1
|
Du Y, Yang Z, Shi H, Chen Z, Chen R, Zhou F, Peng X, Hong T, Jiang L. E3 ubiquitin ligase UBR5 promotes gemcitabine resistance in pancreatic cancer by inducing O-GlcNAcylation-mediated EMT via destabilization of OGA. Cell Death Dis 2024; 15:340. [PMID: 38755129 PMCID: PMC11099055 DOI: 10.1038/s41419-024-06729-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/18/2024]
Abstract
Pancreatic cancer (PC) is among the deadliest malignancies, with an extremely poor diagnosis and prognosis. Gemcitabine (GEM) remains the first-line drug for treating PC; however, only a small percentage of patients benefit from current immunotherapies or targeted therapies. Resistance to GEM is prevalent and affects long-term survival. We found that ubiquitin-protein ligase E3 module N-recognition 5 (UBR5) is a therapeutic target against GEM resistance. UBR5 was markedly upregulated in clinical GEM-resistant PC samples and GEM-resistant PC cells. UBR5 knockdown markedly increased GEM sensitivity in GEM-resistant PC cell lines. UBR5-mediated GEM resistance was accompanied by activation of epithelial-mesenchymal transition (EMT) and could be mitigated by inhibiting EMT. Further analysis revealed that UBR5 promoted GEM resistance in PC cells by enhancing O-GlcNAcylation-mediated EMT. In addition, UBR5 knockdown resulted in increased O-GlcNAase (OGA) levels, an essential negatively regulated enzyme in the O-GlcNAcylation process. We identified a negative association between OGA and UBR5 levels, which further supported the hypothesis that O-GlcNAcylation-mediated GEM resistance induced by UBR5 is OGA-dependent in PC cells. Mechanistic studies revealed that UBR5 acts as an E3 ubiquitin ligase of OGA and regulates O-GlcNAcylation by binding and modulating OGA, facilitating its degradation and ubiquitination. Additionally, high-throughput compound library screening using three-dimensional protein structure analysis and drug screening identified a Food and Drug Administration drug, Y-39983 dihydrochloride, as a potent GEM sensitiser and UBR5 inhibitor. The combination of Y-39983 dihydrochloride and GEM attenuated tumour growth in a mouse xenograft tumour model. Collectively, these data demonstrated that UBR5 plays a pivotal role in the sensitisation of PC to GEM and provides a potential therapeutic strategy to overcome GEM resistance.
Collapse
Affiliation(s)
- Yunyan Du
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Key Laboratory of Drug Targets and Drug Screening of Jiangxi Province, Nanchang University, Nanchang, 330006, China
| | - Zhangjian Yang
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Key Laboratory of Drug Targets and Drug Screening of Jiangxi Province, Nanchang University, Nanchang, 330006, China
| | - Hao Shi
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Key Laboratory of Drug Targets and Drug Screening of Jiangxi Province, Nanchang University, Nanchang, 330006, China
| | - Zhihan Chen
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Key Laboratory of Drug Targets and Drug Screening of Jiangxi Province, Nanchang University, Nanchang, 330006, China
| | - Rong Chen
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Key Laboratory of Drug Targets and Drug Screening of Jiangxi Province, Nanchang University, Nanchang, 330006, China
| | - Fan Zhou
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Xiaogang Peng
- Jiangxi Province Key Laboratory of Molecular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330000, China
| | - Tao Hong
- Key Laboratory of Drug Targets and Drug Screening of Jiangxi Province, Nanchang University, Nanchang, 330006, China.
- Department of Neurosurgery, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China.
| | - Liping Jiang
- School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
- Key Laboratory of Drug Targets and Drug Screening of Jiangxi Province, Nanchang University, Nanchang, 330006, China.
| |
Collapse
|
2
|
Schafte K, Bruna S. The influence of intergenerational trauma on epigenetics and obesity in Indigenous populations - a scoping review. Epigenetics 2023; 18:2260218. [PMID: 37752750 PMCID: PMC10538456 DOI: 10.1080/15592294.2023.2260218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 09/07/2023] [Indexed: 09/28/2023] Open
Abstract
Background: Research has recently begun to examine the potential intergenerational impacts of trauma on obesity.Objective: This scoping review examines the literature on the interactions between intergenerational trauma, epigenetics, and obesity in Indigenous populations. The review was conducted to identify what is known from the literature about how intergenerational trauma may epigenetically influence obesity in Indigenous populations.Methods: Following the PRISMA-ScR guidelines for scoping reviews, online databases were used to identify studies that included discussion of the four focus topics: trauma, epigenetics, obesity, and Indigeneity. The review resulted in six studies that examined those themes. The focus and findings of the selected studies varied from cultural to biological mechanisms and from discussion regarding trauma, epigenetics, obesity, or Indigeneity, but they support three broad statements. First, they support that obesity has genetic and epigenetic factors. Second, intergenerational trauma is prevalent in Indigenous communities. Finally, intergenerational trauma has cultural and biological influences on obesity.Conclusions: Current literature illustrates that intergenerational trauma has behavioural and epigenetic influences that can lead to increased obesity. This scoping review provides a preliminary map of the current literature and understandings of these topics. This review calls for continued studies regarding the connection between trauma, obesity, and epigenetics in Indigenous communities. Future research is vital for practice and policy surrounding individual and communal healing.
Collapse
Affiliation(s)
- Krista Schafte
- Department of Anthropology, Western Washington University, Bellingham, WA, USA
| | - Sean Bruna
- Department of Anthropology, Western Washington University, Bellingham, WA, USA
| |
Collapse
|
3
|
ÇOBANOĞULLARI HAVVA, ERGOREN MAHMUTCERKEZ, DUNDAR MUNIS, BERTELLI MATTEO, TULAY PINAR. Periconceptional Mediterranean diet during pregnancy on children's health. JOURNAL OF PREVENTIVE MEDICINE AND HYGIENE 2022; 63:E65-E73. [PMID: 36479491 PMCID: PMC9710394 DOI: 10.15167/2421-4248/jpmh2022.63.2s3.2748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
During pregnancy, rapid and subtle physiological changes are observed from conception to birth. Nutrition and other lifestyle factors before and during pregnancy have been shown in the literature to influence the health of both mother and child. A healthy and varied diet during pregnancy can provide adequate energy and nutrients for both the mother and the growing fetus. Current research focuses on the periconceptional phase, which includes the early processes of gametogenesis, embryogenesis and placentation. A variety of abnormalities and pregnancy-related problems occur during this period, including congenital defects, fetal loss, miscarriage and preterm birth. A varied and balanced diet during periconception is important to maintain fetal development and growth. To date, numerous studies have been conducted to investigate the effects of consuming different nutrients, foods or food groups during pregnancy on the health of mother and child. For example, the Mediterranean diet is considered as a balanced, nutrient-rich diet due to the low consumption of meat products and fatty foods and the high consumption of vegetables, cheese, olive oil, fish, shellfish and little meat. While many studies have been conducted in the literature to investigate the effects of a Mediterranean diet during pregnancy on fetal health, the results have been inconclusive. The aim of this article is to review the current literature on the Mediterranean diet during pregnancy.
Collapse
Affiliation(s)
- HAVVA ÇOBANOĞULLARI
- Near East University, Faculty of Medicine, Department of Molecular Medicine, Nicosia, Cyprus
| | - MAHMUT CERKEZ ERGOREN
- Near East University, Faculty of Medicine, Department of Medical Genetics, Nicosia, Cyprus
| | - MUNIS DUNDAR
- Erciyes University, Faculty of Medicine, Department of Medical Genetics, Kayseri, Turkey
| | - MATTEO BERTELLI
- MAGISNAT, Peachtree Corners, USA
- MAGI Euregio, Bolzano, Italy
- MAGI’S LAB, Rovereto, Italy
| | - PINAR TULAY
- Near East University, Faculty of Medicine, Department of Medical Genetics, Nicosia, Cyprus
- Near East University, DESAM Research Institute, Nicosia, Cyprus
- Correspondence: Pınar Tulay, Near East University, Faculty of Medicine, Department of Medical Genetics, 99138 Nicosia, Cyprus. E-mail:
| |
Collapse
|
4
|
Blakstad MM, Fawzi WW, Castro MC, Thompson A, Arabi M, Danaei G. Scaling up prenatal nutrition could reduce the global burden of noncommunicable diseases in the next generation: a modeling analysis. Am J Clin Nutr 2022; 116:1291-1302. [PMID: 36192638 PMCID: PMC9630869 DOI: 10.1093/ajcn/nqac214] [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: 12/08/2021] [Revised: 05/05/2022] [Accepted: 08/11/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Nutritional conditions during pregnancy may influence the epigenetic development of an individual and consequently their later-life risk of noncommunicable disease (NCD). Improving nutrition for pregnant females may therefore serve the dual purpose of directly improving pregnancy outcomes and preventing NCDs in the next generation. OBJECTIVES We estimated the impact of prenatal supplementation with iron and folic acid (IFA), multiple micronutrients (MMS), or calcium at 50%, 75%, or 90% coverage on future NCDs by age and sex in 2015. METHODS We used secondary data sources from 132 countries to quantify the cases of diabetes and hypertension and the deaths from selected NCDs that could be averted or delayed by scaling up prenatal micronutrient supplementation. RESULTS Globally, >51,000 NCD deaths, 6 million cases of hypertension, and 3 million cases of diabetes could be prevented per offspring birth cohort if mothers were prenatally supplemented with MMS at 90% coverage. For IFA these numbers would be roughly half. Calcium supplementation at 90% could delay 51,000 deaths per birth cohort. Our model suggests that substantial numbers of NCD deaths and cases of hypertension and diabetes could be prevented in future generations by scaling up micronutrient supplementation for mothers during pregnancy. CONCLUSIONS Highlighting the additional benefits of proven nutrition interventions is critical in ensuring adequate and sustained investments, and programmatic integration. As the double burden of disease continues to grow, population-wide efforts to scale up micronutrient supplementation to pregnant females could help prevent both undernutrition and chronic disease.
Collapse
Affiliation(s)
| | - Wafaie W Fawzi
- Department of Global Health and Population, Harvard TH Chan School of Public Health, Boston, MA, USA,Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA,Department of Nutrition, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Marcia C Castro
- Department of Global Health and Population, Harvard TH Chan School of Public Health, Boston, MA, USA
| | | | | | - Goodarz Danaei
- Department of Global Health and Population, Harvard TH Chan School of Public Health, Boston, MA, USA,Department of Epidemiology, Harvard TH Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
5
|
Paula VG, Souza MRD, Sinzato YK, Villaverde AISB, Corrente JE, Volpato GT, Damasceno DC. Nonpregnant and pregnant adult female rats affected by maternal diabetes environment. Syst Biol Reprod Med 2022; 68:384-395. [DOI: 10.1080/19396368.2022.2115326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Affiliation(s)
- Verônyca Gonçalves Paula
- Laboratory of Experimental Research on Gynecology and Obstetrics, Post Graduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo State, Brazil
| | - Maysa Rocha de Souza
- Laboratory of Experimental Research on Gynecology and Obstetrics, Post Graduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo State, Brazil
| | - Yuri Karen Sinzato
- Laboratory of Experimental Research on Gynecology and Obstetrics, Post Graduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo State, Brazil
| | - Ana Izabel Silva Balbin Villaverde
- Laboratory of Experimental Research on Gynecology and Obstetrics, Post Graduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo State, Brazil
| | - José Eduardo Corrente
- Research Support Office, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo State, Brazil
| | - Gustavo Tadeu Volpato
- Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso State, Brazil
| | - Débora Cristina Damasceno
- Laboratory of Experimental Research on Gynecology and Obstetrics, Post Graduate Course on Tocogynecology, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, São Paulo State, Brazil
| |
Collapse
|
6
|
G N, Zilbauer M. Epigenetics in IBD: a conceptual framework for disease pathogenesis. Frontline Gastroenterol 2022; 13:e22-e27. [PMID: 35812027 PMCID: PMC9234725 DOI: 10.1136/flgastro-2022-102120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 05/16/2022] [Indexed: 02/04/2023] Open
Abstract
The global incidence and prevalence of paediatric inflammatory bowel disease (IBD) is increasing, with a notable emergence in developing countries with historically low rates. This suggests that environmental and epigenetic factors may play an important role in the pathogenesis and progression of IBD. Epigenetics refers to the study of biological mechanisms that result in a change of phenotype, without an change in the underlying DNA sequence. Epigenetic mechanisms drive many biological processes that occur in health, such as development and ageing, and are also implicated in disease, including cancer and other inflammatory diseases. Importantly, identification of cell-type-specific epigenetic mechanisms could lead to the identification of molecular disease subtypes allowing a personalised treatment approach. In this short review, we provide a summary of epigenetic mechanisms operative in mammals, and their potential involvement in IBD pathogenesis. Furthermore, we discuss key challenges associated with investigating epigenetics in IBD and provide potential strategies to overcome these, such as through the use of 'omics' and organoid technologies.
