1
|
Hill KB, Mullen GP, Nagareddy PR, Zimmerman KA, Rudolph MC. Key questions and gaps in understanding adipose tissue macrophages and early-life metabolic programming. Am J Physiol Endocrinol Metab 2024; 327:E478-E497. [PMID: 39171752 PMCID: PMC11482221 DOI: 10.1152/ajpendo.00140.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/08/2024] [Accepted: 08/20/2024] [Indexed: 08/23/2024]
Abstract
The global obesity epidemic, with its associated comorbidities and increased risk of early mortality, underscores the urgent need for enhancing our understanding of the origins of this complex disease. It is increasingly clear that metabolism is programmed early in life and that metabolic programming can have life-long health consequences. As a critical metabolic organ sensitive to early-life stimuli, proper development of adipose tissue (AT) is crucial for life-long energy homeostasis. Early-life nutrients, especially fatty acids (FAs), significantly influence the programming of AT and shape its function and metabolism. Of growing interest are the dynamic responses during pre- and postnatal development to proinflammatory omega-6 (n6) and anti-inflammatory omega-3 (n3) FA exposures in AT. In the US maternal diet, the ratio of "pro-inflammatory" n6- to "anti-inflammatory" n3-FAs has grown dramatically due to the greater prevalence of n6-FAs. Notably, AT macrophages (ATMs) form a significant population within adipose stromal cells, playing not only an instrumental role in AT formation and maintenance but also acting as key mediators of cell-to-cell lipid and cytokine signaling. Despite rapid advances in ATM and immunometabolism fields, research has focused on responses to obesogenic diets and during adulthood. Consequently, there is a significant gap in identifying the mechanisms contributing metabolic health, especially regarding lipid exposures during the establishment of ATM physiology. Our review highlights the current understanding of ATM diversity, their critical role in AT, their potential role in early-life metabolic programming, and the broader implications for metabolism and health.
Collapse
Affiliation(s)
- Kaitlyn B Hill
- Department of Biochemistry and Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Gregory P Mullen
- Department of Biochemistry and Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Prabhakara R Nagareddy
- Department of Internal Medicine, Cardiovascular Section, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Kurt A Zimmerman
- Department of Internal Medicine, Division of Nephrology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| | - Michael C Rudolph
- Department of Biochemistry and Physiology, Harold Hamm Diabetes Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States
| |
Collapse
|
2
|
Röszer T. MicroRNA Profile of Mouse Adipocyte-Derived Extracellular Vesicles. Cells 2024; 13:1298. [PMID: 39120327 PMCID: PMC11311276 DOI: 10.3390/cells13151298] [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: 07/08/2024] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
The post-transcriptional control of gene expression is a complex and evolving field in adipocyte biology, with the premise that the delivery of microRNA (miRNA) species to the obese adipose tissue may facilitate weight loss. Cells shed extracellular vesicles (EVs) that may deliver miRNAs as intercellular messengers. However, we know little about the miRNA profile of EVs secreted by adipocytes during postnatal development. Here, we defined the miRNA cargo of EVs secreted by mouse adipocytes in two distinct phases of development: on postnatal day 6, when adipocytes are lipolytic and thermogenic, and on postnatal day 56, when adipocytes have active lipogenesis. EVs were collected from cell culture supernatants, and their miRNA profile was defined by small RNA sequencing. The most abundant miRNA of mouse adipocyte-derived EVs was mmu-miR-148a-3p. Adipocyte EVs on postnatal day 6 were hallmarked with mmu-miR-98-5p, and some miRNAs were specific to this developmental stage, such as mmu-miR-466i-5p and 12 novel miRNAs. Adipocytes on postnatal day 56 secreted mmu-miR-365-3p, and 16 miRNAs were specific to this developmental stage. The miRNA cargo of adipocyte EVs targeted gene networks of cell proliferation, insulin signaling, interferon response, thermogenesis, and lipogenesis. We provided here a database of miRNAs secreted by developing mouse adipocytes, which may be a tool for further studies on the regulation of gene networks that control mouse adipocyte development.
Collapse
Affiliation(s)
- Tamás Röszer
- Department of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
3
|
Hayasaka K. Pathogenesis and Management of Citrin Deficiency. Intern Med 2024; 63:1977-1986. [PMID: 37952953 PMCID: PMC11309867 DOI: 10.2169/internalmedicine.2595-23] [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: 07/10/2023] [Accepted: 09/22/2023] [Indexed: 11/14/2023] Open
Abstract
Citrin deficiency (CD) is a hereditary disorder caused by SLC25A13 mutations that manifests as neonatal intrahepatic cholestasis caused by CD (NICCD), failure to thrive and dyslipidemia caused by CD (FTTDCD), and adult-onset type 2 citrullinemia (CTLN2). Citrin, an aspartate-glutamate carrier primarily expressed in the liver, is a component of the malate-aspartate shuttle, which is essential for glycolysis. Citrin-deficient hepatocytes have primary defects in glycolysis and de novo lipogenesis and exhibit secondarily downregulated PPARα, leading to impaired β-oxidation. They are unable to utilize glucose and free fatty acids as energy sources, resulting in energy deficiencies. Medium-chain triglyceride (MCT) supplements are effective for treating CD by providing energy to hepatocytes, increasing lipogenesis, and activating the malate-citrate shuttle. However, patients with CD often exhibit growth impairment and irreversible brain and/or liver damage. To improve the quality of life and prevent irreversible damage, MCT supplementation with a diet containing minimal carbohydrates is recommended promptly after the diagnosis.
Collapse
Affiliation(s)
- Kiyoshi Hayasaka
- Department of Pediatrics, Yamagata University School of Medicine, Japan
| |
Collapse
|
4
|
Bai Y, Li J, Wei Y, Chen Z, Liu Z, Guo D, Jia X, Niu Y, Shi B, Zhang X, Zhao Z, Hu J, Han X, Wang J, Liu X, Li S. Proteome Analysis Related to Unsaturated Fatty Acid Synthesis by Interfering with Bovine Adipocyte ACSL1 Gene. Antioxidants (Basel) 2024; 13:641. [PMID: 38929080 PMCID: PMC11200461 DOI: 10.3390/antiox13060641] [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: 04/27/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
Unsaturated fatty acids (UFAs) in beef play a vital role in promoting human health. Long-chain fatty acyl-CoA synthase 1 (ACSL1) is a crucial gene for UFA synthesis in bovine adipocytes. To investigate the protein expression profile during UFA synthesis, we performed a proteomic analysis of bovine adipocytes by RNA interference and non-interference with ACSL1 using label-free techniques. A total of 3558 proteins were identified in both the NC and si-treated groups, of which 1428 were differentially expressed proteins (DEPs; fold change ≥ 1.2 or ≤ 0.83 and p-value < 0.05). The enrichment analysis of the DEPs revealed signaling pathways related to UFA synthesis or metabolism, including cAMP, oxytocin, fatty acid degradation, glycerol metabolism, insulin, and the regulation of lipolysis in adipocytes (p-value < 0.05). Furthermore, based on the enrichment analysis of the DEPs, we screened 50 DEPs that potentially influence the synthesis of UFAs and constructed an interaction network. Moreover, by integrating our previously published transcriptome data, this study established a regulatory network involving differentially expressed long non-coding RNAs (DELs), highlighting 21 DEPs and 13 DELs as key genes involved in UFA synthesis. These findings present potential candidate genes for further investigation into the molecular mechanisms underlying UFA synthesis in bovines, thereby offering insights to enhance the quality of beef and contribute to consumer health in future studies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Zhidong Zhao
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Y.B.); (J.L.); (Y.W.); (Z.C.); (Z.L.); (D.G.); (X.J.); (Y.N.); (B.S.); (X.Z.); (X.H.); (J.W.); (X.L.); (S.L.)
| | - Jiang Hu
- Gansu Key Laboratory of Herbivorous Animal Biotechnology, College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (Y.B.); (J.L.); (Y.W.); (Z.C.); (Z.L.); (D.G.); (X.J.); (Y.N.); (B.S.); (X.Z.); (X.H.); (J.W.); (X.L.); (S.L.)
| | | | | | | | | |
Collapse
|
5
|
Parenti M, Schmidt RJ, Tancredi DJ, Hertz-Picciotto I, Walker CK, Slupsky CM. Neurodevelopment and Metabolism in the Maternal-Placental-Fetal Unit. JAMA Netw Open 2024; 7:e2413399. [PMID: 38805224 PMCID: PMC11134213 DOI: 10.1001/jamanetworkopen.2024.13399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/26/2024] [Indexed: 05/29/2024] Open
Abstract
Importance Disturbances in maternal, placental, and fetal metabolism are associated with developmental outcomes. Associations of maternal, placental, and fetal metabolism with subsequent neurodevelopmental outcomes in the child are understudied. Objective To investigate the metabolic associations within the maternal-placental-fetal unit and subsequent neurodevelopmental outcomes in younger siblings of children with autism spectrum disorder (ASD). Design, Setting, and Participants This cohort study was conducted within a subset of the Markers of Autism Risk in Babies, Learning Early Signs (MARBLES) cohort. MARBLES is a prospective birth cohort of younger siblings of children with ASD assessed for neurodevelopmental outcomes at approximately age 36 months. Participants in MARBLES were recruited through the UC Davis MIND Institute. This subset of the MARBLES cohort included younger siblings born between 2009 and 2015. Maternal third trimester serum, placental tissue, and umbilical cord serum samples were collected from participants. Only pregnancies with at least 2 of these sample types were included in this analysis. Data analysis was conducted from March 1, 2023, to March 15, 2024. Exposures Quantitative metabolomics analysis was conducted on maternal third trimester serum, as well as placental tissue and umbilical cord serum collected at delivery. Main Outcomes and Measures Using the Autism Diagnostic Observation Schedule and Mullen Scales of Early Learning, outcomes were classified as ASD, other nontypical development (non-TD), and typical development (TD). Results This analysis included 100 maternal serum samples, 141 placental samples, and 124 umbilical cord serum samples from 152 pregnancies (median [IQR] maternal age, 34.6 [30.8-38.3] years; median [IQR] gestational age, 39.0 [38.6-39.7] weeks; 87 [57.2%] male infants). There was no evidence that the maternal third trimester serum metabolome was significantly associated with the other metabolomes. The placental and cord serum metabolomes were highly correlated (first latent variate pair: R2 = 0.75; P < .001) and the variate scores for each tissue were significantly associated with reduced risk of non-TD (placenta: relative risk [RR], 0.13; 95% CI, 0.02-0.71; cord: RR, 0.13; 95% CI, 0.03-0.70) but not ASD (placenta: RR, 1.09; 95% CI, 0.42-2.81; cord: RR, 0.63; 95% CI, 0.23-1.73) compared with the TD reference group. Conclusions and Relevance In this cohort study of children with high familial risk of ASD, placental and cord serum metabolism at delivery were highly correlated. Furthermore, placental and cord serum metabolic profiles were associated with risk of non-TD.
Collapse
Affiliation(s)
- Mariana Parenti
- Department of Nutrition, University of California, Davis
- Now with Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, Washington
| | - Rebecca J. Schmidt
- Department of Public Health Sciences, University of California, Davis
- MIND Institute, University of California, Davis, Sacramento
| | - Daniel J. Tancredi
- Department of Pediatrics, School of Medicine, University of California, Davis
| | - Irva Hertz-Picciotto
- Department of Public Health Sciences, University of California, Davis
- MIND Institute, University of California, Davis, Sacramento
| | - Cheryl K. Walker
- MIND Institute, University of California, Davis, Sacramento
- Department of Obstetrics & Gynecology, School of Medicine, University of California, Davis, Sacramento
| | - Carolyn M. Slupsky
- Department of Nutrition, University of California, Davis
- Department of Food Science and Technology, University of California, Davis
| |
Collapse
|
6
|
Kyogashima M, Kamijima K, Takai N, Nakajima T, Mikuma T, Komamura H, Asai K, Ishihara M, Sugiyama E, Tanaka N. Expression of dihomo-γ-linolenic acid and FADS1/2 and ELOVL2/5 in term rabbit placentas. Prostaglandins Leukot Essent Fatty Acids 2024; 202:102629. [PMID: 39002196 DOI: 10.1016/j.plefa.2024.102629] [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: 03/07/2024] [Revised: 06/28/2024] [Accepted: 07/04/2024] [Indexed: 07/15/2024]
Abstract
Long-chain polyunsaturated fatty acids (LCPUFAs) are essential for both fetal and placental development. We characterized the FA composition and gene expression levels of FA-metabolizing enzymes in rabbit placentas. Total FA compositions from term rabbit placentas (n = 7), livers, and plasma (both n = 4) were examined: among LCPUFAs with more than three double bonds, dihomo-γ-linolenic acid (DGLA) was the most abundant (11.4 ± 0.69 %, mean ± SE), while arachidonic acid was the second-most rich component (6.90 ± 0.56 %). DGLA was barely detectable (<1 %) in livers and plasma from term rabbits, which was significantly lower than in placentas (both p < 0.0001). Compared with the liver, transcript levels of the LCPUFA-metabolizing enzymes FADS2 and ELOVL5 were 7- and 4.5-fold higher in placentas (both p < 0.05), but levels of FADS1 and ELOVL2 were significantly lower (both p < 0.01). Our results suggest a placenta-specific enzyme expression pattern and LCPUFA profile in term rabbits, which may support a healthy pregnancy.
Collapse
Affiliation(s)
- M Kyogashima
- Department of Pharmaceutical Sciences, Nihon Pharmaceutical University, 10281 Komuro, Ina-Machi, Kita-Adachi-Gun, Saitama, 362-0806, Japan.
| | - K Kamijima
- Operation Department, Kitayama Labes Co., Ltd., Ina, Nagano, 396-0025, Japan
| | - N Takai
- Operation Department, Kitayama Labes Co., Ltd., Ina, Nagano, 396-0025, Japan
| | - T Nakajima
- Department of Global Medical Research Promotion, Shinshu University Graduate School of Medicine, Matsumoto, Nagano 390-8621, Japan
| | - T Mikuma
- Department of Pharmaceutical Sciences, Nihon Pharmaceutical University, 10281 Komuro, Ina-Machi, Kita-Adachi-Gun, Saitama, 362-0806, Japan
| | - H Komamura
- Department of Food and Health Sciences, Faculty of Health and Human Development, University of Nagano 380-8525, Japan
| | - K Asai
- Department of Food and Health Sciences, Faculty of Health and Human Development, University of Nagano 380-8525, Japan
| | - M Ishihara
- Department of Food and Health Sciences, Faculty of Health and Human Development, University of Nagano 380-8525, Japan
| | - E Sugiyama
- Department of Food and Health Sciences, Faculty of Health and Human Development, University of Nagano 380-8525, Japan
| | - N Tanaka
- Department of Global Medical Research Promotion, Shinshu University Graduate School of Medicine, Matsumoto, Nagano 390-8621, Japan
| |
Collapse
|
7
|
Gyurina K, Yarmak M, Sasi-Szabó L, Molnár S, Méhes G, Röszer T. Loss of Uncoupling Protein 1 Expression in the Subcutaneous Adipose Tissue Predicts Childhood Obesity. Int J Mol Sci 2023; 24:16706. [PMID: 38069028 PMCID: PMC10706300 DOI: 10.3390/ijms242316706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 11/21/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Stimulation of thermogenesis by inducing uncoupling protein 1 (UCP1) expression in adipocytes is thought to promote weight loss by increasing energy expenditure, and it is postulated that the human newborn has thermogenic subcutaneous fat depots. However, it remains unclear whether a relevant number of UCP1-expressing (UCP1+) adipocytes exist in the early postnatal life. Here we studied the distribution of UCP1 and the expression of thermogenic genes in the subcutaneous adipose tissues of the human fetus, infant and child. We show that the deep layer of human fetal and neonatal subcutaneous fat, particularly the abdominal wall, is rich in UCP1+ adipocytes. These adipocytes develop in the late third trimester and persist throughout childhood, expressing a panel of genes linked to mitochondrial biogenesis and thermogenesis. During the early childhood adiposity rebound-a critical phase that determines obesity risk later in life-the absence of adipose tissue UCP1 expression in children with normal body mass index (BMI) correlates with an obesity-associated gene expression signature. Finally, UCP1 expression is negatively correlated with BMI z-score and adipocyte size in infants and children. Overall, our results show that the absence of UCP1 expression in adipose tissue is an early indicator of adipose tissue expansion in children.
Collapse
Affiliation(s)
- Katalin Gyurina
- Institute and University Clinics of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary (L.S.-S.)
| | - Mariia Yarmak
- Institute and University Clinics of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary (L.S.-S.)
| | - László Sasi-Szabó
- Institute and University Clinics of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary (L.S.-S.)
| | - Sarolta Molnár
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (S.M.)
| | - Gábor Méhes
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary; (S.M.)
| | - Tamás Röszer
- Institute and University Clinics of Pediatrics, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary (L.S.-S.)
