1
|
Sandovici I, Fernandez-Twinn DS, Campbell N, Cooper WN, Sekita Y, Zvetkova I, Ferland-McCollough D, Prosser HM, Oyama LM, Pantaleão LC, Cimadomo D, Barbosa de Queiroz K, Cheuk CSK, Smith NM, Kay RG, Antrobus R, Hoelle K, Ma MKL, Smith NH, Geyer SH, Reissig LF, Weninger WJ, Siddle K, Willis AE, Lam BYH, Bushell M, Ozanne SE, Constância M. Overexpression of Igf2-derived Mir483 inhibits Igf1 expression and leads to developmental growth restriction and metabolic dysfunction in mice. Cell Rep 2024; 43:114750. [PMID: 39283743 DOI: 10.1016/j.celrep.2024.114750] [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: 01/10/2023] [Revised: 06/04/2024] [Accepted: 08/28/2024] [Indexed: 09/28/2024] Open
Abstract
Mir483 is a conserved and highly expressed microRNA in placental mammals, embedded within the Igf2 gene. Its expression is dysregulated in a number of human diseases, including metabolic disorders and certain cancers. Here, we investigate the developmental regulation and function of Mir483 in vivo. We find that Mir483 expression is dependent on Igf2 transcription and the regulation of the Igf2/H19 imprinting control region. Transgenic Mir483 overexpression in utero causes fetal, but not placental, growth restriction through insulin-like growth factor 1 (IGF1) and IGF2 and also causes cardiovascular defects leading to fetal death. Overexpression of Mir483 post-natally results in growth stunting through IGF1 repression, increased hepatic lipid production, and excessive adiposity. IGF1 infusion rescues the post-natal growth restriction. Our findings provide insights into the function of Mir483 as a growth suppressor and metabolic regulator and suggest that it evolved within the INS-IGF2-H19 transcriptional region to limit excessive tissue growth through repression of IGF signaling.
Collapse
Affiliation(s)
- Ionel Sandovici
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK; Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK; Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Denise S Fernandez-Twinn
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Niamh Campbell
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK; Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Wendy N Cooper
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK; Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Yoichi Sekita
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK; Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Ilona Zvetkova
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | | | - Haydn M Prosser
- The Wellcome Trust Sanger Institute, Genome Campus, Hinxton, UK
| | - Lila M Oyama
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK; Departmento de Fisiologia, Universidade Federal de São Paulo, Escola Paulista de Medicina, São Paulo, Brazil
| | - Lucas C Pantaleão
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Danilo Cimadomo
- Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK; Laboratory of Developmental Biology, Department of Biology and Biotechnology "Lazzaro Spallanzani," University of Pavia, Pavia, Italy
| | - Karina Barbosa de Queiroz
- Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Cecilia S K Cheuk
- Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK; Nuffield Department of Women's & Reproductive Health, University of Oxford, Oxford, UK
| | - Nicola M Smith
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Richard G Kay
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Robin Antrobus
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK
| | - Katharina Hoelle
- Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK
| | - Marcella K L Ma
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Noel H Smith
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Stefan H Geyer
- Center for Anatomy and Cell Biology, Division of Anatomy, Medical University of Vienna, Vienna, Austria
| | - Lukas F Reissig
- Center for Anatomy and Cell Biology, Division of Anatomy, Medical University of Vienna, Vienna, Austria
| | - Wolfgang J Weninger
- Center for Anatomy and Cell Biology, Division of Anatomy, Medical University of Vienna, Vienna, Austria
| | - Kenneth Siddle
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Anne E Willis
- Medical Research Council Toxicology Unit, University of Leicester, Leicester, UK
| | - Brian Y H Lam
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK
| | - Martin Bushell
- Medical Research Council Toxicology Unit, University of Leicester, Leicester, UK
| | - Susan E Ozanne
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK; Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK
| | - Miguel Constância
- Medical Research Council Metabolic Diseases Unit, Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, UK; Department of Obstetrics and Gynaecology and National Institute for Health Research Cambridge Biomedical Research Centre, Cambridge, UK; Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, UK.
| |
Collapse
|
2
|
Basak S, Mallick R, Navya Sree B, Duttaroy AK. Placental Epigenome Impacts Fetal Development: Effects of Maternal Nutrients and Gut Microbiota. Nutrients 2024; 16:1860. [PMID: 38931215 PMCID: PMC11206482 DOI: 10.3390/nu16121860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/07/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Evidence is emerging on the role of maternal diet, gut microbiota, and other lifestyle factors in establishing lifelong health and disease, which are determined by transgenerationally inherited epigenetic modifications. Understanding epigenetic mechanisms may help identify novel biomarkers for gestation-related exposure, burden, or disease risk. Such biomarkers are essential for developing tools for the early detection of risk factors and exposure levels. It is necessary to establish an exposure threshold due to nutrient deficiencies or other environmental factors that can result in clinically relevant epigenetic alterations that modulate disease risks in the fetus. This narrative review summarizes the latest updates on the roles of maternal nutrients (n-3 fatty acids, polyphenols, vitamins) and gut microbiota on the placental epigenome and its impacts on fetal brain development. This review unravels the potential roles of the functional epigenome for targeted intervention to ensure optimal fetal brain development and its performance in later life.
Collapse
Affiliation(s)
- Sanjay Basak
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad 500007, India; (S.B.); (B.N.S.)
| | - Rahul Mallick
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland;
| | - Boga Navya Sree
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad 500007, India; (S.B.); (B.N.S.)
| | - Asim K. Duttaroy
- Department of Nutrition, Institute of Medical Sciences, Faculty of Medicine, University of Oslo, 0317 Oslo, Norway
| |
Collapse
|
3
|
Jiao P, Lu H, Hao L, Degen AA, Cheng J, Yin Z, Mao S, Xue Y. Nutrigenetic and Epigenetic Mechanisms of Maternal Nutrition-Induced Glucolipid Metabolism Changes in the Offspring. Nutr Rev 2024:nuae048. [PMID: 38781288 DOI: 10.1093/nutrit/nuae048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024] Open
Abstract
Maternal nutrition during pregnancy regulates the offspring's metabolic homeostasis, including insulin sensitivity and the metabolism of glucose and lipids. The fetus undergoes a crucial period of plasticity in the uterus; metabolic changes in the fetus during pregnancy caused by maternal nutrition not only influence fetal growth and development but also have a long-term or even life-long impact for the offspring. Epigenetic modifications, such as DNA methylation, histone modification, and non-coding RNAs, play important roles in intergenerational and transgenerational effects. In this context, this narrative review comprehensively summarizes and analyzes the molecular mechanisms underlying how maternal nutrition, including a high-fat diet, polyunsaturated fatty acid diet, methyl donor nutrient supplementation, feed restriction, and protein restriction during pregnancy, impacts the genes involved in glucolipid metabolism in the liver, adipose tissue, hypothalamus, muscle, and oocytes of the offspring in terms of the epigenetic modifications. This will provide a foundation for the further exploration of nutrigenetic and epigenetic mechanisms for integrative mother-child nutrition and promotion of the offspring's health through the regulation of maternal nutrition during pregnancy. Note: This paper is part of the Nutrition Reviews Special Collection on Precision Nutrition.
Collapse
Affiliation(s)
- Peng Jiao
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Huizhen Lu
- Biotechnology Center, Anhui Agricultural University, Hefei, China
| | - Lizhuang Hao
- Key Laboratory of Plateau Grazing Animal Nutrition and Feed Science of Qinghai Province, Qinghai Plateau Yak Research Center, Qinghai Academy of Science and Veterinary Medicine of Qinghai University, Xining, China
| | - A Allan Degen
- Desert Animal Adaptations and Husbandry, Wyler Department of Dryland Agriculture, Blaustein Institutes for Desert Research, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Jianbo Cheng
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Zongjun Yin
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Shengyong Mao
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yanfeng Xue
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| |
Collapse
|
4
|
Tzeng HT, Lee WC. Impact of Transgenerational Nutrition on Nonalcoholic Fatty Liver Disease Development: Interplay between Gut Microbiota, Epigenetics and Immunity. Nutrients 2024; 16:1388. [PMID: 38732634 PMCID: PMC11085251 DOI: 10.3390/nu16091388] [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: 03/15/2024] [Revised: 04/25/2024] [Accepted: 05/01/2024] [Indexed: 05/13/2024] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) has emerged as the most prevalent pediatric liver disorder, primarily attributed to dietary shifts in recent years. NAFLD is characterized by the accumulation of lipid species in hepatocytes, leading to liver inflammation that can progress to steatohepatitis, fibrosis, and cirrhosis. Risk factors contributing to NAFLD encompass genetic variations and metabolic disorders such as obesity, diabetes, and insulin resistance. Moreover, transgenerational influences, resulting in an imbalance of gut microbial composition, epigenetic modifications, and dysregulated hepatic immune responses in offspring, play a pivotal role in pediatric NAFLD development. Maternal nutrition shapes the profile of microbiota-derived metabolites in offspring, exerting significant influence on immune system regulation and the development of metabolic syndrome in offspring. In this review, we summarize recent evidence elucidating the intricate interplay between gut microbiota, epigenetics, and immunity in fetuses exposed to maternal nutrition, and its impact on the onset of NAFLD in offspring. Furthermore, potential therapeutic strategies targeting this network are also discussed.
Collapse
Affiliation(s)
- Hong-Tai Tzeng
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan;
| | - Wei-Chia Lee
- Division of Urology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 33332, Taiwan
| |
Collapse
|
5
|
Pantaleão LC, Loche E, Fernandez-Twinn DS, Dearden L, Córdova-Casanova A, Osmond C, Salonen MK, Kajantie E, Niu Y, de Almeida-Faria J, Thackray BD, Mikkola TM, Giussani DA, Murray AJ, Bushell M, Eriksson JG, Ozanne SE. Programming of cardiac metabolism by miR-15b-5p, a miRNA released in cardiac extracellular vesicles following ischemia-reperfusion injury. Mol Metab 2024; 80:101875. [PMID: 38218535 PMCID: PMC10832484 DOI: 10.1016/j.molmet.2024.101875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/22/2023] [Accepted: 01/08/2024] [Indexed: 01/15/2024] Open
Abstract
OBJECTIVE We investigated the potential involvement of miRNAs in the developmental programming of cardiovascular diseases (CVD) by maternal obesity. METHODS Serum miRNAs were measured in individuals from the Helsinki Birth Cohort (with known maternal body mass index), and a mouse model was used to determine causative effects of maternal obesity during pregnancy and ischemia-reperfusion on offspring cardiac miRNA expression and release. RESULTS miR-15b-5p levels were increased in the sera of males born to mothers with higher BMI and in the hearts of adult mice born to obese dams. In an ex-vivo model of perfused mouse hearts, we demonstrated that cardiac tissue releases miR-15b-5p, and that some of the released miR-15b-5p was contained within small extracellular vesicles (EVs). We also demonstrated that release was higher from hearts exposed to maternal obesity following ischaemia/reperfusion. Over-expression of miR-15b-5p in vitro led to loss of outer mitochondrial membrane stability and to repressed fatty acid oxidation in cardiomyocytes. CONCLUSIONS These findings suggest that miR-15-b could play a mechanistic role in the dysregulation of cardiac metabolism following exposure to an in utero obesogenic environment and that its release in cardiac EVs following ischaemic damage may be a novel factor contributing to inter-organ communication between the programmed heart and peripheral tissues.
Collapse
Affiliation(s)
- Lucas C Pantaleão
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Elena Loche
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Denise S Fernandez-Twinn
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Laura Dearden
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Adriana Córdova-Casanova
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Clive Osmond
- MRC Lifecourse Epidemiology Unit, University of Southampton, UK
| | - Minna K Salonen
- Finnish Institute for Health and Welfare, Public Health Unit, Finland
| | - Eero Kajantie
- Finnish Institute for Health and Welfare, Public Health Unit, Finland; Clinical Medicine Research Unit, MRC Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland; Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Youguo Niu
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK
| | - Juliana de Almeida-Faria
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK
| | - Benjamin D Thackray
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK; Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK
| | - Tuija M Mikkola
- Finnish Institute for Health and Welfare, Public Health Unit, Finland; Folkhalsan Research Center, Helsinki, Finland; Faculty of Medicine, University of Helsinki, Finland
| | - Dino A Giussani
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK
| | - Andrew J Murray
- Department of Physiology, Development, and Neuroscience, University of Cambridge, Cambridge, UK
| | - Martin Bushell
- CRUK Beatson Institute, Garscube Estate, Switchback Road, Bearsden, Glasgow, G61 1BD, UK
| | - Johan G Eriksson
- Folkhalsan Research Center, Helsinki, Finland; Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, Finland; Singapore Institute for Clinical Sciences, Agency for Science Technology and Research, Singapore, Singapore; Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Susan E Ozanne
- Wellcome-MRC Institute of Metabolic Science and Medical Research Council Metabolic Diseases Unit, University of Cambridge, Cambridge, UK.
| |
Collapse
|
6
|
Mouzaki M, Woo JG, Divanovic S. Gestational and Developmental Contributors of Pediatric MASLD. Semin Liver Dis 2024; 44:43-53. [PMID: 38423068 DOI: 10.1055/s-0044-1782210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Pediatric metabolic dysfunction-associated steatotic liver disease (MASLD) is common and can be seen as early as in utero. A growing body of literature suggests that gestational and early life exposures modify the risk of MASLD development in children. These include maternal risk factors, such as poor cardiometabolic health (e.g., obesity, gestational diabetes, rapid weight gain during pregnancy, and MASLD), as well as periconceptional dietary exposures, degree of physical activity, intestinal microbiome, and smoking. Paternal factors, such as diet and obesity, also appear to play a role. Beyond gestation, early life dietary exposures, as well as the rate of infant weight gain, may further modify the risk of future MASLD development. The mechanisms linking parental health and environmental exposures to pediatric MASLD are complex and not entirely understood. In conclusion, investigating gestational and developmental contributors to MASLD is critical and may identify future interventional targets for disease prevention.
