1
|
Liu X, Liu X, Jin M, Huang N, Song Z, Li N, Huang T. Association between birth weight/joint exposure to ambient air pollutants and type 2 diabetes: a cohort study in the UK Biobank. INTERNATIONAL JOURNAL OF ENVIRONMENTAL HEALTH RESEARCH 2024; 34:2888-2898. [PMID: 37936397 DOI: 10.1080/09603123.2023.2278634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 10/30/2023] [Indexed: 11/09/2023]
Abstract
Early life events and environmental factors are associated with type 2 diabetes (T2D) development. We assessed the combined effect of birth weight andambient air pollutants, and effect of their interaction on T2D risk. Totally, 6,474 T2D incidents were recorded over an 8.7-year follow-up period. The adjusted hazard ratios (aHRs) with 95% confidence intervals (CIs) were 1.31 (1.26, 1.36) for each kilogram decrease in birth weight, and 1.08 (1.05, 1.11) for each standard deviation increase in air pollution score (APS). Birth weight<3000 g amplified the T2D risk associated with high APS. A combination of the lowest birth weight (<2500 g) and the highest quintile of APS led to over two-fold increase in T2D risk (aHR: 2.17; 95% CI: 1.79-2.64). There was a significant additive interaction between them. In conclusion, ambient air pollutants increase the risk for T2D, particularly in populations with low birth weight.
Collapse
Affiliation(s)
- Xiaojing Liu
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China
- Institute of Reproductive and Child Health, Peking University/Key Laboratory of Reproductive Health, Beijing, China
| | - Xiaowen Liu
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China
- Institute of Reproductive and Child Health, Peking University/Key Laboratory of Reproductive Health, Beijing, China
| | - Ming Jin
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China
- Institute of Reproductive and Child Health, Peking University/Key Laboratory of Reproductive Health, Beijing, China
| | - Ninghao Huang
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Zimin Song
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China
| | - Nan Li
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China
- Institute of Reproductive and Child Health, Peking University/Key Laboratory of Reproductive Health, Beijing, China
| | - Tao Huang
- Department of Epidemiology & Biostatistics, School of Public Health, Peking University, Beijing, China
- Ministry of Education, Key Laboratory of Molecular Cardiovascular Sciences (Peking University), Beijing, China
- Center for Intelligent Public Health, Institute for Artificial Intelligence, Peking University, Beijing, China
| |
Collapse
|
2
|
Fitness Changes in Adolescent Girls Following In-School Combined Aerobic and Resistance Exercise: Interaction With Birthweight. Pediatr Exerc Sci 2022; 34:76-83. [PMID: 34894627 DOI: 10.1123/pes.2021-0034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 09/21/2021] [Accepted: 10/12/2021] [Indexed: 11/18/2022]
Abstract
PURPOSE To assess the efficacy of a supervised in-school combined resistance and aerobic training program in adolescent girls and investigate whether responses differ according to birthweight. METHODS Participants (girls aged 13-17 y) were randomized either to an intervention replacing physical education (PE) classes with 2 × 60-minute training sessions per week (n = 58) or to a control group that continued to attend 2 × 60 minutes per week of curriculum PE (n = 41). We measured muscular fitness (handgrip, standing long jump, and sit-ups), cardiorespiratory fitness (20-m shuttle run), skinfolds, and lean body mass preintervention and postintervention and determined effect size (Hedge's g) differences between changes in these measures. We also compared changes within lower (<3000 g) and normal birthweight intervention and PE control subgroups. RESULTS The intervention group showed greater improvements in all the fitness measures and lean body mass (g = 0.22-0.48) and lower skinfold increases (g = 0.41) than PE controls. Within the intervention group, improvements in all fitness measures were larger in lower birthweight (g = 0.53-0.94) than in normal birthweight girls (g = 0.02-0.39). CONCLUSION Replacing curriculum PE with supervised training improved muscular and cardiorespiratory fitness and body composition outcomes in adolescent females. Our findings suggest an enhanced adaptive response to training in participants with lower birthweight which warrants further investigation.
Collapse
|
3
|
Buckels EJ, Bloomfield FH, Oliver MH, Spiroski AM, Harding JE, Jaquiery AL. Sexually dimorphic changes in the endocrine pancreas and skeletal muscle in young adulthood following intra-amniotic IGF-I treatment of growth-restricted fetal sheep. Am J Physiol Endocrinol Metab 2021; 321:E530-E542. [PMID: 34459219 DOI: 10.1152/ajpendo.00111.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Fetal growth restriction (FGR) is associated with decreased insulin secretory capacity and decreased insulin sensitivity in muscle in adulthood. We investigated whether intra-amniotic IGF-I treatment in late gestation mitigated the adverse effects of FGR on the endocrine pancreas and skeletal muscle at 18 mo of age. Singleton-bearing ewes underwent uterine artery embolization between 103 and 107 days of gestational age, followed by 5 once-weekly intra-amniotic injections of 360-µg IGF-I (FGRI) or saline (FGRS) and were compared with an unmanipulated control group (CON). We measured offspring pancreatic endocrine cell mass and pancreatic and skeletal muscle mRNA expression at 18 mo of age (n = 7-9/sex/group). Total α-cell mass was increased ∼225% in FGRI males versus CON and FGRS males, whereas β-cell mass was not different between groups of either sex. Pancreatic mitochondria-related mRNA expression was increased in FGRS females versus CON (NRF1, MTATP6, UCP2), and FGRS males versus CON (TFAM, NRF1, UCP2) but was largely unchanged in FGRI males versus CON. In skeletal muscle, mitochondria-related mRNA expression was decreased in FGRS females versus CON (PPARGC1A, TFAM, NRF1, UCP2, MTATP6), FGRS males versus CON (NRF1 and UCP2), and FGRI females versus CON (TFAM and UCP2), with only MTATP6 expression decreased in FGRI males versus CON. Although the window during which IGF-I treatment was delivered was limited to the final 5 wk of gestation, IGF-I therapy of FGR altered the endocrine pancreas and skeletal muscle in a sex-specific manner in young adulthood.NEW & NOTEWORTHY Fetal growth restriction (FGR) is associated with compromised metabolic function throughout adulthood. Here, we explored the long-term effects of fetal IGF-I therapy on the adult pancreas and skeletal muscle. This is the first study demonstrating that IGF-I therapy of FGR has sex-specific long-term effects at both the tissue and molecular level on metabolically active tissues in adult sheep.
Collapse
Affiliation(s)
- Emma J Buckels
- The Liggins Institute, University of Auckland, Auckland, New Zealand
| | | | - Mark H Oliver
- The Liggins Institute, University of Auckland, Auckland, New Zealand
| | | | - Jane E Harding
- The Liggins Institute, University of Auckland, Auckland, New Zealand
| | - Anne L Jaquiery
- The Liggins Institute, University of Auckland, Auckland, New Zealand
| |
Collapse
|
4
|
Bovine Milk Fat Globule Epidermal Growth Factor VIII activates PI3K/Akt signaling pathway and attenuates sarcopenia in rat model induced by d-galactose. FOOD BIOSCI 2021. [DOI: 10.1016/j.fbio.2020.100847] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
5
|
Chang EI, Hetrick B, Wesolowski SR, McCurdy CE, Rozance PJ, Brown LD. A Two-Week Insulin Infusion in Intrauterine Growth Restricted Fetal Sheep at 75% Gestation Increases Skeletal Myoblast Replication but Did Not Restore Muscle Mass or Increase Fiber Number. Front Endocrinol (Lausanne) 2021; 12:785242. [PMID: 34917036 PMCID: PMC8670988 DOI: 10.3389/fendo.2021.785242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/02/2021] [Indexed: 01/14/2023] Open
Abstract
Intrauterine growth restricted (IUGR) fetuses are born with lower skeletal muscle mass, fewer proliferating myoblasts, and fewer myofibers compared to normally growing fetuses. Plasma concentrations of insulin, a myogenic growth factor, are lower in IUGR fetuses. We hypothesized that a two-week insulin infusion at 75% gestation would increase myoblast proliferation and fiber number in IUGR fetal sheep. Catheterized control fetuses received saline (CON-S, n=6), and the IUGR fetuses received either saline (IUGR-S, n=7) or insulin (IUGR-I, 0.014 ± 0.001 units/kg/hr, n=11) for 14 days. Fetal arterial blood gases and plasma amino acid levels were measured. Fetal skeletal muscles (biceps femoris, BF; and flexor digitorum superficialis, FDS) and pancreases were collected at necropsy (126 ± 2 dGA) for immunochemistry analysis, real-time qPCR, or flow cytometry. Insulin concentrations in IUGR-I and IUGR-S were lower vs. CON-S (P ≤ 0.05, group). Fetal arterial PaO2, O2 content, and glucose concentrations were lower in IUGR-I vs. CON-S (P ≤ 0.01) throughout the infusion period. IGF-1 concentrations tended to be higher in IUGR-I vs. IUGR-S (P=0.06), but both were lower vs. CON-S (P ≤ 0.0001, group). More myoblasts were in S/G2 cell cycle stage in IUGR-I vs. both IUGR-S and CON-S (145% and 113%, respectively, P ≤ 0.01). IUGR-I FDS muscle weighed 40% less and had 40% lower fiber number vs. CON-S (P ≤ 0.05) but were not different from IUGR-S. Myonuclear number per fiber and the mRNA expression levels of muscle regulatory factors were not different between groups. While the pancreatic β-cell mass was lower in both IUGR-I and IUGR-S compared to CON-S, the IUGR groups were not different from each other indicating that feedback inhibition by endogenous insulin did not reduce β-cell mass. A two-week insulin infusion at 75% gestation promoted myoblast proliferation in the IUGR fetus but did not increase fiber or myonuclear number. Myoblasts in the IUGR fetus retain the capacity to proliferate in response to mitogenic stimuli, but intrinsic defects in the fetal myoblast by 75% gestation may limit the capacity to restore fiber number.
Collapse
MESH Headings
- Animals
- Drug Administration Schedule
- Female
- Fetal Development/drug effects
- Fetal Development/physiology
- Fetal Growth Retardation/drug therapy
- Fetal Growth Retardation/pathology
- Hypoglycemic Agents/administration & dosage
- Infusions, Intravenous
- Insulin/administration & dosage
- Muscle Development/drug effects
- Muscle Development/physiology
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/pathology
- Muscle Fibers, Skeletal/physiology
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/pathology
- Muscle, Skeletal/physiology
- Myoblasts, Skeletal/drug effects
- Myoblasts, Skeletal/pathology
- Myoblasts, Skeletal/physiology
- Pregnancy
- Sheep
Collapse
Affiliation(s)
- Eileen I. Chang
- Perinatal Research Center, Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Byron Hetrick
- Department of Human Physiology, University of Oregon, Eugene, OR, United States
| | - Stephanie R. Wesolowski
- Perinatal Research Center, Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Carrie E. McCurdy
- Department of Human Physiology, University of Oregon, Eugene, OR, United States
| | - Paul J. Rozance
- Perinatal Research Center, Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Laura D. Brown
- Perinatal Research Center, Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO, United States
- *Correspondence: Laura D. Brown,
| |
Collapse
|
6
|
Kakadia JH, Jain BB, Biggar K, Sutherland A, Nygard K, Li C, Nathanielsz PW, Jansson T, Gupta MB. Hyperphosphorylation of fetal liver IGFBP-1 precedes slowing of fetal growth in nutrient-restricted baboons and may be a mechanism underlying IUGR. Am J Physiol Endocrinol Metab 2020; 319:E614-E628. [PMID: 32744097 PMCID: PMC7642856 DOI: 10.1152/ajpendo.00220.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
In cultured fetal liver cells, insulin-like growth factor (IGF) binding protein (IGFBP)-1 hyperphosphorylation in response to hypoxia and amino acid deprivation is mediated by inhibition of mechanistic target of rapamycin (mTOR) and activation of amino acid response (AAR) signaling and casein kinase (CK)2. We hypothesized that fetal liver mTOR inhibition, activation of AAR and CK2, and IGFBP-1 hyperphosphorylation occur before development of intrauterine growth restriction (IUGR). Pregnant baboons were fed a control (C) or a maternal nutrient restriction (MNR; 70% calories of control) diet starting at gestational day (GD) 30 (term GD 185). Umbilical blood and fetal liver tissue were obtained at GD 120 (C, n = 7; MNR, n = 10) and 165 (C, n = 7; MNR, n = 8). Fetal weights were unchanged at GD 120 but decreased at GD 165 in the MNR group (-13%, P = 0.03). IGFBP-1 phosphorylation, as determined by parallel reaction monitoring mass spectrometry (PRM-MS), immunohistochemistry, and/or Western blot, was enhanced in MNR fetal liver and umbilical plasma at GD 120 and 165. IGF-I receptor autophosphorylationTyr1135 (-64%, P = 0.05) was reduced in MNR fetal liver at GD 120. Furthermore, fetal liver CK2 (α/α'/β) expression, CK2β colocalization, proximity with IGFBP-1, and CK2 autophosphorylationTyr182 were greater at GD 120 and 165 in MNR vs. C. Additionally, mTOR complex (mTORC)1 (p-P70S6KThr389, -52%, P = 0.05) and mTORC2 (p-AktSer473, -56%, P < 0.001) activity were decreased and AAR was activated (p-GCN2Thr898, +117%, P = 0.02; p-eIF2αSer51, +294%, P = 0.002; p-ERKThr202, +111%, P = 0.03) in MNR liver at GD 120. Our data suggest that fetal liver IGFBP-1 hyperphosphorylation, mediated by mTOR inhibition and both AAR and CK2 activation, is a key link between restricted nutrient and oxygen availability and the development of IUGR.