- Institute of Neurobiology, Ulm University, 89081 Ulm, Germany
| |
Collapse
|
8
|
Rehman SU, Ali R, Zhang H, Zafar MH, Wang M. Research progress in the role and mechanism of Leucine in regulating animal growth and development. Front Physiol 2023; 14:1252089. [PMID: 38046946 PMCID: PMC10691278 DOI: 10.3389/fphys.2023.1252089] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 11/08/2023] [Indexed: 12/05/2023] Open
Abstract
Leucine, a branched-chain amino acid, is essential in regulating animal growth and development. Recent research has uncovered the mechanisms underlying Leucine's anabolic effects on muscle and other tissues, including its ability to stimulate protein synthesis by activating the mTORC1 signaling pathway. The co-ingestion of carbohydrates and essential amino acids enhances Leucine's anabolic effects. Moreover, Leucine has been shown to benefit lipid metabolism, and insulin sensitivity, making it a promising strategy for preventing and treating metabolic diseases, including type 2 diabetes and obesity. While emerging evidence indicates that epigenetic mechanisms may mediate Leucine's effects on growth and development, more research is needed to elucidate its mechanisms of action fully. Specific studies have demonstrated that Leucine promotes muscle growth and metabolic health in animals and humans, making it a promising therapeutic agent. However, it is essential to note that Leucine supplementation may cause digestive issues or interact with certain medications, and More study is required to determine definitively optimal dosages. Therefore, it is important to understand how Leucine interacts with other nutrients, dietary factors, and lifestyle habits to maximize its benefits. Overall, Leucine's importance in human nutrition is far-reaching, and its potential to prevent muscle loss and enhance athletic performance warrants further investigation.
Collapse
Affiliation(s)
| | | | | | | | - Mengzhi Wang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, China
| |
Collapse
|
9
|
Nargis T, Lin X, Giordano E, Ijaz L, Suhail S, Gurzenda EM, Kiefer D, Quadro L, Hanna N, Hussain MM. Characterization of lipoproteins in human placenta and fetal circulation as well as gestational changes in lipoprotein assembly and secretion in human and mouse placentas. Biochim Biophys Acta Mol Cell Biol Lipids 2023; 1868:159357. [PMID: 37315736 PMCID: PMC10529644 DOI: 10.1016/j.bbalip.2023.159357] [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: 01/27/2023] [Revised: 04/28/2023] [Accepted: 06/07/2023] [Indexed: 06/16/2023]
Abstract
In the maternal circulation, apoB-containing low-density lipoproteins (LDL) and apoA1-containing high-density lipoproteins (HDL) transport lipids. The production of lipoproteins in the placenta has been suggested, but the directionality of release has not been resolved. We compared apolipoprotein concentrations and size-exclusion chromatography elution profiles of lipoproteins in maternal/fetal circulations, and in umbilical arteries/veins; identified placental lipoprotein-producing cells; and studied temporal induction of lipoprotein-synthesizing machinery during pregnancy. We observed that maternal and fetal lipoproteins are different with respect to concentrations and elution profiles. Surprisingly, concentrations and elution profiles of lipoproteins in umbilical arteries and veins were similar indicating their homeostatic control. Human placental cultures synthesized apoB100-containing LDL-sized and apoA1-containing HDL-sized particles. Immunolocalization techniques revealed that ApoA1 was present mainly in syncytiotrophoblasts. MTP, a critical protein for lipoprotein assembly, was in these trophoblasts. ApoB was in the placental stroma indicating that trophoblasts secrete apoB-containing lipoproteins into the stroma. ApoB and MTP expressions increased in placentas from the 2nd trimester to term, whereas apoA1 expression was unchanged. Thus, our studies provide new information regarding the timing of lipoprotein gene induction during gestation, the cells involved in lipoprotein assembly and the gel filtration profiles of human placental lipoproteins. Next, we observed that mouse placenta produces MTP, apoB100, apoB48 and apoA1. The expression of genes gradually increased and peaked in late gestation. This information may be useful in identifying transcription factors regulating the induction of these genes in gestation and the importance of placental lipoprotein assembly in fetal development.
Collapse
Affiliation(s)
- Titli Nargis
- Department of Foundations of Medicine, NYU Long Island School of Medicine, Mineola, NY, USA
| | - Xinhua Lin
- Department of Pediatrics, NYU Long Island School of Medicine, Mineola, New York, USA
| | - Elena Giordano
- Food Science Department, Rutgers Center for Lipid Research and Institute of Food Nutrition and Health, Rutgers University, New Brunswick, NJ, USA
| | - Laraib Ijaz
- Department of Foundations of Medicine, NYU Long Island School of Medicine, Mineola, NY, USA
| | - Sarah Suhail
- Department of Foundations of Medicine, NYU Long Island School of Medicine, Mineola, NY, USA
| | - Ellen M Gurzenda
- Department of Pediatrics, NYU Long Island School of Medicine, Mineola, New York, USA
| | - Daniel Kiefer
- Department of Obstetrics and Gynecology, NYU Long Island School of Medicine, Mineola, New York, USA
| | - Loredana Quadro
- Food Science Department, Rutgers Center for Lipid Research and Institute of Food Nutrition and Health, Rutgers University, New Brunswick, NJ, USA
| | - Nazeeh Hanna
- Department of Pediatrics, NYU Long Island School of Medicine, Mineola, New York, USA
| | - M Mahmood Hussain
- Department of Foundations of Medicine, NYU Long Island School of Medicine, Mineola, NY, USA.
| |
Collapse
|
10
|
Klöppel E, Souza MR, Barco VS, Gallego FQ, Sinzato YK, Corrente JE, Rodrigues T, Volpato GT, Damasceno DC. Calcium Supplementation on Glucose Tolerance, Oxidative Stress, and Reproductive Outcomes of Diabetic Rats and Their Offspring. Reprod Sci 2023; 30:2813-2828. [PMID: 37002533 DOI: 10.1007/s43032-023-01217-9] [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: 10/14/2022] [Accepted: 03/09/2023] [Indexed: 04/03/2023]
Abstract
Diabetes mellitus increases the risk of obstetric complications, morbidity, and infant mortality. Controlled nutritional therapy with micronutrients has been employed. However, the effect of calcium (Ca2+) supplementation on diabetic pregnancy is unclear. We aimed to evaluate whether diabetic rats supplemented with Ca2+ during pregnancy present better glucose tolerance, redox status, embryonic and fetal development, newborn weight, and the prooxidant and antioxidant balance of male and female pups. For this, newborn rats received the beta-cytotoxic drug streptozotocin for inducing diabetes on the day of birth. In adulthood, these rats were mated and treated with Ca2+ twice a day from day 0 to day 20 of pregnancy. On day 17, the pregnant rats were submitted to the oral glucose tolerance test (OGTT). At the end of pregnancy, they were anesthetized and killed to collect blood and pancreas samples. The uterine horns were exposed for an evaluation of maternal reproductive outcomes and embryofetal development, and the offspring's liver samples were collected for redox status measurement. Nondiabetic and diabetic rats supplemented with Ca2+ showed no influence on glucose tolerance, redox status, insulin synthesis, serum calcium levels, and embryofetal losses. The reduced rate of newborns classified as adequate for gestational age (AGA) and higher rates of LGA (large) and small (LGA) newborns and higher -SH and GSH-Px antioxidant activities in female pups were observed in diabetic dams, regardless of supplementation. Thus, maternal supplementation caused no improvement in glucose tolerance, oxidative stress biomarkers, embryofetal growth and development, and antioxidants in pups from diabetic mothers.
Collapse
Affiliation(s)
- Eduardo Klöppel
- Laboratory of Experimental Research On Gynecology and Obstetrics, Postgraduate Course On Tocogynecology, Botucatu Medical School, São Paulo State University_UNESP, Botucatu, São Paulo State, Brazil
| | - Maysa Rocha Souza
- Laboratory of Experimental Research On Gynecology and Obstetrics, Postgraduate Course On Tocogynecology, Botucatu Medical School, São Paulo State University_UNESP, Botucatu, São Paulo State, Brazil
- Laboratory of System Physiology and Reproductive Toxicology, Federal University of Mato Grosso_UFMT, Mato Grosso State, Barra Do Garças, Brazil
| | - Vinícius Soares Barco
- Laboratory of Experimental Research On Gynecology and Obstetrics, Postgraduate Course On Tocogynecology, Botucatu Medical School, São Paulo State University_UNESP, Botucatu, São Paulo State, Brazil
| | - Franciane Quintanilha Gallego
- Laboratory of Experimental Research On Gynecology and Obstetrics, Postgraduate Course On Tocogynecology, Botucatu Medical School, São Paulo State University_UNESP, Botucatu, São Paulo State, Brazil
| | - Yuri Karen Sinzato
- Laboratory of Experimental Research On Gynecology and Obstetrics, Postgraduate Course On Tocogynecology, Botucatu Medical School, São Paulo State University_UNESP, Botucatu, São Paulo State, Brazil
| | - José Eduardo Corrente
- Research Support Office, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu, São Paulo State, Brazil
| | - Tiago Rodrigues
- Center for Natural and Human Sciences (CCNH), Federal University of ABC (UFABC), Santo André, São Paulo, SP, Brazil
| | - Gustavo Tadeu Volpato
- Laboratory of Experimental Research On Gynecology and Obstetrics, Postgraduate Course On Tocogynecology, Botucatu Medical School, São Paulo State University_UNESP, Botucatu, São Paulo State, Brazil
- Laboratory of System Physiology and Reproductive Toxicology, Federal University of Mato Grosso_UFMT, Mato Grosso State, Barra Do Garças, Brazil
| | - Débora Cristina Damasceno
- Laboratory of Experimental Research On Gynecology and Obstetrics, Postgraduate Course On Tocogynecology, Botucatu Medical School, São Paulo State University_UNESP, Botucatu, São Paulo State, Brazil.
| |
Collapse
|
11
|
Shi Y, Wang CC, Wu L, Zhang Y, Xu A, Wang Y. Pathophysiological Insight into Fatty Acid-Binding Protein-4: Multifaced Roles in Reproduction, Pregnancy, and Offspring Health. Int J Mol Sci 2023; 24:12655. [PMID: 37628833 PMCID: PMC10454382 DOI: 10.3390/ijms241612655] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 08/04/2023] [Accepted: 08/06/2023] [Indexed: 08/27/2023] Open
Abstract
Fatty acid-binding protein-4 (FABP4), commonly known as adipocyte-fatty acid-binding protein (A-FABP), is a pleiotropic adipokine that broadly affects immunity and metabolism. It has been increasingly recognized that FABP4 dysfunction is associated with various metabolic syndromes, including obesity, diabetes, cardiovascular diseases, and metabolic inflammation. However, its explicit roles within the context of women's reproduction and pregnancy remain to be investigated. In this review, we collate recent studies probing the influence of FABP4 on female reproduction, pregnancy, and even fetal health. Elevated circulating FABP4 levels have been found to correlate with impaired reproductive function in women, such as polycystic ovary syndrome and endometriosis. Throughout pregnancy, FABP4 affects maternal-fetal interface homeostasis by affecting both glycolipid metabolism and immune tolerance, leading to adverse pregnancy outcomes, including miscarriage, gestational obesity, gestational diabetes, and preeclampsia. Moreover, maternal FABP4 levels exhibit a substantial linkage with the metabolic health of offspring. Herein, we discuss the emerging significance and potential application of FABP4 in reproduction and pregnancy health and delve into its underlying mechanism at molecular levels.
Collapse
Affiliation(s)
- Yue Shi
- The Second Clinical Medical School, Beijing University of Chinese Medicine, Beijing 100078, China; (Y.S.); (Y.Z.)
| | - Chi-Chiu Wang
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong;
- Li Ka Shing Institute of Health Sciences, School of Biomedical Sciences, Chinese University of Hong Kong-Sichuan University Joint Laboratory in Reproductive Medicine, The Chinese University of Hong Kong, Hong Kong
| | - Liqun Wu
- Department of Pediatrics, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100078, China;
| | - Yunqing Zhang
- The Second Clinical Medical School, Beijing University of Chinese Medicine, Beijing 100078, China; (Y.S.); (Y.Z.)
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong;
- Department of Medicine, The University of Hong Kong, Hong Kong
| | - Yao Wang
- Department of Obstetrics and Gynaecology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong;
| |
Collapse
|
12
|
Fossee E, Zamora AN, Peterson KE, Cantoral A, Perng W, Téllez-Rojo MM, Torres-Olascoaga LA, Jansen EC. Prenatal dietary patterns in relation to adolescent offspring adiposity and adipokines in a Mexico City cohort. J Dev Orig Health Dis 2023; 14:371-380. [PMID: 36655507 PMCID: PMC10202837 DOI: 10.1017/s2040174422000678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Maternal diet during pregnancy has been associated with obesity among offspring. The extent to which trimester-specific dietary patterns are associated with markers of adiposity during adolescence remains unclear. We examined associations between prenatal diet patterns with adolescent offspring measures of adiposity and adipokines in 384 mother-adolescent dyads from the Mexico City ELEMENT cohort. Trimester-specific diet patterns were derived from principal component analysis of food frequency questionnaire data. Adolescent anthropometry and serum leptin and adiponectin were measured at 10-17 years. Three maternal diet patterns were identified: Prudent Diet (PD), high in fish and vegetables, the High Meat and Fat Diet (HMFD), high in pork and processed meats, and the Transitioning Mexican Diet (TMD), high in corn tortillas and sugar-sweetened beverages. Multiple linear regression was used to estimate sex-stratified associations among quartiles of diet patterns with adiposity and adipokines, adjusting for maternal marital status, education, and parity. First trimester TMD was associated with greater anthropometric measures and higher leptin in females, while third trimester HMFD was associated higher body fat percentage, triceps thickness, waist circumference, and leptin, but lower adiponectin among males. Contrary to expectation, there were positive associations between the trimester 1 PD pattern and anthropometric measurements in females, and for trimester 2 HMFD and TMD patterns with adipokines among males. Findings suggest maternal diet patterns may influence offspring adiposity markers during adolescence in a sex-specific manner.
Collapse
Affiliation(s)
- Erica Fossee
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Astrid N. Zamora
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Karen E. Peterson
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | | | - Wei Perng
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
- Department of Epidemiology, Colorado School of Public Health, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA
| | - Martha M. Téllez-Rojo
- Center for Research on Nutrition and Health, National Institute of Public Health, Cuernavaca, Mexico
| | - Libni A. Torres-Olascoaga
- Center for Research on Nutrition and Health, National Institute of Public Health, Cuernavaca, Mexico
| | - Erica C. Jansen
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| |
Collapse
|
13
|
Guerrero-Santoro J, Morizane M, Oh SY, Mishima T, Goff JP, Bildirici I, Sadovsky E, Ouyang Y, Tyurin VA, Tyurina YY, Kagan VE, Sadovsky Y. The lipase cofactor CGI58 controls placental lipolysis. JCI Insight 2023; 8:168717. [PMID: 37212279 DOI: 10.1172/jci.insight.168717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/12/2023] [Indexed: 05/23/2023] Open
Abstract
In eutherians, the placenta plays a critical role in the uptake, storage, and metabolism of lipids. These processes govern the availability of fatty acids to the developing fetus, where inadequate supply has been associated with substandard fetal growth. Whereas lipid droplets are essential for the storage of neutral lipids in the placenta and many other tissues, the processes that regulate placental lipid droplet lipolysis remain largely unknown. To assess the role of triglyceride lipases and their cofactors in determining placental lipid droplet and lipid accumulation, we assessed the role of patatin like phospholipase domain containing 2 (PNPLA2) and comparative gene identification-58 (CGI58) in lipid droplet dynamics in the human and mouse placenta. While both proteins are expressed in the placenta, the absence of CGI58, not PNPLA2, markedly increased placental lipid and lipid droplet accumulation. These changes were reversed upon restoration of CGI58 levels selectively in the CGI58-deficient mouse placenta. Using co-immunoprecipitation, we found that, in addition to PNPLA2, PNPLA9 interacts with CGI58. PNPLA9 was dispensable for lipolysis in the mouse placenta yet contributed to lipolysis in human placental trophoblasts. Our findings establish a crucial role for CGI58 in placental lipid droplet dynamics and, by extension, in nutrient supply to the developing fetus.