Collapse
Affiliation(s)
- Natasha G
- Paediatrics, Cambridge University, Cambridge, UK
| | | |
Collapse
|
7
|
Guth S, McGinnis E, Copeland W, Hudziak J. A Pilot Trial of a Health Promotion and Illness Prevention Paradigm in the Perinatal Period. Matern Child Health J 2022; 26:1203-1210. [PMID: 35064428 PMCID: PMC9762324 DOI: 10.1007/s10995-021-03354-6] [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] [Accepted: 12/09/2021] [Indexed: 10/19/2022]
Abstract
OBJECTIVES This is a pilot study of the Vermont Family Based Approach, an innovative health promotion program designed to address behavioral health prevention in primary care, adapted for perinatal women. We aimed to determine the acceptability of the intervention across socioeconomic strata, and to identify if participation improves perinatal mental health. METHODS Recruitment occurred at a general obstetrics practice. Women 12-25 weeks gestation were paired with a wellness coach who administered a wellness assessment and used motivational interviewing to facilitate individualized plans based on evidence-based domains of health promotion. Participants were offered access to free, co-located wellness activities through the peripartum, and referred to behavioral health services if appropriate. RESULTS 93 women consented; 16 Medicaid Insured women (MI) and 30 Privately Insured (PI) were randomized to the intervention. Of all activities, yoga and parenting activities were most appealing, with 58% of women attending. PI (M = 12.30, SD = 11.71) attended significantly more activities than MI (M = 3.81, SD = 12.30; p = .001). Trauma exposure was inversely associated with attendance (p = .004). Randomization to the intervention was not associated with improvements in internalizing symptoms or perceived stress at 12 months postpartum, however, attending three or more wellness activities was associated with a decrease in perceived stress between baseline and 12 months postpartum. CONCLUSION This program appeared acceptable and engaging to women with private insurance, but less so with Medicaid. The trial failed to demonstrate improvement in internalizing scores, but of those randomized to the intervention, higher engagement was associated with decreased stress one year after giving birth.
Collapse
Affiliation(s)
- Sarah Guth
- Vermont Center for Children, Youth and Families in the Psychiatry Department at the University of Vermont, Robert Larner College of Medicine, Burlington, VT, USA.
| | - Ellen McGinnis
- Vermont Center for Children, Youth and Families in the Psychiatry Department at the University of Vermont, Robert Larner College of Medicine, Burlington, VT, USA
| | - William Copeland
- Vermont Center for Children, Youth and Families in the Psychiatry Department at the University of Vermont, Robert Larner College of Medicine, Burlington, VT, USA
| | - James Hudziak
- Vermont Center for Children, Youth and Families in the Psychiatry Department at the University of Vermont, Robert Larner College of Medicine, Burlington, VT, USA
| |
Collapse
|
8
|
Epigenetic Effect of Maternal Methyl-Group Donor Intake on Offspring’s Health and Disease. Life (Basel) 2022; 12:life12050609. [PMID: 35629277 PMCID: PMC9145757 DOI: 10.3390/life12050609] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/02/2022] [Accepted: 04/15/2022] [Indexed: 01/16/2023] Open
Abstract
Maternal exposure to some dietary and environmental factors during embryonic development can affect offspring’s phenotype and, furthermore, the risk of developing diseases later in life. One potential mechanism responsible for this early programming may be the modification of the epigenome, such as DNA methylation. Methyl-group donors are essential for DNA methylation and are shown to have an important role in fetal development and later health. The main goal of the present review is to summarize the available literature data on the epigenetic effect (DNA methylation) of maternal methyl-group donor availability on reproductivity, perinatal outcome, and later health of the offspring. In our literature search, we found evidence for the association between alterations in DNA methylation patterns caused by different maternal methyl-group donor (folate, choline, methionine, betaine) intake and reproductivity, birth weight, neural tube defect, congenital heart defect, cleft lip and palate, brain development, and the development of obesity and associated non-communicable diseases in later life. We can conclude that maternal methyl-group donor availability could affect offspring’s health via alterations in DNA methylation and may be a major link between early environmental exposure and the development of diseases in the offspring. However, still, further studies are necessary to confirm the associations and causal relationships.
Collapse
|
9
|
Xu J, Xu HM, Yang MF, Liang YJ, Peng QZ, Zhang Y, Tian CM, Wang LS, Yao J, Nie YQ, Li DF. New Insights Into the Epigenetic Regulation of Inflammatory Bowel Disease. Front Pharmacol 2022; 13:813659. [PMID: 35173618 PMCID: PMC8841592 DOI: 10.3389/fphar.2022.813659] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/06/2022] [Indexed: 01/10/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disease of the colonic mucosa. Environmental factors, genetics, intestinal microbiota, and the immune system are all involved in the pathophysiology of IBD. Lately, accumulating evidence has shown that abnormal epigenetic changes in DNA methylation, histone markers, and non-coding RNA expression greatly contribute to the development of the entire disease. Epigenetics regulates many functions, such as maintaining the homeostasis of the intestinal epithelium and regulating the immune system of the immune cells. In the present study, we systematically summarized the latest advances in epigenetic modification of IBD and how epigenetics reveals new mechanisms of IBD. Our present review provided new insights into the pathophysiology of IBD. Moreover, exploring the patterns of DNA methylation and histone modification through epigenetics can not only be used as biomarkers of IBD but also as a new target for therapeutic intervention in IBD patients.
Collapse
Affiliation(s)
- Jing Xu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Hao-ming Xu
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Mei-feng Yang
- Department of Hematology, Yantian District People’s Hospital, Shenzhen, China
| | | | - Quan-zhou Peng
- Department of Pathology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Yuan Zhang
- Department of Medical Administration, Huizhou Institute of Occupational Diseases Control and Prevention, Huizhou, China
| | - Cheng-mei Tian
- Department of Emergency, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Li-sheng Wang
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Jun Yao
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Yu-qiang Nie
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - De-feng Li
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| |
Collapse
|
10
|
Zhang S, Ou K, Huang J, Fang L, Wang C, Wang Q. Prenatal EGCG exposure-induced heart mass reduction in adult male mice and underlying mechanisms. Food Chem Toxicol 2021; 157:112588. [PMID: 34600025 DOI: 10.1016/j.fct.2021.112588] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/24/2021] [Accepted: 09/28/2021] [Indexed: 12/17/2022]
Abstract
Epigallocatechin-3-gallate (EGCG), which is a major polyphenol in tea, has an unclear effect on cardiac development. In the present study, mice (C57BL/6) were exposed in utero to EGCG dissolved in drinking water (3 μg/ml) for 16 days. A significant decrease in the heart/body weight ratio was observed in adult males but not in adult females. The protein expression levels of TGF-β1 and its downstream transcription factors SMAD3 and SMAD4 were significantly decreased in male hearts. The PI3K/AKT signaling pathway was inhibited, the expression of proapoptotic proteins, such as BAX, Cleaved Caspase3 and Cleaved Caspase9, was elevated, and the level of antiapoptotic proteins, such as BCL-2, was decreased. A reduced heart/body weight ratio may be associated with the loss of cardiac fibers and an increase in myocardial apoptosis. The cardiac levels of aromatic hydrocarbon receptor and androgen receptor were elevated only in males, which may explain the sexual dimorphism in the effects. The promoter methylation levels of pik3r1, tgf-β, smad4 were elevated, and those of ahr were reduced, explaining the mechanism underlying the cardiac histological alteration caused by prenatal exposure to EGCG. The results suggest that ingestion of EGCG during pregnancy may be a risk factor for cardiac development in offspring.
Collapse
Affiliation(s)
- Shenli Zhang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Kunlin Ou
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Jie Huang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Lu Fang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Chonggang Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China
| | - Qin Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, PR China.
| |
Collapse
|
11
|
De Smidt JJA, Odendaal HJ, Nel DG, Nolan H, Du Plessis C, Brink LT, Oelofse A. The effects of in utero exposure to teratogens on organ size: a prospective paediatric study. J Dev Orig Health Dis 2021; 12:748-757. [PMID: 33198841 PMCID: PMC8536468 DOI: 10.1017/s2040174420001002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
In low-income countries, prospective data on combined effects of in utero teratogen exposure are lacking and necessitates new research. The aim of the present study was to explore the effect of in utero teratogen exposure on the size of the kidneys and pancreas 5 years after birth in a low-income paediatric population. Data was collected from 500 mother-child pairs from a low-income setting. Anthropometric measurements included body weight, (BW) body height, mid-upper arm and waist circumference (WC). Clinical measurements included blood pressure (BP), mean arterial pressure and heart rate. Ultrasound measurements included pancreas, and kidney measurements at age 5 years. The main outcome of interest was the effect of maternal smoking and alcohol consumption on ultrasound measurements of organ size at age 5 years. Left and right kidney length measurements were significantly lower in smoking exposed children compared to controls (p = 0.04 and p = 0.03). Pancreas body measurements were significantly lower in smoking exposed children (p = 0.04). Multiple regression analyses were used to examine the associations between the independent variables (IDVs), maternal age, body mass index (BMI), mid-upper arm circumference (MUAC) and BW of the child, on the dependent variables (DVs) kidney lengths and kidney volumes. Also, the association between in utero exposure to alcohol and nicotine and pancreas size. WC was strongest (r = 0.28; p < 0.01) associated with pancreas head [F (4, 454) = 13.44; R2 = 0.11; p < 0.01] and tail (r = 0.30; p < 0.01) measurements at age 5 years, with in utero exposure, sex of the child and BMI as covariates. Kidney length and pancreas body measurements are affected by in utero exposure to nicotine at age 5 years and might contribute to cardiometabolic risk in later life. Also, findings from this study report on ultrasound reference values for kidney and pancreas measurements of children at age 5 years from a low-income setting.
