarelli
- Department of Environmental Health Sciences, Columbia University Mailman School of Public
Health, New York, NY, United States of America
| | - Michael I Goran
- Department of Pediatrics, Children’s Hospital Los Angeles, Los Angeles, CA, United
States of America
| | - Tanya L Alderete
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO,
United States of America
| |
Collapse
|
17
|
Gialeli G, Panagopoulou O, Liosis G, Siahanidou T. Potential Epigenetic Effects of Human Milk on Infants' Neurodevelopment. Nutrients 2023; 15:3614. [PMID: 37630804 PMCID: PMC10460013 DOI: 10.3390/nu15163614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Revised: 08/12/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
The advantages of human milk feeding, especially in preterm babies, are well recognized. Infants' feeding with breast milk lowers the likelihood of developing a diverse range of non-communicable diseases later in life and it is also associated with improved neurodevelopmental outcomes. Although the precise mechanisms through which human milk feeding is linked with infants' neurodevelopment are still unknown, potential epigenetic effects of breast milk through its bioactive components, including non-coding RNAs, stem cells and microbiome, could at least partly explain this association. Micro- and long-non-coding RNAs, enclosed in milk exosomes, as well as breast milk stem cells, survive digestion, reach the circulation and can cross the blood-brain barrier. Certain non-coding RNAs potentially regulate genes implicated in brain development and function, whereas nestin-positive stem cells can possibly differentiate into neural cells or/and act as epigenetic regulators in the brain. Furthermore, breast milk microbiota contributes to the establishment of infant's gut microbiome, which is implicated in brain development via epigenetic modifications and key molecules' regulation. This narrative review provides an updated analysis of the relationship between breast milk feeding and infants' neurodevelopment via epigenetics, pointing out how breast milk's bioactive components could have an impact on the neurodevelopment of both full-term and preterm babies.
Collapse
Affiliation(s)
- Giannoula Gialeli
- First Department of Pediatrics, Medical School, National & Kapodistrian University of Athens, 11527 Athens, Greece; (G.G.); (O.P.)
| | - Ourania Panagopoulou
- First Department of Pediatrics, Medical School, National & Kapodistrian University of Athens, 11527 Athens, Greece; (G.G.); (O.P.)
| | - Georgios Liosis
- Neonatal Intensive Care Unit, “Elena Venizelou” General and Maternal Hospital, 11521 Athens, Greece;
| | - Tania Siahanidou
- First Department of Pediatrics, Medical School, National & Kapodistrian University of Athens, 11527 Athens, Greece; (G.G.); (O.P.)
| |
Collapse
|
18
|
Holzhausen EA, Kupsco A, Chalifour BN, Patterson WB, Schmidt KA, Mokhtari P, Baccarelli AA, Goran MI, Alderete TL. Influence of technical and maternal-infant factors on the measurement and expression of extracellular miRNA in human milk. Front Immunol 2023; 14:1151870. [PMID: 37492577 PMCID: PMC10363855 DOI: 10.3389/fimmu.2023.1151870] [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: 01/26/2023] [Accepted: 06/09/2023] [Indexed: 07/27/2023] Open
Abstract
Breast milk contains thousands of bioactive compounds including extracellular vesicle microRNAs (EV-miRNAs), which may regulate pathways such as infant immune system development and metabolism. We examined the associations between the expression of EV-miRNAs and laboratory variables (i.e., batch effects, sample characteristics), sequencing quality indicators, and maternal-infant characteristics. The study included 109 Latino mother-infant dyads from the Southern California Mother's Milk Study. Mothers were age 28.0 ± 5.6 and 23-46 days postpartum. We used principal components analysis to evaluate whether EV-miRNA expression was associated with factors of interest. Then, we used linear models to estimate relationships between these factors and specific EV-miRNA counts and analyzed functional pathways associated with those EV-miRNAs. Finally, we explored which maternal-infant characteristics predicted sequencing quality indicators. Sequencing quality indicators, predominant breastfeeding, and breastfeedings/day were associated with EV-miRNA principal components. Maternal body mass index and breast milk collection timing predicted proportion of unmapped reads. Expression of 2 EV-miRNAs were associated with days postpartum, 23 EV-miRNAs were associated with breast milk collection time, 23 EV-miRNAs were associated with predominant breastfeeding, and 38 EV-miRNAs were associated with breastfeedings/day. These EV-miRNAs were associated with pathways including Hippo signaling pathway and ECM-receptor interaction, among others. This study identifies several important factors that may contribute to breast milk EV-miRNA expression. Future studies should consider these findings in the design and analysis of breast milk miRNA research.
Collapse
Affiliation(s)
- Elizabeth A. Holzhausen
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Allison Kupsco
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, United States
| | - Bridget N. Chalifour
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - William B. Patterson
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| | - Kelsey A. Schmidt
- Department of Pediatrics, Children’s Hospital Los Angeles, Los Angeles, CA, United States
| | - Pari Mokhtari
- Department of Pediatrics, Children’s Hospital Los Angeles, Los Angeles, CA, United States
| | - Andrea A. Baccarelli
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY, United States
| | - Michael I. Goran
- Department of Pediatrics, Children’s Hospital Los Angeles, Los Angeles, CA, United States
| | - Tanya L. Alderete
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, United States
| |
Collapse
|
19
|
Reniker LN, Frazer LC, Good M. Key biologically active components of breast milk and their beneficial effects. Semin Pediatr Surg 2023; 32:151306. [PMID: 37276783 PMCID: PMC10330649 DOI: 10.1016/j.sempedsurg.2023.151306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Maternal breast milk is the penultimate nutritional source for term and preterm neonates. Its composition is highly complex and includes multiple factors that enhance the development of nearly every neonatal organ system leading to both short- and long-term health benefits. Intensive research is focused on identifying breast milk components that enhance infant health. However, this research is complicated by the significant impact of maternal factors and the processing of pumped breast milk on bioactive ingredients. Optimizing enteral nutrition is particularly important for preterm neonates who miss the transplacental acquisition of nutrients in the third trimester of pregnancy and are at risk for illnesses associated with gut barrier dysfunction, including sepsis and necrotizing enterocolitis. In this review, we will discuss the health benefits of breast milk and its bioactive components.
