1
|
Yang H, Wuren T, Zhai B, Liu Y, Er D. Milk-derived exosomes in the regulation of nutritional and immune functions. Food Sci Nutr 2024; 12:7048-7059. [PMID: 39479690 PMCID: PMC11521659 DOI: 10.1002/fsn3.4323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 03/31/2024] [Accepted: 06/27/2024] [Indexed: 11/02/2024] Open
Abstract
Milk-derived exosomes (MDEs), being a component of milk, have the potential to support immune system maturation in offspring and enhance immune cell proliferation. Through the transport and transmission of essential signaling molecules, MDEs contribute to the regulation of intergenerational and intraspecies communication, thereby impacting nutrient uptake and metabolic functions. A comprehensive comprehension of MDE functionalities is imperative for enhancing the quality of the dairy industry. A systematic search of the databases PubMed/Medline, Web of Science, and Scopus utilizing predetermined keywords resulted in the identification of 418 articles, of which 67 were chosen for inclusion in this review, which specifically explores the intersection of immune response and nutrition. This article provides a critical analysis of the classification of extracellular vesicles, the mechanisms underlying the biosynthesis of microvesicular dietary exosomes (MDEs), the components of MDEs, and their relevance in the contexts of nutrition and immune modulation. The primary aim of this review was to offer valuable scholarly insights to support the advancement and practical application of MDEs.
Collapse
Affiliation(s)
- Hui Yang
- College of Basic Medical ScienceQinghai UniversityXiningQinghaiPR China
| | - Tana Wuren
- Research Center for High Altitude MedicineQinghai UniversityXiningQinghaiPR China
| | - Bin‐tao Zhai
- Key Laboratory of Veterinary Pharmaceutical Development, Lanzhou Institute of Husbandry and Pharmaceutical SciencesChinese Academy of Agricultural SciencesLanzhouGansuPR China
| | - Yang Liu
- College of Life ScienceNingxia UniversityYinchuanNingxiaPR China
| | - Demtu Er
- College of Veterinary MedicineInner Mongolia Agricultural UniversityHohhotInner MongoliaPR China
| |
Collapse
|
2
|
Owen MD, Kennedy MG, Quilang RC, Scott EM, Forbes K. The role of microRNAs in pregnancies complicated by maternal diabetes. Clin Sci (Lond) 2024; 138:1179-1207. [PMID: 39289953 PMCID: PMC11409017 DOI: 10.1042/cs20230681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 08/14/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024]
Abstract
With the global prevalence of diabetes increasing, more people of reproductive age are experiencing hyperglycaemic pregnancies. Maternal Type 1 (T1DM) or Type 2 (T2DM) diabetes mellitus, and gestational diabetes mellitus (GDM) are associated with maternal cardiovascular and metabolic complications. Pregnancies complicated by maternal diabetes also increase the risk of short- and long-term health complications for the offspring, including altered fetal growth and the onset of T2DM and cardiometabolic diseases throughout life. Despite advanced methods for improving maternal glucose control, the prevalence of adverse maternal and offspring outcomes associated with maternal diabetes remains high. The placenta is a key organ at the maternal-fetal interface that regulates fetal growth and development. In pregnancies complicated by maternal diabetes, altered placental development and function has been linked to adverse outcomes in both mother and fetus. Emerging evidence suggests that microRNAs (miRNAs) are key molecules involved in mediating these changes. In this review, we describe the role of miRNAs in normal pregnancy and discuss how miRNA dysregulation in the placenta and maternal circulation is associated with suboptimal placental development and pregnancy outcomes in individuals with maternal diabetes. We also discuss evidence demonstrating that miRNA dysregulation may affect the long-term health of mothers and their offspring. As such, miRNAs are potential candidates as biomarkers and therapeutic targets in diabetic pregnancies at risk of adverse outcomes.
Collapse
Affiliation(s)
- Manon D Owen
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, U.K
| | - Margeurite G Kennedy
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, U.K
- Anthony Nolan Research Institute, Royal Free Hospital, Hampstead, London, U.K
- UCL Cancer Institute, Royal Free Campus, London, U.K
| | - Rachel C Quilang
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, U.K
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Eleanor M Scott
- Division of Clinical and Population Sciences, Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, U.K
| | - Karen Forbes
- Discovery and Translational Science Department, Leeds Institute of Cardiovascular and Metabolic Medicine, Faculty of Medicine and Health, University of Leeds, Leeds, U.K
| |
Collapse
|
3
|
Chen R, Yang H, Dai J, Zhang M, Lu G, Zhang M, Yu H, Zheng M, He Q. The biological functions of maternal-derived extracellular vesicles during pregnancy and lactation and its impact on offspring health. Clin Nutr 2023; 42:493-504. [PMID: 36857958 DOI: 10.1016/j.clnu.2023.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 01/25/2023] [Accepted: 02/09/2023] [Indexed: 02/18/2023]
Abstract
During pregnancy and lactation, mothers provide not only nutrients, but also many bioactive components for their offspring through placenta and breast milk, which are essential for offspring development. Extracellular vesicles (EVs) are nanovesicles containing a variety of biologically active molecules and participate in the intercellular communication. In the past decade, an increasing number of studies have reported that maternal-derived EVs play a crucial role in offspring growth, development, and immune system establishment. Hereby, we summarized the characteristics of EVs; biological functions of maternal-derived EVs during pregnancy, including implantation, decidualization, placentation, embryo development and birth of offspring; biological function of breast milk-derived EVs (BMEs) on infant oral and intestinal diseases, immune system, neurodevelopment, and metabolism. In summary, emerging studies have revealed that maternal-derived EVs play a pivotal role in offspring health. As such, maternal-derived EVs may be used as promising biomarkers in offspring disease diagnosis and treatment. However, existing research on maternal-derived EVs and offspring health is largely limited to animal and cellular studies. Evidence from human studies is needed.
Collapse
Affiliation(s)
- Rui Chen
- School of Public Health, Wuhan University, Wuhan, China
| | | | - Jie Dai
- School of Public Health, Wuhan University, Wuhan, China
| | - Minzhe Zhang
- School of Public Health, Wuhan University, Wuhan, China
| | - Gaolei Lu
- School of Public Health, Wuhan University, Wuhan, China
| | - Minjie Zhang
- School of Public Health, Wuhan University, Wuhan, China
| | - Hongjie Yu
- School of Public Health, Wuhan University, Wuhan, China
| | - Miaobing Zheng
- School of Nutrition and Exercise, Deakin University, Melbourne, Australia
| | - Qiqiang He
- School of Public Health, Wuhan University, Wuhan, China; Wuhan University Shenzhen Research Institute, Shenzhen, China; Hubei Biomass-Resource Chemistry and Environmental Biotechnology Key Laboratory, Wuhan University, Wuhan, China.
| |
Collapse
|
4
|
Melnik BC, John SM, Carrera-Bastos P, Cordain L, Leitzmann C, Weiskirchen R, Schmitz G. The Role of Cow's Milk Consumption in Breast Cancer Initiation and Progression. Curr Nutr Rep 2023; 12:122-140. [PMID: 36729355 PMCID: PMC9974716 DOI: 10.1007/s13668-023-00457-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2022] [Indexed: 02/03/2023]
Abstract
PURPOSE OF REVIEW This review evaluates cow milk's impact on breast carcinogenesis by linking recent epidemiological evidence and new insights into the molecular signaling of milk and its constituents in breast cancer (BCa) pathogenesis. RECENT FINDINGS Recent prospective cohort studies support the association between cow's milk consumption and the risk of estrogen receptor-α-positive (ER+) BCa. Milk is a complex biological fluid that increases systemic insulin-like growth factor 1 (IGF-1), insulin and estrogen signaling, and interacting hormonal promoters of BCa. Further potential oncogenic components of commercial milk include exosomal microRNAs (miR-148a-3p, miR-21-5p), bovine meat and milk factors, aflatoxin M1, bisphenol A, pesticides, and micro- and nanoplastics. Individuals with BRCA1 loss-of-function mutations and FTO and IGF1 gain-of-function polymorphisms enhancing IGF-1/mTORC1 signaling may be at increased risk for milk-induced ER+ BCa. Recent prospective epidemiological and pathobiochemical studies identify commercial milk consumption as a critical risk factor of ER+ BCa. Large meta-analyses gathering individuals of different ethnic origins with milk derived from dairy cows of varying genetic backgrounds and diverse feeding procedures as well as missing data on thermal processing of milk (pasteurization versus ultra-heat treatment) make multi-national meta-analyses unsuitable for BCa risk estimations in susceptible populations. Future studies are required that consider all vulnerable periods of breast carcinogenesis to cow's milk exposure, beginning during the perinatal period and puberty, since these are the most critical periods of mammary gland morphogenesis. Notwithstanding the need for better studies including detailed information on milk processing and vulnerable periods of human breast carcinogenesis, the available evidence suggests that dietary guidelines on milk consumption may have to be reconsidered.
