1
|
Nogueira Silva Lima MT, Delayre-Orthez C, Howsam M, Jacolot P, Niquet-Léridon C, Okwieka A, Anton PM, Perot M, Barbezier N, Mathieu H, Ghinet A, Fradin C, Boulanger E, Jaisson S, Gillery P, Tessier FJ. Early- and life-long intake of dietary advanced glycation end-products (dAGEs) leads to transient tissue accumulation, increased gut sensitivity to inflammation, and slight changes in gut microbial diversity, without causing overt disease. Food Res Int 2024; 195:114967. [PMID: 39277266 DOI: 10.1016/j.foodres.2024.114967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/13/2024] [Accepted: 08/20/2024] [Indexed: 09/17/2024]
Abstract
Dietary advanced glycation end-products (dAGEs) accumulate in organs and are thought to initiate chronic low-grade inflammation (CLGI), induce glycoxidative stress, drive immunosenescence, and influence gut microbiota. Part of the toxicological interest in glycation products such as dietary carboxymethyl-lysine (dCML) relies on their interaction with receptor for advanced glycation end-products (RAGE). It remains uncertain whether early or lifelong exposure to dAGEs contributes physiological changes and whether such effects are reversible or permanent. Our objective was to examine the physiological changes in Wild-Type (WT) and RAGE KO mice that were fed either a standard diet (STD - 20.8 ± 5.1 µg dCML/g) or a diet enriched with dCML (255.2 ± 44.5 µg dCML/g) from the perinatal period for up to 70 weeks. Additionally, an early age (6 weeks) diet switch (dCML→STD) was explored to determine whether potential harmful effects of dCML could be reversed. Previous dCML accumulation patterns described by our group were confirmed here, with significant RAGE-independent accumulation of dCML in kidneys, ileum and colon over the 70-week dietary intervention (respectively 3-fold, 17-fold and 20-fold increases compared with controls). Diet switching returned tissue dCML concentrations to their baseline levels. The dCML-enriched diet had no significative effect on endogenous glycation, inflammation, oxidative stress or senescence parameters. The relative expression of TNFα, VCAM1, IL6, and P16 genes were all upregulated (∼2-fold) in an age-dependent manner, most notably in the kidneys of WT animals. RAGE knockout seemed protective in this regard, diminishing age-related renal expression of TNFα. Significant increases in TNFα expression were detectable in the intestinal tract of the Switch group (∼2-fold), suggesting a higher sensitivity to inflammation perhaps related to the timing of the diet change. Minor fluctuations were observed at family level within the caecal microbiota, including Eggerthellaceae, Anaerovoracaceae and Marinifilaceae communities, indicating slight changes in composition. Despite chronic dCML consumption resulting in higher free CML levels in tissues, there were no substantial increases in parameters related to inflammageing. Age was a more important factor in inflammation status, notably in the kidneys, while the early-life dietary switch may have influenced intestinal susceptibility to inflammation. This study affirms the therapeutic potential of RAGE modulation and corroborates evidence for the disruptive effect of dietary changes occurring too early in life. Future research should prioritize the potential influence of dAGEs on disease aetiology and development, notably any exacerbating effects they may have upon existing health conditions.
Collapse
Affiliation(s)
- M T Nogueira Silva Lima
- U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Institut Pasteur de Lille, University Lille, Inserm, CHU Lille, F-59000 Lille, France
| | - C Delayre-Orthez
- Institut Polytechnique UniLaSalle, Université d'Artois, ULR 7519, Equipe PETALES, 60000 Beauvais, France
| | - M Howsam
- U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Institut Pasteur de Lille, University Lille, Inserm, CHU Lille, F-59000 Lille, France
| | - P Jacolot
- Institut Polytechnique UniLaSalle, Université d'Artois, ULR 7519, Equipe PETALES, 60000 Beauvais, France
| | - C Niquet-Léridon
- Institut Polytechnique UniLaSalle, Université d'Artois, ULR 7519, Equipe PETALES, 60000 Beauvais, France
| | - A Okwieka
- University of Reims Champagne-Ardenne, Laboratory of Biochemistry and Molecular Biology, CNRS/URCA UMR 7369 MEDyC, Faculté de Médecine, 51095 Reims, France
| | - P M Anton
- Institut Polytechnique UniLaSalle, Université d'Artois, ULR 7519, Equipe PETALES, 60000 Beauvais, France
| | - M Perot
- Institut Polytechnique UniLaSalle, Université d'Artois, ULR 7519, Equipe PETALES, 60000 Beauvais, France
| | - N Barbezier
- Institut Polytechnique UniLaSalle, Université d'Artois, ULR 7519, Equipe PETALES, 60000 Beauvais, France
| | - H Mathieu
- Institut Polytechnique UniLaSalle, Université d'Artois, ULR 7519, Equipe PETALES, 60000 Beauvais, France
| | - A Ghinet
- U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Institut Pasteur de Lille, University Lille, Inserm, CHU Lille, F-59000 Lille, France; Junia, Health and Environment, Laboratory of Sustainable Chemistry and Health, 59000 Lille, France
| | - C Fradin
- U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Institut Pasteur de Lille, University Lille, Inserm, CHU Lille, F-59000 Lille, France
| | - E Boulanger
- U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Institut Pasteur de Lille, University Lille, Inserm, CHU Lille, F-59000 Lille, France
| | - S Jaisson
- University of Reims Champagne-Ardenne, Laboratory of Biochemistry and Molecular Biology, CNRS/URCA UMR 7369 MEDyC, Faculté de Médecine, 51095 Reims, France; University Hospital of Reims, Laboratory of Biochemistry-Pharmacology-Toxicology, 51092 Reims, France
| | - P Gillery
- University of Reims Champagne-Ardenne, Laboratory of Biochemistry and Molecular Biology, CNRS/URCA UMR 7369 MEDyC, Faculté de Médecine, 51095 Reims, France; University Hospital of Reims, Laboratory of Biochemistry-Pharmacology-Toxicology, 51092 Reims, France
| | - F J Tessier
- U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Institut Pasteur de Lille, University Lille, Inserm, CHU Lille, F-59000 Lille, France.
| |
Collapse
|
2
|
Mendoza-Romero HN, Biddinger JE, Bedenbaugh MN, Simerly RB. Microglia are Required for Developmental Specification of AgRP Innervation in the Hypothalamus of Offspring Exposed to Maternal High Fat Diet During Lactation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.12.607566. [PMID: 39185162 PMCID: PMC11343114 DOI: 10.1101/2024.08.12.607566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Nutritional fluctuations that occur early in life dictate metabolic adaptations that will affect susceptibility to weight gain and obesity later in life. The postnatal period in mice represents a time of dynamic changes in hypothalamic development and maternal consumption of a high fat diet during the lactation period (MHFD) changes the composition of milk and leads to enhanced susceptibility to obesity in offspring. Agouti-related peptide (AgRP) neurons in the arcuate nucleus of the hypothalamus (ARH) react to changes in multiple metabolic signals and distribute neuroendocrine information to other brain regions, such as the paraventricular hypothalamic nucleus (PVH), which is known to integrate a variety of signals that regulate body weight. Development of neural projections from AgRP neurons to the PVH occurs during the lactation period and these projections are reduced in MHFD offspring, but underlying developmental mechanisms remain largely unknown. Microglia are the resident immune cells of the central nervous system and are involved in refinement of neural connections and modulation of synaptic transmission. Because high fat diet exposure causes activation of microglia in adults, a similar activation may occur in offspring exposed to MHFD and play a role in sculpting hypothalamic feeding circuitry. Genetically targeted axonal labeling and immunohistochemistry were used to visualize AgRP axons and microglia in postnatal mice derived from MHFD dams and morphological changes quantified. The results demonstrate regionally localized changes to microglial morphology in the PVH of MHFD offspring that suggest enhanced surveillance activity and are temporally restricted to the period when AgRP neurons innervate the PVH. In addition, axon labeling experiments confirm a significant decrease in AgRP innervation of the PVH in MHFD offspring and provide direct evidence of synaptic pruning of AgRP inputs to the PVH. Microglial depletion with the Colony-stimulating factor 1 receptor inhibitor PLX5622 determined that the decrease in AgRP innervation observed in MHFD offspring is dependent on microglia, and that microglia are required for weight gain that emerges as early as weaning in offspring of MHFD dams. However, these changes do not appear to be dependent on the degree of microglial mediated synaptic pruning. Together, these findings suggest that microglia are activated by exposure to MHFD and interact directly with AgRP axons during development to permanently alter their density, with implications for developmental programming of metabolic phenotype.
Collapse
Affiliation(s)
| | - Jessica E. Biddinger
- Dept of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Michelle N. Bedenbaugh
- Dept of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Richard B. Simerly
- Dept of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| |
Collapse
|
3
|
Kyono Y, Nishiyama M, Kawamura A, Oikawa S, Tokumoto S, Yamaguchi H, Tomioka K, Nozu K, Mishina H, Nagase H. Association of perinatal factors with neurodevelopmental referrals in a population-based cohort study in Japan. Sci Rep 2024; 14:3492. [PMID: 38347064 PMCID: PMC10861581 DOI: 10.1038/s41598-024-54167-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Accepted: 02/09/2024] [Indexed: 02/15/2024] Open
Abstract
Although the causes of neurodevelopmental disorders remain unknown, several environmental risk factors have attracted considerable attention. We conducted a retrospective, longitudinal, population-based cohort study using data from infant health examinations of children born to mothers with pregnancies between April 1, 2014 and March 31, 2016 in Kobe City to identify the perinatal factors associated with neurodevelopmental referrals in 3-year-old children. There were 15,223 and 1283 children in the normal and referral groups, respectively. Neurodevelopmental referrals at the health checkup for 3-year-old children were significantly associated with the lack of social support during pregnancy (adjusted odds ratio [aOR] 1.99, 99% CI 1.14-3.45, p = 0.001), history of psychiatric consultation (aOR 1.56, 99% CI 1.10-2.22, p = 0.001), no social assistance post-delivery (aOR 1.49, 99% CI 1.03-2.16, p = 0.006), Edinburgh Post-natal Depression Scale (EPDS) score ≥ 9 (aOR 1.36, 99% CI 1.01-1.84, p = 0.008), infant gender (male) (aOR 2.51, 99% CI 2.05-3.06, p < 0.001), and cesarean delivery (aOR 1.39, 99% CI 1.11-1.75, p < 0.001). In conclusion, this exploratory study in the general Japanese population identified six perinatal factors associated with neurodevelopmental referrals in 3-year-old children: infant gender (male), cesarean section, maternal history of psychiatric consultation, EPDS score ≥ 9, lack of social support during pregnancy, and no social assistance post-delivery.
Collapse
Affiliation(s)
- Yuki Kyono
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Masahiro Nishiyama
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan.
- Department of Neurology, Hyogo Prefectural Kobe Children's Hospital, Kobe, Japan.
| | - Aoi Kawamura
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Shizuka Oikawa
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Shoichi Tokumoto
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Hiroshi Yamaguchi
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Kazumi Tomioka
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | - Kandai Nozu
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| | | | - Hiroaki Nagase
- Department of Pediatrics, Kobe University Graduate School of Medicine, 7-5-2, Kusunoki-Cho, Chuo-Ku, Kobe, Hyogo, 650-0017, Japan
| |
Collapse
|
4
|
Adibi JJ, Zhao Y, Koistinen H, Mitchell RT, Barrett ES, Miller R, O'Connor TG, Xun X, Liang HW, Birru R, Smith M, Moog NK. Molecular pathways in placental-fetal development and disruption. Mol Cell Endocrinol 2024; 581:112075. [PMID: 37852527 PMCID: PMC10958409 DOI: 10.1016/j.mce.2023.112075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 09/11/2023] [Accepted: 09/24/2023] [Indexed: 10/20/2023]
Abstract
The first trimester of pregnancy ranks high in priority when minimizing harmful exposures, given the wide-ranging types of organogenesis occurring between 4- and 12-weeks' gestation. One way to quantify potential harm to the fetus in the first trimester is to measure a corollary effect on the placenta. Placental biomarkers are widely present in maternal circulation, cord blood, and placental tissue biopsied at birth or at the time of pregnancy termination. Here we evaluate ten diverse pathways involving molecules expressed in the first trimester human placenta based on their relevance to normal fetal development and to the hypothesis of placental-fetal endocrine disruption (perturbation in development that results in abnormal endocrine function in the offspring), namely: human chorionic gonadotropin (hCG), thyroid hormone regulation, peroxisome proliferator activated receptor protein gamma (PPARγ), leptin, transforming growth factor beta, epiregulin, growth differentiation factor 15, small nucleolar RNAs, serotonin, and vitamin D. Some of these are well-established as biomarkers of placental-fetal endocrine disruption, while others are not well studied and were selected based on discovery analyses of the placental transcriptome. A literature search on these biomarkers summarizes evidence of placenta-specific production and regulation of each biomarker, and their role in fetal reproductive tract, brain, and other specific domains of fetal development. In this review, we extend the theory of fetal programming to placental-fetal programming.