Collapse
Affiliation(s)
- Laura N Reniker
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA, 27599
| | - Lauren C Frazer
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA, 27599
| | - Misty Good
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA, 27599.
| |
Collapse
|
20
|
Maqueda JJ, Giovanazzi A, Rocha AM, Rocha S, Silva I, Saraiva N, Bonito N, Carvalho J, Maia L, Wauben MHM, Oliveira C. Adapter dimer contamination in sRNA-sequencing datasets predicts sequencing failure and batch effects and hampers extracellular vesicle-sRNA analysis. JOURNAL OF EXTRACELLULAR BIOLOGY 2023; 2:e91. [PMID: 38938917 PMCID: PMC11080836 DOI: 10.1002/jex2.91] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/21/2023] [Accepted: 05/10/2023] [Indexed: 06/29/2024]
Abstract
Small RNA (sRNA) profiling of Extracellular Vesicles (EVs) by Next-Generation Sequencing (NGS) often delivers poor outcomes, independently of reagents, platforms or pipelines used, which contributes to poor reproducibility of studies. Here we analysed pre/post-sequencing quality controls (QC) to predict issues potentially biasing biological sRNA-sequencing results from purified human milk EVs, human and mouse EV-enriched plasma and human paraffin-embedded tissues. Although different RNA isolation protocols and NGS platforms were used in these experiments, all datasets had samples characterized by a marked removal of reads after pre-processing. The extent of read loss between individual samples within a dataset did not correlate with isolated RNA quantity or sequenced base quality. Rather, cDNA electropherograms revealed the presence of a constant peak whose intensity correlated with the degree of read loss and, remarkably, with the percentage of adapter dimers, which were found to be overrepresented sequences in high read-loss samples. The analysis through a QC pipeline, which allowed us to monitor quality parameters in a step-by-step manner, provided compelling evidence that adapter dimer contamination was the main factor causing batch effects. We concluded this study by summarising peer-reviewed published workflows that perform consistently well in avoiding adapter dimer contamination towards a greater likelihood of sequencing success.
Collapse
Affiliation(s)
- Joaquín J. Maqueda
- BIOINF2BIO, LDAPortoPortugal
- i3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
- Ipatimup – Institute of Molecular Pathology and Immunology of the University of PortoPortoPortugal
| | - Alberta Giovanazzi
- Department of Biomolecular Health SciencesFaculty of Veterinary Medicine Utrecht UniversityUtrechtThe Netherlands
| | - Ana Mafalda Rocha
- i3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
- Ipatimup – Institute of Molecular Pathology and Immunology of the University of PortoPortoPortugal
| | - Sara Rocha
- i3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
- Ipatimup – Institute of Molecular Pathology and Immunology of the University of PortoPortoPortugal
| | - Isabel Silva
- i3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
- IBMC ‐ Instituto de Biologia Molecular e CelularUniversity of PortoPortoPortugal
| | - Nadine Saraiva
- IPOC – Instituto Português de Oncologia Francisco GentilCoimbraPortugal
| | - Nuno Bonito
- IPOC – Instituto Português de Oncologia Francisco GentilCoimbraPortugal
| | - Joana Carvalho
- i3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
- Ipatimup – Institute of Molecular Pathology and Immunology of the University of PortoPortoPortugal
| | - Luis Maia
- i3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
- ICBAS‐UP ‐ Instituto de Ciências Biomédicas Abel SalazarUniversity of PortoPortoPortugal
- CHUPorto – Department of NeurologyCentro Hospitalar Universitário do PortoPortoPortugal
| | - Marca H. M. Wauben
- Department of Biomolecular Health SciencesFaculty of Veterinary Medicine Utrecht UniversityUtrechtThe Netherlands
| | - Carla Oliveira
- BIOINF2BIO, LDAPortoPortugal
- i3S – Instituto de Investigação e Inovação em SaúdeUniversidade do PortoPortoPortugal
- Ipatimup – Institute of Molecular Pathology and Immunology of the University of PortoPortoPortugal
- FMUP – Faculty of MedicineUniversity of PortoPortoPortugal
| |
Collapse
|
21
|
Abbas MA, Al-Saigh NN, Saqallah FG. Regulation of adipogenesis by exosomal milk miRNA. Rev Endocr Metab Disord 2023; 24:297-316. [PMID: 36692804 DOI: 10.1007/s11154-023-09788-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/12/2023] [Indexed: 01/25/2023]
Abstract
Milk is a rich source of miRNA packaged in exosomes. Evidence for the systemic uptake and tissue distribution of milk exosomes was reported in newborn and adult humans and animals. Breastfeeding in infants was associated with a reduced risk of obesity. Numerous adipogenesis-related miRNAs have been detected in human milk exosomes. It has been demonstrated that ingested exosomal milk miRNAs may alter gene expression in offspring to regulate their metabolism and growth. In humans, consumption of other species' milk, such as cows and goats, is continued through adulthood. Since miRNAs are conserved, the concern of cross-species transfer of adipogenic miRNA has been raised in recent years, and the increase in obesity worldwide was attributed partially to dairy milk consumption by humans. However, evidence is still weak. Research emphasizes the need for an adequate number of exosomal milk's miRNAs to reach the target cell for biological action to be achieved. It was reported that obese women's milk had less miRNA-148a and miRNA-30b, which may affect the fat acquisition of their babies. Some exosomal milk miRNAs, such as miRNA-29, miRNA-148, miRNA-30b and miRNA-125b, may have epigenetic effects on milk recipients. Moreover, the ability of milk exosomes to cross the gastrointestinal barrier makes them a promising oral drug delivery tool. Yet, exosomes may also be tagged with specific ligands which target certain tissues. Thus, milk exosomes can be engineered and loaded with certain miRNAs responsible for adipocyte differentiation, conversion, or browning. Modifications in the miRNA cargo of exosomes can benefit human health and be an alternative to traditional drugs.
Collapse
Affiliation(s)
- Manal A Abbas
- Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, 19328, Jordan.
- Pharmacological and Diagnostic Research Center, Al-Ahliyya Amman University, Amman, 19328, Jordan.
| | - Noor Nadhim Al-Saigh
- Department of Basic Medical Sciences, Faculty of Medicine, Ibn Sina University for Medical Siences, Amman, 11104, Jordan
| | - Fadi G Saqallah
- Discipline of Pharmaceutical Chemistry, School of Pharmaceutical Sciences, Universiti Sains Malaysia, 11800, Penang, Malaysia
| |
Collapse
|
22
|
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
|
23
|
Spiers HVM, Stadler LKJ, Smith H, Kosmoliaptsis V. Extracellular Vesicles as Drug Delivery Systems in Organ Transplantation: The Next Frontier. Pharmaceutics 2023; 15:891. [PMID: 36986753 PMCID: PMC10052210 DOI: 10.3390/pharmaceutics15030891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/12/2023] Open
Abstract
Extracellular vesicles are lipid bilayer-delimited nanoparticles excreted into the extracellular space by all cells. They carry a cargo rich in proteins, lipids and DNA, as well as a full complement of RNA species, which they deliver to recipient cells to induce downstream signalling, and they play a key role in many physiological and pathological processes. There is evidence that native and hybrid EVs may be used as effective drug delivery systems, with their intrinsic ability to protect and deliver a functional cargo by utilising endogenous cellular mechanisms making them attractive as therapeutics. Organ transplantation is the gold standard for treatment for suitable patients with end-stage organ failure. However, significant challenges still remain in organ transplantation; prevention of graft rejection requires heavy immunosuppression and the lack of donor organs results in a failure to meet demand, as manifested by growing waiting lists. Pre-clinical studies have demonstrated the ability of EVs to prevent rejection in transplantation and mitigate ischemia reperfusion injury in several disease models. The findings of this work have made clinical translation of EVs possible, with several clinical trials actively recruiting patients. However, there is much to be uncovered, and it is essential to understand the mechanisms behind the therapeutic benefits of EVs. Machine perfusion of isolated organs provides an unparalleled platform for the investigation of EV biology and the testing of the pharmacokinetic and pharmacodynamic properties of EVs. This review classifies EVs and their biogenesis routes, and discusses the isolation and characterisation methods adopted by the international EV research community, before delving into what is known about EVs as drug delivery systems and why organ transplantation represents an ideal platform for their development as drug delivery systems.