Collapse
Affiliation(s)
- Jennifer Guerrero-Santoro
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mayumi Morizane
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Soo-Young Oh
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Takuya Mishima
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Julie P Goff
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Ibrahim Bildirici
- Department of Obstetrics and Gynecology, Washington University School of Medicine in St. Louis, St. Louis, Missouri, USA
| | - Elena Sadovsky
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yingshi Ouyang
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Vladimir A Tyurin
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health
| | - Yulia Y Tyurina
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health
| | - Valerian E Kagan
- Center for Free Radical and Antioxidant Health, Department of Environmental and Occupational Health
- Department of Chemistry
- Department of Pharmacology and Chemical Biology
- Department of Radiation Oncology; and
| | - Yoel Sadovsky
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
14
|
Varshney S, Adela R, Kachhawa G, Dada R, Kulshreshtha V, Kumari R, Agarwal R, Khadgawat R. Disrupted placental vitamin D metabolism and calcium signaling in gestational diabetes and pre-eclampsia patients. Endocrine 2023; 80:191-200. [PMID: 36477942 DOI: 10.1007/s12020-022-03272-9] [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: 09/26/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Gestational diabetes (GDM) and pre-eclampsia (PE) represents the unrecognized risk factors for reduced bone content in neonates. The present study is planned to explore the components of vitamin D metabolism and calcium transport in placenta of GDM and PE cases and its effect on the neonatal bone mass determination using bone densitometry system. METHODS We have collected serum and placenta tissues from GDM (n = 20), PE (n = 20), and healthy pregnancies (n = 20). In the present study, we found mRNA expression of oxidative stress markers, vitamin D metabolic components and calcium channels, calcium channel binding proteins, plasma membrane calcium ATPase, ATP synthase and Ca2+ release genes; Ryanodine receptors genes were assessed by quantitative real-time PCR (qRT-PCR) in placental tissue of GDM, PE, and healthy pregnancies. RESULTS We observed high level of oxidative stress in both GDM and PE placenta compared to normal pregnancies. CYP2R1 and VDR mRNA expression was significantly downregulated and upregulation of CYP27B1 and CYP24A1 in GDM and PE compared with healthy cases. Similarly, calcium transporters were downregulated in GDM and PE placental tissues. In addition, CYP24A1, VDR, CaBP28K, TRPV5 and PMCA3 mRNA expression were correlated with BMC of neonates. DISCUSSION Oxidative stress is probably relevant to disrupted vitamin D homeostasis and calcium transport in the placenta of GDM and PE cases. The altered regulatory mechanism of CYP24A1 and VDR could indicates more pronounced serum 25(OH)D reduction. Additionally, reduced BMC in the neonates of these cases might be as consequences of modified CYP24A1, VDR, CaBP28K, TRPV5 and PMCA3 mRNA expression.
Collapse
Affiliation(s)
- Shweta Varshney
- Department of Endocrinology & Metabolism, All India Institute of Medical Science, New Delhi, India
| | - Ramu Adela
- Department of Endocrinology & Metabolism, All India Institute of Medical Science, New Delhi, India
- Department of Pharmacy Practice, National Institute of Pharmaceutical Education and Research-Guwahati, Guwahati, Assam, India
| | - Garima Kachhawa
- Department of Obstetrics & Gynecology, All India Institute of Medical Science, New Delhi, India
| | - Reema Dada
- Department of Anatomy, All India Institute of Medical Science, New Delhi, India
| | - Vidushi Kulshreshtha
- Department of Obstetrics & Gynecology, All India Institute of Medical Science, New Delhi, India
| | - Rajesh Kumari
- Department of Obstetrics & Gynecology, All India Institute of Medical Science, New Delhi, India
| | - Ramesh Agarwal
- Department of Neonatology, All India Institute of Medical Science, New Delhi, India
| | - Rajesh Khadgawat
- Department of Endocrinology & Metabolism, All India Institute of Medical Science, New Delhi, India.
| |
Collapse
|
15
|
Klöppel E, Sinzato YK, Rodrigues T, Gallego FQ, Karki B, Volpato GT, Corrente JE, Roy S, Damasceno DC. Benefits of Vitamin D Supplementation on Pregnancy of Rats with Pregestational Diabetes and Their Offspring. Reprod Sci 2023; 30:1241-1256. [PMID: 35999443 DOI: 10.1007/s43032-022-01056-0] [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: 12/06/2021] [Accepted: 07/30/2022] [Indexed: 11/30/2022]
Abstract
Studies on vitamin D supplementation have been performed in experimental and clinical investigations considering gestational diabetes and/or vitamin D deficiency in pregnancy. However, the results are controversial and few present the effects and mechanisms of this micronutrient on pregestational diabetes. The objective of this study was to evaluate the effect of vitamin D on the pregnancy of rats with pre-existing diabetes and their fetuses. Pregestational diabetes was induced in Sprague-Dawley rats at birth. The adult diabetic and nondiabetic rats were orally administered with vitamin D (cholecalciferol) throughout the pregnancy. The diabetes status was monitored during pregnancy by an oral glucose tolerance test (OGTT). At the end of the pregnancy, pancreas and blood samples were collected for morphological analyses and lipid peroxidation measurements, respectively. The influence of vitamin D treatment on reproductive outcomes, fetal growth, and development were compared to those of untreated diabetic and nondiabetic pregnant rats. P < 0.05 was considered a significant statistical limit. The diabetic rats given vitamin D had a greater number of insulin-positive cells, contributing to reduced blood glucose levels and thiobarbituric acid reactive substance concentrations (TBARS-an indicator of membrane lipid peroxidation), and increased reduced thiol group levels, contributing to suitable intrauterine conditions for better fetal development, which was confirmed by higher fetal viability rates. Thus, this study shows the effects and mechanisms of vitamin D supplementation on pre-existing diabetes in pregnant rats, confirming its beneficial effects on maternal redox status and glycemic control, and the decline of adverse maternal-fetal repercussions.
Collapse
Affiliation(s)
- Eduardo Klöppel
- Laboratory of Experimental Research On Gynecology and Obstetrics, Postgraduate Course On Tocogynecology, Botucatu Medical School, Sao Paulo State University (Unesp), Botucatu, Sao Paulo State, Brazil
| | - Yuri K Sinzato
- Laboratory of Experimental Research On Gynecology and Obstetrics, Postgraduate Course On Tocogynecology, Botucatu Medical School, Sao Paulo State University (Unesp), Botucatu, Sao Paulo State, Brazil
| | - Tiago Rodrigues
- Federal University of ABC (UFABC), Santo André, São Paulo State, Brazil
| | - Franciane Q Gallego
- Laboratory of Experimental Research On Gynecology and Obstetrics, Postgraduate Course On Tocogynecology, Botucatu Medical School, Sao Paulo State University (Unesp), Botucatu, Sao Paulo State, Brazil
| | - Barshana Karki
- Laboratory of Experimental Research On Gynecology and Obstetrics, Postgraduate Course On Tocogynecology, Botucatu Medical School, Sao Paulo State University (Unesp), Botucatu, Sao Paulo State, Brazil
- , Boston, MA, USA
| | - Gustavo T Volpato
- Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso (UFMT), Barra do Garças, Mato Grosso State, Brazil
| | - José E Corrente
- Research Support Office, Botucatu Medical School, São Paulo State University (Unesp), Botucatu, São Paulo State, Brazil
| | - Sayon Roy
- Department of Ophthalmology, Boston University School of Medicine, Boston, MA, USA
| | - Débora C Damasceno
- Laboratory of Experimental Research On Gynecology and Obstetrics, Postgraduate Course On Tocogynecology, Botucatu Medical School, Sao Paulo State University (Unesp), Botucatu, Sao Paulo State, Brazil.
| |
Collapse
|
16
|
Sekovanić A, Dorotić A, Pašalić D, Orct T, Kljaković-Gašpić Z, Grgec AS, Stasenko S, Mioč T, Piasek M, Jurasović J. The effects of maternal cigarette smoking on cadmium and lead levels, miRNA expression and biochemical parameters across the feto-placental unit. Heliyon 2022; 8:e12568. [PMID: 36636214 PMCID: PMC9830161 DOI: 10.1016/j.heliyon.2022.e12568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/12/2022] [Accepted: 12/15/2022] [Indexed: 12/25/2022] Open
Abstract
Several miRNAs have been previously identified to be associated with cigarette smoke and/or the toxic metals cadmium (Cd) and lead (Pb). The aim of this study was to investigate the associations of maternal cigarette smoking with cadmium (Cd) and lead (Pb) levels, candidate miRNA expression and biochemical parameters across the feto-placental unit. miRNAs were isolated according to protocols provided by manufacturer from 72 healthy postpartum women using Qiagens' kits based on phenol/guanidine samples lysis and silica-membrane purification of total RNA. Candidate miRNAs (miR-1537, miR-190b, miR-16, miR-21, and miR-146a) were quantified by real-time PCR. Biochemical parameters were analyzed in plasma samples by standardized and harmonized enzymatic methods using appropriate calibrators, while CRP was determined by immunoturbidimetric method. Concentration of Cd and Pb in whole blood and placenta samples were measured by inductively coupled plasma mass spectroscopy. Cd levels in smokers were higher in all of the analyzed compartments of the feto-placental unit, Pb in maternal blood and placenta than non-smokers. Smokers also had a higher expression of miR-16 in maternal and miR-146a in cord plasma, and lower expression of miR-21 in the placenta in comparison to non-smokers. Urate concentrations in the maternal plasma of smokers were lower than this value in non-smokers. The study has demonstrated that maternal smoking was associated with toxic metals (Cd and Pb) levels, urate concentration and alteration of miRNA expression. Given that the effects of maternal smoking on miRNA expression are inadequate, all compartments of the feto-placental unit should be analyzed to obtain a complete picture. This paper is the first to report on the results of expression of cellular and circulating miRNAs simultaneously in maternal and fetal compartments and in the placenta.
Collapse
Affiliation(s)
- Ankica Sekovanić
- Analytical Toxicology and Mineral Metabolism Unit, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia
| | - Adrijana Dorotić
- Department of Medical Laboratory Diagnostics, University Hospital Sveti Duh, 10000 Zagreb, Croatia
| | - Daria Pašalić
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, University of Zagreb, School of Medicine, 10000 Zagreb, Croatia
- Corresponding author.
| | - Tatjana Orct
- Analytical Toxicology and Mineral Metabolism Unit, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia
| | - Zorana Kljaković-Gašpić
- Analytical Toxicology and Mineral Metabolism Unit, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia
| | - Antonija Sulimanec Grgec
- Analytical Toxicology and Mineral Metabolism Unit, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia
| | - Sandra Stasenko
- Clinical Department of Obstetrics and Gynecology, Merkur University Hospital, 10000 Zagreb, Croatia
| | - Tatjana Mioč
- Clinical Department of Obstetrics and Gynecology, Merkur University Hospital, 10000 Zagreb, Croatia
| | - Martina Piasek
- Analytical Toxicology and Mineral Metabolism Unit, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia
| | - Jasna Jurasović
- Analytical Toxicology and Mineral Metabolism Unit, Institute for Medical Research and Occupational Health, 10000 Zagreb, Croatia
| |
Collapse
|
17
|
Barco VS, Gallego FQ, Paula VG, Cruz LL, Karki B, Volpato GT, Damasceno DC. Effects of diabetes between generations on the pre-embryos of rats. AN ACAD BRAS CIENC 2022; 94:e20220717. [PMID: 36515329 DOI: 10.1590/0001-3765202220220717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 09/15/2022] [Indexed: 12/14/2022] Open
Abstract
Pregestational hyperglycemia cause adverse effects on mothers and their offspring. We aimed to evaluate the maternal hyperglycemia influence on pre-embryos from diabetic rats and on their generations (daughters and granddaughters). Diabetes was induced in Sprague-Dawley rats. The mothers and their female pups were submitted to oral glucose tolerance test in adulthood. In day 4 of pregnancy, pre-embryos were collected for morphological analysis. The diabetic mother, daughter and granddaughter rats showed glucose intolerance and their pre-embryos presented developmental delay, degeneration and losses compared to the nondiabetic group. Thus, maternal diabetes transgenerationally affects embryos at early development, which contributes for embryofetal losses.
Collapse
Affiliation(s)
- Vinícius S Barco
- Programa de Pós-Graduação em Tocoginecologia, Universidade Estadual Paulista (UNESP), Faculdade de Medicina de Botucatu, Laboratório de Pesquisa Experimental em Ginecologia e Obstetrícia, Distrito de Rubião Júnior, s/n, 18610-879 Botucatu, SP, Brazil
| | - Franciane Q Gallego
- Programa de Pós-Graduação em Tocoginecologia, Universidade Estadual Paulista (UNESP), Faculdade de Medicina de Botucatu, Laboratório de Pesquisa Experimental em Ginecologia e Obstetrícia, Distrito de Rubião Júnior, s/n, 18610-879 Botucatu, SP, Brazil
| | - Verônyca G Paula
- Programa de Pós-Graduação em Tocoginecologia, Universidade Estadual Paulista (UNESP), Faculdade de Medicina de Botucatu, Laboratório de Pesquisa Experimental em Ginecologia e Obstetrícia, Distrito de Rubião Júnior, s/n, 18610-879 Botucatu, SP, Brazil
| | - Larissa L Cruz
- Programa de Pós-Graduação em Tocoginecologia, Universidade Estadual Paulista (UNESP), Faculdade de Medicina de Botucatu, Laboratório de Pesquisa Experimental em Ginecologia e Obstetrícia, Distrito de Rubião Júnior, s/n, 18610-879 Botucatu, SP, Brazil.,Universidade Federal do Mato Grosso (UFTM), Instituto de Ciências Biológicas e da Saúde, Laboratório de Fisiologia de Sistemas e Toxicologia Reprodutiva, Av. Valdon Varjão, s/n, 78600-000 Barra do Garças, MT, Brazil
| | - Barshana Karki
- Programa de Pós-Graduação em Tocoginecologia, Universidade Estadual Paulista (UNESP), Faculdade de Medicina de Botucatu, Laboratório de Pesquisa Experimental em Ginecologia e Obstetrícia, Distrito de Rubião Júnior, s/n, 18610-879 Botucatu, SP, Brazil
| | - Gustavo T Volpato
- Universidade Federal do Mato Grosso (UFTM), Instituto de Ciências Biológicas e da Saúde, Laboratório de Fisiologia de Sistemas e Toxicologia Reprodutiva, Av. Valdon Varjão, s/n, 78600-000 Barra do Garças, MT, Brazil
| | - Débora C Damasceno
- Programa de Pós-Graduação em Tocoginecologia, Universidade Estadual Paulista (UNESP), Faculdade de Medicina de Botucatu, Laboratório de Pesquisa Experimental em Ginecologia e Obstetrícia, Distrito de Rubião Júnior, s/n, 18610-879 Botucatu, SP, Brazil
| |
Collapse
|
18
|
Habra H, Kachman M, Padmanabhan V, Burant C, Karnovsky A, Meijer J. Alignment and Analysis of a Disparately Acquired Multibatch Metabolomics Study of Maternal Pregnancy Samples. J Proteome Res 2022; 21:2936-2946. [PMID: 36367990 DOI: 10.1021/acs.jproteome.2c00371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Untargeted liquid chromatography-mass spectrometry metabolomics studies are typically performed under roughly identical experimental settings. Measurements acquired with different LC-MS protocols or following extended time intervals harbor significant variation in retention times and spectral abundances due to altered chromatographic, spectrometric, and other factors, raising many data analysis challenges. We developed a computational workflow for merging and harmonizing metabolomics data acquired under disparate LC-MS conditions. Plasma metabolite profiles were collected from two sets of maternal subjects three years apart using distinct instruments and LC-MS procedures. Metabolomics features were aligned using metabCombiner to generate lists of compounds detected across all experimental batches. We applied data set-specific normalization methods to remove interbatch and interexperimental variation in spectral intensities, enabling statistical analysis on the assembled data matrix. Bioinformatics analyses revealed large-scale metabolic changes in maternal plasma between the first and third trimesters of pregnancy and between maternal plasma and umbilical cord blood. We observed increases in steroid hormones and free fatty acids from the first trimester to term of gestation, along with decreases in amino acids coupled to increased levels in cord blood. This work demonstrates the viability of integrating nonidentically acquired LC-MS metabolomics data and its utility in unconventional metabolomics study designs.
Collapse
Affiliation(s)
- Hani Habra
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Maureen Kachman
- Michigan Regional Comprehensive Metabolomics Resource Core, University of Michigan, Ann Arbor, Michigan 48105, United States
| | - Vasantha Padmanabhan
- Department of Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, Michigan 48109, United States
- Department of Obstetrics & Gynecology, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Charles Burant
- Michigan Regional Comprehensive Metabolomics Resource Core, University of Michigan, Ann Arbor, Michigan 48105, United States
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Alla Karnovsky
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
| | - Jennifer Meijer
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, Michigan 48109, United States
- Department of Medicine, Geisel School of Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire 03756, United States
| |
Collapse
|
19
|
Hoang AC, Sasi-Szabó L, Pál T, Szabó T, Diedrich V, Herwig A, Landgraf K, Körner A, Röszer T. Mitochondrial RNA stimulates beige adipocyte development in young mice. Nat Metab 2022; 4:1684-1696. [PMID: 36443525 PMCID: PMC9771821 DOI: 10.1038/s42255-022-00683-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 10/10/2022] [Indexed: 11/30/2022]
Abstract
Childhood obesity is a serious public health crisis and a critical factor that determines future obesity prevalence. Signals affecting adipocyte development in early postnatal life have a strong potential to trigger childhood obesity; however, these signals are still poorly understood. We show here that mitochondrial (mt)RNA efflux stimulates transcription of nuclear-encoded genes for mitobiogenesis and thermogenesis in adipocytes of young mice and human infants. While cytosolic mtRNA is a potential trigger of the interferon (IFN) response, young adipocytes lack such a response to cytosolic mtRNA due to the suppression of IFN regulatory factor (IRF)7 expression by vitamin D receptor signalling. Adult and obese adipocytes, however, strongly express IRF7 and mount an IFN response to cytosolic mtRNA. In turn, suppressing IRF7 expression in adult adipocytes restores mtRNA-induced mitobiogenesis and thermogenesis and eventually mitigates obesity. Retrograde mitochondrion-to-nucleus signalling by mtRNA is thus a mechanism to evoke thermogenic potential during early adipocyte development and to protect against obesity.
