1
|
Vidyadharan VA, Betancourt A, Smith C, Blesson CS, Yallampalli C. Maternal Low-Protein Diet Leads to Mitochondrial Dysfunction and Impaired Energy Metabolism in the Skeletal Muscle of Male Rats. Int J Mol Sci 2024; 25:12860. [PMID: 39684571 DOI: 10.3390/ijms252312860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/18/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
A prenatal low-protein (LP) diet disrupts glucose homeostasis in adult offspring. Skeletal muscles are one of the main sites of glucose clearance, and mitochondria residing in the muscle fibers are central to glucose homeostasis. Our previous studies indicated that impaired mitochondrial health is central to dysregulated glucose metabolism in the gastrocnemius muscle of the LP-programmed female rats. In addition, dysfunctional mitochondria are often an indicator of underlying irregularities in energy metabolism and metabolic inflexibility. Therefore, this study examined the mitochondrial function and metabolic flexibility in the skeletal muscles of prenatal LP-programmed adult male rats. Pregnant Wistar rats were randomly allotted to a control diet (20% protein) or an isocaloric LP diet (6% protein). Standard laboratory rat chow was given to the dams and the pups after delivery and weaning. Gene and protein expressions, mtDNA copy number, and electron microscopy were assessed in gastrocnemius (GS) muscle, and the mitochondrial oxygen consumption rate was determined using isolated flexor digitorum brevis muscle fibers. The genes associated with mitochondrial outer membrane fusion, mitofusin1 and 2 (Mfn1 and Mfn2), fission (Fis1), and biogenesis (Pgc1B, Nrf1, and Esrra) were lower in the LP group. Further, our functional studies showed that the ATP-linked oxygen consumption rate (OCR), maximal, spare respiratory, and non-mitochondrial respiration-associated OCRs were lower in the LP rats. Further, the mRNA and protein expressions of Ndufb8, a key factor involved in the complex-I catalytic activity, were downregulated in the LP group. In addition, the expression of genes linked to mitochondrial pyruvate transport (Mpc1) and metabolism (Pdha1) was lower in the LP group. In contrast, the expression of mitochondrial fatty acid transporters (Cpt1a and Cpt2) was higher in the LP when compared to the control group. However, electron microscopic analysis exhibited no difference in the mitochondrial ultrastructure in the LP muscle compared to the control. Altogether, our results indicate that the LP diet affects the mitochondrial complex-I integrity and dynamics and leads to altered expression of genes associated with substrate oxidation and mitochondrial dysfunction in the skeletal muscle of the male LP offspring.
Collapse
Affiliation(s)
- Vipin A Vidyadharan
- Basic Sciences Perinatology Research Laboratories, Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ancizar Betancourt
- Basic Sciences Perinatology Research Laboratories, Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Craig Smith
- Agilent Technologies Inc., Santa Clara, CA 95051, USA
| | - Chellakkan S Blesson
- Reproductive Endocrinology and Infertility Division, Baylor College of Medicine, Houston, TX 77030, USA
- Family Fertility Center, Texas Children's Hospital, Houston, TX 77030, USA
| | - Chandra Yallampalli
- Basic Sciences Perinatology Research Laboratories, Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
2
|
Sun H, Chen M, Liao J, He L, Wan B, Yin J, Zhang X. The maternal lifestyle in pregnancy: Implications for foetal skeletal muscle development. J Cachexia Sarcopenia Muscle 2024; 15:1641-1650. [PMID: 39155495 PMCID: PMC11446712 DOI: 10.1002/jcsm.13556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/20/2024] [Accepted: 07/15/2024] [Indexed: 08/20/2024] Open
Abstract
The world is facing a global nutrition crisis, as evidenced by the rising incidence of metabolic disorders such as obesity, insulin resistance and chronic inflammation. Skeletal muscle is the largest tissue in humans and plays an important role in movement and host metabolism. Muscle fibre formation occurs mainly during the embryonic stage. Therefore, maternal lifestyle, especially nutrition and exercise during pregnancy, has a critical influence on foetal skeletal muscle development and the subsequent metabolic health of the offspring. In this review, the influence of maternal obesity, malnutrition and micronutrient intake on foetal skeletal muscle development is systematically summarized. We also aim to describe how maternal exercise shapes foetal muscle development and metabolic health in the offspring. The role of maternal gut microbiota and its metabolites on foetal muscle development is further discussed, although this field is still in its 'infancy'. This review will provide new insights to reduce the global crisis of metabolic disorders and highlight current gaps to promote further research.
Collapse
Affiliation(s)
- Haijun Sun
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and TechnologyChina Agricultural UniversityBeijingChina
| | - Meixia Chen
- Institute of Animal Husbandry and Veterinary MedicineBeijing Academy of Agriculture and Forestry SciencesBeijingChina
| | - Jialong Liao
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and TechnologyChina Agricultural UniversityBeijingChina
| | - Linjuan He
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and TechnologyChina Agricultural UniversityBeijingChina
| | - Boyang Wan
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and TechnologyChina Agricultural UniversityBeijingChina
| | - Jingdong Yin
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and TechnologyChina Agricultural UniversityBeijingChina
- Frontiers Science Center for Molecular Design Breeding (MOE)BeijingChina
| | - Xin Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and TechnologyChina Agricultural UniversityBeijingChina
- Frontiers Science Center for Molecular Design Breeding (MOE)BeijingChina
| |
Collapse
|
3
|
Hill DJ, Hill TG. Maternal diet during pregnancy and adaptive changes in the maternal and fetal pancreas have implications for future metabolic health. Front Endocrinol (Lausanne) 2024; 15:1456629. [PMID: 39377073 PMCID: PMC11456468 DOI: 10.3389/fendo.2024.1456629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 08/28/2024] [Indexed: 10/09/2024] Open
Abstract
Fetal and neonatal development is a critical period for the establishment of the future metabolic health and disease risk of an individual. Both maternal undernutrition and overnutrition can result in abnormal fetal organ development resulting in inappropriate birth size, child and adult obesity, and increased risk of Type 2 diabetes and cardiovascular diseases. Inappropriate adaptive changes to the maternal pancreas, placental function, and the development of the fetal pancreas in response to nutritional stress during pregnancy are major contributors to a risk trajectory in the offspring. This interconnected maternal-placental-fetal metabolic axis is driven by endocrine signals in response to the availability of nutritional metabolites and can result in cellular stress and premature aging in fetal tissues and the inappropriate expression of key genes involved in metabolic control as a result of long-lasting epigenetic changes. Such changes result is insufficient pancreatic beta-cell mass and function, reduced insulin sensitivity in target tissues such as liver and white adipose and altered development of hypothalamic satiety centres and in basal glucocorticoid levels. Whilst interventions in the obese mother such as dieting and increased exercise, or treatment with insulin or metformin in mothers who develop gestational diabetes, can improve metabolic control and reduce the risk of a large-for-gestational age infant, their effectiveness in changing the adverse metabolic trajectory in the child is as yet unclear.
Collapse
Affiliation(s)
- David J. Hill
- Lawson Health Research Institute, St. Joseph’s Health Care, London, ON, Canada
- Departments of Medicine, Physiology and Pharmacology, Western University, London, ON, Canada
| | - Thomas G. Hill
- Oxford Centre for Diabetes, Endocrinology, and Metabolism, Wellcome Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
4
|
Vidyadharan VA, Blesson CS, Tanchico D, Betancourt A, Smith C, Yallampalli C. Low Protein Programming Causes Increased Mitochondrial Fusion and Decreased Oxygen Consumption in the Hepatocytes of Female Rats. Nutrients 2023; 15:1568. [PMID: 37049409 PMCID: PMC10097083 DOI: 10.3390/nu15071568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/15/2023] [Accepted: 03/22/2023] [Indexed: 04/14/2023] Open
Abstract
The liver is one of the major organs involved in the regulation of glucose and lipid homeostasis. The effectiveness of metabolic activity in hepatocytes is determined by the quality and quantity of its mitochondria. Mitochondrial function is complex, and they act via various dynamic networks, which rapidly adapt to changes in the cellular milieu. Our present study aims to investigate the effects of low protein programming on the structure and function of mitochondria in the hepatocytes of adult females. Pregnant rats were fed with a control or isocaloric low-protein diet from gestational day 4 until delivery. A normal laboratory chow was given to all dams after delivery and to pups after weaning. The rats were euthanized at 4 months of age and the livers were collected from female offspring for investigating the mitochondrial structure, mtDNA copy number, mRNA, and proteins expression of genes associated with mitochondrial function. Primary hepatocytes were isolated and used for the analysis of the mitochondrial bioenergetics profiles. The mitochondrial ultrastructure showed that the in utero low-protein diet exposure led to increased mitochondrial fusion. Accordingly, there was an increase in the mRNA and protein levels of the mitochondrial fusion gene Opa1 and mitochondrial biogenesis genes Pgc1a and Essra, but Fis1, a fission gene, was downregulated. Low protein programming also impaired the mitochondrial function of the hepatocytes with a decrease in basal respiration ATP-linked respiration and proton leak. In summary, the present study suggests that the hepatic mitochondrial dysfunction induced by an in utero low protein diet might be a potential mechanism linking glucose intolerance and insulin resistance in adult offspring.