Collapse
Affiliation(s)
- Harry V M Spiers
- Department of Surgery, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
- NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Lukas K J Stadler
- Department of Surgery, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
- NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Hugo Smith
- Department of Surgery, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Vasilis Kosmoliaptsis
- Department of Surgery, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
- NIHR Blood and Transplant Research Unit in Organ Donation and Transplantation, University of Cambridge, Cambridge CB2 0QQ, UK
| |
Collapse
|
24
|
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
|
25
|
Human Milk-Derived Levels of let-7g-5p May Serve as a Diagnostic and Prognostic Marker of Low Milk Supply in Breastfeeding Women. Nutrients 2023; 15:nu15030567. [PMID: 36771276 PMCID: PMC9920885 DOI: 10.3390/nu15030567] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 01/13/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
Low milk supply (LMS) is associated with early breastfeeding cessation; however, the biological underpinnings in the mammary gland are not understood. MicroRNAs (miRNAs) are small non-coding RNAs that post-transcriptionally downregulate gene expression, and we hypothesized the profile of miRNAs secreted into milk reflects lactation performance. Longitudinal changes in milk miRNAs were measured using RNAseq in women with LMS (n = 47) and adequate milk supply (AMS; n = 123). Relationships between milk miRNAs, milk supply, breastfeeding outcomes, and infant weight gain were assessed, and interactions between milk miRNAs, maternal diet, smoking status, and BMI were determined. Women with LMS had lower milk volume (p = 0.003), were more likely to have ceased breast feeding by 24 wks (p = 0.0003) and had infants with a lower mean weight-for-length z-score (p = 0.013). Milk production was significantly associated with milk levels of miR-16-5p (R = -0.14, adj p = 0.044), miR-22-3p (R = 0.13, adj p = 0.044), and let-7g-5p (R = 0.12, adj p = 0.046). Early milk levels of let-7g-5p were significantly higher in mothers with LMS (adj p = 0.0025), displayed an interaction between lactation stage and milk supply (p < 0.001), and were negatively related to fruit intake (p = 0.015). Putative targets of let-7g-5p include genes important to hormone signaling, RNA regulation, ion transport, and the extracellular matrix, and down-regulation of two targets (PRLR and IGF2BP1/IMP1) was confirmed in mammary cells overexpressing let-7g-5p in vitro. Our data provide evidence that milk-derived miRNAs reflect lactation performance in women and warrant further investigation to assess their utility for predicting LMS risk and early breastfeeding cessation.
Collapse
|
26
|
Kondracka A, Gil-Kulik P, Kondracki B, Frąszczak K, Oniszczuk A, Rybak-Krzyszkowska M, Staniczek J, Kwaśniewska A, Kocki J. Occurrence, Role, and Challenges of MicroRNA in Human Breast Milk: A Scoping Review. Biomedicines 2023; 11:biomedicines11020248. [PMID: 36830785 PMCID: PMC9953053 DOI: 10.3390/biomedicines11020248] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
MicroRNAs are non-coding segments of RNA involved in the epigenetic modulation of various biological processes. Their occurrence in biological fluids, such as blood, saliva, tears, and breast milk, has drawn attention to their potential influence on health and disease development. Hundreds of microRNAs have been isolated from breast milk, yet the evidence on their function remains inconsistent and inconclusive. The rationale for the current scoping review is to map the evidence on the occurrence, characterization techniques, and functional roles of microRNAs in breast milk. The review of the sources of this evidence highlights the need to address methodological challenges to achieve future advances in understanding microRNAs in breast milk, particularly their role in conditions such as neoplasms. Nonetheless, remarkable progress has been made in characterizing the microRNA profiles of human breast milk.
Collapse
Affiliation(s)
- Adrianna Kondracka
- Department of Obstetrics and Pathology of Pregnancy, Medical University of Lublin, 20-059 Lublin, Poland
| | - Paulina Gil-Kulik
- Department of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
| | - Bartosz Kondracki
- Department of Cardiology, Medical University of Lublin, 20-059 Lublin, Poland
- Correspondence:
| | - Karolina Frąszczak
- Department of Oncological Gynecology and Gynecology, Medical University of Lublin, 20-059 Lublin, Poland
| | - Anna Oniszczuk
- Department of Inorganic Chemistry, Medical University of Lublin, 20-059 Lublin, Poland
| | | | - Jakub Staniczek
- Department of Gynecology, Obstetrics and Gynecologic Oncology, Medical University of Silesia, 40-055 Katowice, Poland
| | - Anna Kwaśniewska
- Department of Obstetrics and Pathology of Pregnancy, Medical University of Lublin, 20-059 Lublin, Poland
| | - Janusz Kocki
- Department of Clinical Genetics, Medical University of Lublin, 11 Radziwillowska Str., 20-080 Lublin, Poland
| |
Collapse
|
27
|
Cho YE, Vorn R, Chimenti M, Crouch K, Shaoshuai C, Narayanaswamy J, Harken A, Schmidt R, Gill J, Lee H. Extracellular vesicle miRNAs in breast milk of obese mothers. Front Nutr 2022; 9:976886. [PMID: 36313069 PMCID: PMC9597365 DOI: 10.3389/fnut.2022.976886] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 09/26/2022] [Indexed: 11/18/2022] Open
Abstract
Background Breast milk has abundant extracellular vesicles (EVs) containing various biological molecules (cargo), including miRNAs. EVs are not degraded in the gastrointestinal system and circulation; thus, breast milk EVs (bEVs) are expected to interact with other organs in breastfed infants and modify the gene expression of recipient cells using miRNAs. Maternal pre-pregnancy BMI is a critical factor influencing the composition of breast milk. Thus, in mothers with obesity, miRNAs in bEVs can be altered, which might be associated with adverse health outcomes in infants. In this study, we examined 798 miRNAs to determine which miRNAs are altered in the bEVs of mothers with obesity and their potential impact on breastfed infants. Methods We recruited healthy nursing mothers who were either of normal weight (BMI < 25) or with obesity (BMI ≥ 30) based on their pre-pregnancy BMI, and delivered a singleton baby in the prior 6 months. EVs were isolated from breast milk with ultracentrifugation. bEV characteristics were examined by flow cytometry and fluorescence imaging of EV markers. A total of 798 miRNAs were screened using a NanoString human miRNA panel to find differentially expressed miRNAs in bEVs of mothers with obesity compared to mothers of normal weight. Results We included 65 nursing mothers: 47 of normal weight and 18 with obesity based on pre-pregnancy BMI. After bEV isolation, we confirmed the expression of various EV markers. Out of 37 EV markers, CD326 (EpCaM) was the most highly expressed in bEVs. The most abundant miRNAs in bEVs include miR-30b-5p, miR-4454, miR-494-3p, and let-7 miRNAs. Target genes of the top 10 miRNAs were associated with cancer, prolactin pathway, EGFR, ErbB, and FoxO signaling pathway. In bEVs of mothers with obesity, 19 miRNAs were differentially expressed (adjusted p < 0.05 cut-off), which include miR-575, miR-630, miR-642a-3p, and miR-652-5p. These miRNAs and their target genes were associated with neurological diseases and psychological disorders. Conclusion In this study, we characterized bEVs and demonstrated altered miRNAs in bEVs of mothers with obesity and identified the pathways of their potential target genes. Our findings will provide insight for future studies investigating the role of bEVs in breastfed infants.