Collapse
Affiliation(s)
- Jennifer J Adibi
- Department of Epidemiology, University of Pittsburgh School of Public Health, USA; Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Yaqi Zhao
- St. Jude's Research Hospital, Memphis, TN, USA
| | - Hannu Koistinen
- Department of Clinical Chemistry, University of Helsinki, Helsinki, Finland
| | - Rod T Mitchell
- Department of Paediatric Endocrinology, Royal Hospital for Children and Young People, Edinburgh BioQuarter, Edinburgh, UK
| | - Emily S Barrett
- Environmental and Population Health Bio-Sciences, Rutgers University School of Public Health, Piscataway, NJ, USA
| | - Richard Miller
- Department of Obstetrics and Gynecology, University of Rochester Medical Center, Rochester, NY, USA
| | - Thomas G O'Connor
- Department of Psychiatry, University of Rochester Medical Center, Rochester, NY, USA
| | - Xiaoshuang Xun
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - Hai-Wei Liang
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - Rahel Birru
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - Megan Smith
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Nora K Moog
- Department of Medical Psychology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| |
Collapse
|
5
|
Wang R, Feng W, Wang Y, Jiang Y, Lin Y, Chen X. Maternal obstructive sleep apnea aggravates metabolic dysfunction-associated fatty liver disease via HMGB1-TLR4 signaling-mediated endoplasmic reticulum stress in male offspring rats. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166889. [PMID: 37730152 DOI: 10.1016/j.bbadis.2023.166889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/09/2023] [Accepted: 09/15/2023] [Indexed: 09/22/2023]
Abstract
AIMS/HYPOTHESIS Maternal obstructive sleep apnea (MOSA) may inflict long-term metabolic effects on offspring. We hypothesize that MOSA increases the propensity for metabolic dysregulation in offspring and thus facilitates the development of metabolic dysfunction-associated fatty liver disease (MAFLD). This study aims to test the hypothesis and explore the underlying mechanism. METHODS The MOSA rat model of upper airway obstruction was established and fecundated. The postweaning male offspring (n = 171) from both the control group and MOSA group were randomly fed the normal chow diet (NCD, n = 89) or high-fat diet (HFD, n = 82) for the next 5 months. Liver function, lipid profile, glucose, and insulin levels were measured. Expression levels of fibrosis-related proteins and endoplasmic reticulum (ER) stress-related proteins in liver tissues were assessed using immunohistochemistry and western blotting. RESULTS MOSA increased body and liver weight in male offspring, along with augmented liver organ coefficient. Serum levels of aminotransferases, low-density lipoprotein, high-density lipoprotein, triglycerides, total cholesterol, total bile acid, fasting glucose, and insulin increased significantly. MOSA exacerbated HFD-induced hepatic steatosis and fibrosis. These effects were driven by the overactivated double-stranded RNA-activated protein kinase (PKR)-like eukaryotic initiation factor 2(PERK)-activating transcription factor (ATF)4-C/EBP homologous protein (CHOP) signaling pathway-induced ER stress, and hyperacetylation and release of high mobility group box-1(HMGB1) elicited above signaling in a TLR4-dependent manner. CONCLUSIONS These findings indicate that MOSA can exert prolonged adverse effects manifested as metabolic dysfunction in male offspring. Therefore, surveillance and management of OSA during pregnancy may be necessary to prevent and alleviate MAFLD in offspring.
Collapse
Affiliation(s)
- Ruhua Wang
- Department of Gastroenterology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120, China
| | - Wei Feng
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangdong Key Laboratory of Vascular Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510180, China
| | - Yan Wang
- Department of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yonghong Jiang
- Department of Gastroenterology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120, China
| | - Yiguang Lin
- Central Laboratory, Fist Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China..
| | - Xueqing Chen
- Department of Gastroenterology, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510120, China.
| |
Collapse
|
6
|
Kim K, Varghese M, Sun H, Abrishami S, Bowers E, Bridges D, Meijer JL, Singer K, Gregg B. The Influence of Maternal High Fat Diet During Lactation on Offspring Hematopoietic Priming. Endocrinology 2023; 165:bqad182. [PMID: 38048597 PMCID: PMC11032250 DOI: 10.1210/endocr/bqad182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/06/2023]
Abstract
Obesity and metabolic diseases are rising among women of reproductive age, increasing offspring metabolic risk. Maternal nutritional interventions during lactation present an opportunity to modify offspring outcomes. We previously demonstrated in mice that adult male offspring have metabolic impairments and increased adipose tissue macrophages (ATM) when dams are fed high fat diet (HFD) during the postnatal lactation window (HFD PN). We sought to understand the effect of HFD during lactation on early-life inflammation. HFD PN offspring were evaluated at postnatal day 16 to 19 for tissue weight and gene expression. Profiling of adipose tissue and bone marrow immune cells was conducted through lipidomics, in vitro myeloid colony forming unit assays, and flow cytometry. HFD PN mice had more visceral gonadal white adipose tissue (GWAT) and subcutaneous fat. Adipose tissue RNA sequencing demonstrated enrichment of inflammation, chemotaxis, and fatty acid metabolism and concordant changes in GWAT lipidomics. Bone marrow (BM) of both HFD PN male and female offspring had increased monocytes (CD45+Ly6G-CD11b+CD115+) and B cells (CD45+Ly6G-CD11b-CD19+). Similarly, serum from HFD PN offspring enhanced in vitro BM myeloid colonies in a toll-like receptor 4-dependent manner. We identified that male HFD PN offspring had increased GWAT pro-inflammatory CD11c+ ATMs (CD45+CD64+). Maternal exposure to HFD alters milk lipids enhancing adiposity and myeloid inflammation even in early life. Future studies are needed to understand the mechanisms driving this pro-inflammatory state of both BM and ATMs, the causes of the sexually dimorphic phenotypes, and the feasibility of intervening in this window to improve metabolic health.
Collapse
Affiliation(s)
- Katherine Kim
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mita Varghese
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Haijing Sun
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Simin Abrishami
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Emily Bowers
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Dave Bridges
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Jennifer L Meijer
- Department of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, NH 03756, USA
- Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
| | - Kanakadurga Singer
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Brigid Gregg
- Department of Pediatrics, Michigan Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| |
Collapse
|
7
|
Besset D, Selmaoui B, Delanaud S, Bessarion L, Chardon K, de Seze R, Leke A, Stéphan-Blanchard E. Influence of radiofrequency electromagnetic fields exposure on sleep patterns in preterm neonates. Int J Radiat Biol 2023; 100:427-432. [PMID: 37972294 DOI: 10.1080/09553002.2023.2277365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 09/29/2023] [Indexed: 11/19/2023]
Abstract
PURPOSE The study objective was to assess the influence of radiofrequency electromagnetic fields (RF-EMF) exposure on sleep patterns in preterm newborns. We hypothesized that an increase in RF-EMF exposure levels would alter infants' sleep structure parameters. MATERIALS AND METHODS Individual, continuous measurements of RF-EMF levels were performed in 29 hospitalized preterm newborns throughout the first 21 days after birth. The last day, overnight sleep structure was recorded by polysomnography. Relationships between both chronic (three-week period) and acute (polysomnographic period) RF-EMF levels with sleep parameters were computed. RESULTS At median levels, the main chronic effect was an increase in indeterminate sleep with RF-EMF exposure. At the highest exposure levels found in our study, an increase in RF-EMF levels increased sleep fragmentation. No significant relationship was found between acute RF-EMF levels and sleep parameters. CONCLUSIONS Despite no consolidated disruption in sleep structure, this study is the first to show that some sleep parameters seem to have a certain sensitivity to chronic - but not acute - RF-EMF exposure in preterm newborns. Further studies are needed to confirm our results and examine possible mid- to long-term, sleep-related cardiorespiratory and neurodevelopmental outcomes.
Collapse
Affiliation(s)
- Dimitri Besset
- PériTox (UMR_I 01), UPJV/INERIS, Jules Verne University of Picardy, Amiens, France
| | - Brahim Selmaoui
- PériTox (UMR_I 01), INERIS/UPJV, INERIS, Verneuil en Halatte, France
| | - Stéphane Delanaud
- PériTox (UMR_I 01), UPJV/INERIS, Jules Verne University of Picardy, Amiens, France
| | - Lisa Bessarion
- PériTox (UMR_I 01), UPJV/INERIS, Jules Verne University of Picardy, Amiens, France
- Neonatal Intensive Care Unit, CHU Amiens-Picardie, Amiens, France
| | - Karen Chardon
- PériTox (UMR_I 01), UPJV/INERIS, Jules Verne University of Picardy, Amiens, France
| | - René de Seze
- PériTox (UMR_I 01), INERIS/UPJV, INERIS, Verneuil en Halatte, France
| | - André Leke
- PériTox (UMR_I 01), UPJV/INERIS, Jules Verne University of Picardy, Amiens, France
- Neonatal Intensive Care Unit, CHU Amiens-Picardie, Amiens, France
| | | |
Collapse
|
8
|
Chardon K, Delanaud S, Tourneux P, Stephan Blanchard E. How to Control Exposure to Fifth-Generation Radiofrequencies in Preterm Newborns in Incubator. Neonatology 2023; 120:666-669. [PMID: 37343536 DOI: 10.1159/000530658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 04/06/2023] [Indexed: 06/23/2023]
Abstract
Infant and family centered development care reduces infant distress and supports the parent and infant's individual abilities. However, a new environmental factor is daily encountered: the radiofrequency electromagnetic fields (RF EMFs) with the most recent fifth-generation (5G) technology. Currently, the effects of RF EMF during development are discussed in animal models. The neonatal intensive care units are not spared from this stressor. The objective of this study was to evaluate the efficacy of a novel, electromagnetically insulating incubator cover to prevent the preterm infant from RF EMF exposure. A personal dosimeter was placed on the mattress of a closed incubator. Periods of exposure to low, medium, and high levels of 5G RF were delivered in the presence or absence of the incubator cover. The use of a silver-copper cover reduced the intensity of 5G radiofrequency levels from 52% to 57% (p < 0.0001), allowing to easily apply the precautionary principle.
Collapse
Affiliation(s)
- Karen Chardon
- PeriTox Laboratory, UMR-I 01 INERIS, Picardie Jules Verne University, Amiens, France
| | - Stéphane Delanaud
- PeriTox Laboratory, UMR-I 01 INERIS, Picardie Jules Verne University, Amiens, France
| | - Pierre Tourneux
- PeriTox Laboratory, UMR-I 01 INERIS, Picardie Jules Verne University, Amiens, France
- Neonatal Intensive Care Unit, Amiens University Hospital, Amiens, France
| | | |
Collapse
|
9
|
Lutz TA. Mammalian models of diabetes mellitus, with a focus on type 2 diabetes mellitus. Nat Rev Endocrinol 2023; 19:350-360. [PMID: 36941447 DOI: 10.1038/s41574-023-00818-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/21/2023] [Indexed: 03/23/2023]
Abstract
Although no single animal model replicates all aspects of diabetes mellitus in humans, animal models are essential for the study of energy balance and metabolism control as well as to investigate the reasons for their imbalance that could eventually lead to overt metabolic diseases such as type 2 diabetes mellitus. The most frequently used animal models in diabetes mellitus research are small rodents that harbour spontaneous genetic mutations or that can be manipulated genetically or by other means to influence their nutrient metabolism and nutrient handling. Non-rodent species, including pigs, cats and dogs, are also useful models in diabetes mellitus research. This Review will outline the advantages and disadvantages of selected animal models of diabetes mellitus to build a basis for their most appropriate use in biomedical research.
Collapse
Affiliation(s)
- Thomas A Lutz
- Institute of Veterinary Physiology, Vetsuisse Faculty University of Zurich, Zurich, Switzerland.
| |
Collapse
|
10
|
Pacheco-Fuentes H, Ton R, Griffith SC. Short- and long-term consequences of heat exposure on mitochondrial metabolism in zebra finches (Taeniopygia castanotis). Oecologia 2023; 201:637-648. [PMID: 36894790 PMCID: PMC10038956 DOI: 10.1007/s00442-023-05344-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 02/20/2023] [Indexed: 03/11/2023]
Abstract
Understanding the consequences of heat exposure on mitochondrial function is crucial as mitochondria lie at the core of metabolic processes, also affecting population dynamics. In adults, mitochondrial metabolism varies with temperature but can also depend on thermal conditions experienced during development. We exposed zebra finches to two alternative heat treatments during early development: "constant", maintained birds at ambient 35 °C from parental pair formation to fledglings' independence, while "periodic" heated broods at 40 °C, 6 h daily at nestling stage. Two years later, we acclimated birds from both experiments at 25 °C for 21 days, before exposing them to artificial heat (40 °C, 5 h daily for 10 days). After both conditions, we measured red blood cells' mitochondrial metabolism using a high-resolution respirometer. We found significantly decreased mitochondrial metabolism for Routine, Oxidative Phosphorylation (OxPhos) and Electron Transport System maximum capacity (ETS) after the heat treatments. In addition, the birds exposed to "constant" heat in early life showed lower oxygen consumption at the Proton Leak (Leak) stage after the heat treatment as adults. Females showed higher mitochondrial respiration for Routine, ETS and Leak independent of the treatments, while this pattern was reversed for OxPhos coupling efficiency (OxCE). Our results show that short-term acclimation involved reduced mitochondrial respiration, and that the reaction of adult birds to heat depends on the intensity, pattern and duration of temperature conditions experienced at early-life stages. Our study provides insight into the complexity underlying variation in mitochondrial metabolism and raises questions on the adaptive value of long-lasting physiological adjustments triggered by the early-life thermal environment.
Collapse
Affiliation(s)
| | - Riccardo Ton
- School of Natural Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - Simon C Griffith
- School of Natural Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| |
Collapse
|
11
|
Reid BM, Sokol N, Aubuchon-Endsley NL, Stroud LR. Maternal prenatal cortisol and the interaction of income and pre-pregnancy body mass index are independently associated with newborn cortisol. Dev Psychobiol 2023; 65:e22354. [PMID: 36567656 PMCID: PMC9940703 DOI: 10.1002/dev.22354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 09/21/2022] [Accepted: 10/30/2022] [Indexed: 12/14/2022]
Abstract
While extensive research has supported the developmental programming hypothesis regarding contributions of prenatal psychosocial or nutritional adversity to offspring stress physiology, fewer studies consider both exposures together with maternal stress physiology. This study examined newborn cortisol output during a stressor as a function of maternal pre-pregnancy health status and nutritional history (pre-pregnancy body mass index [PPBMI]), economic resources (household income), and maternal cortisol awakening response (mCAR) in late pregnancy. Participants were 102 mother-infant pairs from an economically and racial/ethnically diverse sample. Offspring salivary cortisol response to a neurobehavioral exam was assessed at 1 month. Income and maternal PPBMI were positively associated with mCAR in late pregnancy. mCAR was positively related to 1-month newborn cortisol response. The interaction of income and PPBMI was positively associated with newborn cortisol output during an exam at 1-month. Mothers with the highest PPBMI and lowest income had offspring with higher cortisol responses than offspring of mothers with higher income and lower PPBMI. There was no evidence of indirect mediation effects of predictors (PPBMI, income, and interaction) on infant cortisol via mCAR. The differential effects of the interaction of PPBMI and income suggest that these exposures influence infant cortisol output in the context of one another, independent of maternal pregnancy cortisol.