Collapse
Affiliation(s)
- Jenica H Kakadia
- Department of Biochemistry, University of Western Ontario, London, Ontario, Canada
| | - Bhawani B Jain
- Department of Biochemistry, University of Western Ontario, London, Ontario, Canada
| | - Kyle Biggar
- Institute of Biochemistry, Carleton University, Ottawa, Ontario, Canada
| | - Austen Sutherland
- Department of Biochemistry, University of Western Ontario, London, Ontario, Canada
| | - Karen Nygard
- Biotron Integrated Microscopy Facility, University of Western Ontario, London, Ontario, Canada
| | - Cun Li
- University of Wyoming, Laramie, Wyoming
- Southwest National Primate Research Center, San Antonio, Texas
| | - Peter W Nathanielsz
- University of Wyoming, Laramie, Wyoming
- Southwest National Primate Research Center, San Antonio, Texas
| | - Thomas Jansson
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Madhulika B Gupta
- Department of Biochemistry, University of Western Ontario, London, Ontario, Canada
- Department of Pediatrics, University of Western Ontario, London, Ontario, Canada
- Children's Health Research Institute, London, Ontario, Canada
| |
Collapse
|
7
|
Spiroski AM, Oliver MH, Jaquiery AL, Gunn TD, Harding JE, Bloomfield FH. Effects of intrauterine insulin-like growth factor-1 therapy for fetal growth restriction on adult metabolism and body composition are sex specific. Am J Physiol Endocrinol Metab 2020; 318:E568-E578. [PMID: 32101029 DOI: 10.1152/ajpendo.00481.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Fetal growth restriction (FGR) is associated with compromised growth and metabolic function throughout life. Intrauterine therapy of FGR with intra-amniotic insulin-like growth factor-1 (IGF1) enhances fetal growth and alters perinatal metabolism and growth in a sex-specific manner, but the adult effects are unknown. We investigated the effects of intra-amniotic IGF1 treatment of FGR on adult growth and body composition, adrenergic sensitivity, and glucose-insulin axis regulation. Placental embolization-induced FGR was treated with four weekly doses of 360 µg intra-amniotic IGF1 (FGRI) or saline (FGRS). Offspring were raised to adulthood (18 mo: FGRI, n = 12 females, 12 males; FGRS, n = 13 females, 10 males) alongside offspring from unembolized and untreated sheep (CON; n = 12 females, 21 males). FGRI females had increased relative lean mass compared with CON but not FGRS (P < 0.05; 70.6 ± 8.2% vs. 61.4 ± 8.2% vs. 67.6 ± 8.2%), decreased abdominal adipose compared with CON and FGRS (P < 0.05; 43.7 ± 1.2% vs. 49.3 ± 0.9% vs. 48.5 ± 1.0%), increased glucose utilization compared with FGRS but not CON (P < 0.05; 9.6 ± 1.0 vs. 6.0 ± 0.9 vs. 7.6 ± 0.9 mg·kg-1·min-1), and increased β-hydroxybutyric acid:nonesterified fatty acid ratio in response to adrenaline compared with CON and FGRS (P < 0.05; 3.9 ± 1.4 vs. 1.1 ± 1.4 vs. 1.8 ± 1.4). FGRS males were smaller and lighter compared with CON but not FGRI (P < 0.05; 86.8 ± 6.3 vs. 93.5 ± 6.1 vs. 90.7 ± 6.3 kg), with increased peak glucose concentration (10%) in response to a glucose load but few other differences. These effects of intra-amniotic IGF1 therapy on adult body composition, glucose-insulin axis function, and adrenergic sensitivity could indicate improved metabolic regulation during young adulthood in female FGR sheep.
Collapse
Affiliation(s)
| | - Mark Hope Oliver
- The Liggins Institute, University of Auckland, Auckland, New Zealand
| | | | - Travis Dane Gunn
- The Liggins Institute, University of Auckland, Auckland, New Zealand
| | | | | |
Collapse
|
8
|
Zhang J, Xu W, Han H, Zhang L, Wang T. Dietary Leucine Supplementation Restores Serum Glucose Levels, and Modifying Hepatic Gene Expression Related to the Insulin Signal Pathway in IUGR Piglets. Animals (Basel) 2019; 9:ani9121138. [PMID: 31847151 PMCID: PMC6941017 DOI: 10.3390/ani9121138] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/09/2019] [Accepted: 12/11/2019] [Indexed: 12/15/2022] Open
Abstract
Simple Summary Intrauterine malnutrition may compromise the size and structure of fetal organs and tissues, which leads to lower birth weight and a slower rate of growth after weaning. Intrauterine growth restriction/retardation (IUGR) impairs pancreas function, resulting in the decreased glucose levels in serum. Leucine, one of branched chain amino acids, is an essential amino acid and the substrate of protein synthesis. Leucine also acts as a major regulator of hormone signal transduction, like insulin. Dietary branched chain amino acids or leucine have beneficial effects on the glucose metabolism and glycogen synthesis of muscle. Leucine supplementation improves the insulin sensitivity in liver and muscle and then influences the systemic glucose homeostasis. However, it is still unclear whether leucine supplementation would alter insulin sensitivity in IUGR neonatal piglets. Our results showed that dietary leucine supplementation restored serum glucose concentrations, increased insulin and creatinine concentrations, and enhanced protein kinase adenosine monophosphate-activated γ 3-subunit and glucose transporter type 2 expression. These findings suggest that leucine might play a positive role in hepatic lipid metabolism and glucose metabolism in IUGR. Abstract This study aimed to investigate the effects of leucine with different levels on the insulin resistance in intrauterine growth restriction/retardation (IUGR) piglets. Thirty-two weaned piglets were arranged in a 2 × 2 factorial design and four treatments (n = 8) were as follow: (1) normal weaned piglets fed a basal diet (CONT), (2) IUGR weaned piglets fed a basal diet (IUGR), (3) normal weaned piglets fed a basal diet with the addition of 0.35% l-leucine (C-LEU), and (4) IUGR fed a basal diet with the addition of 0.35% l-leucine (I-LEU) for a 21-days trial. The results showed that compared to the IUGR group, the I-LEU group had higher final body weight and body weight gain, higher serum glucose concentrations, and higher serum insulin concentrations (p < 0.05). The gene expression of phosphatidylinositol 3-kinase p110 gamma, protein kinase adenosine monophosphate-activated γ 3-subunit, glycogen synthase kinase-3 alpha, and glucose transporter type 2 were increased in the I-LEU group as compared to the IUGR group (p < 0.05). It was concluded that dietary leucine supplementation restored serum glucose concentrations, increased insulin and creatinine concentrations, and enhanced protein kinase adenosine monophosphate-activated γ 3-subunit and glucose transporter type 2 expression, suggesting that leucine might play a positive role in hepatic lipid metabolism and glucose metabolism in IUGR.
Collapse
Affiliation(s)
| | | | | | | | - Tian Wang
- Correspondence: ; Tel./Fax: +86-25-84395156
| |
Collapse
|
9
|
Cao Y, Sun W, Xu G. Fuzhu jiangtang granules combined with metformin reduces insulin resistance in skeletal muscle of diabetic rats via PI3K/Akt signaling. PHARMACEUTICAL BIOLOGY 2019; 57:660-668. [PMID: 31545909 PMCID: PMC6764404 DOI: 10.1080/13880209.2019.1659831] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 08/05/2019] [Accepted: 08/11/2019] [Indexed: 06/10/2023]
Abstract
Context: Fuzhu Jiangtang Granules (FJG) are a traditional Chinese used in the treatment of diabetes mellitus. However, the antidiabetic mechanism of FJG is not clear. Objective: This study evaluates and determines the antidiabetic mechanism of FJG using a Zucker diabetic fatty (ZDF) rat model. Materials and methods: ZDF (fa/fa) rats were divided into four groups (n = 6): diabetes mellitus (DM), metformin (Met, 0.134 g/kg b.w./day), FJG (0.64 g/kg b.w./day), and combination (Com, 0.134 g/kg b.w./day of Met and 0.64 g/kg b.w./day of FJG). Six ZDF (fa/+) rats served as a normal control. After 6 weeks, biochemical parameters gene and protein expression were detected. Results: The FBG, bodyweight, triglyceride (TG), total cholesterol (TC), free fatty acid (FFA), insulin levels, and HOMA-IR were lower in the FJG than in the DM group (p < 0.05, p < 0.01). In an oral glucose tolerance test, the AUC in the FJG group was significantly lower (p < 0.01). The levels of superoxide dismutase and catalase were higher in the FJG than in the DM group (p < 0.01); the malondialdehyde content and TNF-α were significantly decreased in the FJG group (p < 0.01). FJG increased the mRNA expression of IR and GLUT4 significantly (p < 0.05, p < 0.01). The protein levels of IR, p-IRS1 tyr989, m-PI3Kp85, p-Akt and GLUT4 were increased in the FJG (p < 0.05, p < 0.01), but the protein levels of p-IRS1 ser1101/612/307 were significantly decreased in the JG group (p < 0.01). Discussion and conclusions: The antidiabetic mechanism of FJG may be related to regulation of the insulin-signaling pathway in skeletal muscle. These aspects require further research.
Collapse
Affiliation(s)
- Yunsong Cao
- Department of Nephrology, Dongfang Hospital, Beijing University of Traditional Chinese Medicine, Beijing, China
| | - Wen Sun
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, China
| | - Guangyuan Xu
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, China
- Department of Traditional Chinese Medicine, Fuxing Hospital, the Eighth Clinical Medical College, Capital Medical University, Beijing, China
| |
Collapse
|
10
|
Chang EI, Wesolowski SR, Gilje EA, Baker PR, Reisz JA, D’Alessandro A, Hay, WW, Rozance PJ, Brown LD. Skeletal muscle amino acid uptake is lower and alanine production is greater in late gestation intrauterine growth-restricted fetal sheep hindlimb. Am J Physiol Regul Integr Comp Physiol 2019; 317:R615-R629. [PMID: 31483682 PMCID: PMC6879841 DOI: 10.1152/ajpregu.00115.2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
In a sheep model of intrauterine growth restriction (IUGR) produced from placental insufficiency, late gestation fetuses had smaller skeletal muscle mass, myofiber area, and slower muscle protein accretion rates compared with normally growing fetuses. We hypothesized that IUGR fetal muscle develops adaptations that divert amino acids (AAs) from protein accretion and activate pathways that conserve substrates for other organs. We placed hindlimb arterial and venous catheters into late gestation IUGR (n = 10) and control (CON, n = 8) fetal sheep and included an external iliac artery flow probe to measure hindlimb AA uptake rates. Arterial and venous plasma samples and biceps femoris muscle were analyzed by mass spectrometry-based metabolomics. IUGR fetuses had greater abundance of metabolites enriched within the alanine, aspartate, and glutamate metabolism pathway compared with CON. Net uptake rates of branched-chain AA (BCAA) were lower by 42%-73%, and muscle ammoniagenic AAs (alanine, glycine, and glutamine) were lower by 107%-158% in IUGR hindlimbs versus CON. AA uptake rates correlated with hindlimb weight; the smallest hindlimbs showed net release of ammoniagenic AAs. Gene expression levels indicated a decrease in BCAA catabolism in IUGR muscle. Plasma purines were lower and plasma uric acid was higher in IUGR versus CON, possibly a reflection of ATP conservation. We conclude that IUGR skeletal muscle has lower BCAA uptake and develops adaptations that divert AAs away from protein accretion into alternative pathways that sustain global energy production and nitrogen disposal in the form of ammoniagenic AAs for metabolism in other organs.
Collapse
Affiliation(s)
- Eileen I. Chang
- 1Department of Pediatrics, Section of Neonatology, Perinatal Research Center, University of Colorado School of Medicine, Aurora, Colorado
| | - Stephanie R. Wesolowski
- 1Department of Pediatrics, Section of Neonatology, Perinatal Research Center, University of Colorado School of Medicine, Aurora, Colorado
| | - Elizabeth A. Gilje
- 1Department of Pediatrics, Section of Neonatology, Perinatal Research Center, University of Colorado School of Medicine, Aurora, Colorado
| | - Peter R. Baker
- 2Department of Pediatrics, Section of Genetics and Metabolism, University of Colorado School of Medicine, Aurora, Colorado
| | - Julie A. Reisz
- 3Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado
| | - Angelo D’Alessandro
- 3Department of Biochemistry and Molecular Genetics, University of Colorado School of Medicine, Aurora, Colorado
| | - William W. Hay,
- 1Department of Pediatrics, Section of Neonatology, Perinatal Research Center, University of Colorado School of Medicine, Aurora, Colorado
| | - Paul J. Rozance
- 1Department of Pediatrics, Section of Neonatology, Perinatal Research Center, University of Colorado School of Medicine, Aurora, Colorado
| | - Laura D. Brown
- 1Department of Pediatrics, Section of Neonatology, Perinatal Research Center, University of Colorado School of Medicine, Aurora, Colorado
| |
Collapse
|
11
|
Horton DM, Saint DA, Gatford KL, Kind KL, Owens JA. Sex-specific programming of adult insulin resistance in guinea pigs by variable perinatal growth induced by spontaneous variation in litter size. Am J Physiol Regul Integr Comp Physiol 2019; 316:R352-R361. [PMID: 30735437 DOI: 10.1152/ajpregu.00341.2018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Intrauterine growth restriction (IUGR) and subsequent neonatal catch-up growth are implicated in programming of insulin resistance later in life. Spontaneous IUGR in the guinea pig, due to natural variation in litter size, produces offspring with asymmetric IUGR and neonatal catch-up growth. We hypothesized that spontaneous IUGR and/or accelerated neonatal growth would impair insulin sensitivity in adult guinea pigs. Insulin sensitivity of glucose metabolism was determined by hyperinsulinemic-euglycemic clamp (HEC) in 38 (21 male, 17 female) young adult guinea pigs from litters of two-to-four pups. A subset (10 male, 8 female) were infused with d-[3-3H]glucose before and during the HEC to determine rates of basal and insulin-stimulated glucose utilization, storage, glycolysis, and endogenous glucose production. n males, the insulin sensitivity of whole body glucose uptake ( r = 0.657, P = 0.002) and glucose utilization ( r = 0.884, P = 0.004) correlated positively and independently with birth weight, but not with neonatal fractional growth rate (FGR10-28). In females, the insulin sensitivity of whole body and partitioned glucose metabolism was not related to birth weight, but that of endogenous glucose production correlated negatively and independently with FGR10-28 ( r = -0.815, P = 0.025). Thus, perinatal growth programs insulin sensitivity of glucose metabolism in the young adult guinea pig and in a sex-specific manner; impaired insulin sensitivity, including glucose utilization, occurs after IUGR in males and impaired hepatic insulin sensitivity after rapid neonatal growth in females.
