1
|
Engin A. The Unrestrained Overeating Behavior and Clinical Perspective. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:167-198. [PMID: 39287852 DOI: 10.1007/978-3-031-63657-8_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Obesity-related co-morbidities decrease life quality, reduce working ability, and lead to early death. In the adult population, eating addiction manifests with excessive food consumption and the unrestrained overeating behavior, which is associated with increased risk of morbidity and mortality and defined as the binge eating disorder (BED). This hedonic intake is correlated with fat preference and the total amount of dietary fat consumption is the most potent risk factor for weight gain. Long-term BED leads to greater sensitivity to the rewarding effects of palatable foods and results in obesity fatefully. Increased plasma concentrations of non-esterified free fatty acids and lipid-overloaded hypertrophic adipocytes may cause insulin resistance. In addition to dietary intake of high-fat diet, sedentary lifestyle leads to increased storage of triglycerides not only in adipose tissue but also ectopically in other tissues. Lipid-induced apoptosis, ceramide accumulation, reactive oxygen species overproduction, endoplasmic reticulum stress, and mitochondrial dysfunction play role in the pathogenesis of lipotoxicity. Food addiction and BED originate from complex action of dopaminergic, opioid, and cannabinoid systems. BED may also be associated with both obesity and major depressive disorder. For preventing morbidity and mortality, as well as decreasing the impact of obesity-related comorbidities in appropriately selected patients, opiate receptor antagonists and antidepressant combination are recommended. Pharmacotherapy alongside behavioral management improves quality of life and reduces the obesity risk; however, the number of licensed drugs is very few. Thus, stereotactic treatment is recommended to break down the refractory obesity and binge eating in obese patient. As recent applications in the field of non-invasive neuromodulation, transcranial magnetic stimulation and transcranial direct current stimulation are thought to be important in image-guided deep brain stimulation in humans. Chronic overnutrition most likely provides repetitive and persistent signals that up-regulate inhibitor of nuclear factor kappa B (NF-κB) kinase beta subunit/NF-κB (IKKβ/NF-κB) in the hypothalamus before the onset of obesity. However, how the mechanisms of high-fat diet-induced peripheral signals affect the hypothalamic arcuate nucleus remain largely unknown.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
2
|
Sharma S, Bhonde R. Dilemma of Epigenetic Changes Causing or Reducing Metabolic Disorders in Offsprings of Obese Mothers. Horm Metab Res 2023; 55:665-676. [PMID: 37813098 DOI: 10.1055/a-2159-9128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/11/2023]
Abstract
Maternal obesity is associated with fetal complications predisposing later to the development of metabolic syndrome during childhood and adult stages. High-fat diet seems to influence individuals and their subsequent generations in mediating weight gain, insulin resistance, obesity, high cholesterol, diabetes, and cardiovascular disorder. Research evidence strongly suggests that epigenetic alteration is the major contributor to the development of metabolic syndrome through DNA methylation, histone modifications, and microRNA expression. In this review, we have discussed the outcome of recent studies on the adverse and beneficial effects of nutrients and vitamins through epigenetics during pregnancy. We have further discussed about the miRNAs altered during maternal obesity. Identification of new epigenetic modifiers such as mesenchymal stem cells condition media (MSCs-CM)/exosomes for accelerating the reversal of epigenetic abnormalities for the development of new treatments is yet another aspect of the present review.
Collapse
Affiliation(s)
- Shikha Sharma
- Institute for Stem Cell Science and Regenerative Medicine, Bangalore, India
| | - Ramesh Bhonde
- Stem Cells and Regenerative Medicine, Dr. D. Y. Patil Vidyapeeth Pune (Deemed University), Pune, India
| |
Collapse
|
3
|
He Y, Liu Y, Guan P, He L, Zhou X. Serine Administration Improves Selenium Status, Oxidative Stress, and Mitochondrial Function in Longissimus Dorsi Muscle of Piglets with Intrauterine Growth Retardation. Biol Trace Elem Res 2023; 201:1740-1747. [PMID: 35661959 DOI: 10.1007/s12011-022-03304-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/26/2022] [Indexed: 01/16/2023]
Abstract
Intrauterine growth retardation (IUGR) causes oxidative stress in the skeletal muscle. Serine and selenoproteins are involved in anti-oxidative processes; however, whether IUGR affects selenium status and whether serine has beneficial effects remain elusive. Here, we investigated the effects of serine administration on selenium nutritional status and oxidative stress in the longissimus dorsi muscle of piglets with IUGR. Six newborn Min piglets having normal birth weight were administered saline, and 12 IUGR piglets were either administered saline or 0.8% serine. The results showed a lower selenium content in skeletal muscle in IUGR piglets, which was restored after serine administration. IUGR piglets showed a disturbed expression of genes encoding selenoproteins, with decreased expression of GPX2, GPX4, TXNRD1, and TXNRD3 and increased expression of DIO1, DIO2, SELF, SELM, SELP, and SELW. Notably, serine administration restored the expression levels of these genes. In accordance with the changes in gene expression, the activity of GPX, TXNRD, and DIO and the content of GSH and SELP were also altered, whereas serine administration restored their contents and activities. Moreover, we observed severe oxidative stress in the skeletal muscle of IUGR piglets, as indicated by decreased GSH content and increased MDA and PC content, whereas serine administration alleviated these changes. In conclusion, our results indicate that IUGR piglets showed a disturbed expression of genes encoding selenoproteins, accompanied by severe oxidative stress. Serine administration can improve selenium status, oxidative stress, and mitochondrial function in the longissimus dorsi muscle of piglets with IUGR. These results suggest that serine could potentially be used in the treatment of IUGR in piglets.
Collapse
Affiliation(s)
- Yiwen He
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, 410125, China
| | - Yonghui Liu
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, 410125, China
| | - Peng Guan
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, 410125, China
| | - Liuqin He
- Hunan Provincial Key Laboratory of Animal Intestinal Function and Regulation, College of Life Sciences, Hunan Normal University, Changsha, 410081, China
| | - Xihong Zhou
- Key Laboratory of Agro-Ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, The Chinese Academy of Sciences, Changsha, 410125, China.
| |
Collapse
|
4
|
Shamardl HAMA, Ibrahim NA, Merzeban DH, Elamir AM, Golam RM, Elsayed AM. Resveratrol and Dulaglutide ameliorate adiposity and liver dysfunction in rats with diet-induced metabolic syndrome: Role of SIRT-1 / adipokines / PPARγ and IGF-1. Daru 2023:10.1007/s40199-023-00458-y. [PMID: 36991247 DOI: 10.1007/s40199-023-00458-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 03/05/2023] [Indexed: 03/31/2023] Open
Abstract
BACKGROUND Adiposity and non-alcoholic fatty liver disease (NAFLD) are common characteristics of metabolic syndrome (MS). Understanding the underlying pathogenesis is crucial for the development of new remedies. Resveratrol controls obesity and glycemic disorders in patients with MS. OBJECTIVES This study aimed to evaluate the effect of resveratrol and dulaglutide on adipose tissues and liver in rats with MS, declaring their possible mechanisms. METHODS Rats allocated as Control, MS (induced by a high fat/ high sucrose diet for eight weeks), MS + Resveratrol (30 mg/kg/day orally), and MS + Dulaglutide (0.6 mg/kg twice weekly SC); drugs administration was in the last four weeks. Serum biochemical measurements were done. Liver and visceral fat were processed for biochemistry, histopathology, and immunohistochemistry. RESULTS MS results demonstrated significantly increased systolic and diastolic blood pressure, anthropometric measurements, serum levels of alanine aminotransferase (ALT), glycemic indices, and lipids with decreased HDL-C. Tissue levels of leptin, malondialdehyde (MDA), and TNF-α reactivity significantly increased. Expression of adiponectin, PPARγ, and insulin growth factor-1 (IGF-1) decreased. Also, Western blotting mRNA gene expression of liver SIRT-1 was down-regulated. Resveratrol and dulaglutide significantly and effectively reversed MS complexity, ameliorating all findings, particularly NAFLD and adiposity-induced inflammation. Resveratrol significantly appears superior to dulaglutide regarding the effects on hemodynamics, lipids, adipokines, IGF-1 levels, and adipocyte size. Parallel, dulaglutide has more influence on glycemic control. CONCLUSION Protective effects of the drugs may be through correlations between SIRT-1/adipokines/IGF-1 and PPARγ, improving the cross-talk between insulin resistance, obesity markers, liver dysfunction, and TNF-α. Promising multi-beneficial therapies of resveratrol or dulaglutide in MS are recommended clinically for this purpose. Showing the Experimental Design.
Collapse
Affiliation(s)
| | - Noha A Ibrahim
- Histology and Cell Biology Department, Faculty of Medicine, Fayoum University, Fayoum, 19052, Egypt
| | - Dina H Merzeban
- Medical Physiology DepartmentFaculty of Medicine, Fayoum University, Fayoum, 19052, Egypt
| | - Azza M Elamir
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Fayoum University, Fayoum, 19052, Egypt
| | - Rehab M Golam
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Fayoum University, Fayoum, 19052, Egypt
| | - Asmaa M Elsayed
- Histology and Cell Biology Department, Faculty of Medicine, Fayoum University, Fayoum, 19052, Egypt
| |
Collapse
|
5
|
Stemness of Normal and Cancer Cells: The Influence of Methionine Needs and SIRT1/PGC-1α/PPAR-α Players. Cells 2022; 11:cells11223607. [PMID: 36429035 PMCID: PMC9688847 DOI: 10.3390/cells11223607] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/09/2022] [Accepted: 11/09/2022] [Indexed: 11/16/2022] Open
Abstract
Stem cells are a population of undifferentiated cells with self-renewal and differentiation capacities. Normal and cancer stem cells share similar characteristics in relation to their stemness properties. One-carbon metabolism (OCM), a network of interconnected reactions, plays an important role in this dependence through its role in the endogenous synthesis of methionine and S-adenosylmethionine (SAM), the universal donor of methyl groups in eukaryotic cells. OCM genes are differentially expressed in stem cells, compared to their differentiated counterparts. Furthermore, cultivating stem cells in methionine-restricted conditions hinders their stemness capacities through decreased SAM levels with a subsequent decrease in histone methylation, notably H3K4me3, with a decrease in stem cell markers. Stem cells' reliance on methionine is linked to several mechanisms, including high methionine flux or low endogenous methionine biosynthesis. In this review, we provide an overview of the recent discoveries concerning this metabolic dependence and we discuss the mechanisms behind them. We highlight the influence of SIRT1 on SAM synthesis and suggest a role of PGC-1α/PPAR-α in impaired stemness produced by methionine deprivation. In addition, we discuss the potential interest of methionine restriction in regenerative medicine and cancer treatment.
Collapse
|
6
|
Rachakatla A, Kalashikam RR. Calorie Restriction-Regulated Molecular Pathways and Its Impact on Various Age Groups: An Overview. DNA Cell Biol 2022; 41:459-468. [PMID: 35451872 DOI: 10.1089/dna.2021.0922] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Calorie restriction (CR) if planned properly with regular exercise at different ages can result in healthy weight loss. CR can also have different beneficial effects on improving lifespan and decreasing the age-associated diseases by regulating physiological, biochemical, and molecular markers. The different pathways regulated by CR include:(1) AMP-activated protein kinase (AMPK), which involves PGC-1α, SIRT1, and SIRT3. AMPK also effects myocyte enhancer factor 2 (MEF2), peroxisome proliferator-activated receptor delta, and peroxisome proliferator-activated receptor alpha, which are involved in mitochondrial biogenesis and lipid oxidation; (2) Forkhead box transcription factor's signaling is related to the DNA repair, lipid metabolism, protection of protein structure, autophagy, and resistance to oxidative stress; (3) Mammalian target of rapamycin (mTOR) signaling, which involves key factors, such as S6 protein kinase-1 (S6K1), mTOR complex-1 (mTORC1), and 4E-binding protein (4E-BP). Under CR conditions, AMPK activation and mTOR inhibition helps in the activation of Ulk1 complex along with the acetyltransferase Mec-17, which is necessary for autophagy; (4) Insulin-like growth factor-1 (IGF-1) pathway downregulation protects against cancer and slows the aging process; (5) Nuclear factor kappa B pathway downregulation decreases the inflammation; and (6) c-Jun N-terminal kinase and p38 kinase regulation as a response to the stress. The acute and chronic CR both shows antidepression and anxiolytic action by effecting ghrelin/GHS-R1a signaling. CR also regulates GSK3β kinase and protects against age-related brain atrophy. CR at young age may show many deleterious effects by effecting different mechanisms. Parental CR before or during conception will also affect the health and development of the offspring by causing many epigenetic modifications that show transgenerational transmission. Maternal CR is associated with intrauterine growth retardation effecting the offspring in their adulthood by developing different metabolic syndromes. The epigenetic changes with response to paternal food supply also linked to offspring health. CR at middle and old age provides a significant preventive impact against the development of age-associated diseases.
Collapse
|
7
|
Liu N, Li X, Wang Y, Zhu B, Tian Y, Lang J, Yang J. Photocatalyst prepared by NiCo2O4/CNQDs modified carbon fabric heterojunctions enhanced visible-light-driven photocatalytic degradation of Methyl Orange. CrystEngComm 2022. [DOI: 10.1039/d2ce00183g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
As this point, A novel photocatalyst was reported by us, cobalt nickel tetroxide (NiCo2O4)/g-C3N4 quantum dots (CNQDs) heterojunctions on carbon cloth (CC). NiCo2O4 nanosheets and NiCo2O4/CNQDs were grown on carbon...