Collapse
Affiliation(s)
- Vipin A. Vidyadharan
- Basic Sciences Perinatology Research Laboratories, Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Chellakkan S. Blesson
- Reproductive Endocrinology and Infertility Division, Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX 77030, USA
- Family Fertility Center, Texas Children’s Hospital, Houston, TX 77030, USA
| | - Daren Tanchico
- Basic Sciences Perinatology Research Laboratories, Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ancizar Betancourt
- Basic Sciences Perinatology Research Laboratories, Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Craig Smith
- Agilent Technologies Inc., Santa Clara, CA 95051, USA
| | - Chandra Yallampalli
- Basic Sciences Perinatology Research Laboratories, Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
5
|
Effects of Parental Dietary Restriction on Offspring Fitness in Drosophila melanogaster. Nutrients 2023; 15:nu15051273. [PMID: 36904272 PMCID: PMC10005678 DOI: 10.3390/nu15051273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/18/2023] [Accepted: 03/02/2023] [Indexed: 03/08/2023] Open
Abstract
Dietary restriction (DR) is a well-established strategy to increase lifespan and stress resistance in many eukaryotic species. In addition, individuals fed a restricted diet typically reduce or completely shut down reproduction compared to individuals fed a full diet. Although the parental environment can lead to changes epigenetically in offspring gene expression, little is known about the role of the parental (F0) diet on the fitness of their offspring (F1). This study investigated the lifespan, stress resistance, development, body weight, fecundity, and feeding rate in offspring from parental flies exposed to a full or restricted diet. The offspring flies of the parental DR showed increases in body weight, resistance to various stressors, and lifespan, but the development and fecundity were unaffected. Interestingly, parental DR reduced the feeding rate of their offspring. This study suggests that the effect of DR can extend beyond the exposed individual to their offspring, and it should be considered in both theoretical and empirical studies of senescence.
Collapse
|
6
|
Pezzotta A, Perico L, Morigi M, Corna D, Locatelli M, Zoja C, Benigni A, Remuzzi G, Imberti B. Low Nephron Number Induced by Maternal Protein Restriction Is Prevented by Nicotinamide Riboside Supplementation Depending on Sirtuin 3 Activation. Cells 2022; 11:cells11203316. [PMID: 36291179 PMCID: PMC9600228 DOI: 10.3390/cells11203316] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 10/13/2022] [Accepted: 10/19/2022] [Indexed: 11/16/2022] Open
Abstract
A reduced nephron number at birth, due to critical gestational conditions, including maternal malnutrition, is associated with the risk of developing hypertension and chronic kidney disease in adulthood. No interventions are currently available to augment nephron number. We have recently shown that sirtuin 3 (SIRT3) has an important role in dictating proper nephron endowment. The present study explored whether SIRT3 stimulation, by means of supplementation with nicotinamide riboside (NR), a precursor of the SIRT3 co-substrate nicotinamide adenine dinucleotide (NAD+), was able to improve nephron number in a murine model of a low protein (LP) diet. Our findings show that reduced nephron number in newborn mice (day 1) born to mothers fed a LP diet was associated with impaired renal SIRT3 expression, which was restored through supplementation with NR. Glomerular podocyte density, as well as the rarefaction of renal capillaries, also improved through NR administration. In mechanistic terms, the restoration of SIRT3 expression through NR was mediated by the induction of proliferator-activated receptor γ (PPARγ) coactivator-1α (PGC-1α). Moreover, NR restored SIRT3 activity, as shown by the reduction of the acetylation of optic atrophy 1 (OPA1) and superoxide dismutase 2 (SOD2), which resulted in improved mitochondrial morphology and protection against oxidative damage in mice born to mothers fed the LP diet. Our results provide evidence that it is feasible to prevent nephron mass shortage at birth through SIRT3 boosting during nephrogenesis, thus providing a therapeutic option to possibly limit the long-term sequelae of reduced nephron number in adulthood.
Collapse
|
7
|
Vipin VA, Blesson CS, Yallampalli C. Maternal low protein diet and fetal programming of lean type 2 diabetes. World J Diabetes 2022; 13:185-202. [PMID: 35432755 PMCID: PMC8984567 DOI: 10.4239/wjd.v13.i3.185] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/30/2021] [Accepted: 02/10/2022] [Indexed: 02/06/2023] Open
Abstract
Maternal nutrition is found to be the key factor that determines fetal health in utero and metabolic health during adulthood. Metabolic diseases have been primarily attributed to impaired maternal nutrition during pregnancy, and impaired nutrition has been an immense issue across the globe. In recent years, type 2 diabetes (T2D) has reached epidemic proportion and is a severe public health problem in many countries. Although plenty of research has already been conducted to tackle T2D which is associated with obesity, little is known regarding the etiology and pathophysiology of lean T2D, a variant of T2D. Recent studies have focused on the effects of epigenetic variation on the contribution of in utero origins of lean T2D, although other mechanisms might also contribute to the pathology. Observational studies in humans and experiments in animals strongly suggest an association between maternal low protein diet and lean T2D phenotype. In addition, clear sex-specific disease prevalence was observed in different studies. Consequently, more research is essential for the understanding of the etiology and pathophysiology of lean T2D, which might help to develop better disease prevention and treatment strategies. This review examines the role of protein insufficiency in the maternal diet as the central driver of the developmental programming of lean T2D.
Collapse
Affiliation(s)
- Vidyadharan Alukkal Vipin
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX 77030, United States
| | - Chellakkan Selvanesan Blesson
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX 77030, United States
- Family Fertility Center, Texas Children's Hospital, Houston, TX 77030, United States
| | - Chandra Yallampalli
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX 77030, United States
| |
Collapse
|
8
|
Prenatal Low-Protein Diet Affects Mitochondrial Structure and Function in the Skeletal Muscle of Adult Female Offspring. Nutrients 2022; 14:nu14061158. [PMID: 35334815 PMCID: PMC8954615 DOI: 10.3390/nu14061158] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 03/03/2022] [Accepted: 03/08/2022] [Indexed: 02/06/2023] Open
Abstract
Gestational low-protein (LP) diet leads to glucose intolerance and insulin resistance in adult offspring. We had earlier demonstrated that LP programming affects glucose disposal in females. Mitochondrial health is crucial for normal glucose metabolism in skeletal muscle. In this study, we sought to analyze mitochondrial structure, function, and associated genes in skeletal muscles to explore the molecular mechanism of insulin resistance LP-programmed female offspring. On day four of pregnancy, rats were assigned to a control diet containing 20% protein or an isocaloric 6% protein-containing diet. Standard laboratory diet was given to the dams after delivery until the end of weaning and to pups after weaning. Gestational LP diet led to changes in mitochondrial ultrastructure in the gastrocnemius muscles, including a nine-fold increase in the presence of giant mitochondria along with unevenly formed cristae. Further, functional analysis showed that LP programming caused impaired mitochondrial functions. Although the mitochondrial copy number did not show significant changes, key genes involved in mitochondrial structure and function such as Fis1, Opa1, Mfn2, Nrf1, Nrf2, Pgc1b, Cox4b, Esrra, and Vdac were dysregulated. Our study shows that prenatal LP programming induced disruption in mitochondrial ultrastructure and function in the skeletal muscle of female offspring.
Collapse
|
9
|
Sciascia QL, Prehn C, Adamski J, Daş G, Lang IS, Otten W, Görs S, Metges CC. The Effect of Dietary Protein Imbalance during Pregnancy on the Growth, Metabolism and Circulatory Metabolome of Neonatal and Weaned Juvenile Porcine Offspring. Nutrients 2021; 13:nu13093286. [PMID: 34579160 PMCID: PMC8471113 DOI: 10.3390/nu13093286] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 09/12/2021] [Accepted: 09/16/2021] [Indexed: 11/16/2022] Open
Abstract
Protein imbalance during pregnancy affects women in underdeveloped and developing countries and is associated with compromised offspring growth and an increased risk of metabolic diseases in later life. We studied in a porcine model the glucose and urea metabolism, and circulatory hormone and metabolite profile of offspring exposed during gestation, to maternal isoenergetic low-high (LP-HC), high-low (HP-LC) or adequate (AP) protein-carbohydrate ratio diets. At birth, LP-HC were lighter and the plasma acetylcarnitine to free carnitine ratios at 1 day of life was lower compared to AP offspring. Plasma urea concentrations were lower in 1 day old LP-HC offspring than HP-LC. In the juvenile period, increased insulin concentrations were observed in LP-HC and HP-LC offspring compared to AP, as was body weight from HP-LC compared to LP-HC. Plasma triglyceride concentrations were lower in 80 than 1 day old HP-LC offspring, and glucagon concentrations lower in 80 than 1 day old AP and HP-LC offspring. Plasma urea and the ratio of glucagon to insulin were lower in all 80 than 1 day old offspring. Aminoacyl-tRNA, arginine and phenylalanine, tyrosine and tryptophan metabolism, histidine and beta-alanine metabolism differed between 1 and 80 day old AP and HP-LC offspring. Maternal protein imbalance throughout pregnancy did not result in significant consequences in offspring metabolism compared to AP, indicating enormous plasticity by the placenta and developing offspring.
Collapse
Affiliation(s)
- Quentin L. Sciascia
- Institute of Nutritional Physiology ‘Oskar Kellner’, Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany; (Q.L.S.); (G.D.); (I.S.L.); (S.G.)
| | - Cornelia Prehn
- Metabolomics and Proteomics Core (MPC), Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany;
| | - Jerzy Adamski
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health, 85764 Neuherberg, Germany;
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Gürbüz Daş
- Institute of Nutritional Physiology ‘Oskar Kellner’, Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany; (Q.L.S.); (G.D.); (I.S.L.); (S.G.)
| | - Iris S. Lang
- Institute of Nutritional Physiology ‘Oskar Kellner’, Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany; (Q.L.S.); (G.D.); (I.S.L.); (S.G.)
| | - Winfried Otten
- Institute of Behavioural Physiology, Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany;
| | - Solvig Görs
- Institute of Nutritional Physiology ‘Oskar Kellner’, Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany; (Q.L.S.); (G.D.); (I.S.L.); (S.G.)
| | - Cornelia C. Metges
- Institute of Nutritional Physiology ‘Oskar Kellner’, Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany; (Q.L.S.); (G.D.); (I.S.L.); (S.G.)