Collapse
Affiliation(s)
- Bodo C Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, D-49076, Osnabrück, 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) at the University of Osnabrück, Lower-Saxonian Institute of Occupational Dermatology (NIB), Osnabrück, Germany
| | - Pedro Carrera-Bastos
- Center for Primary Health Care Research, Lund University/Region Skåne, Skåne University Hospital, 205 02, Malmö, Sweden
- Faculty of Biomedical and Health Sciences, Universidad Europea de Madrid, 28670, Madrid, Spain
- Centro de Estudios Avanzados en Nutrición (CEAN), 11007, Cádiz, Spain
| | | | - Claus Leitzmann
- Institute of Nutrition, University of Giessen, 35390, Giessen, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry (IFMPEGKC), RWTH University Hospital Aachen, D-52074, Aachen, Germany
| | - Gerd Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital of Regensburg, University of Regensburg, D-93053, Regensburg, Germany
| |
Collapse
|
5
|
Zeng Y, Wu Y, Zhang Q, Xiao X. Non-coding RNAs: The link between maternal malnutrition and offspring metabolism. Front Nutr 2022; 9:1022784. [DOI: 10.3389/fnut.2022.1022784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/27/2022] [Indexed: 11/11/2022] Open
Abstract
Early life nutrition is associated with the development and metabolism in later life, which is known as the Developmental Origin of Health and Diseases (DOHaD). Epigenetics have been proposed as an important explanation for this link between early life malnutrition and long-term diseases. Non-coding RNAs (ncRNAs) may play a role in this epigenetic programming. The expression of ncRNAs (such as long non-coding RNA H19, microRNA-122, and circular RNA-SETD2) was significantly altered in specific tissues of offspring exposed to maternal malnutrition. Changes in these downstream targets of ncRNAs lead to abnormal development and metabolism. This review aims to summarize the existing knowledge on ncRNAs linking the maternal nutrition condition and offspring metabolic diseases, such as obesity, type 2 diabetes (T2D) and non-alcoholic fatty liver disease (NAFLD).
Collapse
|
6
|
Kochhar P, Dwarkanath P, Ravikumar G, Thomas A, Crasta J, Thomas T, Kurpad AV, Mukhopadhyay A. Placental expression of miR-21-5p, miR-210-3p and miR-141-3p: relation to human fetoplacental growth. Eur J Clin Nutr 2021; 76:730-738. [PMID: 34611295 DOI: 10.1038/s41430-021-01017-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 09/06/2021] [Accepted: 09/21/2021] [Indexed: 12/29/2022]
Abstract
BACKGROUND/OBJECTIVES Dysregulation of microRNAs (miRNAs) and their target genes in placental tissue is associated with foetal growth restriction. We aimed to evaluate associations of placental miR-21-5p, miR-141-3p and miR-210-3p expression with maternal, placental and newborn parameters and with placental expression of their potential target genes PTEN, VEGF, FLT and ENG in a set of well-characterized small- (SGA) and appropriate- (AGA) for gestational age full-term singleton pregnancies. SUBJECTS/METHODS Placental samples (n = 80) from 26 SGA and 54 AGA were collected from full-term singleton pregnancies. Placental transcript abundances of miR-21-5p, miR-141-3p and miR-210-3p were assessed after normalization to a reference miRNA, mir-16-5p by real-time quantitative PCR. Placental transcript abundances of PTEN, VEGF, FLT and ENG were assessed after normalizing to a panel of reference genes. RESULTS Placental miR-21-5p transcript abundance was negatively associated with placental weight (n = 80, r = -0.222, P = 0.047) and this association was specific to the AGA births (n = 54, r = -0.292, P = 0.032). Placental transcript abundances of miR-210-3p and miR-141-3p were not associated with placental weight or birth weight in all 80 births. However, placental miR-210-3p transcript abundance was positively associated with birth weight specifically in the SGA births (n = 26, r = 0.449, P = 0.021). Placental transcript abundance of miR-21-5p was negatively associated with PTEN transcript abundance (Spearman's ρ = -0.245, P = 0.028) while that of miR-141-3p was positively associated with FLT (Spearman's ρ = 0.261, P = 0.019) and ENG (Spearman's ρ = 0.259, P = 0.020) transcript abundances in all 80 births. CONCLUSION We conclude that placental miR-21-5p and miR-210-3p may be involved in fetoplacental growth. However, this regulation is unlikely to be mediated through placental expression of PTEN, VEGF, FLT or ENG.
Collapse
Affiliation(s)
- P Kochhar
- Division of Nutrition, St. John's Research Institute, A recognized research centre of University of Mysore, Bangalore, India
| | - P Dwarkanath
- Division of Nutrition, St. John's Research Institute, A recognized research centre of University of Mysore, Bangalore, India
| | - G Ravikumar
- Department of Pathology, St John's Medical College Hospital, Bangalore, India
| | - A Thomas
- Department of Obstetrics and Gynaecology, St John's Medical College Hospital, Bangalore, India
| | - J Crasta
- Department of Pathology, St John's Medical College Hospital, Bangalore, India
| | - T Thomas
- Department of Biostatistics, St. John's Medical College Hospital, Bangalore, India
| | - A V Kurpad
- Division of Nutrition, St. John's Research Institute, A recognized research centre of University of Mysore, Bangalore, India
| | - A Mukhopadhyay
- Division of Nutrition, St. John's Research Institute, A recognized research centre of University of Mysore, Bangalore, India.
| |
Collapse
|
7
|
Wehbe Z, Kreydiyyeh S. Cow's milk may be delivering potentially harmful undetected cargoes to humans. Is it time to reconsider dairy recommendations? Nutr Rev 2021; 80:874-888. [PMID: 34338770 DOI: 10.1093/nutrit/nuab046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Mammalian evolution has shaped milk into a species-specific vehicle for post-natal development, continuing what began within the mother's womb. Increased consumption of the mother's breast milk is associated with the most adequate metabolic programming and lowers the incidence of the diseases of civilization during adulthood. An abundance of short sequences of RNA, known as microRNA, exists in mammalian breast milk, enclosed within robust small extracellular vesicles known as exosomes. These microRNAs can epigenetically regulate over 60% of human genes. When cow's milk is consumed by humans, the bovine exosomes are transported through the gastrointestinal tract, detected intact in the blood stream, and taken up by target cells, where they alter protein expression. The aim of this review was to highlight the role of dairy exosomes and microRNA, and of the type of dairy product consumed, in human diseases. Given that microRNAs are involved in a vast array of physiological processes and associated with several diseases, perhaps caution should be practiced with regard to human consumption of dairy, particularly for individuals within developmentally critical time frames, such as pregnant and lactating mothers, and young children.
Collapse
Affiliation(s)
- Zena Wehbe
- Z. Wehbe and S. Kreydiyyeh are with the Department of Biology, Faculty of Arts and Sciences, American University of Beirut, Beirut, Lebanon
| | - Sawsan Kreydiyyeh
- Z. Wehbe and S. Kreydiyyeh are with the Department of Biology, Faculty of Arts and Sciences, American University of Beirut, Beirut, Lebanon
| |
Collapse
|
8
|
Howe CG, Foley HB, Kennedy EM, Eckel SP, Chavez TA, Faham D, Grubbs BH, Al-Marayati L, Lerner D, Suglia S, Bastain TM, Marsit CJ, Breton CV. Extracellular vesicle microRNA in early versus late pregnancy with birth outcomes in the MADRES study. Epigenetics 2021; 17:269-285. [PMID: 33734019 DOI: 10.1080/15592294.2021.1899887] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Circulating miRNA may contribute to the development of adverse birth outcomes. However, few studies have investigated extracellular vesicle (EV) miRNA, which play important roles in intercellular communication, or compared miRNA at multiple time points in pregnancy. In the current study, 800 miRNA were profiled for EVs from maternal plasma collected in early (median: 12.5 weeks) and late (median: 31.8 weeks) pregnancy from 156 participants in the MADRES Study, a health disparity pregnancy cohort. Associations between miRNA and birth weight, birth weight for gestational age (GA), and GA at birth were examined using covariate-adjusted robust linear regression. Differences by infant sex and maternal BMI were also investigated. Late pregnancy measures of 13 miRNA were associated with GA at birth (PFDR<0.050). Negative associations were observed for eight miRNA (miR-4454+ miR-7975, miR-4516, let-7b-5p, miR-126-3p, miR-29b-3p, miR-15a-5p, miR-15b-5p, miR-19b-3p) and positive associations for five miRNA (miR-212-3p, miR-584-5p, miR-608, miR-210-3p, miR-188-5p). Predicted target genes were enriched (PFDR<0.050) in pathways involved in organogenesis and placental development. An additional miRNA (miR-107), measured in late pregnancy, was positively associated with GA at birth in infants born to obese women (PFDR for BMI interaction = 0.011). In primary analyses, the associations between early pregnancy miRNA and birth outcomes were not statistically significant (PFDR≥0.05). However, sex-specific associations were observed for early pregnancy measures of 37 miRNA and GA at birth (PFDR for interactions<0.050). None of the miRNA were associated with fetal growth measures (PFDR≥0.050). Our findings suggest that EV miRNA in both early and late pregnancy may influence gestational duration.