Collapse
Affiliation(s)
- Brie M. Reid
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Center for Behavioral and Preventive Medicine, The Miriam Hospital, Providence, Rhode Island, USA
| | - Natasha Sokol
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Center for Behavioral and Preventive Medicine, The Miriam Hospital, Providence, Rhode Island, USA
| | | | - Laura R. Stroud
- Department of Psychiatry and Human Behavior, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Center for Behavioral and Preventive Medicine, The Miriam Hospital, Providence, Rhode Island, USA
| |
Collapse
|
12
|
Gonzalez-Nahm S, Marchesoni J, Maity A, Maguire RL, House JS, Tucker R, Atkinson T, Murphy SK, Hoyo C. Maternal Mediterranean Diet Adherence and Its Associations with Maternal Prenatal Stressors and Child Growth. Curr Dev Nutr 2022; 6:nzac146. [PMID: 36406812 PMCID: PMC9665863 DOI: 10.1093/cdn/nzac146] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/19/2022] [Accepted: 09/23/2022] [Indexed: 11/16/2022] Open
Abstract
Background Psychosocial and physiologic stressors, such as depression and obesity, during pregnancy can have negative consequences, such as increased systemic inflammation, contributing to chronic disease for both mothers and their unborn children. These conditions disproportionately affect racial/ethnic minorities. The effects of recommended dietary patterns in mitigating the effects of these stressors remain understudied. Objectives We aimed to evaluate the relations between maternal Mediterranean diet adherence (MDA) and maternal and offspring outcomes during the first decade of life in African Americans, Hispanics, and Whites. Methods This study included 929 mother-child dyads from the NEST (Newborn Epigenetics STudy), a prospective cohort study. FFQs were used to estimate MDA in pregnant women. Weight and height were measured in children between birth and age 8 y. Multivariable linear regression models were used to examine associations between maternal MDA, inflammatory cytokines, and pregnancy and postnatal outcomes. Results More than 55% of White women reported high MDA during the periconceptional period compared with 22% of Hispanic and 18% of African American women (P < 0.05). Higher MDA was associated with lower likelihood of depressive mood (β = -0.45; 95% CI: -0.90, -0.18; P = 0.02) and prepregnancy obesity (β = -0.29; 95% CI: -0.57, -0.0002; P = 0.05). Higher MDA was also associated with lower body size at birth, which was maintained to ages 3-5 and 6-8 y-this association was most apparent in White children (3-5 y: β = -2.9, P = 0.02; 6-8 y: β = -3.99, P = 0.01). Conclusions If replicated in larger studies, our data suggest that MDA provides a potent avenue by which effects of prenatal stressors on maternal and fetal outcomes can be mitigated to reduce ethnic disparities in childhood obesity.
Collapse
Affiliation(s)
- Sarah Gonzalez-Nahm
- Department of Nutrition, University of Massachusetts Amherst, Amherst, MA, USA
| | - Joddy Marchesoni
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Arnab Maity
- Department of Statistics, North Carolina State University, Raleigh, NC, USA
| | - Rachel L Maguire
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - John S House
- National Institute of Environmental Health Sciences, Durham, NC, USA
| | - Rachel Tucker
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Tamara Atkinson
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| | - Susan K Murphy
- Department of Obstetrics and Gynecology, Duke University, Durham, NC, USA
| | - Cathrine Hoyo
- Department of Biological Sciences, North Carolina State University, Raleigh, NC, USA
| |
Collapse
|
13
|
Aldawood N, Jalouli M, Alrezaki A, Nahdi S, Alamri A, Alanazi M, Manoharadas S, Alwasel S, Harrath AH. Fetal programming: in utero exposure to acrylamide leads to intergenerational disrupted ovarian function and accelerated ovarian aging. Aging (Albany NY) 2022; 14:6887-6904. [PMID: 36069806 PMCID: PMC9512500 DOI: 10.18632/aging.204269] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 07/26/2022] [Indexed: 12/03/2022]
Abstract
In this study we investigated the effects of multigenerational exposures to acrylamide (ACR) on ovarian function. Fifty-day-old Wistar albino female rats were divided into the control and ACR-treated groups (2.5, 10, and 20 mg/kg/day) from day 6 of pregnancy until delivery. The obtained females of the first (AF1) and second generation (AF2) were euthanized at 4 weeks of age, and plasma and ovary samples were collected. We found that in utero multigenerational exposure to ACR reduced fertility and ovarian function in AF1 through inducing histopathological changes as evidenced by the appearance of cysts and degenerating follicles, oocyte vacuolization, and pyknosis in granulosa cells. TMR red positive cells confirmed by TUNEL assay were mostly detected in the stroma of the treated groups. Estradiol and IGF-1 concentrations significantly decreased as a result of decreased CYP19 gene and its protein expression. However, ACR exposure in AF2 led to early ovarian aging as evidenced by high estradiol and progesterone levels among all treated groups compared to control group, corresponding to the upregulation of the CYP19 gene and protein expression. The apoptotic cells of the stroma were greatly detected compared to that in the control group, whereas no significant difference was reported in ESR1 and ESR2 gene expression. This study confirms the developmental adverse effects of ACR on ovarian function and fertility in at least two consecutive generations. It emphasizes the need for more effective strategies during pregnancy, such as eating healthy foods and avoiding consumption of ACR-rich products, including fried foods and coffee.
Collapse
Affiliation(s)
- Nouf Aldawood
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Maroua Jalouli
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Abdulkarem Alrezaki
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Saber Nahdi
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah Alamri
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Mohamed Alanazi
- Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Salim Manoharadas
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Saleh Alwasel
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Abdel Halim Harrath
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
14
|
Allethrin Promotes Apoptosis and Autophagy Associated with the Oxidative Stress-Related PI3K/AKT/mTOR Signaling Pathway in Developing Rat Ovaries. Int J Mol Sci 2022; 23:ijms23126397. [PMID: 35742842 PMCID: PMC9224321 DOI: 10.3390/ijms23126397] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 02/04/2023] Open
Abstract
The increased concern regarding the reduction in female fertility and the impressive numbers of women undergoing fertility treatment support the existence of environmental factors beyond inappropriate programming of developing ovaries. Among these factors are pyrethroids, which are currently some of the most commonly used pesticides worldwide. The present study was performed to investigate the developmental effects of the pyrethroid-based insecticide allethrin on ovarian function in rat offspring in adulthood. We mainly focused on the roles of oxidative stress, apoptosis, autophagy and the related pathways in ovarian injury. Thirty-day-old Wistar albino female rats were intragastrically administered 0 (control), 34.2 or 68.5 mg/kg body weight allethrin after breeding from Day 6 of pregnancy until delivery. We found that allethrin-induced ovarian histopathological damage was accompanied by elevations in oxidative stress and apoptosis. Interestingly, the number of autophagosomes in allethrin-treated ovaries was higher, and this increase was correlated with the upregulated expression of genes and proteins related to the autophagic marker LC-3. Furthermore, allethrin downregulated the expression of PI3K, AKT and mTOR in allethrin-treated ovaries compared with control ovaries. Taken together, the findings of this study suggest that exposure to the pyrethroid-based insecticide allethrin adversely affects both the follicle structure and function in rat offspring during adulthood. Specifically, allethrin can induce excessive oxidative stress and defective autophagy-related apoptosis, probably through inactivation of the PI3K/AKT/mTOR signaling pathway, and these effects may contribute to ovarian dysfunction and impaired fertility in female offspring.
Collapse
|
15
|
Grzęda E, Matuszewska J, Ziarniak K, Gertig-Kolasa A, Krzyśko- Pieczka I, Skowrońska B, Sliwowska JH. Animal Foetal Models of Obesity and Diabetes - From Laboratory to Clinical Settings. Front Endocrinol (Lausanne) 2022; 13:785674. [PMID: 35197931 PMCID: PMC8858803 DOI: 10.3389/fendo.2022.785674] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/11/2022] [Indexed: 12/26/2022] Open
Abstract
The prenatal period, during which a fully formed newborn capable of surviving outside its mother's body is built from a single cell, is critical for human development. It is also the time when the foetus is particularly vulnerable to environmental factors, which may modulate the course of its development. Both epidemiological and animal studies have shown that foetal programming of physiological systems may alter the growth and function of organs and lead to pathology in adulthood. Nutrition is a particularly important environmental factor for the pregnant mother as it affects the condition of offspring. Numerous studies have shown that an unbalanced maternal metabolic status (under- or overnutrition) may cause long-lasting physiological and behavioural alterations, resulting in metabolic disorders, such as obesity and type 2 diabetes (T2DM). Various diets are used in laboratory settings in order to induce maternal obesity and metabolic disorders, and to alter the offspring development. The most popular models are: high-fat, high-sugar, high-fat-high-sugar, and cafeteria diets. Maternal undernutrition models are also used, which results in metabolic problems in offspring. Similarly to animal data, human studies have shown the influence of mothers' diets on the development of children. There is a strong link between the maternal diet and the birth weight, metabolic state, changes in the cardiovascular and central nervous system of the offspring. The mechanisms linking impaired foetal development and adult diseases remain under discussion. Epigenetic mechanisms are believed to play a major role in prenatal programming. Additionally, sexually dimorphic effects on offspring are observed. Therefore, further research on both sexes is necessary.
Collapse
Affiliation(s)
- Emilia Grzęda
- Laboratory of Neurobiology, Faculty of Veterinary Medicine and Animal Science, Poznań University of Life Sciences, Poznań, Poland
| | - Julia Matuszewska
- Laboratory of Neurobiology, Faculty of Veterinary Medicine and Animal Science, Poznań University of Life Sciences, Poznań, Poland
| | - Kamil Ziarniak
- Laboratory of Neurobiology, Faculty of Veterinary Medicine and Animal Science, Poznań University of Life Sciences, Poznań, Poland
- Molecular and Cell Biology Unit, Poznań University of Medical Sciences, Poznań, Poland
| | - Anna Gertig-Kolasa
- Department of Paediatric Diabetes and Obesity, Poznań University of Medical Sciences, Poznań, Poland
| | - Izabela Krzyśko- Pieczka
- Department of Paediatric Diabetes and Obesity, Poznań University of Medical Sciences, Poznań, Poland
| | - Bogda Skowrońska
- Department of Paediatric Diabetes and Obesity, Poznań University of Medical Sciences, Poznań, Poland
| | - Joanna H. Sliwowska
- Laboratory of Neurobiology, Faculty of Veterinary Medicine and Animal Science, Poznań University of Life Sciences, Poznań, Poland
- *Correspondence: Joanna H. Sliwowska,
| |
Collapse
|
16
|
Grilo LF, Tocantins C, Diniz MS, Gomes RM, Oliveira PJ, Matafome P, Pereira SP. Metabolic Disease Programming: From Mitochondria to Epigenetics, Glucocorticoid Signalling and Beyond. Eur J Clin Invest 2021; 51:e13625. [PMID: 34060076 DOI: 10.1111/eci.13625] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/22/2021] [Accepted: 05/24/2021] [Indexed: 12/11/2022]
Abstract
Embryonic and foetal development are critical periods of development in which several environmental cues determine health and disease in adulthood. Maternal conditions and an unfavourable intrauterine environment impact foetal development and may programme the offspring for increased predisposition to metabolic diseases and other chronic pathologic conditions throughout adult life. Previously, non-communicable chronic diseases were only associated with genetics and lifestyle. Now the origins of non-communicable chronic diseases are associated with early-life adaptations that produce long-term dysfunction. Early-life environment sets the long-term health and disease risk and can span through multiple generations. Recent research in developmental programming aims at identifying the molecular mechanisms responsible for developmental programming outcomes that impact cellular physiology and trigger adulthood disease. The identification of new therapeutic targets can improve offspring's health management and prevent or overcome adverse consequences of foetal programming. This review summarizes recent biomedical discoveries in the Developmental Origins of Health and Disease (DOHaD) hypothesis and highlight possible developmental programming mechanisms, including prenatal structural defects, metabolic (mitochondrial dysfunction, oxidative stress, protein modification), epigenetic and glucocorticoid signalling-related mechanisms suggesting molecular clues for the causes and consequences of programming of increased susceptibility of offspring to metabolic disease after birth. Identifying mechanisms involved in DOHaD can contribute to early interventions in pregnancy or early childhood, to re-set the metabolic homeostasis and break the chain of subsequent events that could lead to the development of disease.
Collapse
Affiliation(s)
- Luís F Grilo
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,PhD Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Carolina Tocantins
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Mariana S Diniz
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Rodrigo Mello Gomes
- Department of Physiological Sciences, Biological Sciences Institute, Federal University of Goiás, Goiânia, Brazil
| | - Paulo J Oliveira
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Paulo Matafome
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal.,Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Clinical Academic Center of Coimbra (CACC), Coimbra, Portugal.,Department of Complementary Sciences, Instituto Politécnico de Coimbra, Coimbra Health School (ESTeSC), Coimbra, Portugal
| | - Susana P Pereira
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,Laboratory of Metabolism and Exercise (LametEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, University of Porto, Porto, Portugal
| |
Collapse
|
17
|
Fernández-Pacheco C, Millán P, Rodríguez M, Formoso-Rafferty N, Sánchez-Rodríguez A, Lorenzo PL, Arias-Álvarez M, García-García RM, Rebollar PG. Influence of Different Regimes of Moderate Maternal Feed Restriction during Pregnancy of Primiparous Rabbit Does on Long-Term Metabolic Energy Homeostasis, Productive Performance and Welfare. Animals (Basel) 2021; 11:2736. [PMID: 34573702 PMCID: PMC8470312 DOI: 10.3390/ani11092736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 11/16/2022] Open
Abstract
In this study, a maternal feed restriction (MFR; 105 g/d) in primiparous rabbit does was applied from day 0 to 7 post artificial insemination (AI) (R07, n = 96), from day 7 to 21 post AI (R721, n = 92), from day 0 to 21 post AI (R021, n = 94) or fed ad libitum during whole pregnancy (Control, n= 92). Feed intake (FI) was measured after MFR was over. On day 28 of gestation, fetoplacental development was evaluated (n = 11/group) and the productive parameters of the remaining dams were analyzed. Plasma free tri-iodothyronine (T3) and thyroxine, glucose, insulin, non-esterified fatty acids (NEFA), and corticosterone were analyzed during gestation and lactation (n = 5/group). After MFR, all groups significantly increased their voluntary FI. The longer MFR was, the lower the weight and length of the fetuses, but no long-term effects over litter performance were observed. R021 groups had the lowest T3 and the highest NEFA concentrations during pregnancy and showed insulin resistance at the end of gestation, but during lactation, energy homeostasis was balanced in all groups. MFR did not affect corticosterone concentrations. In conclusion, the ration setting applied slightly involved the energy homeostasis and metabolism of the animals, but their overall metabolic condition, productive performance and welfare were not compromised.