Collapse
Affiliation(s)
- Dane M Horton
- Robinson Research Institute, The University of Adelaide , Adelaide, South Australia , Australia.,Adelaide Medical School, The University of Adelaide , Adelaide, South Australia , Australia
| | - David A Saint
- Adelaide Medical School, The University of Adelaide , Adelaide, South Australia , Australia
| | - Kathryn L Gatford
- Robinson Research Institute, The University of Adelaide , Adelaide, South Australia , Australia.,Adelaide Medical School, The University of Adelaide , Adelaide, South Australia , Australia
| | - Karen L Kind
- Robinson Research Institute, The University of Adelaide , Adelaide, South Australia , Australia.,School of Animal and Veterinary Sciences, The University of Adelaide , Adelaide, South Australia , Australia
| | - Julie A Owens
- Robinson Research Institute, The University of Adelaide , Adelaide, South Australia , Australia.,Adelaide Medical School, The University of Adelaide , Adelaide, South Australia , Australia.,Office of the Deputy Vice-Chancellor Research, Deakin University, Waurn Ponds, Geelong, Victoria , Australia
| |
Collapse
|
12
|
Copper or/and arsenic induces autophagy by oxidative stress-related PI3K/AKT/mTOR pathways and cascaded mitochondrial fission in chicken skeletal muscle. J Inorg Biochem 2018; 188:1-8. [DOI: 10.1016/j.jinorgbio.2018.08.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Revised: 07/21/2018] [Accepted: 08/01/2018] [Indexed: 12/23/2022]
|
13
|
Yates DT, Petersen JL, Schmidt TB, Cadaret CN, Barnes TL, Posont RJ, Beede KA. ASAS-SSR Triennnial Reproduction Symposium: Looking Back and Moving Forward-How Reproductive Physiology has Evolved: Fetal origins of impaired muscle growth and metabolic dysfunction: Lessons from the heat-stressed pregnant ewe. J Anim Sci 2018; 96:2987-3002. [PMID: 29701769 PMCID: PMC6095381 DOI: 10.1093/jas/sky164] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 04/24/2018] [Indexed: 12/11/2022] Open
Abstract
Intrauterine growth restriction (IUGR) is the second leading cause of perinatal mortality and predisposes offspring to metabolic disorders at all stages of life. Muscle-centric fetal adaptations reduce growth and yield metabolic parsimony, beneficial for IUGR fetal survival but detrimental to metabolic health after birth. Epidemiological studies have reported that IUGR-born children experience greater prevalence of insulin resistance and obesity, which progresses to diabetes, hypertension, and other metabolic disorders in adulthood that reduce quality of life. Similar adaptive programming in livestock results in decreased birth weights, reduced and inefficient growth, decreased carcass merit, and substantially greater mortality rates prior to maturation. High rates of glucose consumption and metabolic plasticity make skeletal muscle a primary target for nutrient-sparing adaptations in the IUGR fetus, but at the cost of its contribution to proper glucose homeostasis after birth. Identifying the mechanisms underlying IUGR pathophysiology is a fundamental step in developing treatments and interventions to improve outcomes in IUGR-born humans and livestock. In this review, we outline the current knowledge regarding the adaptive restriction of muscle growth and alteration of glucose metabolism that develops in response to progressively exacerbating intrauterine conditions. In addition, we discuss the evidence implicating developmental changes in β adrenergic and inflammatory systems as key mechanisms for dysregulation of these processes. Lastly, we highlight the utility and importance of sheep models in developing this knowledge.
Collapse
Affiliation(s)
- Dustin T Yates
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE
| | - Jessica L Petersen
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE
| | - Ty B Schmidt
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE
| | - Caitlin N Cadaret
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE
| | - Taylor L Barnes
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE
| | - Robert J Posont
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE
| | - Kristin A Beede
- Department of Animal Science, University of Nebraska-Lincoln, Lincoln, NE
| |
Collapse
|
14
|
Prabakaran AD, Karakkat JV, Vijayan R, Chalissery J, Ibrahim MF, Kaimala S, Adeghate EA, Al-Marzouqi AH, Ansari SA, Mensah-Brown E, Emerald BS. Identification of early indicators of altered metabolism in normal development using a rodent model system. Dis Model Mech 2018; 11:dmm.031815. [PMID: 29434026 PMCID: PMC5897726 DOI: 10.1242/dmm.031815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2017] [Accepted: 01/15/2018] [Indexed: 11/20/2022] Open
Abstract
Although the existence of a close relationship between the early maternal developmental environment, fetal size at birth and the risk of developing disease in adulthood has been suggested, most studies, however, employed experimentally induced intrauterine growth restriction as a model to link this with later adult disease. Because embryonic size variation also occurs under normal growth and differentiation, elucidating the molecular mechanisms underlying these changes and their relevance to later adult disease risk becomes important. The birth weight of rat pups vary according to the uterine horn positions. Using birth weight as a marker, we compared two groups of rat pups – lower birth weight (LBW, 5th to 25th percentile) and average birth weight (ABW, 50th to 75th percentile) – using morphological, biochemical and molecular biology, and genetic techniques. Our results show that insulin metabolism, Pi3k/Akt and Pparγ signaling and the genes regulating growth and metabolism are significantly different in these groups. Methylation at the promoter of the InsII (Ins2) gene and DNA methyltransferase 1 in LBW pups are both increased. Additionally, the Dnmt1 repressor complex, which includes Hdac1, Rb (Rb1) and E2f1, was also upregulated in LBW pups. We conclude that the Dnmt1 repressor complex, which regulates the restriction point of the cell cycle, retards the rate at which cells traverse the G1 or G0 phase of the cell cycle in LBW pups, thereby slowing down growth. This regulatory mechanism mediated by Dnmt1 might contribute to the production of small-size pups and altered physiology and pathology in adult life. Summary: This study suggests an important link between the early embryonic environment and later adult physiology and pathology. At least one process by which this might be coordinated is through the regulatory mechanisms mediated by Dnmt1.
Collapse
Affiliation(s)
- Ashok Daniel Prabakaran
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, PO Box 17666, Abu Dhabi, UAE
| | - Jimsheena Valiyakath Karakkat
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, PO Box 17666, Abu Dhabi, UAE
| | - Ranjit Vijayan
- Department of Biology, College of Science, United Arab Emirates University, Al Ain, PO Box 17666, Abu Dhabi, UAE
| | - Jisha Chalissery
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, PO Box 17666, Abu Dhabi, UAE
| | - Marwa F Ibrahim
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, PO Box 17666, Abu Dhabi, UAE
| | - Suneesh Kaimala
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, PO Box 17666, Abu Dhabi, UAE
| | - Ernest A Adeghate
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, PO Box 17666, Abu Dhabi, UAE
| | - Ahmed Hassan Al-Marzouqi
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, PO Box 17666, Abu Dhabi, UAE.,Department of Basic Medical Sciences, College of Medicine, Mohammed Bin Rashid University of Medicine and Health Sciences, Dubai Healthcare City, PO Box 505055, Dubai, UAE
| | - Suraiya Anjum Ansari
- Department of Biochemistry, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, PO Box 17666, Abu Dhabi, UAE
| | - Eric Mensah-Brown
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, PO Box 17666, Abu Dhabi, UAE
| | - Bright Starling Emerald
- Department of Anatomy, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, PO Box 17666, Abu Dhabi, UAE
| |
Collapse
|
15
|
Soto SM, Blake AC, Wesolowski SR, Rozance PJ, Barthel KB, Gao B, Hetrick B, McCurdy CE, Garza NG, Hay WW, Leinwand LA, Friedman JE, Brown LD. Myoblast replication is reduced in the IUGR fetus despite maintained proliferative capacity in vitro. J Endocrinol 2017; 232:475-491. [PMID: 28053000 PMCID: PMC5440081 DOI: 10.1530/joe-16-0123] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 01/04/2017] [Indexed: 02/04/2023]
Abstract
Adults who were affected by intrauterine growth restriction (IUGR) suffer from reductions in muscle mass and insulin resistance, suggesting muscle growth may be restricted by molecular events that occur during fetal development. To explore the basis of restricted fetal muscle growth, we used a sheep model of progressive placental insufficiency-induced IUGR to assess myoblast proliferation within intact skeletal muscle in vivo and isolated myoblasts stimulated with insulin in vitro Gastrocnemius and soleus muscle weights were reduced by 25% in IUGR fetuses compared to those in controls (CON). The ratio of PAX7+ nuclei (a marker of myoblasts) to total nuclei was maintained in IUGR muscle compared to CON, but the fraction of PAX7+ myoblasts that also expressed Ki-67 (a marker of cellular proliferation) was reduced by 23%. Despite reduced proliferation in vivo, fetal myoblasts isolated from IUGR biceps femoris and cultured in enriched media in vitro responded robustly to insulin in a dose- and time-dependent manner to increase proliferation. Similarly, insulin stimulation of IUGR myoblasts upregulated key cell cycle genes and DNA replication. There were no differences in the expression of myogenic regulatory transcription factors that drive commitment to muscle differentiation between CON and IUGR groups. These results demonstrate that the molecular machinery necessary for transcriptional control of proliferation remains intact in IUGR fetal myoblasts, indicating that in vivo factors such as reduced insulin and IGF1, hypoxia and/or elevated counter-regulatory hormones may be inhibiting muscle growth in IUGR fetuses.
Collapse
Affiliation(s)
- Susan M Soto
- Department of PediatricsUniversity of Colorado School of Medicine, Perinatal Research Center, Aurora, Colorado, USA
| | - Amy C Blake
- Department of PediatricsUniversity of Colorado School of Medicine, Perinatal Research Center, Aurora, Colorado, USA
| | - Stephanie R Wesolowski
- Department of PediatricsUniversity of Colorado School of Medicine, Perinatal Research Center, Aurora, Colorado, USA
| | - Paul J Rozance
- Department of PediatricsUniversity of Colorado School of Medicine, Perinatal Research Center, Aurora, Colorado, USA
| | - Kristen B Barthel
- Department of MolecularCellular, and Developmental Biology, University of Colorado Boulder, BioFrontiers Institute, Boulder, Colorado, USA
| | - Bifeng Gao
- Department of MedicineUniversity of Colorado School of Medicine, Aurora, Colorado, USA
| | - Byron Hetrick
- Department of Human PhysiologyUniversity of Oregon, Eugene, Oregon, USA
| | - Carrie E McCurdy
- Department of Human PhysiologyUniversity of Oregon, Eugene, Oregon, USA
| | - Natalia G Garza
- Department of PediatricsUniversity of Colorado School of Medicine, Perinatal Research Center, Aurora, Colorado, USA
| | - William W Hay
- Department of PediatricsUniversity of Colorado School of Medicine, Perinatal Research Center, Aurora, Colorado, USA
| | - Leslie A Leinwand
- Department of MolecularCellular, and Developmental Biology, University of Colorado Boulder, BioFrontiers Institute, Boulder, Colorado, USA
| | - Jacob E Friedman
- Department of PediatricsUniversity of Colorado School of Medicine, Perinatal Research Center, Aurora, Colorado, USA
| | - Laura D Brown
- Department of PediatricsUniversity of Colorado School of Medicine, Perinatal Research Center, Aurora, Colorado, USA
| |
Collapse
|
16
|
De Matteo R, Hodgson DJ, Bianco-Miotto T, Nguyen V, Owens JA, Harding R, Allison BJ, Polglase G, Black MJ, Gatford KL. Betamethasone-exposed preterm birth does not impair insulin action in adult sheep. J Endocrinol 2017; 232:175-187. [PMID: 27821470 DOI: 10.1530/joe-16-0300] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Accepted: 11/07/2016] [Indexed: 12/13/2022]
Abstract
Preterm birth is associated with increased risk of type 2 diabetes (T2D) in adulthood; however, the underlying mechanisms are poorly understood. We therefore investigated the effect of preterm birth at ~0.9 of term after antenatal maternal betamethasone on insulin sensitivity, secretion and key determinants in adulthood, in a clinically relevant animal model. Glucose tolerance and insulin secretion (intravenous glucose tolerance test) and whole-body insulin sensitivity (hyperinsulinaemic euglycaemic clamp) were measured and tissue collected in young adult sheep (14 months old) after epostane-induced preterm (9M, 7F) or term delivery (11M, 6F). Glucose tolerance and disposition, insulin secretion, β-cell mass and insulin sensitivity did not differ between term and preterm sheep. Hepatic PRKAG2 expression was greater in preterm than in term males (P = 0.028), but did not differ between preterm and term females. In skeletal muscle, SLC2A4 (P = 0.019), PRKAA2 (P = 0.021) and PRKAG2 (P = 0.049) expression was greater in preterm than in term overall and in males, while INSR (P = 0.047) and AKT2 (P = 0.043) expression was greater in preterm than in term males only. Hepatic PRKAG2 expression correlated positively with whole-body insulin sensitivity in males only. Thus, preterm birth at 0.9 of term after betamethasone does not impair insulin sensitivity or secretion in adult sheep, and has sex-specific effects on gene expression of the insulin signalling pathway. Hence, the increased risk of T2D in preterm humans may be due to factors that initiate preterm delivery or in early neonatal exposures, rather than preterm birth per se.