Collapse
Affiliation(s)
- Caitlin G Howe
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Lebanon, NH, USA
| | - Helen B Foley
- Department of Preventive Medicine, Keck School of Medicine at the University of Southern California, Los Angeles, CA, USA
| | - Elizabeth M Kennedy
- Gangarosa Department of Environmental Health, Emory Rollins School of Public Health, Atlanta, GA, USA
| | - Sandrah P Eckel
- Department of Preventive Medicine, Keck School of Medicine at the University of Southern California, Los Angeles, CA, USA
| | - Thomas A Chavez
- Department of Preventive Medicine, Keck School of Medicine at the University of Southern California, Los Angeles, CA, USA
| | - Dema Faham
- Department of Preventive Medicine, Keck School of Medicine at the University of Southern California, Los Angeles, CA, USA
| | - Brendan H Grubbs
- Department of Obstetrics and Gynecology, Keck School of Medicine, Los Angeles, CA, USA
| | - Laila Al-Marayati
- Department of Obstetrics and Gynecology, Keck School of Medicine, Los Angeles, CA, USA.,Eisner Health, Los Angeles, CA, USA
| | | | - Shakira Suglia
- Department of Epidemiology, Emory Rollins School of Public Health, Atlanta, GA, USA
| | - Theresa M Bastain
- Department of Preventive Medicine, Keck School of Medicine at the University of Southern California, Los Angeles, CA, USA
| | - Carmen J Marsit
- Gangarosa Department of Environmental Health, Emory Rollins School of Public Health, Atlanta, GA, USA.,Department of Epidemiology, Emory Rollins School of Public Health, Atlanta, GA, USA
| | - Carrie V Breton
- Department of Preventive Medicine, Keck School of Medicine at the University of Southern California, Los Angeles, CA, USA
| |
Collapse
|
9
|
Melnik BC. Lifetime Impact of Cow's Milk on Overactivation of mTORC1: From Fetal to Childhood Overgrowth, Acne, Diabetes, Cancers, and Neurodegeneration. Biomolecules 2021; 11:404. [PMID: 33803410 PMCID: PMC8000710 DOI: 10.3390/biom11030404] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/04/2021] [Accepted: 03/04/2021] [Indexed: 02/07/2023] Open
Abstract
The consumption of cow's milk is a part of the basic nutritional habits of Western industrialized countries. Recent epidemiological studies associate the intake of cow's milk with an increased risk of diseases, which are associated with overactivated mechanistic target of rapamycin complex 1 (mTORC1) signaling. This review presents current epidemiological and translational evidence linking milk consumption to the regulation of mTORC1, the master-switch for eukaryotic cell growth. Epidemiological studies confirm a correlation between cow's milk consumption and birthweight, body mass index, onset of menarche, linear growth during childhood, acne vulgaris, type 2 diabetes mellitus, prostate cancer, breast cancer, hepatocellular carcinoma, diffuse large B-cell lymphoma, neurodegenerative diseases, and all-cause mortality. Thus, long-term persistent consumption of cow's milk increases the risk of mTORC1-driven diseases of civilization. Milk is a highly conserved, lactation genome-controlled signaling system that functions as a maternal-neonatal relay for optimized species-specific activation of mTORC1, the nexus for regulation of eukaryotic cell growth, and control of autophagy. A deeper understanding of milk´s impact on mTORC1 signaling is of critical importance for the prevention of common diseases of civilization.
Collapse
Affiliation(s)
- Bodo C Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, Am Finkenhügel 7a, D-49076 Osnabrück, Germany
| |
Collapse
|
10
|
MicroRNA-21-Enriched Exosomes as Epigenetic Regulators in Melanomagenesis and Melanoma Progression: The Impact of Western Lifestyle Factors. Cancers (Basel) 2020; 12:cancers12082111. [PMID: 32751207 PMCID: PMC7464294 DOI: 10.3390/cancers12082111] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 07/16/2020] [Accepted: 07/24/2020] [Indexed: 02/06/2023] Open
Abstract
DNA mutation-induced activation of RAS-BRAF-MEK-ERK signaling associated with intermittent or chronic ultraviolet (UV) irradiation cannot exclusively explain the excessive increase of malignant melanoma (MM) incidence since the 1950s. Malignant conversion of a melanocyte to an MM cell and metastatic MM is associated with a steady increase in microRNA-21 (miR-21). At the epigenetic level, miR-21 inhibits key tumor suppressors of the RAS-BRAF signaling pathway enhancing proliferation and MM progression. Increased MM cell levels of miR-21 either result from endogenous upregulation of melanocytic miR-21 expression or by uptake of miR-21-enriched exogenous exosomes. Based on epidemiological data and translational evidence, this review provides deeper insights into environmentally and metabolically induced exosomal miR-21 trafficking beyond UV-irradiation in melanomagenesis and MM progression. Sources of miR-21-enriched exosomes include UV-irradiated keratinocytes, adipocyte-derived exosomes in obesity, airway epithelium-derived exosomes generated by smoking and pollution, diet-related exosomes and inflammation-induced exosomes, which may synergistically increase the exosomal miR-21 burden of the melanocyte, the transformed MM cell and its tumor environment. Several therapeutic agents that suppress MM cell growth and proliferation attenuate miR-21 expression. These include miR-21 antagonists, metformin, kinase inhibitors, beta-blockers, vitamin D, and plant-derived bioactive compounds, which may represent new options for the prevention and treatment of MM.
Collapse
|
11
|
Gerson KD, Haviland MJ, Neo D, Hecht JL, Baccarelli AA, Brennan KJM, Dereix AE, Ralston SJ, Hacker MR, Burris HH. Pregnancy-associated changes in cervical noncoding RNA. Epigenomics 2020; 12:1013-1025. [PMID: 32808540 PMCID: PMC7546170 DOI: 10.2217/epi-2019-0231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 03/20/2020] [Indexed: 12/16/2022] Open
Abstract
Aim: To identify pregnancy-associated changes in cervical noncoding RNA (ncRNA), including miRNA and long noncoding RNA (lncRNA), and their potential effects on biologic processes. Materials & methods: We enrolled 21 pregnant women with term deliveries (≥37 weeks' gestation) in a prospective cohort and collected cervical swabs before 28 weeks' gestation. We enrolled 21 nonpregnant controls. We analyzed miRNA, lncRNA and mRNA expression, applying a Bonferroni correction. Results: Five miRNA and three lncRNA were significantly differentially (>twofold change) expressed. Putative miRNA targets are enriched in genes mediating organogenesis, glucocorticoid signaling, cell adhesion and ncRNA machinery. Conclusion: Differential cervical ncRNA expression occurs in the setting of pregnancy. Gene ontology classification reveals biological pathways through which miRNA may play a biologic role in normal pregnancy physiology.