Collapse
|
8
|
Zhang H, Zheng P, Chen D, Yu B, He J, Mao X, Yu J, Luo Y, Luo J, Huang Z, Yan H. Dietary Arginine Supplementation Improves Intestinal Mitochondrial Functions in Low-Birth-Weight Piglets but Not in Normal-Birth-Weight Piglets. Antioxidants (Basel) 2021; 10:antiox10121995. [PMID: 34943098 PMCID: PMC8698761 DOI: 10.3390/antiox10121995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 12/16/2022] Open
Abstract
Our previous studies revealed that L-arginine supplementation had beneficial effects on intestinal barrier functions of low-birth-weight (LBW) piglets, which were associated with the enhanced antioxidant capacity. Moreover, mitochondrial functions are closely related to the redox state. This study was to explore potential mechanisms of L-arginine-induced beneficial effects against intestinal dysfunction by regulating mitochondrial function of LBW piglets. Twenty 4-day-old normal birth weight (NBW) piglets (BW: 2.08 ± 0.09 kg) and 20 LBW siblings (BW: 1.16 ± 0.07 kg) were artificially fed either a basal diet or a basal diet supplemented with 1.0% L-arginine for 21 d, respectively. Growth performance, intestinal morphology, redox status, mitochondrial morphology, and mitochondrial functions were examined. Data were subjected to two-way analysis of variance. LBW piglets presented lower (p < 0.05) ADG, shorter (p < 0.05) intestinal villus height, lower (p < 0.05) jejunal adenosine triphosphate (ATP) content and higher (p < 0.05) concentrations of Ca2+ and 8-OH-dG in jejunal mitochondria, compared with NBW piglets. Supplementation with 1.0% L-arginine significantly increased (p < 0.05) ADG, the activities of CAT, SOD, and GPx, intestinal villus height and mRNA abundances of ZO-1 (2-fold) in the jejunum of LBW piglets, but not in NBW piglets. Furthermore, the concentrations of ATP and the transcription of COX IV, COX V genes were up-regulated (p < 0.05) and the concentration of Ca2+ and 8-OH-dG were decreased (p < 0.05) in arginine-treated LBW piglets. The results suggest that mitochondrial morphology is affected, and mitochondrial functions are impaired in the jejunum of LBW piglets. While supplementation with 1.0% L-arginine relieved intestinal dysfunction through enhancing antioxidant capacity and improving mitochondrial functions via repairing mitochondrial morphology, normalizing mitochondrial calcium, and increasing ATP concentration in the jejunum of LBW piglets. However, supplementation with L-arginine has no significant beneficial effects on intestinal health in NBW piglets.
Collapse
Affiliation(s)
- Hao Zhang
- Animal Nutrition Insititute, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (D.C.); (B.Y.); (J.H.); (X.M.); (J.Y.); (Y.L.); (J.L.); (Z.H.); (H.Y.)
| | - Ping Zheng
- Animal Nutrition Insititute, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (D.C.); (B.Y.); (J.H.); (X.M.); (J.Y.); (Y.L.); (J.L.); (Z.H.); (H.Y.)
- Key Laboratory for Animal Disease-Resistance Nutrition, China Ministry of Education, Chengdu 611130, China
- Correspondence: ; Tel.: +86-028-86290922
| | - Daiwen Chen
- Animal Nutrition Insititute, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (D.C.); (B.Y.); (J.H.); (X.M.); (J.Y.); (Y.L.); (J.L.); (Z.H.); (H.Y.)
- Key Laboratory for Animal Disease-Resistance Nutrition, China Ministry of Education, Chengdu 611130, China
| | - Bing Yu
- Animal Nutrition Insititute, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (D.C.); (B.Y.); (J.H.); (X.M.); (J.Y.); (Y.L.); (J.L.); (Z.H.); (H.Y.)
- Key Laboratory for Animal Disease-Resistance Nutrition, China Ministry of Education, Chengdu 611130, China
| | - Jun He
- Animal Nutrition Insititute, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (D.C.); (B.Y.); (J.H.); (X.M.); (J.Y.); (Y.L.); (J.L.); (Z.H.); (H.Y.)
- Key Laboratory for Animal Disease-Resistance Nutrition, China Ministry of Education, Chengdu 611130, China
| | - Xiangbing Mao
- Animal Nutrition Insititute, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (D.C.); (B.Y.); (J.H.); (X.M.); (J.Y.); (Y.L.); (J.L.); (Z.H.); (H.Y.)
- Key Laboratory for Animal Disease-Resistance Nutrition, China Ministry of Education, Chengdu 611130, China
| | - Jie Yu
- Animal Nutrition Insititute, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (D.C.); (B.Y.); (J.H.); (X.M.); (J.Y.); (Y.L.); (J.L.); (Z.H.); (H.Y.)
- Key Laboratory for Animal Disease-Resistance Nutrition, China Ministry of Education, Chengdu 611130, China
| | - Yuheng Luo
- Animal Nutrition Insititute, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (D.C.); (B.Y.); (J.H.); (X.M.); (J.Y.); (Y.L.); (J.L.); (Z.H.); (H.Y.)
- Key Laboratory for Animal Disease-Resistance Nutrition, China Ministry of Education, Chengdu 611130, China
| | - Junqiu Luo
- Animal Nutrition Insititute, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (D.C.); (B.Y.); (J.H.); (X.M.); (J.Y.); (Y.L.); (J.L.); (Z.H.); (H.Y.)
- Key Laboratory for Animal Disease-Resistance Nutrition, China Ministry of Education, Chengdu 611130, China
| | - Zhiqing Huang
- Animal Nutrition Insititute, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (D.C.); (B.Y.); (J.H.); (X.M.); (J.Y.); (Y.L.); (J.L.); (Z.H.); (H.Y.)
| | - Hui Yan
- Animal Nutrition Insititute, Sichuan Agricultural University, Chengdu 611130, China; (H.Z.); (D.C.); (B.Y.); (J.H.); (X.M.); (J.Y.); (Y.L.); (J.L.); (Z.H.); (H.Y.)
| |
Collapse
|
9
|
Zhang Y, Yuan H, Peng M, Hu Z, Fan Z, Xu J, He L, Wang Y, Wang W, Su Y, Liu C, Zhang H, Zhao K. Folic acid deficiency damages male reproduction via endoplasmic reticulum stress-associated PERK pathway induced by Caveolin-1 in mice. Syst Biol Reprod Med 2021; 67:383-394. [PMID: 34474604 DOI: 10.1080/19396368.2021.1954724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Folic acid is critical to maintaining normal male reproductive function. Endoplasmic reticulum (ER) stress plays a crucial role in folic acid deficiency. Studies have shown that Caveolin-1 (Cav-1) is involved in ER stress, but the specific mechanism in male reproduction is still unclear. This study aimed to investigate the effects of folic acid deficiency on spermatogenesis and elucidate the underlying mechanisms. C57BL/6 mice fed with folic acid deficiency induced diet(0.3 mg/kg) were used. A significant decrease in the sperm concentration in the folic acid deficiency group was observed. Meanwhile, folic acid deficiency decreased Cav-1 expression in the testis tissue and increased endoplasmic reticulum stress-related PERK, eIF2α, ATF4, CHOP gene expression. Our results suggest that folic acid deficiency can affect male reproduction through the Cav-1-PERK-eIFα-ATF4-CHOP pathway.Abbreviations: ATF4: activating transcription factor 4; Ca2+: calcium ion; Cav-1: Caveolin-1; CCK-8: cell counting kit-8; CHOP: CCAAT-enhancer-binding protein homologous protein; DNA: Deoxyribonucleic acid; DSB: double strand breakage; eIF2α: eukaryotic Initiation Factor 2 alpha; ER: endoplasmic reticulum; FD: folic acid deficiency; FITC: fluorescein isothiocyanate; HE: hematoxylin and eosin; H3K4me3: histone H3 lysine 4 trimethylation; PERK: protein kinase RNA-like endoplasmic reticulum kinase; PI: propidium iodide; RT-qPCR: quantitative reverse transcription PCR; TUNEL: TdT mediated dUTP Nick End Labeling.
Collapse
Affiliation(s)
- Yuan Zhang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongfang Yuan
- Department of Obstetrics And Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Meilin Peng
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhiyong Hu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zunpan Fan
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Xu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liting He
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yongfeng Wang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Wang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yufang Su
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chunyan Liu
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huiping Zhang
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kai Zhao
- Institute of Reproductive Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
10
|
Li Z, Kosgei VJ, Bison A, Alberto JM, Umoret R, Maskali F, Brunaud L, Guéant JL, Guéant-Rodriguez RM. Programming by Methyl Donor Deficiency during Pregnancy and Lactation Produces Cardiomyopathy in Adult Rats Subjected to High Fat Diet. Mol Nutr Food Res 2021; 65:e2100065. [PMID: 33991387 DOI: 10.1002/mnfr.202100065] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/12/2021] [Indexed: 12/15/2022]
Abstract
SCOPE Vitamin B12 and folate (methyl donors) deficiency is frequent during pregnancy. Experimental rat models with methyl donor deficit during pregnancy and lactation (Initial methyl donor deficit (iMDD)) produce impaired myocardium fatty acid oxidation and mitochondrial energy metabolism at weaning. METHODS AND RESULTS The consequences of iMDD on heart of rat pups under normal diet after weaning and high fat diet (HF) between day (D) 50 and D185 are investigated. iMDD/HF induces increased histological fibrosis and increased B-type natriuretic peptide blood level. Inflammation is evidenced by increased protein expression of NFkB, Caspase1, and IL1β and fibrosis by increased expression of αSMA, col1a1, and col1a2 in females, but not in males. Fibrosis is related to increased angiotensin at D50 and D185 and increased protein expression of TGFB1 and AT1 angiotensin receptors at D185. The limited fibrosis in males is consistent with increased expression of AT2, the antagonist receptor of AT1. The increased expression of GLUT4 and decreased expression of PGC1α and PPARα reflect a shift from fatty acid oxidation to glycolysis. CONCLUSION Developmental programming by iMDD produces cardiomyopathy in female offspring exposed to HF. The cardiomyopathy is linked to inflammation and fibrosis through angiotensin-AT2 and TGFB1 pathways and alteration of energy metabolism.
Collapse
Affiliation(s)
- Zhen Li
- UMR Inserm 1256 N-GERE (Nutrition, Génetique et Exposition aux Risques Environmentaux), Université de Lorraine, Vandoeuvre-lès-Nancy, 54500, France
| | - Viola J Kosgei
- UMR Inserm 1256 N-GERE (Nutrition, Génetique et Exposition aux Risques Environmentaux), Université de Lorraine, Vandoeuvre-lès-Nancy, 54500, France
| | - Anais Bison
- UMR Inserm 1256 N-GERE (Nutrition, Génetique et Exposition aux Risques Environmentaux), Université de Lorraine, Vandoeuvre-lès-Nancy, 54500, France
| | - Jean-Marc Alberto
- UMR Inserm 1256 N-GERE (Nutrition, Génetique et Exposition aux Risques Environmentaux), Université de Lorraine, Vandoeuvre-lès-Nancy, 54500, France
| | - Remi Umoret
- UMR Inserm 1256 N-GERE (Nutrition, Génetique et Exposition aux Risques Environmentaux), Université de Lorraine, Vandoeuvre-lès-Nancy, 54500, France
| | - Fatiha Maskali
- Nancyclotep-GIE, CHRU of Nancy, Rue du Morvan, Vandoeuve-Lès-Nancy, 54500, France
| | - Laurent Brunaud
- UMR Inserm 1256 N-GERE (Nutrition, Génetique et Exposition aux Risques Environmentaux), Université de Lorraine, Vandoeuvre-lès-Nancy, 54500, France
| | - Jean-Louis Guéant
- UMR Inserm 1256 N-GERE (Nutrition, Génetique et Exposition aux Risques Environmentaux), Université de Lorraine, Vandoeuvre-lès-Nancy, 54500, France.,Departments of Digestive Diseases, Nutrition and Endocrinology and Molecular Medicine and National Center of Inborn Errors of Metabolism, University Hospital Center, Université de Lorraine, Vandoeuvre-lès-Nancy, 54500, France
| | - Rosa-Maria Guéant-Rodriguez
- UMR Inserm 1256 N-GERE (Nutrition, Génetique et Exposition aux Risques Environmentaux), Université de Lorraine, Vandoeuvre-lès-Nancy, 54500, France.,Departments of Digestive Diseases, Nutrition and Endocrinology and Molecular Medicine and National Center of Inborn Errors of Metabolism, University Hospital Center, Université de Lorraine, Vandoeuvre-lès-Nancy, 54500, France
| |
Collapse
|
11
|
Sarli PM, Manousopoulou A, Efthymiou E, Zouridis A, Potiris A, Pervanidou P, Panoulis K, Vlahos N, Deligeoroglou E, Garbis SD, Eleftheriades M. Liver Proteome Profile of Growth Restricted and Appropriately Grown Newborn Wistar Rats Associated With Maternal Undernutrition. Front Endocrinol (Lausanne) 2021; 12:684220. [PMID: 34127923 PMCID: PMC8195994 DOI: 10.3389/fendo.2021.684220] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/04/2021] [Indexed: 12/22/2022] Open
Abstract
Background Fetal growth restriction (FGR) has been associated with adverse perinatal outcomes and epigenetic modifications that impact gene expression leading to permanent changes of fetal metabolic pathways and thereby influence development of disease in childhood and adult life. In this study, we investigated the result of maternal food restriction on liver protein expression in Wistar male newborn pups. Materials & Methods Ten (n = 10) timed pregnant Wistar rats on their 14th day of gestation were randomly assigned to either control (n = 4) or food restricted group (n = 6). The control group had ad libitum access to food. In the food restricted group, maternal diet was limited in a moderate fashion (50%) from day 15 of pregnancy until delivery. All rats delivered spontaneously on day 21 and newborn pups were immediately weighed. Pups born to normally nourished mothers were considered as controls, while pups born to food restricted mothers were subdivided into two groups, based on their birth weight: growth restricted (FGR) and appropriately grown (non-FGR). Rats were euthanized immediately after birth and liver tissues of 11 randomly selected male offspring (FGR n = 4, non-FGR n = 4, control n = 3) were collected and analyzed using quantitative proteomics. Results In total 6,665 proteins were profiled. Of these, 451 and 751 were differentially expressed in FGR and non-FGR vs. control, respectively, whereas 229 proteins were commonly expressed. Bioinformatics analysis of the differentially expressed proteins (DEPs) in FGR vs. control revealed induction of the super-pathway of cholesterol biosynthesis and inhibition of thyroid hormone metabolism, fatty acid beta oxidation and apelin liver signaling pathway. Analysis of DEPs in non-FGR vs. control groups showed inhibition of thyroid hormone metabolism, fatty acid beta oxidation, and apelin liver signaling pathway. Conclusion This study demonstrates the impact of prenatal food restriction on the proteomic liver profile of FGR and non-FGR offspring underlying the importance of both prenatal adversities and birth weight on liver-dependent postnatal disease.