Collapse
Affiliation(s)
- Carlota Fernández-Pacheco
- Department of Physiology, Faculty of Veterinary, Complutense University of Madrid, Avenida Puerta de Hierro s/n, 28040 Madrid, Spain; (C.F.-P.); (P.M.); (A.S.-R.); (P.L.L.); (R.M.G.-G.)
| | - Pilar Millán
- Department of Physiology, Faculty of Veterinary, Complutense University of Madrid, Avenida Puerta de Hierro s/n, 28040 Madrid, Spain; (C.F.-P.); (P.M.); (A.S.-R.); (P.L.L.); (R.M.G.-G.)
| | - María Rodríguez
- Department of Agrarian Production, ETSIAAB, Technical University of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain; (M.R.); (N.F.-R.)
| | - Nora Formoso-Rafferty
- Department of Agrarian Production, ETSIAAB, Technical University of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain; (M.R.); (N.F.-R.)
| | - Ana Sánchez-Rodríguez
- Department of Physiology, Faculty of Veterinary, Complutense University of Madrid, Avenida Puerta de Hierro s/n, 28040 Madrid, Spain; (C.F.-P.); (P.M.); (A.S.-R.); (P.L.L.); (R.M.G.-G.)
| | - Pedro L. Lorenzo
- Department of Physiology, Faculty of Veterinary, Complutense University of Madrid, Avenida Puerta de Hierro s/n, 28040 Madrid, Spain; (C.F.-P.); (P.M.); (A.S.-R.); (P.L.L.); (R.M.G.-G.)
| | - María Arias-Álvarez
- Department of Animal Production, Faculty of Veterinary, Complutense University of Madrid, Avenida Puerta de Hierro s/n, 28040 Madrid, Spain;
| | - Rosa M. García-García
- Department of Physiology, Faculty of Veterinary, Complutense University of Madrid, Avenida Puerta de Hierro s/n, 28040 Madrid, Spain; (C.F.-P.); (P.M.); (A.S.-R.); (P.L.L.); (R.M.G.-G.)
| | - Pilar G. Rebollar
- Department of Agrarian Production, ETSIAAB, Technical University of Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain; (M.R.); (N.F.-R.)
| |
Collapse
|
18
|
The effect of season of birth on brain epigenome-wide DNA methylation of older adults. J Dev Orig Health Dis 2021; 13:367-377. [PMID: 34308828 DOI: 10.1017/s2040174421000453] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Perinatal light exposure predisposes towards health and behaviour in adulthood. Season of birth is associated with psychiatric, allergic, cardiovascular and metabolic problems. It has been proposed that early-life environmental light disrupts the development of biological rhythms which, in turn, influence later-life health. However, the mechanisms linking perinatal seasonal light to later-life biological rhythm and health in humans are unknown. In this study, we investigated the association between season of birth and epigenome-wide DNA methylation of two postmortem human brain regions (16 hypothalamus, 14 temporal cortex). We did not find statistically significant differences at the whole epigenome level, either because we lacked statistical power or that no association exists. However, when we examined 24 CpG sites that had the highest significance or differential methylation, we identified regions which may be associated with circadian rhythm entrainment, cholinergic neurotransmission and neural development. Amongst methylation of the core clock genes, we identified that hypothalamus Neuronal PAS Domain Protein 2 (NPAS2) gene has hypermethylated regions in long photoperiod-born individuals. In addition, we found nominal associations between season of birth and genes linked to chronotype and narcolepsy. Season of birth-related brain DNA methylation profile was different than a previously reported blood methylation profile, suggesting a tissue-specific mechanism of perinatal light programming. Overall, we are the first to analyse the relationship between season of birth and human brain DNA methylation. Further studies with larger sample sizes are required to confirm an imprinting effect of perinatal light on the circadian clock.
Collapse
|
19
|
Clyburn C, Travagli RA, Arnold AC, Browning KN. DMV extrasynaptic NMDA receptors regulate caloric intake in rats. JCI Insight 2021; 6:139785. [PMID: 33764905 PMCID: PMC8262316 DOI: 10.1172/jci.insight.139785] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 03/24/2021] [Indexed: 11/17/2022] Open
Abstract
Acute high-fat diet (aHFD) exposure induces a brief period of hyperphagia before caloric balance is restored. Previous studies have demonstrated that this period of regulation is associated with activation of synaptic N-methyl-D-aspartate (NMDA) receptors on dorsal motor nucleus of the vagus (DMV) neurons, which increases vagal control of gastric functions. Our aim was to test the hypothesis that activation of DMV synaptic NMDA receptors occurs subsequent to activation of extrasynaptic NMDA receptors. Sprague-Dawley rats were fed a control or high-fat diet for 3-5 days prior to experimentation. Whole-cell patch-clamp recordings from gastric-projecting DMV neurons; in vivo recordings of gastric motility, tone, compliance, and emptying; and food intake studies were used to assess the effects of NMDA receptor antagonism on caloric regulation. After aHFD exposure, inhibition of extrasynaptic NMDA receptors prevented the synaptic NMDA receptor-mediated increase in glutamatergic transmission to DMV neurons, as well as the increase in gastric tone and motility, while chronic extrasynaptic NMDA receptor inhibition attenuated the regulation of caloric intake. After aHFD exposure, the regulation of food intake involved synaptic NMDA receptor-mediated currents, which occurred in response to extrasynaptic NMDA receptor activation. Understanding these events may provide a mechanistic basis for hyperphagia and may identify novel therapeutic targets for the treatment of obesity.
Collapse
|
20
|
Asiedu B, Nyakudya TT, Lembede BW, Chivandi E. Early-life exposure to alcohol and the risk of alcohol-induced liver disease in adulthood. Birth Defects Res 2021; 113:451-468. [PMID: 33577143 DOI: 10.1002/bdr2.1881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 01/13/2021] [Accepted: 01/19/2021] [Indexed: 11/10/2022]
Abstract
Alcohol consumption remains prevalent among pregnant and nursing mothers despite the well-documented adverse effects this may have on the offspring. Moderate-to-high levels of alcohol consumption in pregnancy result in fetal alcohol syndrome (FAS) disorders, with brain defects being chief among the abnormalities. Recent findings indicate that while light-to-moderate levels may not cause FAS, it may contribute to epigenetic changes that make the offspring prone to adverse health outcomes including metabolic disorders and an increased propensity in the adolescent-onset of drinking alcohol. On the one hand, prenatal alcohol exposure (PAE) causes epigenetic changes that affect lipid and glucose transcript regulating genes resulting in metabolic abnormalities. On the other hand, it can program offspring for increased alcohol intake, enhance its palatability, and increase acceptance of alcohol's flavor through associative learning, making alcohol a plausible second hit for the development of alcohol-induced liver disease. Adolescent drinking results in alcohol dependence and abuse in adulthood. Adolescent drinking results in alcohol dependence and abuse in adulthood. Alterations on the opioid system, particularly, the mu-opioid system, has been implicated in the mechanism that induces increased alcohol consumption and acceptance. This review proposes a mechanism that links PAE to the development of alcoholism and eventually to alcoholic liver disease (ALD), which results from prolonged alcohol consumption. While PAE may not lead to ALD development in childhood, there are chances that it may lead to ALD in adulthood.
Collapse
Affiliation(s)
- Bernice Asiedu
- Faculty of Health Sciences, School of Physiology, University of the Witwatersrand, Johannesburg, South Africa
| | - Trevor Tapiwa Nyakudya
- Department of Physiology, Faculty of Health Sciences, School of Medicine, University of Pretoria, Gezina, South Africa
| | - Busisani Wiseman Lembede
- Faculty of Health Sciences, School of Physiology, University of the Witwatersrand, Johannesburg, South Africa
| | - Eliton Chivandi
- Faculty of Health Sciences, School of Physiology, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
21
|
Pham A, Zhang J, Feng L. Exposure to perfluorobutane sulfonate and perfluorooctanesulfonic acid disrupts the production of angiogenesis factors and stress responses in human placental syncytiotrophoblast. Reprod Toxicol 2020; 98:269-277. [PMID: 33144174 DOI: 10.1016/j.reprotox.2020.10.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 10/14/2020] [Accepted: 10/25/2020] [Indexed: 12/18/2022]
Abstract
Poly- and per-fluoroalkyl substances (PFAS) have attracted widespread attention in recent years due to their bioaccumulation, toxicity, and ubiquitous nature. We and others have reported that maternal exposure to PFAS is associated with adverse birth outcomes due to altered placental functions. In this study, we investigated the effects of two major PFAS compounds, perfluorobutane sulfonate (PFBS) and perfluorooctanesulfonic acid (PFOS), on the regulation of the production of angiogenic factors and stress response in placental multinucleated syncytial BeWo cells using qRT-PCR and ELISA. Using this in vitro model, we showed that 1) PFOS or PFBS treatment did not seem to interrupt BeWo cell fusion through syncytins; 2) Exposure to PFOS at 10 μM decreased a potent angiogenic factor PlGF gene expression, which is implicated in preeclampsia; 3) Exposure to either PFOS or PFBS significantly decreased the production of CGB7 and hCG except hCG secretion in PFOS (10 nM) and PFBS (100 nM) treatment groups; 4) Exposure to PFOS (10 μM) increased the gene expression of the stress response molecules CRH while neither PFOS nor PFBS treatment affected a stress mitigation factor 11β-HSD2 expression. Our results demonstrate that exposure to PFOS or PFBS impacts several key pathways involved in placental cell functions. PFOS seems more potent than PFBS. These novel findings provide a potential explanation for the adverse reproductive complications associated with prenatal exposure to PFOS or PFBS, including preeclampsia and contribute to our knowledge of the reproductive toxicity of PFAS, specifically PFOS and PFBS.
Collapse
Affiliation(s)
- Angela Pham
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, United States
| | - Jun Zhang
- MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Jiao Tong University School of Medicine, Shanghai, China
| | - Liping Feng
- Department of Obstetrics and Gynecology, Duke University School of Medicine, Durham, NC, United States; MOE-Shanghai Key Laboratory of Children's Environmental Health, Xinhua Hospital, Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
22
|
Desai M, Ferrini MG, Han G, Narwani K, Ross MG. Maternal High Fat Diet Programs Male Mice Offspring Hyperphagia and Obesity: Mechanism of Increased Appetite Neurons via Altered Neurogenic Factors and Nutrient Sensor AMPK. Nutrients 2020; 12:nu12113326. [PMID: 33138074 PMCID: PMC7693487 DOI: 10.3390/nu12113326] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/20/2020] [Accepted: 10/26/2020] [Indexed: 01/01/2023] Open
Abstract
Maternal high-fat (HF) is associated with offspring hyperphagia and obesity. We hypothesized that maternal HF alters fetal neuroprogenitor cell (NPC) and hypothalamic arcuate nucleus (ARC) development with preferential differentiation of neurons towards orexigenic (NPY/AgRP) versus anorexigenic (POMC) neurons, leading to offspring hyperphagia and obesity. Furthermore, these changes may involve hypothalamic bHLH neuroregulatory factors (Hes1, Mash1, Ngn3) and energy sensor AMPK. Female mice were fed either a control or a high fat (HF) diet prior to mating, and during pregnancy and lactation. HF male newborns were heavier at birth and exhibited decreased protein expression of hypothalamic bHLH factors, pAMPK/AMPK and POMC with increased AgRP. As adults, these changes persisted though with increased ARC pAMPK/AMPK. Importantly, the total NPY neurons were increased, which was consistent with the increased food intake and adult fat mass. Further, NPCs from HF newborn hypothalamic tissue showed similar changes with preferential NPC neuronal differentiation towards NPY. Lastly, the role of AMPK was further confirmed with in vitro treatment of Control NPCs with pharmacologic AMPK modulators. Thus, the altered ARC development of HF offspring results in excess appetite and reduced satiety leading to obesity. The underlying mechanism may involve AMPK/bHLH pathways.
Collapse
Affiliation(s)
- Mina Desai
- Department of Obstetrics and Gynecology, Perinatal Research Laboratory, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (G.H.); (K.N.); (M.G.R.)
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA 90024, USA
- Correspondence: ; Tel.: +310-974-9540
| | - Monica G. Ferrini
- Department of Health and Life Sciences, Charles R. Drew University, Los Angeles, CA 90059, USA;
- Department of Internal Medicine, Charles R. Drew University, Los Angeles, CA 90059, USA
| | - Guang Han
- Department of Obstetrics and Gynecology, Perinatal Research Laboratory, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (G.H.); (K.N.); (M.G.R.)
| | - Kavita Narwani
- Department of Obstetrics and Gynecology, Perinatal Research Laboratory, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (G.H.); (K.N.); (M.G.R.)
| | - Michael G. Ross
- Department of Obstetrics and Gynecology, Perinatal Research Laboratory, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, CA 90502, USA; (G.H.); (K.N.); (M.G.R.)
- Department of Obstetrics and Gynecology, David Geffen School of Medicine, University of California, Los Angeles, CA 90024, USA
- Department of Obstetrics and Gynecology, Charles R. Drew University, Los Angeles, CA 90059, USA
| |
Collapse
|
23
|
Early life overnutrition impairs plasticity of non-neuronal brainstem cells and drives obesity in offspring across development in rats. Int J Obes (Lond) 2020; 44:2405-2418. [PMID: 32999409 DOI: 10.1038/s41366-020-00658-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 07/17/2020] [Accepted: 08/15/2020] [Indexed: 12/11/2022]
Abstract
BACKGROUND The prevalence of adolescent obesity has increased dramatically, becoming a serious public health concern. While previous evidence suggests that in utero- and early postnatal overnutrition increases adult-onset obesity risk, the neurobiological mechanisms underlying this outcome are not well understood. Non-neuronal cells play an underestimated role in the physiological responses to metabolic/nutrient signals. Hypothalamic glial-mediated inflammation is now considered a contributing factor in the development and perpetuation of obesity; however, attention on the role of gliosis and microglia activation in other nuclei is still needed. METHODS/RESULTS Here, we demonstrate that early life consumption of high-fat/sucrose diet (HFSD) is sufficient to increase offspring body weight, hyperleptinemia and potentially maladaptive cytoarchitectural changes in the brainstem dorsal-vagal-complex (DVC), an essential energy balance processing hub, across postnatal development. Our data demonstrate that pre- and postnatal consumption of HFSD result in increased body weight, hyperleptinemia and dramatically affects the non-neuronal landscape, and therefore the plasticity of the DVC in the developing offspring. CONCLUSIONS Current findings are very provocative, considering the importance of the DVC in appetite regulation, suggesting that HFSD-consumption during early life may contribute to subsequent obesity risk via DVC cytoarchitectural changes.