Collapse
Affiliation(s)
- Young Eun Cho
- College of Nursing, The University of Iowa, Iowa City, IA, United States,*Correspondence: Young-Eun Cho,
| | - Rany Vorn
- School of Nursing, Johns Hopkins University, Baltimore, MD, United States
| | - Michael Chimenti
- College of Medicine The University of Iowa, Iowa City, IA, United States
| | - Keith Crouch
- College of Nursing, The University of Iowa, Iowa City, IA, United States
| | - Chen Shaoshuai
- College of Nursing, The University of Iowa, Iowa City, IA, United States
| | | | - Alaria Harken
- College of Nursing, The University of Iowa, Iowa City, IA, United States
| | - Reegan Schmidt
- College of Nursing, The University of Iowa, Iowa City, IA, United States
| | - Jessica Gill
- School of Nursing, Johns Hopkins University, Baltimore, MD, United States,Department of Neurology and Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Hyangkyu Lee
- Mo-Im Kim Nursing Research Institute, College of Nursing, Yonsei University, Seoul, South Korea
| |
Collapse
|
28
|
Human Milk Extracellular Vesicles: A Biological System with Clinical Implications. Cells 2022; 11:cells11152345. [PMID: 35954189 PMCID: PMC9367292 DOI: 10.3390/cells11152345] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/25/2022] [Accepted: 07/28/2022] [Indexed: 12/10/2022] Open
Abstract
The consumption of human milk by a breastfeeding infant is associated with positive health outcomes, including lower risk of diarrheal disease, respiratory disease, otitis media, and in later life, less risk of chronic disease. These benefits may be mediated by antibodies, glycoproteins, glycolipids, oligosaccharides, and leukocytes. More recently, human milk extracellular vesicles (hMEVs) have been identified. HMEVs contain functional cargos, i.e., miRNAs and proteins, that may transmit information from the mother to promote infant growth and development. Maternal health conditions can influence hMEV composition. This review summarizes hMEV biogenesis and functional contents, reviews the functional evidence of hMEVs in the maternal–infant health relationship, and discusses challenges and opportunities in hMEV research.
Collapse
|
29
|
Dobre EG, Constantin C, Neagu M. Skin Cancer Research Goes Digital: Looking for Biomarkers within the Droplets. J Pers Med 2022; 12:jpm12071136. [PMID: 35887633 PMCID: PMC9323323 DOI: 10.3390/jpm12071136] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/12/2022] [Accepted: 07/12/2022] [Indexed: 12/24/2022] Open
Abstract
Skin cancer, which includes the most frequent malignant non-melanoma carcinomas (basal cell carcinoma, BCC, and squamous cell carcinoma, SCC), along with the difficult to treat cutaneous melanoma (CM), pose important worldwide issues for the health care system. Despite the improved anti-cancer armamentarium and the latest scientific achievements, many skin cancer patients fail to respond to therapies, due to the remarkable heterogeneity of cutaneous tumors, calling for even more sophisticated biomarker discovery and patient monitoring approaches. Droplet digital polymerase chain reaction (ddPCR), a robust method for detecting and quantifying low-abundance nucleic acids, has recently emerged as a powerful technology for skin cancer analysis in tissue and liquid biopsies (LBs). The ddPCR method, being capable of analyzing various biological samples, has proved to be efficient in studying variations in gene sequences, including copy number variations (CNVs) and point mutations, DNA methylation, circulatory miRNome, and transcriptome dynamics. Moreover, ddPCR can be designed as a dynamic platform for individualized cancer detection and monitoring therapy efficacy. Here, we present the latest scientific studies applying ddPCR in dermato-oncology, highlighting the potential of this technology for skin cancer biomarker discovery and validation in the context of personalized medicine. The benefits and challenges associated with ddPCR implementation in the clinical setting, mainly when analyzing LBs, are also discussed.
Collapse
Affiliation(s)
- Elena-Georgiana Dobre
- Faculty of Biology, University of Bucharest, Splaiul Independentei 91–95, 050095 Bucharest, Romania;
- Correspondence:
| | - Carolina Constantin
- Immunology Department, “Victor Babes” National Institute of Pathology, 050096 Bucharest, Romania;
- Pathology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
| | - Monica Neagu
- Faculty of Biology, University of Bucharest, Splaiul Independentei 91–95, 050095 Bucharest, Romania;
- Immunology Department, “Victor Babes” National Institute of Pathology, 050096 Bucharest, Romania;
- Pathology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
| |
Collapse
|
30
|
Ali NB, Abdull Razis AF, Ooi DJ, Chan KW, Ismail N, Foo JB. Theragnostic Applications of Mammal and Plant-Derived Extracellular Vesicles: Latest Findings, Current Technologies, and Prospects. Molecules 2022; 27:3941. [PMID: 35745063 PMCID: PMC9228370 DOI: 10.3390/molecules27123941] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/26/2022] [Accepted: 02/11/2022] [Indexed: 11/16/2022] Open
Abstract
The way cells communicate is not fully understood. However, it is well-known that extracellular vesicles (EVs) are involved. Researchers initially thought that EVs were used by cells to remove cellular waste. It is now clear that EVs function as signaling molecules released by cells to communicate with one another, carrying a cargo representing the mother cell. Furthermore, these EVs can be found in all biological fluids, making them the perfect non-invasive diagnostic tool, as their cargo causes functional changes in the cells upon receiving, unlike synthetic drug carriers. EVs last longer in circulation and instigate minor immune responses, making them the perfect drug carrier. This review sheds light on the latest development in EVs isolation, characterization and, application as therapeutic cargo, novel drug loading techniques, and diagnostic tools. We also address the advancement in plant-derived EVs, their characteristics, and applications; since plant-derived EVs only recently gained focus, we listed the latest findings. Although there is much more to learn about, EV is a wide field of research; what scientists have discovered so far is fascinating. This paper is suitable for those new to the field seeking to understand EVs and those already familiar with it but wanting to review the latest findings.