Collapse
Affiliation(s)
- Polyxeni-Maria Sarli
- Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Antigoni Manousopoulou
- Beckman Research Institute, City of Hope National Medical Center, Duarte, CA, United States
| | - Elias Efthymiou
- Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Andreas Zouridis
- Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasios Potiris
- Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiota Pervanidou
- First Department of Paediatrics, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Panoulis
- Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Nikolaos Vlahos
- Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Efthymios Deligeoroglou
- Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Spiros D. Garbis
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Makarios Eleftheriades
- Second Department of Obstetrics and Gynaecology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
12
|
Sirt1-PPARS Cross-Talk in Complex Metabolic Diseases and Inherited Disorders of the One Carbon Metabolism. Cells 2020; 9:cells9081882. [PMID: 32796716 PMCID: PMC7465293 DOI: 10.3390/cells9081882] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 08/07/2020] [Accepted: 08/07/2020] [Indexed: 12/15/2022] Open
Abstract
Sirtuin1 (Sirt1) has a NAD (+) binding domain and modulates the acetylation status of peroxisome proliferator-activated receptor-γ coactivator-1α (PGC1α) and Fork Head Box O1 transcription factor (Foxo1) according to the nutritional status. Sirt1 is decreased in obese patients and increased in weight loss. Its decreased expression explains part of the pathomechanisms of the metabolic syndrome, diabetes mellitus type 2 (DT2), cardiovascular diseases and nonalcoholic liver disease. Sirt1 plays an important role in the differentiation of adipocytes and in insulin signaling regulated by Foxo1 and phosphatidylinositol 3′-kinase (PI3K) signaling. Its overexpression attenuates inflammation and macrophage infiltration induced by a high fat diet. Its decreased expression plays a prominent role in the heart, liver and brain of rat as manifestations of fetal programming produced by deficit in vitamin B12 and folate during pregnancy and lactation through imbalanced methylation/acetylation of PGC1α and altered expression and methylation of nuclear receptors. The decreased expression of Sirt1 produced by impaired cellular availability of vitamin B12 results from endoplasmic reticulum stress through subcellular mislocalization of ELAVL1/HuR protein that shuttles Sirt1 mRNA between the nucleus and cytoplasm. Preclinical and clinical studies of Sirt1 agonists have produced contrasted results in the treatment of the metabolic syndrome. A preclinical study has produced promising results in the treatment of inherited disorders of vitamin B12 metabolism.
Collapse
|
13
|
Harb Z, Deckert V, Bressenot AM, Christov C, Guéant-Rodriguez RM, Raso J, Alberto JM, de Barros JPP, Umoret R, Peyrin-Biroulet L, Lagrost L, Bronowicki JP, Guéant JL. The deficit in folate and vitamin B12 triggers liver macrovesicular steatosis and inflammation in rats with dextran sodium sulfate-induced colitis. J Nutr Biochem 2020; 84:108415. [PMID: 32645655 DOI: 10.1016/j.jnutbio.2020.108415] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 03/21/2020] [Accepted: 05/02/2020] [Indexed: 02/06/2023]
Abstract
The risks of nonalcoholic steatohepatitis (NASH) and deficiency in vitamin B12 and folate (methyl donor deficiency, MDD) are increased in inflammatory bowel disease (IBD). We investigated the influence of MDD on NASH in rats with DSS-induced colitis. Two-month-old male Wistar rats were subjected to MDD diet and/or ingestion of DSS and compared to control animals. We studied steatosis, inflammation, fibrosis, plasma levels of metabolic markers, cytokines and lipopolysaccharide, and inflammatory pathways in liver. MDD triggered a severe macrovesicular steatosis with inflammation in DSS animals that was not observed in animals subjected to DSS or MDD only. The macrovesicular steatosis was closely correlated to folate, vitamin B12, homocysteine plasma level and liver S-adenosyl methionine/S-adenosyl homocysteine (SAM/SAH) ratio. Liver inflammation was evidenced by activation of nuclear factor kappa B (NFκB) pathway and nuclear translocation of NFκB phospho-p65. MDD worsened the increase of interleukin 1-beta (IL-1β) and abolished the increase of IL10 produced by DSS colitis. It increased monocyte chemoattractant protein 1 (MCP-1). MDD triggers liver macrovesicular steatosis and inflammation through imbalanced expression of IL-1β vs. IL10 and increase of MCP-1 in DSS colitis. Our results suggest evaluating whether IBD patients with MDD and increase of MCP-1 are at higher risk of NASH.
Collapse
Affiliation(s)
- Zeinab Harb
- INSERM U1256, Nutrition Génétique et Exposition aux Risques Environnementaux, Medical Faculty, University of Lorraine and Regional University Hospital Center of Nancy, Vandœuvre les Nancy, France
| | - Valérie Deckert
- INSERM UMR1231 Lipides, Nutrition, Cancer, University of Bourgogne Franche-Comté, LipSTIC LabEx, Dijon, France
| | - Aude Marchal Bressenot
- INSERM U1256, Nutrition Génétique et Exposition aux Risques Environnementaux, Medical Faculty, University of Lorraine and Regional University Hospital Center of Nancy, Vandœuvre les Nancy, France; Division of Anatomo-Pathology, Robert Debré University Hospital, Reims
| | - Christo Christov
- INSERM U1256, Nutrition Génétique et Exposition aux Risques Environnementaux, Medical Faculty, University of Lorraine and Regional University Hospital Center of Nancy, Vandœuvre les Nancy, France
| | - Rosa-Maria Guéant-Rodriguez
- INSERM U1256, Nutrition Génétique et Exposition aux Risques Environnementaux, Medical Faculty, University of Lorraine and Regional University Hospital Center of Nancy, Vandœuvre les Nancy, France; Biochemical and Molecular biology lab, Regional University Hospital Center of Nancy, Vandoeuvre les Nancy, France
| | - Jérémie Raso
- INSERM U1256, Nutrition Génétique et Exposition aux Risques Environnementaux, Medical Faculty, University of Lorraine and Regional University Hospital Center of Nancy, Vandœuvre les Nancy, France
| | - Jean Marc Alberto
- INSERM U1256, Nutrition Génétique et Exposition aux Risques Environnementaux, Medical Faculty, University of Lorraine and Regional University Hospital Center of Nancy, Vandœuvre les Nancy, France
| | - Jean-Paul Pais de Barros
- INSERM UMR1231 Lipides, Nutrition, Cancer, University of Bourgogne Franche-Comté, LipSTIC LabEx, Dijon, France
| | - Remy Umoret
- INSERM U1256, Nutrition Génétique et Exposition aux Risques Environnementaux, Medical Faculty, University of Lorraine and Regional University Hospital Center of Nancy, Vandœuvre les Nancy, France
| | - Laurent Peyrin-Biroulet
- INSERM U1256, Nutrition Génétique et Exposition aux Risques Environnementaux, Medical Faculty, University of Lorraine and Regional University Hospital Center of Nancy, Vandœuvre les Nancy, France; Division of Hepatogastroenterology, Regional University Hospital Center of Nancy, Vandoeuvre les Nancy, France
| | - Laurent Lagrost
- INSERM UMR1231 Lipides, Nutrition, Cancer, University of Bourgogne Franche-Comté, LipSTIC LabEx, Dijon, France
| | - Jean-Pierre Bronowicki
- INSERM U1256, Nutrition Génétique et Exposition aux Risques Environnementaux, Medical Faculty, University of Lorraine and Regional University Hospital Center of Nancy, Vandœuvre les Nancy, France; Division of Hepatogastroenterology, Regional University Hospital Center of Nancy, Vandoeuvre les Nancy, France
| | - Jean-Louis Guéant
- INSERM U1256, Nutrition Génétique et Exposition aux Risques Environnementaux, Medical Faculty, University of Lorraine and Regional University Hospital Center of Nancy, Vandœuvre les Nancy, France; Biochemical and Molecular biology lab, Regional University Hospital Center of Nancy, Vandoeuvre les Nancy, France; Division of Hepatogastroenterology, Regional University Hospital Center of Nancy, Vandoeuvre les Nancy, France.
| |
Collapse
|
14
|
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: 18] [Impact Index Per Article: 4.5] [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
|
15
|
Oussalah A, Levy J, Berthezène C, Alpers DH, Guéant JL. Health outcomes associated with vegetarian diets: An umbrella review of systematic reviews and meta-analyses. Clin Nutr 2020; 39:3283-3307. [PMID: 32204974 DOI: 10.1016/j.clnu.2020.02.037] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 02/22/2020] [Accepted: 02/28/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND Several meta-analyses evaluated the association between vegetarian diets and health outcomes. To integrate the large amount of the available evidence, we performed an umbrella review of published meta-analyses that investigated the association between vegetarian diets and health outcomes. METHODS We performed an umbrella review of the evidence across meta-analyses of observational and interventional studies. PubMed, Embase, Cochrane Database of Systematic Reviews, and ISI Web of Knowledge. Additional articles were retrieved from primary search references. Meta-analyses of observational or interventional studies that assessed at least one health outcome in association with vegetarian diets. We estimated pooled effect sizes (ESs) using four different random-effect models: DerSimonian and Laird, maximum likelihood, empirical Bayes, and restricted maximum likelihood. We assessed heterogeneity using I2 statistics and publication bias using funnel plots, radial plots, normal Q-Q plots, and the Rosenthal's fail-safe N test. RESULTS The umbrella review identified 20 meta-analyses of observational and interventional research with 34 health outcomes. The majority of the meta-analyses (80%) were classified as moderate or high-quality reviews, based on the AMSTAR2 criteria. By comparison with omnivorous diets, vegetarian diets were associated with a significantly lower concentration of blood total cholesterol (pooled ES = -0.549 mmol/L; 95% CI: -0.773 to -0.325; P < 0.001), LDL-cholesterol (pooled ES = -0.467 mmol/L; 95% CI: -0.600 to -0.335); P < 0.001), and HDL-cholesterol (pooled ES = -0.082 mmol/L; 95% CI: -0.095 to -0.069; P < 0.001). In comparison to omnivorous diets, vegetarian diets were associated with a reduced risk of negative health outcomes with a pooled ES of 0.886 (95% CI: 0.848 to 0.926; P < 0.001). In comparison to omnivores, Seventh-day Adventists (SDA) vegetarians had a significantly reduced risk of negative health outcomes with a pooled ES of 0.721 (95% CI: 0.625 to 0.832; P < 0.001). Non-SDA vegetarians had no significant reduction of negative health outcomes when compared to omnivores (pooled ES = 0.973; 95% CI: 0.873 to 1.083; P = 0.51). Vegetarian diets were associated with harmful outcomes on one-carbon metabolism markers (lower concentrations of vitamin B12 and higher concentrations of homocysteine), in comparison to omnivorous diets. CONCLUSIONS Vegetarian diets are associated with beneficial effects on the blood lipid profile and a reduced risk of negative health outcomes, including diabetes, ischemic heart disease, and cancer risk. Among vegetarians, SDA vegetarians could represent a subgroup with a further reduced risk of negative health outcomes. Vegetarian diets have adverse outcomes on one-carbon metabolism. The effect of vegetarian diets among pregnant and lactating women requires specific attention. Well-designed prospective studies are warranted to evaluate the consequences of the prevalence of vitamin B12 deficiency during pregnancy and infancy on later life and of trace element deficits on cancer risks. PROSPERO REGISTRATION NUMBER CRD42018092470.
Collapse
Affiliation(s)
- Abderrahim Oussalah
- University of Lorraine, INSERM UMR_S 1256, Nutrition, Genetics, and Environmental Risk Exposure (NGERE), Faculty of Medicine of Nancy, F-54000, Nancy, France; Department of Molecular Medicine, Division of Biochemistry, Molecular Biology, and Nutrition, University Hospital of Nancy, F-54000, Nancy, France; Reference Center for Inborn Errors of Metabolism (ORPHA67872), University Hospital of Nancy, F-54000, Nancy, France.
| | - Julien Levy
- University of Lorraine, INSERM UMR_S 1256, Nutrition, Genetics, and Environmental Risk Exposure (NGERE), Faculty of Medicine of Nancy, F-54000, Nancy, France
| | - Clémence Berthezène
- University of Lorraine, INSERM UMR_S 1256, Nutrition, Genetics, and Environmental Risk Exposure (NGERE), Faculty of Medicine of Nancy, F-54000, Nancy, France
| | - David H Alpers
- Department of Internal Medicine, Washington University School of Medicine, St Louis, MO, 63130, USA
| | - Jean-Louis Guéant
- University of Lorraine, INSERM UMR_S 1256, Nutrition, Genetics, and Environmental Risk Exposure (NGERE), Faculty of Medicine of Nancy, F-54000, Nancy, France; Department of Molecular Medicine, Division of Biochemistry, Molecular Biology, and Nutrition, University Hospital of Nancy, F-54000, Nancy, France; Reference Center for Inborn Errors of Metabolism (ORPHA67872), University Hospital of Nancy, F-54000, Nancy, France.
| |
Collapse
|
16
|
Ramírez-Alarcón K, Sánchez-Agurto Á, Lamperti L, Martorell M. Epigenetics, Maternal Diet and Metabolic Programming. ACTA ACUST UNITED AC 2019. [DOI: 10.2174/1874196701907010045] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Background:
The maternal environment influences embryonic and fetal life. Nutritional deficits or excesses alter the trajectory of fetus/offspring’s development. The concept of “developmental programming” and “developmental origins of health and disease” consists of the idea that maternal diet may remodel the genome and lead to epigenetic changes. These changes are induced during early life, permanently altering the phenotype in the posterior adult stage, favoring the development of metabolic diseases such as obesity, dyslipidemia, hypertension, hyperinsulinemia, and metabolic syndrome. In this review, it is aimed to overview epigenetics, maternal diet and metabolic programming factors and determine which of these might affect future generations.