Collapse
|
24
|
Hernandez-Trejo M, Sámano R, Chico-Barba G, Pizano-Zarate ML, Herrera-González NE. Neonatal adiposity may increase plasmatic cytokines. PLoS One 2020; 15:e0238370. [PMID: 32886687 PMCID: PMC7473588 DOI: 10.1371/journal.pone.0238370] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 08/15/2020] [Indexed: 11/18/2022] Open
Abstract
Maternal health and nutritional status before and during gestation may affect neonates' immune system and energy balance as they develop. The objective of this study was to associate certain clinical markers of maternal adiposity (body mass index and gestational weight gain) and neonatal adiposity (birth weight, abdominal circumference, and waist/height index) with the levels of pro- and anti-inflammatory cytokines in umbilical cord blood at birth: IL-1β, IL-1Rα, IL-4, IL-6, IL-10, IFN-γ, and TNF-α. An exploratory cross-sectional study was conducted with a convenience sample of women from one hospital recruited shortly before giving birth through scheduled cesarean section. Of 31 the pregnant women who agreed to participate and met the inclusion criteria, twenty-nine newborns from these women were analyzed. Three cases of tobacco smoking during pregnancy were identified as an unexpected maternal risk factor and were included in the analysis. Typical of the population treated at this hospital, ten of our participants had diabetes during pregnancy, and nine of them had a pre-pregnancy BMI> 25. Non-parametric statistical analyses and a generalized linear model with gamma scale response with a log link were performed. Results: Correlation analyses, differences in medians, and a prediction model all showed positive and significant results between cytokine levels in cord blood and neonatal abdominal circumference, birth weight, and waist-height index. For maternal variables, smoking during pregnancy showed significant associations with cytokine levels in cord blood. Conclusion: This study found a variety of associations suggesting that increased neonatal adiposity increases pro-inflammatory cytokine levels at birth.
Collapse
Affiliation(s)
- Maria Hernandez-Trejo
- Neurobiología del Desarrollo, Instituto Nacional de Perinatología, Secretaría de Salud, Ciudad de México, México
- * E-mail:
| | - Reyna Sámano
- Nutrición y Bioprogramación, Instituto Nacional de Perinatología, Secretaría de Salud, Ciudad de México, México
| | - Gabriela Chico-Barba
- Nutrición y Bioprogramación, Instituto Nacional de Perinatología, Secretaría de Salud, Ciudad de México, México
| | - Maria Luisa Pizano-Zarate
- Nutrición y Bioprogramación, Instituto Nacional de Perinatología, Secretaría de Salud, Ciudad de México, México
| | - Norma Estela Herrera-González
- Sección de Posgrado e Investigación, Escuela Superior de Medicina del Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Casco de Santo Tomás, Ciudad de México, México
| |
Collapse
|
25
|
Age-related changes in cerebral congenital toxoplasmosis: Histopathological and immunohistochemical evaluation. J Neuroimmunol 2020; 348:577384. [PMID: 32919146 DOI: 10.1016/j.jneuroim.2020.577384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 08/20/2020] [Accepted: 09/01/2020] [Indexed: 11/24/2022]
Abstract
Congenital toxoplasmosis is a widespread worldwide disease producing varying degrees of damage to the fetus including ocular and neurological impairment. However, the underlying mechanisms are not yet clear. Therefore, the current study aimed to investigate the progress of congenital cerebral toxoplasmosis in experimentally infected offspring animal model at different age groups till become adults. To fulfill this aim, the offspring of Me49 T. gondii infected pregnant mice were divided into groups; embryo, infant, young and adult phases. Blood and brain samples were collected for further hormonal and histopathological studies and immunohistochemical staining of glial fibrillary acidic protein (GFAP) and synaptophysin (SYN). Our results showed several encephalitic changes in the infected groups ranging from gliosis to reduced cortical cell number and fibrinoid degeneration of the brain. We showed increased expression of GFAP and SYN indicating activation of astrocytes and modification of the synaptic function, respectively. These changes started intrauterine following congenital infection and increased progressively afterward. Moreover, infected mice had elevated corticosterone levels. In conclusion, the current study provided new evidences for the cellular changes especially in the infected embryo and highlighted the role of GFAP and SYN that may be used as indicators for T. gondii-related neuropathy.
Collapse
|
26
|
Kislal S, Shook LL, Edlow AG. Perinatal exposure to maternal obesity: Lasting cardiometabolic impact on offspring. Prenat Diagn 2020; 40:1109-1125. [PMID: 32643194 DOI: 10.1002/pd.5784] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 06/25/2020] [Accepted: 07/05/2020] [Indexed: 12/11/2022]
Abstract
Evidence from epidemiological, clinical, and animal model studies clearly demonstrates that prenatal and lactational maternal obesity and high-fat diet consumption are associated with cardiometabolic morbidity in offspring. Fetal and offspring sex may be an important effect modifier. Adverse offspring cardiometabolic outcomes observed in the setting of maternal obesity include an increased risk for obesity, features of metabolic syndrome (hypertension, hyperglycemia and insulin resistance, hyperlipidemia, increased adiposity), and non-alcoholic fatty liver disease. This review article synthesizes human and animal data linking maternal obesity and high-fat diet consumption in pregnancy and lactation to adverse cardiometabolic outcomes in offspring. We review key mechanisms underlying skeletal muscle, adipose tissue, pancreatic, liver, and central brain reward programming in obesity-exposed offspring, and how such malprogramming contributes to offspring cardiometabolic morbidity.
Collapse
Affiliation(s)
- Sezen Kislal
- Vincent Center for Reproductive Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Lydia L Shook
- Division of Maternal-Fetal Medicine, Department of Ob/Gyn, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Andrea G Edlow
- Vincent Center for Reproductive Biology, Massachusetts General Hospital Research Institute, Massachusetts General Hospital, Boston, Massachusetts, USA.,Division of Maternal-Fetal Medicine, Department of Ob/Gyn, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
27
|
Sun S, Corbeels K, Desmet L, Segers A, Wang Q, Van Der Schueren B, Depoortere I. Involvement of the GHSR in the developmental programming and metabolic disturbances induced by maternal undernutrition. J Nutr Biochem 2020; 85:108468. [PMID: 32750410 DOI: 10.1016/j.jnutbio.2020.108468] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 07/03/2020] [Accepted: 07/13/2020] [Indexed: 12/18/2022]
Abstract
The mismatch between maternal undernutrition and adequate nutrition after birth increases the risk of developing metabolic diseases. We aimed to investigate whether the hyperghrelinemia during maternal undernourishment rewires the hypothalamic development of the offspring and contributes to the conversion to an obese phenotype when fed a high-fat diet (HFD). Pregnant C57BL/6 J, wild type (WT) and ghrelin receptor (GHSR)-/- mice were assigned to either a normal nourished (NN) group, or an undernutrition (UN) (30% food restricted) group. All pups were fostered by NN Swiss mice. After weaning, pups were fed a normal diet, followed by a HFD from week 9. Plasma ghrelin levels peaked at postnatal day 15 (P15) in both C57BL/6 J UN and NN pups. Hypothalamic Ghsr mRNA expression was upregulated at P15 in UN pups compared to NN pups and inhibited agouti-related peptide (AgRP) projections. Adequate lactation increased body weight of UN WT but not of GHSR-/- pups compared to NN littermates. After weaning with a HFD, body weight and food intake was higher in WT UN pups but lower in GHSR-/- UN pups than in NN controls. The GHSR prevented a decrease in ambulatory activity and oxygen consumption in UN offspring during ad libitum feeding. Maternal undernutrition triggers developmental changes in the hypothalamus in utero which were further affected by adequate feeding after birth during the postnatal period by affecting GHSR signaling. The GHSR contributes to the hyperphagia and the increase in body weight when maternal undernutrition is followed by an obesity prone life environment.
Collapse
Affiliation(s)
- Shu Sun
- Gut Peptide Research Lab, Targid, KU Leuven,3000 Leuven, Belgium
| | - Katrien Corbeels
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven,3000 Leuven, Belgium
| | - Louis Desmet
- Gut Peptide Research Lab, Targid, KU Leuven,3000 Leuven, Belgium
| | - Anneleen Segers
- Gut Peptide Research Lab, Targid, KU Leuven,3000 Leuven, Belgium
| | - Qiaoling Wang
- Gut Peptide Research Lab, Targid, KU Leuven,3000 Leuven, Belgium
| | - Bart Van Der Schueren
- Clinical and Experimental Endocrinology, Department of Chronic Diseases, Metabolism and Ageing (CHROMETA), KU Leuven,3000 Leuven, Belgium
| | - Inge Depoortere
- Gut Peptide Research Lab, Targid, KU Leuven,3000 Leuven, Belgium.
| |
Collapse
|
28
|
Ottolina J, Schimberni M, Makieva S, Bartiromo L, Fazia T, Bernardinelli L, Viganò P, Candiani M, Gentilini D. Early-life factors, in-utero exposures and endometriosis risk: a meta-analysis. Reprod Biomed Online 2020; 41:279-289. [PMID: 32532666 DOI: 10.1016/j.rbmo.2020.04.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 03/25/2020] [Accepted: 04/07/2020] [Indexed: 02/07/2023]
Abstract
This meta-analysis aimed to offer a general picture of the available data on the effects of early-life factors on the risk of developing endometriosis in adult life. An advanced, systematic search of the online medical databases PubMed, EMBASE and CINAHL was limited to full-length manuscripts published in English in peer-reviewed journals up to February 2019. Log of relative risk (RR) was employed to calculate the pooled effect sizes using both fixed and random effects modelling and I-squared tests to assess heterogeneity. Funnel plots were used to investigation publication bias. The meta-analysis was registered in PROSPERO (ID CRD42019138668). Six studies that included a total of 2360 women affected by endometriosis were analysed. The pooled results showed that the risk of developing endometriosis in adult life was significantly increased by being born prematurely (logRR 0.21, 95% CI -0.03 to 0.40), having a low birthweight (logRR 0.35, 95% CI -0.15 to 0.54), being formula-fed (logRR 0.65, 95% CI -0.35 to 0.95) and having been exposed to diethylstilbestrol (DES) in utero (logRR 0.65, 95% CI 0.26 to 1.04. Among intrauterine and early neonatal exposures, prematurity, birthweight, formula feeding and DES were risk factors for the development of endometriosis in adult life.
Collapse
Affiliation(s)
- Jessica Ottolina
- Gynecol/Obstet Unit, San Raffaele Scientific Institute, Milan, Italy.
| | - Matteo Schimberni
- Gynecol/Obstet Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Sofia Makieva
- Reproductive Sciences Laboratory, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | | | - Teresa Fazia
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Luisa Bernardinelli
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Paola Viganò
- Reproductive Sciences Laboratory, Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Massimo Candiani
- Gynecol/Obstet Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Davide Gentilini
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy; Istituto Auxologico Italiano IRCCS, Bioinformatics and Statistical Genomics Unit, Cusano MilaninoMilan, Italy
| |
Collapse
|
29
|
Sex-dependent metabolic effects of pregestational exercise on prenatally stressed mice. J Dev Orig Health Dis 2020; 12:271-279. [PMID: 32406352 DOI: 10.1017/s2040174420000343] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Stressful events during the prenatal period have been related to hyperactive hypothalamic-pituitary-adrenal (HPA) axis responses as well as metabolic changes in adult life. Moreover, regular exercise may contribute to the improvement of the symptoms associated with stress and stress-related chronic diseases. Therefore, this study aims to investigate the effects of exercise, before the gestation period, on the metabolic changes induced by prenatal stress in adult mice. Female Balb/c mice were divided into three groups: control (CON), prenatal restraint stress (PNS) and exercise before the gestational period plus PNS (EX + PNS). When adults, the plasmatic biochemical analysis, oxidative stress, gene expression of metabolic-related receptors and sex differences were assessed in the offspring. Prenatal stress decreased neonatal and adult body weight when compared to the pregestational exercise group. Moreover, prenatal stress was associated with reduced body weight in adult males. PNS and EX + PNS females showed decreased hepatic catalase. Pregestational exercise prevented the stress-induced cholesterol increase in females but did not prevent the liver mRNA expression reduction on the peroxisome proliferator-activated receptors (PPARs) α and γ in PNS females. Conversely, PNS and EX + PNS males showed an increased PPARα mRNA expression. In conclusion, pregestational exercise prevented some effects of prenatal stress on metabolic markers in a sex-specific manner.
Collapse
|
30
|
Joseph S, Alava B, Antolic A, Richards EM, Wood CE, Keller-Wood M. Fetal ovine skeletal and cardiac muscle transcriptomics are differentially altered by increased maternal cortisol during gestation. Physiol Genomics 2020; 52:178-190. [PMID: 32116114 DOI: 10.1152/physiolgenomics.00096.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
We have previously found that in utero exposure to excess maternal cortisol (1 mg/kg/day) in late gestation increases the incidence of stillbirth during labor and produces fetal bradycardia at birth. In the interventricular septum, mitochondrial DNA (mt-DNA) was decreased, and transcriptomics and metabolomics were consistent with altered mitochondrial metabolism. The present study uses transcriptomics to model effects of increased maternal cortisol on fetal biceps femoris. Transcriptomic modeling revealed that pathways related to mitochondrial metabolism were downregulated, whereas pathways for regulation of reactive oxygen species and activation of the apoptotic cascade were upregulated. Mt-DNA and the protein levels of cytochrome C were significantly decreased in the biceps femoris. RT-PCR validation of the pathways confirmed a significant decrease in SLC2A4 mRNA levels and a significant increase in PDK4, TXNIP, ANGPTL4 mRNA levels, suggesting that insulin sensitivity of the biceps femoris muscle may be reduced in cortisol offspring. We also tested for changes in gene expression in diaphragm by rt-PCR. PDK4, TXNIP, and ANGPTL4 mRNA were also increased in the diaphragm, but SLC2A4, cytochrome C protein, and mt-DNA were unchanged. Comparison of the change in gene expression in biceps femoris to that in cardiac interventricular septum and left ventricle showed few common genes and little overlap in specific metabolic or signaling pathways, despite reduction in mt-DNA in both heart and biceps femoris. Our results suggest that glucocorticoid exposure alters expression of nuclear genes important to mitochondrial activity and oxidative stress in both cardiac and skeletal muscle tissues, but that these effects are tissue-specific.