Collapse
Affiliation(s)
- Nada Basheir Ali
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia;
| | - Ahmad Faizal Abdull Razis
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia;
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia; (K.W.C.); (N.I.)
| | - Der Jiun Ooi
- Department of Oral Biology and Biomedical Sciences, Faculty of Dentistry, MAHSA University, Bandar Saujana Putra, Jenjarom 42610, Selangor, Malaysia
| | - Kim Wei Chan
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia; (K.W.C.); (N.I.)
| | - Norsharina Ismail
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia; (K.W.C.); (N.I.)
| | - Jhi Biau Foo
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya 47500, Selangor, Malaysia;
| |
Collapse
|
31
|
Abstract
PURPOSE OF REVIEW Milk-derived extracellular vesicles (MDEVs) are nanovesicles that carry microRNA (miRNA) DNA, RNA, proteins and lipids. MDEVs have a potential of therapeutic targets, based on their properties and cargo profile. The present review summarizes recent studies on MDEVs, their cargo and potential role in mammalian development. RECENT FINDINGS The detailed characterization of their miRNA cargo leads to the conclusion of their potential importance in the regulation of gene expression, immune function, development and infant growth.While their miRNAs are important regulatory elements and their profile expression was characterized in various mammalian milk sources, little is known about their effect on infant health and development. MiRNA activity in breast milk is likely influenced by the overall ecosystem of the early environment, including maternal characteristics, behaviors, and health. SUMMARY MDEVs may have an important role in early child development and infant future health. Understanding benefits of MDEVs characteristics have potential role on gut maturation, immune system development and the prevention of metabolic disorders.
Collapse
Affiliation(s)
- Regina Golan-Gerstl
- Department of Pediatrics, Hadassah Medical Center and Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | | |
Collapse
|
32
|
Hicks SD, Confair A, Warren K, Chandran D. Levels of Breast Milk MicroRNAs and Other Non-Coding RNAs Are Impacted by Milk Maturity and Maternal Diet. Front Immunol 2022; 12:785217. [PMID: 35095859 PMCID: PMC8796169 DOI: 10.3389/fimmu.2021.785217] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 12/15/2021] [Indexed: 12/21/2022] Open
Abstract
There is emerging evidence that non-coding RNAs (ncRNAs) within maternal breast milk (MBM) impart unique metabolic and immunologic effects on developing infants. Most studies examining ncRNAs in MBM have focused on microRNAs. It remains unclear whether microRNA levels are related to other ncRNAs, or whether they are impacted by maternal characteristics. This longitudinal cohort study examined 503 MBM samples from 192 mothers to: 1) identify the most abundant ncRNAs in MBM; 2) examine the impact of milk maturity on ncRNAs; and 3) determine whether maternal characteristics affect ncRNAs. MBM was collected at 0, 1, and 4 months post-delivery. High throughput sequencing quantified ncRNAs within the lipid fraction. There were 3069 ncRNAs and 238 microRNAs with consistent MBM presence (≥10 reads in ≥10% samples). Levels of 17 ncRNAs and 11 microRNAs accounted for 80% of the total RNA content. Most abundant microRNAs displayed relationships ([R]>0.2, adj p< 0.05) with abundant ncRNAs. A large proportion of ncRNAs (1269/3069; 41%) and microRNAs (206/238; 86%) were affected by MBM maturity. The majority of microRNAs (111/206; 54%) increased from 0-4 months. Few ncRNAs and microRNAs were affected (adj p < 0.05) by maternal age, race, parity, body mass index, gestational diabetes, or collection time. However, nearly half of abundant microRNAs (4/11) were impacted by diet. To our knowledge this is the largest study of MBM ncRNAs, and the first to demonstrate a relationship between MBM microRNAs and maternal diet. Such knowledge could guide nutritional interventions aimed at optimizing metabolic and immunologic microRNA profiles within MBM.
Collapse
Affiliation(s)
- Steven D Hicks
- Division of Academic General Pediatrics, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, United States
| | - Alexandra Confair
- Division of Academic General Pediatrics, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, United States
| | - Kaitlyn Warren
- Division of Academic General Pediatrics, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, United States
| | - Desirae Chandran
- Division of Academic General Pediatrics, Department of Pediatrics, Penn State College of Medicine, Hershey, PA, United States
| |
Collapse
|
33
|
Raymond F, Lefebvre G, Texari L, Pruvost S, Metairon S, Cottenet G, Zollinger A, Mateescu B, Billeaud C, Picaud JC, Silva-Zolezzi I, Descombes P, Bosco N. Longitudinal Human Milk miRNA Composition over the First 3 mo of Lactation in a Cohort of Healthy Mothers Delivering Term Infants. J Nutr 2022; 152:94-106. [PMID: 34510208 DOI: 10.1093/jn/nxab282] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 07/12/2021] [Accepted: 07/28/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) are small noncoding RNAs involved in posttranscriptional regulation. miRNAs can be secreted and found in many body fluids, and although they are particularly abundant in breastmilk, their functions remain elusive. Human milk (HM) miRNAs start to raise considerable interest, but a comprehensive understanding of the repertoire and expression profiles along lactation has not been well characterized. OBJECTIVES This study aimed to characterize the longitudinal profile of HM miRNA between the second week and third month postpartum. METHODS We used a new sensitive technology to measure HM miRNAs in a cohort of 44 French mothers [mean ± SD age: 31 ± 3.5; BMI (in kg/m2) 21.8 ± 2.3] who delivered at term and provided HM samples at 3 time points (17 ± 3 d, 60 ± 3 d, and 90 ± 3 d) during follow-up visits. RESULTS We detected 685 miRNAs, of which 35 showed a high and stable expression along the lactation period analyzed. We also described for the first time a set of 11 miRNAs with a dynamic expression profile. To gain insight into the potential functional relevance of this set of miRNAs, we selected miR-3126 and miR-3184 to treat undifferentiated Caco-2 human intestinal cells and then assessed differentially expressed genes and modulation of related biological pathways. CONCLUSIONS Overall, our study provides new insights into HM miRNA composition and, to our knowledge, the first description of its longitudinal dynamics in mothers who delivered at term. Our in vitro results obtained in undifferentiated Caco-2 human intestinal cells transfected with HM miRNAs also provide further support to the hypothesized mother-to-neonate signaling role of HM miRNAs. This trial was registered at clinicaltrials.gov as NCT01894893.