Collapse
Affiliation(s)
- R De Matteo
- Department of Anatomy and Developmental BiologyMonash University, Clayton, Victoria, Australia
| | - D J Hodgson
- Robinson Research InstituteUniversity of Adelaide, Adelaide, South Australia, Australia
- Adelaide Medical SchoolUniversity of Adelaide, Adelaide, South Australia, Australia
| | - T Bianco-Miotto
- Robinson Research InstituteUniversity of Adelaide, Adelaide, South Australia, Australia
- School of AgricultureFood and Wine, University of Adelaide, Adelaide, South Australia, Australia
| | - V Nguyen
- Department of Anatomy and Developmental BiologyMonash University, Clayton, Victoria, Australia
| | - J A Owens
- Robinson Research InstituteUniversity of Adelaide, Adelaide, South Australia, Australia
- Adelaide Medical SchoolUniversity of Adelaide, Adelaide, South Australia, Australia
| | - R Harding
- Department of Anatomy and Developmental BiologyMonash University, Clayton, Victoria, Australia
| | - B J Allison
- Department of Obstetrics & GynaecologyMonash University, Clayton, Victoria, Australia
- The Ritchie CentreHudson Institute of Medical Research, Clayton, Victoria, Australia
| | - G Polglase
- Department of Obstetrics & GynaecologyMonash University, Clayton, Victoria, Australia
- The Ritchie CentreHudson Institute of Medical Research, Clayton, Victoria, Australia
| | - M J Black
- Department of Anatomy and Developmental BiologyMonash University, Clayton, Victoria, Australia
| | - K L Gatford
- Robinson Research InstituteUniversity of Adelaide, Adelaide, South Australia, Australia
- Adelaide Medical SchoolUniversity of Adelaide, Adelaide, South Australia, Australia
| |
Collapse
|
17
|
Brown LD, Hay WW. Impact of placental insufficiency on fetal skeletal muscle growth. Mol Cell Endocrinol 2016; 435:69-77. [PMID: 26994511 PMCID: PMC5014698 DOI: 10.1016/j.mce.2016.03.017] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 03/03/2016] [Accepted: 03/14/2016] [Indexed: 02/07/2023]
Abstract
Intrauterine growth restriction (IUGR) caused by placental insufficiency is one of the most common and complex problems in perinatology, with no known cure. In pregnancies affected by placental insufficiency, a poorly functioning placenta restricts nutrient supply to the fetus and prevents normal fetal growth. Among other significant deficits in organ development, the IUGR fetus characteristically has less lean body and skeletal muscle mass than their appropriately-grown counterparts. Reduced skeletal muscle growth is not fully compensated after birth, as individuals who were born small for gestational age (SGA) from IUGR have persistent reductions in muscle mass and strength into adulthood. The consequences of restricted muscle growth and accelerated postnatal "catch-up" growth in the form of adiposity may contribute to the increased later life risk for visceral adiposity, peripheral insulin resistance, diabetes, and cardiovascular disease in individuals who were formerly IUGR. This review will discuss how an insufficient placenta results in impaired fetal skeletal muscle growth and how lifelong reductions in muscle mass might contribute to increased metabolic disease risk in this vulnerable population.
Collapse
Affiliation(s)
- Laura D Brown
- Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus F441, Perinatal Research Center, 13243 East 23rd Avenue, Aurora, CO 80045, United States.
| | - William W Hay
- Department of Pediatrics, University of Colorado School of Medicine, Anschutz Medical Campus F441, Perinatal Research Center, 13243 East 23rd Avenue, Aurora, CO 80045, United States.
| |
Collapse
|
18
|
Lee GH, Oh KJ, Kim HR, Han HS, Lee HY, Park KG, Nam KH, Koo SH, Chae HJ. Effect of BI-1 on insulin resistance through regulation of CYP2E1. Sci Rep 2016; 6:32229. [PMID: 27576594 PMCID: PMC5006057 DOI: 10.1038/srep32229] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Accepted: 08/04/2016] [Indexed: 12/21/2022] Open
Abstract
Diet-induced obesity is a major contributing factor to the progression of hepatic insulin resistance. Increased free fatty acids in liver enhances endoplasmic reticulum (ER) stress and production of reactive oxygen species (ROS), both are directly responsible for dysregulation of hepatic insulin signaling. BI-1, a recently studied ER stress regulator, was examined to investigate its association with ER stress and ROS in insulin resistance models. To induce obesity and insulin resistance, BI-1 wild type and BI-1 knock-out mice were fed a high-fat diet for 8 weeks. The BI-1 knock-out mice had hyperglycemia, was associated with impaired glucose and insulin tolerance under high-fat diet conditions. Increased activity of NADPH-dependent CYP reductase-associated cytochrome p450 2E1 (CYP2E1) and exacerbation of ER stress in the livers of BI-1 knock-out mice was also observed. Conversely, stable expression of BI-1 in HepG2 hepatocytes was shown to reduce palmitate-induced ER stress and CYP2E1-dependent ROS production, resulting in the preservation of intact insulin signaling. Stable expression of CYP2E1 led to increased ROS production and dysregulation of insulin signaling in hepatic cells, mimicking palmitate-mediated hepatic insulin resistance. We propose that BI-1 protects against obesity-induced hepatic insulin resistance by regulating CYP2E1 activity and ROS production.
Collapse
Affiliation(s)
- Geum-Hwa Lee
- Department of Pharmacology and New Drug Development Institute, Medical School, Chonbuk National University, Jeonju, 561-181, Republic of Korea
| | - Kyoung-Jin Oh
- Division of Life Sciences, Korea University, 145 Anam-Ro, Seongbuk-Gu, Seoul, 136-713, Republic of Korea.,Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, 305-806, Republic of Korea
| | - Hyung-Ryong Kim
- Department of Dental Pharmacology and Wonkwang Dental Research Institute, School of Dentistry, Wonkwang University, Iksan, 570-749, Republic of Korea
| | - Hye-Sook Han
- Division of Life Sciences, Korea University, 145 Anam-Ro, Seongbuk-Gu, Seoul, 136-713, Republic of Korea
| | - Hwa-Young Lee
- Department of Pharmacology and New Drug Development Institute, Medical School, Chonbuk National University, Jeonju, 561-181, Republic of Korea
| | - Keun-Gyu Park
- Department of Internal Medicine, Kyungpook National University School of Medicine, Daegu, 700-721, Republic of Korea
| | - Ki-Hoan Nam
- Laboratory Animal Resource Center, KRIBB, Ochang-eup, 363-883, Republic of Korea
| | - Seung-Hoi Koo
- Division of Life Sciences, Korea University, 145 Anam-Ro, Seongbuk-Gu, Seoul, 136-713, Republic of Korea
| | - Han-Jung Chae
- Department of Pharmacology and New Drug Development Institute, Medical School, Chonbuk National University, Jeonju, 561-181, Republic of Korea
| |
Collapse
|
19
|
Wang J, Cao M, Yang M, Lin Y, Che L, Fang Z, Xu S, Feng B, Li J, Wu D. Intra-uterine undernutrition amplifies age-associated glucose intolerance in pigs via altered DNA methylation at muscle GLUT4 promoter. Br J Nutr 2016; 116:390-401. [PMID: 27265204 DOI: 10.1017/s0007114516002166] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The present study aimed to investigate the effect of maternal malnutrition on offspring glucose tolerance and the epigenetic mechanisms involved. In total, twelve primiparous Landrace×Yorkshire gilts were fed rations providing either 100 % (control (CON)) or 75 % (undernutrition (UN)) nutritional requirements according to the National Research Council recommendations, throughout gestation. Muscle samples of offspring were collected at birth (dpn1), weaning (dpn28) and adulthood (dpn189). Compared with CON pigs, UN pigs showed lower serum glucose concentrations at birth, but showed higher serum glucose and insulin concentrations as well as increased area under the blood glucose curve during intravenous glucose tolerance test at dpn189 (P<0·05). Compared with CON pigs, GLUT-4 gene and protein expressions were decreased at dpn1 and dpn189 in the muscle of UN pigs, which was accompanied by increased methylation at the GLUT4 promoter (P<0·05). These alterations in methylation concurred with increased mRNA levels of DNA methyltransferase (DNMT) 1 at dpn1 and dpn28, DNMT3a at dpn189 and DNMT3b at dpn1 in UN pigs compared with CON pigs (P<0·05). Interestingly, although the average methylation levels at the muscle GLUT4 promoter were decreased at dpn189 compared with dpn1 in pigs exposed to a poor maternal diet (P<0·05), the methylation differences in individual CpG sites were more pronounced with age. Our results indicate that in utero undernutrition persists to silence muscle GLUT4 likely through DNA methylation during the ageing process, which may lead to the amplification of age-associated glucose intolerance.
Collapse
Affiliation(s)
- Jun Wang
- Institute of Animal Nutrition,Sichuan Agricultural University,No. 211,Huimin Road,Wenjiang District,Chengdu,Sichuan 611130,People's Republic of China
| | - Meng Cao
- Institute of Animal Nutrition,Sichuan Agricultural University,No. 211,Huimin Road,Wenjiang District,Chengdu,Sichuan 611130,People's Republic of China
| | - Mei Yang
- Institute of Animal Nutrition,Sichuan Agricultural University,No. 211,Huimin Road,Wenjiang District,Chengdu,Sichuan 611130,People's Republic of China
| | - Yan Lin
- Institute of Animal Nutrition,Sichuan Agricultural University,No. 211,Huimin Road,Wenjiang District,Chengdu,Sichuan 611130,People's Republic of China
| | - Lianqiang Che
- Institute of Animal Nutrition,Sichuan Agricultural University,No. 211,Huimin Road,Wenjiang District,Chengdu,Sichuan 611130,People's Republic of China
| | - Zhengfeng Fang
- Institute of Animal Nutrition,Sichuan Agricultural University,No. 211,Huimin Road,Wenjiang District,Chengdu,Sichuan 611130,People's Republic of China
| | - Shengyu Xu
- Institute of Animal Nutrition,Sichuan Agricultural University,No. 211,Huimin Road,Wenjiang District,Chengdu,Sichuan 611130,People's Republic of China
| | - Bin Feng
- Institute of Animal Nutrition,Sichuan Agricultural University,No. 211,Huimin Road,Wenjiang District,Chengdu,Sichuan 611130,People's Republic of China
| | - Jian Li
- Institute of Animal Nutrition,Sichuan Agricultural University,No. 211,Huimin Road,Wenjiang District,Chengdu,Sichuan 611130,People's Republic of China
| | - De Wu
- Institute of Animal Nutrition,Sichuan Agricultural University,No. 211,Huimin Road,Wenjiang District,Chengdu,Sichuan 611130,People's Republic of China
| |
Collapse
|
20
|
Brøns C, Saltbæk PN, Friedrichsen M, Chen Y, Vaag A. Endocrine and metabolic diurnal rhythms in young adult men born small vs appropriate for gestational age. Eur J Endocrinol 2016; 175:29-40. [PMID: 27252486 DOI: 10.1530/eje-16-0177] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 04/20/2016] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Sleep disturbances and alterations of diurnal endocrine rhythms are associated with increased risk of type 2 diabetes (T2D). We previously showed that young men born small for gestational age (SGA) and with increased risk of T2D have elevated fat and decreased glucose oxidation rates during nighttime. In this study, we investigated whether SGA men have an altered diurnal profile of hormones, substrates and inflammatory markers implicated in T2D pathophysiology compared with matched individuals born appropriate for gestational age (AGA). METHODS We collected hourly blood samples for 24 h, to measure levels of glucose, free fatty acids (FFA), triglycerides (TG), insulin, C-peptide, leptin, resistin, ghrelin, plasminogen activator inhibitor-1 (PAI-1), incretins (GLP-1 and GIP), and inflammatory markers (TNF-α and IL-6) in 13 young men born SGA and 11 young men born AGA. RESULTS Repeated measurements analyses were used to analyze the diurnal variations and differences between groups. The SGA subjects had increased 24-h glucose (P=0.03), glucagon (P=0.03) and resistin (P=0.003) levels with no difference in diurnal rhythms compared with AGA controls. We found significant diurnal variations in levels of blood glucose, plasma TG, FFA, insulin, C-peptide, GLP-1, GIP, leptin, visfatin, TNF-α, IL-6 and PAI-1. The variation in FFA levels differed between the groups during the evening. Plasma ghrelin and glucagon levels did not display diurnal variations. CONCLUSIONS Young men born SGA exhibit elevated 24-h blood glucose, and plasma glucagon and resistin levels with no major differences in diurnal rhythms of these or other key metabolic hormones, substrates or inflammatory markers implicated in the origin of adiposity and T2D.
Collapse
Affiliation(s)
- Charlotte Brøns
- Department of Endocrinology (Diabetes and Metabolism)Rigshospitalet, Copenhagen, Denmark Steno Diabetes CenterGentofte, Denmark
| | - Pernille N Saltbæk
- Department of Endocrinology (Diabetes and Metabolism)Rigshospitalet, Copenhagen, Denmark
| | - Martin Friedrichsen
- Department of Endocrinology (Diabetes and Metabolism)Rigshospitalet, Copenhagen, Denmark Steno Diabetes CenterGentofte, Denmark
| | - Yan Chen
- Department of Endocrinology (Diabetes and Metabolism)Rigshospitalet, Copenhagen, Denmark
| | - Allan Vaag
- Department of Endocrinology (Diabetes and Metabolism)Rigshospitalet, Copenhagen, Denmark Steno Diabetes CenterGentofte, Denmark
| |
Collapse
|
21
|
Martin-Gronert MS, Fernandez-Twinn DS, Bushell M, Siddle K, Ozanne SE. Cell-autonomous programming of rat adipose tissue insulin signalling proteins by maternal nutrition. Diabetologia 2016; 59:1266-75. [PMID: 26965244 PMCID: PMC4861755 DOI: 10.1007/s00125-016-3905-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 02/03/2016] [Indexed: 01/10/2023]
Abstract
AIMS/HYPOTHESIS Individuals with a low birthweight have an increased risk of developing type 2 diabetes mellitus in adulthood. This is associated with peripheral insulin resistance. Here, we aimed to determine whether changes in insulin signalling proteins in white adipose tissue (WAT) can be detected prior to the onset of impaired glucose tolerance, determine whether these changes are cell-autonomous and identify the underlying mechanisms involved. METHODS Fourteen-month-old male rat offspring born to dams fed a standard protein (20%) diet or a low (8%) protein diet throughout gestation and lactation were studied. Fat distribution and adipocyte size were determined. Protein content and mRNA expression of key insulin signalling molecules were analysed in epididymal WAT and in pre-adipocytes that had undergone in vitro differentiation. RESULTS The offspring of low protein fed dams (LP offspring) had reduced visceral WAT mass, altered fat distribution and a higher percentage of small adipocytes in epididymal WAT. This was associated with reduced levels of IRS1, PI3K p110β, Akt1 and PKCζ proteins and of phospho-Akt Ser473. Corresponding mRNA transcript levels were unchanged. Similarly, in vitro differentiated adipocytes from LP offspring showed reduced protein levels of IRβ, IRS1, PI3K p85α and p110β subunits, and Akt1. Levels of Akt Ser473 and IRS1 Tyr612 phosphorylation were reduced, while IRS1 Ser307 phosphorylation was increased. CONCLUSIONS/INTERPRETATION Maternal protein restriction during gestation and lactation changes the distribution and morphology of WAT and reduces the levels of key insulin signalling proteins in the male offspring. This phenotype is retained in in vitro differentiated adipocytes, suggesting that programming occurs via cell-autonomous mechanism(s).