Collapse
Affiliation(s)
- Kristin D Gerson
- Department of Obstetrics & Gynecology, Maternal Child Health Research Center, Center for Research on Reproduction & Women's Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| | - Miriam J Haviland
- Department of Obstetrics & Gynecology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Dayna Neo
- Department of Obstetrics & Gynecology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
| | - Jonathan L Hecht
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Department of Pathology, Harvard Medical School, Boston, MA 02115, USA
| | - Andrea A Baccarelli
- Department of Environmental Health, Columbia University Mailman School of Public Health, New York, NY 10032, USA
| | - Kasey JM Brennan
- Department of Environmental Health, Columbia University Mailman School of Public Health, New York, NY 10032, USA
| | - Alexandra E Dereix
- Department of Environmental Health, Columbia University Mailman School of Public Health, New York, NY 10032, USA
| | - Steven J Ralston
- Department of Obstetrics & Gynecology, Pennsylvania Hospital, Philadelphia, PA 19107, USA
- Department of Obstetrics & Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Michele R Hacker
- Department of Obstetrics & Gynecology, Maternal Child Health Research Center, Center for Research on Reproduction & Women's Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
- Department Obstetrics, Gynecology & Reproductive Biology, Harvard Medical School, Boston, MA 02115, USA
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA 02115, USA
| | - Heather H Burris
- Department of Pediatrics, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Neonatology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA
- Maternal Child Health Research Center, Center for Research on Reproduction & Women’s Health, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104, USA
| |
Collapse
|
12
|
Li J, Fu Z, Jiang H, Chen L, Wu X, Ding H, Xia Y, Wang X, Tang Q, Wu W. IGF2-derived miR-483-3p contributes to macrosomia through regulating trophoblast proliferation by targeting RB1CC1. Mol Hum Reprod 2019; 24:444-452. [PMID: 29939354 DOI: 10.1093/molehr/gay027] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 06/22/2018] [Indexed: 12/18/2022] Open
Abstract
STUDY QUESTION What is the role of insulin-like growth factor 2 (IGF2)-derived miR-483-3p in macrosomia? SUMMARY ANSWER IGF2-derived intronic miR-483-3p is overexpressed in macrosomia placentas, and miR-483-3p prompts HTR-8/SVneo extravillous trophoblast cell line proliferation through down-regulation of its target RB1 inducible coiled-coil 1 (RB1CC1). WHAT IS KNOWN ALREADY Macrosomia is a common pregnancy-associated disease and causes a number of adverse maternal and perinatal outcomes. The development of macrosomia is reportedly attributable to over proliferation of the placental cells. MicroRNAs (miRNAs) play an important role in the development of fetal and placenta by regulating their target genes. Here, we investigated the role of IGF2-derived intronic miR-483-3p in macrosomia. STUDY DESIGN, SIZE, DURATION The expression of IGF2, miR-483-3p and its target gene in placental tissues from 30 pregnant women who had macrosomia was compared to those of 30 gestation-matched healthy pregnant controls. For in vitro studies, the human first trimester extravillous trophoblast cell line, HTR-8/SVneo cell was used. PARTICIPANTS/MATERIALS, SETTING, METHODS Placenta tissues were collected from pregnant women who had macrosomia without diabetes or other complications (n = 30) and healthy pregnant controls (n = 30). HTR-8/SVneo cells were transfected with specific miRNA mimics or inhibitors. MiRNA and mRNA isolated from placenta tissues or cells were measured by quantitative real-time PCR. Protein was measured by western blot. Cell proliferation was assayed using a colorimetric proliferation assay method. Cell cycle and apoptosis were analyzed by flow cytometry. The putative targets of miR-483-3p were predicted using the TargetScan, miRanda, miRDB and DIANA algorithms. Dual luciferase reporter assay was used to measure the relationship of miR-483-3p and RB1CC1. MAIN RESULTS AND THE ROLE OF CHANCE IGF2-derived miR-483-3p was overexpressed in macrosomia placentas. miR-483-3p promoted proliferation in HTR-8/SVneo cells and had a positive relationship with its host gene IGF2. Subsequently, RB1CC1 was confirmed as a direct target of miR-483-3p, which may be an important mediator of cell growth regulation for miR-483-3p. LARGE SCALE DATA N/A. LIMITATIONS, REASONS FOR CAUTION The level of IGF2 and its intronic miR-483-3p in the serum of these participants was not investigated. Further studies are required to understand the mechanisms underlying the cause of the increase of IGF2 and miR-483-3p in macrosomia. WIDER IMPLICATIONS OF THE FINDINGS These findings give a new insight into the role of intronic miRNA and its host gene in the development of macrosomia. Furthermore, it may offer a new target for prognostic and therapeutic intervention for macrosomia. STUDY FUNDING/COMPETING INTEREST(S) This work was supported by awards from National Natural Science Foundation of China (Nos. 81401213, 81673217, 81703260), Jiangsu Provincial Medical Youth Talent (No. QNRC2016110), Jiangsu Overseas Visiting Scholar Program for University Prominent Young & Middle-aged Teachers and Presidents, the Priority Academic Program for the Development of Jiangsu Higher Education Institutions (Public Health and Preventive Medicine), the Education Department of Jiangsu Province (No. 16KJB330010), the Science and Technology Department of Jiangsu Province (No. BK20160227), the China Postdoctoral Science Foundation funded project (No. 2016M601892). The authors declare no competing financial interests.
Collapse
Affiliation(s)
- Jing Li
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China.,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China.,Department of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ziqiang Fu
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China.,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Hua Jiang
- State Key Laboratory of Reproductive Medicine, Department of Gynecology, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Liping Chen
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Nantong University, Nantong, China
| | - Xian Wu
- Interdisciplinary Toxicology Program, University of Georgia, Athens, GA, USA
| | - Hongjuan Ding
- State Key Laboratory of Reproductive Medicine, Department of Obstetrics, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Yankai Xia
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China.,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xinru Wang
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China.,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Qiuqin Tang
- State Key Laboratory of Reproductive Medicine, Department of Obstetrics, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Wei Wu
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China.,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
13
|
Hocaoglu M, Demirer S, Senturk H, Turgut A, Komurcu-Bayrak E. Differential expression of candidate circulating microRNAs in maternal blood leukocytes of the patients with preeclampsia and gestational diabetes mellitus. Pregnancy Hypertens 2019; 17:5-11. [PMID: 31487656 DOI: 10.1016/j.preghy.2019.04.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Revised: 03/20/2019] [Accepted: 04/15/2019] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Preeclampsia (PE) is diagnosed in women presenting with new onset hypertension accompanied by proteinuria. Gestational diabetes mellitus (GDM) is the carbohydrate intolerance that can occur in pregnancy. Neutrophil activation is related to PE and GDM. Circulating microRNAs (miRs) are small, noncoding RNA molecules. The aim of this study was to verify the expression levels of three candidate miRs in blood leukocytes of the patients with PE, GDM, and PE-GDM compared to healthy controls. STUDY DESIGN We selected miR-21-3p, miR-155-5p, and miR-16-5p which have been associated with GDM and PE. Using real-time quantitative PCR, the expression levels of miR-21-3p, miR-155-5p, miR-16-5p were analyzed in PE (n = 23), GDM (n = 19), PE, and GDM (n = 9) compared to healthy controls (n = 28). MAIN OUTCOME MEASURES The relative expression of the target miR in patient samples was compared to the calibrator and the results were expressed as relative quantification values. RESULTS There was a significant decrease in the expression levels of miR-21-3p in GDM and PE and miR-155-5p in PE group. No significant differences were observed in the expression levels of miRs in PE-GDM group. On receiving operator characteristic (ROC) analysis, areas under the curve (AUC) of the expression ratio of miR-21-3p in GDM was 0.73, and miR-21-3p, miR-155-5p in PE were 0.69 and 0.81, respectively. CONCLUSIONS Our findings indicated that decreased miR-21-3p and miR-155-5p expression levels are associated with PE and miR-21-3p levels are associated with GDM. Our study for the first time revealed that miR-21-3p, miR-16-5p and miR155-5p are not related to PE-GDM group.
Collapse
Affiliation(s)
- Meryem Hocaoglu
- Department of Obstetrics and Gynecology, Goztepe Training and Research Hospital, Istanbul Medeniyet University, Istanbul, Turkey.
| | - Selin Demirer
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Hilal Senturk
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Abdulkadir Turgut
- Department of Obstetrics and Gynecology, Goztepe Training and Research Hospital, Istanbul Medeniyet University, Istanbul, Turkey; Department of Obstetrics and Gynecology, Faculty of Medicine, Istanbul Medeniyet University, Istanbul, Turkey
| | - Evrim Komurcu-Bayrak
- Department of Genetics, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey.
| |
Collapse
|
14
|
Melnik BC, Schmitz G. Exosomes of pasteurized milk: potential pathogens of Western diseases. J Transl Med 2019; 17:3. [PMID: 30602375 PMCID: PMC6317263 DOI: 10.1186/s12967-018-1760-8] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2018] [Accepted: 12/21/2018] [Indexed: 12/16/2022] Open
Abstract
Milk consumption is a hallmark of western diet. According to common believes, milk consumption has beneficial effects for human health. Pasteurization of cow's milk protects thermolabile vitamins and other organic compounds including bioactive and bioavailable exosomes and extracellular vesicles in the range of 40-120 nm, which are pivotal mediators of cell communication via systemic transfer of specific micro-ribonucleic acids, mRNAs and regulatory proteins such as transforming growth factor-β. There is compelling evidence that human and bovine milk exosomes play a crucial role for adequate metabolic and immunological programming of the newborn infant at the beginning of extrauterine life. Milk exosomes assist in executing an anabolic, growth-promoting and immunological program confined to the postnatal period in all mammals. However, epidemiological and translational evidence presented in this review indicates that continuous exposure of humans to exosomes of pasteurized milk may confer a substantial risk for the development of chronic diseases of civilization including obesity, type 2 diabetes mellitus, osteoporosis, common cancers (prostate, breast, liver, B-cells) as well as Parkinson's disease. Exosomes of pasteurized milk may represent new pathogens that should not reach the human food chain.