Collapse
Affiliation(s)
- Serene Joseph
- Department of Pharmacodynamics, University of Florida, Gainesville, Florida
| | - Bryan Alava
- Department of Pharmacodynamics, University of Florida, Gainesville, Florida
| | - Andrew Antolic
- Department of Pharmacodynamics, University of Florida, Gainesville, Florida
| | - Elaine M Richards
- Department of Pharmacodynamics, University of Florida, Gainesville, Florida.,Department of Pharmacodynamics, Physiology and Functional Genomics, University of Florida, Gainesville, Florida
| | - Charles E Wood
- Department of Pharmacodynamics, Physiology and Functional Genomics, University of Florida, Gainesville, Florida.,Donald H Barron Reproductive and Perinatal Biology Research Program, University of Florida, Gainesville, Florida
| | - Maureen Keller-Wood
- Department of Pharmacodynamics, University of Florida, Gainesville, Florida.,Donald H Barron Reproductive and Perinatal Biology Research Program, University of Florida, Gainesville, Florida
| |
Collapse
|
31
|
Reeves J, Smith C, Dierenfeld ES, Whitehouse-Tedd K. Captivity-induced metabolic programming in an endangered felid: implications for species conservation. Sci Rep 2020; 10:3630. [PMID: 32107441 PMCID: PMC7046719 DOI: 10.1038/s41598-020-60577-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 02/12/2020] [Indexed: 11/08/2022] Open
Abstract
Reintroduction of captive-bred individuals into the wild is an important conservation activity. However, environmental conditions can influence developmental programming, potentially causing metabolic disorders in adults. These effects are investigated here for the first time in an endangered species. Using body weight and feed intake data for Iberian lynx (Lynx pardinus) (n = 22), we compared the growth of captive versus wild born and/or reared individuals. Captive-born individuals gained weight as a function of calorie intake, unlike wild-born individuals. When compared with females reared in the wild, captive-reared females achieved a larger body size, without evidence of obesity. Captivity-associated changes to metabolic programming may compromise survival in the wild if an increased body size incurs a greater energy requirement. Large body size may also confer a competitive advantage over smaller, wild-born individuals, disrupting the social organisation of existing wild populations, and inferring long-term implications for the phenotypic composition of wild populations.
Collapse
Affiliation(s)
- Jessica Reeves
- Iberian Lynx Captive Breeding Centre "El Acebuche", Parque Nacional de Doñana, Matalascañas, 21760, Huelva, Spain
| | - Carl Smith
- School of Animal, Rural and Environmental Sciences, Nottingham Trent University, Southwell, NG25 0QF, Nottinghamshire, United Kingdom
- Department of Ecology & Vertebrate Zoology, University of Łódź, 12/16 Banacha Street, 90-237, Łódź, Poland
- Institute of Vertebrate Biology, Academy of Sciences of the Czech Republic, Květná 8, 603 65, Brno, Czech Republic
| | - Ellen S Dierenfeld
- School of Animal, Rural and Environmental Sciences, Nottingham Trent University, Southwell, NG25 0QF, Nottinghamshire, United Kingdom
- Ellen S. Dierenfeld LLC, St. Louis, MO, United States of America
| | - Katherine Whitehouse-Tedd
- School of Animal, Rural and Environmental Sciences, Nottingham Trent University, Southwell, NG25 0QF, Nottinghamshire, United Kingdom.
| |
Collapse
|
32
|
Méquinion M, Foldi CJ, Andrews ZB. The Ghrelin-AgRP Neuron Nexus in Anorexia Nervosa: Implications for Metabolic and Behavioral Adaptations. Front Nutr 2020; 6:190. [PMID: 31998738 PMCID: PMC6962137 DOI: 10.3389/fnut.2019.00190] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 12/11/2019] [Indexed: 11/13/2022] Open
Abstract
Anorexia Nervosa (AN) is viewed as primarily a psychiatric disorder owing to the considerable behavioral and genetic overlap with mood disorders and other psychiatric traits. However, the recent reconceptualization of AN as one of both psychiatric and metabolic etiology suggests that metabolic circuits conveying hunger, or sensitive to signals of hunger, may be a critical nexus linking metabolic dysfunction to mood disturbances. Within the brain, hunger is primarily percieved by Agouti-related (AgRP) neurons and hunger increases plasma concentrations of the hormone ghrelin, which targets ghrelin receptors on AgRP neurons to facilitate metabolic adaptations to low energy availability. However, beyond the fundamental role in maintaining hunger signaling, AgRP neurons regulate a diverse range of behaviors such as motivation, locomotor activity, negative reinforcement, anxiety, and obsession and a key factor involved in the manifestation of these behavioral changes in response to activation is the presence or absence of food availability. These changes can be considered adaptive in that they promote affective food-seeking strategies in environments with limited food availability. However, it also suggests that these neurons, so well-studied for their metabolic control, shape mood-related behaviors in a context-dependent manner and dysfunctional control leads not only to metabolic problems but also potentially mood-related problems. The purpose of this review is to underline the potential role of AgRP neurons and ghrelin signaling in both the metabolic and behavioral changes observed in anorexia nervosa. We aim to highlight the most recent studies on AgRP neurons and ghrelin signaling and integrate their metabolic and behavioral roles in normal function and highlight how dysfunction may contribute to the development of AN.
Collapse
Affiliation(s)
| | | | - Zane B. Andrews
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
33
|
Abstract
Many animal models that are currently used in appetite and obesity research share at least some main features of human obesity and its comorbidities. Hence, even though no animal model replicates all aspects of "common" human obesity, animal models are imperative in studying the control of energy balance and reasons for its imbalance that may eventually lead to overt obesity. The most frequently used animal models are small rodents that may be based on mutations or manipulations of individual or several genes and on the exposure to obesogenic diets or other manipulations that predispose the animals to gaining or maintaining excessive weight. Characteristics include hyperphagia or changes in energy metabolism and at least in some models the frequent comorbidities of obesity, like hyperglycemia, insulin resistance, or diabetes-like syndromes. Some of the most frequently used animal models of obesity research involve animals with monogenic mutations of the leptin pathway which in fact are useful to study specific mechanistic aspects of eating controls, but typically do not recapitulate "common" obesity in the human population. Hence, this review will mention advantages and disadvantages of respective animal models in order to build a basis for the most appropriate use in biomedical research.
Collapse
Affiliation(s)
- Thomas A Lutz
- Institute of Veterinary Physiology, Vetsuisse Faculty University of Zurich, Zurich, Switzerland.
| |
Collapse
|
34
|
Chen L, Merkhan MM, Forsyth NR, Wu P. Chorionic and amniotic membrane-derived stem cells have distinct, and gestational diabetes mellitus independent, proliferative, differentiation, and immunomodulatory capacities. Stem Cell Res 2019; 40:101537. [PMID: 31422237 DOI: 10.1016/j.scr.2019.101537] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 06/25/2019] [Accepted: 08/12/2019] [Indexed: 12/19/2022] Open
Abstract
Placental membrane-derived mesenchymal stem cells (MSCs), with the advantages of being non-invasive and having fewer ethical issues, are a promising source for cell therapy. Gestational diabetes (GDM) alters the uterine environment and may affect the therapeutic potential of MSCs derived from placenta. Therefore, we evaluated the biological properties of amniotic (AMSCs) and chorionic membrane MSCs (CMSCs) from human GDM placenta in order to explore their therapeutic potential. In comparison of GDM-/Healthy- CMSCs and AMSCs, the immunophenotypes and typical stellate morphology of MSC were similar in CMSCs irrespective of disease state while the MSC morphology in GDM-AMSCs was less evident. GDM- and Healthy- CMSCs displayed an enhanced proliferation rate and tri-lineage differentiation capacity compared with AMSCs. Notably, GDM-CMSCs had a significantly increased adipogenic ability than Healthy-CMSCs accompanied by increased transcriptional responsiveness of PPARγ and ADIPOQ induction. The secretome effect of Healthy- and GDM- CMSCs/AMSCs by using conditioned media and coculture experiments, suggests that GDM- and Healthy- CMSCs provided an equivalent immunoregulatory effect on suppressing T-cells activation but a reduced effect of GDM-CMSCs on macrophage regulation. However, Healthy- and GDM- CMSCs displayed a superior immunomodulatory capacity in regulation of both T-cells and macrophages than AMSCs. In summary, we highlight the importance of the maternal GDM intrauterine environment during pregnancy and its impact on CMSCs/AMSCs proliferation ability, CMSCs adipogenic potential, and macrophage regulatory capacity.
Collapse
Affiliation(s)
- Liyun Chen
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, UK.
| | - Marwan M Merkhan
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, UK; College of Pharmacy, University of Mosul, Mosul, Iraq
| | - Nicholas R Forsyth
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, UK.
| | - Pensee Wu
- Guy Hilton Research Centre, School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent, UK; Academic Unit of Obstetrics and Gynaecology, University Hospital of North Midlands, Stoke-on-Trent, UK; Keele Cardiovascular Research Group, Institute for Applied Clinical Sciences and Centre for Prognosis Research, Institute of Primary Care and Health Sciences, Keele University, Stoke-on-Trent, UK.
| |
Collapse
|
35
|
Epigenetic regulation of POMC; implications for nutritional programming, obesity and metabolic disease. Front Neuroendocrinol 2019; 54:100773. [PMID: 31344387 DOI: 10.1016/j.yfrne.2019.100773] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 07/18/2019] [Accepted: 07/20/2019] [Indexed: 01/07/2023]
Abstract
Proopiomelanocortin (POMC) is a key mediator of satiety. Epigenetic marks such as DNA methylation may modulate POMC expression and provide a biological link between early life exposures and later phenotype. Animal studies suggest epigenetic marks at POMC are influenced by maternal energy excess and restriction, prenatal stress and Triclosan exposure. Postnatal factors including energy excess, folate, vitamin A, conjugated linoleic acid and leptin may also affect POMC methylation. Recent human studies suggest POMC DNA methylation is influenced by maternal nutrition in early pregnancy and associated with childhood and adult obesity. Studies in children propose a link between POMC DNA methylation and elevated lipids and insulin, independent of body habitus. This review brings together evidence from animal and human studies and suggests that POMC is sensitive to nutritional programming and is associated with a wide range of weight-related and metabolic outcomes.
Collapse
|
36
|
Clyburn C, Howe CA, Arnold AC, Lang CH, Travagli RA, Browning KN. Perinatal high-fat diet alters development of GABA A receptor subunits in dorsal motor nucleus of vagus. Am J Physiol Gastrointest Liver Physiol 2019; 317:G40-G50. [PMID: 31042399 PMCID: PMC6689732 DOI: 10.1152/ajpgi.00079.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/22/2019] [Accepted: 04/29/2019] [Indexed: 01/31/2023]
Abstract
Perinatal high-fat diet (pHFD) exposure increases the inhibition of dorsal motor nucleus of the vagus (DMV) neurons, potentially contributing to the dysregulation of gastric functions. The aim of this study was to test the hypothesis that pHFD increases the inhibition of DMV neurons by disrupting GABAA receptor subunit development. In vivo gastric recordings were made from adult anesthetized Sprague-Dawley rats fed a control or pHFD (14 or 60% kcal from fat, respectively) from embryonic day 13 (E13) to postnatal day 42 (P42), and response to brainstem microinjection of benzodiazepines was assessed. Whole cell patch clamp recordings from DMV neurons assessed the functional expression of GABAA α subunits, whereas mRNA and protein expression were measured via qPCR and Western blotting, respectively. pHFD decreased basal antrum and corpus motility, whereas brainstem microinjection of L838,417 (positive allosteric modulator of α2/3 subunit-containing GABAA receptors) produced a larger decrease in gastric tone and motility. GABAergic miniature inhibitory postsynaptic currents in pHFD DMV neurons were responsive to L838,417 throughout development, unlike control DMV neurons, which were responsive only at early postnatal timepoints. Brainstem mRNA and protein expression of the GABAA α1,2, and 3 subunits, however, did not differ between control and pHFD rats. This study suggests that pHFD exposure arrests the development of synaptic GABAA α2/3 receptor subunits on DMV neurons and that functional synaptic expression is maintained into adulthood, although cellular localization may differ. The tonic activation of slower GABAA α2/3 subunit-containing receptors implies that such developmental changes may contribute to the observed decreased gastric motility. NEW & NOTEWORTHY Vagal neurocircuits involved in the control of gastric functions, satiation, and food intake are subject to significant developmental regulation postnatally, with immature GABAA receptors expressing slower α2/3-subunits, whereas mature GABAA receptor express faster α1-subunits. After perinatal high-fat diet exposure, this developmental regulation of dorsal motor nucleus of the vagus (DMV) neurons is disrupted, increasing their tonic GABAergic inhibition, decreasing efferent output, and potentially decreasing gastric motility.