- Chair of Nutritional Physiology and Animal Nutrition, Faculty of Agriculture and Environmental Sciences, University of Rostock, 18059 Rostock, Germany
- Correspondence: ; Tel.: +49-38208-68-650
| |
Collapse
|
10
|
Callet T, Li H, Coste P, Glise S, Heraud C, Maunas P, Mercier Y, Turonnet N, Zunzunegui C, Panserat S, Bolliet V, Marandel L. Modulation of Energy Metabolism and Epigenetic Landscape in Rainbow Trout Fry by a Parental Low Protein/High Carbohydrate Diet. BIOLOGY 2021; 10:biology10070585. [PMID: 34202225 PMCID: PMC8301017 DOI: 10.3390/biology10070585] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 06/17/2021] [Accepted: 06/20/2021] [Indexed: 12/13/2022]
Abstract
Simple Summary While the effects of parental diets on their progeny have been highly described in mammals, such studies are lacking in fish. To explore such a question in a high trophic level teleost fish, two-year old male and female rainbow trout were fed either a control diet (0% carbohydrate and 63.89% protein) or a high-carbohydrate diet (35% carbohydrate and 42.96% protein), for a complete reproductive cycle for females and for a period of 5 months for males. Neither the maternal nor the paternal high-carbohydrate diet alone had induced significant effects on their progeny. Nevertheless, when both parents were fed the high-carbohydrate diet, the energy metabolism and mitochondrial dynamics of their progeny were altered. Moreover, the epigenetic landscape was also highly affected. Even though, offspring growth was only slightly affected at the early stage of life; the effect of parental high-carbohydrate diet should be explored over the long term. Abstract It is now recognized that parental diets could highly affect offspring metabolism and growth. Studies in fish are, however, lacking. In particular, the effect of a parental diet high in carbohydrate (HC) and low in protein (LP) on progeny has never been examined in higher trophic level teleost fish. Thus, two-year old male and female rainbow trout (Oncorhynchus mykiss) were fed either a control diet (0% carbohydrate and 63.89% protein) or a diet containing 35% carbohydrate and 42.96% protein (HC/LP) for a complete reproductive cycle for females and over a 5-month period for males. Cross-fertilizations were then carried out. To evaluate the effect of the parental diet on their offspring, different phenotypic and metabolic traits were recorded for offspring before their first feeding and again three weeks later. When considering the paternal and maternal HC/LP nutrition independently, fry phenotypes and transcriptomes were only slightly affected. The combination of the maternal and paternal HC/LP diets altered the energy metabolism and mitochondrial dynamics of their progeny, demonstrating the existence of a synergistic effect. The global DNA methylation of whole fry was also highly affected by the HC/LP parental diet, indicating that it could be one of the fundamental mechanisms responsible for the effects of nutritional programming.
Collapse
Affiliation(s)
- Thérèse Callet
- INRAE, Université de Pau et des Pays de L’Adour, E2S UPPA, NUMEA, 64310 Saint-Pée-sur-Nivelle, France; (T.C.); (H.L.); (C.H.); (P.M.); (Y.M.); (N.T.); (C.Z.); (S.P.)
| | - Hongyan Li
- INRAE, Université de Pau et des Pays de L’Adour, E2S UPPA, NUMEA, 64310 Saint-Pée-sur-Nivelle, France; (T.C.); (H.L.); (C.H.); (P.M.); (Y.M.); (N.T.); (C.Z.); (S.P.)
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan 430072, China
- University of Chinese Academy of Sciences, Beijing 100190, China
| | - Pascale Coste
- INRAE, Université de Pau et des Pays de L’Adour, E2S UPPA, ECOBIOP, 64310 Saint-Pée-sur-Nivelle, France; (P.C.); (S.G.); (V.B.)
| | - Stéphane Glise
- INRAE, Université de Pau et des Pays de L’Adour, E2S UPPA, ECOBIOP, 64310 Saint-Pée-sur-Nivelle, France; (P.C.); (S.G.); (V.B.)
| | - Cécile Heraud
- INRAE, Université de Pau et des Pays de L’Adour, E2S UPPA, NUMEA, 64310 Saint-Pée-sur-Nivelle, France; (T.C.); (H.L.); (C.H.); (P.M.); (Y.M.); (N.T.); (C.Z.); (S.P.)
| | - Patrick Maunas
- INRAE, Université de Pau et des Pays de L’Adour, E2S UPPA, NUMEA, 64310 Saint-Pée-sur-Nivelle, France; (T.C.); (H.L.); (C.H.); (P.M.); (Y.M.); (N.T.); (C.Z.); (S.P.)
| | - Yvan Mercier
- INRAE, Université de Pau et des Pays de L’Adour, E2S UPPA, NUMEA, 64310 Saint-Pée-sur-Nivelle, France; (T.C.); (H.L.); (C.H.); (P.M.); (Y.M.); (N.T.); (C.Z.); (S.P.)
| | - Nicolas Turonnet
- INRAE, Université de Pau et des Pays de L’Adour, E2S UPPA, NUMEA, 64310 Saint-Pée-sur-Nivelle, France; (T.C.); (H.L.); (C.H.); (P.M.); (Y.M.); (N.T.); (C.Z.); (S.P.)
| | - Chloé Zunzunegui
- INRAE, Université de Pau et des Pays de L’Adour, E2S UPPA, NUMEA, 64310 Saint-Pée-sur-Nivelle, France; (T.C.); (H.L.); (C.H.); (P.M.); (Y.M.); (N.T.); (C.Z.); (S.P.)
| | - Stéphane Panserat
- INRAE, Université de Pau et des Pays de L’Adour, E2S UPPA, NUMEA, 64310 Saint-Pée-sur-Nivelle, France; (T.C.); (H.L.); (C.H.); (P.M.); (Y.M.); (N.T.); (C.Z.); (S.P.)
| | - Valérie Bolliet
- INRAE, Université de Pau et des Pays de L’Adour, E2S UPPA, ECOBIOP, 64310 Saint-Pée-sur-Nivelle, France; (P.C.); (S.G.); (V.B.)
| | - Lucie Marandel
- INRAE, Université de Pau et des Pays de L’Adour, E2S UPPA, NUMEA, 64310 Saint-Pée-sur-Nivelle, France; (T.C.); (H.L.); (C.H.); (P.M.); (Y.M.); (N.T.); (C.Z.); (S.P.)
- Correspondence:
| |
Collapse
|
11
|
Kim J, Choi A, Kwon YH. Maternal Protein Restriction Altered Insulin Resistance and Inflammation-Associated Gene Expression in Adipose Tissue of Young Adult Mouse Offspring in Response to a High-Fat Diet. Nutrients 2020; 12:nu12041103. [PMID: 32316103 PMCID: PMC7230574 DOI: 10.3390/nu12041103] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/09/2020] [Accepted: 04/14/2020] [Indexed: 12/27/2022] Open
Abstract
Maternal protein restriction is associated with increased risk of insulin resistance and inflammation in adulthood offspring. Here, we investigated whether maternal protein restriction could alter the risk of metabolic syndrome in postweaning high-fat (HF)-diet-challenged offspring, with focus on epididymal adipose tissue gene expression profile. Female ICR mice were fed a control (C) or a low-protein (LP) diet for two weeks before mating and throughout gestation and lactation, and their male offspring were fed an HF diet for 22 weeks (C/HF and LP/HF groups). A subset of offspring of control dams was fed a low-fat control diet (C/C group). In response to postweaning HF diet, serum insulin level and the homeostasis model assessment of insulin resistance (HOMA-IR) were increased in control offspring. Maternal LP diet decreased HOMA-IR and adipose tissue inflammation, and increased serum adiponectin level in the HF-diet-challenged offspring. Accordingly, functional analysis revealed that differentially expressed genes (DEGs) enriched in cytokine production were downregulated in the LP/HF group compared to the C/HF group. We also observed the several annotated gene ontology terms associated with innate immunity and phagocytosis in down-regulated DEGs between LP/HF and C/C groups. In conclusion, maternal protein restriction alleviated insulin resistance and inflammation in young offspring mice fed a HF diet but may impair development of immune system in offspring.
Collapse
Affiliation(s)
- Juhae Kim
- Department of Food and Nutrition, Seoul National University, Seoul 08826, Korea; (J.K.); (A.C.)
| | - Alee Choi
- Department of Food and Nutrition, Seoul National University, Seoul 08826, Korea; (J.K.); (A.C.)
| | - Young Hye Kwon
- Department of Food and Nutrition, Seoul National University, Seoul 08826, Korea; (J.K.); (A.C.)
- Research Institute of Human Ecology, Seoul National University, Seoul 08826, Korea
- Correspondence: ; Tel.: +82-2-880-6833
| |
Collapse
|
12
|
Abstract
Amino acids are not only the building blocks of proteins, an indispensable component of cells, but also play versatile roles in regulating cell metabolism, proliferation, differentiation and growth by themselves or through their derivatives. At the whole body level, the bioavailability and metabolism of amino acids, interacting with other macronutrients, is critical for the physiological processes of reproduction including gametogenesis, fertilization, implantation, placentation, fetal growth and development. In fertilization and early pregnancy, histotroph in oviductal and uterine secretions provides nutrients and microenvironment for conceptus (embryo and extraembryonic membranes) development. These nutrients include select amino acids in histotroph (arginine, leucine and glutamine of particular interest) that stimulate conceptus growth and development, as well as interactions between maternal uterus and the conceptus, thus impacting maintenance of pregnancy, placental growth, development and functions, fetal growth and development, and consequential pregnancy outcomes. Gestational protein undernutrition causes fetal growth restriction and predisposes cardiovascular, metabolic diseases and others in offspring via multiple mechanisms, whereas the supplementation of glycine, leucine and taurine during pregnancy partially rescues growth restriction and beneficially modulates fetal programming. Thus, amino acids are essential for the fertility of humans and all animals.
