1
|
Zhang H, Zhou W, Gao P, Li Z, Li C, Li J, Bian J, Gong L, He C, Han L, Wang M. Ellagic Acid Protects against Alcohol-Related Liver Disease by Modulating the Hepatic Circadian Rhythm Signaling through the Gut Microbiota-NPAS2 Axis. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:25103-25117. [PMID: 39495286 DOI: 10.1021/acs.jafc.4c06992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2024]
Abstract
Alcohol-related liver disease (ALD) encompasses a spectrum of hepatic disorders resulting from alcohol abuse, which constitutes the predominant etiology of morbidity and mortality associated with hepatic pathologies globally. Excessive alcohol consumption disrupts the integrity of the intestinal barrier and perturbs the balance of gut microbiota, thereby facilitating the progression of ALD. Ellagic acid (EA) has been extensively reported to be an effective intervention for alleviating liver symptoms. However, the target molecules of EA in improving ALD and its underlying mechanism remain elusive. First, our study indicates that EA ameliorated ALD through the hepatic circadian rhythm signaling by up-regulating neuronal PAS domain protein 2 (NPAS2). Furthermore, analysis of the intestinal microbiome showed that EA significantly enhanced the abundance of beneficial bacteria, which was positively correlated with NPAS2 expression and negatively correlated with liver injury. Finally, antibiotic treatment and fecal microbiota transplantation (FMT) experiments established a causal relationship between the reshaped microbiota and NPAS2 in the amelioration of ALD. In summary, our study demonstrates novel evidence that EA attenuated ALD by modulating the hepatic circadian rhythm signaling pathway via the gut microbiota-NPAS2 axis, providing valuable insights for EA and microbiome-targeted interventions against ALD.
Collapse
Affiliation(s)
- Hongbo Zhang
- Department of Nutrition and Health, College of Food Science and Engineering, Northwest A&F University, Yang ling, Shaanxi 712100, China
| | - Wenxu Zhou
- Department of Nutrition and Health, College of Food Science and Engineering, Northwest A&F University, Yang ling, Shaanxi 712100, China
| | - Pan Gao
- Department of Nutrition and Health, College of Food Science and Engineering, Northwest A&F University, Yang ling, Shaanxi 712100, China
| | - Zibin Li
- Department of Nutrition and Health, College of Food Science and Engineering, Northwest A&F University, Yang ling, Shaanxi 712100, China
| | - Chaoyue Li
- Department of Nutrition and Health, College of Food Science and Engineering, Northwest A&F University, Yang ling, Shaanxi 712100, China
| | - Jie Li
- College of Animal Science and Technology, Northwest A&F University, Yang ling, Shaanxi 712100, China
| | - Ji Bian
- Kolling Institute, Sydney Medical School, Royal North Shore Hospital, University of Sydney, St. Leonards, Sydney, NSW 2065, Australia
| | - Lan Gong
- UNSW Microbiome Research Centre, St George and Sutherland Clinical Campus, University of New South Wales, Sydney, NSW 2052, Australia
| | - Caian He
- Department of Nutrition and Health, College of Food Science and Engineering, Northwest A&F University, Yang ling, Shaanxi 712100, China
| | - Lin Han
- Department of Nutrition and Health, College of Food Science and Engineering, Northwest A&F University, Yang ling, Shaanxi 712100, China
| | - Min Wang
- Department of Nutrition and Health, College of Food Science and Engineering, Northwest A&F University, Yang ling, Shaanxi 712100, China
| |
Collapse
|
2
|
Bolte E, Dean T, Garcia B, Seferovic MD, Sauter K, Hummel G, Bucher M, Li F, Hicks J, Qin X, Suter MA, Barrozo ER, Jochum M, Shope C, Friedman JE, Gannon M, Wesolowski SR, McCurdy CE, Kievit P, Aagaard KM. Initiation of metformin in early pregnancy results in fetal bioaccumulation, growth restriction, and renal dysmorphology in a primate model. Am J Obstet Gynecol 2024; 231:352.e1-352.e16. [PMID: 38871238 PMCID: PMC11344684 DOI: 10.1016/j.ajog.2024.06.002] [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: 01/17/2024] [Revised: 05/27/2024] [Accepted: 06/04/2024] [Indexed: 06/15/2024]