Collapse
Affiliation(s)
- Marialena Mouzaki
- Divisions of Gastroenterology, Hepatology and Nutrition, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Jessica G Woo
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Biostatistics and Epidemiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Senad Divanovic
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| |
Collapse
|
7
|
La Colla A, Cámara CA, Campisano S, Chisari AN. Mitochondrial dysfunction and epigenetics underlying the link between early-life nutrition and non-alcoholic fatty liver disease. Nutr Res Rev 2023; 36:281-294. [PMID: 35067233 DOI: 10.1017/s0954422422000038] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Early-life malnutrition plays a critical role in foetal development and predisposes to metabolic diseases later in life, according to the concept of 'developmental programming'. Different types of early nutritional imbalance, including undernutrition, overnutrition and micronutrient deficiency, have been related to long-term metabolic disorders. Accumulating evidence has demonstrated that disturbances in nutrition during the period of preconception, pregnancy and primary infancy can affect mitochondrial function and epigenetic mechanisms. Moreover, even though multiple mechanisms underlying non-alcoholic fatty liver disease (NAFLD) have been described, in the past years, special attention has been given to mitochondrial dysfunction and epigenetic alterations. Mitochondria play a key role in cellular metabolic functions. Dysfunctional mitochondria contribute to oxidative stress, insulin resistance and inflammation. Epigenetic mechanisms have been related to alterations in genes involved in lipid metabolism, fibrogenesis, inflammation and tumorigenesis. In accordance, studies have reported that mitochondrial dysfunction and epigenetics linked to early-life nutrition can be important contributing factors in the pathogenesis of NAFLD. In this review, we summarise the current understanding of the interplay between mitochondrial dysfunction, epigenetics and nutrition during early life, which is relevant to developmental programming of NAFLD.
Collapse
Affiliation(s)
- Anabela La Colla
- Departamento de Química y Bioquímica, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata, 7600 Mar del Plata, Argentina
| | - Carolina Anahí Cámara
- Departamento de Química y Bioquímica, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata, 7600 Mar del Plata, Argentina
| | - Sabrina Campisano
- Departamento de Química y Bioquímica, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata, 7600 Mar del Plata, Argentina
| | - Andrea Nancy Chisari
- Departamento de Química y Bioquímica, Facultad de Ciencias Exactas y Naturales, Universidad Nacional de Mar del Plata, 7600 Mar del Plata, Argentina
| |
Collapse
|
8
|
Lee JY, Lee HJ, Jang YH, Kim H, Im K, Yang S, Hoh JK, Ahn JH. Maternal pre-pregnancy obesity affects the uncinate fasciculus white matter tract in preterm infants. Front Pediatr 2023; 11:1225960. [PMID: 38034827 PMCID: PMC10684693 DOI: 10.3389/fped.2023.1225960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 10/23/2023] [Indexed: 12/02/2023] Open
Abstract
Background A growing body of evidence suggests an association between a higher maternal pre-pregnancy body mass index (BMI) and adverse long-term neurodevelopmental outcomes for their offspring. Despite recent attention to the effects of maternal obesity on fetal and neonatal brain development, changes in the brain microstructure of preterm infants born to mothers with pre-pregnancy obesity are still not well understood. This study aimed to detect the changes in the brain microstructure of obese mothers in pre-pregnancy and their offspring born as preterm infants using diffusion tensor imaging (DTI). Methods A total of 32 preterm infants (born to 16 mothers with normal BMI and 16 mothers with a high BMI) at <32 weeks of gestation without brain injury underwent brain magnetic resonance imaging at term-equivalent age (TEA). The BMI of all pregnant women was measured within approximately 12 weeks before pregnancy or the first 2 weeks of gestation. We analyzed the brain volume using a morphologically adaptive neonatal tissue segmentation toolbox and calculated the major white matter (WM) tracts using probabilistic maps of the Johns Hopkins University neonatal atlas. We investigated the differences in brain volume and WM microstructure between preterm infants of mothers with normal and high BMI. The DTI parameters were compared among groups using analysis of covariance adjusted for postmenstrual age at scan and multiple comparisons. Results Preterm infants born to mothers with a high BMI showed significantly increased cortical gray matter volume (p = 0.001) and decreased WM volume (p = 0.003) after controlling for postmenstrual age and multiple comparisons. We found a significantly lower axial diffusivity in the uncinate fasciculus (UNC) in mothers with high BMI than that in mothers with normal BMI (1.690 ± 0.066 vs. 1.762 ± 0.101, respectively; p = 0.005). Conclusion Our study is the first to demonstrate that maternal obesity impacts perinatal brain development patterns in preterm infants at TEA, even in the absence of apparent brain injury. These findings provide evidence for the detrimental effects of maternal obesity on brain developmental trajectories in offspring and suggest potential neurodevelopmental outcomes based on an altered UNC WM microstructure, which is known to be critical for language and social-emotional functions.
Collapse
Affiliation(s)
- Joo Young Lee
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science and Engineering, Seoul, Republic of Korea
| | - Hyun Ju Lee
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Republic of Korea
- Division of Neonatology and Development Medicine, Hanyang University Hospital, Seoul, Republic of Korea
| | - Yong Hun Jang
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science and Engineering, Seoul, Republic of Korea
| | - Hyuna Kim
- Department of Translational Medicine, Hanyang University Graduate School of Biomedical Science and Engineering, Seoul, Republic of Korea
| | - Kiho Im
- Fetal Neonatal Neuroimaging and Developmental Science Center, Boston Children's Hospital and Harvard Medical School, Boston, MA, United States
- Division of Newborn Medicine, Boston Children's Hospital and Harvard Medical School, Boston, MA, United States
| | - Seung Yang
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Republic of Korea
- Department of Pediatrics, Hanyang University Hospital, Seoul, Republic of Korea
| | - Jeong-Kyu Hoh
- Department of Obstetrics and Gynecology, Hanyang University College of Medicine, Seoul, Republic of Korea
- Department of Obstetrics and Gynecology, Hanyang University Hospital, Seoul, Republic of Korea
| | - Ja-Hye Ahn
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Republic of Korea
- Division of Neonatology and Development Medicine, Hanyang University Hospital, Seoul, Republic of Korea
| |
Collapse
|
9
|
He T, Chen Q, Yuan Z, Yang Y, Cao K, Luo J, Dong G, Peng X, Yang Z. Effects of maternal high-fat diet on fetal growth, placental nutrient transporters and circular RNA expression profiles. Food Funct 2023; 14:9391-9406. [PMID: 37791601 DOI: 10.1039/d3fo02202a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Epidemiological and experimental studies suggest that there is a strong correlation between maternal high-fat diet and fetal-placental development. The current study aims to investigate the effects of maternal high-fat diet on fetal growth, placental nutrient transporters and circular RNA expression profiles in a mouse model. Forty C57BL/6 female mice were randomly assigned to two groups, fed either a control (10% fat for energy) diet (CON) or a high-fat (60% fat for energy) diet (HFD) for 4 weeks before mating and throughout pregnancy, and were killed on day 19.5 of pregnancy. The serum glucose, total cholesterol and low-density lipoprotein, the glucolipid metabolism-related hormones, and the insulin resistance index were significantly increased. High-throughput sequencing showed that differentially expressed circRNAs (DE circRNAs) in the placenta can regulate various biological processes, cellular components, and molecular functions through various energy metabolism pathways, and mmu-let-7g-5p was found to target and bind to multiple DE circRNAs. In addition, this study also predicted that various circRNAs with protein coding functions can regulate maternal placental nutrient transport. In general, the ceRNA (circRNAs-miRNAs-mRNAs) regulatory network of maternal placental nutrient transport constructed in this study is of great significance for further understanding the effect of maternal nutrition on fetal growth in the future.
Collapse
Affiliation(s)
- Tianle He
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Qingyun Chen
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Zhidong Yuan
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou 341000, China
| | - Yulian Yang
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Kai Cao
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Ju Luo
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Guozhong Dong
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Xie Peng
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| | - Zhenguo Yang
- Laboratory for Bio-feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing 400715, China.
| |
Collapse
|
10
|
High-fat diet during pregnancy lowers fetal weight and has a long-lasting adverse effect on brown adipose tissue in the offspring. J Dev Orig Health Dis 2023; 14:261-271. [PMID: 36189641 DOI: 10.1017/s2040174422000551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Maternal obesity and malnutrition during gestation and lactation have been recognized to increase the risk of obesity and metabolic disorders in the offspring across their lifespan. However, the gestational period during which malnutrition exerts a decisive effect is unclear. Brown adipose tissue (BAT) plays a critical role in energy metabolism owing to its high efficiency in oxidizing glucose and fatty acids. This study aimed to determine the impact of maternal high-fat diet (HFD) consumption only during pregnancy on BAT and energy metabolism in offspring mice. Dams were fed an HFD or a normal chow diet from embryonic day 2.5. HFD consumption during pregnancy induced glucose intolerance and hypertension in dams. In the offspring of HFD-fed dams, maternal HFD lowered fetal weight without affecting placental weight, whereas HFD consumption after birth exacerbated oxygen consumption and cold-induced thermogenesis at 12 months of age, accompanied by increased lipid droplet size in BAT. These data demonstrate that HFD consumption only during pregnancy exerts a long-lasting effect on BAT. Collectively, these findings indicate the importance of nutrition during pregnancy with respect to the energy metabolism of the offspring, and pregnant women should thus ensure proper nutrition during pregnancy to ensure normal energy metabolism in the offspring.
Collapse
|
11
|
Kang K, Zeng L, Ma J, Shi L, Hu R, Zou H, Peng Q, Wang L, Xue B, Wang Z. High energy diet of beef cows during gestation promoted growth performance of calves by improving placental nutrients transport. Front Vet Sci 2022; 9:1053730. [PMID: 36504847 PMCID: PMC9730878 DOI: 10.3389/fvets.2022.1053730] [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: 09/26/2022] [Accepted: 11/08/2022] [Indexed: 11/25/2022] Open
Abstract
The aim of this study was to explore the effects of dietary energy level during gestation on growth performance and serum parameters in offspring using beef cattle as research objects. Additionally, the gene expressions associated with nutrients transport in the placenta were evaluated. Eighteen Simmental crossbred cows (body weight = 338.44 ± 16.03 kg and 760 ± 6 days of age) were randomly assigned to 3 dietary treatment groups: low energy (LE, metabolic energy = 8.76 MJ/kg), medium (ME, 9.47 MJ/kg) and high (HE, 10.18 MJ/kg). The dietary treatments were introduced from day 45 before expected date of parturition. The pre-experiment lasted for 15 days and formal experiment lasted for 30 days. Growth performance data and blood samples of calves were collected at birth and day 30 post-birth. The placental tissue was collected at parturition. The results indicated that the birth weight and average daily gain of calves in HE group were higher (P < 0.05) than those in LE group. After parturition, the serum contents of glucose, total protein, cortisol and leptin in neonatal calves were significantly increased (P < 0.05) with the elevation of dietary energy levels. At 30 days postpartum, the glucose, glutathione peroxidase, growth hormone, insulin-like growth factor 1 and leptin concentrations of HE group were significantly increased (P < 0.05) as compared with LE group, while the serum amyloid protein A displayed an opposite trend between two groups. With the increase of dietary energy concentration, placental mRNA expressions of vascular endothelial growth factor A, glucose transporter 1 and 3 were significantly up-regulated (P < 0.05). Furthermore, the amino acid transporter solute carrier family 38 member 1, hydroxysteroid 11-beta dehydrogenase 2, insulin-like growth factor 1 and 2 mRNA expressions of HE group were higher (P < 0.05) than those of LE and ME groups. In conclusion, the improved growth performance of calves from the high energy ration supplemented beef cows may be attributed to the increased placental nutrients transport, which may lead to the increased nutrient supply to the fetus.
Collapse
|
12
|
Zhao D, Liu Y, Jia S, He Y, Wei X, Liu D, Ma W, Luo W, Gu H, Yuan Z. Influence of maternal obesity on the multi-omics profiles of the maternal body, gestational tissue, and offspring. Biomed Pharmacother 2022; 151:113103. [PMID: 35605294 DOI: 10.1016/j.biopha.2022.113103] [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: 03/28/2022] [Revised: 04/25/2022] [Accepted: 05/10/2022] [Indexed: 11/28/2022] Open
Abstract
Epidemiological studies show that obesity during pregnancy affects more than half of the pregnancies in the developed countries and is associated with obstetric problems and poor outcomes. Obesity tends to increase the incidence of complications. Furthermore, the resulting offspring are also adversely affected. However, the molecular mechanisms of obesity leading to poor pregnancy outcomes remain unclear. Omics methods are used for genetic diagnosis and marker discovery. The aim of this review was to summarize the maternal and fetal pathophysiological alterations induced by gestational obesity,identified using multi-omics detection techniques, and to generalize the biological functions and potential mechanisms of the differentially expressed molecules.