Collapse
Affiliation(s)
- J. J. A. De Smidt
- Department of Medical Biosciences, University of the Western Cape, Cape Town, South Africa
| | - H. J. Odendaal
- Department of Obstetrics and Gynaecology, Stellenbosch University, Stellenbosch, South Africa
| | - D. G. Nel
- Department of Statistics and Actuarial Science, Stellenbosch University, Stellenbosch, South Africa
| | - H. Nolan
- Department of Obstetrics and Gynaecology, Stellenbosch University, Stellenbosch, South Africa
| | - C. Du Plessis
- Department of Obstetrics and Gynaecology, Stellenbosch University, Stellenbosch, South Africa
| | - L. T. Brink
- Department of Obstetrics and Gynaecology, Stellenbosch University, Stellenbosch, South Africa
| | - A. Oelofse
- Department of Medical Biosciences, University of the Western Cape, Cape Town, South Africa
| |
Collapse
|
12
|
Rivera P, Guerra-Cantera S, Vargas A, Díaz F, García-Úbeda R, Tovar R, Ramírez-López MT, Argente J, de Fonseca FR, Suárez J, Chowen JA. Maternal hypercaloric diet affects factors involved in lipid metabolism and the endogenous cannabinoid systems in the hypothalamus of adult offspring: sex-specific response of astrocytes to palmitic acid and anandamide. Nutr Neurosci 2020; 25:931-944. [PMID: 32954972 DOI: 10.1080/1028415x.2020.1821519] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Aim: We aimed to investigate whether maternal malnutrition during gestation/lactation induces long-lasting changes on inflammation, lipid metabolism and endocannabinoid signaling in the adult offspring hypothalamus and the role of hypothalamic astrocytes in these changes.Methods: We analyzed the effects of a free-choice hypercaloric palatable diet (P) during (pre)gestation, lactation and/or post-weaning on inflammation, lipid metabolism and endogenous cannabinoid signaling in the adult offspring hypothalamus. We also evaluated the response of primary hypothalamic astrocytes to palmitic acid and anandamide.Results: Postnatal exposure to a P diet induced factors involved in hypothalamic inflammation (Tnfa and Il6) and gliosis (Gfap, vimentin and Iba1) in adult offspring, being more significant in females. In contrast, maternal P diet reduced factors involved in astrogliosis (vimentin), fatty acid oxidation (Cpt1a) and monounsaturated fatty acid synthesis (Scd1). These changes were accompanied by an increase in the expression of the genes for the cannabinoid receptor (Cnr1) and Nape-pld, an enzyme involved in endocannabinoid synthesis, in females and a decrease in the endocannabinoid degradation enzyme Faah in males. These changes suggest that the maternal P diet results in sex-specific alterations in hypothalamic endocannabinoid signaling and lipid metabolism. This hypothesis was tested in hypothalamic astrocyte cultures, where palmitic acid (PA) and the polyunsaturated fatty acid N-arachidonoylethanolamine (anandamide or AEA) were found to induce similar changes in the endocannabinoid system (ECS) and lipid metabolism.Conclusion: These results stress the importance of both maternal diet and sex in long term metabolic programming and suggest a possible role of hypothalamic astrocytes in this process.
Collapse
Affiliation(s)
- Patricia Rivera
- Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain
| | - Santiago Guerra-Cantera
- Department of Endocrinology, Fundación Investigación Biomédica del Hospital Infantil Universitario Niño Jesús, Instituto de Investigación Biomédica la Princesa, Madrid, Spain.,CIBEROBN (Centro de Investigación Biomédica en Red sobre Fisiopatología de la Obesidad y Nutrición), Instituto de Salud Carlos III, Madrid, Spain
| | - Antonio Vargas
- Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain
| | - Francisca Díaz
- Department of Endocrinology, Fundación Investigación Biomédica del Hospital Infantil Universitario Niño Jesús, Instituto de Investigación Biomédica la Princesa, Madrid, Spain.,CIBEROBN (Centro de Investigación Biomédica en Red sobre Fisiopatología de la Obesidad y Nutrición), Instituto de Salud Carlos III, Madrid, Spain
| | - Rocío García-Úbeda
- Department of Endocrinology, Fundación Investigación Biomédica del Hospital Infantil Universitario Niño Jesús, Instituto de Investigación Biomédica la Princesa, Madrid, Spain
| | - Rubén Tovar
- Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain
| | | | - Jesús Argente
- Department of Endocrinology, Fundación Investigación Biomédica del Hospital Infantil Universitario Niño Jesús, Instituto de Investigación Biomédica la Princesa, Madrid, Spain.,CIBEROBN (Centro de Investigación Biomédica en Red sobre Fisiopatología de la Obesidad y Nutrición), Instituto de Salud Carlos III, Madrid, Spain.,IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain.,Department of Pediatrics, Universidad Autonóma de Madrid, Madrid, Spain
| | - Fernando Rodríguez de Fonseca
- Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain
| | - Juan Suárez
- Hospital Regional Universitario de Málaga, Instituto de Investigación Biomédica de Málaga (IBIMA), Universidad de Málaga, Málaga, Spain
| | - Julie A Chowen
- Department of Endocrinology, Fundación Investigación Biomédica del Hospital Infantil Universitario Niño Jesús, Instituto de Investigación Biomédica la Princesa, Madrid, Spain.,CIBEROBN (Centro de Investigación Biomédica en Red sobre Fisiopatología de la Obesidad y Nutrición), Instituto de Salud Carlos III, Madrid, Spain.,IMDEA Food Institute, CEI UAM + CSIC, Madrid, Spain
| |
Collapse
|
13
|
He S, Stein AD. Early-Life Nutrition Interventions and Associated Long-Term Cardiometabolic Outcomes: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Adv Nutr 2020; 12:461-489. [PMID: 33786595 PMCID: PMC8009753 DOI: 10.1093/advances/nmaa107] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 06/08/2020] [Accepted: 08/10/2020] [Indexed: 12/14/2022] Open
Abstract
Early-life nutrition interventions can have lifelong cardiometabolic benefits. Most evidence on this topic is derived from observational studies. We evaluated the association of randomized controlled nutritional trials in early life and long-term cardiometabolic outcomes. Through literature search of PubMed, CABI Global Health, Embase, and Cochrane, with manual reference check and weekly alert from PubMed, we identified 8312 records, and included 53 records from 40 cohorts in 21 countries. The total number of participants was 33,551. Interventions were initiated as early as conception, and the longest until 7 y (except 1 study from infancy to 20 y). The cohorts were followed up for between 3 and 73 y. We identified 7 types of interventions (protein-energy supplements, long-chain PUFAs, single micronutrient, multiple micronutrients, infant and young child feeding, dietary counseling, and other) and 4 categories of cardiometabolic outcomes (biomarkers, cardiovascular, body size and composition, and subclinical/clinical outcomes). Most findings were null. Fasting glucose concentration was 0.04 mmol/L lower (95% CI: -0.05, -0.02 mmol/L; I2 = 0%) in the intervention groups than in the control groups (15 studies). BMI (kg/m2) was 0.20 higher (95% CI: 0.12, 0.28; I2 = 54%) in the intervention groups than control groups (14 studies). No significant effect was observed for total cholesterol (12 studies) or blood pressure (17 studies). Ongoing and personalized dietary counseling was associated with lower glucose and cholesterol, better endothelial function, and reduced risk of metabolic syndrome. The timing of intervention mattered, with earlier initiation conferring greater benefit (improved lipid profile and marginally lower glucose concentration) based on 2 studies. In sum, glucose concentration was lower following early-life nutrition interventions, but there is a risk of unintended consequences, including higher BMI. Maternal and child nutrition interventions must be evidence-based and tailored to each population to promote long-term cardiometabolic health.
Collapse
Affiliation(s)
- Siran He
- Nutrition and Health Sciences Program, Laney Graduate School, Emory University, Atlanta, GA, USA
| | | |
Collapse
|
14
|
Family health competence: Attachment, detachment and health practices in the early years of parenthood. Soc Sci Med 2020; 266:113351. [PMID: 32932003 DOI: 10.1016/j.socscimed.2020.113351] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Revised: 08/21/2020] [Accepted: 09/02/2020] [Indexed: 11/24/2022]
Abstract
During the first years of a baby's life, parents develop ways of caring that affect the child's health later in life. In this paper, we focus on eating and sleeping, as social practices that mediate between socioeconomic and cultural conditions and health outcomes, such as weight status. We argue for an analysis of what we call 'family health competence', meaning emerging know-hows and resources relevant to healthy living produced, embodied and shared by household members, to understand the development of health practices of first-time parents and their children. In an ethnographic panel study in the Netherlands, we follow households pre-birth until the first child turns age four. Our analysis suggests that across different families, competences develop enabling parents to balance a) attaching and b) detaching in particular ways. Parents learn how to observe and interpret their new-borns, bracket doubt, build trust, manage time pressures and mobilize support networks. These competences are partly class and gender-specific while there is also significant diversity within class and gender. The competence to balance attachment and detachment can be understood as the effect of contradictory social norms and institutional (labour market and care) provisions typical for late-modern welfare states.
Collapse
|
15
|
Science and Healthy Meals in the World: Nutritional Epigenomics and Nutrigenetics of the Mediterranean Diet. Nutrients 2020; 12:nu12061748. [PMID: 32545252 PMCID: PMC7353392 DOI: 10.3390/nu12061748] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 06/07/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023] Open
Abstract
The Mediterranean Diet (MD), UNESCO Intangible Cultural Heritage of Humanity, has become a scientific topic of high interest due to its health benefits. The aim of this review is to pick up selected studies that report nutrigenomic or nutrigenetic data and recapitulate some of the biochemical/genomic/genetic aspects involved in the positive health effects of the MD. These include (i) the antioxidative potential of its constituents with protective effects against several diseases; (ii) the epigenetic and epigenomic effects exerted by food components, such as Indacaxanthin, Sulforaphane, and 3-Hydroxytyrosol among others, and their involvement in the modulation of miRNA expression; (iii) the existence of predisposing or protective human genotypes due to allelic diversities and the impact of the MD on disease risk. A part of the review is dedicated to the nutrigenomic effects of the main cooking methods used in the MD and also to a comparative analysis of the nutrigenomic properties of the MD and other diet regimens and non-MD-related aliments. Taking all the data into account, the traditional MD emerges as a diet with a high antioxidant and nutrigenomic modulation power, which is an example of the “Environment-Livings-Environment” relationship and an excellent patchwork of interconnected biological actions working toward human health.
Collapse
|
16
|
The Effects of Maternal and Postnatal Dietary Methyl Nutrients on Epigenetic Changes that Lead to Non-Communicable Diseases in Adulthood. Int J Mol Sci 2020; 21:ijms21093290. [PMID: 32384688 PMCID: PMC7246552 DOI: 10.3390/ijms21093290] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 04/30/2020] [Accepted: 05/01/2020] [Indexed: 12/13/2022] Open
Abstract
The risk for non-communicable diseases in adulthood can be programmed by early nutrition. This programming is mediated by changes in expression of key genes in various metabolic pathways during development, which persist into adulthood. These developmental modifications of genes are due to epigenetic alterations in DNA methylation patterns. Recent studies have demonstrated that DNA methylation can be affected by maternal or early postnatal diets. Because methyl groups for methylation reactions come from methionine cycle nutrients (i.e., methionine, choline, betaine, folate), deficiency or supplementation of these methyl nutrients can directly change epigenetic regulation of genes permanently. Although many studies have described the early programming of adult diseases by maternal and infant nutrition, this review discusses studies that have associated early dietary methyl nutrient manipulation with direct effects on epigenetic patterns that could lead to chronic diseases in adulthood. The maternal supply of methyl nutrients during gestation and lactation can alter epigenetics, but programming effects vary depending on the timing of dietary intervention, the type of methyl nutrient manipulated, and the tissue responsible for the phenotype. Moreover, the postnatal manipulation of methyl nutrients can program epigenetics, but more research is needed on whether this approach can rescue maternally programmed offspring.