Abstract
BACKGROUND In recent years, pragmatic metformin use in pregnancy has stretched to include prediabetes mellitus, type 2 diabetes mellitus, gestational diabetes mellitus, and (most recently) preeclampsia. However, with its expanded use, concerns of unintended harm have been raised. OBJECTIVE This study developed an experimental primate model and applied ultrahigh performance liquid chromatography coupled to triple-quadrupole mass spectrometry for direct quantitation of maternal and fetal tissue metformin levels with detailed fetal biometry and histopathology. STUDY DESIGN Within 30 days of confirmed conception (defined as early pregnancy), 13 time-bred (timed-mated breeding) Rhesus dams with pregnancies designated for fetal necropsy were initiated on twice-daily human dose-equivalent 10 mg/kg metformin or vehicle control. Pregnant dams were maintained as pairs and fed either a control chow or 36% fat Western-style diet. Metformin or placebo vehicle control was delivered in various treats while the animals were separated via a slide. A cesarean delivery was performed at gestational day 145, and amniotic fluid and blood were collected, and the fetus and placenta were delivered. The fetus was immediately necropsied by trained primate center personnel. All fetal organs were dissected, measured, sectioned, and processed per clinical standards. Fluid and tissue metformin levels were assayed using validated ultrahigh performance liquid chromatography coupled to triple-quadrupole mass spectrometry in selected reaction monitoring against standard curves. RESULTS Among 13 pregnancies at gestational day 145 with fetal necropsy, 1 dam and its fetal tissues had detectable metformin levels despite being allocated to the vehicle control group (>1 μmol metformin/kg maternal weight or fetal or placental tissue), whereas a second fetus allocated to the vehicle control group had severe fetal growth restriction (birthweight of 248.32 g [<1%]) and was suspected of having a fetal congenital condition. After excluding these 2 fetal pregnancies from further analyses, 11 fetuses from dams initiated on either vehicle control (n=4: 3 female and 1 male fetuses) or 10 mg/kg metformin (n=7: 5 female and 2 male fetuses) were available for analyses. Among dams initiated on metformin at gestational day 30 (regardless of maternal diet), significant bioaccumulation within the fetal kidney (0.78-6.06 μmol/kg; mean of 2.48 μmol/kg), liver (0.16-0.73 μmol/kg; mean of 0.38 μmol/kg), fetal gut (0.28-1.22 μmol/kg; mean of 0.70 μmol/kg), amniotic fluid (0.43-3.33 μmol/L; mean of 1.88 μmol/L), placenta (0.16-1.00 μmol/kg; mean of 0.50 μmol/kg), fetal serum (0.00-0.66 μmol/L; mean of 0.23 μmol/L), and fetal urine (4.10-174.10 μmol/L; mean of 38.5 μmol/L) was observed, with fetal levels near biomolar equivalent to maternal levels (maternal serum: 0.18-0.86 μmol/L [mean of 0.46 μmol/L]; maternal urine: 42.60-254.00 μmol/L [mean of 149.30 μmol/L]). Western-style diet feeding neither accelerated nor reduced metformin bioaccumulations in maternal or fetal serum, urine, amniotic fluid, placenta, or fetal tissues. In these 11 animals, fetal bioaccumulation of metformin was associated with less fetal skeletal muscle (57% lower cross-sectional area of gastrocnemius) and decreased liver, heart, and retroperitoneal fat masses (P<.05), collectively driving lower delivery weight (P<.0001) without changing the crown-rump length. Sagittal sections of fetal kidneys demonstrated delayed maturation, with disorganized glomerular generations and increased cortical thickness. This renal dysmorphology was not accompanied by structural or functional changes indicative of renal insufficiency. CONCLUSION Our study demonstrates fetal bioaccumulation of metformin with associated fetal growth restriction and renal dysmorphology after maternal initiation of the drug within 30 days of conception in primates. Given these results and the prevalence of metformin use during pregnancy, additional investigation of any potential immediate and enduring effects of prenatal metformin use is warranted.