Collapse
Affiliation(s)
- Duan Zhao
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, China.
| | - Yusi Liu
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, China.
| | - Shanshan Jia
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, China.
| | - Yiwen He
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, China.
| | - Xiaowei Wei
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, China.
| | - Dan Liu
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, China.
| | - Wei Ma
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, China.
| | - Wenting Luo
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, China.
| | - Hui Gu
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, China.
| | - Zhengwei Yuan
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital, China Medical University, Shenyang 110004, China.
| |
Collapse
|
13
|
Hatmal MM, Al-Hatamleh MAI, Olaimat AN, Alshaer W, Hasan H, Albakri KA, Alkhafaji E, Issa NN, Al-Holy MA, Abderrahman SM, Abdallah AM, Mohamud R. Immunomodulatory Properties of Human Breast Milk: MicroRNA Contents and Potential Epigenetic Effects. Biomedicines 2022; 10:1219. [PMID: 35740242 PMCID: PMC9219990 DOI: 10.3390/biomedicines10061219] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 05/15/2022] [Accepted: 05/17/2022] [Indexed: 02/07/2023] Open
Abstract
Infants who are exclusively breastfed in the first six months of age receive adequate nutrients, achieving optimal immune protection and growth. In addition to the known nutritional components of human breast milk (HBM), i.e., water, carbohydrates, fats and proteins, it is also a rich source of microRNAs, which impact epigenetic mechanisms. This comprehensive work presents an up-to-date overview of the immunomodulatory constituents of HBM, highlighting its content of circulating microRNAs. The epigenetic effects of HBM are discussed, especially those regulated by miRNAs. HBM contains more than 1400 microRNAs. The majority of these microRNAs originate from the lactating gland and are based on the remodeling of cells in the gland during breastfeeding. These miRNAs can affect epigenetic patterns by several mechanisms, including DNA methylation, histone modifications and RNA regulation, which could ultimately result in alterations in gene expressions. Therefore, the unique microRNA profile of HBM, including exosomal microRNAs, is implicated in the regulation of the genes responsible for a variety of immunological and physiological functions, such as FTO, INS, IGF1, NRF2, GLUT1 and FOXP3 genes. Hence, studying the HBM miRNA composition is important for improving the nutritional approaches for pregnancy and infant's early life and preventing diseases that could occur in the future. Interestingly, the composition of miRNAs in HBM is affected by multiple factors, including diet, environmental and genetic factors.
Collapse
Affiliation(s)
- Ma’mon M. Hatmal
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Mohammad A. I. Al-Hatamleh
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu 16150, Malaysia;
| | - Amin N. Olaimat
- Department of Clinical Nutrition and Dietetics, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan; (A.N.O.); (M.A.A.-H.)
| | - Walhan Alshaer
- Cell Therapy Center (CTC), The University of Jordan, Amman 11942, Jordan;
| | - Hanan Hasan
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan;
| | - Khaled A. Albakri
- Faculty of Medicine, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Enas Alkhafaji
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, The University of Jordan, Amman 11942, Jordan;
| | - Nada N. Issa
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Murad A. Al-Holy
- Department of Clinical Nutrition and Dietetics, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan; (A.N.O.); (M.A.A.-H.)
| | - Salim M. Abderrahman
- Department of Biology and Biotechnology, Faculty of Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Atiyeh M. Abdallah
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar;
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu 16150, Malaysia;
| |
Collapse
|
14
|
Doan TNA, Akison LK, Bianco-Miotto T. Epigenetic Mechanisms Responsible for the Transgenerational Inheritance of Intrauterine Growth Restriction Phenotypes. Front Endocrinol (Lausanne) 2022; 13:838737. [PMID: 35432208 PMCID: PMC9008301 DOI: 10.3389/fendo.2022.838737] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 03/02/2022] [Indexed: 12/20/2022] Open
Abstract
A poorly functioning placenta results in impaired exchanges of oxygen, nutrition, wastes and hormones between the mother and her fetus. This can lead to restriction of fetal growth. These growth restricted babies are at increased risk of developing chronic diseases, such as type-2 diabetes, hypertension, and kidney disease, later in life. Animal studies have shown that growth restricted phenotypes are sex-dependent and can be transmitted to subsequent generations through both the paternal and maternal lineages. Altered epigenetic mechanisms, specifically changes in DNA methylation, histone modifications, and non-coding RNAs that regulate expression of genes that are important for fetal development have been shown to be associated with the transmission pattern of growth restricted phenotypes. This review will discuss the subsequent health outcomes in the offspring after growth restriction and the transmission patterns of these diseases. Evidence of altered epigenetic mechanisms in association with fetal growth restriction will also be reviewed.
Collapse
Affiliation(s)
- Thu Ngoc Anh Doan
- School of Agriculture, Food and Wine, Waite Research Institute, University of Adelaide, Adelaide, SA, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
| | - Lisa K. Akison
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
| | - Tina Bianco-Miotto
- School of Agriculture, Food and Wine, Waite Research Institute, University of Adelaide, Adelaide, SA, Australia
- Robinson Research Institute, University of Adelaide, Adelaide, SA, Australia
- *Correspondence: Tina Bianco-Miotto,
| |
Collapse
|
15
|
Kuiper-Makris C, Selle J, Nüsken E, Dötsch J, Alejandre Alcazar MA. Perinatal Nutritional and Metabolic Pathways: Early Origins of Chronic Lung Diseases. Front Med (Lausanne) 2021; 8:667315. [PMID: 34211985 PMCID: PMC8239134 DOI: 10.3389/fmed.2021.667315] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 05/12/2021] [Indexed: 12/12/2022] Open
Abstract
Lung development is not completed at birth, but expands beyond infancy, rendering the lung highly susceptible to injury. Exposure to various influences during a critical window of organ growth can interfere with the finely-tuned process of development and induce pathological processes with aberrant alveolarization and long-term structural and functional sequelae. This concept of developmental origins of chronic disease has been coined as perinatal programming. Some adverse perinatal factors, including prematurity along with respiratory support, are well-recognized to induce bronchopulmonary dysplasia (BPD), a neonatal chronic lung disease that is characterized by arrest of alveolar and microvascular formation as well as lung matrix remodeling. While the pathogenesis of various experimental models focus on oxygen toxicity, mechanical ventilation and inflammation, the role of nutrition before and after birth remain poorly investigated. There is accumulating clinical and experimental evidence that intrauterine growth restriction (IUGR) as a consequence of limited nutritive supply due to placental insufficiency or maternal malnutrition is a major risk factor for BPD and impaired lung function later in life. In contrast, a surplus of nutrition with perinatal maternal obesity, accelerated postnatal weight gain and early childhood obesity is associated with wheezing and adverse clinical course of chronic lung diseases, such as asthma. While the link between perinatal nutrition and lung health has been described, the underlying mechanisms remain poorly understood. There are initial data showing that inflammatory and nutrient sensing processes are involved in programming of alveolarization, pulmonary angiogenesis, and composition of extracellular matrix. Here, we provide a comprehensive overview of the current knowledge regarding the impact of perinatal metabolism and nutrition on the lung and beyond the cardiopulmonary system as well as possible mechanisms determining the individual susceptibility to CLD early in life. We aim to emphasize the importance of unraveling the mechanisms of perinatal metabolic programming to develop novel preventive and therapeutic avenues.
Collapse
Affiliation(s)
- Celien Kuiper-Makris
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics—Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jaco Selle
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics—Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Eva Nüsken
- Department of Pediatric and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Jörg Dötsch
- Department of Pediatric and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Miguel A. Alejandre Alcazar
- Department of Pediatric and Adolescent Medicine, Translational Experimental Pediatrics—Experimental Pulmonology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Excellence Cluster on Stress Responses in Aging-associated Diseases (CECAD), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Member of the German Centre for Lung Research (DZL), Institute for Lung Health, University of Giessen and Marburg Lung Centre (UGMLC), Gießen, Germany
| |
Collapse
|
16
|
Hicks JA, Liu HC. Centennial Review: Metabolic microRNA - shifting gears in the regulation of metabolic pathways in poultry. Poult Sci 2021; 100:100856. [PMID: 33652542 PMCID: PMC7936154 DOI: 10.1016/j.psj.2020.11.033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 11/16/2020] [Accepted: 11/22/2020] [Indexed: 01/01/2023] Open
Abstract
Over 20 yr ago, a small noncoding class of RNA termed microRNA (miRNA) that was able to recognize sequences in mRNAs and inhibit their translation was discovered in Caenorhabditis elegans. In the intervening years, miRNA have been discovered in most eukaryotes and are now known to regulate the majority of protein-coding genes. It has been discovered that disruption of miRNA function often leads to the development of pathological conditions. One physiological system under extensive miRNA-mediated regulation is metabolism. Metabolism is one of the most dynamic of biological networks within multiple organs, including the liver, muscle, and adipose tissue, working in concert to respond to ever-changing nutritional cues and energy demands. Therefore, it is not surprising that miRNA regulate virtually all aspects of eukaryotic metabolism and have been linked to metabolic disorders, such as obesity, fatty liver diseases, and diabetes, just to name a few. Chickens, and birds in general, face their own unique metabolic challenges, particularly after hatching, when their metabolism must completely transform from using lipid-rich yolk to carbohydrate-rich feed as fuel in a very short period of time. Furthermore, commercial poultry breeds have undergone extensive selection over the last century for more desirable production traits, which has resulted in numerous metabolic consequences. Here, we review the current knowledge of miRNA-mediated regulation of metabolic development and function in chickens.
Collapse
Affiliation(s)
- Julie A Hicks
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA
| | - Hsiao-Ching Liu
- Department of Animal Science, North Carolina State University, Raleigh, NC 27695, USA.
| |
Collapse
|
17
|
Ellur G, Sukhdeo SV, Khan MT, Sharan K. Maternal high protein-diet programs impairment of offspring's bone mass through miR-24-1-5p mediated targeting of SMAD5 in osteoblasts. Cell Mol Life Sci 2021; 78:1729-1744. [PMID: 32734584 PMCID: PMC11071892 DOI: 10.1007/s00018-020-03608-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 12/25/2022]
Abstract
Maternal nutrition is crucial for the offspring's skeleton development and the onset of osteoporosis later in life. While maternal low protein diet has been shown to regulate bone mass negatively, the effect of a high protein diet (HP) remains unexplored. Here, we found that C57BL/6 mice fed with HP delivered offspring with decreased skeletal mineralization at birth and reduced bone mass throughout their life due to a decline in their osteoblast maturation. A small RNA sequencing study revealed that miR-24-1-5p was highly upregulated in HP group osteoblasts. Target prediction and validation studies identified SMAD-5 as a direct target of miR-24-1-5p. Furthermore, mimic and inhibitor studies showed a negative correlation between miR-24-1-5p expression and osteoblast function. Moreover, ex vivo inhibition of miR-24-1-5p reversed the reduced maturation and SMAD-5 expression in the HP group osteoblasts. Together, we show that maternal HP diminishes the bone mass of the offspring through miR-24-1-5p.
Collapse
Affiliation(s)
- Govindraj Ellur
- Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysore, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shinde Vijay Sukhdeo
- Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysore, India
| | - Md Touseef Khan
- Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysore, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Kunal Sharan
- Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysore, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
18
|
Savva C, Helguero LA, González-Granillo M, Couto D, Melo T, Li X, Angelin B, Domingues MR, Kutter C, Korach-André M. Obese mother offspring have hepatic lipidic modulation that contributes to sex-dependent metabolic adaptation later in life. Commun Biol 2021; 4:14. [PMID: 33398027 PMCID: PMC7782679 DOI: 10.1038/s42003-020-01513-z] [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: 06/24/2020] [Accepted: 11/24/2020] [Indexed: 02/05/2023] Open
Abstract
With the increasing prevalence of obesity in women of reproductive age, there is an urgent need to understand the metabolic impact on the fetus. Sex-related susceptibility to liver diseases has been demonstrated but the underlying mechanism remains unclear. Here we report that maternal obesity impacts lipid metabolism differently in female and male offspring. Males, but not females, gained more weight and had impaired insulin sensitivity when born from obese mothers compared to control. Although lipid mass was similar in the livers of female and male offspring, sex-specific modifications in the composition of fatty acids, triglycerides and phospholipids was observed. These overall changes could be linked to sex-specific regulation of genes controlling metabolic pathways. Our findings revised the current assumption that sex-dependent susceptibility to metabolic disorders is caused by sex-specific postnatal regulation and instead we provide molecular evidence supporting in utero metabolic adaptations in the offspring of obese mothers.
Collapse
Affiliation(s)
- Christina Savva
- Department of Medicine, Cardio Metabolic Unit (CMU) and KI/AZ Integrated Cardio Metabolic Center (ICMC), Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
- Clinical Department of Endocrinology, Metabolism and Diabetes, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Luisa A Helguero
- Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Marcela González-Granillo
- Department of Medicine, Cardio Metabolic Unit (CMU) and KI/AZ Integrated Cardio Metabolic Center (ICMC), Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
- Clinical Department of Endocrinology, Metabolism and Diabetes, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Daniela Couto
- CESAM, Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Santiago University Campus, Aveiro, Portugal
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Santiago University Campus, Aveiro, Portugal
| | - Tânia Melo
- CESAM, Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Santiago University Campus, Aveiro, Portugal
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Santiago University Campus, Aveiro, Portugal
| | - Xidan Li
- Department of Medicine, Cardio Metabolic Unit (CMU) and KI/AZ Integrated Cardio Metabolic Center (ICMC), Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Bo Angelin
- Department of Medicine, Cardio Metabolic Unit (CMU) and KI/AZ Integrated Cardio Metabolic Center (ICMC), Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
- Clinical Department of Endocrinology, Metabolism and Diabetes, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Maria Rosário Domingues
- CESAM, Centre for Environmental and Marine Studies, Department of Chemistry, University of Aveiro, Santiago University Campus, Aveiro, Portugal
- Mass Spectrometry Centre, LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Santiago University Campus, Aveiro, Portugal
| | - Claudia Kutter
- Department of Microbiology, Tumor and Cell Biology, Science for Life Laboratory, Karolinska Institute, Stockholm, Sweden
| | - Marion Korach-André
- Department of Medicine, Cardio Metabolic Unit (CMU) and KI/AZ Integrated Cardio Metabolic Center (ICMC), Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden.