Collapse
Affiliation(s)
- Bodo C. Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, Am Finkenhügel 7A, 49076 Osnabrück, Germany
| | - Gerd Schmitz
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, University of Regensburg, Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| |
Collapse
|
15
|
Guo D, Jiang H, Chen Y, Yang J, Fu Z, Li J, Han X, Wu X, Xia Y, Wang X, Chen L, Tang Q, Wu W. Elevated microRNA-141-3p in placenta of non-diabetic macrosomia regulate trophoblast proliferation. EBioMedicine 2018; 38:154-161. [PMID: 30420300 PMCID: PMC6306401 DOI: 10.1016/j.ebiom.2018.11.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 10/21/2018] [Accepted: 11/01/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Several studies have reported microRNAs (miRNAs) could regulate the placental development, though the role and mechanism of miRNAs in the development of non-diabetic macrosomia (NDFMS) remains unclear. METHODS To identify the aberrantly expressed key miRNAs in placenta of NDFMS, we employed a strategy consisting of initial screening with miRNA microarray and further validation with quantitative RT-PCR assay (qRT-PCR). In vitro cellular model and a mouse pregnancy model were used to delineate the functional effects of key miRNA on proliferation, invasion, and migration. FINDINGS miR-141-3p was identified as the key miRNA with expression level significantly higher in placentas of NDFMS compared with those from normal controls. Overexpressed miR-141-3p in HTR-8/SVneo cells contributed to increased cell proliferation, invasion, and migration. miR-141-3p inhibition in HTR-8/SVneo cells resulted in decreased cell proliferation and invasion. Significantly increased infant birth weight was observed in late pregnancy of C57BL/6J mice treated with miR-141-3p agomir. However, no significant difference was found in early pregnancy of C57BL/6J mice treated with miR-141-3p agomir. INTERPRETATION miR-141-3p could stimulate placental cell proliferation to participate in the occurrence and development of NDFMS.
Collapse
Affiliation(s)
- Dan Guo
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China; Department of Preventive Health Branch, The Affiliated Jiangning Hospital With Nanjing Medical University, Nanjing, China
| | - Hua Jiang
- Department of Gynecology, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China
| | - Yiqiu Chen
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jing Yang
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Ziqiang Fu
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Jing Li
- Department of Public Health, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiumei Han
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xian Wu
- Interdisciplinary Toxicology Program, University of Georgia, Athens, GA, USA
| | - Yankai Xia
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xinru Wang
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Liping Chen
- Department of Gynecology and Obstetrics, Second Affiliated Hospital of Nantong University, Nantong, China.
| | - Qiuqin Tang
- Department of Obstetrics, The Affiliated Obstetrics and Gynecology Hospital of Nanjing Medical University, Nanjing Maternity and Child Health Care Hospital, Nanjing, China.
| | - Wei Wu
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing, China; Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, China; National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA.
| |
Collapse
|
16
|
Diabetes in Pregnancy and MicroRNAs: Promises and Limitations in Their Clinical Application. Noncoding RNA 2018; 4:ncrna4040032. [PMID: 30424584 PMCID: PMC6316501 DOI: 10.3390/ncrna4040032] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 10/29/2018] [Accepted: 11/05/2018] [Indexed: 12/12/2022] Open
Abstract
Maternal diabetes is associated with an increased risk of complications for the mother and her offspring. The latter have an increased risk of foetal macrosomia, hypoglycaemia, respiratory distress syndrome, preterm delivery, malformations and mortality but also of life-long development of obesity and diabetes. Epigenetics have been proposed as an explanation for this long-term risk, and microRNAs (miRNAs) may play a role, both in short- and long-term outcomes. Gestation is associated with increasing maternal insulin resistance, as well as β-cell expansion, to account for the increased insulin needs and studies performed in pregnant rats support a role of miRNAs in this expansion. Furthermore, several miRNAs are involved in pancreatic embryonic development. On the other hand, maternal diabetes is associated with changes in miRNA both in maternal and in foetal tissues. This review aims to summarise the existing knowledge on miRNAs in gestational and pre-gestational diabetes, both as diagnostic biomarkers and as mechanistic players, in the development of gestational diabetes itself and also of short- and long-term complications for the mother and her offspring.
Collapse
|
17
|
Badon SE, Littman AJ, Chan KCG, Tadesse MG, Stapleton PL, Bammler TK, Sorensen TK, Williams MA, Enquobahrie DA. Physical activity and epigenetic biomarkers in maternal blood during pregnancy. Epigenomics 2018; 10:1383-1395. [PMID: 30324807 PMCID: PMC6275564 DOI: 10.2217/epi-2017-0169] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 06/20/2018] [Indexed: 12/13/2022] Open
Abstract
AIM Investigate associations of leisure time physical activity (LTPA) with DNA methylation and miRNAs during pregnancy. Patients & methods: LTPA, candidate DNA methylation and circulating miRNAs were measured (average 15 weeks gestation) in pregnant women (n = 92). RESULTS Each additional hour of prepregnancy LTPA duration was associated with hypermethylation in C1orf212 (β = 0.137, 95% CI: 0.004-0.270) and higher circulating miR-146b-5p (β = 0.084, 95% CI: 0.017-0.151). Each additional metabolic equivalent hour of early-pregnancy LTPA energy expenditure was associated with higher circulating miR-21-3p (β = 0.431, 95% CI: 0.089-0.772) in women carrying female offspring, and lower circulating miR-146b-5p (β = -0.285, 95% CI: -0.528 to -0.043) and miR-517-5p (β = -0.406, 95% CI: -0.736 to -0.076) in women carrying male offspring. CONCLUSION Our findings suggest that LTPA may influence maternal epigenetic biomarkers, possibly in an offspring sex-specific manner.
Collapse
Affiliation(s)
- Sylvia E Badon
- Department of Epidemiology, University of Washington, Seattle, WA 98185, USA
| | - Alyson J Littman
- Department of Epidemiology, University of Washington, Seattle, WA 98185, USA
- Seattle Epidemiologic Research & Information Center, VA Puget Sound, Seattle, WA 98108, USA
| | | | - Mahlet G Tadesse
- Department of Mathematics & Statistics, Georgetown University, Washington, DC 20057, USA
| | - Patricia L Stapleton
- Department of Occupational & Environmental Health Sciences, University of Washington, Seattle, WA 98195, USA
| | - Theo K Bammler
- Department of Occupational & Environmental Health Sciences, University of Washington, Seattle, WA 98195, USA
| | - Tanya K Sorensen
- Center for Perinatal Studies, Swedish Medical Center, Seattle, WA 98104, USA
| | - Michelle A Williams
- Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA 02115, USA
| | | |
Collapse
|
18
|
Hiraku Y. [Environmental Factors and MicroRNA: Application for DOHaD Research and Future Perspectives]. Nihon Eiseigaku Zasshi 2018; 73:105-109. [PMID: 29848859 DOI: 10.1265/jjh.73.105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In Japan, the prevalence of low birth weight (< 2,500 g) has been increasing, probably owing to leanness, exposure to toxic chemicals and smoking. Epidemiological studies revealed that low birth weight poses risks of hypertension, coronary heart diseases and diabetes. Although the precise mechanism has not been understood, there is an urgent need for appropriate public health interventions. MicroRNA (miRNA) is a small RNA consisting of approximately 22 nucleotides and distributed in a wide variety of organs and body fluids. miRNAs are involved in the pathogenesis of various human diseases and expected to be their potential biomarkers. The interest on the study on miRNA in the research field of developmental origins of health and disease (DOHaD) has been growing, and the number of related papers has been increasing. There are several molecular epidemiological studies on the relationship between maternal miRNA and fetal development. The effects of smoking and dietary factors on miRNA expression and fetal development have been investigated in epidemiological and experimental studies. However, the role of maternal miRNA in fetal development has not been well understood so far. In this review, the current status of studies on miRNA expression in DOHaD research is described and future perspectives are discussed.
Collapse
Affiliation(s)
- Yusuke Hiraku
- Department of Environmental and Molecular Medicine, Mie University Graduate School of Medicine
| |
Collapse
|
19
|
Nahavandi S, Seah JM, Shub A, Houlihan C, Ekinci EI. Biomarkers for Macrosomia Prediction in Pregnancies Affected by Diabetes. Front Endocrinol (Lausanne) 2018; 9:407. [PMID: 30108547 PMCID: PMC6079223 DOI: 10.3389/fendo.2018.00407] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Accepted: 06/29/2018] [Indexed: 12/16/2022] Open
Abstract
Large birthweight, or macrosomia, is one of the commonest complications for pregnancies affected by diabetes. As macrosomia is associated with an increased risk of a number of adverse outcomes for both the mother and offspring, accurate antenatal prediction of fetal macrosomia could be beneficial in guiding appropriate models of care and interventions that may avoid or reduce these associated risks. However, current prediction strategies which include physical examination and ultrasound assessment, are imprecise. Biomarkers are proving useful in various specialties and may offer a new avenue for improved prediction of macrosomia. Prime biomarker candidates in pregnancies with diabetes include maternal glycaemic markers (glucose, 1,5-anhydroglucitol, glycosylated hemoglobin) and hormones proposed implicated in placental nutrient transfer (adiponectin and insulin-like growth factor-1). There is some support for an association of these biomarkers with birthweight and/or macrosomia, although current evidence in this emerging field is still limited. Thus, although biomarkers hold promise, further investigation is needed to elucidate the potential clinical utility of biomarkers for macrosomia prediction for pregnancies affected by diabetes.