Collapse
Affiliation(s)
| | - László Sasi-Szabó
- Institute of Pediatrics, Clinical Centre, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tibor Pál
- Institute of Pediatrics, Clinical Centre, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamás Szabó
- Institute of Pediatrics, Clinical Centre, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | | | - Annika Herwig
- Institute of Neurobiology, Ulm University, Ulm, Germany
| | - Kathrin Landgraf
- Center for Pediatric Research, University Hospital for Children and Adolescents, University of Leipzig, Leipzig, Germany
| | - Antje Körner
- Center for Pediatric Research, University Hospital for Children and Adolescents, University of Leipzig, Leipzig, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Center München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Tamás Röszer
- Institute of Neurobiology, Ulm University, Ulm, Germany.
- Institute of Pediatrics, Clinical Centre, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| |
Collapse
|
20
|
Röszer T. Metabolic impact of adipose tissue macrophages in the early postnatal life. J Leukoc Biol 2022; 112:1515-1524. [PMID: 35899927 PMCID: PMC9796690 DOI: 10.1002/jlb.3mr0722-201r] [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: 05/29/2022] [Revised: 07/02/2022] [Indexed: 01/07/2023] Open
Abstract
Adipose tissue macrophages (ATMs) play key roles in metabolic inflammation, insulin resistance, adipose tissue fibrosis, and immune disorders associated with obesity. Research on ATM biology has mostly been conducted in the setting of adult obesity, since adipocyte hypertrophy is associated with a significant increase in ATM number. Signals that control ATM activation toward a proinflammatory or a proresolving phenotype also determine the developmental program and lipid metabolism of adipocytes after birth. ATMs are present at birth and actively participate in the synthesis of mediators, which induce lipolysis, mitobiogenesis, and mitochondrial uncoupling in adipocytes. ATMs in the newborn and the infant promote a lipolytic and fatty acid oxidizing adipocyte phenotype, which is essential to support the lipid-fueled metabolism, to maintain nonshivering thermogenesis and counteract an excessive adipose tissue expansion. Since adipose tissue metabolism in the early postnatal life determines obesity status in adulthood, early-life ATM functions may have a life-long impact.
Collapse
Affiliation(s)
- Tamás Röszer
- Division of Pediatric Obesity, Children's Hospital and Institute of PediatricsUniversity of DebrecenDebrecenHungary,Institute of NeurobiologyUlm UniversityUlmGermany
| |
Collapse
|
21
|
Catlin NR, Bowman CJ, Campion SN, Lewis EM, Nowland WS, Stethem C, Cappon GD. The postnatal resolution of developmental toxicity induced by pharmacological diacylglycerol acyltransferase 2 (DGAT2) inhibition during gestation in rats. Toxicol Sci 2022; 189:225-236. [PMID: 35866640 DOI: 10.1093/toxsci/kfac077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Ervogastat (PF-06865571) is a small molecule diacylglycerol acyltransferase 2 (DGAT2) inhibitor being developed for the oral treatment of non-alcoholic steatohepatitis (NASH) with liver fibrosis. DGAT2 is a key enzyme in triglyceride synthesis in tissues and in regulating energy metabolism. Fertility and developmental toxicity studies with ervogastat were conducted in female rats and rabbits. There were no effects on female rat fertility or rabbit embryo-fetal development. Administration of ervogastat to pregnant rats during organogenesis reduced fetal weight and caused higher incidences of bent bones in fetuses that were shown to resolve by postnatal day 28 and were therefore considered to be transient variations secondary to developmental delay. Extended dosing in rats through the end of gestation and lactation (pre- and post-natal development study) caused impaired skin development, reduced offspring viability and growth retardation. The spectrum of developmental effects in rats is consistent with the intended pharmacology (altered triglyceride metabolism) and the transient nature of the skeletal findings, along with the late gestational window of sensitivity for the effects on skin barrier development, reduce the concern for potential adverse developmental effects following unintended early gestational exposure to ervogastat in humans where treatment can be discontinued once pregnancy is determined.
Collapse
Affiliation(s)
- Natasha R Catlin
- Drug Safety Research, Development, & Medical, Pfizer Worldwide Research & Development, Groton, CT, USA
| | - Christopher J Bowman
- Drug Safety Research, Development, & Medical, Pfizer Worldwide Research & Development, Groton, CT, USA
| | - Sarah N Campion
- Drug Safety Research, Development, & Medical, Pfizer Worldwide Research & Development, Groton, CT, USA
| | - Elise M Lewis
- Charles River Laboratories, Inc, Safety Assessment, Horsham, PA, USA
| | - William S Nowland
- Drug Safety Research, Development, & Medical, Pfizer Worldwide Research & Development, Groton, CT, USA
| | - Christine Stethem
- Drug Safety Research, Development, & Medical, Pfizer Worldwide Research & Development, Groton, CT, USA
| | - Gregg D Cappon
- Drug Safety Research, Development, & Medical, Pfizer Worldwide Research & Development, Groton, CT, USA
| |
Collapse
|
22
|
Fayezi S, Mehdizadeh A, Germeyer A, Strowitzki T, Fayyazpour P, Nowrouzi Z, Zarezadeh R. Maternal erythrocyte fatty acid composition as a predictive marker for pregnancy health. Biofactors 2022; 48:763-778. [PMID: 35357720 DOI: 10.1002/biof.1840] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/14/2022] [Indexed: 12/11/2022]
Abstract
Pregnancy is accompanied by a surge in demand for fatty acids (FAs) in order to support maternal health, as well as fetal growth and development. Of particular demand is essential for long-chain polyunsaturated FAs. FAs are primarily obtained from dietary sources and are distributed in the body. In comparison with the use of self-reporting approaches, measuring the FA levels within different blood compartments can present a more accurate image of nutritional, and thus tissue, FA composition. Hence, the FA profile of plasma or serum is commonly used for physiological analyses. Nevertheless, plasma and serum FAs are not yet incorporated into cell membranes, and consequently may not be a suitable reflection of the FA status of body tissues. The evaluation of erythrocyte FA levels offers a superior possibility for the following reasons: the biological fluctuation of erythrocyte FA composition is low, phospholipids account for almost all the lipid content of erythrocytes, and the FA profiles of erythrocytes represent those of tissues. Here, we elaborate on whether the status of maternal erythrocyte FAs can serve as a prognostic biomarker for reproductive health and fetomaternal complications, including embryonic and fetoplacental development, gestational length, and preeclampsia. In addition, factors with the potential of altering the maternal erythrocyte FAs such as maternal diet, lifestyle habits, genetics, and body composition are discussed.
Collapse
Affiliation(s)
- Shabnam Fayezi
- Department of Gynecologic Endocrinology and Fertility Disorders, Women's Hospital, Ruprecht-Karls University of Heidelberg, Heidelberg, Germany
- Department of Reproductive Biology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Mehdizadeh
- Hematology and Oncology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ariane Germeyer
- Department of Gynecologic Endocrinology and Fertility Disorders, Women's Hospital, Ruprecht-Karls University of Heidelberg, Heidelberg, Germany
| | - Thomas Strowitzki
- Department of Gynecologic Endocrinology and Fertility Disorders, Women's Hospital, Ruprecht-Karls University of Heidelberg, Heidelberg, Germany
| | - Parisa Fayyazpour
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Student's Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zahra Nowrouzi
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Student's Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Zarezadeh
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Student's Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
- Endocrine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
23
|
Allman BR, Spray BJ, Lan RS, Andres A, Børsheim E. Circulating long-chain acylcarnitine concentrations are not affected by exercise training in pregnant women with obesity. J Appl Physiol (1985) 2022; 132:470-476. [PMID: 34989648 PMCID: PMC8816616 DOI: 10.1152/japplphysiol.00712.2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
The purpose of this study was to determine the effect of exercise during pregnancy in sedentary women with obesity on longitudinal changes in long-chain acylcarnitine (LC-AC) concentrations. We hypothesized that exercise training would significantly decrease circulating LC-ACs throughout gestation compared with a nonexercise control group. Pregnant women with obesity considered otherwise healthy [n = 80, means ± SD; body mass index (BMI): 36.9 ± 5.7 kg/m2] were randomized into an exercise (n = 40, aerobic/resistance 3 times/wk, ∼13th gestation week until birth) or a nonexercise control (n = 40) group. At gestation week 12.2 ± 0.5 and 36.0 ± 0.4, a submaximal exercise test was conducted, and indirect calorimetry was used to measure relative resting energy expenditure (REE), as well as respiratory exchange ratio (RER) at rest. Fasting blood samples were collected and analyzed for LC-AC concentrations. Fitness improved with prenatal exercise training; however, exercise training did not affect circulating LC-AC. When groups were collapsed, LC-ACs decreased during gestation (combined groups, P < 0.001), whereas REE (kcal/kg/day, P = 0.008) increased. However, average REE relative to fat-free mass (FFM) (kcal/kg FFM/day) and RER did not change. There was an inverse relationship between the change in RER and all LC-ACs (except C18:2) throughout gestation (C14: r = -0.26, P = 0.04; C16: r = -0.27, P = 0.03; C18:1: r = -0.28, P = 0.02). In summary, a moderate-intensity exercise intervention during pregnancy in women with obesity did not alter LC-ACs concentrations versus control, indicating that the balance between long-chain fatty acid availability and oxidation neither improved nor worsened with an exercise intervention.NEW & NOTEWORTHY This research showed that a moderate-intensity prenatal exercise program, consisting of aerobic and resistance training, did not negatively impact normal alterations in substrate supply and demand for the mother and the offspring throughout gestation. Findings provide support for metabolic safety of exercise during pregnancy.
Collapse
Affiliation(s)
- Brittany R. Allman
- 1Arkansas Children’s Nutrition Center, Little Rock, Arkansas,2Arkansas Children’s Research Institute, Little Rock, Arkansas,3Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | | | - Renny S. Lan
- 1Arkansas Children’s Nutrition Center, Little Rock, Arkansas,3Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Aline Andres
- 1Arkansas Children’s Nutrition Center, Little Rock, Arkansas,2Arkansas Children’s Research Institute, Little Rock, Arkansas,3Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Elisabet Børsheim
- 1Arkansas Children’s Nutrition Center, Little Rock, Arkansas,2Arkansas Children’s Research Institute, Little Rock, Arkansas,3Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas,4Department of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| |
Collapse
|
24
|
Röszer T. Co-Evolution of Breast Milk Lipid Signaling and Thermogenic Adipose Tissue. Biomolecules 2021; 11:1705. [PMID: 34827703 PMCID: PMC8615456 DOI: 10.3390/biom11111705] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/06/2021] [Accepted: 11/10/2021] [Indexed: 12/16/2022] Open
Abstract
Breastfeeding is a unique and defining behavior of mammals and has a fundamental role in nourishing offspring by supplying a lipid-rich product that is utilized to generate heat and metabolic fuel. Heat generation from lipids is a feature of newborn mammals and is mediated by the uncoupling of mitochondrial respiration in specific fat depots. Breastfeeding and thermogenic adipose tissue have a shared evolutionary history: both have evolved in the course of homeothermy evolution; breastfeeding mammals are termed "thermolipials", meaning "animals with warm fat". Beyond its heat-producing capacity, thermogenic adipose tissue is also necessary for proper lipid metabolism and determines adiposity in offspring. Recent advances have demonstrated that lipid metabolism in infants is orchestrated by breast milk lipid signals, which establish mother-to-child signaling and control metabolic development in the infant. Breastfeeding rates are declining worldwide, and are paralleled by an alarming increase in childhood obesity, which at least in part may have its roots in the impaired metabolic control by breast milk lipid signals.
Collapse
Affiliation(s)
- Tamás Röszer
- Institute of Neurobiology, Faculty of Science, Ulm University, 89081 Ulm, Germany
| |
Collapse
|
25
|
Kuentzel KB, Bradić I, Akhmetshina A, Korbelius M, Rainer S, Kolb D, Gauster M, Vujić N, Kratky D. Defective Lysosomal Lipolysis Causes Prenatal Lipid Accumulation and Exacerbates Immediately after Birth. Int J Mol Sci 2021; 22:10416. [PMID: 34638755 PMCID: PMC8508985 DOI: 10.3390/ijms221910416] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/24/2021] [Accepted: 09/24/2021] [Indexed: 12/03/2022] Open
Abstract
Cholesterol and fatty acids are essential lipids that are critical for membrane biosynthesis and fetal organ development. Cholesteryl esters (CE) are degraded by hormone-sensitive lipase (HSL) in the cytosol and by lysosomal acid lipase (LAL) in the lysosome. Impaired LAL or HSL activity causes rare pathologies in humans, with HSL deficiency presenting less severe clinical manifestations. The infantile form of LAL deficiency, a lysosomal lipid storage disorder, leads to premature death. However, the importance of defective lysosomal CE degradation and its consequences during early life are incompletely understood. We therefore investigated how defective CE catabolism affects fetus and infant maturation using Lal and Hsl knockout (-/-) mouse models. This study demonstrates that defective lysosomal but not neutral lipolysis alters placental and fetal cholesterol homeostasis and exhibits an initial disease pathology already in utero as Lal-/- fetuses accumulate hepatic lysosomal lipids. Immediately after birth, LAL deficiency exacerbates with massive hepatic lysosomal lipid accumulation, which continues to worsen into young adulthood. Our data highlight the crucial role of LAL during early development, with the first weeks after birth being critical for aggravating LAL deficiency.
Collapse
Affiliation(s)
- Katharina B. Kuentzel
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (K.B.K.); (I.B.); (A.A.); (M.K.); (S.R.); (N.V.)
| | - Ivan Bradić
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (K.B.K.); (I.B.); (A.A.); (M.K.); (S.R.); (N.V.)
| | - Alena Akhmetshina
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (K.B.K.); (I.B.); (A.A.); (M.K.); (S.R.); (N.V.)
| | - Melanie Korbelius
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (K.B.K.); (I.B.); (A.A.); (M.K.); (S.R.); (N.V.)
| | - Silvia Rainer
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (K.B.K.); (I.B.); (A.A.); (M.K.); (S.R.); (N.V.)
| | - Dagmar Kolb
- Gottfried Schatz Research Center, Cell Biology, Histology and Embryology, Medical University of Graz, 8010 Graz, Austria; (D.K.); (M.G.)
- Core Facility Ultrastructural Analysis, Medical University of Graz, 8010 Graz, Austria
- BioTechMed-Graz, 8010 Graz, Austria
| | - Martin Gauster
- Gottfried Schatz Research Center, Cell Biology, Histology and Embryology, Medical University of Graz, 8010 Graz, Austria; (D.K.); (M.G.)
| | - Nemanja Vujić
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (K.B.K.); (I.B.); (A.A.); (M.K.); (S.R.); (N.V.)
| | - Dagmar Kratky
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (K.B.K.); (I.B.); (A.A.); (M.K.); (S.R.); (N.V.)
- BioTechMed-Graz, 8010 Graz, Austria
| |
Collapse
|
26
|
Hoang AC, Yu H, Röszer T. Transcriptional Landscaping Identifies a Beige Adipocyte Depot in the Newborn Mouse. Cells 2021; 10:2368. [PMID: 34572017 PMCID: PMC8470180 DOI: 10.3390/cells10092368] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 08/31/2021] [Accepted: 09/05/2021] [Indexed: 12/12/2022] Open
Abstract
The present study sought to identify gene networks that are hallmarks of the developing inguinal subcutaneous adipose tissue (iWAT) and the interscapular brown adipose tissue (BAT) in the mouse. RNA profiling revealed that the iWAT of postnatal (P) day 6 mice expressed thermogenic and lipid catabolism transcripts, along with the abundance of transcripts associated with the beige adipogenesis program. This was an unexpected finding, as thermogenic BAT was believed to be the only site of nonshivering thermogenesis in the young mouse. However, the transcriptional landscape of BAT in P6 mice suggests that it is still undergoing differentiation and maturation, and that the iWAT temporally adopts thermogenic and lipolytic potential. Moreover, P6 iWAT and adult (P56) BAT were similar in their expression of immune gene networks, but P6 iWAT was unique in the abundant expression of antimicrobial proteins and virus entry factors, including a possible receptor for SARS-CoV-2. In summary, postnatal iWAT development is associated with a metabolic shift from thermogenesis and lipolysis towards fat storage. However, transcripts of beige-inducing signal pathways including β-adrenergic receptors and interleukin-4 signaling were underrepresented in young iWAT, suggesting that the signals for thermogenic fat differentiation may be different in early postnatal life and in adulthood.