Collapse
Affiliation(s)
- Haijun Gao
- Department of Obstetrics & Gynecology, Howard University College of Medicine, Washington, DC, USA.
| |
Collapse
|
13
|
Seidel U, Huebbe P, Rimbach G. Taurine: A Regulator of Cellular Redox Homeostasis and Skeletal Muscle Function. Mol Nutr Food Res 2018; 63:e1800569. [DOI: 10.1002/mnfr.201800569] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 08/10/2018] [Indexed: 12/18/2022]
Affiliation(s)
- Ulrike Seidel
- Institute of Human Nutrition and Food ScienceUniversity of Kiel Kiel Germany
| | - Patricia Huebbe
- Institute of Human Nutrition and Food ScienceUniversity of Kiel Kiel Germany
| | - Gerald Rimbach
- Institute of Human Nutrition and Food ScienceUniversity of Kiel Kiel Germany
| |
Collapse
|
14
|
Larsen LH, Sandø-Pedersen S, Ørstrup LKH, Grunnet N, Quistorff B, Mortensen OH. Gestational Protein Restriction in Wistar Rats; Effect of Taurine Supplementation on Properties of Newborn Skeletal Muscle. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 975 Pt 1:413-433. [PMID: 28849472 DOI: 10.1007/978-94-024-1079-2_34] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Taurine ameliorates changes occurring in newborn skeletal muscle as a result of gestational protein restriction in C57BL/6 mice, but taurine supplementation effects may be exaggerated in C57BL/6 mice due to their inherent excessive taurinuria.We examined if maternal taurine supplementation could ameliorate changes in gene expression levels, properties of mitochondria, myogenesis, and nutrient transport and sensing, in male newborn skeletal muscle caused by a maternal low protein (LP) diet in Wistar rats.LP diet resulted in an 11% non-significant decrease in birth weight, which was not rescued by taurine supplementation (LP-Tau). LP-Tau offspring had significantly lower birth weight compared to controls. Gene expression profiling revealed 895 significantly changed genes, mainly an LP-induced down-regulation of genes involved in protein translation. Taurine fully or partially rescued 32% of these changes, but with no distinct pattern as to which genes were rescued.Skeletal muscle taurine content in LP-Tau offspring was increased, but no changes in mRNA levels of the taurine synthesis pathway were observed. Taurine transporter mRNA levels, but not protein levels, were increased by LP diet.Nutrient sensing signaling pathways were largely unaffected in LP or LP-Tau groups, although taurine supplementation caused a decrease in total Akt and AMPK protein levels. PAT4 amino acid transporter mRNA was increased by LP, and normalized by taurine supplementation.In conclusion, gestational protein restriction in rats decreased genes involved in protein translation in newborn skeletal muscle and led to changes in nutrient transporters. Taurine partly rescued these changes, hence underscoring the importance of taurine in development.
Collapse
Affiliation(s)
- Lea Hüche Larsen
- Department of Biomedical Sciences, Cellular and Metabolic Research Section, University of Copenhagen, København, Denmark
| | - Sofie Sandø-Pedersen
- Department of Biomedical Sciences, Cellular and Metabolic Research Section, University of Copenhagen, København, Denmark
| | - Laura Kofoed Hvidsten Ørstrup
- Department of Biomedical Sciences, Cellular and Metabolic Research Section, University of Copenhagen, København, Denmark
| | - Niels Grunnet
- Department of Biomedical Sciences, Cellular and Metabolic Research Section, University of Copenhagen, København, Denmark
| | - Bjørn Quistorff
- Department of Biomedical Sciences, Cellular and Metabolic Research Section, University of Copenhagen, København, Denmark
| | - Ole Hartvig Mortensen
- Department of Biomedical Sciences, Cellular and Metabolic Research Section, University of Copenhagen, København, Denmark.
| |
Collapse
|
15
|
Strategies to reduce non-communicable diseases in the offspring: negative and positive in utero programming. J Dev Orig Health Dis 2018; 9:642-652. [PMID: 30111388 DOI: 10.1017/s2040174418000569] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Non-communicable diseases (NCDs) are a major problem as they are the leading cause of death and represent a substantial economic cost. The 'Developmental Origins of Health and Disease Hypothesis' proposes that adverse stimuli at different life stages can increase the predisposition to these diseases. In fact, adverse in utero programming is a major origin of these diseases due to the high malleability of embryonic development. This review provides a comprehensive analysis of the scientific literature on in utero programming and NCDs highlighting potential medical strategies to prevent these diseases based upon this programming. We fully address the concept and mechanisms involved in this programming (anatomical disruptions, epigenetic modifications and microbiota alterations). We also examine the negative role of in utero programming on the increased predisposition of NCDs in the offspring, which introduces the passive medical approach that consists of avoiding adverse stimuli including an unhealthy diet and environmental chemicals. Finally, we extensively discuss active medical approaches that target the causes of NCDs and have the potential to significantly and rapidly reduce the incidence of NCDs. These approaches can be classified as direct in utero programming modifications and personalized lifestyle pregnancy programs; they could potentially provide transgenerational NCDs protection. Active strategies against NCDs constitute a promising tool for the reduction in NCDs.
Collapse
|
16
|
Shen L, Gan M, Zhang S, Ma J, Tang G, Jiang Y, Li M, Wang J, Li X, Che L, Zhu L. Transcriptome Analyses Reveal Adult Metabolic Syndrome With Intrauterine Growth Restriction in Pig Models. Front Genet 2018; 9:291. [PMID: 30158951 PMCID: PMC6103486 DOI: 10.3389/fgene.2018.00291] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Accepted: 07/12/2018] [Indexed: 12/23/2022] Open
Abstract
Epidemiological data have indicated that intrauterine growth retardation (IUGR) is a risk factor for the adult metabolic syndrome in pigs. However, the causative genetic mechanism leading to the phenotype in adulthood has not been well characterized. In the present study, both normal and IUGR adult pigs were used as models to survey the differences in global gene expression in livers through transcriptome sequencing. The transcriptome libraries generated 104.54 gb of data. In normal and IUGR pigs, 16,948 and 17,078 genes were expressed, respectively. A total of 1,322 differentially expressed genes (DEGs) were identified. Enrichment analysis of the DEGs revealed that the top overrepresented gene ontology (GO) terms and pathways were related to oxidoreductase activity, ATPase activity, amino catabolic process, glucose metabolism, and insulin signaling pathway. The increased gluconeogenesis (GNG) and decreased glycogen synthesis in the liver contributed to the glucose intolerance observed in IUGR. The reduced expression of insulin signaling genes (such as PI3K and AKT) indicated an elevated risk of diabetes in adulthood. Together, these findings provide a comprehensive understanding of the molecular mechanisms of adult IUGR pigs and valuable information for future studies of therapeutic intervention in IUGR metabolic syndrome.
Collapse
Affiliation(s)
- Linyuan Shen
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China.,Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - Mailin Gan
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Shunhua Zhang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Jideng Ma
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Guoqing Tang
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Yanzhi Jiang
- College of Life Science, Sichuan Agricultural University, Chengdu, China
| | - Mingzhou Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Jinyong Wang
- Chongqing Academy of Animal Science, Chongqing, China
| | - Xuewei Li
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| | - Lianqiang Che
- Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu, China
| | - Li Zhu
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
17
|
Chen S, Zhang M, Bo L, Li S, Hu L, Zhao X, Sun C. Metabolomic analysis of the toxic effect of chronic exposure of cadmium on rat urine. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2018; 25:3765-3774. [PMID: 29168138 DOI: 10.1007/s11356-017-0774-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 11/15/2017] [Indexed: 06/07/2023]
Abstract
This study aimed to assess the toxic effect of chronic exposure to cadmium through a metabolomic approach based on ultra-performance liquid chromatography/mass spectrometry (UPLC-MS). Forty male Sprague-Dawley rats were randomly assigned to the following groups: control, low-dose cadmium chloride (CdCl2) (0.13 mg/kg body weight (bw)), middle-dose CdCl2 (0.8/kg bw), and high-dose CdCl2 (4.9 mg/kg bw). The rats continuously received CdCl2 via drinking water for 24 weeks. Rat urine samples were then collected at different time points to establish the metabolomic profiles. Multiple statistical analyses with principal component analysis and partial least squares-discriminant analysis were used to investigate the metabolomic profile changes in the urine samples and screen for potential biomarkers. Thirteen metabolites were identified from the metabolomic profiles of rat urine after treatment. Compared with the control group, the treated groups showed significantly increased intensities of phenylacetylglycine, guanidinosuccinic acid, 4-pyridoxic acid, 4-aminohippuric acid, 4-guanidinobutanoic acid, allantoic acid, dopamine, LysoPC(18:2(9Z,12Z)), and L-urobilinogen. By contrast, the intensities of creatinine, L-carnitine, taurine, and pantothenic acid in the treated groups were significantly decreased. These results indicated that Cd disrupts energy and lipid metabolism. Meanwhile, Cd causes liver and kidney damage via induction of oxidative stress; serum biochemical indices (e.g., creatinine and urea nitrogen) also support the aforementioned results.
Collapse
Affiliation(s)
- Shuai Chen
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, 194 Xuefu Road, Harbin, Heilongjiang, 150081, China
| | - Meiyan Zhang
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, 194 Xuefu Road, Harbin, Heilongjiang, 150081, China
| | - Lu Bo
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, 194 Xuefu Road, Harbin, Heilongjiang, 150081, China
| | - Siqi Li
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, 194 Xuefu Road, Harbin, Heilongjiang, 150081, China
| | - Liyan Hu
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, 194 Xuefu Road, Harbin, Heilongjiang, 150081, China
| | - Xiujuan Zhao
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, 194 Xuefu Road, Harbin, Heilongjiang, 150081, China.
| | - Changhao Sun
- Department of Nutrition and Food Hygiene, Public Health College, Harbin Medical University, 194 Xuefu Road, Harbin, Heilongjiang, 150081, China.
| |
Collapse
|
18
|
Li XW, Gao HY, Liu J. The role of taurine in improving neural stem cells proliferation and differentiation. Nutr Neurosci 2017; 20:409-415. [PMID: 26906683 DOI: 10.1080/1028415x.2016.1152004] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
OBJECTIVES Taurine is one of the most abundant amino acids in the central nervous system and has important functions in the promotion of brain development. This study aimed to determine the mechanistic role of taurine in improving neuronal proliferation, stem cell proliferation, and neural differentiation. METHODS The data for this review were primarily retrieved from the PubMed database from 1985 to 2015 in English. The search string included the keywords taurine, brain development, neuronal, stem cell, proliferation, differentiation, and others. Relevant publications were identified, retrieved, and reviewed. RESULTS This review introduces the source, function, and mechanisms of taurine in brain development and provides additional detail regarding the mechanistic role of taurine in improving neuronal proliferation, stem cell proliferation, and neural differentiation. Many studies concerning these aspects are discussed. CONCLUSIONS Taurine plays an important role in brain development, including neuronal proliferation, stem cell proliferation, and differentiation, via several mechanisms. Taurine can be directly used in clinical applications to improve brain development because it has no toxic effects on humans.