Collapse
|
17
|
Stephen BJ, Pareek N, Saeed M, Kausar MA, Rahman S, Datta M. Xeno-miRNA in Maternal-Infant Immune Crosstalk: An Aid to Disease Alleviation. Front Immunol 2020; 11:404. [PMID: 32269563 PMCID: PMC7109445 DOI: 10.3389/fimmu.2020.00404] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 02/20/2020] [Indexed: 12/12/2022] Open
Abstract
Human milk is a complex liquid that contains multifaceted compounds which provide nutrition to infants and helps to develop their immune system. The presence of secretory immunoglobulins (IgA), leucocytes, lysozyme, lactoferrin, etc., in breast milk and their role in imparting passive immunity to infants as well as modulating development of an infant's immune system is well-established. Breast milk miRNAs (microRNAs) have been found to be differentially expressed in diverse tissues and biological processes during various molecular functions. Lactation is reported to assist mothers and their offspring to adapt to an ever-changing food supply. It has been observed that certain subtypes of miRNAs exist that are codified by non-human genomes but are still present in circulation. They have been termed as xeno-miRNA (XenomiRs). XenomiRs in humans have been found from various exogenous sources. Route of entry in human systems have been mainly dietary. The possibility of miRNAs taken up into mammalian circulation through diet, and thereby effecting gene expression, is a distinct possibility. This mechanism suggests an interesting possibility that dietary foods may modulate the immune strength of infants via highly specific post-transcriptional regulatory information present in mother's milk. This serves as a major breakthrough in understanding the fundamentals of nutrition and cross-organism communication. In this review, we elaborate and understand the complex crosstalk of XenomiRs present in mother's milk and their plausible role in modulating the infant immune system against infectious and inflammatory diseases.
Collapse
Affiliation(s)
| | - Nidhi Pareek
- Department of Microbiology, School of Life Sciences, Central University of Rajasthan, Ajmer, India
| | - Mohd Saeed
- Department of Biology, College of Sciences, University of Ha'il, Ha'il, Saudi Arabia
| | - Mohd Adnan Kausar
- Department of Biochemistry, College of Medicines, University of Ha'il, Ha'il, Saudi Arabia
| | - Safikur Rahman
- Department of Botany, Munshi Singh College, Babasaheb Bhimrao Ambedkar Bihar University, Muzaffarpur, India
| | - Manali Datta
- Amity Institute of Biotechnology, Amity University Rajasthan, Jaipur, India
| |
Collapse
|
18
|
Blin G, Liand M, Mauduit C, Chehade H, Benahmed M, Simeoni U, Siddeek B. Maternal Exposure to High-Fat Diet Induces Long-Term Derepressive Chromatin Marks in the Heart. Nutrients 2020; 12:E181. [PMID: 31936461 PMCID: PMC7019950 DOI: 10.3390/nu12010181] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/30/2019] [Accepted: 01/07/2020] [Indexed: 02/07/2023] Open
Abstract
Heart diseases are a leading cause of death. While the link between early exposure to nutritional excess and heart disease risk is clear, the molecular mechanisms involved are poorly understood. In the developmental programming field, increasing evidence is pointing out the critical role of epigenetic mechanisms. Among them, polycomb repressive complex 2 (PRC2) and DNA methylation play a critical role in heart development and pathogenesis. In this context, we aimed at evaluating the role of these epigenetic marks in the long-term cardiac alterations induced by early dietary challenge. Using a model of rats exposed to maternal high-fat diet during gestation and lactation, we evaluated cardiac alterations at adulthood. Expression levels of PRC2 components, its histone marks di- and trimethylated histone H3 (H3K27me2/3), associated histone mark (ubiquitinated histone H2A, H2AK119ub1) and target genes were measured by Western blot. Global DNA methylation level and DNA methyl transferase 3B (DNMT3B) protein levels were measured. Maternal high-fat diet decreased H3K27me3, H2Ak119ub1 and DNA methylation levels, down-regulated the enhancer of zeste homolog 2 (EZH2), and DNMT3B expression. The levels of the target genes, isl lim homeobox 1 (Isl1), six homeobox 1 (Six1) and mads box transcription enhancer factor 2, polypeptide C (Mef2c), involved in cardiac pathogenesis were up regulated. Overall, our data suggest that the programming of cardiac alterations by maternal exposure to high-fat diet involves the derepression of pro-fibrotic and pro-hypertrophic genes through the induction of EZH2 and DNMT3B deficiency.
Collapse
Affiliation(s)
- Guillaume Blin
- Woman-Mother-Child Department, Division of Pediatrics, DOHaD Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Rue du Bugnon 27, 1011 Lausanne, Switzerland; (G.B.); (M.L.); (H.C.); (U.S.)
| | - Marjorie Liand
- Woman-Mother-Child Department, Division of Pediatrics, DOHaD Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Rue du Bugnon 27, 1011 Lausanne, Switzerland; (G.B.); (M.L.); (H.C.); (U.S.)
| | - Claire Mauduit
- INSERM U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Team 5, 06204 Nice, France; (C.M.); (M.B.)
| | - Hassib Chehade
- Woman-Mother-Child Department, Division of Pediatrics, DOHaD Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Rue du Bugnon 27, 1011 Lausanne, Switzerland; (G.B.); (M.L.); (H.C.); (U.S.)
| | - Mohamed Benahmed
- INSERM U1065, Centre Méditerranéen de Médecine Moléculaire (C3M), Team 5, 06204 Nice, France; (C.M.); (M.B.)
| | - Umberto Simeoni
- Woman-Mother-Child Department, Division of Pediatrics, DOHaD Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Rue du Bugnon 27, 1011 Lausanne, Switzerland; (G.B.); (M.L.); (H.C.); (U.S.)
| | - Benazir Siddeek
- Woman-Mother-Child Department, Division of Pediatrics, DOHaD Laboratory, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Rue du Bugnon 27, 1011 Lausanne, Switzerland; (G.B.); (M.L.); (H.C.); (U.S.)
| |
Collapse
|
19
|
Zhang H, Jin Y, Wang M, Loor JJ, Wang H. N-Carbamylglutamate and l-arginine supplementation improve hepatic antioxidant status in intrauterine growth-retarded suckling lambs. RSC Adv 2020; 10:11173-11181. [PMID: 35495302 PMCID: PMC9050450 DOI: 10.1039/c9ra09316h] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 03/01/2020] [Indexed: 12/26/2022] Open
Abstract
The influence of dietary supplementation of l-arginine (Arg) or N-carbamylglutamate (NCG) on the hepatic antioxidant status in intrauterine-growth-retarded (IUGR) suckling lambs remains unclear. The current work aimed to investigate the regulatory mechanisms whereby dietary Arg or NCG alter hepatic antioxidant status in suckling lambs suffering from IUGR. Forty-eight newborn Hu lambs of normal birth weight (CON) and IUGR were allocated randomly into four groups of 12 animals each: CON (4.25 ± 0.14 kg), IUGR (3.01 ± 0.12 kg), IUGR + 1% Arg (2.99 ± 0.13 kg), or IUGR + 0.1% NCG (3.03 ± 0.11 kg). All lambs were raised for a period of 21 days from 7 to 28 days after birth. Compared with the IUGR suckling animals, glutathione peroxidase (GSH-Px), superoxide dismutase (SOD), and reduced glutathione (GSH) content were greater (P < 0.05), and protein carbonyl and malondialdehyde (MDA) levels were reduced (P < 0.05) in the livers of both IUGR + 1% Arg and 0.1% NCG suckling animals. Relative to IUGR suckling lambs, supplementing with Arg or NCG markedly reduced (P < 0.05) reactive oxygen species (ROS) levels, apoptosis, and necrosis in liver. Relative to IUGR suckling lambs, protein and mRNA expression of GSH-Px1, SOD2, catalase (CAT), heme oxygenase-1 (HO-1), inducible nitric oxide (NO) synthase (iNOS), and epithelial NO synthase (eNOS) increased in IUGR animals receiving Arg or NCG (P < 0.05). Both Arg and NCG can protect neonates from IUGR-induced hepatic oxidative damage through promoting the expression of antioxidative enzymes (including SOD, CAT, and GSH-Px), phase II metabolizing enzymes, and activation of the NO pathway. The influence of dietary supplementation of l-arginine (Arg) or N-carbamylglutamate (NCG) on the hepatic antioxidant status in intrauterine-growth-retarded (IUGR) suckling lambs remains unclear.![]()
Collapse
Affiliation(s)
- Hao Zhang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition
- College of Animal Science and Technology
- Yangzhou University
- Yangzhou 225009
- P. R. China
| | - Yaqian Jin
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition
- College of Animal Science and Technology
- Yangzhou University
- Yangzhou 225009
- P. R. China
| | - Mengzhi Wang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition
- College of Animal Science and Technology
- Yangzhou University
- Yangzhou 225009
- P. R. China
| | - Juan J. Loor
- Department of Animal Sciences and Division of Nutritional Sciences
- University of Illinois
- Urbana
- USA
| | - Hongrong Wang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition
- College of Animal Science and Technology
- Yangzhou University
- Yangzhou 225009
- P. R. China
| |
Collapse
|
20
|
Neto JGO, Boechat SK, Romão JS, Pazos-Moura CC, Oliveira KJ. Treatment with cinnamaldehyde reduces the visceral adiposity and regulates lipid metabolism, autophagy and endoplasmic reticulum stress in the liver of a rat model of early obesity. J Nutr Biochem 2019; 77:108321. [PMID: 31869758 DOI: 10.1016/j.jnutbio.2019.108321] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 11/18/2019] [Accepted: 12/06/2019] [Indexed: 12/11/2022]
Abstract
Nutrition at early stages of life contributes to the alarming incidence of childhood obesity, insulin resistance and hepatoesteatosis. Cinnamaldehyde, major component of cinnamon, increases insulin sensitivity and modulates adiposity and lipid metabolism. The aim of this study was to analyze the impact of cinnamaldehyde treatment during adolescence in a rat model of early obesity. Litter size reduction was used to induce overfeeding and early obesity. At postnatal day 30 (adolescence), the male Wistar rats received cinnamaldehyde by gavage (40 mg/kg of body weight/day) for 29 days and were studied at the end of treatment at 60 days old or 4 months thereafter (180 days old). At 60 days of age, the treatment with cinnamaldehyde promoted reduced visceral adiposity, serum triacylglycerol, and attenuation of energy efficiency and insulin resistance. In the liver, it reduced lipid synthesis, stimulated autophagy and reduced ER stress. At 180 days of age, animals treated with cinnamaldehyde during the adolescence exhibited normalization of visceral adiposity and energy efficiency, and attenuation of hyperphagia, serum hypertriglyceridemia and hepatic triacylglycerol content, with molecular markers indicative of reduced hepatic synthesis. However, the beneficial effect observed at 60 days of age on glucose homeostasis, autophagy and ER stress was lost. Therefore, the cinnamaldehyde supplementation during the adolescence has short- and long-term metabolic beneficial effects, highlighting its potential as an adjuvant in the treatment of early obesity.