Collapse
Affiliation(s)
- Courtney Clyburn
- Department of Neural and Behavioral Sciences, Penn State College of Medicine , Hershey, Pennsylvania
| | - Caitlin A Howe
- Department of Neural and Behavioral Sciences, Penn State College of Medicine , Hershey, Pennsylvania
| | - Amy C Arnold
- Department of Neural and Behavioral Sciences, Penn State College of Medicine , Hershey, Pennsylvania
| | - Charles H Lang
- Department of Cellular and Molecular Physiology, Penn State College of Medicine , Hershey, Pennsylvania
| | - R Alberto Travagli
- Department of Neural and Behavioral Sciences, Penn State College of Medicine , Hershey, Pennsylvania
| | - Kirsteen N Browning
- Department of Neural and Behavioral Sciences, Penn State College of Medicine , Hershey, Pennsylvania
| |
Collapse
|
37
|
Rosin JM, Kurrasch DM. Emerging roles for hypothalamic microglia as regulators of physiological homeostasis. Front Neuroendocrinol 2019; 54:100748. [PMID: 31059719 DOI: 10.1016/j.yfrne.2019.100748] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/24/2019] [Accepted: 05/02/2019] [Indexed: 01/22/2023]
Abstract
The hypothalamus is a crucial brain region that responds to external stressors and functions to maintain physiological homeostatic processes, such as core body temperature and energy balance. The hypothalamus regulates homeostasis by producing hormones that thereby influence the production of other hormones that then control the internal milieu of the body. Microglia are resident macrophages and phagocytic immune cells of the central nervous system (CNS), classically known for surveying the brain's environment, responding to neural insults, and disposing of cellular debris. Recent evidence has shown that microglia are also responsive to external stressors and can influence both the development and function of the hypothalamus in a sex-dependent manner. This emerging microglia-hypothalamic interaction raises the intriguing notion that microglia might play an unappreciated role in hypothalamic control of physiological homeostasis. In this review, we briefly outline how the hypothalamus regulates physiological homeostasis and then describe how this literature overlaps with our understanding of microglia's role in the CNS. We also outline the current literature demonstrating how microglia loss or activation affects the hypothalamus, and ultimately homeostasis. We conclude by proposing how microglia could be key regulators of homeostatic processes by sensing cues external to the CNS and transmitting them through the hypothalamus.
Collapse
Affiliation(s)
- Jessica M Rosin
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Deborah M Kurrasch
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada; Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
38
|
Cordner ZA, Khambadkone SG, Boersma GJ, Song L, Summers TN, Moran TH, Tamashiro KLK. Maternal high-fat diet results in cognitive impairment and hippocampal gene expression changes in rat offspring. Exp Neurol 2019; 318:92-100. [PMID: 31051155 DOI: 10.1016/j.expneurol.2019.04.018] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/13/2019] [Accepted: 04/29/2019] [Indexed: 01/01/2023]
Abstract
Consumption of a high-fat diet has long been known to increase risk for obesity, diabetes, and the metabolic syndrome. Further evidence strongly suggests that these same metabolic disorders are associated with an increased risk of cognitive impairment later in life. Now faced with an expanding global burden of obesity and increasing prevalence of dementia due to an aging population, understanding the effects of high-fat diet consumption on cognition is of increasingly critical importance. Further, the developmental origins of many adult onset neuropsychiatric disorders have become increasingly clear, indicating a need to investigate effects of various risk factors, including diet, across the lifespan. Here, we use a rat model to assess the effects of maternal diet during pregnancy and lactation on cognition and hippocampal gene expression of offspring. Behaviorally, adult male offspring of high-fat fed dams had impaired object recognition memory and impaired spatial memory compared to offspring of chow-fed dams. In hippocampus, we found decreased expression of Insr, Lepr, and Slc2a1 (GLUT1) among offspring of high-fat fed dams at postnatal day 21. The decreased expression of Insr and Lepr persisted at postnatal day 150. Together, these data provide additional evidence to suggest that maternal exposure to high-fat diet during pregnancy and lactation can have lasting effects on the brain, behavior, and cognition on adult offspring.
Collapse
Affiliation(s)
- Zachary A Cordner
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA
| | - Seva G Khambadkone
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA; Cellular & Molecular Medicine Graduate Program, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA
| | - Gretha J Boersma
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA
| | - Lin Song
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA
| | - Tyler N Summers
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA
| | - Timothy H Moran
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA; Cellular & Molecular Medicine Graduate Program, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA
| | - Kellie L K Tamashiro
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA; Cellular & Molecular Medicine Graduate Program, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA.
| |
Collapse
|
39
|
Argaev Frenkel L, Rozenfeld H, Rozenberg K, Sampson SR, Rosenzweig T. N-Acetyl-l-Cysteine Supplement in Early Life or Adulthood Reduces Progression of Diabetes in Nonobese Diabetic Mice. Curr Dev Nutr 2019; 3:nzy097. [PMID: 30993256 PMCID: PMC6459986 DOI: 10.1093/cdn/nzy097] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 11/26/2018] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Oxidative stress contributes to the pathologic process leading to the development, progression, and complications of type 1 diabetes (T1D). OBJECTIVE The aim of this study was to investigate the effect of the antioxidant N-acetyl-l-cysteine (NAC), supplemented during early life or adulthood on the development of T1D. METHODS NAC was administered to nonobese diabetic (NOD) female mice during pregnancy and lactation, and the development of diabetes was followed in offspring. In an additional set of experiments, offspring of untreated mice were given NAC during adulthood, and the development of T1D was followed. Morbidity rate, insulitis and serum cytokines were measured in the 2 sets of experiments. In addition, markers of oxidative stress, glutathione, lipid peroxidation, total antioxidant capacity and activity of antioxidant enzymes, were followed. RESULTS Morbidity rate was reduced in both treatment protocols. A decrease in interferon γ, tumor necrosis factor α, interleukin 1α, and other type 1 diabetes-associated proinflammatory cytokines was found in mice supplemented with NAC in adulthood or during early life compared with control NOD mice. The severity of insulitis was higher in control NOD mice than in treated groups. NAC administration significantly reduced oxidative stress, as determined by reduced lipid peroxidation and increased total antioxidant capacity in serum and pancreas of mice treated in early life or in adulthood and increased pancreatic glutathione when administrated in adulthood. The activity of antioxidant enzymes was not affected in mice given NAC in adulthood, whereas an increase in the activity of superoxide dismutase and catalase was demonstrated in the pancreas of their offspring. CONCLUSION NAC decreased morbidity of NOD mice by attenuating the immune response, presumably by eliminating oxidative stress, and might be beneficial in reducing morbidity rates of T1D in high-risk individuals.
Collapse
Affiliation(s)
- Lital Argaev Frenkel
- Department of Molecular Biology, School of Health Sciences, Ariel University, Ariel, Israel
- Department of Nutrition Sciences, School of Health Sciences, Ariel University, Ariel, Israel
| | - Hava Rozenfeld
- Department of Molecular Biology, School of Health Sciences, Ariel University, Ariel, Israel
- Department of Nutrition Sciences, School of Health Sciences, Ariel University, Ariel, Israel
| | - Konstantin Rozenberg
- Department of Molecular Biology, School of Health Sciences, Ariel University, Ariel, Israel
- Department of Nutrition Sciences, School of Health Sciences, Ariel University, Ariel, Israel
| | - Sanford R Sampson
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Tovit Rosenzweig
- Department of Molecular Biology, School of Health Sciences, Ariel University, Ariel, Israel
- Department of Nutrition Sciences, School of Health Sciences, Ariel University, Ariel, Israel
| |
Collapse
|
40
|
McMenamin CA, Clyburn C, Browning KN. High-Fat Diet During the Perinatal Period Induces Loss of Myenteric Nitrergic Neurons and Increases Enteric Glial Density, Prior to the Development of Obesity. Neuroscience 2019; 393:369-380. [PMID: 30454864 DOI: 10.1016/j.neuroscience.2018.09.033] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 09/19/2018] [Accepted: 09/24/2018] [Indexed: 02/07/2023]
Abstract
Diet-induced obesity induces peripheral inflammation accompanied by a loss of myenteric neurons. Few studies, however, have investigated the effects of a high-fat diet (HFD) on either the development of myenteric neurons or prior to the occurrence of obesity. The present study assessed the effects of maternal HFD on the density and neurochemical phenotype of myenteric ganglia in the upper gastrointestinal tract. Sprague-Dawley rats were fed either a control or HFD (14% or 60% kcal from fat, respectively) from embryonic day 13; the fundus, corpus and duodenum were fixed thereafter at postnatal 2, 4, 6 and 12 weeks of age for subsequent immunohistochemical studies. While myenteric ganglion size did not differ throughout the study, HFD exposure decreased the number of nitrergic neurons by 6 weeks of age in all regions. This decrease was accompanied by a loss of PGP-immunoreactive neurons, suggesting a decline in myenteric neuronal number. HFD also increased myenteric plexus glial cell density in all regions by 4 weeks of age. These changes occurred in the absence of an increase in serum or gastric inflammatory markers. The present study suggests that exposure to a HFD during the perinatal time period results in glial proliferation and loss of inhibitory nitrergic neurons prior to the onset of obesity, suggesting that dietary alterations may affect gastrointestinal functions independently of increased adiposity or glycemic dysregulation.
Collapse
Affiliation(s)
- Caitlin A McMenamin
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA 17033, United States
| | - Courtney Clyburn
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA 17033, United States
| | - Kirsteen N Browning
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, PA 17033, United States.
| |
Collapse
|
41
|
Enteric Neuronal Degeneration: Is it Due to Your Mother's Diet? Neuroscience 2019; 393:366-368. [PMID: 30454863 DOI: 10.1016/j.neuroscience.2018.10.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 10/08/2018] [Indexed: 11/20/2022]
|
42
|
Nyakudya TT, Mukwevho E, Erlwanger KH. The protective effect of neonatal oral administration of oleanolic acid against the subsequent development of fructose-induced metabolic dysfunction in male and female rats. Nutr Metab (Lond) 2018; 15:82. [PMID: 30479649 PMCID: PMC6245863 DOI: 10.1186/s12986-018-0314-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 10/22/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Consumption of fructose-rich diets has been implicated in the increasing global prevalence of metabolic syndrome (MetS). Interventions during periods of early ontogenic developmental plasticity can cause epigenetic changes which program metabolism for positive or negative health benefits later in life. The phytochemical oleanolic acid (OA) possesses anti-diabetic and anti-obesity effects. We investigated the potential protective effects of neonatal administration of OA on the subsequent development of high fructose diet-induced metabolic dysfunction in rats. METHOD Male and female (N = 112) suckling rats were randomly assigned to four groups and administered orally: distilled water (DW), oleanolic acid (OA; 60 mg/kg), high-fructose solution (HF; 20% w/v) or OA + HF for 7 days. The rats were weaned onto normal commercial rat chow up to day 55. From day 56, half of the rats in each treatment group were continued on plain water and the rest on a high fructose solution as drinking fluid for 8 weeks. On day 110, the rats were subjected to an oral glucose tolerance test and then euthanased on day 112. Tissue and blood samples were collected to determine the effects of the treatments on visceral fat pad mass, fasting plasma levels of cholesterol, insulin, glucose, triglycerides, insulin resistance (HOMA-IR) and glucose tolerance. RESULTS Rats which consumed fructose as neonates and then later as adults (HF + F) and those which consumed fructose only in adulthood (DW + F) had significant increases in terminal body mass (females only), visceral fat mass (males and females), serum triglycerides (females only), epididymal fat (males only), fasting plasma glucose (males and females), impaired glucose metabolism (females only), β-cell dysfunction and insulin resistance (males and females) compared to the other treatment groups (P < 0.05). There were no differences in fasting serum cholesterol levels across all treatment groups in both male and female rats (P > 0.05). CONCLUSION We conclude that neonatal oral administration of OA during the critical window of developmental plasticity protected against the development of health outcomes associated with fructose-induced metabolic disorders in the rats.
Collapse
Affiliation(s)
- Trevor T. Nyakudya
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193 South Africa
- Department of Human Anatomy and Physiology, Faculty of Health Sciences, University of Johannesburg, Doornfontein, Johannesburg, 2028 South Africa
| | - Emmanuel Mukwevho
- Department of Biochemistry, Faculty of Natural Sciences & Agriculture, North West University, Mafikeng, Mmabatho, 2735 South Africa
| | - Kennedy H. Erlwanger
- School of Physiology, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, 2193 South Africa
| |
Collapse
|
43
|
Kosińska M, Sierzputowska-Pieczara M, Gadzinowski J, Cygan D, Szpecht D. Percentile charts of twin birthweight. Pediatr Int 2018; 60:948-953. [PMID: 30074674 DOI: 10.1111/ped.13669] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 06/28/2018] [Accepted: 08/01/2018] [Indexed: 11/28/2022]
Abstract
BACKGROUND The birthweight of multiples is naturally lower than that of singletons. Given that the incidence of twin pregnancy has risen in recent years, it seems reasonable to create standards of birthweight separately for twins. This could help in the objective assessment of small and large for gestational age twin newborns. The main goal of this study was therefore to construct and present up-to-date birthweight references. METHODS The present percentile charts for twins are based on a cohort retrospective study of 757 pairs of twins (767 boys and 709 girls) born between weeks 25 and 39 of gestation. Mean and standard deviation were calculated for the subsequent weeks of gestation. Percentiles were read for the subsequent gestational age. The obtained curves were smoothed with a fifth-degree polynomial function. The significance of differences between the 50th percentile values for twins and singletons was estimated using median test. RESULTS In both sexes, a continuous observable trend occurs of a significantly lower average birthweight for twins. Differences increase with increasing gestational age and are greater in girls. The estimated 50th percentile for twins was greater than the estimated 10th percentile for singletons. This supports the notion of discordant growth as a physiological adaptation that promotes maturity. CONCLUSIONS Percentile charts for singletons are not applicable for twins. This indicates the importance of applying separate percentile charts for twins, enabling objective evaluation of their health status and identifying deviations from normality.
Collapse
Affiliation(s)
- Magdalena Kosińska
- Department of Human Developmental Biology, Institute of Anthropology, Adam Mickiewicz University, Poznań, Poland
| | | | - Janusz Gadzinowski
- Department of Neonatology, University of Medical Sciences, Poznań, Poland
| | | | - Dawid Szpecht
- Department of Neonatology, University of Medical Sciences, Poznań, Poland
| |
Collapse
|
44
|
Rosin JM, Kurrasch DM. Bisphenol A and microglia: could microglia be responsive to this environmental contaminant during neural development? Am J Physiol Endocrinol Metab 2018; 315:E279-E285. [PMID: 29812986 DOI: 10.1152/ajpendo.00443.2017] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
There is a growing interest in the functional role of microglia in the developing brain. In our laboratory, we have become particularly intrigued as to whether fetal microglia in the embryonic brain are susceptible to maternal challenges in utero (e.g., maternal infection, stress) and, if so, whether their precocious activation could then adversely influence brain development. One such challenge that is newly arising in this field is whether microglia might be downstream targets to endocrine-disrupting chemicals, such as the plasticizer bisphenol A (BPA), which functions in part by mimicking estrogen structure and function. A growing body of evidence demonstrates that gestational exposure to BPA has adverse effects on brain development, although the exact mechanisms are still emerging. Given that microglia express estrogen receptors and steroid-producing enzymes, microglia might be an unappreciated target of BPA. Mechanistically, we propose that BPA binding to estrogen receptors within microglia initiates transcription of downstream target genes, which then leads to activation of microglia that can then perhaps adversely influence brain development. Here, we first briefly outline the current understanding of how microglia may influence brain development and then describe how this literature overlaps with our understanding of BPA's effects during similar time points. We also outline the current literature demonstrating that BPA exposure affects microglia. We conclude by discussing our thoughts on the mechanisms through which exposure to BPA could disrupt normal microglia functions, ultimately affecting brain development that could potentially lead to lasting behavioral effects and perhaps even neuroendocrine diseases such as obesity.