Scope and Approach:
Nutrients interfere with the epigenome by influencing the supply and use of methyl groups through DNA transmethylation and demethylation mechanisms. They also influence the remodeling of chromatin and arginine or lysine residues at the N-terminal tails of histone, thus altering miRNA expression. Fats, proteins, B vitamins and folates act as important cofactors in methylation processes. The metabolism of carbon in the methyl groups of choline, folic acid and methionine to S-Adenosyl Methionine (SAM), acts as methyl donors to methyl DNA, RNA, and proteins. B-complex vitamins are important since they act as coenzymes during this process.
Key Findings and Conclusion:
Nutrients, during pregnancy, potentially influence susceptibility to diseases in adulthood. Additionally, the deficit or excess of nutrients alter the epigenetic machinery, affecting genes and influencing the genome of the offspring and therefore, predisposing the development of chronic diseases in adults.
Collapse
|
17
|
Ghemrawi R, Arnold C, Battaglia-Hsu SF, Pourié G, Trinh I, Bassila C, Rashka C, Wiedemann A, Flayac J, Robert A, Dreumont N, Feillet F, Guéant JL, Coelho D. SIRT1 activation rescues the mislocalization of RNA-binding proteins and cognitive defects induced by inherited cobalamin disorders. Metabolism 2019; 101:153992. [PMID: 31672445 DOI: 10.1016/j.metabol.2019.153992] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/16/2019] [Accepted: 09/20/2019] [Indexed: 01/01/2023]
Abstract
BACKGROUND The molecular consequences of inborn errors of vitamin B12 or cobalamin metabolism are far from being understood. Moreover, innovative therapeutic strategies are needed for the treatment of neurological outcomes that are usually resistant to conventional treatments. Our previous findings suggest a link between SIRT1, cellular stress and RNA binding proteins (RBP) mislocalization in the pathological mechanisms triggered by impaired vitamin B12 metabolism. OBJECTIVES AND METHODS The goal of this study was to investigate the effects of the pharmacological activation of SIRT1 using SRT1720 on the molecular mechanisms triggered by impaired methionine synthase activity. Experiments were performed in vitro with fibroblasts from patients with the cblG and cblC inherited defects of vitamin B12 metabolism and in vivo with an original transgenic mouse model of methionine synthase deficiency specific to neuronal cells. Subcellular localization of the RBPs HuR, HnRNPA1, RBM10, SRSF1 and Y14 was investigated by immunostaining and confocal microscopy in patient fibroblasts. RBPs methylation and phosphorylation were studied by co-immunoprecipitation and proximity ligation assay. Cognitive performance of the transgenic mice treated with SRT1720 was measured with an aquatic maze. RESULTS Patient fibroblasts with cblC and cblG defects of vitamin B12 metabolism presented with endoplasmic reticulum stress, altered methylation, phosphorylation and subcellular localization of HuR, HnRNPA1 and RBM10, global mRNA mislocalization and increased HnRNPA1-dependent skipping of IRF3 exons. Incubation of fibroblasts with cobalamin, S-adenosyl methionine and okadaic acid rescued the localization of the RBPs and mRNA. The SIRT1 activating compound SRT1720 inhibited ER stress and rescued RBP and mRNA mislocalization and IRF3 splicing. Treatment with this SIRT1 agonist prevented all these hallmarks in patient fibroblasts but it also improved the deficient hippocampo-dependent learning ability of methionine synthase conditional knock-out mice. CONCLUSIONS By unraveling the molecular mechanisms triggered by inborn errors of cbl metabolism associating ER stress, RBP mislocalization and mRNA trafficking, our study opens novel therapeutic perspectives for the treatment of inborn errors of vitamin B12 metabolism.
Collapse
Affiliation(s)
- Rose Ghemrawi
- Université de Lorraine, Inserm, UMRS 1256, NGERE - Nutrition, Genetics, and Environmental Risk Exposure, F-54000 Nancy, France
| | - Carole Arnold
- Université de Lorraine, Inserm, UMRS 1256, NGERE - Nutrition, Genetics, and Environmental Risk Exposure, F-54000 Nancy, France.
| | - Shyue-Fang Battaglia-Hsu
- Université de Lorraine, Inserm, UMRS 1256, NGERE - Nutrition, Genetics, and Environmental Risk Exposure, F-54000 Nancy, France.
| | - Grégory Pourié
- Université de Lorraine, Inserm, UMRS 1256, NGERE - Nutrition, Genetics, and Environmental Risk Exposure, F-54000 Nancy, France.
| | - Isabelle Trinh
- Université de Lorraine, Inserm, UMRS 1256, NGERE - Nutrition, Genetics, and Environmental Risk Exposure, F-54000 Nancy, France.
| | - Christine Bassila
- Université de Lorraine, Inserm, UMRS 1256, NGERE - Nutrition, Genetics, and Environmental Risk Exposure, F-54000 Nancy, France
| | - Charif Rashka
- Université de Lorraine, Inserm, UMRS 1256, NGERE - Nutrition, Genetics, and Environmental Risk Exposure, F-54000 Nancy, France.
| | - Arnaud Wiedemann
- Université de Lorraine, Inserm, UMRS 1256, NGERE - Nutrition, Genetics, and Environmental Risk Exposure, F-54000 Nancy, France; Université de Lorraine, CHRU-Nancy, National Center of Inborn Errors of Metabolism, F-54000 Nancy, France
| | - Justine Flayac
- Université de Lorraine, Inserm, UMRS 1256, NGERE - Nutrition, Genetics, and Environmental Risk Exposure, F-54000 Nancy, France.
| | - Aurélie Robert
- Université de Lorraine, Inserm, UMRS 1256, NGERE - Nutrition, Genetics, and Environmental Risk Exposure, F-54000 Nancy, France.
| | - Natacha Dreumont
- Université de Lorraine, Inserm, UMRS 1256, NGERE - Nutrition, Genetics, and Environmental Risk Exposure, F-54000 Nancy, France.
| | - François Feillet
- Université de Lorraine, Inserm, UMRS 1256, NGERE - Nutrition, Genetics, and Environmental Risk Exposure, F-54000 Nancy, France; Université de Lorraine, CHRU-Nancy, National Center of Inborn Errors of Metabolism, F-54000 Nancy, France.
| | - Jean-Louis Guéant
- Université de Lorraine, Inserm, UMRS 1256, NGERE - Nutrition, Genetics, and Environmental Risk Exposure, F-54000 Nancy, France; Université de Lorraine, CHRU-Nancy, National Center of Inborn Errors of Metabolism, F-54000 Nancy, France.
| | - David Coelho
- Université de Lorraine, Inserm, UMRS 1256, NGERE - Nutrition, Genetics, and Environmental Risk Exposure, F-54000 Nancy, France; Université de Lorraine, CHRU-Nancy, National Center of Inborn Errors of Metabolism, F-54000 Nancy, France.
| |
Collapse
|
18
|
Mladenović D, Radosavljević T, Hrnčić D, Rasic-Markovic A, Stanojlović O. The effects of dietary methionine restriction on the function and metabolic reprogramming in the liver and brain - implications for longevity. Rev Neurosci 2019; 30:581-593. [PMID: 30817309 DOI: 10.1515/revneuro-2018-0073] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 10/26/2018] [Indexed: 02/05/2023]
Abstract
Methionine is an essential sulphur-containing amino acid involved in protein synthesis, regulation of protein function and methylation reactions. Dietary methionine restriction (0.12-0.17% methionine in food) extends the life span of various animal species and delays the onset of aging-associated diseases and cancers. In the liver, methionine restriction attenuates steatosis and delays the development of non-alcoholic steatohepatitis due to antioxidative action and metabolic reprogramming. The limited intake of methionine stimulates the fatty acid oxidation in the liver and the export of lipoproteins as well as inhibits de novo lipogenesis. These effects are mediated by various signaling pathways and effector molecules, including sirtuins, growth hormone/insulin-like growth factor-1 axis, sterol regulatory element binding proteins, adenosine monophosphate-dependent kinase and general control nonderepressible 2 pathway. Additionally, methionine restriction stimulates the synthesis of fibroblast growth factor-21 in the liver, which increases the insulin sensitivity of peripheral tissues. In the brain, methionine restriction delays the onset of neurodegenerative diseases and increases the resistance to various forms of stress through antioxidative effects and alterations in lipid composition. This review aimed to summarize the morphological, functional and molecular changes in the liver and brain caused by the methionine restriction, with possible implications in the prolongation of maximal life span.
Collapse
Affiliation(s)
- Dušan Mladenović
- Institute of Pathophysiology 'Ljubodrag Buba Mihailovic', Faculty of Medicine, University of Belgrade, Dr Subotica 9, 11000 Belgrade, Serbia
| | - Tatjana Radosavljević
- Institute of Pathophysiology 'Ljubodrag Buba Mihailovic', Faculty of Medicine, University of Belgrade, Dr Subotica 9, 11000 Belgrade, Serbia
| | - Dragan Hrnčić
- Institute of Medical Physiology 'Richard Burian', Faculty of Medicine, University of Belgrade, Višegradska 26/II, 11000 Belgrade, Serbia
| | - Aleksandra Rasic-Markovic
- Institute of Medical Physiology 'Richard Burian', Faculty of Medicine, University of Belgrade, Višegradska 26/II, 11000 Belgrade, Serbia
| | - Olivera Stanojlović
- Institute of Medical Physiology 'Richard Burian', Faculty of Medicine, University of Belgrade, Višegradska 26/II, 11000 Belgrade, Serbia
| |
Collapse
|
19
|
Polyphenols and IUGR Pregnancies: Effects of Maternal Hydroxytyrosol Supplementation on Hepatic Fat Accretion and Energy and Fatty Acids Profile of Fetal Tissues. Nutrients 2019; 11:nu11071534. [PMID: 31284510 PMCID: PMC6682965 DOI: 10.3390/nu11071534] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 06/29/2019] [Accepted: 07/01/2019] [Indexed: 12/12/2022] Open
Abstract
Maternal supplementation with hydroxytyrosol, a polyphenol present in olive leaves and fruits, is a highly promising strategy to improve the oxidative and metabolic status of fetuses at risk of intrauterine growth restriction, which may diminish the appearance of low-birth-weight neonates. The present study aimed to determine whether hydroxytyrosol, by preventing lipid peroxidation, may influence the fat accretion and energy homeostasis in the liver, as well as the fatty acid composition in the liver and muscle. The results indicate that hydroxytyrosol treatment significantly decreased the energy content of the fetal liver, without affecting fat accretion, and caused significant changes in the availability of fatty acids. There were significant increases in the amount of total polyunsaturated fatty acids, omega-3 and omega-6, which are highly important for adequate fetal tissue development. However, there were increases in the omega-6/omega-3 ratio and the desaturation index, which make further studies necessary to determine possible effects on the pro/anti-inflammatory status of the fetuses.
Collapse
|
20
|
Reddy VS, Trinath J, Reddy GB. Implication of homocysteine in protein quality control processes. Biochimie 2019; 165:19-31. [PMID: 31269461 DOI: 10.1016/j.biochi.2019.06.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 06/26/2019] [Indexed: 12/22/2022]
Abstract
Homocysteine (Hcy) is a key metabolite generated during methionine metabolism. The elevated levels of Hcy in the blood are reffered to as hyperhomocystenimeia (HHcy). The HHcy is caused by impaired metabolism/deficiency of either folate or B12 or defects in Hcy metabolism. Accumulating evidence suggests that HHcy is associated with cardiovascular and brain diseases including atherosclerosis, endothelial injury, and stroke etc. Vitamin B12 (cobalamin; B12) is a water-soluble vitamin essential for two metabolic reactions. It acts as a co-factor for methionine synthase and L-methylmalonyl-CoA mutase. Besides, it is also vital for DNA synthesis and maturation of RBC. Deficiency of B12 is associated with haematological and neurological disorders. Hyperhomocysteinemia (HHcy)-induced toxicity is thought to be mediated by the accumulation of Hcy and its metabolites, homocysteinylated proteins. Cellular protein quality control (PQC) is essential for the maintenance of proteome integrity, and cell viability and its failure contributes to the development of multiple diseases. Chaperones, unfolded protein response (UPR), ubiquitin-proteasome system (UPS), and autophagy are analogous strategies of PQC that maintain cellular proteome integrity. Recently, multiple studies reported that HHcy responsible for perturbation of PQC by reducing chaperone levels, activating UPR, and impairing autophagy. Besides, HHcy also induce cytotoxicity, inflammation, protein aggregation and apoptosis. It has been shown that some of the factors including altered SIRT1-HSF1 axis and irreversible homocysteinylation of proteins are responsible for folate and/or B12 deficiency or HHcy-induced impairment of PQC. Therefore, this review highlights the current understanding of HHcy in the context of cellular PQC and their pathophysiological and clinical consequences, epigenomic changes, therapeutic implications of B12, and chemical chaperones based on cell culture and experimental animal models.
Collapse
Affiliation(s)
- V Sudhakar Reddy
- Biochemistry Division, National Institute of Nutrition, Hyderabad, India.
| | - Jamma Trinath
- Department of Biological Sciences, BITS-Pilani, 500078, Hyderabad Campus, Hyderabad, Telangana, India
| | | |
Collapse
|
21
|
Cerf ME. Cardiac Glucolipotoxicity and Cardiovascular Outcomes. ACTA ACUST UNITED AC 2018; 54:medicina54050070. [PMID: 30344301 PMCID: PMC6262512 DOI: 10.3390/medicina54050070] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 10/04/2018] [Accepted: 10/05/2018] [Indexed: 01/12/2023]
Abstract
Cardiac insulin signaling can be impaired due to the altered fatty acid metabolism to induce insulin resistance. In diabetes and insulin resistance, the metabolic, structural and ultimately functional alterations in the heart and vasculature culminate in diabetic cardiomyopathy, coronary artery disease, ischemia and eventually heart failure. Glucolipotoxicity describes the combined, often synergistic, adverse effects of elevated glucose and free fatty acid concentrations on heart structure, function, and survival. The quality of fatty acid shapes the cardiac structure and function, often influencing survival. A healthy fatty acid balance is therefore critical for maintaining cardiac integrity and function.