Collapse
Affiliation(s)
- Erin Bolte
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Tyler Dean
- Oregon National Primate Research Center, Beaverton, OR
| | - Brandon Garcia
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Maxim D Seferovic
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | | | - Gwendolynn Hummel
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX
| | - Matthew Bucher
- Department of Human Physiology, University of Oregon, Eugene OR
| | - Feng Li
- Department of Pathology and Immunology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - John Hicks
- Department of Pathology and Immunology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Xuan Qin
- Department of Pathology and Immunology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Melissa A Suter
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Enrico R Barrozo
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Michael Jochum
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Cynthia Shope
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Jacob E Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | - Maureen Gannon
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, TN
| | | | | | - Paul Kievit
- Oregon National Primate Research Center, Beaverton, OR
| | - Kjersti M Aagaard
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX; Oregon National Primate Research Center, Beaverton, OR.
| |
Collapse
|
3
|
Ding L, Weger BD, Liu J, Zhou L, Lim Y, Wang D, Xie Z, Liu J, Ren J, Zheng J, Zhang Q, Yu M, Weger M, Morrison M, Xiao X, Gachon F. Maternal high fat diet induces circadian clock-independent endocrine alterations impacting the metabolism of the offspring. iScience 2024; 27:110343. [PMID: 39045103 PMCID: PMC11263959 DOI: 10.1016/j.isci.2024.110343] [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: 11/24/2023] [Revised: 05/02/2024] [Accepted: 06/19/2024] [Indexed: 07/25/2024] Open
Abstract
Maternal obesity has long-term effects on offspring metabolic health. Among the potential mechanisms, prior research has indicated potential disruptions in circadian rhythms and gut microbiota in the offspring. To challenge this hypothesis, we implemented a maternal high fat diet regimen before and during pregnancy, followed by a standard diet after birth. Our findings confirm that maternal obesity impacts offspring birth weight and glucose and lipid metabolisms. However, we found minimal impact on circadian rhythms and microbiota that are predominantly driven by the feeding/fasting cycle. Notably, maternal obesity altered rhythmic liver gene expression, affecting mitochondrial function and inflammatory response without disrupting the hepatic circadian clock. These changes could be explained by a masculinization of liver gene expression similar to the changes observed in polycystic ovarian syndrome. Intriguingly, such alterations seem to provide the first-generation offspring with a degree of protection against obesity when exposed to a high fat diet.
Collapse
Affiliation(s)
- Lu Ding
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Benjamin D. Weger
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Jieying Liu
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
- Department of Medical Research Center, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China
| | - Liyuan Zhou
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100022, China
| | - Yenkai Lim
- Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Dongmei Wang
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Ziyan Xie
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Jing Liu
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Jing Ren
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Jia Zheng
- Department of Endocrinology, Peking University First Hospital, Beijing 100034, China
| | - Qian Zhang
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Miao Yu
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Meltem Weger
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Mark Morrison
- Frazer Institute, Faculty of Medicine, The University of Queensland, Woolloongabba, QLD 4102, Australia
- Australian Infectious Diseases Research Centre, St. Lucia, QLD 4072, Australia
| | - Xinhua Xiao
- Key Laboratory of Endocrinology of National Health Commission, Diabetes Research Center of Chinese Academy of Medical Sciences, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Frédéric Gachon
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, QLD 4072, Australia
| |
Collapse
|
4
|
Nelson BN, Friedman JE. Developmental Programming of the Fetal Immune System by Maternal Western-Style Diet: Mechanisms and Implications for Disease Pathways in the Offspring. Int J Mol Sci 2024; 25:5951. [PMID: 38892139 PMCID: PMC11172957 DOI: 10.3390/ijms25115951] [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: 04/18/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Maternal obesity and over/undernutrition can have a long-lasting impact on offspring health during critical periods in the first 1000 days of life. Children born to mothers with obesity have reduced immune responses to stimuli which increase susceptibility to infections. Recently, maternal western-style diets (WSDs), high in fat and simple sugars, have been associated with skewing neonatal immune cell development, and recent evidence suggests that dysregulation of innate immunity in early life has long-term consequences on metabolic diseases and behavioral disorders in later life. Several factors contribute to abnormal innate immune tolerance or trained immunity, including changes in gut microbiota, metabolites, and epigenetic modifications. Critical knowledge gaps remain regarding the mechanisms whereby these factors impact fetal and postnatal immune cell development, especially in precursor stem cells in bone marrow and fetal liver. Components of the maternal microbiota that are transferred from mothers consuming a WSD to their offspring are understudied and identifying cause and effect on neonatal innate and adaptive immune development needs to be refined. Tools including single-cell RNA-sequencing, epigenetic analysis, and spatial location of specific immune cells in liver and bone marrow are critical for understanding immune system programming. Considering the vital role immune function plays in offspring health, it will be important to understand how maternal diets can control developmental programming of innate and adaptive immunity.