Collapse
Affiliation(s)
- Sofia Nahavandi
- Department of Medicine, The University of Melbourne, Melbourne, VIC, Australia
| | - Jas-mine Seah
- Department of Endocrinology, Austin Health, Melbourne, VIC, Australia
| | - Alexis Shub
- Department of Medicine, The University of Melbourne, Melbourne, VIC, Australia
- Mercy Hospital for Women, Mercy Health, Melbourne, VIC, Australia
| | - Christine Houlihan
- Department of Medicine, The University of Melbourne, Melbourne, VIC, Australia
- Department of Endocrinology, Austin Health, Melbourne, VIC, Australia
- Mercy Hospital for Women, Mercy Health, Melbourne, VIC, Australia
| | - Elif I. Ekinci
- Department of Medicine, The University of Melbourne, Melbourne, VIC, Australia
- Department of Endocrinology, Austin Health, Melbourne, VIC, Australia
| |
Collapse
|
20
|
Antischistosomiasis Liver Fibrosis Effects of Chlorogenic Acid through IL-13/miR-21/Smad7 Signaling Interactions In Vivo and In Vitro. Antimicrob Agents Chemother 2017; 61:AAC.01347-16. [PMID: 27872076 DOI: 10.1128/aac.01347-16] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 11/17/2016] [Indexed: 12/23/2022] Open
Abstract
This study investigated the antischistosomiasis liver fibrosis effects of chlorogenic acid (CGA) on interleukin 13 (IL-13)/microRNA-21 (miR-21)/Smad7 signaling interactions in the hepatic stellate LX2 cell line and schistosome-infected mice. The transfection was based on the ability of the GV273-miR-21-enhanced green fluorescent protein (EGFP) and GV369-miR-21-EGFP lentiviral system to up- or downregulate the miR-21 gene in LX2 cells. The mRNA expression of miR-21, Smad7, and connective tissue growth factor (CTGF) and the protein expression of Smad7, CTGF, Smad1, phosphor-Smad1 (p-Smad1), Smad2, p-Smad2, Smad2/3, p-Smad2/3, transforming growth factor β (TGF-β) receptor I, and α-smooth muscle actin (α-SMA) was assayed. Pathological manifestation of hepatic tissue was assessed for the degree of liver fibrosis in animals. The results showed that CGA could inhibit the mRNA expression of miR-21, promote Smad7, and inhibit CTGF mRNA expression. Meanwhile, CGA could significantly lower the protein levels of CTGF, p-Smad1, p-Smad2, p-Smad2/3, TGF-β receptor I, and α-SMA and elevate the Smad7 protein level. In vivo, with treatment with CGA, the signaling molecules of IL-13/miR-21/Smad7 interactions were markedly regulated. CGA could also reduce the degree of liver fibrosis in pathological manifestations. In conclusion, CGA could inhibit schistosomiasis-induced hepatic fibrosis through IL-13/miR-21/Smad7 signaling interactions in LX2 cells and schistosome-infected mice and might serve as an antifibrosis agent for treating schistosomiasis liver fibrosis.
Collapse
|
21
|
Zheng J, Zhang Q, Mul JD, Yu M, Xu J, Qi C, Wang T, Xiao X. Maternal high-calorie diet is associated with altered hepatic microRNA expression and impaired metabolic health in offspring at weaning age. Endocrine 2016; 54:70-80. [PMID: 27106801 DOI: 10.1007/s12020-016-0959-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 04/11/2016] [Indexed: 12/22/2022]
Abstract
High-calorie diet (HCD) feeding in mice predisposes offspring for impaired glucose homeostasis and obesity. However, the mechanisms underlying these detrimental effects of maternal nutrition, especially during early life of offspring, are incompletely understood. MicroRNAs (miRNAs) are small non-coding RNAs that can regulate target gene expression. Here we hypothesized that impaired metabolic health in offspring from HCD-fed dams at weaning is associated with dysregulated expression of hepatic miRNAs. Dams were fed a chow diet (CD; 11.4 % kcal fat, 62.8 % from carbohydrate, 25.8 % from protein) or HCD (58 % kcal from fat; 25.6 % from carbohydrate, 16.4 % from protein) during gestation and lactation, and metabolic health was assessed in male offspring at weaning. Hepatic levels of miRNAs and target genes were investigated in offspring from CD- or HCD-fed dams using gene and protein expression. Maternal HCD feeding impaired metabolic health in offspring compared to offspring from CD-fed dams. Microarray analysis indicated that expressions of miR-615-5p, miR-3079-5p, miR-124*, and miR-101b* were downregulated, whereas miR-143* was upregulated, in livers from offspring from HCD-fed dams. Our functional enrichment analysis indicated that the target genes of these differentially expressed miRNAs, including tumor necrosis factor-α (TNF-α) and mitogen-activated protein kinase 1 (MAPK1), were mapped to inflammatory pathways. Finally, we verified that both mRNA and protein levels of the pro-inflammatory modulators TNF-α and MAPK1 were significantly increased in livers of offspring from HCD-fed dams at weaning. Maternal HCD feeding predisposes offspring to a higher body weight and impaired glucose metabolism at weaning. To the best of knowledge, our study is the first to show that maternal HCD consumption impairs metabolic health, modulates hepatic miRNA expression, and increases markers of hepatic inflammation in offspring as early as at weaning age.
Collapse
Affiliation(s)
- Jia Zheng
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Diabetes Research Center of Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Dongcheng District, Beijing, 100730, China
| | - Qian Zhang
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Diabetes Research Center of Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Dongcheng District, Beijing, 100730, China
| | - Joram D Mul
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Miao Yu
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Diabetes Research Center of Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Dongcheng District, Beijing, 100730, China
| | - Jianping Xu
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Diabetes Research Center of Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Dongcheng District, Beijing, 100730, China
| | - Cuijuan Qi
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Diabetes Research Center of Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Dongcheng District, Beijing, 100730, China
| | - Tong Wang
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Diabetes Research Center of Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Dongcheng District, Beijing, 100730, China
| | - Xinhua Xiao
- Department of Endocrinology, Key Laboratory of Endocrinology, Ministry of Health, Peking Union Medical College Hospital, Diabetes Research Center of Chinese Academy of Medical Sciences and Peking Union Medical College, No. 1 Shuaifuyuan, Wangfujing Street, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
22
|
Yang F, Wang Y, Xue J, Ma Q, Zhang J, Chen YF, Shang ZZ, Li QQ, Zhang SL, Zhao L. Effect of Corilagin on the miR-21/smad7/ERK signaling pathway in a schistosomiasis-induced hepatic fibrosis mouse model. Parasitol Int 2016; 65:308-15. [DOI: 10.1016/j.parint.2016.03.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 02/28/2016] [Accepted: 03/01/2016] [Indexed: 12/28/2022]
|
23
|
ZHANG JITAI, CAI QIANYING, JI SISI, ZHANG HENGXIN, WANG YUHUAN, YAN HONGTAO, YANG XINJUN. Decreased miR-143 and increased miR-21 placental expression levels are associated with macrosomia. Mol Med Rep 2016; 13:3273-80. [DOI: 10.3892/mmr.2016.4892] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 01/08/2016] [Indexed: 11/05/2022] Open
|
24
|
Li J, Chen L, Qiuqin Tang, Wu W, Hao Gu, Lou Liu, Jie Wu, Hua Jiang, Hongjuan Ding, Xia Y, Chen D, Hu Y, Wang X. The role, mechanism and potentially novel biomarker of microRNA-17-92 cluster in macrosomia. Sci Rep 2015; 5:17212. [PMID: 26598317 PMCID: PMC4657041 DOI: 10.1038/srep17212] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 10/26/2015] [Indexed: 12/16/2022] Open
Abstract
Macrosomia is one of the most common perinatal complications of pregnancy and has life-long health implications for the infant. microRNAs (miRNAs) have been identified to regulate placental development, yet the role of miRNAs in macrosomia remains poorly understood. Here we investigated the role of miR-17-92 cluster in macrosomia. The expression levels of five miRNAs in miR-17-92 cluster were significantly elevated in placentas of macrosomia, which may due to the up-regulation of miRNA-processing enzyme Drosha and Dicer. Cell cycle pathway was identified to be the most relevant pathways regulated by miR-17-92 cluster miRNAs. Importantly, miR-17-92 cluster increased proliferation, attenuated cell apoptosis and accelerated cells entering S phase by targeting SMAD4 and RB1 in HTR8/SVneo cells. Furthermore, we found that expression of miR-17-92 cluster in serum had a high diagnostic sensitivity and specificity for macrosomia (AUC: 80.53%; sensitivity: 82.61%; specificity: 69.57%). Our results suggested that miR-17-92 cluster contribute to macrosomia development by targeting regulators of cell cycle pathway. Our findings not only provide a novel insight into the molecular mechanisms of macrosomia, but also the clinical value of miR-17-92 cluster as a predictive biomarker for macrosomia.