Collapse
Affiliation(s)
- Marlon E Cerf
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa.
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Stellenbosch, Tygerberg 7505, South Africa.
| |
Collapse
|
22
|
Battaglia-Hsu SF, Ghemrawi R, Coelho D, Dreumont N, Mosca P, Hergalant S, Gauchotte G, Sequeira JM, Ndiongue M, Houlgatte R, Alberto JM, Umoret R, Robert A, Paoli J, Jung M, Quadros EV, Guéant JL. Inherited disorders of cobalamin metabolism disrupt nucleocytoplasmic transport of mRNA through impaired methylation/phosphorylation of ELAVL1/HuR. Nucleic Acids Res 2018; 46:7844-7857. [PMID: 30016500 PMCID: PMC6125644 DOI: 10.1093/nar/gky634] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 06/26/2018] [Accepted: 07/12/2018] [Indexed: 12/12/2022] Open
Abstract
The molecular mechanisms that underlie the neurological manifestations of patients with inherited diseases of vitamin B12 (cobalamin) metabolism remain to date obscure. We observed transcriptomic changes of genes involved in RNA metabolism and endoplasmic reticulum stress in a neuronal cell model with impaired cobalamin metabolism. These changes were related to the subcellular mislocalization of several RNA binding proteins, including the ELAVL1/HuR protein implicated in neuronal stress, in this cell model and in patient fibroblasts with inborn errors of cobalamin metabolism and Cd320 knockout mice. The decreased interaction of ELAVL1/HuR with the CRM1/exportin protein of the nuclear pore complex and its subsequent mislocalization resulted from hypomethylation at R-217 produced by decreased S-adenosylmethionine and protein methyl transferase CARM1 and dephosphorylation at S221 by increased protein phosphatase PP2A. The mislocalization of ELAVL1/HuR triggered the decreased expression of SIRT1 deacetylase and genes involved in brain development, neuroplasticity, myelin formation, and brain aging. The mislocalization was reversible upon treatment with siPpp2ca, cobalamin, S-adenosylmethionine, or PP2A inhibitor okadaic acid. In conclusion, our data highlight the key role of the disruption of ELAVL1/HuR nuclear export, with genomic changes consistent with the effects of inborn errors of Cbl metabolisms on brain development, neuroplasticity and myelin formation.
Collapse
Affiliation(s)
- Shyue-Fang Battaglia-Hsu
- INSERM UMRS 954 NGERE – Nutrition, Genetics, and Environmental Risk Exposure and National Center of Inborn Errors of Metabolism, Faculty of Medicine of Nancy, University of Lorraine and University Regional Hospital Center of Nancy, Vandoeuvre-lès-Nancy, F-54000, France
| | - Rose Ghemrawi
- INSERM UMRS 954 NGERE – Nutrition, Genetics, and Environmental Risk Exposure and National Center of Inborn Errors of Metabolism, Faculty of Medicine of Nancy, University of Lorraine and University Regional Hospital Center of Nancy, Vandoeuvre-lès-Nancy, F-54000, France
| | - David Coelho
- INSERM UMRS 954 NGERE – Nutrition, Genetics, and Environmental Risk Exposure and National Center of Inborn Errors of Metabolism, Faculty of Medicine of Nancy, University of Lorraine and University Regional Hospital Center of Nancy, Vandoeuvre-lès-Nancy, F-54000, France
| | - Natacha Dreumont
- INSERM UMRS 954 NGERE – Nutrition, Genetics, and Environmental Risk Exposure and National Center of Inborn Errors of Metabolism, Faculty of Medicine of Nancy, University of Lorraine and University Regional Hospital Center of Nancy, Vandoeuvre-lès-Nancy, F-54000, France
| | - Pauline Mosca
- INSERM UMRS 954 NGERE – Nutrition, Genetics, and Environmental Risk Exposure and National Center of Inborn Errors of Metabolism, Faculty of Medicine of Nancy, University of Lorraine and University Regional Hospital Center of Nancy, Vandoeuvre-lès-Nancy, F-54000, France
| | - Sébastien Hergalant
- INSERM UMRS 954 NGERE – Nutrition, Genetics, and Environmental Risk Exposure and National Center of Inborn Errors of Metabolism, Faculty of Medicine of Nancy, University of Lorraine and University Regional Hospital Center of Nancy, Vandoeuvre-lès-Nancy, F-54000, France
| | - Guillaume Gauchotte
- INSERM UMRS 954 NGERE – Nutrition, Genetics, and Environmental Risk Exposure and National Center of Inborn Errors of Metabolism, Faculty of Medicine of Nancy, University of Lorraine and University Regional Hospital Center of Nancy, Vandoeuvre-lès-Nancy, F-54000, France
| | - Jeffrey M Sequeira
- Division of Hematology/Oncology, Department of Medicine, SUNY-Downstate Medical Center, Brooklyn, New York, NY, USA
| | - Mariam Ndiongue
- INSERM UMRS 954 NGERE – Nutrition, Genetics, and Environmental Risk Exposure and National Center of Inborn Errors of Metabolism, Faculty of Medicine of Nancy, University of Lorraine and University Regional Hospital Center of Nancy, Vandoeuvre-lès-Nancy, F-54000, France
| | - Rémi Houlgatte
- INSERM UMRS 954 NGERE – Nutrition, Genetics, and Environmental Risk Exposure and National Center of Inborn Errors of Metabolism, Faculty of Medicine of Nancy, University of Lorraine and University Regional Hospital Center of Nancy, Vandoeuvre-lès-Nancy, F-54000, France
| | - Jean-Marc Alberto
- INSERM UMRS 954 NGERE – Nutrition, Genetics, and Environmental Risk Exposure and National Center of Inborn Errors of Metabolism, Faculty of Medicine of Nancy, University of Lorraine and University Regional Hospital Center of Nancy, Vandoeuvre-lès-Nancy, F-54000, France
| | - Remy Umoret
- INSERM UMRS 954 NGERE – Nutrition, Genetics, and Environmental Risk Exposure and National Center of Inborn Errors of Metabolism, Faculty of Medicine of Nancy, University of Lorraine and University Regional Hospital Center of Nancy, Vandoeuvre-lès-Nancy, F-54000, France
| | - Aurélie Robert
- INSERM UMRS 954 NGERE – Nutrition, Genetics, and Environmental Risk Exposure and National Center of Inborn Errors of Metabolism, Faculty of Medicine of Nancy, University of Lorraine and University Regional Hospital Center of Nancy, Vandoeuvre-lès-Nancy, F-54000, France
| | - Justine Paoli
- INSERM UMRS 954 NGERE – Nutrition, Genetics, and Environmental Risk Exposure and National Center of Inborn Errors of Metabolism, Faculty of Medicine of Nancy, University of Lorraine and University Regional Hospital Center of Nancy, Vandoeuvre-lès-Nancy, F-54000, France
| | - Martin Jung
- Department of Medical Biochemistry and Molecular Biology, Saarland University, Building 44, Homburg 66421, Germany
| | - Edward V Quadros
- Division of Hematology/Oncology, Department of Medicine, SUNY-Downstate Medical Center, Brooklyn, New York, NY, USA
| | - Jean-Louis Guéant
- INSERM UMRS 954 NGERE – Nutrition, Genetics, and Environmental Risk Exposure and National Center of Inborn Errors of Metabolism, Faculty of Medicine of Nancy, University of Lorraine and University Regional Hospital Center of Nancy, Vandoeuvre-lès-Nancy, F-54000, France
| |
Collapse
|
23
|
Li Y. Epigenetic Mechanisms Link Maternal Diets and Gut Microbiome to Obesity in the Offspring. Front Genet 2018; 9:342. [PMID: 30210530 PMCID: PMC6119695 DOI: 10.3389/fgene.2018.00342] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 08/09/2018] [Indexed: 12/14/2022] Open
Abstract
Nutrition is the most important environmental factor that can influence early developmental processes through regulation of epigenetic mechanisms during pregnancy and neonatal periods. Maternal diets or nutritional compositions contribute to the establishment of the epigenetic profiles in the fetus that have a profound impact on individual susceptibility to certain diseases or disorders in the offspring later in life. Obesity is considered a global epidemic that impairs human life quality and also increases risk of development of many human diseases such as diabetes and cardiovascular diseases. Studies have shown that maternal nutrition status is closely associated with obesity in progenies indicating obesity has a developmental origin. Maternal diets may also impact the early establishment of the fetal and neonatal microbiome leading to specific epigenetic signatures that may potentially predispose to the development of late-life obesity. This article will review the association of different maternal dietary statuses including essential nutritional quantity and specific dietary components with gut microbiome in determining epigenetic impacts on offspring susceptibility to obesity.
Collapse
Affiliation(s)
- Yuanyuan Li
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham, Birmingham, AL, United States
- Comprehensive Cancer Center, University of Alabama at Birmingham, Birmingham, AL, United States
- Nutrition Obesity Research Center, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
24
|
Abstract
The present review will present the recent published results and discuss the main effects of nutrients, mainly fatty acids, on the expression of genes involved in lipid metabolism. In this sense, the review focuses in two phases: prenatal life and finishing phase, showing how nutrients can modulate gene expression affecting marbling and fatty acid profile in meat from ruminants. Adiposity in ruminants starts to be affected by nutrients during prenatal life when maternal nutrition affects the differentiation and proliferation of adipose cells enhancing the marbling potential. Therefore, several fetal programming studies were carried out in the last two decades in order to better understand how nutrients affect long-term expression of genes involved in adipogenesis and lipogenesis. In addition, during the finishing phase, marbling becomes largely dependent on starch digestion and glucose metabolism, being important to create alternatives to increase these metabolic processes, and modulates gene expression. Different lipid sources and their fatty acids may also influence the expression of genes responsible to encode enzymes involved in fat tissue deposition, influencing meat quality. In conclusion, the knowledge shows that gene expression is a metabolic factor affecting marbling and fatty acid profile in ruminant meat and diets and their nutrients have direct effect on how these genes are expressed.
Collapse
|
25
|
Wnt Signaling Pathways Are Dysregulated in Rat Female Cerebellum Following Early Methyl Donor Deficiency. Mol Neurobiol 2018; 56:892-906. [PMID: 29804229 DOI: 10.1007/s12035-018-1128-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Accepted: 05/14/2018] [Indexed: 10/16/2022]
Abstract
Gestational methyl donor (especially B9 and B12 vitamins) deficiency is involved in birth defects and brain development retardation. The underlying molecular mechanisms that are dysregulated still remain poorly understood, in particular in the cerebellum. As evidenced from previous data, females are more affected than males. In this study, we therefore took advantage of a validated rat nutritional model and performed a microarray analysis on female progeny cerebellum, in order to identify which genes and molecular pathways were disrupted in response to methyl donor deficiency. We found that cerebellum development is altered in female pups, with a decrease of the granular cell layer thickness at postnatal day 21. Furthermore, we investigated the involvement of the Wnt signaling pathway, a major molecular pathway involved in neuronal development and later on in synaptic assembly and neurotransmission processes. We found that Wnt canonical pathway was disrupted following early methyl donor deficiency and that neuronal targets were selectively enriched in the downregulated genes. These results could explain the structural brain defects previously observed and highlighted new genes and a new molecular pathway affected by nutritional methyl donor deprivation.
Collapse
|
26
|
Quantification of fetal organ volume and fat deposition following in utero exposure to maternal Western Diet using MRI. PLoS One 2018; 13:e0192900. [PMID: 29447203 PMCID: PMC5814025 DOI: 10.1371/journal.pone.0192900] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 01/16/2018] [Indexed: 12/17/2022] Open
Abstract
PURPOSE To examine the feasibility of using MRI to identify differences in liver size and fat deposition in fetal guinea pigs exposed to an in utero environment influenced by maternal consumption of a Western diet. MATERIALS AND METHODS Female guinea pigs fed either an energy-dense Western Diet (WD), comprised of increased saturated fats and simple sugars, or a Control Diet (CD) from weaning through pregnancy, underwent MR scanning near term (~ 60 days; term ~ 69 days). Maternal weights were collected at mating and at MR scanning. T1-weighted, T2-weighted, and IDEAL water-fat images were acquired at 3 Tesla. The images were used to segment maternal adipose tissue, fetal liver, fetal brain, fetal adipose tissue, and total fetal volumes and to measure maternal and fetal hepatic fat fractions. RESULTS Weights of WD sows were lower prior to pregnancy (P = .04), however their weight gain over pregnancy did not differ from the CD group (P = .98). The WD sows had less total adipose tissue (TAT) at MR scanning (P = .04), while hepatic fat content was significantly elevated (P = .04). When controlling for litter size, WD fetuses had larger livers (P = .02), smaller brains (P = .01), and increased total adipose tissue volume (P = .01) when normalized by fetal volume. The WD fetuses also had increased hepatic fat fractions compared to CD fetal livers (P < .001). CONCLUSION Maternal Western Diet consumption prior to and during pregnancy induces differences in maternal liver fat content, fetal liver volume and liver fat storage, as well as changes in fetal adipose tissue deposition that can be measured in utero using MRI.
Collapse
|
27
|
In rats gestational iron deficiency does not change body fat or hepatic mitochondria in the aged offspring. J Dev Orig Health Dis 2017; 9:232-240. [PMID: 28870272 DOI: 10.1017/s2040174417000721] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mitochondrial dysfunction and resulting changes in adiposity have been observed in the offspring of animals fed a high fat (HF) diet. As iron is an important component of the mitochondria, we have studied the offspring of female rats fed complete (Con) or iron-deficient (FeD) rations for the duration of gestation to test for similar effects. The FeD offspring were ~12% smaller at weaning and remained so because of a persistent reduction in lean tissue mass. The offspring were fed a complete (stock) diet until 52 weeks of age after which some animals from each litter were fed a HF diet for a further 12 weeks. The HF diet increased body fat when compared with animals fed the stock diet, however, prenatal iron deficiency did not change the ratio of fat:lean in either the stock or HF diet groups. The HF diet caused triglyceride to accumulate in the liver, however, there was no effect of prenatal iron deficiency. The activity of the mitochondrial electron transport complexes was similar in all groups including those challenged with a HF diet. HF feeding increased the number of copies of mitochondrial DNA and the prevalence of the D-loop mutation, however, neither parameter was affected by prenatal iron deficiency. This study shows that the effects of prenatal iron deficiency differ from other models in that there is no persistent effect on hepatic mitochondria in aged animals exposed to an increased metabolic load.