Collapse
MESH Headings
- Adipocytes, Beige/metabolism
- Adipose Tissue, Brown/metabolism
- Adipose Tissue, White/metabolism
- Animals
- Animals, Newborn
- Biomarkers/metabolism
- Cell Cycle/genetics
- Gene Expression Regulation, Developmental
- Gene Ontology
- Gene Regulatory Networks
- Male
- Mice, Inbred C57BL
- Models, Biological
- Muscle Development/genetics
- Neuropeptides/metabolism
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Signal Transduction
- Transcription, Genetic
- Mice
Collapse
Affiliation(s)
| | | | - Tamás Röszer
- Institute of Neurobiology, Ulm University, 89081 Ulm, Germany; (A.C.H.); (H.Y.)
| |
Collapse
|
27
|
Vidarsdottir H, Halldorsson TI, Geirsson RT, Bjarnason R, Franzson L, Valdimarsdottir UA, Thorkelsson T. Mode of delivery was associated with transient changes in the metabolomic profile of neonates. Acta Paediatr 2021; 110:2110-2118. [PMID: 33636029 DOI: 10.1111/apa.15822] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 02/22/2021] [Accepted: 02/25/2021] [Indexed: 11/28/2022]
Abstract
AIMS To estimate potential differences in neonatal metabolomic profiles at birth and at the time of newborn screening by delivery mode. METHODS A prospective study at Women's Clinic at Landspitali-The National University Hospital of Iceland. Women having normal vaginal birth or elective caesarean section from November 2013 to April 2014 were offered participation. Blood samples from mothers before birth and umbilical cord at birth were collected and amino acids and acylcarnitines measured by tandem mass spectrometry. Results from the Newborn screening programme in Iceland were collected. Amino acids and acylcarnitines from different samples were compared by delivery mode. RESULTS Eighty three normal vaginal births and 32 elective caesarean sections were included. Mean differences at birth were higher for numerous amino acids, and some acylcarnitines in neonates born vaginally compared to elective caesarean section. Maternal blood samples and newborn screening results showed small differences that lost significance after correction for multiple testing. Many amino acids and some acylcarnitines were numerically higher in cord blood compared to maternal. Many amino acids and most acylcarnitines were numerically higher in newborn screening results compared to cord blood. CONCLUSION We observed transient yet distinct differences in metabolomic profiles between neonates by delivery mode.
Collapse
Affiliation(s)
- Harpa Vidarsdottir
- Faculty of Medicine School of Health Sciences University of Iceland Reykjavik Iceland
- Department of Neonatology Astrid Lindgren Children's Hospital Karolinska University Hospital Stockholm Sweden
| | | | - Reynir Tomas Geirsson
- Faculty of Medicine School of Health Sciences University of Iceland Reykjavik Iceland
- Women's Clinic Landspitali – The National University Hospital of Iceland Reykjavik Iceland
| | - Ragnar Bjarnason
- Faculty of Medicine School of Health Sciences University of Iceland Reykjavik Iceland
- Children's Hospital Iceland Landspitali – The National University Hospital of Iceland Reykjavik Iceland
| | - Leifur Franzson
- Faculty of Pharmaceutical Sciences School of Health Science University of Iceland Reykjavik Iceland
- Department of Genetics and Molecular Medicine Landspitali – The National University Hospital of Iceland Reykjavik Iceland
| | - Unnur Anna Valdimarsdottir
- Center for Public Health Science School of Health Science University of Iceland Reykjavik Iceland
- Department of Medical Epidemiology and Biostatistics Karolinska Institutet Stockholm Sweden
- Department of Epidemiology Harvard T H Chan School of Public Health Boston MA USA
| | - Thordur Thorkelsson
- Faculty of Medicine School of Health Sciences University of Iceland Reykjavik Iceland
- Children's Hospital Iceland Landspitali – The National University Hospital of Iceland Reykjavik Iceland
| |
Collapse
|
28
|
Auclair N, Sané AT, Ahmarani L, Patey N, Beaulieu JF, Peretti N, Spahis S, Levy E. Sar1b mutant mice recapitulate gastrointestinal abnormalities associated with chylomicron retention disease. J Lipid Res 2021; 62:100085. [PMID: 33964306 PMCID: PMC8175419 DOI: 10.1016/j.jlr.2021.100085] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/15/2021] [Accepted: 04/17/2021] [Indexed: 11/17/2022] Open
Abstract
Chylomicron retention disease (CRD) is an autosomal recessive disorder associated with biallelic Sar1b mutations leading to defects in intracellular chylomicron (CM) trafficking and secretion. To date, a direct cause-effect relationship between CRD and Sar1b mutation has not been established, but genetically modified animal models provide an opportunity to elucidate unrecognized aspects of these mutations. To examine the physiological role and molecular mechanisms of Sar1b function, we generated mice expressing either a targeted deletion or mutation of human Sar1b using the CRISPR-Cas9 system. We found that deletion or mutation of Sar1b in mice resulted in late-gestation lethality of homozygous embryos. Moreover, compared with WT mice, heterozygotes carrying a single disrupted Sar1b allele displayed lower plasma levels of triglycerides, total cholesterol, and HDL-cholesterol, along with reduced CM secretion following gastric lipid gavage. Similarly, decreased expression of apolipoprotein B and microsomal triglyceride transfer protein was observed in correlation with the accumulation of mucosal lipids. Inefficient fat absorption in heterozygotes was confirmed via an increase in fecal lipid excretion. Furthermore, genetically modified Sar1b affected intestinal lipid homeostasis as demonstrated by enhanced fatty acid β-oxidation and diminished lipogenesis through the modulation of transcription factors. This is the first reported mammalian animal model with human Sar1b genetic defects, which reproduces some of the characteristic CRD features and provides a direct cause-effect demonstration.
Collapse
Affiliation(s)
- Nickolas Auclair
- Research Center, CHU Ste-Justine, Université de Montréal, Montreal, Quebec, Canada; Department of Pharmacology & Physiology, Université de Montréal, Montreal, Quebec, Canada
| | - Alain T Sané
- Research Center, CHU Ste-Justine, Université de Montréal, Montreal, Quebec, Canada
| | - Lena Ahmarani
- Research Center, CHU Ste-Justine, Université de Montréal, Montreal, Quebec, Canada; Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada
| | - Nathalie Patey
- Research Center, CHU Ste-Justine, Université de Montréal, Montreal, Quebec, Canada; Department of Pathology, Université de Montréal, Montreal, Quebec, Canada
| | - Jean-François Beaulieu
- Laboratory of Intestinal Physiopathology, Department of Immunology and Cell Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Noel Peretti
- Department of Pediatric Gastroenterology-Hepatology and Nutrition, Laboratory INSERM 1060 Cardiovascular Metabolism Endocrinology and Nutrition CarMEN, Lyon, France
| | - Schohraya Spahis
- Research Center, CHU Ste-Justine, Université de Montréal, Montreal, Quebec, Canada; Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada
| | - Emile Levy
- Research Center, CHU Ste-Justine, Université de Montréal, Montreal, Quebec, Canada; Department of Pharmacology & Physiology, Université de Montréal, Montreal, Quebec, Canada; Department of Nutrition, Université de Montréal, Montreal, Quebec, Canada.
| |
Collapse
|
29
|
da Costa Faria NR, Chaves-Filho AB, Alcantara LCJ, de Siqueira IC, Calcagno JI, Miyamoto S, de Filippis AMB, Yoshinaga MY. Plasma lipidome profiling of newborns with antenatal exposure to Zika virus. PLoS Negl Trop Dis 2021; 15:e0009388. [PMID: 33930014 PMCID: PMC8115770 DOI: 10.1371/journal.pntd.0009388] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 05/12/2021] [Accepted: 04/14/2021] [Indexed: 02/07/2023] Open
Abstract
The 2015–2016 Zika virus (ZIKV) outbreak in Brazil was remarkably linked to the incidence of microcephaly and other deleterious clinical manifestations, including eye abnormalities, in newborns. It is known that ZIKV targets the placenta, triggering an inflammatory profile that may cause placental insufficiency. Transplacental lipid transport is delicately regulated during pregnancy and deficiency on the delivery of lipids such as arachidonic and docosahexaenoic acids may lead to deficits in both brain and retina during fetal development. Here, plasma lipidome profiles of ZIKV exposed microcephalic and normocephalic newborns were compared to non-infected controls. Our results reveal major alterations in circulating lipids from both ZIKV exposed newborns with and without microcephaly relative to controls. In newborns with microcephaly, the plasma concentrations of hydroxyoctadecadienoic acid (HODE), primarily as 13-HODE isomer, derived from linoleic acid were higher as compared to normocephalic ZIKV exposed newborns and controls. Total HODE concentrations were also positively associated with levels of other oxidized lipids and several circulating free fatty acids in newborns, indicating a possible plasma lipidome signature of microcephaly. Moreover, higher concentrations of lysophosphatidylcholine in ZIKV exposed normocephalic newborns relative to controls suggest a potential disruption of polyunsaturated fatty acids transport across the blood-brain barrier of fetuses. The latter data is particularly important given the neurocognitive and neurodevelopmental abnormalities observed in follow-up studies involving children with antenatal ZIKV exposure, but normocephalic at birth. Taken together, our data reveal that plasma lipidome alterations associated with antenatal exposure to ZIKV could contribute to identification and monitoring of the wide spectrum of clinical phenotypes at birth and further, during childhood. Antenatal exposure to Zika virus (ZIKV) is linked to a wide range of clinical presentations at birth, from asymptomatic cases to microcephaly, and other neurocognitive and neurodevelopmental abnormalities manifested in the early childhood. Stratification of these clinical phenotypes in newborns with suspected antenatal ZIKV exposure is challenging, but critical to improve early assessment of rehabilitative interventions. In this study, plasma lipidome profiling of 274 lipid species was performed in both normocephalic and microcephalic newborns with antenatal ZIKV exposure and compared to non-infected controls. Multiple lipid species were independent predictors of antenatal ZIKV exposure. More specifically, microcephaly was strongly associated with an oxidized free fatty acid and ZIKV exposed normocephalic newborns exhibited higher plasma concentrations of lysophosphatidylcholine relative to controls. These findings emphasize the need for studies focused on the role of individual lipids in neuropathogenesis of ZIKV and raise the potential of plasma lipidome profiling for early diagnosis of newborns with suspected antenatal ZIKV exposure. To validate the predictive ability of this approach, prospective studies with a larger cohort of newborns are now required.
Collapse
Affiliation(s)
| | | | | | | | - Juan Ignacio Calcagno
- Maternidade Prof. José Maria de Magalhães Netto, State Health Secretary (Salvador), Bahia, Brazil
| | - Sayuri Miyamoto
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | | | - Marcos Yukio Yoshinaga
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
30
|
Ikenoue S, Waffarn F, Ohashi M, Tanaka M, Gillen DL, Buss C, Entringer S, Wadhwa PD. Placental Corticotrophin-Releasing Hormone is a Modulator of Fetal Liver Blood Perfusion. J Clin Endocrinol Metab 2021; 106:646-653. [PMID: 33313841 PMCID: PMC7947764 DOI: 10.1210/clinem/dgaa908] [Citation(s) in RCA: 3] [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] [Received: 06/10/2020] [Indexed: 12/26/2022]
Abstract
CONTEXT Variation in fetal liver blood flow influences fetal growth and postnatal body composition. Placental corticotrophin-releasing hormone has been implicated as a key mediator of placental-fetal perfusion. OBJECTIVE To determine whether circulating levels of placental corticotrophin-releasing hormone across gestation are associated with variations in fetal liver blood flow. DESIGN Prospective cohort study. METHODS Fetal ultrasonography was performed at 30 weeks' gestation to characterize fetal liver blood flow (quantified by subtracting ductus venosus flow from umbilical vein flow). Placental corticotrophin-releasing hormone was measured in maternal circulation at approximately 12, 20, and 30 weeks' gestation. Multiple regression analysis was used to determine the proportion of variation in fetal liver blood flow explained by placental corticotrophin-releasing hormone. Covariates included maternal age, parity, pre-pregnancy body mass index, gestational weight gain, and fetal sex. RESULTS A total of 79 uncomplicated singleton pregnancies were analyzed. Fetal liver blood flow was 68.4 ± 36.0 mL/min (mean ± SD). Placental corticotrophin-releasing hormone concentrations at 12, 20, and 30 weeks were 12.5 ± 8.1, 35.7 ± 24.5, and 247.9 ± 167.8 pg/mL, respectively. Placental corticotrophin-releasing hormone at 30 weeks, but not at 12 and 20 weeks, was significantly and positively associated with fetal liver blood flow at 30 weeks (r = 0.319; P = 0.004) and explained 10.4% of the variance in fetal liver blood flow. CONCLUSIONS Placental corticotrophin-releasing hormone in late gestation is a possible modulator of fetal liver blood flow and may constitute a biochemical marker in clinical investigations of fetal growth and body composition.
Collapse
Affiliation(s)
- Satoru Ikenoue
- Development, Health and Disease Research Program, University of California, Irvine, Irvine, CA 92697, USA
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Feizal Waffarn
- Development, Health and Disease Research Program, University of California, Irvine, Irvine, CA 92697, USA
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA
| | - Masanao Ohashi
- Development, Health and Disease Research Program, University of California, Irvine, Irvine, CA 92697, USA
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA
- Department of Obstetrics and Gynecology, University of Miyazaki, Miyazaki, Japan
| | - Mamoru Tanaka
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Daniel L Gillen
- Department of Statistics, University of California, Irvine, Irvine, CA, USA
| | - Claudia Buss
- Development, Health and Disease Research Program, University of California, Irvine, Irvine, CA 92697, USA
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA
- Institute of Medical Psychology, Charité University Medicine, Berlin, Germany
| | - Sonja Entringer
- Development, Health and Disease Research Program, University of California, Irvine, Irvine, CA 92697, USA
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA
- Institute of Medical Psychology, Charité University Medicine, Berlin, Germany
| | - Pathik D Wadhwa
- Development, Health and Disease Research Program, University of California, Irvine, Irvine, CA 92697, USA
- Department of Pediatrics, University of California, Irvine, Irvine, CA, USA
- Department of Psychiatry and Human Behavior, University of California, Irvine, Irvine, CA, USA
- Department of Obstetrics and Gynecology, University of California, Irvine, Irvine, CA, USA
- Department of Epidemiology, University of California, Irvine, Irvine, CA, USA
- Correspondence: Pathik D. Wadhwa, MD, PhD, UCI Development, Health and Disease Research Program, University of California, Irvine, 3117 Gillespie, Irvine, CA 92697, USA.
| |
Collapse
|
31
|
Chen W, Lu Y, Hu D, Mo J, Ni J. Black mulberry (Morus nigra L.) polysaccharide ameliorates palmitate-induced lipotoxicity in hepatocytes by activating Nrf2 signaling pathway. Int J Biol Macromol 2021; 172:394-407. [PMID: 33450344 DOI: 10.1016/j.ijbiomac.2021.01.059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 01/06/2021] [Accepted: 01/10/2021] [Indexed: 01/05/2023]
Abstract
Black mulberry (Morus nigra L.) has shown health benefits against metabolic disorders. Lipotoxicity is considered as a potentially cause of metabolic syndrome, and there is no effective treatment. However, the protective effect and its mechanism of black mulberry against lipotoxicity are unclear. In this study, three polysaccharide fractions (BP1, BP2, BP3) were isolated from black mulberry by stepwise precipitation with 30%, 60%, and 90% of ethanol and analyzed by GPC, HPLC and FT-IR methods. BP1 exhibited a better protective effect than BP2 and BP3 on palmitic acid (PA)-induced lipotoxicity in HepG2 cells. BP1 effectively reduced PA-induced lipotoxicity by eliminating accumulation of ROS, improving mitochondrial function, reversing glutathione depletion and enhancing antioxidant enzyme activities. Mechanistically, BP1 activated the Nrf2 signaling pathway, a master regulator of the antioxidant defense system, through increasing Nrf2 nuclear translocation and phosphorylation. Collectively, these results demonstrate that BP1 has the great potential for applications in lipid disorders.
Collapse
Affiliation(s)
- Wei Chen
- Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang University, Hangzhou 310058, China; Ningbo Research Institute, Zhejiang University, Ningbo 315100, China.
| | - Yang Lu
- Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang University, Hangzhou 310058, China
| | - Dongwen Hu
- Department of Food Science and Nutrition, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang University, Hangzhou 310058, China
| | - Jianling Mo
- Department of Traditional Chinese Medicine, Sir Run Run Shaw Hospital, School of Medcine, Zhejiang University, Hangzhou 310016, China
| | - Jingdan Ni
- Department of Traditional Chinese Medicine, Sir Run Run Shaw Hospital, School of Medcine, Zhejiang University, Hangzhou 310016, China
| |
Collapse
|
32
|
Mauri M, Calmarza P, Ibarretxe D. Dyslipemias and pregnancy, an update. CLINICA E INVESTIGACION EN ARTERIOSCLEROSIS 2020; 33:41-52. [PMID: 33309071 DOI: 10.1016/j.arteri.2020.10.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Revised: 09/25/2020] [Accepted: 10/05/2020] [Indexed: 12/12/2022]
Abstract
During pregnancy there is a physiological increase in total cholesterol (TC) and triglycerides (TG) plasma concentrations, due to increased insulin resistance, oestrogens, progesterone, and placental lactogen, although their reference values are not exactly known, TG levels can increase up to 300mg/dL, and TC can go as high as 350mg/dL. When the cholesterol concentration exceeds the 95th percentile (familial hypercholesterolaemia (FH) and transient maternal hypercholesterolaemia), there is a predisposition to oxidative stress in foetal vessels, exposing the newborn to a greater fatty streaks formation and a higher risk of atherosclerosis. However, the current treatment of pregnant women with hyperlipidaemia consists of a diet and suspension of lipid-lowering drugs. The most prevalent maternal hypertriglyceridaemia (HTG) is due to secondary causes, like diabetes, obesity, drugs, etc. The case of severe HTG due to genetic causes is less prevalent, and can be a higher risk of maternal-foetal complications, such as, acute pancreatitis (AP), pre-eclampsia, preterm labour, and gestational diabetes. Severe HTG-AP is a rare but potentially lethal pregnancy complication, for the mother and the foetus, usually occurs during the third trimester or in the immediate postpartum period, and there are no specific protocols for its diagnosis and treatment. In conclusion, it is crucial that dyslipidaemia during pregnancy must be carefully evaluated, not just because of the acute complications, but also because of the future cardiovascular morbidity and mortality of the newborn child. That is why the establishment of consensus protocols or guidelines is essential for its management.