Collapse
Affiliation(s)
- Xiang-Wen Li
- a Department of Neonatology & NICU of Bayi Children's Hospital , the Army General Hospital of the Chinese PLA , Beijing 100700 , China
- b Graduate School of the Liaoning Medical College , Jinzhou City 121001 , Liaoning province , China
| | - Hong-Yan Gao
- c Department of Scientific Research , the Army General Hospital of the Chinese PLA , Beijing 100700 , China
| | - Jing Liu
- a Department of Neonatology & NICU of Bayi Children's Hospital , the Army General Hospital of the Chinese PLA , Beijing 100700 , China
| |
Collapse
|
19
|
Tarry-Adkins JL, Fernandez-Twinn DS, Chen JH, Hargreaves IP, Neergheen V, Aiken CE, Ozanne SE. Poor maternal nutrition and accelerated postnatal growth induces an accelerated aging phenotype and oxidative stress in skeletal muscle of male rats. Dis Model Mech 2016; 9:1221-1229. [PMID: 27585884 PMCID: PMC5087829 DOI: 10.1242/dmm.026591] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 08/18/2016] [Indexed: 01/09/2023] Open
Abstract
‘Developmental programming’, which occurs as a consequence of suboptimal in utero and early environments, can be associated with metabolic dysfunction in later life, including an increased incidence of cardiovascular disease and type 2 diabetes, and predisposition of older men to sarcopenia. However, the molecular mechanisms underpinning these associations are poorly understood. Many conditions associated with developmental programming are also known to be associated with the aging process. We therefore utilized our well-established rat model of low birth weight and accelerated postnatal catch-up growth (termed ‘recuperated’) in this study to establish the effects of suboptimal maternal nutrition on age-associated factors in skeletal muscle. We demonstrated accelerated telomere shortening (a robust marker of cellular aging) as evidenced by a reduced frequency of long telomeres (48.5-8.6 kb) and an increased frequency of short telomeres (4.2-1.3 kb) in vastus lateralis muscle from aged recuperated offspring compared to controls. This was associated with increased protein expression of the DNA-damage-repair marker 8-oxoguanine-glycosylase (OGG1) in recuperated offspring. Recuperated animals also demonstrated an oxidative stress phenotype, with decreased citrate synthase activity, increased electron-transport-complex activities of complex I, complex II-III and complex IV (all markers of functional mitochondria), and increased xanthine oxidase (XO), p67phox and nuclear-factor kappa-light-chain-enhancer of activated B-cells (NF-κB). Recuperated offspring also demonstrated increased antioxidant defense capacity, with increased protein expression of manganese superoxide dismutase (MnSOD), copper-zinc superoxide dismutase (CuZnSOD), catalase and heme oxygenase-1 (HO1), all of which are known targets of NF-κB and can be upregulated as a consequence of oxidative stress. Recuperated offspring also had a pro-inflammatory phenotype, as evidenced by increased tumor necrosis factor-α (TNFα) and interleukin-1β (IL1β) protein levels. Taken together, we demonstrate, for the first time to our knowledge, an accelerated aging phenotype in skeletal muscle in the context of developmental programming. These findings may pave the way for suitable interventions in at-risk populations. Summary: Muscle of ‘developmentally programmed’ rat offspring demonstrated accelerated aging and oxidative stress, which could explain why some individuals are at greater risk of developing age-associated muscular dysfunction than others.
Collapse
Affiliation(s)
- Jane L Tarry-Adkins
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Hills Road, Cambridge CB2 OQQ, UK
| | - Denise S Fernandez-Twinn
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Hills Road, Cambridge CB2 OQQ, UK
| | - Jian Hua Chen
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Hills Road, Cambridge CB2 OQQ, UK
| | - Iain P Hargreaves
- Neurometabolic Unit, National Hospital, University College London, London WC1N 3BG, UK
| | - Viruna Neergheen
- Neurometabolic Unit, National Hospital, University College London, London WC1N 3BG, UK
| | - Catherine E Aiken
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Hills Road, Cambridge CB2 OQQ, UK
| | - Susan E Ozanne
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Treatment Centre, Addenbrooke's Hospital, Hills Road, Cambridge CB2 OQQ, UK
| |
Collapse
|
20
|
Zou T, Yu B, Yu J, Mao X, Zheng P, He J, Huang Z, Liu Y, Chen D. Moderately decreased maternal dietary energy intake during pregnancy reduces fetal skeletal muscle mitochondrial biogenesis in the pigs. GENES AND NUTRITION 2016; 11:19. [PMID: 27551320 PMCID: PMC4968452 DOI: 10.1186/s12263-016-0535-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 06/15/2016] [Indexed: 11/10/2022]
Abstract
BACKGROUND Mitochondria are of major importance in oocyte and early embryo, playing a key role in maintaining energy homeostasis. Epidemiological findings indicate that maternal undernutrition-induced mitochondrial dysfunction during pregnancy is associated with the development of metabolic disorders in offspring. Here, we investigated the effects of moderately decreased maternal energy intake during pregnancy on skeletal muscle mitochondrial biogenesis in fetal offspring with pig as a model. METHODS Pregnant Meishan sows were allocated to a standard-energy (SE) intake group as recommended by the National Research Council (NRC; 2012) and a low-energy (LE) intake group. Fetal umbilical vein serum and longissimus muscle samples were collected for further analysis on day 90 of pregnancy. RESULTS Sow and fetal weights and the concentrations of serum growth hormone (GH) and glucose were reduced in LE group. Maternal LE diet decreased the messenger RNA (mRNA) expression of genes involved in mitochondrial biogenesis and function such as peroxisome proliferator-activated receptor gamma coactivator 1α (PPARGC1A), nuclear respiratory factor 1 (NRF1), mitochondrial transcription factor A (TFAM), β subunit of mitochondrial H(+)-ATP synthase (ATB5B), sirtuin 1 (Sirt1), and citrate synthase (CS). The protein expression of PPARGC1A and Sirt1, intracellular NAD(+)-to-NADH ratio, and CS activity was reduced in LE group, and accordingly, mitochondrial DNA (mtDNA) content was decreased. Moreover, copper/zinc superoxide dismutase (CuZn-SOD) expression at both mRNA and protein levels and SOD and catalase (CAT) activities were reduced in LE group as well. CONCLUSIONS The observed decrease in muscle mitochondrial biogenesis and antioxidant defense capacity suggests that moderately decreased maternal energy intake during pregnancy impairs mitochondrial function in fetal pigs.
Collapse
Affiliation(s)
- Tiande Zou
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Cheng du, China
| | - Bing Yu
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Cheng du, China
| | - Jie Yu
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Cheng du, China
| | - Xiangbing Mao
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Cheng du, China
| | - Ping Zheng
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Cheng du, China
| | - Jun He
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Cheng du, China
| | - Zhiqing Huang
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Cheng du, China
| | - Yue Liu
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Cheng du, China
| | - Daiwen Chen
- Key Laboratory for Animal Disease-Resistant Nutrition of the Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Cheng du, China
| |
Collapse
|
21
|
Taurine Enhances Proliferation and Promotes Neuronal Specification of Murine and Human Neural Stem/Progenitor Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2015; 803:457-72. [DOI: 10.1007/978-3-319-15126-7_36] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
22
|
Peñagaricano F, Wang X, Rosa GJ, Radunz AE, Khatib H. Maternal nutrition induces gene expression changes in fetal muscle and adipose tissues in sheep. BMC Genomics 2014; 15:1034. [PMID: 25429728 PMCID: PMC4301459 DOI: 10.1186/1471-2164-15-1034] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 11/19/2014] [Indexed: 01/15/2023] Open
Abstract
Background Maternal nutrition during different stages of pregnancy can induce significant changes in the structure, physiology, and metabolism of the offspring. These changes could have important implications on food animal production especially if these perturbations impact muscle and adipose tissue development. Here, we evaluated the impact of different maternal isoenergetic diets, alfalfa haylage (HY; fiber), corn (CN; starch), and dried corn distillers grains (DG; fiber plus protein plus fat), on the transcriptome of fetal muscle and adipose tissues in sheep. Results Prepartum diets were associated with notable gene expression changes in fetal tissues. In longissimus dorsi muscle, a total of 224 and 823 genes showed differential expression (FDR ≤0.05) in fetuses derived from DG vs. CN and HY vs. CN maternal diets, respectively. Several of these significant genes affected myogenesis and muscle differentiation. In subcutaneous and perirenal adipose tissues, 745 and 208 genes were differentially expressed (FDR ≤0.05), respectively, between CN and DG diets. Many of these genes are involved in adipogenesis, lipogenesis, and adipose tissue development. Pathway analysis revealed that several GO terms and KEGG pathways were enriched (FDR ≤0.05) with differentially expressed genes associated with tissue and organ development, chromatin biology, and different metabolic processes. Conclusions These findings provide evidence that maternal nutrition during pregnancy can alter the programming of fetal muscle and fat tissues in sheep. The ramifications of the observed gene expression changes, in terms of postnatal growth, body composition, and meat quality of the offspring, warrant future investigation. Electronic supplementary material The online version of this article (doi:10.1186/1471-2164-15-1034) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Francisco Peñagaricano
- Department of Animal Sciences, University of Wisconsin-Madison, 1675 Observatory Drive, Madison, WI 53706, USA.