Collapse
Affiliation(s)
- Benjamin N. Nelson
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Jacob E. Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Department of Physiology and Biochemistry, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Pediatrics, Section of Diabetes and Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
5
|
Carroll DT, Elsakr JM, Miller A, Fuhr J, Lindsley SR, Kirigiti M, Takahashi DL, Dean TA, Wesolowski SR, McCurdy CE, Friedman JE, Aagaard KM, Kievit P, Gannon M. Maternal Western-style diet in nonhuman primates leads to offspring islet adaptations including altered gene expression and insulin hypersecretion. Am J Physiol Endocrinol Metab 2023; 324:E577-E588. [PMID: 37134140 PMCID: PMC10259856 DOI: 10.1152/ajpendo.00087.2023] [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: 03/22/2023] [Revised: 04/26/2023] [Accepted: 04/26/2023] [Indexed: 05/05/2023]
Abstract
Maternal overnutrition is associated with increased susceptibility to type 2 diabetes in the offspring. Rodent models have shown that maternal overnutrition influences islet function in offspring. To determine whether maternal Western-style diet (WSD) alters prejuvenile islet function in a model that approximates that of human offspring, we utilized a well-characterized Japanese macaque model. We compared islet function from offspring exposed to WSD throughout pregnancy and lactation and weaned to WSD (WSD/WSD) compared with islets from offspring exposed only to postweaning WSD (CD/WSD) at 1 yr of age. WSD/WSD offspring islets showed increased basal insulin secretion and an exaggerated increase in glucose-stimulated insulin secretion, as assessed by dynamic ex vivo perifusion assays, relative to CD/WSD-exposed offspring. We probed potential mechanisms underlying insulin hypersecretion using transmission electron microscopy to evaluate β-cell ultrastructure, qRT-PCR to quantify candidate gene expression, and Seahorse assay to assess mitochondrial function. Insulin granule density, mitochondrial density, and mitochondrial DNA ratio were similar between groups. However, islets from WSD/WSD male and female offspring had increased expression of transcripts known to facilitate stimulus-secretion coupling and changes in the expression of cell stress genes. Seahorse assay revealed increased spare respiratory capacity in islets from WSD/WSD male offspring. Overall, these results show that maternal WSD feeding confers changes to genes governing insulin secretory coupling and results in insulin hypersecretion as early as the postweaning period. The results suggest a maternal diet leads to early adaptation and developmental programming in offspring islet genes that may underlie future β-cell dysfunction.NEW & NOTEWORTHY Programed adaptations in islets in response to maternal WSD exposure may alter β-cell response to metabolic stress in offspring. We show that islets from maternal WSD-exposed offspring hypersecrete insulin, possibly due to increased components of stimulus-secretion coupling. These findings suggest that islet hyperfunction is programed by maternal diet, and changes can be detected as early as the postweaning period in nonhuman primate offspring.