Collapse
Affiliation(s)
- Marta Mauri
- Unidad de Lípidos, Servicio de Medicina Interna, Hospital de Terrassa, Consorci Sanitari de Terrassa, Terrassa, Barcelona, España
| | - Pilar Calmarza
- Servicio de Bioquímica Clínica, Hospital Universitario Miguel Servet, Universidad de Zaragoza, Zaragoza, España.
| | - Daiana Ibarretxe
- Unidad de Medicina Vascular y Metabolismo (UVASMET), Hospital Universitario de Reus, Universidad Rovira y Virgili, IISPV, CIBERDEM, Reus, Tarragona, España
| |
Collapse
|
33
|
Catlin NR, Bowman CJ, Campion SN, Davenport SD, Esler WP, Kumpf SW, Lewis EM, Nowland WS, Ross TT, Stedman DS, Stethem C, Cappon GD. Inhibition of ACC causes malformations in rats and rabbits: comparison of mammalian findings and alternative assays. Toxicol Sci 2020; 179:183-194. [PMID: 33247737 DOI: 10.1093/toxsci/kfaa169] [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: 11/12/2022] Open
Abstract
Acetyl-CoA carboxylase (ACC) is an enzyme within the de novo lipogenesis (DNL) pathway and plays a role in regulating lipid metabolism. Pharmacologic ACC inhibition has been an area of interest for multiple potential indications including oncology, acne vulgaris, metabolic diseases such as type 2 diabetes mellitus, and non-alcoholic fatty liver disease/non-alcoholic steatohepatitis. A critical role for ACC in de novo synthesis of long-chain fatty acids during fetal development has been demonstrated in studies in mice lacking Acc1, where the absence of Acc1 results in early embryonic lethality. Following positive predictions of developmental toxicity in alternative in vitro assays (positive in murine embryonic stem cell [mESC] assay and rat whole embryo culture, but negative in zebrafish), developmental toxicity (growth retardation and dysmorphogenesis associated with disrupted midline fusion) was observed with the oral administration of the dual ACC1 and 2 inhibitor, PF-05175157, in Sprague Dawley rats and New Zealand White rabbits. The results of these studies are presented here to make comparisons across the assays, as well as mechanistic insights from the mESC assay demonstrating high ACC expression in the mESC and that ACC induced developmental toxicity can be rescued with palmitic acid providing supportive evidence for DNL pathway inhibition as the underlying mechanism. Ultimately, while the battery of alternative approaches and weight-of-evidence case were useful for hazard identification, the embryo-fetal development studies were necessary to inform the risk assessment on the adverse fetal response, as malformations and/or embryo fetal lethality were limited to doses that caused near complete inhibition of DNL.
Collapse
Affiliation(s)
- Natasha R Catlin
- Drug Safety Research and Development, Pfizer Worldwide Research & Development, Groton, CT, USA
| | - Christopher J Bowman
- Drug Safety Research and Development, Pfizer Worldwide Research & Development, Groton, CT, USA
| | - Sarah N Campion
- Drug Safety Research and Development, Pfizer Worldwide Research & Development, Groton, CT, USA
| | - Scott D Davenport
- Drug Safety Research and Development, Pfizer Worldwide Research & Development, Groton, CT, USA
| | - William P Esler
- Internal Medicine Research Unit, Pfizer Worldwide Research & Development, Cambridge, MA, USA
| | - Steven W Kumpf
- Drug Safety Research and Development, Pfizer Worldwide Research & Development, Groton, CT, USA
| | - Elise M Lewis
- Charles River Laboratories, Inc, Safety Assessment, Horsham, PA, USA
| | - William S Nowland
- Drug Safety Research and Development, Pfizer Worldwide Research & Development, Groton, CT, USA
| | - Trenton T Ross
- Internal Medicine Research Unit, Pfizer Worldwide Research & Development, Cambridge, MA, USA
| | - Donald S Stedman
- Drug Safety Research and Development, Pfizer Worldwide Research & Development, Groton, CT, USA
| | - Christine Stethem
- Drug Safety Research and Development, Pfizer Worldwide Research & Development, Groton, CT, USA
| | - Gregg D Cappon
- Drug Safety Research and Development, Pfizer Worldwide Research & Development, Groton, CT, USA
| |
Collapse
|
34
|
Carneros D, Medina-Gómez G, Giralt M, León-Camacho M, Campbell M, Moreno-Aliaga MJ, Villarroya F, Bustos M. Cardiotrophin-1 contributes to metabolic adaptations through the regulation of lipid metabolism and to the fasting-induced fatty acid mobilization. FASEB J 2020; 34:15875-15887. [PMID: 33047392 DOI: 10.1096/fj.202000109r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 09/03/2020] [Accepted: 09/16/2020] [Indexed: 11/11/2022]
Abstract
It is becoming clear that several human pathologies are caused by altered metabolic adaptations. During liver development, there are physiological changes, from the predominant utilization of glucose (fetal life) to the use of lipids (postnatal life). Fasting is another physiological stress that elicits well-known metabolic adjustments. We have reported the metabolic properties of cardiotrophin-1 (CT-1), a member of the interleukin-6 family of cytokines. Here, we aimed at analyzing the role of CT-1 in response to these metabolic changes. We used different in vivo models. Furthermore, a differential study was carried out with wild-type and CT-1 null mice in fed (ad libitum) and food-restricted conditions. We demonstrated that Ct-1 is a metabolic gene induced in the liver via PPARα in response to lipids in mice (neonates- and food-restricted adults). We found that Ct-1 mRNA expression in white adipose tissue directly involved PPARα and PPARγ. Finally, the physiological role of CT-1 in fasting is confirmed by the impaired food restriction-induced adipose tissue lipid mobilization in CT-1 null mice. Our findings support a previously unrecognized physiological role of CT-1 in metabolic adaptations, through the regulation of lipid metabolism and contributes to fasting-induced free fatty acid mobilization.
Collapse
Affiliation(s)
- David Carneros
- Area of Liver, Digestive and Inflammatory Diseases, Institute of Biomedicine of Seville (IBiS), Consejo Superior de Investigaciones Científicas (CSIC), University of Seville, Virgen del Rocio University Hospital, Seville, Spain
| | - Gema Medina-Gómez
- Department of Basic Sciences of Health, Area of Biochemistry and Molecular Biology, Rey Juan Carlos University, Madrid, Spain
| | - Marta Giralt
- Department of Biochemistry and Molecular Biomedicine, Barcelona University, Barcelona, Spain.,CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III (ISCIII), Madrid, Spain
| | - Manuel León-Camacho
- Department of Lipid Characterization and Quality, Instituto de la Grasa (CSIC), Seville, Spain
| | - Mark Campbell
- MRC MDU, Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK
| | - Maria J Moreno-Aliaga
- CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III (ISCIII), Madrid, Spain.,Centre for Nutrition Research and Department of Nutrition, Food Science and Physiology School of Pharmacy and Nutrition, University of Navarra, Navarra's Health Research Institute (IdiSNA), Pamplona, Spain
| | - Francesc Villarroya
- Department of Biochemistry and Molecular Biomedicine, Barcelona University, Barcelona, Spain.,CIBERobn Physiopathology of Obesity and Nutrition, Institute of Health Carlos III (ISCIII), Madrid, Spain
| | - Matilde Bustos
- Area of Liver, Digestive and Inflammatory Diseases, Institute of Biomedicine of Seville (IBiS), Consejo Superior de Investigaciones Científicas (CSIC), University of Seville, Virgen del Rocio University Hospital, Seville, Spain
| |
Collapse
|
35
|
Cheatham CL. Nutritional Factors in Fetal and Infant Brain Development. ANNALS OF NUTRITION AND METABOLISM 2020; 75 Suppl 1:20-32. [PMID: 32564018 DOI: 10.1159/000508052] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 02/12/2020] [Indexed: 11/19/2022]
Abstract
Fetal and infant brain development determine the trajectory of the organism across the lifespan. Optimal maternal and infant nutrition during the period of rapid brain development is vital to the integrity of the neural substrate for subsequent lifelong functions. The goal of this review is to educate the reader on the effects of fetal and infant nutrition on the developing human brain. A review of the literature reveals 6 nutrients that have been studied with respect to maternal nutrition and subsequent offspring brain development: folate, iodine, iron, vitamin D, choline, and docosahexaenoic acid (DHA; 22:6n-3). The research is discussed with a focus on the timing of nutrient needs (preconception, prenatally, and postnatally) as well as potential confounding and unobserved variables.
Collapse
Affiliation(s)
- Carol L Cheatham
- Department of Psychology and Neuroscience and Nutrition Research Institute, University of North Carolina at Chapel Hill, Kannapolis, North Carolina, USA,
| |
Collapse
|
36
|
Davies KL, Camm EJ, Atkinson EV, Lopez T, Forhead AJ, Murray AJ, Fowden AL. Development and thyroid hormone dependence of skeletal muscle mitochondrial function towards birth. J Physiol 2020; 598:2453-2468. [PMID: 32087026 PMCID: PMC7317365 DOI: 10.1113/jp279194] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Accepted: 02/05/2020] [Indexed: 12/12/2022] Open
Abstract
Key points Skeletal muscle energy requirements increase at birth but little is known regarding the development of mitochondria that provide most of the cellular energy as ATP. Thyroid hormones are known regulators of adult metabolism and are important in driving several aspects of fetal development, including muscle fibre differentiation. Mitochondrial density and the abundance of mitochondrial membrane proteins in skeletal muscle increased during late gestation. However, mitochondrial functional capacity, measured as oxygen consumption rate, increased primarily after birth. Fetal hypothyroidism resulted in significant reductions in mitochondrial function and density in skeletal muscle before birth. Mitochondrial function matures towards birth and is dependent on the presence of thyroid hormones, with potential implications for the health of pre‐term and hypothyroid infants.
Abstract Birth is a significant metabolic challenge with exposure to a pro‐oxidant environment and the increased energy demands for neonatal survival. This study investigated the development of mitochondrial density and activity in ovine biceps femoris skeletal muscle during the perinatal period and examined the role of thyroid hormones in these processes. Muscle capacity for oxidative phosphorylation increased primarily after birth but was accompanied by prepartum increases in mitochondrial density and the abundance of electron transfer system (ETS) complexes I–IV and ATP‐synthase as well as by neonatal upregulation of uncoupling proteins. This temporal disparity between prepartum maturation and neonatal upregulation of mitochondrial oxidative capacity may protect against oxidative stress associated with birth while ensuring energy availability to the neonate. Fetal thyroid hormone deficiency reduced oxidative phosphorylation and prevented the prepartum upregulation of mitochondrial density and ETS proteins in fetal skeletal muscle. Overall, the data show that mitochondrial function matures over the perinatal period and is dependent on thyroid hormones, with potential consequences for neonatal viability and adult metabolic health. Skeletal muscle energy requirements increase at birth but little is known regarding the development of mitochondria that provide most of the cellular energy as ATP. Thyroid hormones are known regulators of adult metabolism and are important in driving several aspects of fetal development, including muscle fibre differentiation. Mitochondrial density and the abundance of mitochondrial membrane proteins in skeletal muscle increased during late gestation. However, mitochondrial functional capacity, measured as oxygen consumption rate, increased primarily after birth. Fetal hypothyroidism resulted in significant reductions in mitochondrial function and density in skeletal muscle before birth. Mitochondrial function matures towards birth and is dependent on the presence of thyroid hormones, with potential implications for the health of pre‐term and hypothyroid infants.
Collapse
Affiliation(s)
- K L Davies
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK
| | - E J Camm
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK
| | - E V Atkinson
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK
| | - T Lopez
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK
| | - A J Forhead
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK.,Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, OX3 0BP, UK
| | - A J Murray
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK
| | - A L Fowden
- Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK
| |
Collapse
|
37
|
Guo Z, Luo Y, Zhang P, Chetwynd AJ, Qunhui Xie H, Abdolahpur Monikh F, Tao W, Xie C, Liu Y, Xu L, Zhang Z, Valsami-Jones E, Lynch I, Zhao B. Deciphering the particle specific effects on metabolism in rat liver and plasma from ZnO nanoparticles versus ionic Zn exposure. ENVIRONMENT INTERNATIONAL 2020; 136:105437. [PMID: 31881423 DOI: 10.1016/j.envint.2019.105437] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/10/2019] [Accepted: 12/20/2019] [Indexed: 06/10/2023]
Abstract
Toxicity of ZnO nanoparticles (NPs) are often related to the release of Zn2+ ions due to their dissolution. Studies also suggest that the toxicity of ZnO NPs cannot be solely explained by the release of Zn2+ ions; however, there is a lack of direct evidence of ZnO particulate effects. This study compared the acute toxicity of ZnO NPs and ZnSO4 following intranasal exposure using a combination of metallomics and metabolomics approaches. Significant accumulation of Zn in the liver was only found in the ZnO NP treatment, with 29% of the newly accumulated Zn in the form of ZnO as revealed by X-ray fine structure spectroscopy (XAFS). This is the first direct evidence suggesting the persistence of ZnO NPs in liver upon intranasal exposure. Although both ZnO NPs and ZnSO4 altered the metabolite profiles, with some overlaps and considerable specificity, of both liver and plasma samples, more and distinct metabolites in the liver and opposite effects in the plasma were altered by ZnO NPs compared with ZnSO4, consistent with no accumulation of Zn detected in liver from ZnSO4. Specifically, a large number of antioxidant-related compounds and energetic substrates were exclusively elevated in the liver of ZnO NP-treated animals. These findings provided direct evidence that persistence of ZnO NPs induced particle-specific effects on the antioxidant systems and energy metabolism pathways.
Collapse
Affiliation(s)
- Zhiling Guo
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Yali Luo
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Peng Zhang
- School of Geography, Earth and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, UK.
| | - Andrew J Chetwynd
- School of Geography, Earth and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Heidi Qunhui Xie
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute of Environment and Health, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Fazel Abdolahpur Monikh
- Institute of Environmental Sciences (CML), Leiden University, Leiden 2300 RA, the Netherlands
| | - Wunqun Tao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Changjian Xie
- Institute of High Energy Physics Chinese Academy of Sciences, Beijing 100049, China
| | - Yiyun Liu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Li Xu
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute of Environment and Health, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| | - Zhiyong Zhang
- Institute of High Energy Physics Chinese Academy of Sciences, Beijing 100049, China
| | - Eugenia Valsami-Jones
- School of Geography, Earth and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Iseult Lynch
- School of Geography, Earth and Environmental Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Bin Zhao
- State Key Laboratory of Environmental Chemistry and Ecotoxicology, Research Center for Eco-Environmental Sciences, Chinese Academy of Sciences, Beijing 100085, China; University of Chinese Academy of Sciences, Beijing 100049, China; Institute of Environment and Health, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China.
| |
Collapse
|
38
|
Chattrapiban T, Smit HA, Wijga AH, Brunekreef B, Vonk JM, Gehring U, van Rossem L. The joint effect of maternal smoking during pregnancy and maternal pre-pregnancy overweight on infants' term birth weight. BMC Pregnancy Childbirth 2020; 20:132. [PMID: 32106826 PMCID: PMC7047372 DOI: 10.1186/s12884-020-2816-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 02/17/2020] [Indexed: 11/20/2022] Open
Abstract
Background It is well known that maternal smoking during pregnancy and maternal pre-pregnancy overweight have opposite effects on the infants’ birth weight. We report on the association of the combination between both risk factors and the infants’ birth weight. Methods We studied 3241 infants born at term in the PIAMA birth cohort. Maternal smoking during pregnancy and pre-pregnancy height and weight were self-reported. Multivariable regression analysis was performed to assess the associations between infants of mothers who only smoked during pregnancy, who only had pre-pregnancy overweight and who had both risk factors simultaneously, on term birth weight and the risk of being SGA or LGA. Results Of 3241 infants, 421 infants (13%) were born to smoking, non-overweight mothers, 514 (15.8%) to non-smoking, overweight mothers, 129 (4%) to smoking and overweight mothers and 2177 (67%) to non-smoking, non-overweight mothers (reference group). Infants of mothers who smoked and also had pre-pregnancy overweight had similar term birth weight (− 26.6 g, 95%CI: − 113.0, 59.8), SGA risk (OR = 1.06, 95%CI: 0.56, 2.04), and LGA risk (OR = 1.09, 95%CI: 0.61, 1.96) as the reference group. Conclusions Our findings suggested that the effects of maternal smoking during pregnancy and maternal pre-pregnancy overweight on infants’ birth weight cancel each other out. Therefore, birth weight may not be a good indicator of an infant’s health status in perinatal practice because it may mask potential health risks due to these maternal risk factors when both present together.