| | | | | | | | | |
Collapse
|
23
|
Liu X, Pan S, Li X, Sun Q, Yang X, Zhao R. Maternal low-protein diet affects myostatin signaling and protein synthesis in skeletal muscle of offspring piglets at weaning stage. Eur J Nutr 2014; 54:971-9. [PMID: 25266448 DOI: 10.1007/s00394-014-0773-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 09/19/2014] [Indexed: 11/26/2022]
Abstract
PURPOSE We tested the hypothesis that maternal low-protein (LP) diet during gestation and lactation can program myostatin (MSTN) signaling and protein synthesis in skeletal muscle of offspring at weaning stage (35 days). METHODS Fourteen Meishan sows were fed either LP or standard-protein diets throughout gestation and lactation, male offspring piglets were killed at weaning stage and longissimus dorsi (LD) muscles were taken. The cross-sectional areas (CSA) of LD muscles were measured by hematoxylin and eosin staining. The levels of free amino acids in plasma were measured by amino acid auto-analyzer. Proteins and mRNA were determined by Western blot and RT-qPCR, respectively. RESULTS Body weight, LD muscle weight and the myofiber CSA were significantly decreased (P < 0.05) in LP piglets; meanwhile, the concentration of branched-chain amino acids was also significantly decreased (P < 0.001). MSTN protein content tended to be higher (P = 0.098) in LP piglets, while the expression of MSTN receptors, activin type II receptor-beta and transforming growth factor type-beta type I receptor kinase, was significantly up-regulated (P < 0.05). Furthermore, p38 mitogen-activated protein kinase, the downstream signaling factor of MSTN, was also enhanced significantly (P < 0.05). In addition, key factors of translation initiation, phosphorylated eukaryotic initiation factor 4E and the 70 kDa ribosomal protein S6 kinase, were significantly decreased (P < 0.05) in LP piglets. CONCLUSIONS Our results suggest that maternal LP diet during gestation and lactation affects MSTN signaling and protein synthesis in skeletal muscle of offspring at weaning stage.
Collapse
Affiliation(s)
- Xiujuan Liu
- Key Laboratory of Animal Physiology and Biochemistry, Ministry of Agriculture, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | | | | | | | | | | |
Collapse
|
24
|
Ramos-Mandujano G, Hernández-Benítez R, Pasantes-Morales H. Multiple mechanisms mediate the taurine-induced proliferation of neural stem/progenitor cells from the subventricular zone of the adult mouse. Stem Cell Res 2014; 12:690-702. [PMID: 24681519 DOI: 10.1016/j.scr.2014.02.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Revised: 01/15/2014] [Accepted: 02/27/2014] [Indexed: 12/12/2022] Open
Abstract
Taurine was previously reported to increase the proliferation of neural precursor cells (NPCs) from subventricular zone of the mouse brain. The results of a study that aimed to understand the mechanisms of this effect are presented here. Because taurine was not found in NPC nuclei, direct interactions with nuclear elements seem unlikely. A gene expression profile analysis indicated that genes that are regulated by taurine have roles in i) proliferation, including the Shh and Wnt pathways; ii) cellular adhesion; iii) cell survival; and iv) mitochondrial functioning. Cell cycle analysis of propidium iodide and CFSE-labeled cells using flow cytometry revealed an increase in the number of cells in the S-phase and a decrease in those in the G0/G1 phase in taurine-treated cultures. No changes in the length of the cell cycle were observed. Quantification of the viable, apoptotic, and necrotic cells in cultures using flow cytometry and calcein-AM, annexin-V, and propidium iodide staining showed reductions in the number of apoptotic and necrotic cells (18% to 11% and 13% to 10%, respectively) and increases in the number of viable cells (61% to 69%) in the taurine-treated cultures. Examination of the relative mitochondrial potential values by flow cytometry and rhodamine123 or JC-1 staining showed a 44% increase in the number of cells with higher mitochondrial potential and a 38% increase in the mitochondrial membrane potential in taurine cultures compared with those of controls. Taken together, the results suggest that taurine provides more favorable conditions for cell proliferation by improving mitochondrial functioning.
Collapse
Affiliation(s)
- Gerardo Ramos-Mandujano
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Reyna Hernández-Benítez
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| | - Herminia Pasantes-Morales
- División de Neurociencias, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico.
| |
Collapse
|
25
|
Metabonomic analysis of quercetin against the toxicity of chronic exposure to low-level dichlorvos in rats via ultra-performance liquid chromatography–mass spectrometry. Toxicol Lett 2014; 225:230-9. [DOI: 10.1016/j.toxlet.2013.12.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 12/14/2013] [Accepted: 12/17/2013] [Indexed: 01/16/2023]
|
26
|
Hou L, Hellgren LI, Kongsted AH, Vaag A, Nielsen MO. Pre-natal undernutrition and post-natal overnutrition are associated with permanent changes in hepatic metabolism markers and fatty acid composition in sheep. Acta Physiol (Oxf) 2014; 210:317-29. [PMID: 24313944 DOI: 10.1111/apha.12211] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 10/29/2013] [Accepted: 11/30/2013] [Indexed: 12/26/2022]
Abstract
AIM Determine the impacts of pre- and early-post-natal nutrition on selected markers of hepatic glucose and fat metabolism. METHODS Twin-bearing ewes were fed 100% (NORM) or 50% (LOW) of protein and energy requirements during the last 6-weeks of gestation. Twin-lambs received either a high-carbohydrate high-fat (HCHF) or conventional (CONV) diet from 3 days to 6 months of age (around puberty), whereafter lambs from the four subgroups were slaughtered (16 males/3 females). Remaining lambs (19 females) were fed a moderate diet and slaughtered at 2 years of age (young adults). RESULTS Pre-natal LOW nutrition was associated with increased hepatic triglyceride, ceramide and free fatty acid content in adulthood (not observed in lambs), which was accompanied by up-regulated early-stage insulin signalling as reflected by increased INSRβ and PI3K-p110 protein expression. The HCHF diet increased hepatic triglyceride content in lambs, associated with down-regulated expressions of energy-metabolism-related genes (GLUT1, PPARα, SREBP1c, PEPCK). These post-natal effects were not observed in adult HCHF sheep, after they had received a moderate (body-fat correcting) diet for 1.5 years. Interestingly, pre-natal LOW nutrition induced permanent alterations in hepatic phospholipids' fatty acid composition. Thus, the amount of linoleic acid (C18 : 2 ∆(9,12)) was significantly increased and composition of rumen-derived fatty acids were altered, indicating changed composition of rumenal microbiota. CONCLUSION Hepatic insulin signalling and linoleic and microbial-derived fatty acid content in phospholipids are targets of foetal programming induced by late-gestation undernutrition. Future studies are required to explain their cause-effect associations with increased risks of developing hepatic steatosis and insulin insensitivity in adulthood.
Collapse
Affiliation(s)
- L. Hou
- Faculty of Health and Medical Sciences; Department of Veterinary Clinical and Animal Sciences; University of Copenhagen; Frederiksberg Denmark
- Centre for Fetal Programming; Copenhagen Denmark
| | - L. I. Hellgren
- Centre for Fetal Programming; Copenhagen Denmark
- Center for Biological Sequence Analysis; Technical University of Denmark; Lyngby Denmark
| | - A. H. Kongsted
- Faculty of Health and Medical Sciences; Department of Veterinary Clinical and Animal Sciences; University of Copenhagen; Frederiksberg Denmark
- Centre for Fetal Programming; Copenhagen Denmark
| | - A. Vaag
- Centre for Fetal Programming; Copenhagen Denmark
- Department of Endocrinology; Rigshospitalet; Copenhagen Denmark
| | - M. O. Nielsen
- Faculty of Health and Medical Sciences; Department of Veterinary Clinical and Animal Sciences; University of Copenhagen; Frederiksberg Denmark
- Centre for Fetal Programming; Copenhagen Denmark
| |
Collapse
|
27
|
Oster M, Murani E, Metges CC, Ponsuksili S, Wimmers K. High- and low-protein gestation diets do not provoke common transcriptional responses representing universal target-pathways in muscle and liver of porcine progeny. Acta Physiol (Oxf) 2014; 210:202-14. [PMID: 24188291 DOI: 10.1111/apha.12192] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 07/02/2013] [Accepted: 10/30/2013] [Indexed: 12/27/2022]
Abstract
AIM Maternal diets introduce transcriptional changes in the offspring, highlighting the concept of genetic and physiological plasticity. Our previous analyses investigated stage-dependent transcriptional responses to either maternal high or low protein/carbohydrate ratios in either muscle or liver. Foetal programming is proposed to be mediated by a small number of gatekeeper processes, such as cytoskeleton remodelling and cell-cycle regulation. Here, we conducted an overall analysis of a three-dimensional data set aiming to elucidate, whether there are universally targeted pathways of adaptive transcriptional response to different protein/carbohydrate ratios. METHODS Microarray analyses were performed on liver and skeletal muscle tissue sampled at 94 days post-conception and 1, 28 and 188 days post-natum from offspring (n = 253) of German Landrace gilts that were fed isoenergetic diets containing low, high or adequate protein. RESULTS Cluster analyses revealed a hierarchical influence of tissue, ontogenetic stage and diet on transcript levels. Considering results cumulatively over stages, liver showed only marginal transcriptional differences between the dietary groups, whereas considerable differences appeared in muscle. Considering results cumulatively over tissues, nutrition-responsive transcriptions were observed along ontogenesis. Pathway analyses revealed transcript differences in genes related to tissue remodelling, cell-cycle regulation and mitochondrial function. CONCLUSION The factors tissue, stage and diet impact gene expression in a hierarchical order. Porcine liver appeared to be a tissue that was more resilient to nutritional modulation compared with skeletal muscle tissue. Differential modulation between tissues and dietary groups reveal that there are no universal target-pathways of adaptive transcriptional response to different protein diets.