Collapse
Affiliation(s)
- Darian T Carroll
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Joseph M Elsakr
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Allie Miller
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Jennifer Fuhr
- Department of Veterans Affairs Tennessee Valley, Nashville, Tennessee, United States
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Sarah Rene Lindsley
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, Oregon, United States
| | - Melissa Kirigiti
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, Oregon, United States
| | - Diana L Takahashi
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, Oregon, United States
| | - Tyler A Dean
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, Oregon, United States
| | - Stephanie R Wesolowski
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado, United States
| | - Carrie E McCurdy
- Department of Human Physiology, University of Oregon, Eugene, Oregon, United States
| | - Jacob E Friedman
- Harold Hamm Diabetes Center, University of Oklahoma, Oklahoma City, Oklahoma, United States
| | - Kjersti M Aagaard
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine, Houston, Texas, United States
| | - Paul Kievit
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, Oregon, United States
| | - Maureen Gannon
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
- Department of Veterans Affairs Tennessee Valley, Nashville, Tennessee, United States
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, Tennessee, United States
| |
Collapse
|
6
|
Nash MJ, Dobrinskikh E, Soderborg TK, Janssen RC, Takahashi DL, Dean TA, Varlamov O, Hennebold JD, Gannon M, Aagaard KM, McCurdy CE, Kievit P, Bergman BC, Jones KL, Pietras EM, Wesolowski SR, Friedman JE. Maternal diet alters long-term innate immune cell memory in fetal and juvenile hematopoietic stem and progenitor cells in nonhuman primate offspring. Cell Rep 2023; 42:112393. [PMID: 37058409 PMCID: PMC10570400 DOI: 10.1016/j.celrep.2023.112393] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/30/2023] [Accepted: 03/30/2023] [Indexed: 04/15/2023] Open
Abstract
Maternal overnutrition increases inflammatory and metabolic disease risk in postnatal offspring. This constitutes a major public health concern due to increasing prevalence of these diseases, yet mechanisms remain unclear. Here, using nonhuman primate models, we show that maternal Western-style diet (mWSD) exposure is associated with persistent pro-inflammatory phenotypes at the transcriptional, metabolic, and functional levels in bone marrow-derived macrophages (BMDMs) from 3-year-old juvenile offspring and in hematopoietic stem and progenitor cells (HSPCs) from fetal and juvenile bone marrow and fetal liver. mWSD exposure is also associated with increased oleic acid in fetal and juvenile bone marrow and fetal liver. Assay for transposase-accessible chromatin with sequencing (ATAC-seq) profiling of HSPCs and BMDMs from mWSD-exposed juveniles supports a model in which HSPCs transmit pro-inflammatory memory to myeloid cells beginning in utero. These findings show that maternal diet alters long-term immune cell developmental programming in HSPCs with proposed consequences for chronic diseases featuring altered immune/inflammatory activation across the lifespan.
Collapse
Affiliation(s)
- Michael J Nash
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Evgenia Dobrinskikh
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Taylor K Soderborg
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Rachel C Janssen
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Diana L Takahashi
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Tyler A Dean
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Oleg Varlamov
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Jon D Hennebold
- Division of Reproductive and Developmental Sciences, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Maureen Gannon
- Department of Medicine, Division of Diabetes, Endocrinology, and Metabolism, Vanderbilt University Medical Center, Nashville, TN 37235, USA
| | - Kjersti M Aagaard
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Carrie E McCurdy
- Department of Human Physiology, University of Oregon, Eugene, OR 97403, USA
| | - Paul Kievit
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR 97006, USA
| | - Bryan C Bergman
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Kenneth L Jones
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Eric M Pietras
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Stephanie R Wesolowski
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Jacob E Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| |
Collapse
|
7
|
Developmental Programming in Animal Models: Critical Evidence of Current Environmental Negative Changes. Reprod Sci 2023; 30:442-463. [PMID: 35697921 PMCID: PMC9191883 DOI: 10.1007/s43032-022-00999-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 06/02/2022] [Indexed: 11/23/2022]
Abstract
The Developmental Origins of Health and Disease (DOHaD) approach answers questions surrounding the early events suffered by the mother during reproductive stages that can either partially or permanently influence the developmental programming of children, predisposing them to be either healthy or exhibit negative health outcomes in adulthood. Globally, vulnerable populations tend to present high obesity rates, including among school-age children and women of reproductive age. In addition, adults suffer from high rates of diabetes, hypertension, cardiovascular, and other metabolic diseases. The increase in metabolic outcomes has been associated with the combination of maternal womb conditions and adult lifestyle-related factors such as malnutrition and obesity, smoking habits, and alcoholism. However, to date, "new environmental changes" have recently been considered negative factors of development, such as maternal sedentary lifestyle, lack of maternal attachment during lactation, overcrowding, smog, overurbanization, industrialization, noise pollution, and psychosocial stress experienced during the current SARS-CoV-2 pandemic. Therefore, it is important to recognize how all these factors impact offspring development during pregnancy and lactation, a period in which the subject cannot protect itself from these mechanisms. This review aims to introduce the importance of studying DOHaD, discuss classical programming studies, and address the importance of studying new emerging programming mechanisms, known as actual lifestyle factors, during pregnancy and lactation.