Collapse
Affiliation(s)
- Jessica M Rosin
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary , Calgary, Alberta , Canada
- Alberta Children's Hospital Research Institute, University of Calgary , Calgary, Alberta , Canada
- Hotchkiss Brain Institute, University of Calgary , Calgary, Alberta , Canada
| | - Deborah M Kurrasch
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary , Calgary, Alberta , Canada
- Alberta Children's Hospital Research Institute, University of Calgary , Calgary, Alberta , Canada
- Hotchkiss Brain Institute, University of Calgary , Calgary, Alberta , Canada
| |
Collapse
|
45
|
Mao J, Pennington KA, Talton OO, Schulz LC, Sutovsky M, Lin Y, Sutovsky P. In Utero and Postnatal Exposure to High Fat, High Sucrose Diet Suppressed Testis Apoptosis and Reduced Sperm Count. Sci Rep 2018; 8:7622. [PMID: 29769570 PMCID: PMC5955896 DOI: 10.1038/s41598-018-25950-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 05/02/2018] [Indexed: 01/11/2023] Open
Abstract
Obesity affects male fertility and maternal diabetes affects the offspring sperm epigenome. However, the effects of in utero exposure to maternal glucose intolerance in combination with postnatal high fat, high sucrose (HFHS) diet consumption on offspring spermatogenesis is not clear. The present study was designed to test these effects. One week before and during pregnancy, dams were fed either control or HFHS diet to induce gestational glucose intolerance, and returned to standard diet during lactation. Male offspring from each maternal group were split into control and HFHS-fed groups for eight weeks prior to sacrifice at 11, 19 or 31 weeks of age, and reproductive tissues were harvested for analysis of testicular germ cell apoptosis and sperm output. Postnatal HFHS diet suppressed spermatogonia apoptosis in all age groups and maternal HFHS diet reduced testosterone levels at 11 weeks. At 31 weeks of age, the postnatal HFHS diet increased body weight, and reduced epididymis weight and sperm count. The combination of in utero and postnatal exposure impacted sperm counts most significantly. In summary, HFHS diet during pregnancy puts male offspring at greater risk of infertility, particularly when combined with postnatal high fat diet feeding.
Collapse
Affiliation(s)
- Jiude Mao
- Division of Animal Sciences, University of Missouri, Columbia, MO, 65211, USA.
| | - Kathleen A Pennington
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, MO, 65211, USA.,Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX, USA
| | - Omonseigho O Talton
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, MO, 65211, USA
| | - Laura C Schulz
- Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, MO, 65211, USA
| | - Miriam Sutovsky
- Division of Animal Sciences, University of Missouri, Columbia, MO, 65211, USA
| | - Yan Lin
- Key Laboratory for Animal Disease Resistance, the Ministry of Nutrition of Education of China, Institute of Animal Nutrition, Sichuan Agricultural University, Ya'an, 625001, China
| | - Peter Sutovsky
- Division of Animal Sciences, University of Missouri, Columbia, MO, 65211, USA.,Department of Obstetrics, Gynecology and Women's Health, University of Missouri, Columbia, MO, 65211, USA
| |
Collapse
|
46
|
Altered expression of PGC-1α and PDX1 and their methylation status are associated with fetal glucose metabolism in gestational diabetes mellitus. Biochem Biophys Res Commun 2018; 501:300-306. [PMID: 29730292 DOI: 10.1016/j.bbrc.2018.05.010] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 05/02/2018] [Indexed: 11/23/2022]
Abstract
PURPOSE To investigate the effect of gestational diabetes mellitus (GDM) on the expression and methylation of PGC-1α and PDX1 in placenta and their effects on fetal glucose metabolism. METHODS 20 cases of full-term placenta without pregnancy complications and umbilical cord abnormalities and 20 cases of GDM group were collected. DNA and RNA were isolated from samples of tissue collected from the fetal side of the placenta immediately after delivery. DNA methylation was quantified at 7 CpG sites within the PGC-1α and PDX1 genes using PCR amplification of bisulfite treated DNA and subsequent DNA sequencing. PGC-1α and PDX1 mRNA levels were measured by reverse transcription-quantitative PCR (RT-qPCR). Meanwhile, the placental insulin, blood glucose and HbA1c levels were determined. RESULTS The fetus birth weight and placental weight in GDM group were significantly higher than those in control group (P < 0.05). Insulin, HbA1c and blood glucose levels in GDM group were significantly higher than those in control group (P < 0.01). Insulin content was positively correlated with newborn birth weight and placental weight while HbA1c and blood glucose were positively correlated with insulin concentration (r = 0.92, P < 0.01, r = 0.85, P < 0.01). The levels of PGC-1α and PDX1 mRNA were lower in the GDM group compared to the control group. The methylation level of PGC-1α gene was higher in the GDM group compared to the control group (P < 0.05). Blood glucose was negatively correlated with the expression of PGC-1α and PDX1 mRNA in the placenta (r = -0.42, P < 0.01, r = -0.49, P < 0.01). CONCLUSION The changes of epigenetic modification of PGC-1α gene in pregnant women with gestational diabetes mellitus may be a mechanism of abnormal glucose metabolism in offspring.
Collapse
|
47
|
Clyburn C, Travagli RA, Browning KN. Acute high-fat diet upregulates glutamatergic signaling in the dorsal motor nucleus of the vagus. Am J Physiol Gastrointest Liver Physiol 2018; 314:G623-G634. [PMID: 29368945 PMCID: PMC6008060 DOI: 10.1152/ajpgi.00395.2017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Obesity is associated with dysregulation of vagal neurocircuits controlling gastric functions, including food intake and energy balance. In the short term, however, caloric intake is regulated homeostatically although the precise mechanisms responsible are unknown. The present study examined the effects of acute high-fat diet (HFD) on glutamatergic neurotransmission within central vagal neurocircuits and its effects on gastric motility. Sprague-Dawley rats were fed a control or HFD diet (14% or 60% kcal from fat, respectively) for 3-5 days. Whole cell patch-clamp recordings and brainstem application of antagonists were used to assess the effects of acute HFD on glutamatergic transmission to dorsal motor nucleus of the vagus (DMV) neurons and subsequent alterations in gastric tone and motility. After becoming hyperphagic initially, caloric balance was restored after 3 days following HFD exposure. In control rats, the non- N-methyl-d-aspartate (NMDA) receptor antagonist, 6,7-dinitroquinoxaline-2,3-dione (DNQX), but not the NMDA receptor antagonist, amino-5-phosphonopentanoate (AP5), significantly decreased excitatory synaptic currents and action potential firing rate in gastric-projecting DMV neurons. In contrast, both AP5 and DNQX decreased excitatory synaptic transmission and action potential firing in acute HFD neurons. When microinjected into the brainstem, AP5, but not DNQX, decreased gastric motility and tone in acute HFD rats only. These results suggest that acute HFD upregulates NMDA receptor-mediated currents, increasing DMV neuronal excitability and activating the vagal efferent cholinergic pathway, thus increasing gastric tone and motility. Although such neuroplasticity may be a persistent adaptation to the initial exposure to HFD, it may also be an important mechanism in homeostatic regulation of energy balance. NEW & NOTEWORTHY Vagal neurocircuits are critical to the regulation of gastric functions, including satiation and food intake. Acute high-fat diet upregulates glutamatergic signaling within central vagal neurocircuits via activation of N-methyl-d-aspartate receptors, increasing vagal efferent drive to the stomach. Although it is possible that such neuroplasticity is a persistent adaptation to initial exposure to the high-fat diet, it may also play a role in the homeostatic control of feeding.
Collapse
Affiliation(s)
- Courtney Clyburn
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, Pennsylvania
| | - R. Alberto Travagli
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, Pennsylvania
| | - Kirsteen N. Browning
- Department of Neural and Behavioral Sciences, Penn State College of Medicine, Hershey, Pennsylvania
| |
Collapse
|
48
|
Rudolph MC, Jackman MR, Presby DM, Houck JA, Webb PG, Johnson GC, Soderborg TK, de la Houssaye BA, Yang IV, Friedman JE, MacLean PS. Low Neonatal Plasma n-6/n-3 PUFA Ratios Regulate Offspring Adipogenic Potential and Condition Adult Obesity Resistance. Diabetes 2018; 67:651-661. [PMID: 29138256 PMCID: PMC5860857 DOI: 10.2337/db17-0890] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 11/06/2017] [Indexed: 01/08/2023]
Abstract
Adipose tissue expansion progresses rapidly during postnatal life, influenced by both prenatal maternal factors and postnatal developmental cues. The ratio of omega-6 (n-6) relative to n-3 polyunsaturated fatty acids (PUFAs) is believed to regulate perinatal adipogenesis, but the cellular mechanisms and long-term effects are not well understood. We lowered the fetal and postnatal n-6/n-3 PUFA ratio exposure in wild-type offspring under standard maternal dietary fat amounts to test the effects of low n-6/n-3 ratios on offspring adipogenesis and adipogenic potential. Relative to wild-type pups receiving high perinatal n-6/n-3 ratios, subcutaneous adipose tissue in 14-day-old wild-type pups receiving low n-6/n-3 ratios had more adipocytes that were smaller in size; decreased Pparγ2, Fabp4, and Plin1; several lipid metabolism mRNAs; coincident hypermethylation of the PPARγ2 proximal promoter; and elevated circulating adiponectin. As adults, offspring that received low perinatal n-6/n-3 ratios were diet-induced obesity (DIO) resistant and had a lower positive energy balance and energy intake, greater lipid fuel preference and non-resting energy expenditure, one-half the body fat, and better glucose clearance. Together, the findings support a model in which low early-life n-6/n-3 ratios remodel adipose morphology to increase circulating adiponectin, resulting in a persistent adult phenotype with improved metabolic flexibility that prevents DIO.
Collapse
Affiliation(s)
- Michael C Rudolph
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine, Aurora, CO
| | - Matthew R Jackman
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine, Aurora, CO
| | - David M Presby
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine, Aurora, CO
| | - Julie A Houck
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine, Aurora, CO
| | - Patricia G Webb
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine, Aurora, CO
| | - Ginger C Johnson
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine, Aurora, CO
| | - Taylor K Soderborg
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO
| | - Becky A de la Houssaye
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO
| | - Ivana V Yang
- Division of Biomedical Informatics and Personalized Medicine, University of Colorado School of Medicine, Aurora, CO
| | - Jacob E Friedman
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine, Aurora, CO
- Department of Pediatrics, Section of Neonatology, University of Colorado School of Medicine, Aurora, CO
| | - Paul S MacLean
- Division of Endocrinology, Metabolism and Diabetes, University of Colorado School of Medicine, Aurora, CO
| |
Collapse
|
49
|
Miyaguti NADS, de Oliveira SCP, Gomes-Marcondes MCC. Maternal nutritional supplementation with fish oil and/or leucine improves hepatic function and antioxidant defenses, and minimizes cachexia indexes in Walker-256 tumor-bearing rats offspring. Nutr Res 2018; 51:29-39. [DOI: 10.1016/j.nutres.2017.12.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 10/30/2017] [Accepted: 12/08/2017] [Indexed: 12/12/2022]
|
50
|
McMenamin CA, Travagli RA, Browning KN. Perinatal high fat diet increases inhibition of dorsal motor nucleus of the vagus neurons regulating gastric functions. Neurogastroenterol Motil 2018; 30:10.1111/nmo.13150. [PMID: 28762595 PMCID: PMC5739938 DOI: 10.1111/nmo.13150] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 06/07/2017] [Indexed: 01/02/2023]
Abstract
BACKGROUND Previous studies suggest an increased inhibition of dorsal motor nucleus of the vagus (DMV) neurons following exposure to a perinatal high fat diet (PNHFD); the underlying neural mechanisms, however, remain unknown. This study assessed the effects of PNHFD on inhibitory synaptic inputs to DMV neurons and the vagally dependent control of gastric tone and motility. METHODS Whole-cell patch clamp recordings were made from DMV neurons in thin brainstem slices from Sprague-Dawley rats fed either a control diet or HFD (14 or 60% kcal from fat, respectively) from embryonic day 13 onwards; gastric tone and motility were recorded in in vivo anesthetized rats. KEY RESULTS The non-selective GABAA antagonist, BIC (10 μmol L-1 ), induced comparable inward currents in PNHFD and control DMV neurons, but a larger current in PNHFD neurons at higher concentrations (50 μmol L-1 ). Differences were not apparent in neuronal responses to the phasic GABAA antagonist, gabazine (GBZ), the extrasynaptic GABAA agonist, THIP, the GABA transport blocker, nipecotic acid, or the gliotoxin, fluoroacetate, suggesting that PNHFD altered inhibitory transmission but not GABAA receptor density or function, GABA uptake or glial modulation of synaptic strength. Similarly, the increase in gastric motility and tone following brainstem microinjection of low doses of BIC (1-10 pmoles) and GBZ (0.01-0.1 pmoles) were unchanged in PNHFD rats while higher doses of BIC (25 pmoles) induced a significantly larger increase in gastric tone compared to control. CONCLUSIONS AND INFERENCES These studies suggest that exposure to PNHFD increases the tonic inhibition of DMV neurons, possibly contributing to dysregulated vagal control of gastric functions.
Collapse
Affiliation(s)
| | | | - Kirsteen N. Browning
- Address for correspondence: Kirsteen N Browning, PhD, Department of Neural and Behavioral Science, Penn State College of Medicine, 500 University Drive, MC H109, Hershey, PA 17033, Tel: 717 531 8267,
| |
Collapse
|