Collapse
Affiliation(s)
- Frederic Raymond
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Gregory Lefebvre
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Lorane Texari
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Solenn Pruvost
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Sylviane Metairon
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Geoffrey Cottenet
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Alix Zollinger
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Bogdan Mateescu
- Brain Research Institute, University of Zurich, Zurich, Switzerland
| | - Claude Billeaud
- Neonatology Nutrition, Lactarium Bordeaux-Marmande, Bordeaux, France
| | - Jean-Charles Picaud
- Neonatal Intensive Care Unit, University Hospital Croix Rousse, Lyon, France.,CarMeN unit, Claude Bernard University Lyon 1, 69310 Pierre Benite, France
| | | | - Patrick Descombes
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Nabil Bosco
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland.,Nestlé Research, Singapore
| |
Collapse
|
34
|
Kupsco A, Lee JJ, Prada D, Valvi D, Hu L, Petersen MS, Coull BA, Weihe P, Grandjean P, Baccarelli AA. Marine pollutant exposures and human milk extracellular vesicle-microRNAs in a mother-infant cohort from the Faroe Islands. ENVIRONMENT INTERNATIONAL 2022; 158:106986. [PMID: 34991248 PMCID: PMC8742869 DOI: 10.1016/j.envint.2021.106986] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 10/29/2021] [Accepted: 11/15/2021] [Indexed: 05/26/2023]
Abstract
BACKGROUND/AIMS Early life exposures to marine contaminants can adversely impact child health but modes of action are unclear. Human milk contains extracellular vesicles (EVs) that can transport biologically relevant cargo from mother to infant, including microRNAs (miRNAs), and may partly mediate the effects of pollutants on child health. However, the role of marine pollutants on miRNA expression in milk EVs is unexplored. METHODS We isolated EV RNA from 333 milk samples collected between 2 and 74 days postpartum from a Faroese birth cohort born 1997-2000 and sequenced 2083 miRNAs using a targeted library preparation method. We quantified five perfluoroalkyl substances (PFAS), pesticide metabolite p,p'-dichlorodiphenyldichloroethylene (DDE), and the sum of three major polychlorinated biphenyls (ΣPCBs) in maternal serum at 34 weeks of gestation and maternal hair total mercury (Hg) at birth. We used negative binomial regressions to estimate associations between individual pollutants and 418 reliably expressed EV-miRNAs adjusted for potential confounders. We performed sparse principal components (PCs) analysis to derive the first four components of the EV-miRNA data and examined associations between pollutants and PCs using Bayesian kernel machine regression (BKMR). RESULTS We observed no associations between pollutants and individual EV-miRNA expression after controlling the false discovery rate at 0.1. However, BKMR suggested that Hg was positively associated with PC1 and negatively associated with PC3, while ΣPCBs was negatively associated with PC3, and two PFAS were associated with PC4. Exploration of PC loadings followed by pathway analyses suggested that miRNAs in PC1 (miR-200b-3p, miR-664a-3p, miR-6738-5p, miR-429, miR-1236-5p, miR-4464, and miR-30b-5p) may be related to Hg neurotoxicity, while remaining PCs require further research. CONCLUSIONS Our findings suggest that groups of milk EV-miRNAs may better serve as environmental biomarkers than individual miRNAs. Future studies are needed to elucidate the role of milk EV-miRNAs in child health following prenatal exposures.
Collapse
Affiliation(s)
- Allison Kupsco
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY 10023, United States.
| | - Jenny Jyoung Lee
- Department of Biostatistics, Harvard T.H Chan School of Public Health, Boston, MA, United States
| | - Diddier Prada
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY 10023, United States; Instituto Nacional de Cancerología, San Fernando 22, Colonia Seccion XVI, Tlalpan, Mexico City 14080, Mexico
| | - Damaskini Valvi
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY 10029, United States
| | - Lisa Hu
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY 10023, United States
| | - Maria Skaalum Petersen
- Department of Occupational Medicine and Public Health, The Faroese Hospital System, Tórshavn, Faroe Islands; Center of Health Science, University of The Faroe Islands, Tórshavn, Faroe Islands
| | - Brent A Coull
- Department of Biostatistics, Harvard T.H Chan School of Public Health, Boston, MA, United States; Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Pal Weihe
- Department of Occupational Medicine and Public Health, The Faroese Hospital System, Tórshavn, Faroe Islands; Center of Health Science, University of The Faroe Islands, Tórshavn, Faroe Islands
| | - Philippe Grandjean
- Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, United States; Department of Environmental Medicine, University of Southern Denmark, Odense C, Denmark
| | - Andrea A Baccarelli
- Department of Environmental Health Sciences, Columbia University Mailman School of Public Health, New York, NY 10023, United States
| |
Collapse
|
35
|
Mierziak J, Kostyn K, Boba A, Czemplik M, Kulma A, Wojtasik W. Influence of the Bioactive Diet Components on the Gene Expression Regulation. Nutrients 2021; 13:3673. [PMID: 34835928 PMCID: PMC8619229 DOI: 10.3390/nu13113673] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/13/2021] [Accepted: 10/14/2021] [Indexed: 02/07/2023] Open
Abstract
Diet bioactive components, in the concept of nutrigenetics and nutrigenomics, consist of food constituents, which can transfer information from the external environment and influence gene expression in the cell and thus the function of the whole organism. It is crucial to regard food not only as the source of energy and basic nutriments, crucial for living and organism development, but also as the factor influencing health/disease, biochemical mechanisms, and activation of biochemical pathways. Bioactive components of the diet regulate gene expression through changes in the chromatin structure (including DNA methylation and histone modification), non-coding RNA, activation of transcription factors by signalling cascades, or direct ligand binding to the nuclear receptors. Analysis of interactions between diet components and human genome structure and gene activity is a modern approach that will help to better understand these relations and will allow designing dietary guidances, which can help maintain good health.