Collapse
Affiliation(s)
- Jessika Geisebel Oliveira Neto
- Departamento de Fisiologia e Farmacologia, Universidade Federal Fluminense, Rua Hernani Pires de Melo, 101, São domingos, Niterói, 24210-130, RJ, Brazil
| | - Silvia Karl Boechat
- Departamento de Fisiologia e Farmacologia, Universidade Federal Fluminense, Rua Hernani Pires de Melo, 101, São domingos, Niterói, 24210-130, RJ, Brazil
| | - Juliana Santos Romão
- Departamento de Fisiologia e Farmacologia, Universidade Federal Fluminense, Rua Hernani Pires de Melo, 101, São domingos, Niterói, 24210-130, RJ, Brazil
| | - Carmen Cabanelas Pazos-Moura
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Centro de Ciências da Saúde, Ilha do Fundão, Rio de Janeiro,21949-900, RJ, Brazil
| | - Karen Jesus Oliveira
- Departamento de Fisiologia e Farmacologia, Universidade Federal Fluminense, Rua Hernani Pires de Melo, 101, São domingos, Niterói, 24210-130, RJ, Brazil; Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Centro de Ciências da Saúde, Ilha do Fundão, Rio de Janeiro,21949-900, RJ, Brazil.
| |
Collapse
|
21
|
Lee HS. The interaction between gut microbiome and nutrients on development of human disease through epigenetic mechanisms. Genomics Inform 2019; 17:e24. [PMID: 31610620 PMCID: PMC6808642 DOI: 10.5808/gi.2019.17.3.e24] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 09/22/2019] [Indexed: 02/06/2023] Open
Abstract
Early environmental exposure is recognized as a key factor for long-term health based on the Developmental Origins of Health and Disease hypothesis. It considers that early-life nutrition is now being recognized as a major contributor that may permanently program change of organ structure and function toward the development of diseases, in which epigenetic mechanisms are involved. Recent researches indicate early-life environmental factors modulate the microbiome development and the microbiome might be mediate diet-epigenetic interaction. This review aims to define which nutrients involve microbiome development during the critical window of susceptibility to disease, and how microbiome modulation regulates epigenetic changes and influences human health and future prevention strategies.
Collapse
Affiliation(s)
- Ho-Sun Lee
- Forensic Toxicology Division, Daegu Institute, National Forensic Service, Chilgok 39872, Korea
| |
Collapse
|
22
|
Maternal protein intake during pregnancy is associated with child growth up to 5 years of age, but not through insulin-like growth factor-1: findings from the ROLO study. Br J Nutr 2019; 120:1252-1261. [PMID: 30484757 DOI: 10.1017/s0007114518002611] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Infant protein intake has been associated with child growth, however, research on maternal protein intake during pregnancy is limited. Insulin-like growth factors (IGF) play a role in early fetal development and maternal protein intake may influence child body composition via IGF-1. The aim of this study was to investigate the association of maternal protein intake throughout pregnancy on cord blood IGF-1 and child body composition from birth to 5 years of age. Analysis was carried out on 570 mother-child dyads from the Randomised cOntrol trial of LOw glycaemic index diet study. Protein intake was recorded using 3-d food diaries in each trimester of pregnancy and protein intake per kg of maternal weight (g/d per kg) was calculated. Cord blood IGF-1 was measured at birth. Infant anthropometry was measured at birth, 6 months, 2 and 5 years of age. Mixed modelling, linear regression, and mediation analysis were carried out. Birth weight centiles were positively associated with early-pregnancy protein intake (g/d per kg), while weight centiles from 6 months to 5 years were negatively associated (B=-21·6, P<0·05). These associations were not mediated by IGF-1. Our findings suggest that high protein intake in early-pregnancy may exert an in utero effect on offspring body composition with a higher weight initially at birth but slower growth rates into childhood. Further research is needed to elucidate the exact mechanisms by which dietary protein modulates fetal growth.
Collapse
|
23
|
Rivera P, Ramírez‐López MT, Vargas A, Decara J, Vázquez M, Arco R, Gómez de Heras R, Argente J, Rodríguez de Fonseca F, Chowen JA, Suárez J. Perinatal free-choice of a high-calorie low-protein diet affects leptin signaling through IRS1 and AMPK dephosphorylation in the hypothalami of female rat offspring in adulthood. Acta Physiol (Oxf) 2019; 226:e13244. [PMID: 30589509 DOI: 10.1111/apha.13244] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 12/07/2018] [Accepted: 12/17/2018] [Indexed: 12/17/2022]
Abstract
AIM We aimed to investigate whether a dysregulated maternal diet during gestation and lactation induces long-lasting changes in the hypothalamic control of feeding behavior in the offspring and whether this effect is sex specific. METHODS The study included an analysis of appetite-regulating metabolic hormones and hypothalamic signaling in male and female offspring in adulthood after exposure to a free-choice high-calorie palatable low-protein (P) diet or standard chow (C) during (pre)gestation/lactation (maternal) and/or postweaning (offspring). RESULTS Maternal exposure to the P diet resulted in decreased protein intake and body weight gain in dams and decreased body weight gain in offspring during lactation. The maternal P diet (PC) specifically increased feed efficacy and decreased body weight and cholesterol levels in the female offspring in adulthood, but no changes in adiposity or leptin levels were found. In contrast, P diet exposure after weaning (CP and PP) increased caloric intake, adiposity and circulating levels of leptin in the male and female offspring in adulthood. The hypothalami of the female offspring exposed to the maternal P diet (PC and PP) expressed high levels of the phospho-leptin receptor and low levels of SOCS3, phospho-IRS1 and phospho-AMPK, regardless of the postweaning diet. The hypothalami of the female rats in the PC group also showed increased levels of STAT3 and the orexigenic neuropeptide Agrp. CONCLUSIONS Maternal exposure to a free-choice high-calorie low-protein diet induces a long-term feed efficacy associated with changes in leptin signaling through IRS-1 and AMPK dephosphorylation in the hypothalami of female offspring in adulthood.
Collapse
Affiliation(s)
- Patricia Rivera
- Department of Endocrinology, Fundación Investigación Biomédica del Hospital Infantil Universitario Niño Jesús Instituto de Investigación Biomédica la Princesa Madrid Spain
| | - María T. Ramírez‐López
- Departamento de Psicobiología, Facultad de Psicología Universidad Complutense de Madrid Pozuelo de Alarcón Spain
- Hospital Universitario de Getafe Getafe (Madrid) Spain
| | - Antonio Vargas
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga Universidad de Málaga Málaga Spain
| | - Juan Decara
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga Universidad de Málaga Málaga Spain
| | - Mariam Vázquez
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga Universidad de Málaga Málaga Spain
| | - Rocío Arco
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga Universidad de Málaga Málaga Spain
| | - Raquel Gómez de Heras
- Departamento de Psicobiología, Facultad de Psicología Universidad Complutense de Madrid Pozuelo de Alarcón Spain
| | - Jesús Argente
- Department of Endocrinology, Fundación Investigación Biomédica del Hospital Infantil Universitario Niño Jesús Instituto de Investigación Biomédica la Princesa Madrid Spain
- Centro de Investigación Biomédica en Red Fisiología de la Obesidad y Nutrición (CIBEROBN) Madrid Spain
- IMDEA Food Institute Campus of International Excellence (CEI) UAM + CSIC Madrid Spain
- Department of PediatricsUniversity Autonoma de Madrid Madrid Spain
| | - Fernando Rodríguez de Fonseca
- Departamento de Psicobiología, Facultad de Psicología Universidad Complutense de Madrid Pozuelo de Alarcón Spain
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga Universidad de Málaga Málaga Spain
| | - Julie A. Chowen
- Department of Endocrinology, Fundación Investigación Biomédica del Hospital Infantil Universitario Niño Jesús Instituto de Investigación Biomédica la Princesa Madrid Spain
- Centro de Investigación Biomédica en Red Fisiología de la Obesidad y Nutrición (CIBEROBN) Madrid Spain
- IMDEA Food Institute Campus of International Excellence (CEI) UAM + CSIC Madrid Spain
| | - Juan Suárez
- Unidad de Gestión Clínica de Salud Mental, Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga Universidad de Málaga Málaga Spain
| |
Collapse
|
24
|
Allman BR, Andres A, Børsheim E. The Association of Maternal Protein Intake during Pregnancy in Humans with Maternal and Offspring Insulin Sensitivity Measures. Curr Dev Nutr 2019; 3:nzz055. [PMID: 31139768 PMCID: PMC6533362 DOI: 10.1093/cdn/nzz055] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/16/2019] [Accepted: 04/18/2019] [Indexed: 12/15/2022] Open
Abstract
The purpose of this review is to critically evaluate the studies assessing the relations between protein intake during human pregnancy and insulin sensitivity measures in the mother and offspring, and to get a better understanding of the knowledge gaps that still exist. Overall, there is insufficient evidence to conclude about implications of higher amounts of protein intake during pregnancy on maternal or offspring insulin sensitivity. However, studies show a relation between protein quality and insulin sensitivity, such that animal protein may be associated with negative outcomes and plant protein may be associated with positive insulin sensitivity outcomes. There is an urgent need for standardized studies using comparable terminology to evaluate any potential relations between insulin sensitivity in mothers and offspring and truly low and high maternal protein intake while maintaining eucaloric balance to better inform about optimal protein dosage and quality during this period.
Collapse
Affiliation(s)
- Brittany R Allman
- Arkansas Children's Nutrition Center, Little Rock, AR, USA
- Arkansas Children's Research Institute, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Aline Andres
- Arkansas Children's Nutrition Center, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Elisabet Børsheim
- Arkansas Children's Nutrition Center, Little Rock, AR, USA
- Arkansas Children's Research Institute, Little Rock, AR, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
25
|
Darby JRT, Mohd Dollah MHB, Regnault TRH, Williams MT, Morrison JL. Systematic review: Impact of resveratrol exposure during pregnancy on maternal and fetal outcomes in animal models of human pregnancy complications-Are we ready for the clinic? Pharmacol Res 2019; 144:264-278. [PMID: 31029765 DOI: 10.1016/j.phrs.2019.04.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/15/2019] [Accepted: 04/18/2019] [Indexed: 12/14/2022]
Abstract
Resveratrol (RSV) has been reported to have potential beneficial effects in the complicated pregnancy. Various pregnancy complications lead to a suboptimal in utero environment that impacts fetal growth during critical windows of development. Detrimental structural changes to key organ systems in utero persist into adult life and predispose offspring to an increased risk of chronic non-communicable metabolic diseases such as cardiovascular disease, diabetes and obesity. The aim of this systematic review was to determine the effect of gestational RSV exposure on both maternal and fetal outcomes. Publicly available databases (n = 8) were searched for original studies reporting maternal and/or fetal outcomes after RSV exposure during pregnancy irrespective of species. Of the 115 studies screened, 31 studies were included in this review. RSV exposure occurred for different durations across a range of species (Rats n = 18, Mice n = 7, Japanese Macaques n = 3 and Sheep n = 3), models of complicated pregnancy (eg. maternal dietary manipulations, gestational diabetes, maternal hypoxia, teratogen exposure, etc.), dosages and administration routes. Maternal and fetal outcomes differed not only based on the model of complicated pregnancy assessed but also as a result of species. Given the heterogenic nature of these studies, further investigation assessing RSV exposure during the complicated pregnancy is warranted. In order to make an informed decision regarding the use of RSV to intervene in pregnancy complications, we suggest a minimum data set for consideration in future studies.