- Clinical Department of Endocrinology, Metabolism and Diabetes, Karolinska University Hospital Huddinge, Stockholm, Sweden.
| |
Collapse
|
19
|
Guo Q, Lu Y, Huang Y, Guo Y, Zhu S, Zhang Q, Zhu D, Wang Z, Luo J. Exosomes from β-Cells Promote Differentiation of Induced Pluripotent Stem Cells into Insulin-Producing Cells Through microRNA-Dependent Mechanisms. Diabetes Metab Syndr Obes 2021; 14:4767-4782. [PMID: 34934332 PMCID: PMC8678630 DOI: 10.2147/dmso.s342647] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 12/01/2021] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Exosomes have emerged as potential tools for the differentiation of induced pluripotent stem cells (iPSCs) into insulin-producing cells (IPCs). Exosomal microRNAs are receiving increasing attention in this process. Here, we aimed at investigating the role of exosomes derived from a murine pancreatic β-cell line and identifying signature exosomal miRNAs on iPSCs differentiation. METHODS Exosomes were isolated from MIN6 cells and identified with TEM, NTA and Western blot. PKH67 tracer and transwell assay were used to confirm exosome delivery into iPSCs. qRT-PCR was applied to detect key pancreatic transcription gene expression and exosome-derived miRNA expression. Insulin secretion was determined using FCM and immunofluorescence. The specific exosomal miRNAs were determined via RNA-interference of Ago2. The therapeutic effect of 21 day-exosome-induced IPCs was validated in T1D mice induced by STZ. RESULTS iPSCs cultured in medium containing exosomes showed sustained higher expression of MAFA, Insulin1, Insulin2, Isl1, Neuroud1, Nkx6.1 and NGN3 compared to control iPSCs. In FCM analysis, approximately 52.7% of the differentiated cells displayed insulin expression at the middle stage. Consistent with the gene expression data, immunofluorescence assays showed that Nkx6.1 and insulin expression in iPSCs were significantly upregulated. Intriguingly, the expression of pancreatic markers and insulin was significantly decreased in iPSCs cultured with siAgo2 exosomes. Transplantation of 21 day-induced IPCs intoT1D mice efficiently enhanced glucose tolerance and partially controlled hyperglycemia. The therapeutic effect was significantly attenuated in T1D mice that received iPSCs cultured with siAgo2 exosomes. Of the seven exosomal microRNAs selected for validation, miR-706, miR-709, miR-466c-5p, and miR-423-5p showed dynamic expression during 21 days in culture. CONCLUSION These data indicate that differentiation of exosome-induced iPSCs into functional cells is crucially dependent on the specific miRNAs encased within exosomes, whose functional analysis is likely to provide insight into novel regulatory mechanisms governing iPSCs differentiation into IPCs.
Collapse
Affiliation(s)
- Qingsong Guo
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, People’s Republic of China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, People’s Republic of China
| | - Yuhua Lu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, People’s Republic of China
| | - Yan Huang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, People’s Republic of China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, People’s Republic of China
| | - Yibing Guo
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, People’s Republic of China
| | - Shajun Zhu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, People’s Republic of China
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, People’s Republic of China
| | - Qiuqiang Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, People’s Republic of China
| | - Donghui Zhu
- Nantong University Medical School, Nantong, 226001, People’s Republic of China
| | - Zhiwei Wang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, People’s Republic of China
- Zhiwei Wang Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Xi Si Road, Nantong, 226001, People’s Republic of China Email
| | - Jia Luo
- Department of Pharmacy, Affiliated Hospital of Nantong University, Nantong, 226001, People’s Republic of China
- Correspondence: Jia Luo Department of Pharmacy, Affiliated Hospital of Nantong University, Xi Si Road, Nantong, 226001, People’s Republic of China Email
| |
Collapse
|
20
|
Christoforou ER, Sferruzzi-Perri AN. Molecular mechanisms governing offspring metabolic programming in rodent models of in utero stress. Cell Mol Life Sci 2020; 77:4861-4898. [PMID: 32494846 PMCID: PMC7658077 DOI: 10.1007/s00018-020-03566-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/23/2020] [Accepted: 05/27/2020] [Indexed: 12/13/2022]
Abstract
The results of different human epidemiological datasets provided the impetus to introduce the now commonly accepted theory coined as 'developmental programming', whereby the presence of a stressor during gestation predisposes the growing fetus to develop diseases, such as metabolic dysfunction in later postnatal life. However, in a clinical setting, human lifespan and inaccessibility to tissue for analysis are major limitations to study the molecular mechanisms governing developmental programming. Subsequently, studies using animal models have proved indispensable to the identification of key molecular pathways and epigenetic mechanisms that are dysregulated in metabolic organs of the fetus and adult programmed due to an adverse gestational environment. Rodents such as mice and rats are the most used experimental animals in the study of developmental programming. This review summarises the molecular pathways and epigenetic mechanisms influencing alterations in metabolic tissues of rodent offspring exposed to in utero stress and subsequently programmed for metabolic dysfunction. By comparing molecular mechanisms in a variety of rodent models of in utero stress, we hope to summarise common themes and pathways governing later metabolic dysfunction in the offspring whilst identifying reasons for incongruencies between models so to inform future work. With the continued use and refinement of such models of developmental programming, the scientific community may gain the knowledge required for the targeted treatment of metabolic diseases that have intrauterine origins.
Collapse
Affiliation(s)
- Efthimia R Christoforou
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Downing Site, Cambridge, UK
| | - Amanda N Sferruzzi-Perri
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Downing Site, Cambridge, UK.
| |
Collapse
|
21
|
Carrillo-Lozano E, Sebastián-Valles F, Knott-Torcal C. Circulating microRNAs in Breast Milk and Their Potential Impact on the Infant. Nutrients 2020; 12:E3066. [PMID: 33049923 PMCID: PMC7601398 DOI: 10.3390/nu12103066] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/27/2020] [Accepted: 10/01/2020] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (MiRNAs) are small RNA molecules that can exert regulatory functions in gene expression. MiRNAs have been identified in diverse tissues and biological fluids, both in the context of health and disease. Breastfeeding has been widely recognized for its superior nutritional benefits; however, a number of bioactive compounds have been found to transcend these well-documented nutritional contributions. Breast milk was identified as a rich source of miRNAs. There has been increasing interest about their potential ability to transfer to the offspring as well as what their specific involvement is within the benefits of breast milk in the infant. In comparison to breast milk, formula milk lacks many of the benefits of breastfeeding, which is thought to be a result of the absence of some of these bioactive compounds. In recent years, the miRNA profile of breast milk has been widely studied, along with the possible transfer mechanisms throughout the infant's digestive tract and the role of miRNA-modulated genes and their potential protective and regulatory functions. Nonetheless, to date, the current evidence is not consistent, as many methodological limitations have been identified; hence, discrepancies exits about the biological functions of miRNAs. Further research is needed to provide thorough knowledge in this field.
Collapse
|
22
|
Thompson MD. Developmental Programming of NAFLD by Parental Obesity. Hepatol Commun 2020; 4:1392-1403. [PMID: 33024911 PMCID: PMC7527686 DOI: 10.1002/hep4.1578] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/08/2020] [Accepted: 07/08/2020] [Indexed: 12/11/2022] Open
Abstract
The surge of obesity across generations has become an increasingly relevant issue, with consequences for associated comorbidities in offspring. Data from longitudinal birth cohort studies support an association between maternal obesity and offspring nonalcoholic fatty liver disease (NAFLD), suggesting that perinatal obesity or obesogenic diet exposure reprograms offspring liver and increases NAFLD susceptibility. In preclinical models, offspring exposed to maternal obesogenic diet have increased hepatic steatosis after diet-induced obesity; however, the implications for later NAFLD development and progression are still unclear. Although some models show increased NAFLD incidence and progression in offspring, development of nonalcoholic steatohepatitis with fibrosis may be model dependent. Multigenerational programming of NAFLD phenotypes occurs after maternal obesogenic diet exposure; however, the mechanisms for such programming remain poorly understood. Likewise, emerging data on the role of paternal obesity in offspring NAFLD development reveal incomplete mechanisms. This review will explore the impact of parental obesity and obesogenic diet exposure on offspring NAFLD and areas for further investigation, including the impact of parental diet on disease progression, and consider potential interventions in preclinical models.
Collapse
Affiliation(s)
- Michael D. Thompson
- Division of Endocrinology and DiabetesDepartment of PediatricsWashington University School of MedicineSt. LouisMO
| |
Collapse
|
23
|
Sohel MMH. Macronutrient modulation of mRNA and microRNA function in animals: A review. ACTA ACUST UNITED AC 2020; 6:258-268. [PMID: 33005759 PMCID: PMC7503081 DOI: 10.1016/j.aninu.2020.06.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 06/01/2020] [Accepted: 06/15/2020] [Indexed: 12/15/2022]
Abstract
Dietary macronutrients have been regarded as a basic source of energy and amino acids that are necessary for the maintenance of cellular homeostasis, metabolic programming as well as protein synthesis. Due to the emergence of “nutrigenomics”, a unique discipline that combines nutritional and omics technologies to study the impacts of nutrition on genomics, it is increasingly evident that macronutrients also have a significant role in the gene expression regulation. Gene expression is a complex phenomenon controlled by several signaling pathways and could be influenced by a wide variety of environmental and physiological factors. Dietary macronutrients are the most important environmental factor influencing the expression of both genes and microRNAs (miRNA). miRNA are tiny molecules of 18 to 22 nucleotides long that regulate the expression of genes. Therefore, dietary macronutrients can influence the expression of genes in both direct and indirect manners. Recent advancements in the state-of-the-art technologies regarding molecular genetics, such as next-generation sequencing, quantitative PCR array, and microarray, allowed us to investigate the occurrence of genome-wide changes in the expression of genes in relation to augmented or reduced dietary macronutrient intake. The purpose of this review is to accumulate the current knowledge focusing on macronutrient mediated changes in the gene function. This review will discuss the impact of altered dietary carbohydrate, protein, and fat intake on the expression of coding genes and their functions. In addition, it will also summarize the regulation of miRNA, both cellular and extracellular miRNA, expression modulated by dietary macronutrients.
Collapse
Affiliation(s)
- Md Mahmodul Hasan Sohel
- Department of Genetics, Faculty of Veterinary Medicine, Erciyes University, Kayseri, 38039, Turkey.,Genome and Stem Cell Centre, Erciyes University, Kayseri, 38039, Turkey
| |
Collapse
|
24
|
Zheng J, Zhang L, Wang Z, Zhang J. Maternal high-fat diet regulates glucose metabolism and pancreatic β cell phenotype in mouse offspring at weaning. PeerJ 2020; 8:e9407. [PMID: 32607287 PMCID: PMC7316079 DOI: 10.7717/peerj.9407] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 06/02/2020] [Indexed: 11/20/2022] Open
Abstract
Background Maternal malnutrition is a critical factor in determining the risk of obesity and glucose intolerance in offspring. However, little is known about the effects of a maternal high-fat diet (HFD) on the β cell phenotype in offspring, which is a major factor in glucose homeostasis, especially during the early life of offspring. Methods Dams were randomly fed a HFD (60% kcal from fat) or a chow diet before pregnancy and during gestation and lactation. Glucose metabolism and the β cell phenotype were assessed in male offspring at weaning. Results Dams fed a HFD showed impaired glucose tolerance. A HFD predisposed the offspring to increased impairment of metabolic health, including obesity, glucose intolerance and insulin resistance, compared with offspring from chow diet-fed dams. Furthermore, increased islet sizes and islet densities were observed in male offspring from HFD-fed dams at weaning. There were increases in the insulin-positive area, β cell mass and β cell proliferation in male offspring from HFD-fed dams at weaning age. Next, we further determined whether a maternal HFD could affect β cell apoptosis in mouse offspring and found that there was no significant change in β cell apoptosis between the HFD and control groups. Conclusion Our study is novel in showing that a maternal HFD predisposes offspring to impaired glucose metabolism and has a profound effect on β cell mass and proliferation in offspring mice, which is observed in mice as early as at weaning age. However, further study to clarify the underlying mechanisms is warranted.
Collapse
Affiliation(s)
- Jia Zheng
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Ling Zhang
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Ziwei Wang
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Junqing Zhang
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| |
Collapse
|
25
|
Deodati A, Inzaghi E, Cianfarani S. Epigenetics and In Utero Acquired Predisposition to Metabolic Disease. Front Genet 2020; 10:1270. [PMID: 32082357 PMCID: PMC7000755 DOI: 10.3389/fgene.2019.01270] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 11/18/2019] [Indexed: 01/21/2023] Open
Abstract
Epidemiological evidence has shown an association between prenatal malnutrition and a higher risk of developing metabolic disease in adult life. An inadequate intrauterine milieu affects both growth and development, leading to a permanent programming of endocrine and metabolic functions. Programming may be due to the epigenetic modification of genes implicated in the regulation of key metabolic mechanisms, including DNA methylation, histone modifications, and microRNAs (miRNAs). The expression of miRNAs in organs that play a key role in metabolism is influenced by in utero programming, as demonstrated by both experimental and human studies. miRNAs modulate multiple pathways such as insulin signaling, immune responses, adipokine function, lipid metabolism, and food intake. Liver is one of the main target organs of programming, undergoing structural, functional, and epigenetic changes following the exposure to a suboptimal intrauterine environment. The focus of this review is to provide an overview of the effects of exposure to an adverse in utero milieu on epigenome with a focus on the molecular mechanisms involved in liver programming.