Collapse
Affiliation(s)
- Malgorzata S Martin-Gronert
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Box 289, 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 Hospital, Box 289, Cambridge, CB2 OQQ, UK
| | - Martin Bushell
- MRC Toxicology Unit, University of Leicester, Hodgkin Building, Leicester, UK
| | - Kenneth Siddle
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Box 289, 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 Hospital, Box 289, Cambridge, CB2 OQQ, UK
| |
Collapse
|
22
|
Zeeshan HMA, Lee GH, Kim HR, Chae HJ. Endoplasmic Reticulum Stress and Associated ROS. Int J Mol Sci 2016; 17:327. [PMID: 26950115 PMCID: PMC4813189 DOI: 10.3390/ijms17030327] [Citation(s) in RCA: 596] [Impact Index Per Article: 74.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 01/21/2016] [Accepted: 02/24/2016] [Indexed: 02/07/2023] Open
Abstract
The endoplasmic reticulum (ER) is a fascinating network of tubules through which secretory and transmembrane proteins enter unfolded and exit as either folded or misfolded proteins, after which they are directed either toward other organelles or to degradation, respectively. The ER redox environment dictates the fate of entering proteins, and the level of redox signaling mediators modulates the level of reactive oxygen species (ROS). Accumulating evidence suggests the interrelation of ER stress and ROS with redox signaling mediators such as protein disulfide isomerase (PDI)-endoplasmic reticulum oxidoreductin (ERO)-1, glutathione (GSH)/glutathione disuphide (GSSG), NADPH oxidase 4 (Nox4), NADPH-P450 reductase (NPR), and calcium. Here, we reviewed persistent ER stress and protein misfolding-initiated ROS cascades and their significant roles in the pathogenesis of multiple human disorders, including neurodegenerative diseases, diabetes mellitus, atherosclerosis, inflammation, ischemia, and kidney and liver diseases.
Collapse
Affiliation(s)
- Hafiz Maher Ali Zeeshan
- Department of Pharmacology and New Drug Development Institute, School of Medicine, Chonbuk National University, Jeonju, Chonbuk 561-180, Korea.
| | - Geum Hwa Lee
- Department of Pharmacology and New Drug Development Institute, School of Medicine, Chonbuk National University, Jeonju, Chonbuk 561-180, Korea.
| | - Hyung-Ryong Kim
- Department of Dental Pharmacology and Wonkwang Biomaterial Implant Research Institute, School of Dentistry, Wonkwang University, Iksan, Chonbuk 570-749, Korea.
| | - Han-Jung Chae
- Department of Pharmacology and New Drug Development Institute, School of Medicine, Chonbuk National University, Jeonju, Chonbuk 561-180, Korea.
| |
Collapse
|
23
|
Maternal periodontitis decreases plasma membrane GLUT4 content in skeletal muscle of adult offspring. Life Sci 2016; 148:194-200. [DOI: 10.1016/j.lfs.2016.02.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 02/04/2016] [Accepted: 02/04/2016] [Indexed: 02/07/2023]
|
24
|
Beauchamp B, Harper ME. In utero Undernutrition Programs Skeletal and Cardiac Muscle Metabolism. Front Physiol 2016; 6:401. [PMID: 26779032 PMCID: PMC4701911 DOI: 10.3389/fphys.2015.00401] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2015] [Accepted: 12/07/2015] [Indexed: 12/16/2022] Open
Abstract
In utero undernutrition is associated with increased risk for insulin resistance, obesity, and cardiovascular disease during adult life. A common phenotype associated with low birth weight is reduced skeletal muscle mass. Given the central role of skeletal muscle in whole body metabolism, alterations in its mass as well as its metabolic characteristics may contribute to disease risk. This review highlights the metabolic alterations in cardiac and skeletal muscle associated with in utero undernutrition and low birth weight. These tissues have high metabolic demands and are known to be sites of major metabolic dysfunction in obesity, type 2 diabetes, and cardiovascular disease. Recent research demonstrates that mitochondrial energetics are decreased in skeletal and cardiac muscles of adult offspring from undernourished mothers. These effects apparently lead to the development of a thrifty phenotype, which may represent overall a compensatory mechanism programmed in utero to handle times of limited nutrient availability. However, in an environment characterized by food abundance, the effects are maladaptive and increase adulthood risks of metabolic disease.
Collapse
Affiliation(s)
- Brittany Beauchamp
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa Ottawa, ON, Canada
| | - Mary-Ellen Harper
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa Ottawa, ON, Canada
| |
Collapse
|
25
|
The Link between Fetal Programming, Inflammation, Muscular Strength, and Blood Pressure. Mediators Inflamm 2015; 2015:710613. [PMID: 26491235 PMCID: PMC4600564 DOI: 10.1155/2015/710613] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 08/03/2015] [Indexed: 12/26/2022] Open
Abstract
Hypertension affects one billion individuals worldwide and is considered the leading cause of cardiovascular death, stroke, and myocardial infarction. This increase in the burden of hypertension and cardiovascular diseases (CVD) is principally driven by lifestyle changes such as increased hypercaloric diets and reduced physical activity producing an increase of obesity, insulin resistance, and low-grade inflammation. Visceral adipocytes are the principal source of proinflammatory cytokines and systemic inflammation participates in several steps in the development of CVD. However, maternal and infant malnutrition also persists as a major public health issue in low- to middle-income regions such as Latin America (LA). We propose that the increased rates of cardiovascular and metabolic diseases in these countries could be the result of the discrepancy between a restricted nutritional environment during fetal development and early life, and a nutritionally abundant environment during adulthood. Maternal undernutrition, which may manifest in lower birth weight offspring, appears to accentuate the relative risk of chronic disease at lower levels of adiposity. Therefore, LA populations may be more vulnerable to the pathogenic consequences of obesity than individuals with similar lifestyles in high-income countries, which may be mediated by higher levels of proinflammatory markers and lower levels of muscle mass and strength observed in low birth weight individuals.
Collapse
|
26
|
Mother's nutritional miRNA legacy: Nutrition during pregnancy and its possible implications to develop cardiometabolic disease in later life. Pharmacol Res 2015; 100:322-34. [PMID: 26325301 DOI: 10.1016/j.phrs.2015.08.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 08/18/2015] [Accepted: 08/19/2015] [Indexed: 01/02/2023]
Abstract
Maternal nutrition during pregnancy and lactation influences the offspring's health in the long-term. Indeed, human epidemiological studies and animal model experiments suggest that either an excess or a deficit in maternal nutrition influence offspring development and susceptibility to metabolic disorders. Different epigenetic mechanisms may explain in part the way by which dietary factors in early critical developmental steps might be able to affect the susceptibility to develop metabolic diseases in adulthood. microRNAs are versatile regulators of gene expression and play a major role during tissue homeostasis and disease. Dietary factors have also been shown to modify microRNA expression. However, the role of microRNAs in fetal programming remains largely unstudied. This review evaluates in vivo studies conducted to analyze the effect of maternal diet on the modulation of the microRNA expression in the offspring and their influence to develop metabolic and cardiovascular disease in later life. In overall, the available evidence suggests that nutritional status during pregnancy influence offspring susceptibility to the development of cardiometabolic risk factors, partly through microRNA action. Thus, therapeutic modulation of microRNAs can open up new strategies to combat - later in life - the effects of nutritional insult during critical points of development.
Collapse
|
27
|
Cohen DD. Neuromuscular performance deficits in low birthweight children: a target for physical activity interventions? Dev Med Child Neurol 2015; 57:406-7. [PMID: 25640481 DOI: 10.1111/dmcn.12682] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Daniel D Cohen
- Masira Institute, Faculty of Life Sciences, Universidad de Santander (UDES), Bucaramanga, Santander, Colombia
| |
Collapse
|
28
|
Dunlop K, Cedrone M, Staples JF, Regnault TRH. Altered fetal skeletal muscle nutrient metabolism following an adverse in utero environment and the modulation of later life insulin sensitivity. Nutrients 2015; 7:1202-16. [PMID: 25685986 PMCID: PMC4344584 DOI: 10.3390/nu7021202] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 02/02/2015] [Indexed: 02/07/2023] Open
Abstract
The importance of the in utero environment as a contributor to later life metabolic disease has been demonstrated in both human and animal studies. In this review, we consider how disruption of normal fetal growth may impact skeletal muscle metabolic development, ultimately leading to insulin resistance and decreased insulin sensitivity, a key precursor to later life metabolic disease. In cases of intrauterine growth restriction (IUGR) associated with hypoxia, where the fetus fails to reach its full growth potential, low birth weight (LBW) is often the outcome, and early in postnatal life, LBW individuals display modifications in the insulin-signaling pathway, a critical precursor to insulin resistance. In this review, we will present literature detailing the classical development of insulin resistance in IUGR, but also discuss how this impaired development, when challenged with a postnatal Western diet, may potentially contribute to the development of later life insulin resistance. Considering the important role of the skeletal muscle in insulin resistance pathogenesis, understanding the in utero programmed origins of skeletal muscle deficiencies in insulin sensitivity and how they may interact with an adverse postnatal environment, is an important step in highlighting potential therapeutic options for LBW offspring born of pregnancies characterized by placental insufficiency.
Collapse
Affiliation(s)
- Kristyn Dunlop
- Department of Physiology and Pharmacology, Western University, London, ON N6A-5C1, Canada.
| | - Megan Cedrone
- Department of Biology, Western University, London, ON N6A 5B7, Canada.
| | - James F Staples
- Department of Biology, Western University, London, ON N6A 5B7, Canada.
| | - Timothy R H Regnault
- Department of Physiology and Pharmacology, Western University, London, ON N6A-5C1, Canada.
- Department of Obstetrics and Gynecology, Western University, London, ON N6H-5W9, Canada.
- Lawson Health Research Institute, London, ON N6C-2R5, Canada.
- Children's Health Research Institute, London, ON N6C-2V5, Canada.
| |
Collapse
|
29
|
The developmental origins of sarcopenia: from epidemiological evidence to underlying mechanisms. J Dev Orig Health Dis 2014; 1:150-7. [PMID: 25141783 DOI: 10.1017/s2040174410000097] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Sarcopenia is defined as the loss of skeletal muscle mass and strength with age. There is increasing recognition of the serious health consequences in terms of disability, morbidity and mortality as well as major healthcare costs. Adult determinants of sarcopenia including age, gender, size, levels of physical activity and heritability have been well described. Nevertheless, there remains considerable unexplained variation in muscle mass and strength between older adults that may reflect not only the current rate of loss but the peak attained earlier in life. To date most epidemiological studies of sarcopenia have focused on factors modifying decline in later life; however, a life course approach to understanding sarcopenia, additionally, focuses on factors operating earlier in life including developmental influences. The epidemiological evidence linking low birth weight with lower muscle mass and strength is strong and consistent with replication in a number of different groups including children, young and older adults. However, most of the evidence for the cellular, hormonal, metabolic and molecular mechanisms underlying these associations comes from animal models. The next stage is to translate the understanding of mechanisms from animal muscle to human muscle enabling progress to be made not only in earlier identification of individuals at risk of sarcopenia but also in the development of beneficial interventions across the life course.
Collapse
|
30
|
Yates DT, Clarke DS, Macko AR, Anderson MJ, Shelton LA, Nearing M, Allen RE, Rhoads RP, Limesand SW. Myoblasts from intrauterine growth-restricted sheep fetuses exhibit intrinsic deficiencies in proliferation that contribute to smaller semitendinosus myofibres. J Physiol 2014; 592:3113-25. [PMID: 24860171 PMCID: PMC4214663 DOI: 10.1113/jphysiol.2014.272591] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 05/09/2014] [Indexed: 12/21/2022] Open
Abstract
Intrauterine growth restriction (IUGR) reduces skeletal muscle mass in fetuses and offspring. Our objective was to determine whether myoblast dysfunction due to intrinsic cellular deficiencies or serum factors reduces myofibre hypertrophy in IUGR fetal sheep. At 134 days, IUGR fetuses weighed 67% less (P < 0.05) than controls and had smaller (P < 0.05) carcasses and semitendinosus myofibre areas. IUGR semitendinosus muscles had similar percentages of pax7-positive nuclei and pax7 mRNA but lower (P < 0.05) percentages of myogenin-positive nuclei (7 ± 2% and 13 ± 2%), less myoD and myogenin mRNA, and fewer (P < 0.05) proliferating myoblasts (PNCA-positive-pax7-positive) than controls (44 ± 2% vs. 52 ± 1%). Primary myoblasts were isolated from hindlimb muscles, and after 3 days in growth media (20% fetal bovine serum, FBS), myoblasts from IUGR fetuses had 34% fewer (P < 0.05) myoD-positive cells than controls and replicated 20% less (P < 0.05) during a 2 h BrdU pulse. IUGR myoblasts also replicated less (P < 0.05) than controls during a BrdU pulse after 3 days in media containing 10% control or IUGR fetal sheep serum (FSS). Both myoblast types replicated less (P < 0.05) with IUGR FSS-supplemented media compared to control FSS-supplemented media. In differentiation-promoting media (2% FBS), IUGR and control myoblasts had similar percentages of myogenin-positive nuclei after 5 days and formed similar-sized myotubes after 7 days. We conclude that intrinsic cellular deficiencies in IUGR myoblasts and factors in IUGR serum diminish myoblast proliferation and myofibre size in IUGR fetuses, but intrinsic myoblast deficiencies do not affect differentiation. Furthermore, the persistent reduction in IUGR myoblast replication shows adaptive deficiencies that explain poor muscle growth in IUGR newborn offspring.