Collapse
Affiliation(s)
- Jing Li
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing 211166, China.,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China.,Department of Public Health, Xuzhou Medical College, Xuzhou, Jiangsu, China
| | - Liping Chen
- Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China.,Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Nantong University, Nantong, 226001, China.,Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Qiuqin Tang
- State Key Laboratory of Reproductive Medicine, Department of Obstetrics, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing 210004, China
| | - Wei Wu
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing 211166, China.,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China.,State Key Laboratory of Reproductive Medicine, Wuxi Maternal and Child Health Care Hospital Affiliated to Nanjing Medical University, Wuxi 214002, China
| | - Hao Gu
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing 211166, China.,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Lou Liu
- State Key Laboratory of Reproductive Medicine, Wuxi Maternal and Child Health Care Hospital Affiliated to Nanjing Medical University, Wuxi 214002, China
| | - Jie Wu
- State Key Laboratory of Reproductive Medicine, Wuxi Maternal and Child Health Care Hospital Affiliated to Nanjing Medical University, Wuxi 214002, China
| | - Hua Jiang
- Perinatology Unit, Changzhou Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Changzhou 213003, China
| | - Hongjuan Ding
- State Key Laboratory of Reproductive Medicine, Department of Obstetrics, Nanjing Maternity and Child Health Care Hospital Affiliated to Nanjing Medical University, Nanjing 210004, China
| | - Yankai Xia
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing 211166, China.,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| | - Daozhen Chen
- State Key Laboratory of Reproductive Medicine, Wuxi Maternal and Child Health Care Hospital Affiliated to Nanjing Medical University, Wuxi 214002, China
| | - Yali Hu
- Reproductive Medicine Center, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Xinru Wang
- State Key Laboratory of Reproductive Medicine, Institute of Toxicology, Nanjing Medical University, Nanjing 211166, China.,Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
25
|
MicroRNAs in Breastmilk and the Lactating Breast: Potential Immunoprotectors and Developmental Regulators for the Infant and the Mother. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2015; 12:13981-4020. [PMID: 26529003 PMCID: PMC4661628 DOI: 10.3390/ijerph121113981] [Citation(s) in RCA: 144] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 10/21/2015] [Accepted: 10/27/2015] [Indexed: 12/12/2022]
Abstract
Human milk (HM) is the optimal source of nutrition, protection and developmental programming for infants. It is species-specific and consists of various bioactive components, including microRNAs, small non-coding RNAs regulating gene expression at the post-transcriptional level. microRNAs are both intra- and extra-cellular and are present in body fluids of humans and animals. Of these body fluids, HM appears to be one of the richest sources of microRNA, which are highly conserved in its different fractions, with milk cells containing more microRNAs than milk lipids, followed by skim milk. Potential effects of exogenous food-derived microRNAs on gene expression have been demonstrated, together with the stability of milk-derived microRNAs in the gastrointestinal tract. Taken together, these strongly support the notion that milk microRNAs enter the systemic circulation of the HM fed infant and exert tissue-specific immunoprotective and developmental functions. This has initiated intensive research on the origin, fate and functional significance of milk microRNAs. Importantly, recent studies have provided evidence of endogenous synthesis of HM microRNA within the human lactating mammary epithelium. These findings will now form the basis for investigations of the role of microRNA in the epigenetic control of normal and aberrant mammary development, and particularly lactation performance.
Collapse
|
26
|
AngiomiRs: Potential Biomarkers of Pregnancy's Vascular Pathologies. J Pregnancy 2015; 2015:320386. [PMID: 26550492 PMCID: PMC4621355 DOI: 10.1155/2015/320386] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 09/27/2015] [Indexed: 12/29/2022] Open
Abstract
In recent years, microRNAs (miRNAs) have been the focus of research for their role in posttranscriptional regulation and as potential biomarkers of risk for disease development. Their identification in specific physiological processes, like angiogenesis, a key pathway in placental vascular development in pregnancy, suggests an important role of miRNAs that regulate angiogenesis (angiomiRs). Many complications of pregnancy have in common placental vascular alterations, involving an imbalance in the angiogenesis process in the development of conditions such as preeclampsia, intrauterine growth restriction, and gestational diabetes, complications with the highest rates of morbimortality in pregnancy. Many studies have identified angiomiRs with differential expression profiles in each of these diseases; however, this evidence requires further studies focused on evaluating their potential as biomarkers of risk for the angiomiRs detected, to establish correlations between placental tissue and serum/plasma expression profiles. Therefore, the objective of this review is to highlight the best angiomiRs detected in placental tissue and serum/plasma in each of these three pathologies to show the current data available for potential biomarkers and to propose future research strategies on this topic.
Collapse
|
27
|
Abstract
Based on own translational research of the biochemical and hormonal effects of cow's milk consumption in humans, this review presents milk as a signaling system of mammalian evolution that activates the nutrient-sensitive kinase mechanistic target of rapamycin complex 1 (mTORC1), the pivotal regulator of translation. Milk, a mammary gland-derived secretory product, is required for species-specific gene-nutrient interactions that promote appropriate growth and development of the newborn mammal. This signaling system is highly conserved and tightly controlled by the lactation genome. Milk is sufficient to activate mTORC1, the crucial regulator of protein, lipid, and nucleotide synthesis orchestrating anabolism, cell growth and proliferation. To fulfill its mTORC1-activating function, milk delivers four key metabolic messengers: (1) essential branched-chain amino acids (BCAAs); (2) glutamine; (3) palmitic acid; and (4) bioactive exosomal microRNAs, which in a synergistical fashion promote mTORC1-dependent translation. In all mammals except Neolithic humans, postnatal activation of mTORC1 by milk intake is restricted to the postnatal lactation period. It is of critical concern that persistent hyperactivation of mTORC1 is associated with aging and the development of age-related disorders such as obesity, type 2 diabetes mellitus, cancer, and neurodegenerative diseases. Persistent mTORC1 activation promotes endoplasmic reticulum (ER) stress and drives an aimless quasi-program, which promotes aging and age-related diseases.
Collapse
|
28
|
Melnik BC, John SM, Schmitz G. Milk consumption during pregnancy increases birth weight, a risk factor for the development of diseases of civilization. J Transl Med 2015; 13:13. [PMID: 25592553 PMCID: PMC4302093 DOI: 10.1186/s12967-014-0377-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 12/29/2014] [Indexed: 02/06/2023] Open
Abstract
Antenatal dietary lifestyle intervention and nutrition during pregnancy and early postnatal life are important for appropriate lifelong metabolic programming. Epidemiological evidence underlines the crucial role of increased birth weight as a risk factor for the development of chronic diseases of civilization such as obesity, diabetes and cancer. Obstetricians and general practitioners usually recommend milk consumption during pregnancy as a nutrient enriched in valuable proteins and calcium for bone growth. However, milk is not just a simple nutrient, but has been recognized to function as an endocrine signaling system promoting anabolism and postnatal growth by activating the nutrient-sensitive kinase mTORC1. Moreover, pasteurized cow’s milk transfers biologically active exosomal microRNAs into the systemic circulation of the milk consumer apparently affecting more than 11 000 human genes including the mTORC1-signaling pathway. This review provides literature evidence and evidence derived from translational research that milk consumption during pregnancy increases gestational, placental, fetal and birth weight. Increased birth weight is a risk factor for the development of diseases of civilization thus involving key disciplines of medicine. With regard to the presented evidence we suggest that dietary recommendations promoting milk consumption during pregnancy have to be re-evaluated.