Collapse
Affiliation(s)
- Justyna Mierziak
- Faculty of Biotechnology, University of Wrocław, Przybyszewskiego 63/77, 51-148 Wroclaw, Poland; (A.B.); (M.C.); (A.K.)
| | - Kamil Kostyn
- Department of Genetics, Plant Breeding & Seed Production, Faculty of Life Sciences and Technology, Wroclaw University of Environmental and Life Sciences, pl. Grunwaldzki 24A, 50-363 Wroclaw, Poland;
| | - Aleksandra Boba
- Faculty of Biotechnology, University of Wrocław, Przybyszewskiego 63/77, 51-148 Wroclaw, Poland; (A.B.); (M.C.); (A.K.)
| | - Magdalena Czemplik
- Faculty of Biotechnology, University of Wrocław, Przybyszewskiego 63/77, 51-148 Wroclaw, Poland; (A.B.); (M.C.); (A.K.)
| | - Anna Kulma
- Faculty of Biotechnology, University of Wrocław, Przybyszewskiego 63/77, 51-148 Wroclaw, Poland; (A.B.); (M.C.); (A.K.)
| | - Wioleta Wojtasik
- Faculty of Biotechnology, University of Wrocław, Przybyszewskiego 63/77, 51-148 Wroclaw, Poland; (A.B.); (M.C.); (A.K.)
| |
Collapse
|
36
|
Tingö L, Ahlberg E, Johansson L, Pedersen SA, Chawla K, Sætrom P, Cione E, Simpson MR. Non-Coding RNAs in Human Breast Milk: A Systematic Review. Front Immunol 2021; 12:725323. [PMID: 34539664 PMCID: PMC8440964 DOI: 10.3389/fimmu.2021.725323] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/13/2021] [Indexed: 12/12/2022] Open
Abstract
Breast milk is the primary source of nutrition and hydration for the newborn infant but also plays an important role in the child’s first immune defense. Additionally, several breast milk factors have been implicated in immune-related health outcomes later in life, including immunoglobulins, cytokines, chemokines, growth factors and, more recently, non-coding RNA (ncRNA) species. In this systematic review, we provide a comprehensive summary of the current literature on endogenous ncRNAs found in human breast milk. Thirty (30) relevant studies were identified and, whilst the majority studies focused on microRNAs (miRNAs), there is evidence that breast milk contains high quantities of RNA which also include long-coding RNAs, circular RNAs, as well as other short RNAs and fragmented tRNA and rRNAs. Among studies investigating miRNAs, miR-148a-3p, miR-30a/d-5p, miR-22-3p, miR-146b-5p, miR-200a/c-3p, and the 5p end of the let-7 miRNAs were commonly reported among the top 10 miRNAs in the cell, lipid, and skim milk fractions of breast milk. Methodological difference and small sample sizes limit the possibility of conclusively identifying which maternal and infant characteristics affect the miRNA profile. The highly expressed miRNAs were generally reported to be similar across lactational stage, milk fraction, maternal and infant characteristics, or infant growth and health. All the same, individual studies identify potential differences in miRNA expression levels which should be confirmed by future studies. Stability, uptake, and physiological functions of miRNAs were also considered in several studies. Breast milk miRNAs are relatively resistant to a range of harsh conditions and uptake experiments suggest that extracellular vesicles containing miRNAs and circular RNAs can be taken up by intestinal epithelial cells. Although the evidence regarding the functional effect of breast milk miRNAs is limited, the predicted functions range from metabolic and biosynthetic processes to signaling pathways, cellular adhesion, communication, growth, and differentiation. Finally, this systematic review highlights some of the methodological challenges and knowledge gaps which can help direct future research in this field. In particular, it is important to further investigate the bioavailability of miRNAs in different milk fractions, and to characterize other ncRNAs which are largely unstudied.
Collapse
Affiliation(s)
- Lina Tingö
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden.,Örebro University Food and Health Programme, School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Emelie Ahlberg
- Division of Inflammation and Infection, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Lovisa Johansson
- Division of Neurobiology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Sindre Andre Pedersen
- Library Section for Medicine and Health Sciences, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | - Konika Chawla
- Department of Clinical and Molecular Medicine, NTNU Norwegian University of Science and Technology, Trondheim, Norway.,Bioinformatics Core Facility - BioCore, NTNU Norwegian University of Science and Technology, Trondheim, Norway.,K.G. Jebsen Center for Genetic Epidemiology, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | - Pål Sætrom
- Department of Clinical and Molecular Medicine, NTNU Norwegian University of Science and Technology, Trondheim, Norway.,Bioinformatics Core Facility - BioCore, NTNU Norwegian University of Science and Technology, Trondheim, Norway.,K.G. Jebsen Center for Genetic Epidemiology, NTNU Norwegian University of Science and Technology, Trondheim, Norway.,Department of Computer Science, NTNU Norwegian University of Science and Technology, Trondheim, Norway
| | - Erika Cione
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Melanie Rae Simpson
- Department of Public Health and Nursing, NTNU Norwegian University of Science and Technology, Trondheim, Norway.,Clinic of Laboratory Medicine, St. Olavs Hospital, Trondheim, Norway
| |
Collapse
|
37
|
Extracellular Vesicles in Skin Wound Healing. Pharmaceuticals (Basel) 2021; 14:ph14080811. [PMID: 34451909 PMCID: PMC8400229 DOI: 10.3390/ph14080811] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/13/2021] [Accepted: 08/16/2021] [Indexed: 12/13/2022] Open
Abstract
Each year, millions of individuals suffer from a non-healing wound, abnormal scarring, or injuries accompanied by an infection. For these cases, scientists are searching for new therapeutic interventions, from which one of the most promising is the use of extracellular vesicles (EVs). Naturally, EV-based signaling takes part in all four wound healing phases: hemostasis, inflammation, proliferation, and remodeling. Such an extensive involvement of EVs suggests exploiting their action to modulate the impaired healing phase. Furthermore, next to their natural wound healing capacity, EVs can be engineered for better defined pharmaceutical purposes, such as carrying specific cargo or targeting specific destinations by labelling them with certain surface proteins. This review aims to promote scientific awareness in basic and translational research of EVs by summarizing the current knowledge about their natural role in each stage of skin repair and the most recent findings in application areas, such as wound healing, skin regeneration, and treatment of dermal diseases, including the stem cell-derived, plant-derived, and engineered EVs.
Collapse
|
38
|
Ong SL, Blenkiron C, Haines S, Acevedo-Fani A, Leite JAS, Zempleni J, Anderson RC, McCann MJ. Ruminant Milk-Derived Extracellular Vesicles: A Nutritional and Therapeutic Opportunity? Nutrients 2021; 13:2505. [PMID: 34444665 PMCID: PMC8398904 DOI: 10.3390/nu13082505] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/13/2021] [Accepted: 07/19/2021] [Indexed: 12/12/2022] Open
Abstract
Milk has been shown to contain a specific fraction of extracellular particles that are reported to resist digestion and are purposefully packaged with lipids, proteins, and nucleic acids to exert specific biological effects. These findings suggest that these particles may have a role in the quality of infant nutrition, particularly in the early phase of life when many of the foundations of an infant's potential for health and overall wellness are established. However, much of the current research focuses on human or cow milk only, and there is a knowledge gap in how milk from other species, which may be more commonly consumed in different regions, could also have these reported biological effects. Our review provides a summary of the studies into the extracellular particle fraction of milk from a wider range of ruminants and pseudo-ruminants, focusing on how this fraction is isolated and characterised, the stability and uptake of the fraction, and the reported biological effects of these fractions in a range of model systems. As the individual composition of milk from different species is known to differ, we propose that the extracellular particle fraction of milk from non-traditional and minority species may also have important and distinct biological properties that warrant further study.