Collapse
Affiliation(s)
- M. Oster
- Institute for Genome Biology; Leibniz Institute for Farm Animal Biology; Dummerstorf Germany
| | - E. Murani
- Institute for Genome Biology; Leibniz Institute for Farm Animal Biology; Dummerstorf Germany
| | - C. C. Metges
- Institute for Nutritional Physiology; Leibniz Institute for Farm Animal Biology; Dummerstorf Germany
| | - S. Ponsuksili
- Research Group Functional Genomics; Leibniz Institute for Farm Animal Biology; Dummerstorf Germany
| | - K. Wimmers
- Institute for Genome Biology; Leibniz Institute for Farm Animal Biology; Dummerstorf Germany
| |
Collapse
|
28
|
Abstract
Development of metabolic syndrome is attributed to genes, dietary intake, physical activity and environmental factors. Fetal programming due to maternal nutrition is also an important factor especially in developing countries where intrauterine growth retardation followed by excess nutrition postnatally is causing mismatch predisposing individuals to development of metabolic syndrome and its components. Several epidemiological and animal studies have provided evidence for the link between intrauterine growth retardation and adult metabolic diseases. Deficiency of macronutrients, protein and carbohydrates, during pregnancy and gestation results in lower infant birth weight, a surrogate marker of fetal growth and subsequently insulin resistance, glucose intolerance, hypertension and adiposity in adulthood. The role of micronutrients is less extensively studied but however gaining attention with several recent studies focusing on this aspect. Several mechanisms have been proposed to explain the developmental origin of adult diseases important among them being alteration of hypothalamic pituitary axis, epigenetic regulation of gene expression and oxidative stress. All of these mechanisms may be acting at different time during gestation and contributing to development of metabolic syndrome in adulthood.
Collapse
Affiliation(s)
- Ramakrishnan Lakshmy
- Department of Cardiac Biochemistry, All India Institute of Medical Sciences, New Delhi, 110049, India,
| |
Collapse
|
29
|
Gestational protein restriction induces alterations in placental morphology and mitochondrial function in rats during late pregnancy. J Mol Histol 2013; 44:629-37. [DOI: 10.1007/s10735-013-9522-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 06/26/2013] [Indexed: 01/07/2023]
|
30
|
Matzkin LM, Johnson S, Paight C, Markow TA. Preadult parental diet affects offspring development and metabolism in Drosophila melanogaster. PLoS One 2013; 8:e59530. [PMID: 23555695 PMCID: PMC3608729 DOI: 10.1371/journal.pone.0059530] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2013] [Accepted: 02/15/2013] [Indexed: 02/07/2023] Open
Abstract
When Drosophila melanogaster larvae are reared on isocaloric diets differing in their amounts of protein relative to sugar, emerging adults exhibit significantly different development times and metabolic pools of protein, glycogen and trigylcerides. In the current study, we show that the influence of larval diet experienced during just one generation extends into the next generation, even when that subsequent generation had been shifted to a standard diet during development. Offspring of flies that were reared on high protein relative to sugar underwent metamorphosis significantly faster, had higher reproductive outputs, and different metabolic pool contents compared to the offspring of adults from low protein relative to sugar diets. In addition, isofemale lines differed in the degree to which parental effects were observed, suggesting a genetic component to the observed transgenerational influences.
Collapse
Affiliation(s)
- Luciano M. Matzkin
- Department of Biological Sciences, University of Alabama in Huntsville, Huntsville, Alabama, United States of America
| | - Sarah Johnson
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Christopher Paight
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California San Diego, La Jolla, California, United States of America
| | - Therese A. Markow
- Section of Cell and Developmental Biology, Division of Biological Sciences, University of California San Diego, La Jolla, California, United States of America
- Laboratorio Nacional de Genomica de la Biodiversidad, Centro de Investigaciones y Estudios Avancados, Irapuato, Guanajuato, Mexico
- * E-mail:
| |
Collapse
|
31
|
Roysommuti S, Wyss JM. Perinatal taurine exposure affects adult arterial pressure control. Amino Acids 2012; 46:57-72. [PMID: 23070226 DOI: 10.1007/s00726-012-1417-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2012] [Accepted: 10/04/2012] [Indexed: 12/13/2022]
Abstract
Taurine is an abundant, free amino acid found in mammalian cells that contributes to many physiologic functions from that of a simple cell osmolyte to a programmer of adult health and disease. Taurine's contribution extends from conception throughout life, but its most critical exposure period is during perinatal life. In adults, taurine supplementation prevents or alleviates cardiovascular disease and related complications. In contrast, low taurine consumption coincides with increased risk of cardiovascular disease, obesity and type II diabetes. This review focuses on the effects that altered perinatal taurine exposure has on long-term mechanisms that control adult arterial blood pressure and could thereby contribute to arterial hypertension through its ability to program these cardiovascular regulatory mechanisms very early in life. The modifications of these mechanisms can last a lifetime and transfer to the next generation, suggesting that epigenetic mechanisms underlie the changes. The ability of perinatal taurine exposure to influence arterial pressure control mechanisms and hypertension in adult life appears to involve the regulation of growth and development, the central and autonomic nervous system, the renin-angiotensin system, glucose-insulin interaction and changes to heart, blood vessels and kidney function.
Collapse
Affiliation(s)
- Sanya Roysommuti
- Department of Physiology, Faculty of Medicine, Khon Kaen University, Khon Kaen, 40002, Thailand,
| | | |
Collapse
|
32
|
Maloney CA, Hay SM, Reid MD, Duncan G, Nicol F, Sinclair KD, Rees WD. A methyl-deficient diet fed to rats during the pre- and peri-conception periods of development modifies the hepatic proteome in the adult offspring. GENES AND NUTRITION 2012; 8:181-90. [PMID: 22907820 DOI: 10.1007/s12263-012-0314-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Accepted: 08/02/2012] [Indexed: 01/03/2023]
Abstract
A methyl-deficient diet (MD) lacking folic acid and the associated methyl donors choline and methionine, fed to the laboratory rat during the periods of oocyte and embryo development, has been shown to programme glucose metabolism in the offspring. The hepatic proteome of the male offspring of female rats fed MD diets for 3 weeks prior to mating and for the first 5 days of gestation has been examined by 2-dimensional gel electrophoresis. Three groups of differentially abundant proteins associated with energy metabolism, amino acid metabolism and antioxidant defence were identified in the soluble proteins extracted from the liver from the MD offspring at both 6 and 12 months of age. Altered mitochondrial activity in other programming models leads to a similar pattern of differential protein abundance. Two of the differentially abundant proteins were identified as GAPDH and PGK-1 by mass spectrometry. Western blotting showed that there were multiple isoforms of both proteins with similar molecular weights but different isoelectric points. The differentially abundant spots reduced in the MD offspring corresponded to minor isoforms of GAPDH and PGK-1. The levels of PPAR-alpha, SREBP and glucocorticoid receptor mRNAs associated with other models of prenatal programming were unchanged in the MD offspring. The data suggest that a diet deficient in folic acid and associated methyl donors fed during the peri-conception and early preimplantation periods of mammalian development affects mitochondrial function in the offspring and that the posttranslational modification of proteins may be important.
Collapse
Affiliation(s)
- Christopher A Maloney
- School of Medical Sciences, Faculty of Medicine, The University of New South Wales, Sydney, NSW, 2052, Australia
| | | | | | | | | | | | | |
Collapse
|
33
|
Liu J, Yu B, Mao X, He J, Yu J, Zheng P, Huang Z, Chen D. Effects of intrauterine growth retardation and maternal folic acid supplementation on hepatic mitochondrial function and gene expression in piglets. Arch Anim Nutr 2012; 66:357-71. [PMID: 22889112 DOI: 10.1080/1745039x.2012.710084] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Piglets with intrauterine growth retardation (IUGR) or with normal birth weight (NBW) were selected to evaluate the effects of maternal folic acid supplementation on hepatic mitochondrial function and expression levels of genes involved in mitochondrial DNA (mtDNA) biogenesis and mitochondrial function. During gestation, primiparous Yorkshire sows were fed a Control diet (folic acid 1.3 mg/kg) or a folic acid-supplemented diet (folic acid 30 mg/kg) with 16 replicates per diet. During the 28-d lactation period, sows were fed a common diet. Compared with NBW piglets, hepatic ATP concentrations and mtDNA contents were decreased in IUGR piglets. Furthermore, IUGR piglets exhibited lower membrane potential and decreased oxygen consumption in liver mitochondria, but these parameters were not affected by maternal folic acid supplementation. Intrauterine growth retardation decreased mRNA expression abundance of peroxisomal proliferator-activated receptor-γ coactivator-1α, mitochondrial transcription factor A, uncoupling protein 3, and cytochrome c oxidase subunit I and IV. Impaired antioxidant capacity characterised by increased malondialdehyde content and decreased manganese-superoxide dismutase activity was also observed in IUGR pigs. In IUGR piglets, however, nearly all of these parameters were normalised to the level of NBW piglets when the maternal diet was supplemented with folic acid during pregnancy. Hence, maternal folic acid supplementation was proved to be an effective way to reverse the changes in gene expressions in IUGR pigs, which provided a possible nutritional strategy to improve growth development of IUGR individuals.
Collapse
Affiliation(s)
- Jingbo Liu
- Institute of Animal Nutrition, Sichuan Agricultural University , Ya'an, Sichuan Province, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Liu J, Chen D, Yao Y, Yu B, Mao X, He J, Huang Z, Zheng P. Intrauterine growth retardation increases the susceptibility of pigs to high-fat diet-induced mitochondrial dysfunction in skeletal muscle. PLoS One 2012; 7:e34835. [PMID: 22523560 PMCID: PMC3327708 DOI: 10.1371/journal.pone.0034835] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Accepted: 03/06/2012] [Indexed: 01/07/2023] Open
Abstract
It has been recognized that there is a relationship between prenatal growth restriction and the development of metabolic-related diseases in later life, a process involved in mitochondrial dysfunction. In addition, intrauterine growth retardation (IUGR) increases the susceptibility of offspring to high-fat (HF) diet-induced metabolic syndrome. Recent findings suggested that HF feeding decreased mitochondrial oxidative capacity and impaired mitochondrial function in skeletal muscle. Therefore, we hypothesized that the long-term consequences of IUGR on mitochondrial biogenesis and function make the offspring more susceptible to HF diet-induced mitochondrial dysfunction. Normal birth weight (NBW), and IUGR pigs were allotted to control or HF diet in a completely randomized design, individually. After 4 weeks of feeding, growth performance and molecular pathways related to mitochondrial function were determined. The results showed that IUGR decreased growth performance and plasma insulin concentrations. In offspring fed a HF diet, IUGR was associated with enhanced plasma leptin levels, increased concentrations of triglyceride and malondialdehyde (MDA), and reduced glycogen and ATP contents in skeletal muscle. High fat diet-fed IUGR offspring exhibited decreased activities of lactate dehydrogenase (LDH) and glucose-6-phosphate dehydrogenase (G6PD). These alterations in metabolic traits of IUGR pigs were accompanied by impaired mitochondrial respiration function, reduced mitochondrial DNA (mtDNA) contents, and down-regulated mRNA expression levels of genes responsible for mitochondrial biogenesis and function. In conclusion, our results suggest that IUGR make the offspring more susceptible to HF diet-induced mitochondrial dysfunction.