Collapse
Affiliation(s)
- Dustin T Yates
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ, USA
| | - Derek S Clarke
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ, USA
| | - Antoni R Macko
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ, USA
| | - Miranda J Anderson
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ, USA
| | - Leslie A Shelton
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ, USA
| | - Marie Nearing
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ, USA
| | - Ronald E Allen
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ, USA
| | - Robert P Rhoads
- Department of Animal and Poultry Sciences, Virginia Tech, Blacksburg, VA, USA
| | - Sean W Limesand
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, AZ, USA
| |
Collapse
|
31
|
Beauchamp B, Ghosh S, Dysart MW, Kanaan GN, Chu A, Blais A, Rajamanickam K, Tsai EC, Patti ME, Harper ME. Low birth weight is associated with adiposity, impaired skeletal muscle energetics and weight loss resistance in mice. Int J Obes (Lond) 2014; 39:702-11. [PMID: 25091727 PMCID: PMC4326251 DOI: 10.1038/ijo.2014.120] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 06/07/2014] [Accepted: 06/28/2014] [Indexed: 01/29/2023]
Abstract
BACKGROUND In utero undernutrition is associated with obesity and insulin resistance, although its effects on skeletal muscle remain poorly defined. Therefore, in the current study we explored the effects of in utero food restriction on muscle energy metabolism in mice. METHODS We used an experimental mouse model system of maternal undernutrition during late pregnancy to examine offspring from undernourished dams (U) and control offspring from ad libitum-fed dams (C). Weight loss of 10-week-old offspring on a 4-week 40% calorie-restricted diet was also followed. Experimental approaches included bioenergetic analyses in isolated mitochondria, intact (permeabilized) muscle and at the whole body level. RESULTS U have increased adiposity and decreased glucose tolerance compared to C. Strikingly, when U are put on a 40% calorie-restricted diet they lose half as much weight as calorie-restricted controls. Mitochondria from muscle overall from U had decreased coupled (state 3) and uncoupled (state 4) respiration and increased maximal respiration compared to C. Mitochondrial yield was lower in U than C. In permeabilized fiber preparations from mixed fiber-type muscle, U had decreased mitochondrial content and decreased adenylate-free leak respiration, fatty acid oxidative capacity and state 3 respiratory capacity through complex I. Fiber maximal oxidative phosphorylation capacity did not differ between U and C but was decreased with calorie restriction. CONCLUSIONS Our results reveal that in utero undernutrition alters metabolic physiology through a profound effect on skeletal muscle energetics and blunts response to a hypocaloric diet in adulthood. We propose that mitochondrial dysfunction links undernutrition in utero with metabolic disease in adulthood.
Collapse
Affiliation(s)
- B Beauchamp
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - S Ghosh
- Cardiovascular & Metabolic Disorders Program, Duke-NUS Graduate Medical School, Singapore, Singapore
| | - M W Dysart
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - G N Kanaan
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - A Chu
- 1] Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada [2] Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - A Blais
- 1] Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada [2] Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| | - K Rajamanickam
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - E C Tsai
- Division of Neurosurgery, Department of Surgery, Faculty of Medicine, Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
| | - M-E Patti
- Division of Integrative Physiology and Metabolism, Joslin Diabetes Center, Boston, MA, USA
| | - M-E Harper
- 1] Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada [2] Ottawa Institute of Systems Biology, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
32
|
Abstract
Establishing sufficient skeletal muscle mass is essential for lifelong metabolic health. The intrauterine environment is a major determinant of the muscle mass that is present during the life course of an individual, because muscle fiber number is set at the time of birth. Thus, a compromised intrauterine environment from maternal nutrient restriction or placental insufficiency that restricts muscle fiber number can have permanent effects on the amount of muscle an individual will live with. Reduced muscle mass due to fewer muscle fibers persists even after compensatory or 'catch-up' postnatal growth occurs. Furthermore, muscle hypertrophy can only partially compensate for this limitation in fiber number. Compelling associations link low birth weight and decreased muscle mass to future insulin resistance, which can drive the development of the metabolic syndrome and type 2 diabetes, and the risk of cardiovascular events later in life. There are gaps in knowledge about the origins of reduced muscle growth at the cellular level and how these patterns are set during fetal development. By understanding the nutrient and endocrine regulation of fetal skeletal muscle growth and development, we can direct research efforts toward improving muscle growth early in life to prevent the development of chronic metabolic diseases later in life.
Collapse
Affiliation(s)
- Laura D. Brown
- Department of Pediatrics (Neonatology), University of Colorado School of Medicine, Anschutz Medical Campus F441, Perinatal Research Center, 13243 East 23 Avenue, Aurora, CO 80045, Phone: 303-724-0106, Fax: 303-724-0898
| |
Collapse
|
33
|
Cohen DD, Gómez-Arbeláez D, Camacho PA, Pinzon S, Hormiga C, Trejos-Suarez J, Duperly J, Lopez-Jaramillo P. Low muscle strength is associated with metabolic risk factors in Colombian children: the ACFIES study. PLoS One 2014; 9:e93150. [PMID: 24714401 PMCID: PMC3979680 DOI: 10.1371/journal.pone.0093150] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2013] [Accepted: 02/28/2014] [Indexed: 01/23/2023] Open
Abstract
Purpose In youth, poor cardiorespiratory and muscular strength are associated with elevated metabolic risk factors. However, studies examining associations between strength and risk factors have been done exclusively in high income countries, and largely in Caucasian cohorts. The aim of this study was to assess these interactions in schoolchildren in Colombia, a middle income Latin American country. Methods We measured body mass index, body composition, handgrip strength (HG), cardiorespiratory fitness (CRF) and metabolic risk factors in 669 low-middle socioeconomic status Colombian schoolchildren (mean age 11.52±1.13, 47% female). Associations between HG, CRF and metabolic risk factors were evaluated. Results HG and CRF were inversely associated with blood pressure, HOMA index and a composite metabolic risk score (p<0.001 for all) and HG was also inversely associated with triglycerides and C-reactive protein (CRP) (both p<0.05). Associations between HG and risk factors were marginally weakened after adjusting for CRF, while associations between CRF and these factors were substantially weakened after adjusting for HG. Linear regression analyses showed inverse associations between HG and systolic BP (β = −0.101; p = 0.047), diastolic BP (β = −0.241; p> = 0.001), HOMA (β = −0.164; p = 0.005), triglycerides (β = −0.583; p = 0.026) and CRP (β = −0.183; p = 0.037) but not glucose (p = 0.698) or HDL cholesterol (p = 0.132). The odds ratios for having clustered risk in the weakest quartile compared with the strongest quartile were 3.0 (95% confidence interval: 1.81–4.95). Conclusions In Colombian schoolchildren both poorer handgrip strength/kg body mass and cardiorespiratory fitness were associated with a worse metabolic risk profile. Associations were stronger and more consistent between handgrip and risk factors than between cardiorespiratory fitness and these risk factors. Our findings indicate the addition of handgrip dynamometry to non-invasive youth health surveillance programs would improve the accuracy of the assessment of cardio-metabolic health.
Collapse
Affiliation(s)
- Daniel Dylan Cohen
- Dirección de Investigaciones, Fundación Oftalmológica de Santander (FOSCAL), Floridablanca, Santander, Colombia
- Facultad de la Ciencias de la Salud, Universidad de Santander (UDES), Bucaramanga, Santander, Colombia
| | - Diego Gómez-Arbeláez
- Dirección de Investigaciones, Fundación Oftalmológica de Santander (FOSCAL), Floridablanca, Santander, Colombia
- Facultad de la Ciencias de la Salud, Universidad de Santander (UDES), Bucaramanga, Santander, Colombia
| | - Paul Anthony Camacho
- Dirección de Investigaciones, Fundación Oftalmológica de Santander (FOSCAL), Floridablanca, Santander, Colombia
| | - Sandra Pinzon
- Facultad de la Ciencias de la Salud, Universidad de Santander (UDES), Bucaramanga, Santander, Colombia
| | - Claudia Hormiga
- Dirección de Investigaciones, Fundación Oftalmológica de Santander (FOSCAL), Floridablanca, Santander, Colombia
| | - Juanita Trejos-Suarez
- Facultad de la Ciencias de la Salud, Universidad de Santander (UDES), Bucaramanga, Santander, Colombia
| | - John Duperly
- Facultad de Medicina, Universidad de Los Andes, Bogota, Colombia
| | - Patricio Lopez-Jaramillo
- Dirección de Investigaciones, Fundación Oftalmológica de Santander (FOSCAL), Floridablanca, Santander, Colombia
- Facultad de la Ciencias de la Salud, Universidad de Santander (UDES), Bucaramanga, Santander, Colombia
- * E-mail:
| |
Collapse
|
34
|
Fernandez-Twinn DS, Alfaradhi MZ, Martin-Gronert MS, Duque-Guimaraes DE, Piekarz A, Ferland-McCollough D, Bushell M, Ozanne SE. Downregulation of IRS-1 in adipose tissue of offspring of obese mice is programmed cell-autonomously through post-transcriptional mechanisms. Mol Metab 2014; 3:325-33. [PMID: 24749062 PMCID: PMC3986586 DOI: 10.1016/j.molmet.2014.01.007] [Citation(s) in RCA: 87] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 01/07/2014] [Accepted: 01/13/2014] [Indexed: 01/28/2023] Open
Abstract
We determined the effects of maternal diet-induced obesity on offspring adipose tissue insulin signalling and miRNA expression in the aetiology of insulin resistance in later life. Although body composition and glucose tolerance of 8-week-old male offspring of obese dams were not dysregulated, serum insulin was significantly (p<0.05) elevated. Key insulin signalling proteins in adipose tissue were down-regulated, including the insulin receptor, catalytic (p110β) and regulatory (p85α) subunits of PI3K as well as AKT1 and 2 (all p<0.05). The largest reduction observed was in IRS-1 protein (p<0.001), which was regulated post-transcriptionally. Concurrently, miR-126, which targets IRS-1, was up-regulated (p<0.05). These two features were maintained in isolated primary pre-adipocytes and differentiated adipocytes in-vitro. We have therefore established that maternal diet-induced obesity programs adipose tissue insulin resistance. We hypothesise that maintenance of the phenotype in-vitro strongly suggests that this mechanism is cell autonomous and may drive insulin resistance in later life.
Collapse
Affiliation(s)
- Denise S Fernandez-Twinn
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, Level 4, Box 289, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Maria Z Alfaradhi
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, Level 4, Box 289, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Malgorzata S Martin-Gronert
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, Level 4, Box 289, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Daniella E Duque-Guimaraes
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, Level 4, Box 289, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Ana Piekarz
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, Level 4, Box 289, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - David Ferland-McCollough
- MRC Toxicology Unit, Hodgkin Building, PO Box 138, University of Leicester, Lancaster Road, Leicester LE1 9HN, UK
| | - Martin Bushell
- MRC Toxicology Unit, Hodgkin Building, PO Box 138, University of Leicester, Lancaster Road, Leicester LE1 9HN, UK
| | - Susan E Ozanne
- University of Cambridge Metabolic Research Laboratories and MRC Metabolic Diseases Unit, Institute of Metabolic Science, Level 4, Box 289, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| |
Collapse
|
35
|
Mortensen B, Friedrichsen M, Andersen NR, Alibegovic AC, Højbjerre L, Sonne MP, Stallknecht B, Dela F, Wojtaszewski JFP, Vaag A. Physical inactivity affects skeletal muscle insulin signaling in a birth weight-dependent manner. J Diabetes Complications 2014; 28:71-8. [PMID: 24120282 DOI: 10.1016/j.jdiacomp.2013.09.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Revised: 09/03/2013] [Accepted: 09/05/2013] [Indexed: 10/26/2022]
Abstract
AIMS We investigated whether physical inactivity could unmask defects in insulin and AMPK signaling in low birth weight (LBW) subjects. METHODS Twenty LBW and 20 normal birth weight (NBW) subjects were investigated using the euglycemic-hyperinsulinemic clamp with excision of skeletal muscle biopsies pre and post 9days of bed rest. Employing Western blotting, we investigated skeletal muscle Akt, AS160, GLUT4, and AMPK signaling. RESULTS Peripheral insulin action was similar in the two groups and was decreased to the same extent post bed rest. Insulin and AMPK signaling was unaffected by bed rest in NBW individuals. LBW subjects showed decreased insulin-stimulated Akt phosphorylation and increased AMPK α1 and γ3 protein expression post bed rest. Insulin response of AS160 phosphorylation was lower in LBW subjects both pre and post bed rest. CONCLUSIONS Bed rest-induced insulin resistance is not explained by impaired muscle insulin or AMPK signaling in subjects with or without LBW. Lower muscle insulin signaling in LBW subjects post bed rest despite similar degree of insulin resistance as seen in controls may to some extent support the idea that LBW subjects are at higher risk of developing type 2 diabetes when being physically inactive.