Collapse
Affiliation(s)
- Annalisa Deodati
- Dipartimento Pediatrico Universitario Ospedaliero "Bambino Gesù" Children's Hospital, Tor Vergata University, Rome, Italy
| | - Elena Inzaghi
- Dipartimento Pediatrico Universitario Ospedaliero "Bambino Gesù" Children's Hospital, Tor Vergata University, Rome, Italy
| | - Stefano Cianfarani
- Dipartimento Pediatrico Universitario Ospedaliero "Bambino Gesù" Children's Hospital, Tor Vergata University, Rome, Italy.,Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
26
|
Moradi Sarabi M, Mohammadrezaei Khorramabadi R, Zare Z, Eftekhar E. Polyunsaturated fatty acids and DNA methylation in colorectal cancer. World J Clin Cases 2019; 7:4172-4185. [PMID: 31911898 PMCID: PMC6940323 DOI: 10.12998/wjcc.v7.i24.4172] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/27/2019] [Accepted: 12/13/2019] [Indexed: 02/05/2023] Open
Abstract
Colorectal cancer (CRC) has been designated a major global problem, especially due to its high prevalence in developed countries. CRC mostly occurs sporadically (75%-80%), and only 20%-25% of patients have a family history. Several processes are involved in the development of CRC such as a combination of genetic and epigenetic alterations. Epigenetic changes, including DNA methylation play a vital role in the progression of CRC. Complex interactions between susceptibility genes and environmental factors, such as a diet and sedentary lifestyle, lead to the development of CRC. Clinical and experimental studies have confirmed the beneficial effects of dietary polyunsaturated fatty acids (PUFAs) in preventing CRC. From a mechanistic viewpoint, it has been suggested that PUFAs are pleiotropic agents that alter chromatin remodeling, membrane structure and downstream cell signaling. Moreover, PUFAs can alter the epigenome via modulation of DNA methylation. In this review, we summarize recent investigations linking PUFAs and DNA methylation-associated CRC risk.
Collapse
Affiliation(s)
- Mostafa Moradi Sarabi
- Department of Biochemistry and Genetics, School of Medicine, Lorestan University of Medical Sciences, Khorramabad 381251698, Iran
| | - Reza Mohammadrezaei Khorramabadi
- Department of Biochemistry and Genetics, School of Medicine, Lorestan University of Medical Sciences, Khorramabad 381251698, Iran
| | - Zohre Zare
- Department of Pharmaceutics, School of Pharmacy, Lorestan University of Medical Sciences, Khorramabad 381251698, Iran
| | - Ebrahim Eftekhar
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas 7919915519, Iran
| |
Collapse
|
27
|
Van Hese I, Goossens K, Vandaele L, Opsomer G. Invited review: MicroRNAs in bovine colostrum-Focus on their origin and potential health benefits for the calf. J Dairy Sci 2019; 103:1-15. [PMID: 31677833 DOI: 10.3168/jds.2019-16959] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 09/06/2019] [Indexed: 12/23/2022]
Abstract
Colostrum is the first milk produced by a cow after she gives birth. Compared with mature milk, it has a high concentration of immunoglobulin G. Calves are born without circulating antibodies, thus ingestion of colostrum is necessary to protect the calf against pathogens in the first challenging weeks of life. In addition to the life-saving supply of antibodies, colostrum contains minerals, vitamins, growth factors, and immune cells. Recently, microRNAs (miRNAs) were added to that list. MicroRNAs are short, non-coding RNA molecules that can regulate gene expression at the post-transcriptional level. They are thought to act as key regulators of diverse biological and developmental processes. Colostrum contains higher amounts of miRNAs than mature milk; immune- and development-related miRNAs are prominent. Their expression pattern in milk is likely to be influenced by maternal nutrition and environment. The fat content of the maternal diet appears to have a major effect on expression of miRNAs in milk and in the neonate. The immunological state of the mammary gland seems to affect miRNA expression as well. In cows diagnosed with subclinical mastitis, alterations in the expression of miRNAs in milk have been observed. It is believed that miRNAs in colostrum and milk are signaling molecules passed from mother to newborn. They are packaged in extracellular vesicles, which makes them resistant to the harsh conditions in the gastrointestinal tract. Therefore, they can reach the small intestine, where they are absorbed and transferred into the bloodstream. MicroRNAs are important for the development of the intestines. For example, miRNAs stimulate cell viability, proliferation, and stem cell activity of the intestinal epithelium. Furthermore, miRNAs seem to act as key players in the development of the complete immune system. They can, among other things, regulate B- and T-cell differentiation and affect interleukin production of macrophages. The abundance of miRNAs in colostrum and milk and the possibility for their absorption in the intestines of the neonate supports the hypothesis that these tiny molecules are important for the development of the newborn. The probable relation of diet to the expression of miRNAs by the mother creates a possible avenue to optimize expression of miRNAs and improve neonatal maturation.
Collapse
Affiliation(s)
- I Van Hese
- Animal Sciences Unit, Flanders Research Institute for Agriculture, Fisheries and Food (ILVO), Scheldeweg, Melle, 9090, Belgium; Department of Reproduction, Obstetrics and Herd Health Faculty of Veterinary Medicine, Ghent University, Salisburylaan, Merelbeke, 9820, Belgium.
| | - K Goossens
- Animal Sciences Unit, Flanders Research Institute for Agriculture, Fisheries and Food (ILVO), Scheldeweg, Melle, 9090, Belgium
| | - L Vandaele
- Animal Sciences Unit, Flanders Research Institute for Agriculture, Fisheries and Food (ILVO), Scheldeweg, Melle, 9090, Belgium
| | - G Opsomer
- Department of Reproduction, Obstetrics and Herd Health Faculty of Veterinary Medicine, Ghent University, Salisburylaan, Merelbeke, 9820, Belgium
| |
Collapse
|
28
|
Cleal JK, Bruce KD, Shearer JL, Thomas H, Plume J, Gregory L, Shepard JN, Spiers-Fitzgerald KL, Mani R, Lewis RM, Lillycrop KA, Hanson MA, Byrne CD, Cagampang FR. Maternal Obesity during Pregnancy Alters Daily Activity and Feeding Cycles, and Hypothalamic Clock Gene Expression in Adult Male Mouse Offspring. Int J Mol Sci 2019; 20:E5408. [PMID: 31671625 PMCID: PMC6862679 DOI: 10.3390/ijms20215408] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 10/12/2019] [Accepted: 10/28/2019] [Indexed: 02/07/2023] Open
Abstract
An obesogenic diet adversely affects the endogenous mammalian circadian clock, altering daily activity and metabolism, and resulting in obesity. We investigated whether an obese pregnancy can alter the molecular clock in the offspring hypothalamus, resulting in changes to their activity and feeding rhythms. Female mice were fed a control (C, 7% kcal fat) or high fat diet (HF, 45% kcal fat) before mating and throughout pregnancy. Male offspring were fed the C or HF diet postweaning, resulting in four offspring groups: C/C, C/HF, HF/C, and HF/HF. Daily activity and food intake were monitored, and at 15 weeks of age were killed at six time-points over 24 h. The clock genes Clock, Bmal1, Per2, and Cry2 in the suprachiasmatic nucleus (SCN) and appetite genes Npy and Pomc in the arcuate nucleus (ARC) were measured. Daily activity and feeding cycles in the HF/C, C/HF, and HF/HF offspring were altered, with increased feeding bouts and activity during the day and increased food intake but reduced activity at night. Gene expression patterns and levels of Clock, Bmal1, Per2, and Cry2 in the SCN and Npy and Pomc in the ARC were altered in HF diet-exposed offspring. The altered expression of hypothalamic molecular clock components and appetite genes, together with changes in activity and feeding rhythms, could be contributing to offspring obesity.
Collapse
Affiliation(s)
- Jane K Cleal
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Kimberley D Bruce
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Jasmin L Shearer
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Hugh Thomas
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Jack Plume
- Centre for Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton SO17 1BJ, UK.
| | - Louise Gregory
- Centre for Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton SO17 1BJ, UK.
| | - James N Shepard
- Centre for Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton SO17 1BJ, UK.
| | - Kerry L Spiers-Fitzgerald
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Ravi Mani
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Rohan M Lewis
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Karen A Lillycrop
- Centre for Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton SO17 1BJ, UK.
| | - Mark A Hanson
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Christopher D Byrne
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Felino R Cagampang
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| |
Collapse
|
29
|
González-Becerra K, Ramos-Lopez O, Barrón-Cabrera E, Riezu-Boj JI, Milagro FI, Martínez-López E, Martínez JA. Fatty acids, epigenetic mechanisms and chronic diseases: a systematic review. Lipids Health Dis 2019; 18:178. [PMID: 31615571 PMCID: PMC6792183 DOI: 10.1186/s12944-019-1120-6] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 09/30/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Chronic illnesses like obesity, type 2 diabetes (T2D) and cardiovascular diseases, are worldwide major causes of morbidity and mortality. These pathological conditions involve interactions between environmental, genetic, and epigenetic factors. Recent advances in nutriepigenomics are contributing to clarify the role of some nutritional factors, including dietary fatty acids in gene expression regulation. This systematic review assesses currently available information concerning the role of the different fatty acids on epigenetic mechanisms that affect the development of chronic diseases or induce protective effects on metabolic alterations. METHODS A targeted search was conducted in the PubMed/Medline databases using the keywords "fatty acids and epigenetic". The data were analyzed according to the PRISMA-P guidelines. RESULTS Consumption fatty acids like n-3 PUFA: EPA and DHA, and MUFA: oleic and palmitoleic acid was associated with an improvement of metabolic alterations. On the other hand, fatty acids that have been associated with the presence or development of obesity, T2D, pro-inflammatory profile, atherosclerosis and IR were n-6 PUFA, saturated fatty acids (stearic and palmitic), and trans fatty acids (elaidic), have been also linked with epigenetic changes. CONCLUSIONS Fatty acids can regulate gene expression by modifying epigenetic mechanisms and consequently result in positive or negative impacts on metabolic outcomes.
Collapse
Affiliation(s)
- K González-Becerra
- Institute of Traslational Nutrigenetics and Nutrigenomics, Health Sciences University Center, University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - O Ramos-Lopez
- Department of Nutrition, Food Science, Physiology and Toxicology, Centre for Nutrition Research, University of Navarra, Pamplona, Spain
- Faculty of Medicine and Psychology, Autonomous University of Baja California, Tijuana, B.C., Mexico
| | - E Barrón-Cabrera
- Institute of Traslational Nutrigenetics and Nutrigenomics, Health Sciences University Center, University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - J I Riezu-Boj
- Department of Nutrition, Food Science, Physiology and Toxicology, Centre for Nutrition Research, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - F I Milagro
- Department of Nutrition, Food Science, Physiology and Toxicology, Centre for Nutrition Research, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn), Carlos III Health Institute, Madrid, Spain
| | - E Martínez-López
- Institute of Traslational Nutrigenetics and Nutrigenomics, Health Sciences University Center, University of Guadalajara, Guadalajara, Jalisco, Mexico.
- Department of Molecular Biology in Medicine, Health Sciences University Center, University of Guadalajara, Sierra Mojada 950, 44340, Guadalajara, Jalisco, Mexico.
| | - J A Martínez
- Department of Nutrition, Food Science, Physiology and Toxicology, Centre for Nutrition Research, University of Navarra, Pamplona, Spain
- Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
- Centro de Investigación Biomédica en Red Fisiopatología de la Obesidad y Nutrición (CIBERobn), Carlos III Health Institute, Madrid, Spain
- Madrid Institute of Advanced Studies (IMDEA Food), Madrid, Spain
| |
Collapse
|
30
|
Şanlı E, Kabaran S. Maternal Obesity, Maternal Overnutrition and Fetal Programming: Effects of Epigenetic Mechanisms on the Development of Metabolic Disorders. Curr Genomics 2019; 20:419-427. [PMID: 32476999 PMCID: PMC7235386 DOI: 10.2174/1389202920666191030092225] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 10/12/2019] [Accepted: 10/21/2019] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Maternal obesity and maternal overnutrition, can lead to epigenetic alterations during pregnancy and these alterations can influence fetal and neonatal phenotype which increase the risk of metabolic disorders in later stages of life. OBJECTIVE The effects of maternal obesity on fetal programming and potential mechanisms of maternal epigenetic regulation of gene expression which have persistent effects on fetal health and development were investigated. METHODS Review of the literature was carried out in order to discuss the effects of maternal obesity and epigenetic mechanisms in fetal programming of metabolic disorders. All abstracts and full-text articles were examined and the most relevant articles were included in this review. RESULTS Maternal obesity and maternal overnutrition during fetal period has important overall effects on long-term health. Maternal metabolic alterations during early stages of fetal development can lead to permanent changes in organ structures, cell numbers and metabolism. Epigenetic modifications (DNA methylation, histone modifications, microRNAs) play an important role in disease susceptibility in the later stages of human life. Maternal nutrition alter expression of hypothalamic genes which can increase fetal and neonatal energy intake. Epigenetic modifications may affect the increasing rate of obesity and other metabolic disorders worldwide since the impact of these changes can be passed through generations. CONCLUSION Weight management before and during pregnancy, together with healthy nutritional intakes may improve the maternal metabolic environment, which can reduce the risks of fetal programming of metabolic diseases. Further evidence from long-term follow-up studies are needed in order to determine the role of maternal obesity on epigenetic mechanisms.