Collapse
Affiliation(s)
- Jack R T Darby
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | - Murni H B Mohd Dollah
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia
| | - Timothy R H Regnault
- Departments of Obstetrics and Gynaecology and Physiology and Pharmacology, Western University, 1151 Richmond Street, London, ON, N6A 5C1, Canada; Children's Health Research Institute, 800 Commissioners Road East, London, ON, N6C 2V5, Canada
| | - Marie T Williams
- Health and Alliance for Research in Exercise, Nutrition and Activity (ARENA), School of Health Sciences, University of South Australia, Adelaide, Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, Australia.
| |
Collapse
|
26
|
Guo J, Li B, Zuo Z, Chen M, Wang C. Maternal Supplementation with β‐Carotene During Pregnancy Disturbs Lipid Metabolism and Glucose Homoeostasis in F1 Female Mice. Mol Nutr Food Res 2019; 63:e1900072. [DOI: 10.1002/mnfr.201900072] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Indexed: 12/25/2022]
Affiliation(s)
- Jiaojiao Guo
- State Key Laboratory of Cellular Stress BiologySchool of Life SciencesXiamen University Xiamen 36110 P. R. China
| | - Bingshui Li
- State Key Laboratory of Cellular Stress BiologySchool of Life SciencesXiamen University Xiamen 36110 P. R. China
| | - Zhenghong Zuo
- State Key Laboratory of Cellular Stress BiologySchool of Life SciencesXiamen University Xiamen 36110 P. R. China
| | - Meng Chen
- State Key Laboratory of Cellular Stress BiologySchool of Life SciencesXiamen University Xiamen 36110 P. R. China
- Key Laboratory of Ministry of Education for Subtropical Wetland Ecosystem ResearchXiamen University Xiamen 36110 P. R. China
| | - Chonggang Wang
- State Key Laboratory of Cellular Stress BiologySchool of Life SciencesXiamen University Xiamen 36110 P. R. China
- Key Laboratory of Ministry of Education for Subtropical Wetland Ecosystem ResearchXiamen University Xiamen 36110 P. R. China
| |
Collapse
|
27
|
Enteric Neuronal Degeneration: Is it Due to Your Mother's Diet? Neuroscience 2019; 393:366-368. [PMID: 30454863 DOI: 10.1016/j.neuroscience.2018.10.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 10/08/2018] [Indexed: 11/20/2022]
|
28
|
Abstract
Our social environment, from the microscopic to the macro-social, affects us for the entirety of our lives. One integral line of research to examine how interpersonal and societal environments can get "under the skin" is through the lens of epigenetics. Epigenetic mechanisms are adaptations made to our genome in response to our environment which include tags placed on and removed from the DNA itself to how our DNA is packaged, affecting how our genes are read, transcribed, and interact. These tags are affected by social environments and can persist over time; this may aid us in responding to experiences and exposures, both the enriched and the disadvantageous. From memory formation to immune function, the experience-dependent plasticity of epigenetic modifications to micro- and macro-social environments may contribute to the process of learning from comfort, pain, and stress to better survive in whatever circumstances life has in store.
Collapse
Affiliation(s)
- Sarah M Merrill
- Centre for Molecular Medicine and Therapeutics, British Columbia Children's Hospital, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Nicole Gladish
- Centre for Molecular Medicine and Therapeutics, British Columbia Children's Hospital, Vancouver, BC, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Michael S Kobor
- Centre for Molecular Medicine and Therapeutics, British Columbia Children's Hospital, Vancouver, BC, Canada.
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada.
- Human Early Learning Partnership, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
29
|
Coppedè F, Seghieri M, Stoccoro A, Santini E, Giannini L, Rossi C, Migliore L, Solini A. DNA methylation of genes regulating appetite and prediction of weight loss after bariatric surgery in obese individuals. J Endocrinol Invest 2019; 42:37-44. [PMID: 29603098 DOI: 10.1007/s40618-018-0881-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 03/25/2018] [Indexed: 12/15/2022]
Abstract
PURPOSE Epigenetic traits are influenced by clinical variables; interaction between DNA methylation (DNAmeth) and bariatric surgery-induced weight loss has been scarcely explored. We investigated whether DNAmeth of genes encoding for molecules/hormones regulating appetite, food intake or obesity could predict successful weight outcome following Roux-en-Y gastric bypass (RYGB). METHODS Forty-five obese individuals with no known comorbidities were stratified accordingly to weight decrease one-year after RYGB (excess weight loss, EWL ≥ 50%: good responders, GR; EWL < 50%: worse responders, WR). DNAmeth of leptin (LEP), ghrelin (GHRL), ghrelin receptor (GHSR) and insulin-growth factor-2 (IGF2) was assessed before intervention. Single nucleotide polymorphisms of genes affecting DNAmeth, DNMT3A and DNMT3B, were also determined. RESULTS At baseline, type 2 diabetes was diagnosed by OGTT in 13 patients. Post-operatively, GR (n = 23) and WR (n = 22) achieved an EWL of 67.7 ± 9.6 vs 38.2 ± 9.0%, respectively. Baseline DNAmeth did not differ between GR and WR for any tested genes, even when the analysis was restricted to subjects with no diabetes. A relationship between GHRL and LEP methylation profiles emerged (r = 0.47, p = 0.001). Searching for correlation between DNAmeth of the studied genes with demographic characteristics and baseline biochemical parameters of the studied population, we observed a correlation between IGF2 methylation and folate (r = 0.44, p = 0.003). Rs11683424 for DNMT3A and rs2424913 for DNMT3B did not correlate with DNAmeth of the studied genes. CONCLUSIONS In severely obese subjects, the degree of DNAmeth of some genes affecting obesity and related conditions does not work as predictor of successful response to RYGB.
Collapse
Affiliation(s)
- F Coppedè
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - M Seghieri
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - A Stoccoro
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - E Santini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - L Giannini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - C Rossi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - L Migliore
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - A Solini
- Department of Surgical, Medical, Molecular and Critical Area Pathology, University of Pisa, Via Roma 67, 56126, Pisa, Italy.
| |
Collapse
|
30
|
Lubinsky M. Evolutionary justifications for human reproductive limitations. J Assist Reprod Genet 2018; 35:2133-2139. [PMID: 30116921 DOI: 10.1007/s10815-018-1285-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 08/02/2018] [Indexed: 01/27/2023] Open
Abstract
Common human reproductive inefficiencies have multiple etiologies. Going against chance, many effects, such as polycystic ovaries, endometriosis, and folate metabolic issues, have genetic components, while aneuploid losses arise from diverse mitotic and meiotic errors at different stages, some transitory. This can be advantageous, since greater overall survival with fewer offspring can increase reproductive success. Benefits primarily accrue to mothers, who bear most child related costs, and for whom early losses are less costly than late. Different adaptations to different situations reflect human evolutionary history. For early speciation, periodic climate extremes repeatedly reduced resources, favoring limitations while contracted populations helped fix relevant genes. Later, under better conditions, evolving social cooperation could increase fecundity faster than it added resources, further supporting reproductive suppression through mitotic aneuploidy, with very early losses minimizing maternal costs. The grandmother hypothesis suggests benefits in limiting reproduction as maternal age increased pregnancy risks in order to support grandchildren as they arrived, selecting for maternal age-related meiotic aneuploidy. Finally, with variable short-term agricultural shortages, acute reproductive responses arose through chromatin "nutrient sensor"-regulated epigenetic effects that also shifted some lethal effects earlier, reducing both maternal and mutation load costs. Overall, despite suggestions to the contrary, it is likely that human selective pressures have not decreased with civilization, but that many of the costs have been shifted to early reproduction.
Collapse
|
31
|
Mitanchez D, Chavatte-Palmer P. Review shows that maternal obesity induces serious adverse neonatal effects and is associated with childhood obesity in their offspring. Acta Paediatr 2018; 107:1156-1165. [PMID: 29421859 DOI: 10.1111/apa.14269] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 01/10/2018] [Accepted: 02/02/2018] [Indexed: 12/18/2022]
Abstract
AIM Obesity at the start of pregnancy has been rising worldwide, increasing the risk of maternal complications. We reviewed the independent effects of maternal obesity during pregnancy on neonatal adverse outcomes and the risk of childhood obesity and adverse cardio-metabolic profiles. METHODS We searched MEDLINE for papers published in English between December 2007 and November 2017, focusing primarily on human studies published in the last five years. However, we also chose to highlight examples derived from model animals that could bring mechanistic insight and preventive and therapeutic avenues. RESULTS Our review showed that maternal obesity had independent effects on neonatal adverse outcomes such as macrosomia, perinatal mortality and birth defects. Maternal obesity alone increased the risks for adverse neonatal outcomes, including macrosomia, perinatal mortality, induced preterm birth and birth defects. In association with excess gestational weight gain, mainly early in pregnancy, increased the risks of childhood obesity, higher fat mass and, to a smaller extent, adverse cardio-metabolic profiles. Animal models highlighted sexually dimorphic responses to maternal obesity. CONCLUSION Maternal obesity induced serious adverse neonatal effects and was associated with childhood obesity in their offspring. The peri-conceptional period is critical for metabolic programming, and obese women need close monitoring from conception.
Collapse
Affiliation(s)
- Delphine Mitanchez
- Department of Perinatality; APHP; GHUEP; Armand Trousseau Hospital; Paris France
- Faculty of Medicine; Sorbonne University; Paris France
| | | |
Collapse
|
32
|
Dietary vitamin A impacts DNA methylation patterns of adipogenesis-related genes in suckling rats. Arch Biochem Biophys 2018; 650:75-84. [DOI: 10.1016/j.abb.2018.05.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 04/23/2018] [Accepted: 05/09/2018] [Indexed: 01/07/2023]
|
33
|
Lubinsky M. An epigenetic association of malformations, adverse reproductive outcomes, and fetal origins hypothesis related effects. J Assist Reprod Genet 2018; 35:953-964. [PMID: 29855751 PMCID: PMC6030006 DOI: 10.1007/s10815-018-1197-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2018] [Accepted: 04/25/2018] [Indexed: 12/17/2022] Open
Abstract
VACTERL, the prototype for associated congenital anomalies, also has connections with functional issues such as pregnancy losses, prematurity, growth delays, perinatal difficulties, and parental subfertility. This segues into a broader association with similar connections even in the absence of malformations. DNA methylation disturbances in the ovum are a likely cause, with epigenetic links to individual components and to folate effects before conception, explaining diverse fetal and placental findings and providing a link to fetal origin hypothesis-related effects. The association encompasses the following: (1) Pre- and periconceptual effects, with frequent fertility issues and occasional imprinting disorders. (2) Early malformations. (3) Adverse pregnancy outcomes (APOs), as above. (4) Developmental destabilization that resolves soon after birth. This potentiates other causes of association findings, introducing multiple confounders. (5) Long-term fetal origins hypothesis-related risks. The other findings are exceptional when the same malformations have Mendelian origins, supporting a distinct pathogenesis. Expressions are facilitated by one-carbon metabolic issues, maternal and fetal stress, and decreased embryo size. This may be one of the commonest causes of adverse reproductive outcomes, but multifactorial findings, variable onsets and phenotypes, and interactions with multiple confounders make recognition difficult. This association supports VACTERL as a continuum that includes isolated malformations, extends the fetal origins hypothesis, explains adverse effects linked to maternal obesity, and suggests possible interventions.