Collapse
Affiliation(s)
- Thanin Chattrapiban
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, STR 6.118, Utrecht, 3508, GA, The Netherlands
| | - Henriette A Smit
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, STR 6.118, Utrecht, 3508, GA, The Netherlands
| | - Alet H Wijga
- Center for Prevention and Health Services Research, National Institute of Public Health and the Environment, P.O Box 1, 3720, BA, Bilthoven, The Netherlands
| | - Bert Brunekreef
- Institute for Risk Assessment Science (IRAS), Utrecht University, Yalelaan 2, 3584, CM, Utrecht, The Netherlands
| | - Judith M Vonk
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713, GZ, Groningen, The Netherlands
| | - Ulrike Gehring
- Institute for Risk Assessment Science (IRAS), Utrecht University, Yalelaan 2, 3584, CM, Utrecht, The Netherlands
| | - Lenie van Rossem
- Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, STR 6.118, Utrecht, 3508, GA, The Netherlands. .,Department of Obstretrics and Gynecology, Erasmus MC University Medical Center, Dr. Molewaterplein 50, 3015, GD, Rotterdam, the Netherlands.
| |
Collapse
|
39
|
Millership SJ, Van de Pette M, Withers DJ. Genomic imprinting and its effects on postnatal growth and adult metabolism. Cell Mol Life Sci 2019; 76:4009-4021. [PMID: 31270580 PMCID: PMC6785587 DOI: 10.1007/s00018-019-03197-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 06/10/2019] [Accepted: 06/17/2019] [Indexed: 12/25/2022]
Abstract
Imprinted genes display parent-of-origin-specific expression with this epigenetic system of regulation found exclusively in therian mammals. Historically, defined imprinted gene functions were almost solely focused on pregnancy and the influence on the growth parameters of the developing embryo and placenta. More recently, a number of postnatal functions have been identified which converge on resource allocation, both for animals in the nest and in adults. While many of the prenatal functions of imprinted genes that have so far been described adhere to the "parental conflict" hypothesis, no clear picture has yet emerged on the functional role of imprints on postnatal metabolism. As these roles are uncovered, interest in the potential for these genes to influence postnatal metabolism and associated adult-onset disease outcomes when dysregulated has gathered pace. Here, we review the published data on imprinted genes and their influence on postnatal metabolism, starting in the nest, and then progressing through to adulthood. When observing the functional effects of these genes on adult metabolism, we must always be careful to acknowledge the influence both of direct expression in the relevant metabolic tissue, but also indirect metabolic programming effects caused by their modulation of both in utero and postnatal growth trajectories.
Collapse
Affiliation(s)
- Steven J Millership
- MRC London Institute of Medical Sciences, Du Cane Road, London, W12 0NN, UK.
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK.
| | - Mathew Van de Pette
- MRC London Institute of Medical Sciences, Du Cane Road, London, W12 0NN, UK
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Dominic J Withers
- MRC London Institute of Medical Sciences, Du Cane Road, London, W12 0NN, UK.
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
40
|
Numakura C, Tamiya G, Ueki M, Okada T, Maisawa SI, Kojima-Ishii K, Murakami J, Horikawa R, Tokuhara D, Ito K, Adachi M, Abiko T, Mitsui T, Hayasaka K. Growth impairment in individuals with citrin deficiency. J Inherit Metab Dis 2019; 42:501-508. [PMID: 30715743 DOI: 10.1002/jimd.12051] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Accepted: 01/04/2019] [Indexed: 11/06/2022]
Abstract
Citrin deficiency causes neonatal intrahepatic cholestasis (NICCD), failure to thrive and dyslipidemia (FTTDCD), and adult-onset type II citrullinemia (CTLN2). Owing to a defect in the NADH-shuttle, citrin deficiency impairs hepatic glycolysis and de novo lipogenesis leading to hepatic energy deficit. To investigate the physiological role of citrin, we studied the growth of 111 NICCD-affected subjects (51 males and 60 females) and 12 NICCD-unaffected subjects (five males and seven females), including the body weight, height, and genotype. We constructed growth charts using the lambda-mu-sigma (LMS) method. The NICCD-affected subjects showed statistically significant growth impairment, including low birth weight and length, low body weight until 6 to 9 months of age, low height until 11 to 13 years of age, and low body weight in 7 to 12-year-old males and 8-year-old females. NICCD-unaffected subjects showed similar growth impairment, including low birth weight and height, and growth impairment during adolescence. In the third trimester, de novo lipogenesis is required for deposition of body fat and myelination of the developing central nervous system, and its impairment likely causes low birth weight and length. The growth rate is the highest during the first 6 months of life and slows down after 6 months of age, which is probably associated with the onset and recovery of NICCD. Adolescence is the second catch-up growth period, and the proportion and distribution of body fat change depending on age and sex. Characteristic growth impairment in citrin deficiency suggests a significant role of citrin in the catch-up growth via lipogenesis.
Collapse
Affiliation(s)
- Chikahiko Numakura
- Department of Pediatrics, Yamagata University School of Medicine, Yamagata, Japan
| | - Gen Tamiya
- Tohoku Medical Megabank Organization, Tohoku University, Miyagi, Japan
- Statistical Genetics Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
| | - Masao Ueki
- Statistical Genetics Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
| | - Tomoo Okada
- Department of Nutrition and Health Science, Kanagawa Institute of Technology, Kanagawa, Japan
| | - Shun-Ichi Maisawa
- Department of Pediatrics, Morioka Children's Hospital, Morioka, Japan
| | - Kanako Kojima-Ishii
- Department of Pediatrics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Jun Murakami
- Division of Pediatrics and Perinatology, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Reiko Horikawa
- Division of Endocrinology and Metabolism, National Center for Child Health and Development, Tokyo, Japan
| | - Daisuke Tokuhara
- Department of Pediatrics, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Koichi Ito
- Department of Pediatrics and Neonatology, Graduate School of Medical, Sciences, Nagoya City University, Nagoya, Japan
| | - Masanori Adachi
- Department of Endocrinology and Metabolism, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Takahiro Abiko
- Department of Pediatrics, Yamagata University School of Medicine, Yamagata, Japan
| | - Tetsuo Mitsui
- Department of Pediatrics, Yamagata University School of Medicine, Yamagata, Japan
| | - Kiyoshi Hayasaka
- Department of Pediatrics, Yamagata University School of Medicine, Yamagata, Japan
- Department of Pediatrics, Miyukikai Hospital, Kaminoyama, Japan
| |
Collapse
|
41
|
Bildirici I, Schaiff WT, Chen B, Morizane M, Oh SY, O’Brien M, Sonnenberg-Hirche C, Chu T, Barak Y, Nelson DM, Sadovsky Y. PLIN2 Is Essential for Trophoblastic Lipid Droplet Accumulation and Cell Survival During Hypoxia. Endocrinology 2018; 159:3937-3949. [PMID: 30351430 PMCID: PMC6240902 DOI: 10.1210/en.2018-00752] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 10/17/2018] [Indexed: 12/12/2022]
Abstract
Trophoblast hypoxia and injury, key components of placental dysfunction, are associated with fetal growth restriction and other complications of pregnancy. Accumulation of lipid droplets has been found in hypoxic nonplacental cells. Unique to pregnancy, lipid accumulation in the placenta might perturb lipid transport to the fetus. We tested the hypothesis that hypoxia leads to accumulation of lipid droplets in human trophoblasts and that trophoblastic PLIN proteins play a key role in this process. We found that hypoxia promotes the accumulation of lipid droplets in primary human trophoblasts. A similar accretion of lipid droplets was found in placental villi in vivo from pregnancies complicated by fetal growth restriction. In both situations, these changes were associated with an increased level of cellular triglycerides. Exposure of trophoblasts to hypoxia led to reduced fatty acid efflux and oxidation with no change in fatty acid uptake or synthesis. We further found that hypoxia markedly stimulated PLIN2 mRNA synthesis and protein expression, which colocalized to lipid droplets. Knockdown of PLIN2, but not PLIN3, enhanced trophoblast apoptotic death, and overexpression of PLIN2 promoted cell viability. Collectively, our data indicate that hypoxia enhances trophoblastic lipid retention in the form of lipid droplets and that PLIN2 plays a key role in this process and in trophoblast defense against apoptotic death. These findings also imply that this protective mechanism may lead to diminished trafficking of lipids to the developing fetus.
Collapse
Affiliation(s)
- Ibrahim Bildirici
- Department of Obstetrics and Gynecology, Washington University, St. Louis, Missouri
| | - W Timothy Schaiff
- Department of Obstetrics and Gynecology, Washington University, St. Louis, Missouri
| | - Baosheng Chen
- Department of Obstetrics and Gynecology, Washington University, St. Louis, Missouri
| | - Mayumi Morizane
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Soo-Young Oh
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Matthew O’Brien
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | | | - Tianjiao Chu
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Yaacov Barak
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - D Michael Nelson
- Department of Obstetrics and Gynecology, Washington University, St. Louis, Missouri
| | - Yoel Sadovsky
- Magee-Womens Research Institute, Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania
- Correspondence: Yoel Sadovsky, MD, Magee-Womens Research Institute, 204 Craft Avenue, Pittsburgh, Pennsylvania 15213. E-mail:
| |
Collapse
|
42
|
Millership SJ, Tunster SJ, Van de Pette M, Choudhury AI, Irvine EE, Christian M, Fisher AG, John RM, Scott J, Withers DJ. Neuronatin deletion causes postnatal growth restriction and adult obesity in 129S2/Sv mice. Mol Metab 2018; 18:97-106. [PMID: 30279096 PMCID: PMC6308027 DOI: 10.1016/j.molmet.2018.09.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 09/10/2018] [Indexed: 01/31/2023] Open
Abstract
OBJECTIVE Imprinted genes are crucial for the growth and development of fetal and juvenile mammals. Altered imprinted gene dosage causes a variety of human disorders, with growth and development during these crucial early stages strongly linked with future metabolic health in adulthood. Neuronatin (Nnat) is a paternally expressed imprinted gene found in neuroendocrine systems and white adipose tissue and is regulated by the diet and leptin. Neuronatin expression is downregulated in obese children and has been associated with stochastic obesity in C57BL/6 mice. However, our recent studies of Nnat null mice on this genetic background failed to display any body weight or feeding phenotypes but revealed a defect in glucose-stimulated insulin secretion due to the ability of neuronatin to potentiate signal peptidase cleavage of preproinsulin. Nnat deficiency in beta cells therefore caused a lack of appropriate storage and secretion of mature insulin. METHODS To further explore the potential role of Nnat in the regulation of body weight and adiposity, we studied classical imprinting-related phenotypes such as placental, fetal, and postnatal growth trajectory patterns that may impact upon subsequent adult metabolic phenotypes. RESULTS Here we find that, in contrast to the lack of any body weight or feeding phenotypes on the C57BL/6J background, deletion of Nnat in mice on 129S2/Sv background causes a postnatal growth restriction with reduced adipose tissue accumulation, followed by catch up growth after weaning. This was in the absence of any effect on fetal growth or placental development. In adult 129S2/Sv mice, Nnat deletion was associated with hyperphagia, reduced energy expenditure, and partial leptin resistance. Lack of neuronatin also potentiated obesity caused by either aging or high fat diet feeding. CONCLUSIONS The imprinted gene Nnat plays a key role in postnatal growth, adult energy homeostasis, and the pathogenesis of obesity via catch up growth effects, but this role is dependent upon genetic background.
Collapse
Affiliation(s)
- Steven J Millership
- MRC London Institute of Medical Sciences, Du Cane Road, London, W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Simon J Tunster
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AX, UK
| | | | | | - Elaine E Irvine
- MRC London Institute of Medical Sciences, Du Cane Road, London, W12 0NN, UK
| | - Mark Christian
- Institute of Reproductive and Developmental Biology, Department of Surgery and Cancer, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Amanda G Fisher
- MRC London Institute of Medical Sciences, Du Cane Road, London, W12 0NN, UK
| | - Rosalind M John
- School of Biosciences, Cardiff University, Museum Avenue, Cardiff, CF10 3AX, UK
| | - James Scott
- National Heart and Lung Institute, Department of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK
| | - Dominic J Withers
- MRC London Institute of Medical Sciences, Du Cane Road, London, W12 0NN, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London, W12 0NN, UK.
| |
Collapse
|
43
|
Wesolowski SR, Mulligan CM, Janssen RC, Baker PR, Bergman BC, D'Alessandro A, Nemkov T, Maclean KN, Jiang H, Dean TA, Takahashi DL, Kievit P, McCurdy CE, Aagaard KM, Friedman JE. Switching obese mothers to a healthy diet improves fetal hypoxemia, hepatic metabolites, and lipotoxicity in non-human primates. Mol Metab 2018; 18:25-41. [PMID: 30337225 PMCID: PMC6308036 DOI: 10.1016/j.molmet.2018.09.008] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 09/12/2018] [Accepted: 09/24/2018] [Indexed: 12/20/2022] Open
Abstract
Objective Non-alcoholic fatty liver disease (NAFLD) risk begins in utero in offspring of obese mothers. A critical unmet need in this field is to understand the pathways and biomarkers underlying fetal hepatic lipotoxicity and whether maternal dietary intervention during pregnancy is an effective countermeasure. Methods We utilized a well-established non-human primate model of chronic, maternal, Western-style diet induced obesity (OB-WSD) compared with mothers on a healthy control diet (CON) or a subset of OB-WSD mothers switched to the CON diet (diet reversal; OB-DR) prior to and for the duration of the next pregnancy. Fetuses were studied in the early 3rd trimester. Results Fetuses from OB-WSD mothers had higher circulating triglycerides (TGs) and lower arterial oxygenation suggesting hypoxemia, compared with fetuses from CON and OB-DR mothers. Hepatic TG content, oxidative stress (TBARs), and de novo lipogenic genes were increased in fetuses from OB-WSD compared with CON mothers. Fetuses from OB-DR mothers had lower lipogenic gene expression and TBARs yet persistently higher TGs. Metabolomic profiling of fetal liver and serum (umbilical artery) revealed distinct separation of CON and OB-WSD groups, and an intermediate phenotype in fetuses from OB-DR mothers. Pathway analysis identified decreased tricarboxylic acid cycle intermediates, increased amino acid (AA) metabolism and byproducts, and increased gluconeogenesis, suggesting an increased reliance on AA metabolism to meet energy needs in the liver of fetuses from OB-WSD mothers. Components in collagen synthesis, including serum protein 5-hydroxylysine and hepatic lysine and proline, were positively correlated with hepatic TGs and TBARs, suggesting early signs of fibrosis in livers from the OB-WSD group. Importantly, hepatic gluconeogenic and arginine related intermediates and serum levels of lactate, pyruvate, several AAs, and nucleotide intermediates were normalized in the OB-DR group. However, hepatic levels of CDP-choline and total ceramide levels remained high in fetuses from OB-DR mothers. Conclusions Our data provide new metabolic evidence that, in addition to fetal hepatic steatosis, maternal WSD creates fetal hypoxemia and increases utilization of AAs for energy production and early activation of gluconeogenic pathways in the fetal liver. When combined with hyperlipidemia and limited antioxidant activity, the fetus suffers from hepatic oxidative stress and altered intracellular metabolism which can be improved with maternal diet intervention. Our data reinforce the concept that multiple “first hits” occur in the fetus prior to development of obesity and demonstrate new biomarkers with potential clinical implications for monitoring NAFLD risk in offspring. Maternal WSD increases fetal hypoxemia and utilization of AAs for gluconeogenesis. Maternal WSD increases fetal oxidative stress and precursors to liver fibrosis. Carnosine and l-proline uniquely correlated with fetal TG and oxidative stress. Fetal TGs were correlated with fetal arterial oxygen saturation. Diet reversal in obese WSD mothers prevents fetal hypoxemia and oxidative stress.