Collapse
Affiliation(s)
- Ezgi Şanlı
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Eastern Mediterranean University, Famagusta, T.R. North Cyprus via Mersin 10, Turkey
| | - Seray Kabaran
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Eastern Mediterranean University, Famagusta, T.R. North Cyprus via Mersin 10, Turkey
| |
Collapse
|
31
|
Abstract
Non-communicable diseases (NCD) such as type-2 diabetes and CVD are now highly prevalent in both developed and developing countries. Evidence from both human and animal studies shows that early-life nutrition is an important determinant of NCD risk in later life. The mechanism by which the early-life environment influences future disease risk has been suggested to include the altered epigenetic regulation of gene expression. Epigenetic processes regulate the accessibility of genes to the cellular proteins that control gene transcription, determining where and when a gene is switched on and its level of activity. Epigenetic processes not only play a central role in regulating gene expression but also allow an organism to adapt to the environment. In this review, we will focus on how both maternal and paternal nutrition can alter the epigenome and the evidence that these changes are causally involved in determining future disease risk.
Collapse
Affiliation(s)
- Mark A Burton
- Academic Unit of Human Development and Health, Faculty of Medicine,University of Southampton,Southampton,UK
| | - Karen A Lillycrop
- Centre for Biological Sciences, Faculty of Natural and Environmental Sciences,University of Southampton,Southampton,UK
| |
Collapse
|
32
|
He P, Wei P, Chen X, Lin Y, Peng J. Identification and characterization of microRNAs in the gonad of Trachinotus ovatus using Solexa sequencing. COMPARATIVE BIOCHEMISTRY AND PHYSIOLOGY D-GENOMICS & PROTEOMICS 2019; 30:312-320. [DOI: 10.1016/j.cbd.2019.03.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 03/27/2019] [Accepted: 03/28/2019] [Indexed: 01/27/2023]
|
33
|
Pomar CA, Castro H, Picó C, Serra F, Palou A, Sánchez J. Cafeteria Diet Consumption during Lactation in Rats, Rather than Obesity Per Se, alters miR-222, miR-200a, and miR-26a Levels in Milk. Mol Nutr Food Res 2019; 63:e1800928. [DOI: 10.1002/mnfr.201800928] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 01/09/2019] [Indexed: 01/21/2023]
Affiliation(s)
- Catalina A. Pomar
- Laboratory of Molecular Biology; Nutrition and Biotechnology (Nutrigenomics and Obesity); University of the Balearic Islands; Palma de Mallorca Spain
- CIBER Fisiopatología de la Obesidad y Nutrición; Madrid Spain C.P. 28029
| | - Heriberto Castro
- Laboratory of Molecular Biology; Nutrition and Biotechnology (Nutrigenomics and Obesity); University of the Balearic Islands; Palma de Mallorca Spain
- Universidad Autónoma de Nuevo León; Facultad de Salud Pública y Nutrición; Nuevo León México C.P. 64460
| | - Catalina Picó
- Laboratory of Molecular Biology; Nutrition and Biotechnology (Nutrigenomics and Obesity); University of the Balearic Islands; Palma de Mallorca Spain
- CIBER Fisiopatología de la Obesidad y Nutrición; Madrid Spain C.P. 28029
- Instituto de Investigación Sanitaria Illes Balears; Palma de Mallorca Spain C.P. 07120
| | - Francisca Serra
- Laboratory of Molecular Biology; Nutrition and Biotechnology (Nutrigenomics and Obesity); University of the Balearic Islands; Palma de Mallorca Spain
- CIBER Fisiopatología de la Obesidad y Nutrición; Madrid Spain C.P. 28029
- Instituto de Investigación Sanitaria Illes Balears; Palma de Mallorca Spain C.P. 07120
| | - Andreu Palou
- Laboratory of Molecular Biology; Nutrition and Biotechnology (Nutrigenomics and Obesity); University of the Balearic Islands; Palma de Mallorca Spain
- CIBER Fisiopatología de la Obesidad y Nutrición; Madrid Spain C.P. 28029
- Instituto de Investigación Sanitaria Illes Balears; Palma de Mallorca Spain C.P. 07120
| | - Juana Sánchez
- Laboratory of Molecular Biology; Nutrition and Biotechnology (Nutrigenomics and Obesity); University of the Balearic Islands; Palma de Mallorca Spain
- CIBER Fisiopatología de la Obesidad y Nutrición; Madrid Spain C.P. 28029
- Instituto de Investigación Sanitaria Illes Balears; Palma de Mallorca Spain C.P. 07120
| |
Collapse
|
34
|
Liu Y, Shen J, Yang X, Sun Q, Yang X. Folic Acid Reduced Triglycerides Deposition in Primary Chicken Hepatocytes. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2018; 66:13162-13172. [PMID: 30484310 DOI: 10.1021/acs.jafc.8b05193] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Abdominal fat or fatty liver cause huge economic losses in the poultry industry, and nonalcoholic fatty liver disease (NAFLD) is also a global health issue in humans. More than 90% of de novo lipogenesis in humans and chickens is undertaken by the liver, which is proved to be full of lipids in new-born chickens. Folic acid was thought to have correlation with lipid metabolism. Primary hepatocytes from new-born chickens were employed as a natural model of early stage fatty liver in vitro and further to explore whether folic acid could prevent fatty liver in the current study. We found that folic acid addition reduced triglyceride deposition by suppressing de novo fatty acid synthesis and coordinately promoting triglyceride hydrolysis and exportation in primary chicken hepatocytes from new-born chickens. In addition, lipogenesis suppression was through the PI3K/AKT/SREBP pathway mediated by weakening insulin/IGF signal. Our data suggested that folic acid may be considered as a precautionary strategy for abdominal fat deposition in broilers or fatty liver in laying hens and humans. In addition, mechanism regulation also implied that an IGF2 inhibitor and PI3K inhibitor may be used for the NAFLD precautionary measure to reduce TG deposition.
Collapse
Affiliation(s)
- Yanli Liu
- College of Animal Science and Technology , Northwest A&F University , Yangling , China
| | - Jing Shen
- College of Animal Science and Technology , Northwest A&F University , Yangling , China
| | - Xin Yang
- College of Animal Science and Technology , Northwest A&F University , Yangling , China
| | - Qingzhu Sun
- College of Animal Science and Technology , Northwest A&F University , Yangling , China
| | - Xiaojun Yang
- College of Animal Science and Technology , Northwest A&F University , Yangling , China
| |
Collapse
|
35
|
Moradi Sarabi M, Zahedi SA, Pajouhi N, Khosravi P, Bagheri S, Ahmadvand H, Shahryarhesami S. The effects of dietary polyunsaturated fatty acids on miR-126 promoter DNA methylation status and VEGF protein expression in the colorectal cancer cells. GENES AND NUTRITION 2018; 13:32. [PMID: 30598703 PMCID: PMC6299631 DOI: 10.1186/s12263-018-0623-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 11/29/2018] [Indexed: 02/06/2023]
Abstract
Background There is increasing evidence indicating an aberrant expression of miRNAs in colorectal cancer (CRC) development. Growing evidence has suggested that polyunsaturated fatty acids (PUFAs) could modulate the remodeling of the epigenome. No study has yet been published to examine the direct effect of PUFA on the promoter methylation of miRNAs. This study aimed to examine the potential clinical application of PUFA on the promoter DNA methylation of miR-126 and its angiogenic target molecule (VEGF) in the CRC cells. Methods We investigated the direct effect of 100 μM EPA, DHA, and LA for 24 h on promoter methylation status of miR-126 in a panel of five CRC cell lines (HCT116, HT29/219, Caco2, SW742, and LS180) by methylation-specific PCR (MSP). We also quantified the miR-126 and VEGF transcript expression levels in five CRC cell lines affected by PUFA by real-time PCR. Moreover, we analyzed the protein expression level of VEGF, as a target of miR-126, by western blotting assay. Results MSP analysis showed extensive DNA methylation of the miR-126 promoter in all five CRC cell lines, and among all three PUFAs, only DHA completely demethylated the promoter of miR-126 in HCT116 and Caco2 cell lines. We found that only DHA significantly induces the expression level of miR-126 in HCT116 and Caco2 cell lines, respectively, by 20.1-fold and 1.68-fold (p < 0.05). Our finding indicates that the downregulation of VEGF protein level is also effectively observed only in DHA-treated HCT116 and Caco2 cells compared to control cells (p < 0.05). Conclusions Our results provide evidence that n-3 PUFAs are able to modulate cellular miR-126 DNA methylation and inhibit VEGF expression level in a cell-type specific manner in colorectal cancer cells. DHA always showed higher efficacy than EPA and LA in our experiment. Overall, our results suggest a potential clinical application of n-3 PUFAs as anti-angiogenic agents in CRC therapy.
Collapse
Affiliation(s)
- Mostafa Moradi Sarabi
- 1Department of Biochemistry and Genetics, Lorestan University of Medical Sciences, School of Medicine, Khorramabad, 381251698 Iran.,3Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Seyed Abdollah Zahedi
- 1Department of Biochemistry and Genetics, Lorestan University of Medical Sciences, School of Medicine, Khorramabad, 381251698 Iran
| | - Naser Pajouhi
- 2Department of Physiology, School of Medicine, Lorestan University of Medical Sciences, Khorramabad, Iran.,3Razi Herbal Medicines Research Center, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Peyman Khosravi
- 1Department of Biochemistry and Genetics, Lorestan University of Medical Sciences, School of Medicine, Khorramabad, 381251698 Iran.,4Student Research Committee, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Shahrokh Bagheri
- 1Department of Biochemistry and Genetics, Lorestan University of Medical Sciences, School of Medicine, Khorramabad, 381251698 Iran
| | - Hassan Ahmadvand
- 1Department of Biochemistry and Genetics, Lorestan University of Medical Sciences, School of Medicine, Khorramabad, 381251698 Iran
| | | |
Collapse
|
36
|
Xie R, Sun Y, Wu J, Huang S, Jin G, Guo Z, Zhang Y, Liu T, Liu X, Cao X, Wang B, Cao H. Maternal High Fat Diet Alters Gut Microbiota of Offspring and Exacerbates DSS-Induced Colitis in Adulthood. Front Immunol 2018; 9:2608. [PMID: 30483266 PMCID: PMC6243010 DOI: 10.3389/fimmu.2018.02608] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 10/23/2018] [Indexed: 12/11/2022] Open
Abstract
Background: Accumulating evidence shows that high fat diet is closely associated with inflammatory bowel disease. However, the effects and underlying mechanisms of maternal high fat diet (MHFD) on the susceptibility of offspring to colitis in adulthood lacks confirmation. Methods: C57BL/6 pregnant mice were given either a high fat (60 E% fat, MHFD group) or control diet [10 E% fat, maternal control diet (MCD) group] during gestation and lactation. The intestinal development, mucosal barrier function, microbiota, and mucosal inflammation of 3-week old offspring were assessed. After weaning all mice were fed a control diet until 8 weeks of age when the microbiota was analyzed. Offspring were also treated with 2% DSS solution for 5 days and the severity of colitis was assessed. Results: The offspring in MHFD group were significantly heavier than those in MCD group only at 2–4 weeks of age, while no differences were found in the body weight between two groups at other measured time points. Compared with MCD group, MHFD significantly inhibited intestinal development and disrupted barrier function in 3-week old offspring. Although H&E staining showed no obvious microscopic inflammation in both groups of 3-week old offspring, increased production of inflammatory cytokines indicated low-grade inflammation was induced in MHFD group. Moreover, fecal analysis of the 3-week old offspring indicated that the microbiota compositions and diversity were significantly changed in MHFD group. Interestingly after 5 weeks consumption of control diet in both groups, the microbiota composition of offspring in MHFD group was still different from that in MCD group, although the bacterial diversity was partly recovered at 8 weeks of age. Finally, after DSS treatment in 8-week old offspring, MHFD significantly exacerbated the severity of colitis and increased the production of proinflammatory cytokine. Conclusions: Our data reveal that MHFD in early life can inhibit intestinal development, induce dysbiosis and low-grade inflammation and lead to the disruption of intestinal mucosal barrier in offspring, and enhance DSS-induced colitis in adulthood.
Collapse
Affiliation(s)
- Runxiang Xie
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Yue Sun
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Jingyi Wu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Shumin Huang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Ge Jin
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Zixuan Guo
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Yujie Zhang
- Department of Pathology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Tianyu Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Xiang Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Xiaocang Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| |
Collapse
|
37
|
The Glucose-Regulated MiR-483-3p Influences Key Signaling Pathways in Cancer. Cancers (Basel) 2018; 10:cancers10060181. [PMID: 29867024 PMCID: PMC6025222 DOI: 10.3390/cancers10060181] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 05/25/2018] [Accepted: 05/29/2018] [Indexed: 02/06/2023] Open
Abstract
The hsa-mir-483 gene, located within the IGF2 locus, transcribes for two mature microRNAs, miR-483-5p and miR-483-3p. This gene, whose regulation is mediated by the the CTNNB1/USF1 complex, shows an independent expression from its host gene IGF2. The miR-483-3p affects the Wnt/β-catenin, the TGF-β, and the TP53 signaling pathways by targeting several genes as CTNNB1, SMAD4, IGF1, and BBC3. Accordingly, miR-483-3p is associated with various tissues specific physiological properties as insulin and melanin production, as well as with cellular physiological functions such as wounding, differentiation, proliferation, and survival. Deregulation of miR-483-3p is observed in different types of cancer, and its overexpression can inhibit the pro-apoptotic pathway induced by the TP53 target effectors. As a result, the oncogenic characteristics of miR-483-3p are linked to the effect of some of the most relevant cancer-related genes, TP53 and CTNNB1, as well as to one of the most important cancer hallmark: the aberrant glucose metabolism of tumor cells. In this review, we summarize the recent findings regarding the miR-483-3p, to elucidate its functional role in physiological and pathological contexts, focusing overall on its involvement in cancer and in the TP53 pathway.