Collapse
|
28
|
Periyasamy P, Shinohara T. Age-related cataracts: Role of unfolded protein response, Ca 2+ mobilization, epigenetic DNA modifications, and loss of Nrf2/Keap1 dependent cytoprotection. Prog Retin Eye Res 2017; 60:1-19. [PMID: 28864287 PMCID: PMC5600869 DOI: 10.1016/j.preteyeres.2017.08.003] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 08/26/2017] [Accepted: 08/28/2017] [Indexed: 12/11/2022]
Abstract
Age-related cataracts are closely associated with lens chronological aging, oxidation, calcium imbalance, hydration and crystallin modifications. Accumulating evidence indicates that misfolded proteins are generated in the endoplasmic reticulum (ER) by most cataractogenic stresses. To eliminate misfolded proteins from cells before they can induce senescence, the cells activate a clean-up machinery called the ER stress/unfolded protein response (UPR). The UPR also activates the nuclear factor-erythroid-2-related factor 2 (Nrf2), a central transcriptional factor for cytoprotection against stress. Nrf2 activates nearly 600 cytoprotective target genes. However, if ER stress reaches critically high levels, the UPR activates destructive outputs to trigger programmed cell death. The UPR activates mobilization of ER-Ca2+ to the cytoplasm and results in activation of Ca2+-dependent proteases to cleave various enzymes and proteins which cause the loss of normal lens function. The UPR also enhances the overproduction of reactive oxygen species (ROS), which damage lens constituents and induce failure of the Nrf2 dependent cytoprotection. Kelch-like ECH-associated protein 1 (Keap1) is an oxygen sensor protein and regulates the levels of Nrf2 by the proteasomal degradation. A significant loss of DNA methylation in diabetic cataracts was found in the Keap1 promoter, which overexpresses the Keap1 protein. Overexpressed Keap1 significantly decreases the levels of Nrf2. Lower levels of Nrf2 induces loss of the redox balance toward to oxidative stress thereby leading to failure of lens cytoprotection. Here, this review summarizes the overall view of ER stress, increases in Ca2+ levels, protein cleavage, and loss of the well-established stress protection in somatic lens cells.
Collapse
Affiliation(s)
- Palsamy Periyasamy
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA; Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| | - Toshimichi Shinohara
- Department of Ophthalmology and Visual Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
29
|
Folate and vitamin B12 status is associated with insulin resistance and metabolic syndrome in morbid obesity. Clin Nutr 2017; 37:1700-1706. [PMID: 28780990 DOI: 10.1016/j.clnu.2017.07.008] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Revised: 06/24/2017] [Accepted: 07/06/2017] [Indexed: 12/29/2022]
Abstract
BACKGROUND Low vitamin B12 and high folate during pregnancy are associated with visceral obesity and insulin resistance in offspring. In the general population, high folate exacerbates the increase of methylmalonic acid, a marker of vitamin B12 deficiency. However, the influence of vitamin B12 and folate and their related markers on insulin resistance and metabolic syndrome remains unknown in severe obesity. AIM To evaluate the influence of vitamin B12 and folate on HOMA-IR and components of metabolic syndrome in severe obesity. METHODS 278 consecutive obese patients were assessed prospectively for HOMA-IR, red blood cell (RBC) folates, homocysteine and methylmalonic acid. We compared the associations with the components of metabolic syndrome during the preoperative multidisciplinary evaluation (period-1) and before bariatric surgery (period-2). RESULTS The HOMA-IR was higher in patients with highest tertile of RBC and either lowest tertile of plasma B12 or highest tertile of MMA (p < 0.034 and 0.011, respectively). Lg HOMA-IR was negatively correlated with Lg homocysteine (p < 0.0001) and positively correlated with Lg serum folate (p < 0.001). The independent predictors for HOMA-IR at period 2 were either BMI and homocysteine (model 1 without serum folate, p = 0.010 and p = 0.002, respectively) or BMI and MMA (model 2 without homocysteine, p = 0.030 and p = 0.004, respectively). Age and RBC folate remained independently associated with the number of metabolic syndrome components (p = 0.006 and 0.020, respectively). CONCLUSIONS RBC folate, homocysteine, and MMA predict HOMA-IR in severe obesity. Our findings challenge the benefit of folate fortified food in severe obesity, in particular in patients with a deficit of vitamin B12. The cohort study was registered at clinicaltrials.gov as NCT02663388.
Collapse
|
30
|
Zhang H, Li Y, Su W, Ying Z, Zhou L, Zhang L, Wang T. Resveratrol attenuates mitochondrial dysfunction in the liver of intrauterine growth retarded suckling piglets by improving mitochondrial biogenesis and redox status. Mol Nutr Food Res 2017; 61. [PMID: 27958670 DOI: 10.1002/mnfr.201600653] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 11/24/2016] [Accepted: 12/05/2016] [Indexed: 12/25/2022]
Abstract
SCOPE Emerging evidence has identified mitochondrial biogenesis and oxidative phosphorylation as potential targets for the prevention and treatment of metabolic syndrome. This study investigated the effect of resveratrol (RSV) on hepatic mitochondrial function in intrauterine growth-retarded (IUGR) suckling piglets. METHODS AND RESULTS Seven normal birth weight (NBW) and fourteen IUGR neonatal male piglets were selected. Piglets were fed control diets supplemented with 0 (NBW-CON), 0 (IUGR-CON), and 1.0 (IUGR-RSV) g RSV per kg of milk dry matter from 7 to 21 days of age (n = 7), respectively. Mitochondrial function, swelling, and redox status in the liver were assessed. Compared with NBW, IUGR impaired hepatic mitochondrial biogenesis and energy homeostasis of the control piglets. IUGR control piglets showed overproduction of superoxide radicals, increased concentration of malondialdehyde, and marked swelling in the mitochondria. RSV improved mitochondrial DNA content, ATP production, and fatty acid oxidation in the liver of IUGR piglets, along with an increased activity of sirtuin 1. RSV inhibited mitochondrial superoxide anion accumulation, increased complex III and manganese superoxide dismutase activities, and ameliorated mitochondrial swelling and lipid peroxidation in the IUGR piglets. CONCLUSION RSV may have beneficial effects in improving hepatic mitochondrial function and redox status in the IUGR piglets.
Collapse
Affiliation(s)
- Hao Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Yue Li
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Weipeng Su
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Zhixiong Ying
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Le Zhou
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Lili Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Tian Wang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
31
|
Abstract
Obesity-related co-morbidities decrease life quality, reduce working ability and lead to early death. The total amount of dietary fat consumption may be the most potent food-related risk factor for weight gain. In this respect, dietary intake of high-caloric, high-fat diets due to chronic over-eating and sedentary lifestyle lead to increased storage of triglycerides not only in adipose tissue but also ectopically in other tissues . Increased plasma concentrations of non-esterified free fatty acids and lipid-overloaded hypertrophic adipocytes may cause insulin resistance in an inflammation-independent manner. Even in the absence of metabolic disorders, mismatch between fatty acid uptake and utilization leads to the accumulation of toxic lipid species resulting in organ dysfunction. Lipid-induced apoptosis, ceramide accumulation, reactive oxygen species overproduction, endoplasmic reticulum stress, and mitochondrial dysfunction may play role in the pathogenesis of lipotoxicity. The hypothalamus senses availability of circulating levels of glucose, lipids and amino acids, thereby modifies feeding according to the levels of those molecules. However, the hypothalamus is also similarly vulnerable to lipotoxicity as the other ectopic lipid accumulated tissues. Chronic overnutrition most likely provides repetitive and persistent signals that up-regulate inhibitor of nuclear factor kappa B kinase beta subunit/nuclear factor kappa B (IKKβ/NF-κB) in the hypothalamus before the onset of obesity. However, the mechanisms by which high-fat diet induced peripheral signals affect the hypothalamic arcuate nucleus remain largely unknown. In this chapter, besides lipids and leptin, the role of glucose and insulin on specialized fuel-sensing neurons of hypothalamic neuronal circuits has been debated.
Collapse
|
32
|
Perinatal high methyl donor alters gene expression in IGF system in male offspring without altering DNA methylation. Future Sci OA 2016; 3:FSO164. [PMID: 28344827 PMCID: PMC5351714 DOI: 10.4155/fsoa-2016-0077] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Accepted: 11/15/2016] [Indexed: 01/08/2023] Open
Abstract
Aim: To investigate the effect of a protein restriction and a supplementation with methyl donor nutrients during fetal and early postnatal life on the expression and epigenetic state of imprinted genes from the IGF system. Materials & methods: Pregnant female rats were fed a protein-restricted diet supplemented or not with methyl donor. Results: Gene expression of the Igf2, H19, Igf1, Igf2r and Plagl1 genes in the liver of male offspring at birth and weaning was strongly influenced by maternal diet. Whereas the methylation profiles of the Igf2, H19 and Igf2r genes were remarkably stable, DNA methylation of Plagl1 promoter was slightly modified. Conclusion: DNA methylation of most, but not all, imprinted gene regulatory regions was resistant to methyl group nutritional supply. Fetal environment influences fetal growth and may confer a risk to develop metabolic diseases, possibly through alterations in the epigenetic state of the genome. Imprinted genes constitute a special class of genes that are crucial for the control of fetal and postnatal growth and are closely associated with energy metabolism. In addition, these genes are finely regulated by epigenetic mechanisms that are themselves influenced by environmental factors. This study showed that methyl donor nutrients in maternal diet strongly influenced the expression level of imprinted genes in the liver of rat offspring, despite a mild effect on epigenetic regulation.
Collapse
|
33
|
Su KH, Dai C. Metabolic control of the proteotoxic stress response: implications in diabetes mellitus and neurodegenerative disorders. Cell Mol Life Sci 2016; 73:4231-4248. [PMID: 27289378 PMCID: PMC5599143 DOI: 10.1007/s00018-016-2291-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2015] [Revised: 05/13/2016] [Accepted: 06/07/2016] [Indexed: 12/12/2022]
Abstract
Proteome homeostasis, or proteostasis, is essential to maintain cellular fitness and its disturbance is associated with a broad range of human health conditions and diseases. Cells are constantly challenged by various extrinsic and intrinsic insults, which perturb cellular proteostasis and provoke proteotoxic stress. To counter proteomic perturbations and preserve proteostasis, cells mobilize the proteotoxic stress response (PSR), an evolutionarily conserved transcriptional program mediated by heat shock factor 1 (HSF1). The HSF1-mediated PSR guards the proteome against misfolding and aggregation. In addition to proteotoxic stress, emerging studies reveal that this proteostatic mechanism also responds to cellular energy state. This regulation is mediated by the key cellular metabolic sensor AMP-activated protein kinase (AMPK). In this review, we present an overview of the maintenance of proteostasis by HSF1, the metabolic regulation of the PSR, particularly focusing on AMPK, and their implications in the two major age-related diseases-diabetes mellitus and neurodegenerative disorders.
Collapse
Affiliation(s)
- Kuo-Hui Su
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA
| | - Chengkai Dai
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME, 04609, USA.
| |
Collapse
|
34
|
Melhem H, Hansmannel F, Bressenot A, Battaglia-Hsu SF, Billioud V, Alberto JM, Gueant JL, Peyrin-Biroulet L. Methyl-deficient diet promotes colitis and SIRT1-mediated endoplasmic reticulum stress. Gut 2016; 65:595-606. [PMID: 25608526 DOI: 10.1136/gutjnl-2014-307030] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 01/04/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND Methyl donor deficiency (MDD) aggravates experimental colitis in rats and increases endoplasmic reticulum (ER) stress through decreased sirtuin 1 (SIRT1) in neuronal cells and myocardium. ER stress plays a key role in IBD pathogenesis. AIM We investigated whether the influence of MDD on colitis resulted from an ER stress response triggered by decreased SIRT1 expression. DESIGN The unfolded protein response (UPR), chaperones proteins, heat shock factor protein 1 (HSF1) and SIRT1 were examined in rats with MDD and dextran sulfate sodium (DSS)-induced colitis in a Caco-2 cell model with stable expression of transcobalamin-oleosin (TO) chimera, which impairs cellular availability of vitamin B12, and in IBD. The effects of SIRT1 activation were studied both in vitro and in vivo. RESULTS MDD aggravated DSS-induced colitis clinically, endoscopically and histologically. MDD activated ER stress pathways, with increased phosphorylate-PKR-like ER kinase, P-eiF-2α, P-IRE-1α, activating transcription factor (ATF)6, XBP1-S protein and ATF4 mRNA expression levels in rats. This was accompanied by reduced SIRT1 expression level and greater acetylation of HSF1, in relation with a dramatic decrease of chaperones (binding immunoglobulin protein (BIP), heat shock protein (HSP)27 and HSP90). Adding either vitamin B12, S-adenosylmethionine or an SIRT1 activator (SRT1720) reduced the UPR in vitro. In rats, SIRT1 activation by SRT1720 prevented colitis by reducing HSF1 acetylation and increasing expression of BIP, HSP27 and HSP90. Immunohistochemistry showed impaired expression of SIRT1 in the colonic epithelium of patients with IBD. CONCLUSIONS SIRT1 is a master regulator of ER stress and severity of experimental colitis in case of MDD. It could deserve further interest as a therapeutic target of IBD.