Collapse
Affiliation(s)
| | | | | | - Peter R Baker
- Department of Pediatrics, Section of Genetics and Metabolism, USA
| | - Bryan C Bergman
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, USA
| | - Angelo D'Alessandro
- Department of Biochemistry & Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Travis Nemkov
- Department of Biochemistry & Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | | | - Hua Jiang
- Department of Pediatrics, Section of Genetics and Metabolism, USA
| | - Tyler A Dean
- Division of Diabetes, Obesity & Metabolism, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Diana L Takahashi
- Division of Diabetes, Obesity & Metabolism, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Paul Kievit
- Division of Diabetes, Obesity & Metabolism, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97006, USA
| | - Carrie E McCurdy
- Department of Human Physiology, University of Oregon, Eugene, OR, 97403, USA
| | - Kjersti M Aagaard
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jacob E Friedman
- Department of Pediatrics, Section of Neonatology, USA; Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, USA; Department of Biochemistry & Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA.
| |
Collapse
|
44
|
Archer E, Pavela G, McDonald S, Lavie CJ, Hill JO. Cell-Specific "Competition for Calories" Drives Asymmetric Nutrient-Energy Partitioning, Obesity, and Metabolic Diseases in Human and Non-human Animals. Front Physiol 2018; 9:1053. [PMID: 30147656 PMCID: PMC6097573 DOI: 10.3389/fphys.2018.01053] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 07/16/2018] [Indexed: 12/20/2022] Open
Abstract
The mammalian body is a complex physiologic “ecosystem” in which cells compete for calories (i.e., nutrient-energy). Axiomatically, cell-types with competitive advantages acquire a greater number of consumed calories, and when possible, increase in size and/or number. Thus, it is logical and parsimonious to posit that obesity is the competitive advantages of fat-cells (adipocytes) driving a disproportionate acquisition and storage of nutrient-energy. Accordingly, we introduce two conceptual frameworks. Asymmetric Nutrient-Energy Partitioning describes the context-dependent, cell-specific competition for calories that determines the partitioning of nutrient-energy to oxidation, anabolism, and/or storage; and Effective Caloric Intake which describes the number of calories available to constrain energy-intake via the inhibition of the sensorimotor appetitive cells in the liver and brain that govern ingestive behaviors. Inherent in these frameworks is the independence and dissociation of the energetic demands of metabolism and the neuro-muscular pathways that initiate ingestive behaviors and energy intake. As we demonstrate, if the sensorimotor cells suffer relative caloric deprivation via asymmetric competition from other cell-types (e.g., skeletal muscle- or fat-cells), energy-intake is increased to compensate for both real and merely apparent deficits in energy-homeostasis (i.e., true and false signals, respectively). Thus, we posit that the chronic positive energy balance (i.e., over-nutrition) that leads to obesity and metabolic diseases is engendered by apparent deficits (i.e., false signals) driven by the asymmetric inter-cellular competition for calories and concomitant differential partitioning of nutrient-energy to storage. These frameworks, in concert with our previous theoretic work, the Maternal Resources Hypothesis, provide a parsimonious and rigorous explanation for the rapid rise in the global prevalence of increased body and fat mass, and associated metabolic dysfunctions in humans and other mammals inclusive of companion, domesticated, laboratory, and feral animals.
Collapse
Affiliation(s)
| | - Gregory Pavela
- The University of Alabama at Birmingham, Birmingham, AL, United States
| | | | - Carl J Lavie
- School of Medicine, John Ochsner Heart and Vascular Institute, The University of Queensland, New Orleans, LA, United States
| | - James O Hill
- Center for Human Nutrition, University of Colorado Health Sciences Center, Denver, CO, United States
| |
Collapse
|
45
|
Balogh O, Bruckmaier R, Keller S, Reichler IM. Effect of maternal metabolism on fetal supply: Glucose, non-esterified fatty acids and beta-hydroxybutyrate concentrations in canine maternal serum and fetal fluids at term pregnancy. Anim Reprod Sci 2018; 193:209-216. [PMID: 29716779 DOI: 10.1016/j.anireprosci.2018.04.072] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 04/11/2018] [Accepted: 04/13/2018] [Indexed: 01/19/2023]
Abstract
The progressive adaptations in carbohydrate and lipid metabolism during canine pregnancy are reflected in the concentrations of glucose, non-esterified fatty acids (NEFA) and β-hydroxybutyrate (BHB). The levels of these metabolites in the bitch likely affect fetal concentrations and the composition of amniotic and allantoic fluids (AMF and ALF, respectively). We studied 31 canine parturitions (Cesarean sections) and found that glucose, NEFA and BHB concentrations were significantly higher in maternal serum than in AMF or ALF. Glucose levels in maternal serum, AMF and ALF were closely related (R2 ≥ 0.821, P < 0.0001) as well as serum and AMF BHB levels (R2 = 0.661, P < 0.0001). In maternal serum, increases in NEFA were associated with increased BHB, and both were negatively related to glucose (P ≤ 0.010). To estimate the effect of the metabolic burden of pregnancy, we evaluated these variables in relation to the dam's body weight and to the ratio of litter weight to the dam's body weight (LW/BW). Maternal serum glucose was not influenced by LW/BW, but it was lower in small than in large/giant bitches. Small breed dogs and those with >10% LW/BW had significantly higher serum NEFA and BHB concentrations. Glucose in AMF and ALF was independent of LW/BW (P ≥ 0.399). AMF NEFA was lower and BHB higher, if LW/BW was >10% (P ≤ 0.048). In conclusion, the extent of the metabolic load of pregnancy in bitches depends on breed size and on the ratio of litter weight to dam's body weight. Maternal concentrations of glucose, BHB and NEFA determine the concentrations of these metabolites in fetal fluids.
Collapse
Affiliation(s)
- Orsolya Balogh
- Clinic of Reproductive Medicine, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 260, 8057, Zurich, Switzerland.
| | - Rupert Bruckmaier
- Veterinary Physiology, Vetsuisse Faculty, University of Bern, Bremgartenstrasse 109a, 3001, Bern, Switzerland
| | - Stefanie Keller
- Clinic of Reproductive Medicine, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 260, 8057, Zurich, Switzerland
| | - Iris Margaret Reichler
- Clinic of Reproductive Medicine, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 260, 8057, Zurich, Switzerland
| |
Collapse
|
46
|
Peoples JNR, Maxmillian T, Le Q, Nadtochiy SM, Brookes PS, Porter GA, Davidson VL, Ebert SN. Metabolomics reveals critical adrenergic regulatory checkpoints in glycolysis and pentose-phosphate pathways in embryonic heart. J Biol Chem 2018. [PMID: 29540484 DOI: 10.1074/jbc.ra118.002566] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Cardiac energy demands during early embryonic periods are sufficiently met through glycolysis, but as development proceeds, the oxidative phosphorylation in mitochondria becomes increasingly vital. Adrenergic hormones are known to stimulate metabolism in adult mammals and are essential for embryonic development, but relatively little is known about their effects on metabolism in the embryonic heart. Here, we show that embryos lacking adrenergic stimulation have ∼10-fold less cardiac ATP compared with littermate controls. Despite this deficit in steady-state ATP, neither the rates of ATP formation nor degradation was affected in adrenergic hormone-deficient hearts, suggesting that ATP synthesis and hydrolysis mechanisms were fully operational. We thus hypothesized that adrenergic hormones stimulate metabolism of glucose to provide chemical substrates for oxidation in mitochondria. To test this hypothesis, we employed a metabolomics-based approach using LC/MS. Our results showed glucose 1-phosphate and glucose 6-phosphate concentrations were not significantly altered, but several downstream metabolites in both glycolytic and pentose-phosphate pathways were significantly lower compared with controls. Furthermore, we identified glyceraldehyde-3-phosphate dehydrogenase and glucose-6-phosphate dehydrogenase as key enzymes in those respective metabolic pathways whose activity was significantly (p < 0.05) and substantially (80 and 40%, respectively) lower in adrenergic hormone-deficient hearts. Addition of pyruvate and to a lesser extent ribose led to significant recovery of steady-state ATP concentrations. These results demonstrate that without adrenergic stimulation, glucose metabolism in the embryonic heart is severely impaired in multiple pathways, ultimately leading to insufficient metabolic substrate availability for successful transition to aerobic respiration needed for survival.
Collapse
Affiliation(s)
- Jessica N R Peoples
- From the Burnett School of Biomedical Sciences, Division of Metabolic and Cardiovascular Sciences, University of Central Florida, College of Medicine, Orlando, Florida 32827
| | - Timmi Maxmillian
- From the Burnett School of Biomedical Sciences, Division of Metabolic and Cardiovascular Sciences, University of Central Florida, College of Medicine, Orlando, Florida 32827
| | - Quynh Le
- From the Burnett School of Biomedical Sciences, Division of Metabolic and Cardiovascular Sciences, University of Central Florida, College of Medicine, Orlando, Florida 32827
| | - Sergiy M Nadtochiy
- the Department of Anesthesiology, University of Rochester Medical Center, Rochester, New York 14620, and
| | - Paul S Brookes
- From the Burnett School of Biomedical Sciences, Division of Metabolic and Cardiovascular Sciences, University of Central Florida, College of Medicine, Orlando, Florida 32827
| | - George A Porter
- the Department of Pediatrics, Division of Cardiology, University of Rochester Medical Center, Rochester, New York 14642
| | - Victor L Davidson
- From the Burnett School of Biomedical Sciences, Division of Metabolic and Cardiovascular Sciences, University of Central Florida, College of Medicine, Orlando, Florida 32827
| | - Steven N Ebert
- From the Burnett School of Biomedical Sciences, Division of Metabolic and Cardiovascular Sciences, University of Central Florida, College of Medicine, Orlando, Florida 32827,
| |
Collapse
|
47
|
Chavan-Gautam P, Rani A, Freeman DJ. Distribution of Fatty Acids and Lipids During Pregnancy. Adv Clin Chem 2018; 84:209-239. [PMID: 29478515 DOI: 10.1016/bs.acc.2017.12.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Maternal fatty acid and lipid metabolism undergoes changes during pregnancy to facilitate fetal growth and development. Different types of fatty acids have different roles in maintaining a successful pregnancy and they are incorporated into different forms of lipids for the purpose of storage and transport. This chapter aims to provide an understanding of the distribution and metabolism of fatty acids and lipids in the maternal, placental, and fetal compartments. We further describe how this distribution is altered in maternal obesity, preterm birth, and pregnancy complications such as gestational diabetes mellitus, preeclampsia, and intrauterine growth restriction.
Collapse
Affiliation(s)
- Preeti Chavan-Gautam
- Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth Deemed University, Pune, Maharashtra, India.
| | - Alka Rani
- Interactive Research School for Health Affairs (IRSHA), Bharati Vidyapeeth Deemed University, Pune, Maharashtra, India
| | - Dilys J Freeman
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
48
|
Ros P, Díaz F, Freire-Regatillo A, Argente-Arizón P, Barrios V, Argente J, Chowen JA. Resveratrol Intake During Pregnancy and Lactation Modulates the Early Metabolic Effects of Maternal Nutrition Differently in Male and Female Offspring. Endocrinology 2018; 159:810-825. [PMID: 29186387 DOI: 10.1210/en.2017-00610] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 11/20/2017] [Indexed: 12/22/2022]
Abstract
Poor maternal nutrition can have detrimental long-term consequences on energy homeostasis in the offspring. Resveratrol exerts antioxidant and antiobesity actions, but its impact during development remains largely unknown. We hypothesized that resveratrol intake during pregnancy and lactation could improve the effects of poor maternal nutrition on offspring metabolism. Wistar rats received a low-fat diet (LFD; 10.2% kcal from fat) or high-fat diet (HFD; 61.6% kcal from fat), with half of each group receiving resveratrol in their drinking water (50 mg/L) during pregnancy and lactation. Body weight (BW) of dams was measured at treatment onset and weaning [postnatal day (PND) 21] and of pups at birth and PND21, at which time dams and pups were euthanized. Although HFD dams consumed more energy, their BW at the end of lactation was unaffected. Mean litter size was not modified by maternal diet or resveratrol. At birth, male offspring from HFD and resveratrol (HFD + R) dams weighed less than those from LFD and resveratrol (LFD + R) dams. On PND21, pups of both sexes from HFD dams weighed more, had more visceral adipose tissue (VAT) and subcutaneous adipose tissue (SCAT), and had higher serum leptin levels than those from LFD dams. Resveratrol reduced BW, leptin, VAT, and SCAT, with females being more affected, but increased glycemia. Neuropeptide levels were unaffected by resveratrol. In conclusion, resveratrol intake during pregnancy and lactation decreased BW and adipose tissue content in offspring of dams on an HFD but did not affect offspring from LFD-fed dams, suggesting that the potential protective effects of resveratrol during gestation/lactation are diet dependent.
Collapse
Affiliation(s)
- Purificación Ros
- Hospital Universitario Puerto de Hierro-Majadahonda, Madrid, Spain
- Department of Pediatrics, Universidad Autónoma of Madrid, Madrid, Spain
| | - Francisca Díaz
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Alejandra Freire-Regatillo
- Department of Pediatrics, Universidad Autónoma of Madrid, Madrid, Spain
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Pilar Argente-Arizón
- Department of Pediatrics, Universidad Autónoma of Madrid, Madrid, Spain
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Vicente Barrios
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| | - Jesús Argente
- Department of Pediatrics, Universidad Autónoma of Madrid, Madrid, Spain
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
- Instituto Madrileño de Estudios Avanzados Food Institute, Campus of International Excellence, Universidad Autónoma of Madrid + Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red de la Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
49
|
Cerychova R, Pavlinkova G. HIF-1, Metabolism, and Diabetes in the Embryonic and Adult Heart. Front Endocrinol (Lausanne) 2018; 9:460. [PMID: 30158902 PMCID: PMC6104135 DOI: 10.3389/fendo.2018.00460] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 07/26/2018] [Indexed: 12/12/2022] Open
Abstract
The heart is able to metabolize any substrate, depending on its availability, to satisfy its energy requirements. Under normal physiological conditions, about 95% of ATP is produced by oxidative phosphorylation and the rest by glycolysis. Cardiac metabolism undergoes reprograming in response to a variety of physiological and pathophysiological conditions. Hypoxia-inducible factor 1 (HIF-1) mediates the metabolic adaptation to hypoxia and ischemia, including the transition from oxidative to glycolytic metabolism. During embryonic development, HIF-1 protects the embryo from intrauterine hypoxia, its deletion as well as its forced expression are embryonically lethal. A decrease in HIF-1 activity is crucial during perinatal remodeling when the heart switches from anaerobic to aerobic metabolism. In the adult heart, HIF-1 protects against hypoxia, although its deletion in cardiomyocytes affects heart function even under normoxic conditions. Diabetes impairs HIF-1 activation and thus, compromises HIF-1 mediated responses under oxygen-limited conditions. Compromised HIF-1 signaling may contribute to the teratogenicity of maternal diabetes and diabetic cardiomyopathy in adults. In this review, we discuss the function of HIF-1 in the heart throughout development into adulthood, as well as the deregulation of HIF-1 signaling in diabetes and its effects on the embryonic and adult heart.
Collapse
Affiliation(s)
- Radka Cerychova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology of the Czech Academy of Sciences, Prague, Czechia
- Faculty of Science, Charles University, Prague, Czechia
| | - Gabriela Pavlinkova
- Laboratory of Molecular Pathogenetics, Institute of Biotechnology of the Czech Academy of Sciences, Prague, Czechia
- *Correspondence: Gabriela Pavlinkova
| |
Collapse
|
50
|
White V, Jawerbaum A, Mazzucco MB, Gauster M, Desoye G, Hiden U. IGF2 stimulates fetal growth in a sex- and organ-dependent manner. Pediatr Res 2018; 83:183-189. [PMID: 28910276 DOI: 10.1038/pr.2017.221] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 08/31/2017] [Indexed: 11/09/2022]
Abstract
BackgroundInsulin-like growth factor 2 (IGF2) is a key determinant of fetal growth, and the altered expression of IGF2 is implicated in fetal growth disorders and maternal metabolic derangements including gestational diabetes. Here we studied how increased levels of IGF2 in late pregnancy affect fetal growth.MethodsWe employed a rat model of repeated intrafetal IGF2 administration in late pregnancy, i.e., during GD19-GD21, and measured the consequences on fetal organ weight and expression of insulin/IGF-axis components.ResultsIGF2 treatment tended to increase fetal weight, but only weight increase of the fetal stomach reached significance (+33±9%; P<0.01). Sex-dependent data analysis revealed a sexual dimorphism of IGF2 action. In male fetuses, IGF2 administration significantly increased fetal weight (+13±3%; P<0.05) and weight of fetal stomach (+42±10%; P<0.01), intestine (+26±5%; P<0.05), liver (+13±4%; P<0.05), and pancreas (+25±8%; P<0.05). Weights of heart, lungs, and kidneys were unchanged. In female fetuses, IGF2 increased only stomach weight (+26±9%; P<0.05). Furthermore, gene expression of insulin/IGF axis in the heart, lungs, liver, and stomach was more sensitive toward IGF2 treatment in male than in female fetuses.ConclusionData suggest that elevated circulating IGF2 in late pregnancy predominantly stimulates organ growth of the digestive system, and male fetuses are more susceptible toward the IGF2 effects than female fetuses.
Collapse
Affiliation(s)
- Veronica White
- Laboratory of Reproduction and Metabolism, Center for Pharmacological and Botanical Studies, CEFyBO-CONICET, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Alicia Jawerbaum
- Laboratory of Reproduction and Metabolism, Center for Pharmacological and Botanical Studies, CEFyBO-CONICET, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Maria Belen Mazzucco
- Laboratory of Reproduction and Metabolism, Center for Pharmacological and Botanical Studies, CEFyBO-CONICET, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Martin Gauster
- Institute of Cell Biology, Histology and Embryology, Medical University of Graz, Graz, Austria
| | - Gernot Desoye
- Department of Obstetrics and Gynaecology, Medical University of Graz, Graz, Austria
| | - Ursula Hiden
- Department of Obstetrics and Gynaecology, Medical University of Graz, Graz, Austria
| |
Collapse
|