Collapse
|
38
|
Marques-Rocha JL, Garcia-Lacarte M, Samblas M, Bressan J, Martínez JA, Milagro FI. Regulatory roles of miR-155 and let-7b on the expression of inflammation-related genes in THP-1 cells: effects of fatty acids. J Physiol Biochem 2018; 74:579-589. [PMID: 29790117 DOI: 10.1007/s13105-018-0629-x] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 04/03/2018] [Indexed: 12/21/2022]
Abstract
The main aim of this investigation was to study the regulatory roles of let-7b and miR-155-3p on the expression of inflammation-associated genes in monocytes, macrophages, and lipopolysaccharide (LPS)-activated macrophages (AcM). A second goal was to analyze the potential modulatory roles of different fatty acids, including oleic, palmitic, eicosapentaenoic (EPA), and docosahexaenoic (DHA), on the expression of these miRNAs in the three cell types. This hypothesis was tested in human acute monocytic leukemia cells (THP-1), which were differentiated into macrophages with 2-O-tetradecanoylphorbol-13-acetate (TPA) and further activated with LPS for 24 h. Monocytes, macrophages, and AcM were transfected with a negative control, or mimics for miR-155-3p and miR-let-7b-5p. The expression of both miRNAs and some proinflammatory genes was analyzed by qRT-PCR. Interestingly, let-7b mimic reduced the expression of IL6 and TNF in monocytes, and SERPINE1 expression in LPS-activated macrophages. However, IL6, TNF, and SERPINE1 were upregulated in macrophages by let-7b mimic. IL6 expression was higher in the three types of cells after transfecting with miR-155-3p mimic. Similarly, expression of SERPINE1 was increased by miR-155-3p mimic in monocytes and macrophages. However, TLR4 was downregulated by miR-155-3p in monocytes and macrophages. Regarding the effects of the different fatty acids, oleic acid increased the expression of let-7b in macrophages and AcM and also increased the expression of miR-155 in monocytes when compared with DHA but not when compared with non-treated cells. Overall, these results suggest anti- and proinflammatory roles of let-7b and miR-155-3p in THP-1 cells, respectively, although these outcomes are strongly dependent on the cell type. Noteworthy, oleic acid might exert beneficial anti-inflammatory effects in immune cells (i.e., non-activated and LPS-activated macrophages) by upregulating the expression of let-7b.
Collapse
Affiliation(s)
- J L Marques-Rocha
- Department of Integrated Education of Health, Federal University of Espirito Santo, Vitória, Brazil
| | - M Garcia-Lacarte
- Department of Nutrition, Food Science and Physiology; Centre for Nutrition Research, University of Navarra, c / Irunlarrea 1, 31008, Pamplona, Navarra, Spain
| | - M Samblas
- Department of Nutrition, Food Science and Physiology; Centre for Nutrition Research, University of Navarra, c / Irunlarrea 1, 31008, Pamplona, Navarra, Spain
| | - J Bressan
- Department of Nutrition and Health, Federal University of Viçosa, Viçosa, Brazil
| | - J A Martínez
- Department of Nutrition, Food Science and Physiology; Centre for Nutrition Research, University of Navarra, c / Irunlarrea 1, 31008, Pamplona, Navarra, Spain.,CIBERobn, Fisiopatología de la Obesidad y la Nutrición, Carlos III Health Institute, Madrid, Spain.,IdiSNA, Navarra Institute for Health Research, Pamplona, Spain.,IMDEA Food, Madrid, Spain
| | - F I Milagro
- Department of Nutrition, Food Science and Physiology; Centre for Nutrition Research, University of Navarra, c / Irunlarrea 1, 31008, Pamplona, Navarra, Spain. .,CIBERobn, Fisiopatología de la Obesidad y la Nutrición, Carlos III Health Institute, Madrid, Spain.
| |
Collapse
|
39
|
Marjonen H, Toivonen M, Lahti L, Kaminen-Ahola N. Early prenatal alcohol exposure alters imprinted gene expression in placenta and embryo in a mouse model. PLoS One 2018; 13:e0197461. [PMID: 29763474 PMCID: PMC5953443 DOI: 10.1371/journal.pone.0197461] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 05/02/2018] [Indexed: 12/22/2022] Open
Abstract
Prenatal alcohol exposure (PAE) can harm the embryonic development and cause life-long consequences in offspring’s health. To clarify the molecular mechanisms of PAE we have used a mouse model of early alcohol exposure, which is based on maternal ad libitum ingestion of 10% (v/v) ethanol for the first eight days of gestation (GD 0.5–8.5). Owing to the detected postnatal growth-restricted phenotype in the offspring of this mouse model and both prenatal and postnatal growth restriction in alcohol-exposed humans, we focused on imprinted genes Insulin-like growth factor 2 (Igf2), H19, Small Nuclear Ribonucleoprotein Polypeptide N (Snrpn) and Paternally expressed gene 3 (Peg3), which all are known to be involved in embryonic and placental growth and development. We studied the effects of alcohol on DNA methylation level at the Igf2/H19 imprinting control region (ICR), Igf2 differentially methylated region 1, Snrpn ICR and Peg3 ICR in 9.5 embryonic days old (E9.5) embryos and placentas by using MassARRAY EpiTYPER. To determine alcohol-induced alterations globally, we also examined methylation in long interspersed nuclear elements (Line-1) in E9.5 placentas. We did not observe any significant alcohol-induced changes in DNA methylation levels. We explored effects of PAE on gene expression of E9.5 embryos as well as E9.5 and E16.5 placentas by using quantitative PCR. The expression of growth promoter gene Igf2 was decreased in the alcohol-exposed E9.5 and E16.5 placentas. The expression of negative growth controller H19 was significantly increased in the alcohol-exposed E9.5 embryos compared to controls, and conversely, a trend of decreased expression in alcohol-exposed E9.5 and E16.5 placentas were observed. Furthermore, increased Snrpn expression in alcohol-exposed E9.5 embryos was also detected. Our study indicates that albeit no alterations in the DNA methylation levels of studied sequences were detected by EpiTYPER, early PAE can affect the expression of imprinted genes in both developing embryo and placenta.
Collapse
Affiliation(s)
- Heidi Marjonen
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland
| | - Mia Toivonen
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland
| | - Laura Lahti
- Department of Biological and Environmental Sciences, Division of Genetics, University of Helsinki, Helsinki, Finland
| | - Nina Kaminen-Ahola
- Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland
- * E-mail:
| |
Collapse
|
40
|
Zhao M, Li Y, Yao H, Dou L, Zhang S, Zhao Q, Li L. Sex-specific Alterations in Serology and the Expression of Liver FATP4 Protein in Offspring Exposed to High-Fat Diet during Pregnancy and/or Lactation. Lipids 2018; 53:301-311. [PMID: 29701266 DOI: 10.1002/lipd.12029] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 01/20/2018] [Accepted: 01/30/2018] [Indexed: 11/06/2022]
Abstract
Changes in dietary composition will have a significant impact on the nutritional status of the mother and the offspring. To examine the relevant hormone level changes during lactation and the expression of fatty acid transporters in the placenta and liver under the condition of a high-fat (HF) diet, we established HF animal models and conducted a cross-fostering program to mimic the shift in diet. On gestation day (GD)18, the weight of placenta in the HF group was significantly higher than that in the control group (p < 0.05). HF-fed male pups had a significantly lower serum insulin level, but the same phenomenon was not found in females. On the contrary, serum triacylglycerol (TAG) level presented a tendency to decrease only in female offspring. Oil red O staining showed lipid accumulation in the HF diet offspring livers. The mRNA levels of FATP4 in the placenta in the HF diet group were significantly upregulated compared to the control diet group (p < 0.05). High-fat diet (HFD) consumption also altered the liver mRNA levels of FATP4, SREBP-1, and SCD-1 in the male offspring, while the changes in protein levels of FATP4 were not observed in either sex. In conclusion, maternal HF diet has a profound impact on offspring growth, metabolism, and the risk of metabolic disorders, which would depend on the exposure period of pregnancy and lactation.
Collapse
Affiliation(s)
- Mingqiu Zhao
- Department of Food and Nutrition Hygiene, School of Public Health, Anhui Medical University, Meishan Road, Hefei, Anhui, 230601, China
| | - Yi Li
- Anhui Provincial Hospital, Hefei, Anhui, China
| | - Huihui Yao
- Department of Food and Nutrition Hygiene, School of Public Health, Anhui Medical University, Meishan Road, Hefei, Anhui, 230601, China
| | - Lihua Dou
- Department of Food and Nutrition Hygiene, School of Public Health, Anhui Medical University, Meishan Road, Hefei, Anhui, 230601, China
| | - Shuya Zhang
- Department of Food and Nutrition Hygiene, School of Public Health, Anhui Medical University, Meishan Road, Hefei, Anhui, 230601, China
| | - Qihong Zhao
- Department of Food and Nutrition Hygiene, School of Public Health, Anhui Medical University, Meishan Road, Hefei, Anhui, 230601, China
| | - Li Li
- Department of Food and Nutrition Hygiene, School of Public Health, Anhui Medical University, Meishan Road, Hefei, Anhui, 230601, China
| |
Collapse
|
41
|
Matoušková P, Hanousková B, Skálová L. MicroRNAs as Potential Regulators of Glutathione Peroxidases Expression and Their Role in Obesity and Related Pathologies. Int J Mol Sci 2018; 19:ijms19041199. [PMID: 29662007 PMCID: PMC5979329 DOI: 10.3390/ijms19041199] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2018] [Revised: 04/08/2018] [Accepted: 04/10/2018] [Indexed: 12/19/2022] Open
Abstract
Glutathione peroxidases (GPxs) belong to the eight-member family of phylogenetically related enzymes with different cellular localization, but distinct antioxidant function. Several GPxs are important selenoproteins. Dysregulated GPx expression is connected with severe pathologies, including obesity and diabetes. We performed a comprehensive bioinformatic analysis using the programs miRDB, miRanda, TargetScan, and Diana in the search for hypothetical microRNAs targeting 3′untranslated regions (3´UTR) of GPxs. We cross-referenced the literature for possible intersections between our results and available reports on identified microRNAs, with a special focus on the microRNAs related to oxidative stress, obesity, and related pathologies. We identified many microRNAs with an association with oxidative stress and obesity as putative regulators of GPxs. In particular, miR-185-5p was predicted by a larger number of programs to target six GPxs and thus could play the role as their master regulator. This microRNA was altered by selenium deficiency and can play a role as a feedback control of selenoproteins’ expression. Through the bioinformatics analysis we revealed the potential connection of microRNAs, GPxs, obesity, and other redox imbalance related diseases.
Collapse
Affiliation(s)
- Petra Matoušková
- Faculty of Pharmacy, Department of Biochemical Sciences, Charles University, 500 05, Hradec Králové, Czech Republic.
| | - Barbora Hanousková
- Faculty of Pharmacy, Department of Biochemical Sciences, Charles University, 500 05, Hradec Králové, Czech Republic.
| | - Lenka Skálová
- Faculty of Pharmacy, Department of Biochemical Sciences, Charles University, 500 05, Hradec Králové, Czech Republic.
| |
Collapse
|
42
|
Macrophage Populations in Visceral Adipose Tissue from Pregnant Women: Potential Role of Obesity in Maternal Inflammation. Int J Mol Sci 2018; 19:ijms19041074. [PMID: 29617296 PMCID: PMC5979476 DOI: 10.3390/ijms19041074] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 03/22/2018] [Accepted: 03/26/2018] [Indexed: 12/23/2022] Open
Abstract
Obesity is associated with inflammatory changes and accumulation and phenotype polarization of adipose tissue macrophages (ATMs). Obese pregnant women have alterations in adipose tissue composition, but a detailed description of macrophage population is not available. In this study, we characterized macrophage populations in visceral adipose tissue (VAT) from pregnant women with normal, overweight, and obese pregestational weight. Immunophenotyping of macrophages from VAT biopsies was performed by flow cytometry using CD45 and CD14 as markers of hematopoietic and monocyte linage, respectively, while HLA-DR, CD11c, CD163, and CD206 were used as pro- and anti-inflammatory markers. Adipocyte number and size were evaluated by light microscopy. The results show that pregnant women that were overweight and obese during the pregestational period had adipocyte hypertrophy. Two different macrophage populations in VAT were identified: recruited macrophages (CD45+CD14+), and a novel population lacking CD45, which was considered to be a resident macrophages subset (CD45−CD14+). The number of resident HLA−DRlow/− macrophages showed a negative correlation with body mass index (BMI). Both resident and recruited macrophages from obese women expressed higher CD206 levels. CD11c expression was higher in resident HLA-DR+ macrophages from obese women. A strong correlation between CD206 and CD11c markers and BMI was observed. Our findings show that being overweight and obese in the pregestational period is associated with adipocyte hypertrophy and specific ATMs populations in VAT.
Collapse
|
43
|
Abstract
Developmental programming resulting from maternal malnutrition can lead to an increased risk of metabolic disorders such as obesity, insulin resistance, type 2 diabetes and cardiovascular disorders in the offspring in later life. Furthermore, many conditions linked with developmental programming are also known to be associated with the aging process. This review summarizes the available evidence about the molecular mechanisms underlying these effects, with the potential to identify novel areas of therapeutic intervention. This could also lead to the discovery of new treatment options for improved patient outcomes.
Collapse
|
44
|
Abstract
Nonalcoholic fatty liver disease (NAFLD) is currently the most common cause of chronic liver disease worldwide and is present in a third of the general population and the majority of individuals with obesity and type 2 diabetes. Importantly, NAFLD can progress to severe nonalcoholic steatohepatitis (NASH), associated with liver failure and hepatocellular carcinoma. Recent research efforts have extensively focused on identifying factors contributing to the additional "hit" required to promote NALFD disease progression. The maternal diet, and in particular a high-fat diet (HFD), may be one such hit "priming" the development of severe fatty liver disease, a notion supported by the increasing incidence of NAFLD among children and adolescents in Westernized countries. In recent years, a plethora of key studies have used murine models of maternal obesity to identify fundamental mechanisms such as lipogenesis, mitochondrial function, inflammation, and fibrosis that may underlie the developmental priming of NAFLD. In this chapter, we will address key considerations for constructing experimental models and both conventional and advanced methods of quantifying NAFLD disease status.