Collapse
Affiliation(s)
- Brynjulf Mortensen
- Steno Diabetes Center, Gentofte, Denmark; Molecular Physiology Group, The August Krogh Centre, Department of Nutrition, Exercise and Sports, University of Copenhagen.
| | - Martin Friedrichsen
- Steno Diabetes Center, Gentofte, Denmark; Molecular Physiology Group, The August Krogh Centre, Department of Nutrition, Exercise and Sports, University of Copenhagen
| | - Nicoline R Andersen
- Molecular Physiology Group, The August Krogh Centre, Department of Nutrition, Exercise and Sports, University of Copenhagen
| | | | - Lise Højbjerre
- Department of Biomedical Sciences, University of Copenhagen; Center for Healthy Ageing, University of Copenhagen
| | - Mette P Sonne
- Department of Biomedical Sciences, University of Copenhagen; Center for Healthy Ageing, University of Copenhagen
| | | | - Flemming Dela
- Department of Biomedical Sciences, University of Copenhagen; Center for Healthy Ageing, University of Copenhagen
| | - Jørgen F P Wojtaszewski
- Molecular Physiology Group, The August Krogh Centre, Department of Nutrition, Exercise and Sports, University of Copenhagen
| | - Allan Vaag
- Steno Diabetes Center, Gentofte, Denmark; Rigshospitalet, Department of Endocrinology, Denmark
| |
Collapse
|
36
|
da Silva Aragão R, Guzmán-Quevedo O, Pérez-García G, Toscano AE, Gois Leandro C, Manhães-de-Castro R, Bolaños-Jiménez F. Differential developmental programming by early protein restriction of rat skeletal muscle according to its fibre-type composition. Acta Physiol (Oxf) 2014; 210:70-83. [PMID: 23362831 DOI: 10.1111/apha.12073] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 11/13/2012] [Accepted: 01/25/2013] [Indexed: 12/29/2022]
Abstract
AIMS Differences in fibre-type composition of skeletal muscle have been associated with obesity and insulin resistance. As a poor nutrient environment early in life is a predisposing factor for the development of obesity and related metabolic diseases at adulthood, this study aimed at determining the long-term consequences of maternal undernutrition on the structural and metabolic properties of two skeletal muscles characterized by their different fibre-type composition and metabolic properties. METHODS The fibre-type composition and enzymatic activities of hexokinase (HK), beta-hydroxyacyl-CoA dehydrogenase (β-HAD) and citrate synthase (CS) were measured in soleus and extensor digitorum longus (EDL) muscles from adult rats born to dams fed a control (17% protein) or a low-protein [8% protein (PR)] diet throughout pregnancy and lactation. In addition, the expression levels of several genes regulating glycolysis, fatty acid oxidation and mitochondrial biogenesis were determined by real-time PCR. RESULTS Protein rats exhibited enhanced density of type II fibres along with decreased rate of fatty acid oxidation and glycolysis in soleus but not EDL. Malnourished rats exhibited also a different gene expression profile in soleus and EDL. Altogether, these alterations correspond to a state of energy deficiency and are present in animals which do not show yet any sign of obesity or glucose intolerance. CONCLUSION We conclude that maternal protein restriction alters in the long term the structural and enzymatic properties of offspring skeletal muscle in a fibre-type-dependent manner. These alterations might have a causative role in the development of obesity and related metabolic disorders later in life.
Collapse
Affiliation(s)
- R. da Silva Aragão
- INRA; UMR1280 Physiologie des Adaptations Nutritionnelles; Nantes France
- Université de Nantes; Nantes Atlantique Université; Nantes France
- Departamento de Nutrição; Centro de Ciências da Saúde; Universidade Federal de Pernambuco; Recife Brazil
| | - O. Guzmán-Quevedo
- INRA; UMR1280 Physiologie des Adaptations Nutritionnelles; Nantes France
- Université de Nantes; Nantes Atlantique Université; Nantes France
| | - G. Pérez-García
- INRA; UMR1280 Physiologie des Adaptations Nutritionnelles; Nantes France
- Université de Nantes; Nantes Atlantique Université; Nantes France
| | - A. E. Toscano
- Núcleo de Enfermagem; CAV; Universidade Federal de Pernambuco; Vitória de Santo Antão Brazil
| | - C. Gois Leandro
- Núcleo de Educação Física e Ciências do Esporte; CAV; Universidade Federal de Pernambuco; Vitória de Santo Antão Brazil
| | - R. Manhães-de-Castro
- Departamento de Nutrição; Centro de Ciências da Saúde; Universidade Federal de Pernambuco; Recife Brazil
| | - F. Bolaños-Jiménez
- INRA; UMR1280 Physiologie des Adaptations Nutritionnelles; Nantes France
- Université de Nantes; Nantes Atlantique Université; Nantes France
| |
Collapse
|
37
|
Nicholas LM, Morrison JL, Rattanatray L, Ozanne SE, Kleemann DO, Walker SK, MacLaughlin SM, Zhang S, Martin-Gronert MS, McMillen IC. Differential effects of exposure to maternal obesity or maternal weight loss during the periconceptional period in the sheep on insulin signalling molecules in skeletal muscle of the offspring at 4 months of age. PLoS One 2013; 8:e84594. [PMID: 24386400 PMCID: PMC3873457 DOI: 10.1371/journal.pone.0084594] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2013] [Accepted: 11/25/2013] [Indexed: 11/19/2022] Open
Abstract
Exposure to maternal obesity before and/or throughout pregnancy may increase the risk of obesity and insulin resistance in the offspring in childhood and adult life, therefore, resulting in its transmission into subsequent generations. We have previously shown that exposure to maternal obesity around the time of conception alone resulted in increased adiposity in female lambs. Changes in the abundance of insulin signalling molecules in skeletal muscle and adipose tissue precede the development of insulin resistance and type 2 diabetes. It is not clear, however, whether exposure to maternal obesity results in insulin resistance in her offspring as a consequence of the impact of increased adiposity on skeletal muscle or as a consequence of the programming of specific changes in the abundance of insulin signalling molecules in this tissue. We have used an embryo transfer model in the sheep to investigate the effects of exposure to either maternal obesity or to weight loss in normal and obese mothers preceding and for one week after conception on the expression and abundance of insulin signalling molecules in muscle in the offspring. We found that exposure to maternal obesity resulted in lower muscle GLUT-4 and Ser 9 phospho-GSK3α and higher muscle GSK3α abundance in lambs when compared to lambs conceived in normally nourished ewes. Exposure to maternal weight loss in normal or obese mothers, however, resulted in lower muscle IRS1, PI3K, p110β, aPKCζ, Thr 642 phospho-AS160 and GLUT-4 abundance in the offspring. In conclusion, maternal obesity or weight loss around conception have each programmed specific changes on subsets of molecules in the insulin signalling, glucose transport and glycogen synthesis pathways in offspring. There is a need for a stronger evidence base to ensure that weight loss regimes in obese women seeking to become pregnant minimize the metabolic costs for the next generation.
Collapse
Affiliation(s)
- Lisa M. Nicholas
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Janna L. Morrison
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Leewen Rattanatray
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
- Discipline of Physiology, School of Molecular and Life Sciences, University of Adelaide, Adelaide, South Australia, Australia
| | - Susan E. Ozanne
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Dave O. Kleemann
- Turretfield Research Centre, South Australian Research and Development Institute, Rosedale, South Australia, Australia
| | - Simon K. Walker
- Turretfield Research Centre, South Australian Research and Development Institute, Rosedale, South Australia, Australia
| | - Severence M. MacLaughlin
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Song Zhang
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
| | - Malgorzata S. Martin-Gronert
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Isabella C. McMillen
- Sansom Institute for Health Research, School of Pharmacy and Medical Sciences, University of South Australia, Adelaide, South Australia, Australia
- *
| |
Collapse
|
38
|
Hill DJ, Prapavessis H, Shoemaker JK, Jackman M, Mahmud FH, Clarson C. Relationship between Birth Weight and Metabolic Status in Obese Adolescents. ISRN OBESITY 2013; 2013:490923. [PMID: 24555145 PMCID: PMC3901987 DOI: 10.1155/2013/490923] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Accepted: 07/28/2013] [Indexed: 01/20/2023]
Abstract
Objective. To examine the relationships between birth weight and body mass index, percent body fat, blood lipids, glycemia, insulin resistance, adipokines, blood pressure, and endothelial function in a cohort of obese adolescents. Design and Methods. Ninety-five subjects aged 10-16 years (mean age 13.5 years) with a body mass index >95th centile (mean [±SEM] 33.0 ± 0.6) were utilized from two prospective studies for obesity prevention prior to any interventions. The mean term birth weight was 3527 ± 64 g (range 1899-4990 g;). Results. Body mass index z-score correlated positively with birth weight (r (2) = 0.05, P = 0.03), but not percent body fat. Insulin resistance negatively correlated with birth weight (r (2) = 0.05, P < 0.001), as did fasting plasma insulin (r (2) = 0.05, P < 0.001); both being significantly greater for subjects of small versus large birth weight (Δ Homeostasis Model Assessment = 2.5 and Δ insulin = 10 pmol/L for birth weight <2.5 kg versus >4.5 kg). Adiponectin, but not leptin, blood pressure z-scores or peripheral arterial tomography values positively correlated with birth weight (r (2) = 0.07, P = 0.008). Conclusions. Excess body mass index in obese adolescents was positively related to birth weight. Birth weight was not associated with cardiovascular risk factors but represented a significant determinant of insulin resistance.
Collapse
Affiliation(s)
- David J. Hill
- London Health Sciences Centre, Lawson Health Research Institute, London, ON, Canada N6A 5W9
- Lawson Health Research Institute, St Joseph's Health Care, 268 Grosvenor Street, London, ON, Canada N6A 4V2
- Department of Paediatrics, University of Western Ontario, London, ON, Canada N6A 5A5
- Department of Medicine, University of Western Ontario, London, ON, Canada N6A 5A5
- Department of Physiology and Pharmacology, University of Western Ontario, London, ON, Canada N6A 5A5
| | - Harry Prapavessis
- School of Kinesiology, University of Western Ontario, London, ON, Canada N6A 5A5
| | - J. Kevin Shoemaker
- School of Kinesiology, University of Western Ontario, London, ON, Canada N6A 5A5
| | - Michelle Jackman
- London Health Sciences Centre, Children's Hospital, London, ON, Canada N6A 5W9
- Department of Pediatrics , University of Calgary, Calgary, Alberta, Canada
| | - Farid H. Mahmud
- Department of Paediatrics, University of Toronto and Hospital for Sick Children, Toronto, ON, Canada M5G 1X8
| | - Cheril Clarson
- London Health Sciences Centre, Lawson Health Research Institute, London, ON, Canada N6A 5W9
- Department of Paediatrics, University of Western Ontario, London, ON, Canada N6A 5A5
- London Health Sciences Centre, Children's Hospital, London, ON, Canada N6A 5W9
| |
Collapse
|
39
|
Yoshida Y, Fuchita M, Kimura-Koyanagi M, Kanno A, Matsuda T, Asahara SI, Hashimoto N, Isagawa T, Ogawa W, Aburatani H, Noda T, Seino S, Kasuga M, Kido Y. Contribution of insulin signaling to the regulation of pancreatic beta-cell mass during the catch-up growth period in a low birth weight mouse model. Diabetol Int 2013. [DOI: 10.1007/s13340-013-0127-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
40
|
Epigenetic origins of metabolic disease: The impact of the maternal condition to the offspring epigenome and later health consequences. FOOD SCIENCE AND HUMAN WELLNESS 2013. [DOI: 10.1016/j.fshw.2013.03.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
41
|
Batista TM, Ribeiro RA, da Silva PMR, Camargo RL, Lollo PCB, Boschero AC, Carneiro EM. Taurine supplementation improves liver glucose control in normal protein and malnourished mice fed a high-fat diet. Mol Nutr Food Res 2012; 57:423-34. [DOI: 10.1002/mnfr.201200345] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2012] [Revised: 10/11/2012] [Accepted: 10/23/2012] [Indexed: 01/05/2023]
Affiliation(s)
- Thiago M. Batista
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia; Universidade Estadual de Campinas (UNICAMP); Campinas SP Brazil
| | - Rosane A. Ribeiro
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia; Universidade Estadual de Campinas (UNICAMP); Campinas SP Brazil
- Núcleo em Ecologia e Desenvolvimento Sócio-Ambiental de Macaé (NUPEM); Universidade Federal do Rio de Janeiro (UFRJ); Macaé RJ Brazil
| | - Priscilla M. R. da Silva
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia; Universidade Estadual de Campinas (UNICAMP); Campinas SP Brazil
| | - Rafael L. Camargo
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia; Universidade Estadual de Campinas (UNICAMP); Campinas SP Brazil
| | - Pablo C. B. Lollo
- Departamento de Alimentos e Nutrição, Faculdade de Engenharia de Alimentos; Universidade Estadual de Campinas (UNICAMP); Campinas SP Brazil
| | - Antonio C. Boschero
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia; Universidade Estadual de Campinas (UNICAMP); Campinas SP Brazil
| | - Everardo M. Carneiro
- Departamento de Biologia Estrutural e Funcional, Instituto de Biologia; Universidade Estadual de Campinas (UNICAMP); Campinas SP Brazil
| |
Collapse
|
42
|
Carter LG, Lewis KN, Wilkerson DC, Tobia CM, Ngo Tenlep SY, Shridas P, Garcia-Cazarin ML, Wolff G, Andrade FH, Charnigo RJ, Esser KA, Egan JM, de Cabo R, Pearson KJ. Perinatal exercise improves glucose homeostasis in adult offspring. Am J Physiol Endocrinol Metab 2012; 303:E1061-8. [PMID: 22932781 PMCID: PMC3469606 DOI: 10.1152/ajpendo.00213.2012] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Emerging research has shown that subtle factors during pregnancy and gestation can influence long-term health in offspring. In an attempt to be proactive, we set out to explore whether a nonpharmacological intervention, perinatal exercise, might improve offspring health. Female mice were separated into sedentary or exercise cohorts, with the exercise cohort having voluntary access to a running wheel prior to mating and during pregnancy and nursing. Offspring were weaned, and analyses were performed on the mature offspring that did not have access to running wheels during any portion of their lives. Perinatal exercise caused improved glucose disposal following an oral glucose challenge in both female and male adult offspring (P < 0.05 for both). Blood glucose concentrations were reduced to lower values in response to an intraperitoneal insulin tolerance test for both female and male adult offspring of parents with access to running wheels (P < 0.05 and P < 0.01, respectively). Male offspring from exercised dams showed increased percent lean mass and decreased fat mass percent compared with male offspring from sedentary dams (P < 0.01 for both), but these parameters were unchanged in female offspring. These data suggest that short-term maternal voluntary exercise prior to and during healthy pregnancy and nursing can enhance long-term glucose homeostasis in offspring.
Collapse
Affiliation(s)
- Lindsay G Carter
- Graduate Center for Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, Kentucky 40536-0200, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Type 2 diabetes (T2D), also known as non-insulin dependent diabetes mellitus, arises as a consequence of peripheral insulin resistance in combination with an inability of pancreatic islet β-cells to secrete adequate amounts of insulin. It is widely recognized that the current environment (e.g. an unhealthy diet and sedentary lifestyle) contributes to this process. In recent years, however, the role of the early environment, particularly nutrition, has emerged as an important factor capable of influencing health and disease risk of an individual, including risk of T2D. The impact of early environment on glucose metabolism has been extensively studied. Compelling evidence from epidemiological studies and animal models suggests that early nutrition can affect insulin action as a mediator of glucose homeostasis in peripheral tissues and as an important regulator of appetite and body weight. The early environment can also affect β-cell mass and function, and hence insulin secretion. The molecular mechanisms underlying the relationship between a suboptimal early environment and impaired insulin action and secretion is thought to include epigenetic modifications of the foetal genome, oxidative stress and mitochondrial dysfunction.