Collapse
Affiliation(s)
- Siew Ling Ong
- Smart Foods Innovation Centre of Excellence, Te Ohu Rangahau Kai, AgResearch Ltd., Massey University Campus, Palmerston North 4410, New Zealand;
| | - Cherie Blenkiron
- Department of Molecular Medicine and Pathology, Faculty of Medical and Health Sciences, University of Auckland, Auckland 1051, New Zealand;
- Auckland Cancer Society Research Centre, University of Auckland, Auckland 1051, New Zealand
| | - Stephen Haines
- Beyond Food Innovation Centre of Excellence, AgResearch Ltd., Lincoln 7674, New Zealand;
| | - Alejandra Acevedo-Fani
- Riddet Institute, Massey University, Palmerston North 4442, New Zealand; (A.A.-F.); (J.A.S.L.)
| | - Juliana A. S. Leite
- Riddet Institute, Massey University, Palmerston North 4442, New Zealand; (A.A.-F.); (J.A.S.L.)
| | - Janos Zempleni
- Department of Nutrition and Health Sciences, University of Nebraska-Lincoln, Lincoln, NE 68583, USA;
| | - Rachel C. Anderson
- Smart Foods Innovation Centre of Excellence, Te Ohu Rangahau Kai, AgResearch Ltd., Massey University Campus, Palmerston North 4410, New Zealand;
- Riddet Institute, Massey University, Palmerston North 4442, New Zealand; (A.A.-F.); (J.A.S.L.)
| | - Mark J. McCann
- Smart Foods Innovation Centre of Excellence, Te Ohu Rangahau Kai, AgResearch Ltd., Massey University Campus, Palmerston North 4410, New Zealand;
- Riddet Institute, Massey University, Palmerston North 4442, New Zealand; (A.A.-F.); (J.A.S.L.)
| |
Collapse
|
39
|
Jiang X, You L, Zhang Z, Cui X, Zhong H, Sun X, Ji C, Chi X. Biological Properties of Milk-Derived Extracellular Vesicles and Their Physiological Functions in Infant. Front Cell Dev Biol 2021; 9:693534. [PMID: 34249944 PMCID: PMC8267587 DOI: 10.3389/fcell.2021.693534] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 05/26/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) are released by all cells under pathological and physiological conditions. EVs harbor various biomolecules, including protein, lipid, non-coding RNA, messenger RNA, and DNA. In 2007, mRNA and microRNA (miRNA) carried by EVs were found to have regulatory functions in recipient cells. The biological function of EVs has since then increasingly drawn interest. Breast milk, as the most important nutritional source for infants, contains EVs in large quantities. An increasing number of studies have provided the basis for the hypothesis associated with information transmission between mothers and infants via breast milk-derived EVs. Most studies on milk-derived EVs currently focus on miRNAs. Milk-derived EVs contain diverse miRNAs, which remain stable both in vivo and in vitro; as such, they can be absorbed across different species. Further studies have confirmed that miRNAs derived from milk-derived EVs can resist the acidic environment and enzymatic hydrolysis of the digestive tract; moreover, they can be absorbed by intestinal cells in infants to perform physiological functions. miRNAs derived from milk EVs have been reported in the maturation of immune cells, regulation of immune response, formation of neuronal synapses, and development of metabolic diseases such as obesity and diabetes. This article reviews current status and advances in milk-derived EVs, including their history, biogenesis, molecular contents, and biological functions. The effects of milk-derived EVs on growth and development in both infants and adults were emphasized. Finally, the potential application and future challenges of milk-derived EVs were discussed, providing comprehensive understanding and new insight into milk-derived EVs.
Collapse
Affiliation(s)
- Xue Jiang
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China.,The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, China
| | - Lianghui You
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Zhenxing Zhang
- The First Affiliated Hospital With Nanjing Medical University, Nanjing, China
| | - Xianwei Cui
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Hong Zhong
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Xingzhen Sun
- The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, China
| | - Chenbo Ji
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Xia Chi
- Women's Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| |
Collapse
|
40
|
Melnik BC, Stremmel W, Weiskirchen R, John SM, Schmitz G. Exosome-Derived MicroRNAs of Human Milk and Their Effects on Infant Health and Development. Biomolecules 2021; 11:biom11060851. [PMID: 34200323 PMCID: PMC8228670 DOI: 10.3390/biom11060851] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 05/29/2021] [Accepted: 06/01/2021] [Indexed: 11/16/2022] Open
Abstract
Multiple biologically active components of human milk support infant growth, health and development. Milk provides a wide spectrum of mammary epithelial cell-derived extracellular vesicles (MEVs) for the infant. Although the whole spectrum of MEVs appears to be of functional importance for the growing infant, the majority of recent studies report on the MEV subfraction of milk exosomes (MEX) and their miRNA cargo, which are in the focus of this review. MEX and the dominant miRNA-148a play a key role in intestinal maturation, barrier function and suppression of nuclear factor-κB (NF-κB) signaling and may thus be helpful for the prevention and treatment of necrotizing enterocolitis. MEX and their miRNAs reach the systemic circulation and may impact epigenetic programming of various organs including the liver, thymus, brain, pancreatic islets, beige, brown and white adipose tissue as well as bones. Translational evidence indicates that MEX and their miRNAs control the expression of global cellular regulators such as DNA methyltransferase 1-which is important for the up-regulation of developmental genes including insulin, insulin-like growth factor-1, α-synuclein and forkhead box P3-and receptor-interacting protein 140, which is important for the regulation of multiple nuclear receptors. MEX-derived miRNA-148a and miRNA-30b may stimulate the expression of uncoupling protein 1, the key inducer of thermogenesis converting white into beige/brown adipose tissue. MEX have to be considered as signalosomes derived from the maternal lactation genome emitted to promote growth, maturation, immunological and metabolic programming of the offspring. Deeper insights into milk's molecular biology allow the conclusion that infants are both "breast-fed" and "breast-programmed". In this regard, MEX miRNA-deficient artificial formula is not an adequate substitute for breastfeeding, the birthright of all mammals.
Collapse
Affiliation(s)
- Bodo C. Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, D-49076 Osnabrück, Germany;
- Correspondence: ; Tel.: +49-5241-988060
| | - Wolfgang Stremmel
- Private Praxis for Internal Medicine, Beethovenstraße 2, D-76530 Baden-Baden, Germany;
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, D-52074 Aachen, 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, University of Regensburg, D-93053 Regensburg, Germany;
| |
Collapse
|