Collapse
Affiliation(s)
- Bodo C Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, Sedanstrasse 115, D-49090, Osnabrück, Germany.
| | - Swen Malte John
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabrück, Sedanstrasse 115, D-49090, Osnabrück, Germany.
| | - Gerd Schmitz
- Institute of Clinical Chemistry and Laboratory Medicine, University Clinics of Regensburg, Regensburg, Germany.
| |
Collapse
|
29
|
Melnik BC. The pathogenic role of persistent milk signaling in mTORC1- and milk-microRNA-driven type 2 diabetes mellitus. Curr Diabetes Rev 2015; 11:46-62. [PMID: 25587719 PMCID: PMC4428476 DOI: 10.2174/1573399811666150114100653] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Revised: 01/08/2015] [Accepted: 01/09/2015] [Indexed: 12/12/2022]
Abstract
Milk, the secretory product of the lactation genome, promotes growth of the newborn mammal. Milk delivers insulinotropic amino acids, thus maintains a molecular crosstalk with the pancreatic β-cell of the milk recipient. Homeostasis of β-cells and insulin production depend on the appropriate magnitude of mTORC1 signaling. mTORC1 is activated by branched-chain amino acids (BCAAs), glutamine, and palmitic acid, abundant nutrient signals of cow´s milk. Furthermore, milk delivers bioactive exosomal microRNAs. After milk consumption, bovine microRNA-29b, a member of the diabetogenic microRNA-29- family, reaches the systemic circulation and the cells of the milk consumer. MicroRNA-29b downregulates branchedchain α-ketoacid dehydrogenase, a potential explanation for increased BCAA serum levels, the metabolic signature of insulin resistance and type 2 diabetes mellitus (T2DM). In non-obese diabetic mice, microRNA-29b downregulates the antiapoptotic protein Mcl-1, which leads to early β-cell death. In all mammals except Neolithic humans, milk-driven mTORC1 signaling is physiologically restricted to the postnatal period. In contrast, chronic hyperactivated mTORC1 signaling has been associated with the development of age-related diseases of civilization including T2DM. Notably, chronic hyperactivation of mTORC1 enhances endoplasmic reticulum stress that promotes apoptosis. In fact, hyperactivated β-cell mTORC1 signaling induced early β-cell apoptosis in a mouse model. The EPIC-InterAct Study demonstrated an association between milk consumption and T2DM in France, Italy, United Kingdom, Germany, and Sweden. In contrast, fermented milk products and cheese exhibit an inverse correlation. Since the early 1950´s, refrigeration technology allowed widespread consumption of fresh pasteurized milk, which facilitates daily intake of bioactive bovine microRNAs. Persistent uptake of cow´s milk-derived microRNAs apparently transfers an overlooked epigenetic diabetogenic program that should not reach the human food chain.
Collapse
Affiliation(s)
- Bodo C Melnik
- Department of Dermatology, Environmental Medicine and Health Theory, University of Osnabruck, Sedanstraße 115, D-49090 Osnabrück, Germany.
| |
Collapse
|
30
|
Jiang H, Wen Y, Hu L, Miao T, Zhang M, Dong J. Serum MicroRNAs as Diagnostic Biomarkers for Macrosomia. Reprod Sci 2014; 22:664-71. [PMID: 25519717 DOI: 10.1177/1933719114561557] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Macrosomia is defined as an infant's birth weight of more than 4000 g. Although microRNAs (miRNAs) have been implicated in the pathogenesis of various diseases, the associations between serum miRNAs and macrosomia have been rarely reported. METHODOLOGY We used the Taqman Low Density Array followed by quantitative reverse transcriptase polymerase chain reaction assays to screen for miRNAs associated with macrosomia using serum samples collected 1 week before delivery. RESULTS Profiling results showed that 1 miRNA was significantly upregulated and 10 miRNAs were significantly downregulated in serum samples of macrosomia (ΔΔCt > 3-fold). The expression levels of miR-21 were significantly decreased in macrosomia as compared to the controls in the third trimester. Receiver operating characteristic (ROC) curve analyses showed that the area under the ROC curve for miR-21 was 67.7% (sensitivity = 66.7% and specificity = 70.0%). CONCLUSIONS miR-21 in maternal serum is differentially expressed between macrosomia and controls, and miR-21 could be used as a candidate biomarker to predict macrosomia.
Collapse
Affiliation(s)
- Hua Jiang
- Perinatology Unit, Changzhou Maternity and Child Health Care Hospital affiliated to Nanjing Medical University, Changzhou, P.R. China
| | - Yang Wen
- Department of Epidemiology and Biostatistics, State Key Lab of Reproductive Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Lingmin Hu
- Perinatology Unit, Changzhou Maternity and Child Health Care Hospital affiliated to Nanjing Medical University, Changzhou, P.R. China
| | - Tingting Miao
- Perinatology Unit, Changzhou Maternity and Child Health Care Hospital affiliated to Nanjing Medical University, Changzhou, P.R. China
| | - Ming Zhang
- Perinatology Unit, Changzhou Maternity and Child Health Care Hospital affiliated to Nanjing Medical University, Changzhou, P.R. China
| | - Jing Dong
- Department of Epidemiology and Biostatistics, State Key Lab of Reproductive Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
31
|
Unverdorben L, Hüttenbrenner R, Knabl J, Jeschke U, Hutter S. Galectin-13/PP-13 expression in term placentas of gestational diabetes mellitus pregnancies. Placenta 2014; 36:191-8. [PMID: 25499680 DOI: 10.1016/j.placenta.2014.11.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 11/04/2014] [Accepted: 11/25/2014] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Gestational diabetes mellitus (GDM) is an increasing harm in pregnancy. Inflammatory processes in the placenta seem to have an influence on pathogenesis besides known factors like maternal BMI. Galectin-13 (gal-13) is an immunoregulatory protein, which is suspected to play a role in development of GDM in the placenta. METHODS A total of 40 placentas were obtained from women treated for gestational diabetes mellitus. Placental tissue for control group was obtained from 40 women with normal pregnancy. We investigated the protein expression of gal-13 in term placentas with immunohistochemistry and immunofluorescence. Immunohistochemical staining was analyzed with the semi-quantified IRS score. Gal-13 serum levels were performed with ELISA on a total of 20 probes from women with GDM and healthy control pregnancies in the third trimester. RESULTS Gal-13 was found in syncytiotrophoblast, in nuclei of syncytiotrophoblast and trophoblast cells as well in extravillous trophoblast cells of normal placentas. In GDM placentas, gal-13 expression was significantly decreased in all of these examined cell types (syncytiotrophoblast p = 0.003, nuclei of syncytiotrophoblast p = 0.007; extravillous trophoblast cells p = 0.001). The ELISA showed a significant lower gal-13 serum level in blood from pregnant women with GDM in comparison to healthy controls. DISCUSSION As gal-13 with its anti-inflammatory functions plays a role in regulation of maternal immune system, a lack of gal-13 may contribute to an imbalance in inflammation processes in the placenta during pregnancy and therefore influences development of GDM.
Collapse
Affiliation(s)
- L Unverdorben
- Department of Gynecology and Obstetrics, Ludwig Maximilians University of Munich, Maistraße 11, 80337 Munich, Germany
| | - R Hüttenbrenner
- Department of Gynecology and Obstetrics, Ludwig Maximilians University of Munich, Maistraße 11, 80337 Munich, Germany
| | - J Knabl
- Department of Gynecology and Obstetrics, Klinik Hallerwiese, Diakonie Neuendettelsau, St.-Johannis-Mühlgasse 19, 90419 Nuremberg, Germany
| | - U Jeschke
- Department of Gynecology and Obstetrics, Ludwig Maximilians University of Munich, Maistraße 11, 80337 Munich, Germany.
| | - S Hutter
- Department of Gynecology and Obstetrics, Ludwig Maximilians University of Munich, Maistraße 11, 80337 Munich, Germany
| |
Collapse
|
32
|
Miura K, Higashijima A, Hasegawa Y, Abe S, Miura S, Kaneuchi M, Yoshiura KI, Masuzaki H. Circulating levels of maternal plasma cell-free miR-21 are associated with maternal body mass index and neonatal birth weight. Prenat Diagn 2014; 35:509-11. [PMID: 25273622 DOI: 10.1002/pd.4509] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Accepted: 09/24/2014] [Indexed: 12/28/2022]
Affiliation(s)
- Kiyonori Miura
- Department of Obstetrics and Gynecology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Early second-trimester serum microRNAs as potential biomarker for nondiabetic macrosomia. BIOMED RESEARCH INTERNATIONAL 2014; 2014:394125. [PMID: 25405200 PMCID: PMC4227359 DOI: 10.1155/2014/394125] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 08/19/2014] [Accepted: 08/27/2014] [Indexed: 12/13/2022]
Abstract
Background. Macrosomia has become a worldwide problem with the rapid economic growth in the past few years. However, the detailed mechanism of how the macrosomia happened remains unknown. Growing evidence indicates that miRNAs are involved in maintaining metabolic homeostasis. We hypothesized that serum miRNAs are potential biomarkers for macrosomia. Methods. We performed miRNAs profiling using TLDA chips in the discovery phase in two pooled samples from 30 cases and 30 controls, respectively. Individual qRT-PCR was conducted for the discovery phase samples. To confirm the results, we detected the miRNAs which were differentially expressed in the microarray assays and individual qRT-PCR in external validation phase with another 30 cases and 30 controls. Results. In the discovery stage, miR-194 and miR-376a expression levels were significantly different between macrosomia group and controls (P = 0.048 for miR-194 and P = 0.018 for miR-376a, resp.). Further evaluation of the two miRNAs on a total of 120 serum samples showed that the miR-376a remains significantly lower in macrosomia (P = 0.032). Receiver operating characteristic curve analyses showed that the area under curve for miR-376a was 67.8% (sensitivity = 96.7% and specificity = 40.0%). Conclusions. Serum miR-376a may serve as a potential noninvasive biomarker in detecting macrosomia.
Collapse
|