Collapse
Affiliation(s)
- Kimberley D Bruce
- Division of Endocrinology, Metabolism, and Diabetes, University of Colorado Anschutz Medical Campus, Aurora, CO, USA.
| | - Karen R Jonscher
- Department of Anesthesiology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
45
|
Imam MU, Ismail M. The Impact of Traditional Food and Lifestyle Behavior on Epigenetic Burden of Chronic Disease. GLOBAL CHALLENGES (HOBOKEN, NJ) 2017; 1:1700043. [PMID: 31565292 PMCID: PMC6607231 DOI: 10.1002/gch2.201700043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 07/12/2017] [Indexed: 05/11/2023]
Abstract
Noncommunicable chronic diseases (NCCDs) are the leading causes of morbidity and mortality globally. The mismatch between present day diets and ancestral genome is suggested to contribute to the NCCDs burden, which is promoted by traditional risk factors like unhealthy diets, physical inactivity, alcohol and tobacco. However, epigenetic evidence now suggests that cumulatively inherited epigenetic modifications may have made humans more prone to the effects of present day lifestyle factors. Perinatal starvation was widespread in the 19th century. This together with more recent events like increasing consumption of western and low fiber diets, smoking, harmful use of alcohol, physical inactivity, and environmental pollutants may have programed the human epigenome for higher NCCDs risk. In this review, on the basis of available epigenetic data it is hypothesized that transgenerational effects of lifestyle factors may be contributing to the current global burden of NCCDs. Thus, there is a need to reconsider prevention strategies so that the subsequent generations will not have to pay for our sins and those of our ancestors.
Collapse
Affiliation(s)
- Mustapha U. Imam
- Precision Nutrition Innovation InstituteCollege of Public HealthZhengzhou UniversityZhengzhou450001China
| | - Maznah Ismail
- Laboratory of Molecular BiomedicineInstitute of BioscienceUniversiti Putra MalaysiaSerdangSelangor43400Malaysia
| |
Collapse
|
46
|
Huang Y, Ye T, Liu C, Fang F, Chen Y, Dong Y. Maternal high-fat diet during pregnancy and lactation affects hepatic lipid metabolism in early life of offspring rat. J Biosci 2017; 42:311-319. [DOI: 10.1007/s12038-017-9675-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
47
|
Wilson RA, Deasy W, Hayes A, Cooke MB. High fat diet and associated changes in the expression of micro-RNAs in tissue: Lessons learned from animal studies. Mol Nutr Food Res 2017; 61. [PMID: 28233461 DOI: 10.1002/mnfr.201600943] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 01/15/2017] [Accepted: 02/13/2017] [Indexed: 12/13/2022]
Abstract
Environment and genetic factors play an important role in the development of obesity, and diet is one of the main contributing factors to this disease. High fat intake is associated with body weight gain, leading to obesity and other metabolic diseases. MicroRNAs (miRNAs) are a group of small, noncoding RNAs that are important regulators of gene expression at posttranscriptional level. Studies have shown that high fat intake, independent of body weight status, can significantly impact both negatively and positively the expression of miRNAs and thus the biological function of tissues such as adipose, skeletal, and cardiac muscle, liver, neuronal, and endothelial. This review will summarize the effects of high calorie diet in the form of high fat intake on miRNA expression in various tissues of animal models and of high fat fed offspring. We will also briefly review the impact of different dietary lipids on miRNA expression. Given changes in miRNA expression have been associated with the development of many diseases including obesity, understanding their biological role could have important clinical implications and offer tangible therapeutic targets for the prevention, management, and/or treatment of obesity and other lifestyle-related disorders.
Collapse
Affiliation(s)
- Robin A Wilson
- College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia.,Australian Institute for Musculoskeletal Science (AIMSS), Western Health, Melbourne, VIC, Australia
| | - William Deasy
- College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia.,Australian Institute for Musculoskeletal Science (AIMSS), Western Health, Melbourne, VIC, Australia
| | - Alan Hayes
- College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia.,Australian Institute for Musculoskeletal Science (AIMSS), Western Health, Melbourne, VIC, Australia
| | - Matthew B Cooke
- College of Health and Biomedicine, Victoria University, Melbourne, VIC, Australia.,Australian Institute for Musculoskeletal Science (AIMSS), Western Health, Melbourne, VIC, Australia
| |
Collapse
|
48
|
Mousavi SN, Koohdani F, Shidfar F, Eslaminejad MB, Izadi P, Eshraghian M, Shafieineek L, Tohidinik H. Effects of Maternal Isocaloric Diet Containing Different Amounts of Soy Oil and Extra Virgin Olive Oil on Weight, Serum Glucose, and Lipid Profile of Female Mice Offspring. IRANIAN JOURNAL OF MEDICAL SCIENCES 2017; 42:161-169. [PMID: 28360442 PMCID: PMC5366364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
BACKGROUND Health status of offspring is programmed by maternal diet throughout gestation and lactation. The present study investigates the lasting effects of maternal supplementation with different amounts of soy oil or extra virgin olive oil (EVOO) on weight and biochemical parameters during gestation and lactation of female mice offspring. METHODS Eight weeks old female C57BL/6 mice (n=40) were assigned through simple randomization into four isocaloric dietary groups (16% of calories as soy oil (LSO) or EVOO (LOO) and 45% of calories as soy oil (HSO) or EVOO (HOO)) during three weeks of gestation and lactation. After weaning (at 3 weeks), all offspring received a diet containing 16% of calories as soy oil and were sacrificed at 6 weeks. Two-way ANOVA was used to adjust for confounding variables and repeated measures test for weight gain trend. Statistical analyses were performed with the IBM SPSS package. RESULTS At birth and adolescence, the weight of offspring was significantly higher in the soy oil than the olive oil groups (P<0.001 and P<0.001, respectively). Adolescence weight was significantly higher in the offspring born to mothers fed with 16% oil than those with 45% oil (P=0.001). Serum glucose, triglyceride and total cholesterol were significantly higher in the LSO than LOO (P<0.001, P<0.001 and P<0.001), LSO than HSO (P<0.001, P=0.03 and P<0.001), and LOO than HOO (P<0.001, P<0.001 and P<0.001) dietary groups, respectively. Serum triglyceride and total cholesterol were significantly higher in the offspring of HSO than HOO fed mothers (P<0.001 and P<0.001, respectively). CONCLUSION A maternal diet containing EVOO has better effects on birth weight, as well as weight and serum biochemical parameters in offspring at adolescence.
Collapse
Affiliation(s)
- Seyedeh Neda Mousavi
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Fariba Koohdani
- Department of Cellular and Molecular Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzad Shidfar
- Department of Nutrition, School of Public Health, Colorectal Research Center, Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran,Correspondence: Farzad Shidfar, PhD; Department of Nutrition, School of Public Health, Colorectal Research Center, Rasoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran Tel: +98 21 88622533 Fax: +98 21 88622755
| | - Mohamadreza Baghaban Eslaminejad
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Pantea Izadi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammadreza Eshraghian
- Department of Statistics, School of Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Shafieineek
- Department of Sport Physiology, School of Physical Education and Sport Sciences, University of Tehran, Tehran, Iran
| | - Hamidreza Tohidinik
- School of Public Health, Gonabad University of Medical Sciences, Gonabad, Iran
| |
Collapse
|
49
|
de Paula Simino LA, de Fante T, Figueiredo Fontana M, Oliveira Borges F, Torsoni MA, Milanski M, Velloso LA, Souza Torsoni A. Lipid overload during gestation and lactation can independently alter lipid homeostasis in offspring and promote metabolic impairment after new challenge to high-fat diet. Nutr Metab (Lond) 2017; 14:16. [PMID: 28239403 PMCID: PMC5319047 DOI: 10.1186/s12986-017-0168-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 02/06/2017] [Indexed: 02/06/2023] Open
Abstract
Background Nutritional status in early life is critically involved in the metabolic phenotype of offspring. However the changes triggered by maternal consumption of high-fat diet (HFD) in pre- or postnatal period should be better understood. Here we evaluated whether maternal HFD consumption during gestation and lactation could differently affect liver miR-122 and miR-370 expression leading to metabolic damages observed in offspring. Moreover, we investigate whether early overnutrition program offspring to more harmful response to HFD in later life. Methods Female mice were fed either a standard chow (SC) diet or a HFD three weeks before and during mating, gestation and/or lactation. Offspring were evaluated on the delivery day (d0), in a cross-fostering model at day 28 (d28) and in adult life, after a re-challenge with a HFD (d82). Results In vitro analysis using liver cell line showed that palmitate could induced decrease in miR-122 and increase in miR-370 expression. Newborn pups (d0) from obese dams showed a decrease in lipid oxidation markers (Cpt1a and Acadvl), an increase in triacylglycerol synthesis markers (Agpat and Gpam), as well as lower miR-122 and higher miR-370 hepatic content that was inversely correlated to maternal serum NEFA and TAG. Pups fostered to SC dams presented an increase in body weight and Agpat/Gpam expression at d28 compared to pups fostered to HFD dams and an inverse correlation was observed between miR-122 hepatic expression and offspring serum TAG. In adult life (d82), the reintroduction of HFD resulted in higher body weight gain and hepatic lipid content. These effects were accompanied by impairment in lipid and glucose metabolism, demonstrated by reduced Cpt1a/Acadvl and increased Agpat/Gpam expression, lower glucose tolerance and insulin sensitivity. Conclusion Our data suggest that both gestational and lactation overnutrition results in metabolic changes that can permanently alter lipid homeostasis in offspring. The presence of fatty acids in maternal blood and milk seem to be responsible for modulating the expression of miR-122 and miR-370, which are involved in liver metabolism. These alterations significantly increase susceptibility to obesity and ectopic lipid accumulation and lead to a more harmful response to HFD in offspring. Electronic supplementary material The online version of this article (doi:10.1186/s12986-017-0168-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Laís Angélica de Paula Simino
- Laboratory of Metabolic Disorders, Faculty of Applied Sciences, University of Campinas -UNICAMP, Limeira, São Paulo Brazil
| | - Thaís de Fante
- Laboratory of Metabolic Disorders, Faculty of Applied Sciences, University of Campinas -UNICAMP, Limeira, São Paulo Brazil
| | - Marina Figueiredo Fontana
- Laboratory of Metabolic Disorders, Faculty of Applied Sciences, University of Campinas -UNICAMP, Limeira, São Paulo Brazil
| | - Fernanda Oliveira Borges
- Laboratory of Metabolic Disorders, Faculty of Applied Sciences, University of Campinas -UNICAMP, Limeira, São Paulo Brazil
| | - Márcio Alberto Torsoni
- Laboratory of Metabolic Disorders, Faculty of Applied Sciences, University of Campinas -UNICAMP, Limeira, São Paulo Brazil
| | - Marciane Milanski
- Laboratory of Metabolic Disorders, Faculty of Applied Sciences, University of Campinas -UNICAMP, Limeira, São Paulo Brazil
| | - Lício Augusto Velloso
- Laboratory of Cell Signaling, Faculty of Medical Sciences, University Of Campinas - UNICAMP, Campinas, São Paulo Brazil
| | - Adriana Souza Torsoni
- Laboratory of Metabolic Disorders, Faculty of Applied Sciences, University of Campinas -UNICAMP, Limeira, São Paulo Brazil
| |
Collapse
|
50
|
Ning LJ, He AY, Lu DL, Li JM, Qiao F, Li DL, Zhang ML, Chen LQ, Du ZY. Nutritional background changes the hypolipidemic effects of fenofibrate in Nile tilapia (Oreochromis niloticus). Sci Rep 2017; 7:41706. [PMID: 28139735 PMCID: PMC5282496 DOI: 10.1038/srep41706] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Accepted: 12/29/2016] [Indexed: 12/15/2022] Open
Abstract
Peroxisome proliferation activated receptor α (PPARα) is an important transcriptional regulator of lipid metabolism and is activated by high-fat diet (HFD) and fibrates in mammals. However, whether nutritional background affects PPARα activation and the hypolipidemic effects of PPARα ligands have not been investigated in fish. In the present two-phase study of Nile tilapia (Oreochromis niloticus), fish were first fed a HFD (13% fat) or low-fat diet (LFD; 1% fat) diet for 10 weeks, and then fish from the first phase were fed the HFD or LFD supplemented with 200 mg/kg body weight fenofibrate for 4 weeks. The results indicated that the HFD did not activate PPARα or other lipid catabolism-related genes. Hepatic fatty acid β-oxidation increased significantly in the HFD and LFD groups after the fenofibrate treatment, when exogenous substrates were sufficiently provided. Only in the HFD group, fenofibrate significantly increased hepatic PPARα mRNA and protein expression, and decreased liver and plasma triglyceride concentrations. This is the first study to show that body fat deposition and dietary lipid content affects PPARα activation and the hypolipidemic effects of fenofibrate in fish, and this could be due to differences in substrate availability for lipid catabolism in fish fed with different diets.
Collapse
Affiliation(s)
- Li-Jun Ning
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, China
| | - An-Yuan He
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, China
| | - Dong-Liang Lu
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, China
| | - Jia-Min Li
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, China
| | - Fang Qiao
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, China
| | - Dong-Liang Li
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, China
| | - Mei-Ling Zhang
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, China
| | - Li-Qiao Chen
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, China
| | - Zhen-Yu Du
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai, China
| |
Collapse
|