Collapse
Affiliation(s)
- M S Martin-Gronert
- University of Cambridge Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | | |
Collapse
|
44
|
Brown LD, Rozance PJ, Thorn SR, Friedman JE, Hay WW. Acute supplementation of amino acids increases net protein accretion in IUGR fetal sheep. Am J Physiol Endocrinol Metab 2012; 303:E352-64. [PMID: 22649066 PMCID: PMC3423121 DOI: 10.1152/ajpendo.00059.2012] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Placental insufficiency decreases fetal amino acid uptake from the placenta, plasma insulin concentrations, and protein accretion, thus compromising normal fetal growth trajectory. We tested whether acute supplementation of amino acids or insulin into the fetus with intrauterine growth restriction (IUGR) would increase net fetal protein accretion rates. Late-gestation IUGR and control (CON) fetal sheep received acute, 3-h infusions of amino acids (with euinsulinemia), insulin (with euglycemia and euaminoacidemia), or saline. Fetal leucine metabolism was measured under steady-state conditions followed by a fetal muscle biopsy to quantify insulin signaling. In CON, increasing amino acid delivery rates to the fetus by 100% increased leucine oxidation rates by 100%. In IUGR, amino acid infusion completely suppressed fetal protein breakdown rates but increased leucine oxidation rate by only 25%, resulting in increased protein accretion rates by 150%. Acute insulin infusion, however, had very little effect on amino acid delivery rates, fetal leucine disposal rates, or fetal protein accretion rates in CON or IUGR fetuses despite robust signaling of the fetal skeletal muscle insulin-signaling cascade. These results indicate that, when amino acids are given directly into the fetal circulation independently of changes in insulin concentrations, IUGR fetal sheep have suppressed protein breakdown rates, thus increasing net fetal protein accretion.
Collapse
Affiliation(s)
- Laura D Brown
- Perinatal Research Center, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, USA.
| | | | | | | | | |
Collapse
|
45
|
Perälä MM, Eriksson JG. Early growth and postprandial glucose, insulin, lipid and inflammatory responses in adulthood. Curr Opin Lipidol 2012; 23:327-33. [PMID: 22617752 DOI: 10.1097/mol.0b013e3283541da6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW Epidemiological findings suggest that prenatal and postnatal growth is associated with later health outcomes including cardiovascular disease and type 2 diabetes. It has been suggested that these associations are mediated through classical risk factors, for example dyslipidemia. Despite extensive epidemiological investigations, only limited data are available on the long-term influences of early growth on postprandial responses, although postprandial levels of many risk factors have been proposed to be more important than fasting levels in disease process. This review focuses on recent studies evaluating the effect of early growth on postprandial responses. RECENT FINDINGS Current evidence from postprandial studies shows that individuals who were small at birth or grew slowly during infancy have elevated postprandial insulin and triglyceride responses. However, early growth does not seem to affect postprandial inflammatory markers. It is likely that both liver programming and abnormalities in insulin-sensitive tissues play key roles in explaining these elevated responses. SUMMARY Recent studies suggest that slow growth during early life has an adverse effect on postprandial metabolism, and predicts higher insulin and triglyceride responses. These elevated postprandial responses might be one underlying mechanism explaining the increased risk of cardiovascular disease and type 2 diabetes associated with nonoptimal early growth.
Collapse
Affiliation(s)
- Mia-Maria Perälä
- Department of Chronic Disease Prevention, National Institute for Health and Welfare, University of Helsinki, Helsinki University Central Hospital, Unit of General Practice, Vaasa, Finland.
| | | |
Collapse
|
46
|
De Blasio MJ, Gatford KL, Harland ML, Robinson JS, Owens JA. Placental restriction reduces insulin sensitivity and expression of insulin signaling and glucose transporter genes in skeletal muscle, but not liver, in young sheep. Endocrinology 2012; 153:2142-51. [PMID: 22434080 DOI: 10.1210/en.2011-1955] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Poor growth before birth is associated with impaired insulin sensitivity later in life, increasing the risk of type 2 diabetes. The tissue sites at which insulin resistance first develops after intrauterine growth restriction (IUGR), and its molecular basis, are unclear. We have therefore characterized the effects of placental restriction (PR), a major cause of IUGR, on whole-body insulin sensitivity and expression of molecular determinants of insulin signaling and glucose uptake in skeletal muscle and liver of young lambs. Whole-body insulin sensitivity was measured at 30 d by hyperinsulinaemic euglycaemic clamp and expression of insulin signaling genes (receptors, pathways, and targets) at 43 d in muscle and liver of control (n = 15) and PR (n = 13) lambs. PR reduced size at birth and increased postnatal growth, fasting plasma glucose (+15%, P = 0.004), and insulin (+115%, P = 0.009). PR reduced whole-body insulin sensitivity (-43%, P < 0.001) and skeletal muscle expression of INSR (-36%), IRS1 (-28%), AKT2 (-44%), GLUT4 (-88%), GSK3α (-35%), and GYS1 (-31%) overall (each P < 0.05) and decreased AMPKγ3 expression in females (P = 0.030). PR did not alter hepatic expression of insulin signaling and related genes but increased GLUT2 expression (P = 0.047) in males. Whole-body insulin sensitivity correlated positively with skeletal muscle expression of IRS1, AKT2, HK, AMPKγ2, and AMPKγ3 in PR lambs only (each P < 0.05) but not with hepatic gene expression in control or PR lambs. Onset of insulin resistance after PR and IUGR is accompanied by, and can be accounted for by, reduced expression of insulin signaling and metabolic genes in skeletal muscle but not liver.
Collapse
Affiliation(s)
- Miles J De Blasio
- The Robinson Institute and School of Pediatrics and Reproductive Health, University of Adelaide, Adelaide, South Australia 5005, Australia
| | | | | | | | | |
Collapse
|
47
|
Dufour S, Petersen KF. Disassociation of liver and muscle insulin resistance from ectopic lipid accumulation in low-birth-weight individuals. J Clin Endocrinol Metab 2011; 96:3873-80. [PMID: 21994962 PMCID: PMC3232622 DOI: 10.1210/jc.2011-1747] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
CONTEXT Low birth weight (LBW) is a marker of fetal stress and is associated with an increased prevalence of type 2 diabetes (T2D). Insulin resistance plays a prominent role in the development of T2D; however, the pathogenesis of T2D in LBW is controversial. OBJECTIVE The objective of the study was to assess whole-body and tissue-specific insulin sensitivity and intramyocellular lipid (IMCL) and hepatic lipid content in LBW and matched control subjects. DESIGN These were prospective and pair-matched studies. SETTING The study was conducted at Yale University Center for Clinical Investigation. PARTICIPANTS Young, lean, nonsmoking, sedentary LBW (n = 45) and matched control subjects participated in the study. INTERVENTION Interventions included an oral glucose tolerance test and hyperinsulinemic-euglycemic clamps and (1)H magnetic resonance spectroscopy. MAIN OUTCOMES MEASURES The main outcomes measures included insulin sensitivity index; whole-body and tissue-specific insulin sensitivity; liver lipid and IMCL contents; and fasting concentrations of cortisol, GH, and IGF-I as markers of the hypothalamic-pituitary-adrenal and IGFI/GH axes. RESULTS The LBW subjects were insulin resistant as reflected by a 20% reduction in insulin sensitivity index as compared with the controls (P = 0.0017), which could be attributed to both liver and muscle insulin resistance. There were no differences in IMCL or hepatic triglyceride content between LBW and control groups. In the LBW group, fasting plasma concentrations of cortisol (P = 0.01) and GH (P = 0.01) were increased, and IGF-I concentrations reduced (P < 0.05) a pattern, which may suggest potential dysregulation of the hypothalamic-pituitary-adrenal and IGF-I/GH axes. CONCLUSION These results support the hypothesis that fetal stress and LBW lead to liver and muscle insulin resistance and show that this is independent of lipid deposition in these organs.
Collapse
Affiliation(s)
- Sylvie Dufour
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, Connecticut 06520-8020, USA
| | | |
Collapse
|
48
|
Højbjerre L, Alibegovic AC, Sonne MP, Dela F, Vaag A, Bruun JM, Stallknecht B. Increased lipolysis but diminished gene expression of lipases in subcutaneous adipose tissue of healthy young males with intrauterine growth retardation. J Appl Physiol (1985) 2011; 111:1863-70. [PMID: 21903886 DOI: 10.1152/japplphysiol.00960.2011] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Intrauterine growth retardation (IUGR) is associated with a central fat distribution and risk of developing type 2 diabetes in adults when exposed to a sedentary Western lifestyle. Increased lipolysis is an early defect of metabolism in IUGR subjects, but the sites and molecular mechanisms involved are unknown. Twenty IUGR and 20 control (CON) subjects, aged 20-30 years, were studied before and after 10 days of bed rest using the glucose clamp technique combined with measurements of in vivo metabolism by microdialysis technique and blood flow by (133)Xe washout technique in subcutaneous abdominal (SCAAT) and femoral (SCFAT) adipose tissue. Additionally, mRNA expression of lipases was evaluated in biopsies from SCAAT. Lipolysis in SCAAT was substantially higher in IUGR than in CON subjects despite markedly lower mRNA expression of lipases. Blood flow was higher in IUGR compared with CON in both SCAAT and SCFAT. Whole body insulin sensitivity did not differ between groups and decreased after bed rest. After bed rest, SCAAT lipolysis remained higher in IUGR compared with CON, and SCFAT lipolysis decreased in CON but not in IUGR. Prior to the development of whole body insulin resistance, young men with IUGR are characterized by increased in vivo adipose tissue lipolysis and blood flow with a paradoxically decreased expression of lipases compared with CON, and 10 days of physical inactivity underlined the baseline findings. Subjects with IUGR exhibit primary defects in adipose tissue metabolism.
Collapse
Affiliation(s)
- Lise Højbjerre
- Department of Biomedical Sciences, Faculty of Health Sciences, University of Copenhagen, Denmark
| | | | | | | | | | | | | |
Collapse
|
49
|
Perälä MM, Valsta LM, Kajantie E, Leiviskä J, Eriksson JG. Impact of early growth on postprandial responses in later life. PLoS One 2011; 6:e24070. [PMID: 21904606 PMCID: PMC3164134 DOI: 10.1371/journal.pone.0024070] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2011] [Accepted: 07/29/2011] [Indexed: 12/15/2022] Open
Abstract
Background Low birth weight and slow growth during infancy are associated with increased rates of chronic diseases in adulthood. Associations with risk factors such as fasting glucose and lipids concentrations are weaker than expected based on associations with disease. This could be explained by differences in postprandial responses, which, however, have been little studied. Our aim was to examine the impact of growth during infancy on postprandial responses to a fast-food meal (FF-meal) and a meal, which followed the macro-nutrient composition of the dietary guidelines (REC-meal). Methodology/Principal Findings We recruited 24 overweight 65–75 year-old subjects, 12 with slow growth during infancy (SGI-group) and 12 with normal early growth. All the subjects were born at term. The study meals were isocaloric and both meals were consumed once. Plasma glucose, insulin, triglycerides (TG) and free fatty acids (FFA) were measured in fasting state and over a 4-h period after both meals. Subjects who grew slowly during infancy were also smaller at birth. Fasting glucose, insulin or lipid concentrations did not differ significantly between the groups. The TG responses were higher for the SGI-group both during the FF-meal (P = 0.047) and the REC-meal (P = 0.058). The insulin responses were significantly higher for the SGI-group after the FF-meal (P = 0.036). Glucose and FFA responses did not differ significantly between the groups. Conclusions Small birth size and slow early growth predict postprandial TG and insulin responses. Elevated responses might be one explanation why subjects who were small at birth and experiencing slow growth in infancy are at an increased risk of developing cardiovascular diseases in later life.
Collapse
Affiliation(s)
- Mia-Maria Perälä
- Department of Chronic Disease Prevention, The National Institute for Health and Welfare, Helsinki, Finland.
| | | | | | | | | |
Collapse
|
50
|
Sferruzzi-Perri AN, Vaughan OR, Coan PM, Suciu MC, Darbyshire R, Constancia M, Burton GJ, Fowden AL. Placental-specific Igf2 deficiency alters developmental adaptations to undernutrition in mice. Endocrinology 2011; 152:3202-12. [PMID: 21673101 DOI: 10.1210/en.2011-0240] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The pattern of fetal growth is a major determinant of the subsequent health of the infant. We recently showed in undernourished (UN) mice that fetal growth is maintained until late pregnancy, despite reduced placental weight, through adaptive up-regulation of placental nutrient transfer. Here, we determine the role of the placental-specific transcript of IGF-II (Igf2P0), a major regulator of placental transport capacity in mice, in adapting placental phenotype to UN. We compared the morphological and functional responses of the wild-type (WT) and Igf2P0-deficient placenta in WT mice fed ad libitium or 80% of the ad libitium intake. We observed that deletion of Igf2P0 prevented up-regulation of amino acid transfer normally seen in UN WT placenta. This was associated with a reduction in the proportion of the placenta dedicated to nutrient transport, the labyrinthine zone, and its constituent volume of trophoblast in Igf2P0-deficient placentas exposed to UN on d 16 of pregnancy. Additionally, Igf2P0-deficient placentas failed to up-regulate their expression of the amino acid transporter gene, Slc38a2, and down-regulate phosphoinositide 3-kinase-protein kinase B signaling in response to nutrient restriction on d 19. Furthermore, deleting Igf2P0 altered maternal concentrations of hormones (insulin and corticosterone) and metabolites (glucose) in both nutritional states. Therefore, Igf2P0 plays important roles in adapting placental nutrient transfer capacity during UN, via actions directly on the placenta and/or indirectly through the mother.
Collapse
Affiliation(s)
- A N Sferruzzi-Perri
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Downing Street, Cambridge CB2 3EG, United Kingdom.
| | | | | | | | | | | | | | | |
Collapse
|