Collapse
Affiliation(s)
- Hassan Melhem
- INSERM U954, Faculté de Médecine, Nutrition Génétique et exposition aux risques environnementaux, Université de Lorraine 54 511, Vandœuvre-Lès-Nancy cedex, France
| | - Franck Hansmannel
- INSERM U954, Faculté de Médecine, Nutrition Génétique et exposition aux risques environnementaux, Université de Lorraine 54 511, Vandœuvre-Lès-Nancy cedex, France
| | - Aude Bressenot
- INSERM U954, Faculté de Médecine, Nutrition Génétique et exposition aux risques environnementaux, Université de Lorraine 54 511, Vandœuvre-Lès-Nancy cedex, France
| | - Syue-Fang Battaglia-Hsu
- INSERM U954, Faculté de Médecine, Nutrition Génétique et exposition aux risques environnementaux, Université de Lorraine 54 511, Vandœuvre-Lès-Nancy cedex, France
| | - Vincent Billioud
- INSERM U954, Faculté de Médecine, Nutrition Génétique et exposition aux risques environnementaux, Université de Lorraine 54 511, Vandœuvre-Lès-Nancy cedex, France
| | - Jean Marc Alberto
- INSERM U954, Faculté de Médecine, Nutrition Génétique et exposition aux risques environnementaux, Université de Lorraine 54 511, Vandœuvre-Lès-Nancy cedex, France
| | - Jean Louis Gueant
- INSERM U954, Faculté de Médecine, Nutrition Génétique et exposition aux risques environnementaux, Université de Lorraine 54 511, Vandœuvre-Lès-Nancy cedex, France
| | - Laurent Peyrin-Biroulet
- INSERM U954, Faculté de Médecine, Nutrition Génétique et exposition aux risques environnementaux, Université de Lorraine 54 511, Vandœuvre-Lès-Nancy cedex, France
| |
Collapse
|
35
|
Brun JM, Bernadet MD, Cornuez A, Leroux S, Bodin L, Basso B, Davail S, Jaglin M, Lessire M, Martin X, Sellier N, Morisson M, Pitel F. Influence of grand-mother diet on offspring performances through the male line in Muscovy duck. BMC Genet 2015; 16:145. [PMID: 26690963 PMCID: PMC4687110 DOI: 10.1186/s12863-015-0303-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 12/10/2015] [Indexed: 01/28/2023] Open
Abstract
Background In mammals, multigenerational environmental effects have been documented by either epidemiological studies in human or animal experiments in rodents. Whether such phenomena also occur in birds for more than one generation is still an open question. The objective of this study was to investigate if a methionine deficiency experienced by a mother (G0) could affect her grand-offspring phenotypes (G2 hybrid mule ducks and G2 purebred Muscovy ducks), through their Muscovy sons (G1). Muscovy drakes are used for the production of mule ducks, which are sterile offspring of female common duck (Anas platyrhynchos) and Muscovy drakes (Cairina moschata). In France, mule ducks are bred mainly for the production of “foie gras”, which stems from hepatic steatosis under two weeks of force-feeding (FF). Two groups of female Muscovy ducks received either a methionine deficient diet or a control diet. Their sons were mated to Muscovy or to common duck females to produce Muscovy or Mule ducks, respectively. Several traits were measured in the G2 progenies, concerning growth, feed efficiency during FF, body composition after FF, and quality of foie gras and magret. Results In the G2 mule duck progeny, grand-maternal methionine deficiency (GMMD) decreased 4, 8, and 12 week body weights but increased weight gain and feed efficiency during FF, and abdominal fat weight. The plasmatic glucose and triglyceride contents at the end of FF were higher in the methionine deficient group. In the G2 purebred Muscovy progeny, GMMD tended to decrease 4 week body weight in both sexes, and decreased weight gain between the ages of 4 and 12 weeks, 12 week body weight, and body weight at the end of FF in male offspring only. GMMD tended to increase liver weight and increased the carcass proportion of liver in both sexes. Conclusion Altogether, these results show that the mother’s diet is able to affect traits linked to growth and to lipid metabolism in the offspring of her sons, in Muscovy ducks. Whether this transmission through the father of information induced in the grand-mother by the environment is epigenetic remains to be demonstrated. Electronic supplementary material The online version of this article (doi:10.1186/s12863-015-0303-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jean-Michel Brun
- UMR INRA, Génétique, Physiologie et Systèmes d'Elevage, INRA, 31328, Castanet Tolosan, France. .,INPT ENSAT, Génétique, Physiologie et Systèmes d'Elevage, INRA, 31328, Castanet Tolosan, France. .,INPT ENVT, Génétique, Physiologie et Systèmes d'Elevage, INRA, 31328, Castanet Tolosan, France.
| | - Marie-Dominique Bernadet
- Institut National de la Recherche Agronomique, Unité Expérimentale des Palmipèdes à Foie Gras, UE89, 40280, Benquet, France.
| | - Alexis Cornuez
- Institut National de la Recherche Agronomique, Unité Expérimentale des Palmipèdes à Foie Gras, UE89, 40280, Benquet, France.
| | - Sophie Leroux
- UMR INRA, Génétique, Physiologie et Systèmes d'Elevage, INRA, 31328, Castanet Tolosan, France. .,INPT ENSAT, Génétique, Physiologie et Systèmes d'Elevage, INRA, 31328, Castanet Tolosan, France. .,INPT ENVT, Génétique, Physiologie et Systèmes d'Elevage, INRA, 31328, Castanet Tolosan, France.
| | - Loys Bodin
- UMR INRA, Génétique, Physiologie et Systèmes d'Elevage, INRA, 31328, Castanet Tolosan, France. .,INPT ENSAT, Génétique, Physiologie et Systèmes d'Elevage, INRA, 31328, Castanet Tolosan, France. .,INPT ENVT, Génétique, Physiologie et Systèmes d'Elevage, INRA, 31328, Castanet Tolosan, France.
| | - Benjamin Basso
- UMR INRA, Génétique, Physiologie et Systèmes d'Elevage, INRA, 31328, Castanet Tolosan, France. .,INPT ENSAT, Génétique, Physiologie et Systèmes d'Elevage, INRA, 31328, Castanet Tolosan, France. .,INPT ENVT, Génétique, Physiologie et Systèmes d'Elevage, INRA, 31328, Castanet Tolosan, France. .,Present addresses: ITSAP-Institut de l'Abeille, Site Agroparc, 84914, Avignon, France. .,UMT Protection des Abeilles dans l'Environnement, CS 40506, 84914, Avignon, France.
| | - Stéphane Davail
- UMR5254 IUT des Pays de l'Adour-CNRS, 40004, Mont de Marsan Cedex, France.
| | - Mathilde Jaglin
- UMR5254 IUT des Pays de l'Adour-CNRS, 40004, Mont de Marsan Cedex, France.
| | - Michel Lessire
- Institut National de la Recherche Agronomique, UR83 Unité de Recherche Avicole, 37380, Nouzilly, France.
| | - Xavier Martin
- Institut National de la Recherche Agronomique, Unité Expérimentale des Palmipèdes à Foie Gras, UE89, 40280, Benquet, France.
| | - Nadine Sellier
- Institut National de la Recherche Agronomique, Pôle d'Expérimentation Avicole de Tours, UE1295, 37380, Nouzilly, France.
| | - Mireille Morisson
- UMR INRA, Génétique, Physiologie et Systèmes d'Elevage, INRA, 31328, Castanet Tolosan, France. .,INPT ENSAT, Génétique, Physiologie et Systèmes d'Elevage, INRA, 31328, Castanet Tolosan, France. .,INPT ENVT, Génétique, Physiologie et Systèmes d'Elevage, INRA, 31328, Castanet Tolosan, France.
| | - Frédérique Pitel
- UMR INRA, Génétique, Physiologie et Systèmes d'Elevage, INRA, 31328, Castanet Tolosan, France. .,INPT ENSAT, Génétique, Physiologie et Systèmes d'Elevage, INRA, 31328, Castanet Tolosan, France. .,INPT ENVT, Génétique, Physiologie et Systèmes d'Elevage, INRA, 31328, Castanet Tolosan, France.
| |
Collapse
|
36
|
Bionaz M, Osorio J, Loor JJ. TRIENNIAL LACTATION SYMPOSIUM: Nutrigenomics in dairy cows: Nutrients, transcription factors, and techniques1,2. J Anim Sci 2015; 93:5531-53. [DOI: 10.2527/jas.2015-9192] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- M. Bionaz
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis 97333
| | - J. Osorio
- Department of Animal and Rangeland Sciences, Oregon State University, Corvallis 97333
| | - J. J. Loor
- Department of Animal Sciences and Division of Nutritional Sciences, University of Illinois, Urbana 61801
| |
Collapse
|
37
|
Lee HS. Impact of Maternal Diet on the Epigenome during In Utero Life and the Developmental Programming of Diseases in Childhood and Adulthood. Nutrients 2015; 7:9492-507. [PMID: 26593940 PMCID: PMC4663595 DOI: 10.3390/nu7115467] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2015] [Revised: 10/18/2015] [Accepted: 10/26/2015] [Indexed: 02/06/2023] Open
Abstract
Exposure to environmental factors in early life can influence developmental processes and long-term health in humans. Early life nutrition and maternal diet are well-known examples of conditions shown to influence the risk of developing metabolic diseases, including type 2 diabetes mellitus and cardiovascular diseases, in adulthood. It is increasingly accepted that environmental compounds, including nutrients, can produce changes in the genome activity that, in spite of not altering the DNA sequence, can produce important, stable and, in some instances, transgenerational alterations in the phenotype. Epigenetics refers to changes in gene function that cannot be explained by changes in the DNA sequence, with DNA methylation patterns/histone modifications that can make important contributions to epigenetic memory. The epigenome can be considered as an interface between the genome and the environment that is central to the generation of phenotypes and their stability throughout the life course. To better understand the role of maternal health and nutrition in the initiation and progression of diseases in childhood and adulthood, it is necessary to identify the physiological and/or pathological roles of specific nutrients on the epigenome and how dietary interventions in utero and early life could modulate disease risk through epigenomic alteration.
Collapse
Affiliation(s)
- Ho-Sun Lee
- Epigenetics Group, International Agency for Research on Cancer (IARC), 150 Cours Albert-Thomas, 69372 Cedex 08, France.
| |
Collapse
|
38
|
Eckert JJ, Velazquez MA, Fleming TP. Cell signalling during blastocyst morphogenesis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 843:1-21. [PMID: 25956293 DOI: 10.1007/978-1-4939-2480-6_1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Blastocyst morphogenesis is prepared for even before fertilisation. Information stored within parental gametes can influence both maternal and embryonic gene expression programmes after egg activation at fertilisation. A complex network of intrinsic, cell-cell mediated and extrinsic, embryo-environment signalling mechanisms operates throughout cleavage, compaction and cavitation. These signalling events not only ensure developmental progression, cell differentiation and lineage allocation to inner cell mass (embryo proper) and trophectoderm (future extraembryonic lineages) but also provide a degree of developmental plasticity ensuring survival in prevailing conditions by adaptive responses. Indeed, many cellular functions including differentiation, metabolism, gene expression and gene expression regulation are subject to plasticity with short- or long-term consequences even into adult life. The interplay between intrinsic and extrinsic signals impacting on blastocyst morphogenesis is becoming clearer. This has been best studied in the mouse which will be the focus of this chapter but translational significance to human and domestic animal embryology will be a focus in future years.
Collapse
Affiliation(s)
- Judith J Eckert
- Human Development and Health, Faculty of Medicine, University of Southampton, SO16 6YD, Southampton, UK
| | | | | |
Collapse
|
39
|
Ibeagha-Awemu EM, Zhao X. Epigenetic marks: regulators of livestock phenotypes and conceivable sources of missing variation in livestock improvement programs. Front Genet 2015; 6:302. [PMID: 26442116 PMCID: PMC4585011 DOI: 10.3389/fgene.2015.00302] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Accepted: 09/11/2015] [Indexed: 12/30/2022] Open
Abstract
Improvement in animal productivity has been achieved over the years through careful breeding and selection programs. Today, variations in the genome are gaining increasing importance in livestock improvement strategies. Genomic information alone, however, explains only a part of the phenotypic variance in traits. It is likely that a portion of the unaccounted variance is embedded in the epigenome. The epigenome encompasses epigenetic marks such as DNA methylation, histone tail modifications, chromatin remodeling, and other molecules that can transmit epigenetic information such as non-coding RNA species. Epigenetic factors respond to external or internal environmental cues such as nutrition, pathogens, and climate, and have the ability to change gene expression leading to emergence of specific phenotypes. Accumulating evidence shows that epigenetic marks influence gene expression and phenotypic outcome in livestock species. This review examines available evidence of the influence of epigenetic marks on livestock (cattle, sheep, goat, and pig) traits and discusses the potential for consideration of epigenetic markers in livestock improvement programs. However, epigenetic research activities on farm animal species are currently limited partly due to lack of recognition, funding and a global network of researchers. Therefore, considerable less attention has been given to epigenetic research in livestock species in comparison to extensive work in humans and model organisms. Elucidating therefore the epigenetic determinants of animal diseases and complex traits may represent one of the principal challenges to use epigenetic markers for further improvement of animal productivity.
Collapse
Affiliation(s)
- Eveline M. Ibeagha-Awemu
- Dairy and Swine Research and Development Centre, Agriculture and Agri-Food CanadaSherbrooke, QC, Canada
| | - Xin Zhao
- Department of Animal Science, McGill University, Ste-Anne-De-BellevueQC, Canada
| |
Collapse
|
40
|
Pourié G, Martin N, Bossenmeyer-Pourié C, Akchiche N, Guéant-Rodriguez RM, Geoffroy A, Jeannesson E, El Hajj Chehadeh S, Mimoun K, Brachet P, Koziel V, Alberto JM, Helle D, Debard R, Leininger B, Daval JL, Guéant JL. Folate- and vitamin B12-deficient diet during gestation and lactation alters cerebellar synapsin expression via impaired influence of estrogen nuclear receptor α. FASEB J 2015; 29:3713-25. [PMID: 26018677 DOI: 10.1096/fj.14-264267] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 05/11/2015] [Indexed: 02/06/2023]
Abstract
Deficiency in the methyl donors vitamin B12 and folate during pregnancy and postnatal life impairs proper brain development. We studied the consequences of this combined deficiency on cerebellum plasticity in offspring from rat mothers subjected to deficient diet during gestation and lactation and in rat neuroprogenitor cells expressing cerebellum markers. The major proteomic change in cerebellum of 21-d-old deprived females was a 2.2-fold lower expression of synapsins, which was confirmed in neuroprogenitors cultivated in the deficient condition. A pathway analysis suggested that these proteomic changes were related to estrogen receptor α (ER-α)/Src tyrosine kinase. The influence of impaired ER-α pathway was confirmed by abnormal negative geotaxis test at d 19-20 and decreased phsophorylation of synapsins in deprived females treated by ER-α antagonist 1,3-bis(4-hydroxyphenyl)-4-methyl-5-[4-(2-piperidinylethoxy)phenol]-1H-pyrazole dihydrochloride (MPP). This effect was consistent with 2-fold decreased expression and methylation of ER-α and subsequent decreased ER-α/PPAR-γ coactivator 1 α (PGC-1α) interaction in deficiency condition. The impaired ER-α pathway led to decreased expression of synapsins through 2-fold decreased EGR-1/Zif-268 transcription factor and to 1.7-fold reduced Src-dependent phosphorylation of synapsins. The treatment of neuroprogenitors with either MPP or PP1 (4-(4'-phenoxyanilino)-6,7-dimethoxyquinazoline, 6,7-dimethoxy-N-(4-phenoxyphenyl)-4-quinazolinamine, SKI-1, Src-l1) Src inhibitor produced similar effects. In conclusion, the deficiency during pregnancy and lactation impairs the expression of synapsins through a deregulation of ER-α pathway.