Collapse
Affiliation(s)
- Mark Lubinsky
- , 6003 W. Washington Blvd., Wauwatosa, WI, 53213, USA.
| |
Collapse
|
34
|
Abstract
Obesity poses a severe threat to human health, including the increased prevalence of hypertension, insulin resistance, diabetes mellitus, cancer, inflammation, sleep apnoea and other chronic diseases. Current therapies focus mainly on suppressing caloric intake, but the efficacy of this approach remains poor. A better understanding of the pathophysiology of obesity will be essential for the management of obesity and its complications. Knowledge gained over the past three decades regarding the aetiological mechanisms underpinning obesity has provided a framework that emphasizes energy imbalance and neurohormonal dysregulation, which are tightly regulated by autophagy. Accordingly, there is an emerging interest in the role of autophagy, a conserved homeostatic process for cellular quality control through the disposal and recycling of cellular components, in the maintenance of cellular homeostasis and organ function by selectively ridding cells of potentially toxic proteins, lipids and organelles. Indeed, defects in autophagy homeostasis are implicated in metabolic disorders, including obesity, insulin resistance, diabetes mellitus and atherosclerosis. In this Review, the alterations in autophagy that occur in response to nutrient stress, and how these changes alter the course of obesogenesis and obesity-related complications, are discussed. The potential of pharmacological modulation of autophagy for the management of obesity is also addressed.
Collapse
Affiliation(s)
- Yingmei Zhang
- Department of Cardiology, Fudan University Zhongshan Hospital, Shanghai, China.
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, USA.
| | - James R Sowers
- Diabetes and Cardiovascular Research Center, University of Missouri-Columbia School of Medicine, Columbia, MO, USA
| | - Jun Ren
- Department of Cardiology, Fudan University Zhongshan Hospital, Shanghai, China.
- Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, USA.
| |
Collapse
|
35
|
Plucińska K, Barger SW. Maternal obesity reprograms offspring's executive brain centers in a sex-specific manner?: An Editorial for 'Perinatal high fat diet and early life methyl donor supplementation alter one carbon metabolism and DNA methylation in the brain' on page 362. J Neurochem 2018; 145:358-361. [PMID: 29663393 DOI: 10.1111/jnc.14334] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 03/01/2018] [Indexed: 01/21/2023]
Abstract
This editorial highlights an article by McKee and colleagues in the current issue of Journal of Neurochemistry, in which the authors report epigenetic changes linked to one-carbon metabolism in prefrontal cortex (PFC) of murine offspring from dams fed high-fat diet to mimic maternal obesity. The group found that high-fat diet feeding in utero increases weight gain in offspring and dynamically alters DNA methylation in the PFC of male but not female brains. These epigenetic marks were associated with a shift in brain one-carbon metabolism (folate and methionine) intermediates and were normalized by early-life methyl-donor supplementation in a sex-specific manner.
Collapse
Affiliation(s)
- Kaja Plucińska
- Center for Basic Metabolic Research, Section of Integrative Physiology, University of Copenhagen, Copenhagen, Denmark
| | - Steven W Barger
- Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock AR, USA
| |
Collapse
|
36
|
Geraghty AA, Sexton-Oates A, O’Brien EC, Alberdi G, Fransquet P, Saffery R, McAuliffe FM. A Low Glycaemic Index Diet in Pregnancy Induces DNA Methylation Variation in Blood of Newborns: Results from the ROLO Randomised Controlled Trial. Nutrients 2018; 10:nu10040455. [PMID: 29642382 PMCID: PMC5946240 DOI: 10.3390/nu10040455] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/28/2018] [Accepted: 04/04/2018] [Indexed: 11/30/2022] Open
Abstract
The epigenetic profile of the developing fetus is sensitive to environmental influence. Maternal diet has been shown to influence DNA methylation patterns in offspring, but research in humans is limited. We investigated the impact of a low glycaemic index dietary intervention during pregnancy on offspring DNA methylation patterns using a genome-wide methylation approach. Sixty neonates were selected from the ROLO (Randomised cOntrol trial of LOw glycaemic index diet to prevent macrosomia) study: 30 neonates from the low glycaemic index intervention arm and 30 from the control, whose mothers received no specific dietary advice. DNA methylation was investigated in 771,484 CpG sites in free DNA from cord blood serum. Principal component analysis and linear regression were carried out comparing the intervention and control groups. Gene clustering and pathway analysis were also explored. Widespread variation was identified in the newborns exposed to the dietary intervention, accounting for 11% of the total level of DNA methylation variation within the dataset. No association was found with maternal early-pregnancy body mass index (BMI), infant sex, or birthweight. Pathway analysis identified common influences of the intervention on gene clusters plausibly linked to pathways targeted by the intervention, including cardiac and immune functioning. Analysis in 60 additional samples from the ROLO study failed to replicate the original findings. Using a modest-sized discovery sample, we identified preliminary evidence of differential methylation in progeny of mothers exposed to a dietary intervention during pregnancy.
Collapse
Affiliation(s)
- Aisling A. Geraghty
- UCD Perinatal Research Centre, Obstetrics and Gynaecology, School of Medicine, University College Dublin, National Maternity Hospital, Dublin 2, Ireland; (A.A.G.); (E.C.O.); (G.A.)
| | - Alexandra Sexton-Oates
- Cancer and Disease Epigenetics, Murdoch Children’s Research Institute, Melbourne, Victoria 3052, Australia; (A.S.-O.); (P.F.); (R.S.)
| | - Eileen C. O’Brien
- UCD Perinatal Research Centre, Obstetrics and Gynaecology, School of Medicine, University College Dublin, National Maternity Hospital, Dublin 2, Ireland; (A.A.G.); (E.C.O.); (G.A.)
| | - Goiuri Alberdi
- UCD Perinatal Research Centre, Obstetrics and Gynaecology, School of Medicine, University College Dublin, National Maternity Hospital, Dublin 2, Ireland; (A.A.G.); (E.C.O.); (G.A.)
| | - Peter Fransquet
- Cancer and Disease Epigenetics, Murdoch Children’s Research Institute, Melbourne, Victoria 3052, Australia; (A.S.-O.); (P.F.); (R.S.)
| | - Richard Saffery
- Cancer and Disease Epigenetics, Murdoch Children’s Research Institute, Melbourne, Victoria 3052, Australia; (A.S.-O.); (P.F.); (R.S.)
- Department of Paediatrics, University of Melbourne, Victoria 3010, Australia
| | - Fionnuala M. McAuliffe
- UCD Perinatal Research Centre, Obstetrics and Gynaecology, School of Medicine, University College Dublin, National Maternity Hospital, Dublin 2, Ireland; (A.A.G.); (E.C.O.); (G.A.)
- Correspondence: ; Tel.: +353-1-63732
| |
Collapse
|
37
|
Abstract
PURPOSE OF REVIEW It is becoming increasingly evident that epigenetic mechanisms, particularly DNA methylation, play a role in the regulation of blood lipid levels and lipid metabolism-linked phenotypes and diseases. RECENT FINDINGS Recent genome-wide methylation and candidate gene studies of blood lipids have highlighted several robustly replicated methylation markers across different ethnicities. Furthermore, many of these lipid-related CpG sites associated with blood lipids are also linked to lipid-related phenotypes and diseases. Integrating epigenome-wide association studies (EWAS) data with other layers of molecular data such as genetics or the transcriptome, accompanied by relevant statistical methods (e.g. Mendelian randomization), provides evidence for causal relationships. Recent data suggest that epigenetic changes can be consequences rather than causes of dyslipidemia. There is sparse information on many lipid classes and disorders of lipid metabolism, and also on the interplay of DNA methylation with other epigenetic layers such as histone modifications and regulatory RNAs. SUMMARY The current review provides a literature overview of epigenetic modifications in lipid metabolism and other lipid-related phenotypes and diseases focusing on EWAS of DNA methylation from January 2016 to September 2017. Recent studies strongly support the importance of epigenetic modifications, such as DNA methylation, in lipid metabolism and related diseases for relevant biological insights, reliable biomarkers, and even future therapeutics.
Collapse
Affiliation(s)
- Kirstin Mittelstraß
- Research Unit of Molecular Epidemiology
- Institute of Epidemiology, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
| | - Melanie Waldenberger
- Research Unit of Molecular Epidemiology
- Institute of Epidemiology, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), Neuherberg, Germany
| |
Collapse
|
38
|
Zhao N, Yang S, Jia Y, Sun B, He B, Zhao R. Maternal betaine supplementation attenuates glucocorticoid-induced hepatic lipid accumulation through epigenetic modification in adult offspring rats. J Nutr Biochem 2018; 54:105-112. [DOI: 10.1016/j.jnutbio.2017.12.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 11/15/2017] [Accepted: 12/06/2017] [Indexed: 12/21/2022]
|
39
|
Won SB, Kwon YH. Maternal Consumption of Low-Isoflavone Soy Protein Isolate Confers the Increased Predisposition to Alcoholic Liver Injury in Adult Rat Offspring. Nutrients 2018. [PMID: 29534433 PMCID: PMC5872750 DOI: 10.3390/nu10030332] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Offspring of female rats fed either a casein (CAS) diet or a low-isoflavone soy protein isolate (SPI) diet were compared in an animal model of chronic ethanol consumption to investigate whether maternal diet regulates the adaptive responses of offspring to postnatal ethanol exposure and potentially affects the development of liver disease in later life. Female rats were fed either a CAS or an SPI diet before mating, and during pregnancy and lactation. Male offspring from the same litter were pair-fed either a control or ethanol diet for six weeks (CAS/CON, CAS/EtOH, SPI/CON, and SPI/EtOH groups). Serum aminotransferase activities and hepatic inflammatory indicators were higher in the SPI/EtOH group than in the CAS/EtOH group. Ethanol consumption increased serum homocysteine levels, hepatic S-adenosylmethionine:S-adenosylhomocysteine ratio, and hepatic endoplasmic reticulum stress only in offspring of SPI-fed female rats. Total and high-density lipoprotein (HDL) cholesterol levels and mRNA levels of hepatic genes involved in HDL cholesterol assembly were reduced in the SPI group in response to ethanol consumption. In conclusion, offspring of SPI-fed female rats were more susceptible to the later development of alcoholic liver disease than offspring of CAS-fed female rats. Furthermore, maternal SPI consumption altered one-carbon metabolism and cholesterol metabolism of offspring fed an ethanol diet.
Collapse
Affiliation(s)
- Sae Bom Won
- Department of Food and Nutrition, Seoul National University, Seoul 08826, Korea.
- Research Institute of Human Ecology, Seoul National University, Seoul 08826, Korea.
| | - Young Hye Kwon
- Department of Food and Nutrition, Seoul National University, Seoul 08826, Korea.
- Research Institute of Human Ecology, Seoul National University, Seoul 08826, Korea.
| |
Collapse
|
40
|
Sanchez-Garrido MA, Ruiz-Pino F, Velasco I, Barroso A, Fernandois D, Heras V, Manfredi-Lozano M, Vazquez MJ, Castellano JM, Roa J, Pinilla L, Tena-Sempere M. Intergenerational Influence of Paternal Obesity on Metabolic and Reproductive Health Parameters of the Offspring: Male-Preferential Impact and Involvement of Kiss1-Mediated Pathways. Endocrinology 2018; 159:1005-1018. [PMID: 29309558 DOI: 10.1210/en.2017-00705] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 12/19/2017] [Indexed: 12/12/2022]
Abstract
Obesity and its comorbidities are reaching epidemic proportions worldwide. Maternal obesity is known to predispose the offspring to metabolic disorders, independently of genetic inheritance. This intergenerational transmission has also been suggested for paternal obesity, with a potential negative impact on the metabolic and, eventually, reproductive health of the offspring, likely via epigenetic changes in spermatozoa. However, the neuroendocrine component of such phenomenon and whether paternal obesity sensitizes the offspring to the disturbances induced by high-fat diet (HFD) remain poorly defined. We report in this work the metabolic and reproductive impact of HFD in the offspring from obese fathers, with attention to potential sex differences and alterations of hypothalamic Kiss1 system. Lean and obese male rats were mated with lean virgin female rats; male and female offspring were fed HFD from weaning onward and analyzed at adulthood. The increases in body weight and leptin levels, but not glucose intolerance, induced by HFD were significantly augmented in the male, but not female, offspring from obese fathers. Paternal obesity caused a decrease in luteinizing hormone (LH) levels and exacerbated the drop in circulating testosterone and gene expression of its key biosynthetic enzymes caused by HFD in the male offspring. LH responses to central kisspeptin-10 administration were also suppressed in HFD males from obese fathers. In contrast, paternal obesity did not significantly alter gonadotropin levels in the female offspring fed HFD, although these females displayed reduced LH responses to kisspeptin-10. Our findings suggest that HFD-induced metabolic and reproductive disturbances are exacerbated by paternal obesity preferentially in males, whereas kisspeptin effects are affected in both sexes.