Collapse
Affiliation(s)
- Jingbo Liu
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, Sichuan, People's Republic of China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, People's Republic of China
| | - Daiwen Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, Sichuan, People's Republic of China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, People's Republic of China
| | - Ying Yao
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, Sichuan, People's Republic of China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, People's Republic of China
| | - Bing Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, Sichuan, People's Republic of China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, People's Republic of China
| | - Xiangbing Mao
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, Sichuan, People's Republic of China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, People's Republic of China
| | - Jun He
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, Sichuan, People's Republic of China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, People's Republic of China
| | - Zhiqing Huang
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, Sichuan, People's Republic of China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, People's Republic of China
| | - Ping Zheng
- Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, Sichuan, People's Republic of China
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, People's Republic of China
| |
Collapse
|
35
|
Oster M, Murani E, Metges CC, Ponsuksili S, Wimmers K. A gestational high protein diet affects the abundance of muscle transcripts related to cell cycle regulation throughout development in porcine progeny. PLoS One 2012; 7:e34519. [PMID: 22496824 PMCID: PMC3322122 DOI: 10.1371/journal.pone.0034519] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Accepted: 03/05/2012] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND In various animal models pregnancy diets have been shown to affect offspring phenotype. Indeed, the underlying programming of development is associated with modulations in birth weight, body composition, and continual diet-dependent modifications of offspring metabolism until adulthood, producing the hypothesis that the offspring's transcriptome is permanently altered depending on maternal diet. METHODOLOGY/PRINCIPAL FINDINGS To assess alterations of the offspring's transcriptome due to gestational protein supply, German Landrace sows were fed isoenergetic diets containing protein levels of either 30% (high protein--HP) or 12% (adequate protein--AP) throughout their pregnancy. Offspring muscle tissue (M. longissimus dorsi) was collected at 94 days post conception (dpc), and 1, 28, and 188 days post natum (dpn) for use with Affymetrix GeneChip Porcine Genome Arrays and subsequent statistical and Ingenuity pathway analyses. Numerous transcripts were found to have altered abundance at 94 dpc and 1 dpn; at 28 dpn no transcripts were altered, and at 188 dpn only a few transcripts showed a different abundance between diet groups. However, when assessing transcriptional changes across developmental time points, marked differences were obvious among the dietary groups. Depending on the gestational dietary exposure, short- and long-term effects were observed for mRNA expression of genes related to cell cycle regulation, energy metabolism, growth factor signaling pathways, and nucleic acid metabolism. In particular, the abundance of transcripts related to cell cycle remained divergent among the groups during development. CONCLUSION Expression analysis indicates that maternal protein supply induced programming of the offspring's genome; early postnatal compensation of the slight growth retardation obvious at birth in HP piglets resulted, as did a permanently different developmental alteration and responsiveness to the common environment of the transcriptome. The transcriptome modulations are interpreted as the molecular equivalent of developmental plasticity of the offspring that necessitates adaptation and maintenance of the organismal phenotype.
Collapse
Affiliation(s)
- Michael Oster
- Research Unit Molecular Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Eduard Murani
- Research Unit Molecular Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Cornelia C. Metges
- Research Unit Nutritional Physiology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Siriluck Ponsuksili
- Research Group Functional Genomics, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Klaus Wimmers
- Research Unit Molecular Biology, Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| |
Collapse
|
36
|
Mitochondrial Respiration Is Decreased in Rat Kidney Following Fetal Exposure to a MaternalLow-ProteinDiet. J Nutr Metab 2012; 2012:989037. [PMID: 22536494 PMCID: PMC3321454 DOI: 10.1155/2012/989037] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 01/04/2012] [Indexed: 01/20/2023] Open
Abstract
Maternal protein restriction in rat pregnancy is associated with impaired renal development and age-related loss of renal function in the resulting offspring. Pregnant rats were fed either control or low-protein (LP) diets, and kidneys from their male offspring were collected at 4, 13, or 16 weeks of age. Mitochondrial state 3 and state 4 respiratory rates were decreased by a third in the LP exposed adults. The reduction in mitochondrial function was not explained by complex IV deficiency or altered expression of the complex I subunits that are typically associated with mitochondrial dysfunction. Similarly, there was no evidence that LP-exposure resulted in greater oxidative damage to the kidney, differential expression of ATP synthetase β-subunit, and ATP-ADP translocase 1. mRNA expression of uncoupling protein 2 was increased in adult rats exposed to LP in utero, but there was no evidence of differential expression at the protein level. Exposure to maternal undernutrition is associated with a decrease in mitochondrial respiration in kidneys of adult rats. In the absence of gross disturbances in respiratory chain protein expression, programming of coupling efficiency may explain the long-term impact of the maternal diet.
Collapse
|
37
|
Oster M, Murani E, Metges CC, Ponsuksili S, Wimmers K. A low protein diet during pregnancy provokes a lasting shift of hepatic expression of genes related to cell cycle throughout ontogenesis in a porcine model. BMC Genomics 2012; 13:93. [PMID: 22424151 PMCID: PMC3342123 DOI: 10.1186/1471-2164-13-93] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2011] [Accepted: 03/16/2012] [Indexed: 12/23/2022] Open
Abstract
Background In rodent models and in humans the impact of gestational diets on the offspring's phenotype was shown experimentally and epidemiologically. Adverse environmental conditions during fetal development provoke an intrauterine adaptive response termed 'fetal programming', which may lead to both persistently biased responsiveness to extrinsic factors and permanent consequences for the organismal phenotype. This leads to the hypothesis that the offspring's transcriptome exhibits short-term and long-term changes, depending on the maternal diet. In order to contribute to a comprehensive inventory of genes and functional networks that are targets of nutritional programming initiated during fetal life, we applied whole-genome microarrays for expression profiling in a longitudinal experimental design covering prenatal, perinatal, juvenile, and adult ontogenetic stages in a porcine model. Pregnant sows were fed either a gestational low protein diet (LP, 6% CP) or an adequate protein diet (AP, 12% CP). All offspring was nursed by foster sows receiving standard diets. After weaning, all offspring was fed standard diets ad libitum. Results Analyses of the hepatic gene expression of the offspring at prenatal (94 dies post conceptionem, dpc) and postnatal stages (1, 28, 188 dies post natum, dpn) included comparisons between dietary groups within stages as well as comparisons between ontogenetic stages within diets to separate diet-specific transcriptional changes and maturation processes. We observed differential expression of genes related to lipid metabolism (e.g. Fatty acid metabolism, Biosynthesis of steroids, Synthesis and degradation of ketone bodies, FA elongation in mitochondria, Bile acid synthesis) and cell cycle regulation (e.g. Mitotic roles of PLK, G1/S checkpoint regulation, G2/M DNA damage checkpoint regulation). Notably, at stage 1 dpn no regulation of a distinct pathway was found in LP offspring. Conclusions The transcriptomic modulations point to persistent functional demand on the liver towards cell proliferation in the LP group but not in the AP group at identical nutritional conditions during postnatal life due to divergent 'programming' of the genome. Together with the observation that the offspring of both groups did not differ in body weight but in body composition and fat content, the data indicate that the activity of various genes led to diverse partitioning of nutrients among peripheral and visceral organs and tissues.
Collapse
Affiliation(s)
- Michael Oster
- Research Unit Molecular Biology, Leibniz Institute for Farm Animal Biology, Dummerstorf, Germany
| | | | | | | | | |
Collapse
|
38
|
Maternal obesity during gestation impairs fatty acid oxidation and mitochondrial SIRT3 expression in rat offspring at weaning. PLoS One 2011; 6:e24068. [PMID: 21901160 PMCID: PMC3162035 DOI: 10.1371/journal.pone.0024068] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2011] [Accepted: 07/29/2011] [Indexed: 01/11/2023] Open
Abstract
In utero exposure to maternal obesity increases the offspring's risk of obesity in later life. We have also previously reported that offspring of obese rat dams develop hepatic steatosis, mild hyperinsulinemia, and a lipogenic gene signature in the liver at postnatal day (PND)21. In the current study, we examined systemic and hepatic adaptations in male Sprague-Dawley offspring from lean and obese dams at PND21. Indirect calorimetry revealed decreases in energy expenditure (p<0.001) and increases in RER values (p<0.001), which were further exacerbated by high fat diet (45% kcals from fat) consumption indicating an impaired ability to utilize fatty acids in offspring of obese dams as analyzed by PRCF. Mitochondrial function is known to be associated with fatty acid oxidation (FAO) in the liver. Several markers of hepatic mitochondrial function were reduced in offspring of obese dams. These included SIRT3 mRNA (p = 0.012) and mitochondrial protein content (p = 0.002), electron transport chain complexes (II, III, and ATPase), and fasting PGC-1α mRNA expression (p<0.001). Moreover, hepatic LCAD, a SIRT3 target, was not only reduced 2-fold (p<0.001) but was also hyperacetylated in offspring of obese dams (p<0.005) suggesting decreased hepatic FAO. In conclusion, exposure to maternal obesity contributes to early perturbations in whole body and liver energy metabolism. Mitochondrial dysfunction may be an underlying event that reduces hepatic fatty acid oxidation and precedes the development of detrimental obesity associated co-morbidities such as insulin resistance and NAFLD.
Collapse
|