Collapse
Affiliation(s)
- Grégory Pourié
- *Institut National de la Santé et de la Recherche Médicale, Unité 954, Nutrition-Genetics and Environmental Exposure, Medical Faculty and University Hospital Center, Nancy University, Vandoeuvre lès Nancy, France; Human Nutrition Unit, Unité Mixte de Recherche 1019 Institut National de la Recherche Agronomique/University of Auvergne, Institut National de la Recherche Agronomique Centre of Theix, Saint-Genès Champanelle, France; and Istituto di Ricovero e Cura a Carattere Scientifico, Oasi Maria Santissima-Institute for Research on Mental Retardation and Brain Aging, Troina, Italy
| | - Nicolas Martin
- *Institut National de la Santé et de la Recherche Médicale, Unité 954, Nutrition-Genetics and Environmental Exposure, Medical Faculty and University Hospital Center, Nancy University, Vandoeuvre lès Nancy, France; Human Nutrition Unit, Unité Mixte de Recherche 1019 Institut National de la Recherche Agronomique/University of Auvergne, Institut National de la Recherche Agronomique Centre of Theix, Saint-Genès Champanelle, France; and Istituto di Ricovero e Cura a Carattere Scientifico, Oasi Maria Santissima-Institute for Research on Mental Retardation and Brain Aging, Troina, Italy
| | - Carine Bossenmeyer-Pourié
- *Institut National de la Santé et de la Recherche Médicale, Unité 954, Nutrition-Genetics and Environmental Exposure, Medical Faculty and University Hospital Center, Nancy University, Vandoeuvre lès Nancy, France; Human Nutrition Unit, Unité Mixte de Recherche 1019 Institut National de la Recherche Agronomique/University of Auvergne, Institut National de la Recherche Agronomique Centre of Theix, Saint-Genès Champanelle, France; and Istituto di Ricovero e Cura a Carattere Scientifico, Oasi Maria Santissima-Institute for Research on Mental Retardation and Brain Aging, Troina, Italy
| | - Nassila Akchiche
- *Institut National de la Santé et de la Recherche Médicale, Unité 954, Nutrition-Genetics and Environmental Exposure, Medical Faculty and University Hospital Center, Nancy University, Vandoeuvre lès Nancy, France; Human Nutrition Unit, Unité Mixte de Recherche 1019 Institut National de la Recherche Agronomique/University of Auvergne, Institut National de la Recherche Agronomique Centre of Theix, Saint-Genès Champanelle, France; and Istituto di Ricovero e Cura a Carattere Scientifico, Oasi Maria Santissima-Institute for Research on Mental Retardation and Brain Aging, Troina, Italy
| | - Rosa Maria Guéant-Rodriguez
- *Institut National de la Santé et de la Recherche Médicale, Unité 954, Nutrition-Genetics and Environmental Exposure, Medical Faculty and University Hospital Center, Nancy University, Vandoeuvre lès Nancy, France; Human Nutrition Unit, Unité Mixte de Recherche 1019 Institut National de la Recherche Agronomique/University of Auvergne, Institut National de la Recherche Agronomique Centre of Theix, Saint-Genès Champanelle, France; and Istituto di Ricovero e Cura a Carattere Scientifico, Oasi Maria Santissima-Institute for Research on Mental Retardation and Brain Aging, Troina, Italy
| | - Andréa Geoffroy
- *Institut National de la Santé et de la Recherche Médicale, Unité 954, Nutrition-Genetics and Environmental Exposure, Medical Faculty and University Hospital Center, Nancy University, Vandoeuvre lès Nancy, France; Human Nutrition Unit, Unité Mixte de Recherche 1019 Institut National de la Recherche Agronomique/University of Auvergne, Institut National de la Recherche Agronomique Centre of Theix, Saint-Genès Champanelle, France; and Istituto di Ricovero e Cura a Carattere Scientifico, Oasi Maria Santissima-Institute for Research on Mental Retardation and Brain Aging, Troina, Italy
| | - Elise Jeannesson
- *Institut National de la Santé et de la Recherche Médicale, Unité 954, Nutrition-Genetics and Environmental Exposure, Medical Faculty and University Hospital Center, Nancy University, Vandoeuvre lès Nancy, France; Human Nutrition Unit, Unité Mixte de Recherche 1019 Institut National de la Recherche Agronomique/University of Auvergne, Institut National de la Recherche Agronomique Centre of Theix, Saint-Genès Champanelle, France; and Istituto di Ricovero e Cura a Carattere Scientifico, Oasi Maria Santissima-Institute for Research on Mental Retardation and Brain Aging, Troina, Italy
| | - Sarah El Hajj Chehadeh
- *Institut National de la Santé et de la Recherche Médicale, Unité 954, Nutrition-Genetics and Environmental Exposure, Medical Faculty and University Hospital Center, Nancy University, Vandoeuvre lès Nancy, France; Human Nutrition Unit, Unité Mixte de Recherche 1019 Institut National de la Recherche Agronomique/University of Auvergne, Institut National de la Recherche Agronomique Centre of Theix, Saint-Genès Champanelle, France; and Istituto di Ricovero e Cura a Carattere Scientifico, Oasi Maria Santissima-Institute for Research on Mental Retardation and Brain Aging, Troina, Italy
| | - Khalid Mimoun
- *Institut National de la Santé et de la Recherche Médicale, Unité 954, Nutrition-Genetics and Environmental Exposure, Medical Faculty and University Hospital Center, Nancy University, Vandoeuvre lès Nancy, France; Human Nutrition Unit, Unité Mixte de Recherche 1019 Institut National de la Recherche Agronomique/University of Auvergne, Institut National de la Recherche Agronomique Centre of Theix, Saint-Genès Champanelle, France; and Istituto di Ricovero e Cura a Carattere Scientifico, Oasi Maria Santissima-Institute for Research on Mental Retardation and Brain Aging, Troina, Italy
| | - Patrick Brachet
- *Institut National de la Santé et de la Recherche Médicale, Unité 954, Nutrition-Genetics and Environmental Exposure, Medical Faculty and University Hospital Center, Nancy University, Vandoeuvre lès Nancy, France; Human Nutrition Unit, Unité Mixte de Recherche 1019 Institut National de la Recherche Agronomique/University of Auvergne, Institut National de la Recherche Agronomique Centre of Theix, Saint-Genès Champanelle, France; and Istituto di Ricovero e Cura a Carattere Scientifico, Oasi Maria Santissima-Institute for Research on Mental Retardation and Brain Aging, Troina, Italy
| | - Violette Koziel
- *Institut National de la Santé et de la Recherche Médicale, Unité 954, Nutrition-Genetics and Environmental Exposure, Medical Faculty and University Hospital Center, Nancy University, Vandoeuvre lès Nancy, France; Human Nutrition Unit, Unité Mixte de Recherche 1019 Institut National de la Recherche Agronomique/University of Auvergne, Institut National de la Recherche Agronomique Centre of Theix, Saint-Genès Champanelle, France; and Istituto di Ricovero e Cura a Carattere Scientifico, Oasi Maria Santissima-Institute for Research on Mental Retardation and Brain Aging, Troina, Italy
| | - Jean-Marc Alberto
- *Institut National de la Santé et de la Recherche Médicale, Unité 954, Nutrition-Genetics and Environmental Exposure, Medical Faculty and University Hospital Center, Nancy University, Vandoeuvre lès Nancy, France; Human Nutrition Unit, Unité Mixte de Recherche 1019 Institut National de la Recherche Agronomique/University of Auvergne, Institut National de la Recherche Agronomique Centre of Theix, Saint-Genès Champanelle, France; and Istituto di Ricovero e Cura a Carattere Scientifico, Oasi Maria Santissima-Institute for Research on Mental Retardation and Brain Aging, Troina, Italy
| | - Deborah Helle
- *Institut National de la Santé et de la Recherche Médicale, Unité 954, Nutrition-Genetics and Environmental Exposure, Medical Faculty and University Hospital Center, Nancy University, Vandoeuvre lès Nancy, France; Human Nutrition Unit, Unité Mixte de Recherche 1019 Institut National de la Recherche Agronomique/University of Auvergne, Institut National de la Recherche Agronomique Centre of Theix, Saint-Genès Champanelle, France; and Istituto di Ricovero e Cura a Carattere Scientifico, Oasi Maria Santissima-Institute for Research on Mental Retardation and Brain Aging, Troina, Italy
| | - Renée Debard
- *Institut National de la Santé et de la Recherche Médicale, Unité 954, Nutrition-Genetics and Environmental Exposure, Medical Faculty and University Hospital Center, Nancy University, Vandoeuvre lès Nancy, France; Human Nutrition Unit, Unité Mixte de Recherche 1019 Institut National de la Recherche Agronomique/University of Auvergne, Institut National de la Recherche Agronomique Centre of Theix, Saint-Genès Champanelle, France; and Istituto di Ricovero e Cura a Carattere Scientifico, Oasi Maria Santissima-Institute for Research on Mental Retardation and Brain Aging, Troina, Italy
| | - Brigitte Leininger
- *Institut National de la Santé et de la Recherche Médicale, Unité 954, Nutrition-Genetics and Environmental Exposure, Medical Faculty and University Hospital Center, Nancy University, Vandoeuvre lès Nancy, France; Human Nutrition Unit, Unité Mixte de Recherche 1019 Institut National de la Recherche Agronomique/University of Auvergne, Institut National de la Recherche Agronomique Centre of Theix, Saint-Genès Champanelle, France; and Istituto di Ricovero e Cura a Carattere Scientifico, Oasi Maria Santissima-Institute for Research on Mental Retardation and Brain Aging, Troina, Italy
| | - Jean-Luc Daval
- *Institut National de la Santé et de la Recherche Médicale, Unité 954, Nutrition-Genetics and Environmental Exposure, Medical Faculty and University Hospital Center, Nancy University, Vandoeuvre lès Nancy, France; Human Nutrition Unit, Unité Mixte de Recherche 1019 Institut National de la Recherche Agronomique/University of Auvergne, Institut National de la Recherche Agronomique Centre of Theix, Saint-Genès Champanelle, France; and Istituto di Ricovero e Cura a Carattere Scientifico, Oasi Maria Santissima-Institute for Research on Mental Retardation and Brain Aging, Troina, Italy
| | - Jean-Louis Guéant
- *Institut National de la Santé et de la Recherche Médicale, Unité 954, Nutrition-Genetics and Environmental Exposure, Medical Faculty and University Hospital Center, Nancy University, Vandoeuvre lès Nancy, France; Human Nutrition Unit, Unité Mixte de Recherche 1019 Institut National de la Recherche Agronomique/University of Auvergne, Institut National de la Recherche Agronomique Centre of Theix, Saint-Genès Champanelle, France; and Istituto di Ricovero e Cura a Carattere Scientifico, Oasi Maria Santissima-Institute for Research on Mental Retardation and Brain Aging, Troina, Italy
| |
Collapse
|
41
|
Early-life exposure to high-fat diet may predispose rats to gender-specific hepatic fat accumulation by programming Pepck expression. J Nutr Biochem 2015; 26:433-40. [DOI: 10.1016/j.jnutbio.2014.10.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 09/28/2014] [Accepted: 10/16/2014] [Indexed: 12/25/2022]
|
42
|
Maternal high-fat-diet programs rat offspring liver fatty acid metabolism. Lipids 2015; 50:565-73. [PMID: 25899040 DOI: 10.1007/s11745-015-4018-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 04/02/2015] [Indexed: 12/28/2022]
Abstract
In offspring exposed in utero to a maternal diet high in fat (HF), we have previously demonstrated that despite similar birth weights, HF adult offspring at 6 months of age had significantly higher body weights, greater adiposity, and increased triacylglycerol (TAG) levels as compared to controls. We hypothesized that a maternal HF diet predisposes to offspring adiposity via a programmed increase in the synthesis of monounsaturated fatty acids in the liver and hence increased substrate availability for liver TAG synthesis. We further hypothesized that programmed changes in offspring liver fatty acid metabolism are associated with increased liver expression of the lipogenic enzyme stearoyl-CoA desaturase-1 (SCD-1). Female rats were maintained on a HF diet rich in monounsaturated fatty acids (MUFA) prior to and throughout pregnancy and lactation. After birth, newborns were nursed by the same dam, and all offspring were weaned to control diet. Plasma and liver fatty acid compositions were determined using gas chromatography/mass spectrometry. Fatty acid C16 desaturation indices of palmitoleic/palmitic and (vaccenic + palmitoleic)/palmitic and the C18 desaturation index of oleic/stearic were calculated. Liver protein abundance of SCD-1 was analyzed in newborns and adult offspring. Plasma and liver C16 desaturation indices were decreased in HF newborns, but increased in the adult offspring. Liver SCD-1 expression was increased in the HF adult offspring. These data show that the maternal HF diet during pregnancy and lactation increases offspring liver SCD-1 protein abundance and alters the liver C16 desaturase pathway.
Collapse
|