Collapse
Affiliation(s)
- Miguel Angel Sanchez-Garrido
- Instituto Maimónides de Investigación Biomédica de Cordoba, University of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Cordoba, Spain
| | - Francisco Ruiz-Pino
- Instituto Maimónides de Investigación Biomédica de Cordoba, University of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Cordoba, Spain
- Hospital Universitario Reina Sofia, Cordoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, Spain
| | - Inmaculada Velasco
- Instituto Maimónides de Investigación Biomédica de Cordoba, University of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Cordoba, Spain
| | - Alexia Barroso
- Instituto Maimónides de Investigación Biomédica de Cordoba, University of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Cordoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, Spain
| | - Daniela Fernandois
- Instituto Maimónides de Investigación Biomédica de Cordoba, University of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Cordoba, Spain
| | - Violeta Heras
- Instituto Maimónides de Investigación Biomédica de Cordoba, University of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Cordoba, Spain
| | - Maria Manfredi-Lozano
- Instituto Maimónides de Investigación Biomédica de Cordoba, University of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Cordoba, Spain
| | - Maria Jesus Vazquez
- Instituto Maimónides de Investigación Biomédica de Cordoba, University of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Cordoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, Spain
| | - Juan Manuel Castellano
- Instituto Maimónides de Investigación Biomédica de Cordoba, University of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Cordoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, Spain
| | - Juan Roa
- Instituto Maimónides de Investigación Biomédica de Cordoba, University of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Cordoba, Spain
- Hospital Universitario Reina Sofia, Cordoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, Spain
| | - Leonor Pinilla
- Instituto Maimónides de Investigación Biomédica de Cordoba, University of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Cordoba, Spain
- Hospital Universitario Reina Sofia, Cordoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, Spain
| | - Manuel Tena-Sempere
- Instituto Maimónides de Investigación Biomédica de Cordoba, University of Cordoba, Cordoba, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Cordoba, Cordoba, Spain
- Hospital Universitario Reina Sofia, Cordoba, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, Spain
| |
Collapse
|
41
|
Kereliuk SM, Brawerman GM, Dolinsky VW. Maternal Macronutrient Consumption and the Developmental Origins of Metabolic Disease in the Offspring. Int J Mol Sci 2017; 18:E1451. [PMID: 28684678 PMCID: PMC5535942 DOI: 10.3390/ijms18071451] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 06/14/2017] [Accepted: 06/30/2017] [Indexed: 12/18/2022] Open
Abstract
Recent research aimed at understanding the rise in obesity and cardiometabolic disease in children suggests that suboptimal maternal nutrition conditions organ systems and physiological responses in the offspring contributing to disease development. Understanding the mechanisms by which the macronutrient composition of the maternal diet during pregnancy or lactation affects health outcomes in the offspring may lead to new maternal nutrition recommendations, disease prevention strategies and therapies that reduce the increasing incidence of cardiometabolic disease in children. Recent mechanistic animal model research has identified how excess fats and sugars in the maternal diet alter offspring glucose tolerance, insulin signaling and metabolism. Maternal nutrition appears to influence epigenetic alterations in the offspring and the programming of gene expression in key metabolic pathways. This review is focused on experimental studies in animal models that have investigated mechanisms of how maternal consumption of macronutrients affects cardiometabolic disease development in the offspring. Future research using "-omic" technologies is essential to elucidate the mechanisms of how altered maternal macronutrient consumption influences the development of disease in the offspring.
Collapse
Affiliation(s)
- Stephanie M Kereliuk
- Department of Pharmacology & Therapeutics, University of Manitoba, Winnipeg, MB R3E 3P4, Canada.
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research Theme of the Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 3P4, Canada.
| | - Gabriel M Brawerman
- Department of Pharmacology & Therapeutics, University of Manitoba, Winnipeg, MB R3E 3P4, Canada.
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research Theme of the Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 3P4, Canada.
| | - Vernon W Dolinsky
- Department of Pharmacology & Therapeutics, University of Manitoba, Winnipeg, MB R3E 3P4, Canada.
- Diabetes Research Envisioned and Accomplished in Manitoba (DREAM) Research Theme of the Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 3P4, Canada.
| |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW Crohn's disease and ulcerative colitis, the two major forms of inflammatory bowel disease (IBD), represent chronic diseases of unknown cause, and they are regarded as prototypical complex diseases. Despite all the recent advances, a complete appreciation of the pathogenesis of IBD is still limited. In this review, we present recent information contributing to a better understanding of mechanisms underlying IBD. RECENT FINDINGS Here, we attempt to highlight novel environmental triggers, data on the gut microbiota, its interaction with the host, and the potential influence of diet and food components. We discuss recent findings on defective signaling pathways and the potential effects on the immune response, and we present new data on epigenetic changes, inflammasome, and damage-associated molecular patterns associated with IBD. SUMMARY The continuing identification of several epigenetic, transcriptomic, proteomic, and metabolomic alterations in patients with IBD reflects the complex nature of the disease and suggests the need for innovative approaches such as systems biology for identifying novel relevant targets in IBD.
Collapse
Affiliation(s)
- Heitor S P de Souza
- aServiço de Gastroenterologia e Laboratório Multidisciplinar de Pesquisa, Departamento de Clínica Médica, Hospital Universitário Clementino Fraga Filho, Universidade Federal do Rio de Janeiro bD'Or Institute for Research and Education (IDOR), Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
43
|
Pedroso AP, Souza AP, Dornellas APS, Oyama LM, Nascimento CMO, Santos GMS, Rosa JC, Bertolla RP, Klawitter J, Christians U, Tashima AK, Ribeiro EB. Intrauterine Growth Restriction Programs the Hypothalamus of Adult Male Rats: Integrated Analysis of Proteomic and Metabolomic Data. J Proteome Res 2017; 16:1515-1525. [PMID: 28314371 DOI: 10.1021/acs.jproteome.6b00923] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Programming of hypothalamic functions regulating energy homeostasis may play a role in intrauterine growth restriction (IUGR)-induced adulthood obesity. The present study investigated the effects of IUGR on the hypothalamus proteome and metabolome of adult rats submitted to 50% protein-energy restriction throughout pregnancy. Proteomic and metabolomic analyzes were performed by data independent acquisition mass spectrometry and multiple reaction monitoring, respectively. At age 4 months, the restricted rats showed elevated adiposity, increased leptin and signs of insulin resistance. 1356 proteins were identified and 348 quantified while 127 metabolites were quantified. The restricted hypothalamus showed down-regulation of 36 proteins and 5 metabolites and up-regulation of 21 proteins and 9 metabolites. Integrated pathway analysis of the proteomics and metabolomics data indicated impairment of hypothalamic glucose metabolism, increased flux through the hexosamine pathway, deregulation of TCA cycle and the respiratory chain, and alterations in glutathione metabolism. The data suggest IUGR modulation of energy metabolism and redox homeostasis in the hypothalamus of male adult rats. The present results indicated deleterious consequences of IUGR on hypothalamic pathways involved in pivotal physiological functions. These results provide guidance for future mechanistic studies assessing the role of intrauterine malnutrition in the development of metabolic diseases later in life.
Collapse
Affiliation(s)
- Amanda P Pedroso
- Department of Physiology, Universidade Federal de São Paulo UNIFESP , São Paulo, SP 04021-001, Brazil
| | - Adriana P Souza
- Department of Physiology, Universidade Federal de São Paulo UNIFESP , São Paulo, SP 04021-001, Brazil
| | - Ana P S Dornellas
- Department of Physiology, Universidade Federal de São Paulo UNIFESP , São Paulo, SP 04021-001, Brazil
| | - Lila M Oyama
- Department of Physiology, Universidade Federal de São Paulo UNIFESP , São Paulo, SP 04021-001, Brazil
| | - Cláudia M O Nascimento
- Department of Physiology, Universidade Federal de São Paulo UNIFESP , São Paulo, SP 04021-001, Brazil
| | - Gianni M S Santos
- Division of Applied Statistics, Universidade Federal de São Paulo UNIFESP , São Paulo, SP 04021-001, Brazil
| | - José C Rosa
- Protein Chemistry Center, Department of Molecular and Cell Biology, Ribeirão Preto Medical School, Universidade de São Paulo , Ribeirão Preto, SP 03178-200, Brazil
| | - Ricardo P Bertolla
- Department of Surgery, Universidade Federal de São Paulo UNIFESP , São Paulo, SP 04021-001, Brazil
| | - Jelena Klawitter
- iC42 Clinical Research and Development, Department of Anesthesiology, University of Colorado Denver , Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Uwe Christians
- iC42 Clinical Research and Development, Department of Anesthesiology, University of Colorado Denver , Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Alexandre K Tashima
- Department of Biochemistry, Universidade Federal de São Paulo UNIFESP , São Paulo, SP 04021-001, Brazil
| | - Eliane B Ribeiro
- Department of Physiology, Universidade Federal de São Paulo UNIFESP , São Paulo, SP 04021-001, Brazil
| |
Collapse
|
44
|
Reynolds CM, Segovia SA, Vickers MH. Experimental Models of Maternal Obesity and Neuroendocrine Programming of Metabolic Disorders in Offspring. Front Endocrinol (Lausanne) 2017; 8:245. [PMID: 28993758 PMCID: PMC5622157 DOI: 10.3389/fendo.2017.00245] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 09/06/2017] [Indexed: 12/17/2022] Open
Abstract
Evidence from epidemiological, clinical, and experimental studies have clearly shown that disease risk in later life is increased following a poor early life environment, a process preferentially termed developmental programming. In particular, this work clearly highlights the importance of the nutritional environment during early development with alterations in maternal nutrition, including both under- and overnutrition, increasing the risk for a range of cardiometabolic and neurobehavioral disorders in adult offspring characterized by both adipokine resistance and obesity. Although the mechanistic basis for such developmental programming is not yet fully defined, a common feature derived from experimental animal models is that of alterations in the wiring of the neuroendocrine pathways that control energy balance and appetite regulation during early stages of developmental plasticity. The adipokine leptin has also received significant attention with clear experimental evidence that normal regulation of leptin levels during the early life period is critical for the normal development of tissues and related signaling pathways that are involved in metabolic and cardiovascular homeostasis. There is also increasing evidence that alterations in the epigenome and other underlying mechanisms including an altered gut-brain axis may contribute to lasting cardiometabolic dysfunction in offspring. Ongoing studies that further define the mechanisms between these associations will allow for identification of early risk markers and implementation of strategies around interventions that will have obvious beneficial implications in breaking a programmed transgenerational cycle of metabolic disorders.
Collapse
Affiliation(s)
| | | | - Mark H. Vickers
- Liggins Institute, University of Auckland, Auckland, New Zealand
- *Correspondence: Mark H. Vickers,
| |
Collapse
|