Collapse
|
8
|
Galvan-Martinez DH, Bosquez-Mendoza VM, Ruiz-Noa Y, Ibarra-Reynoso LDR, Barbosa-Sabanero G, Lazo-de-la-Vega-Monroy ML. Nutritional, pharmacological, and environmental programming of NAFLD in early life. Am J Physiol Gastrointest Liver Physiol 2023; 324:G99-G114. [PMID: 36472341 DOI: 10.1152/ajpgi.00168.2022] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the main liver disease worldwide, and its prevalence in children and adolescents has been increasing in the past years. It has been demonstrated that parental exposure to different conditions, both preconceptionally and during pregnancy, can lead to fetal programming of several metabolic diseases, including NAFLD. In this article, we review some of the maternal and paternal conditions that may be involved in early-life programing of adult NAFLD. First, we describe the maternal nutritional factors that have been suggested to increase the risk of NAFLD in the offspring, such as an obesogenic diet, overweight/obesity, and altered lipogenesis. Second, we review the association of certain vitamin supplementation and the use of some drugs during pregnancy, for instance, glucocorticoids, with a higher risk of NAFLD. Furthermore, we discuss the evidence showing that maternal-fetal pathologies, including gestational diabetes mellitus (GDM), insulin resistance (IR), and intrauterine growth restriction (IUGR), as well as the exposure to environmental contaminants, and the impact of microbiome changes, are important factors in early-life programming of NAFLD. Finally, we review how paternal preconceptional conditions, such as exercise and diet (particularly obesogenic diets), may impact fetal growth and liver function. Altogether, the presented evidence supports the hypothesis that both in utero exposure and parental conditions may influence fetal outcomes, including the development of NAFLD in early life and adulthood. The study of these conditions is crucial to better understand the diverse mechanisms involved in NAFLD, as well as for defining new preventive strategies for this disease.
Collapse
Affiliation(s)
| | | | - Yeniley Ruiz-Noa
- Health Sciences Division, Medical Sciences Department, University of Guanajuato, Campus Leon, Mexico
| | | | - Gloria Barbosa-Sabanero
- Health Sciences Division, Medical Sciences Department, University of Guanajuato, Campus Leon, Mexico
| | | |
Collapse
|
9
|
Abruzzese GA, Arbocco FCV, Ferrer MJ, Silva AF, Motta AB. Role of Hormones During Gestation and Early Development: Pathways Involved in Developmental Programming. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1428:31-70. [PMID: 37466768 DOI: 10.1007/978-3-031-32554-0_2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2023]
Abstract
Accumulating evidence suggests that an altered maternal milieu and environmental insults during the intrauterine and perinatal periods of life affect the developing organism, leading to detrimental long-term outcomes and often to adult pathologies through programming effects. Hormones, together with growth factors, play critical roles in the regulation of maternal-fetal and maternal-neonate interfaces, and alterations in any of them may lead to programming effects on the developing organism. In this chapter, we will review the role of sex steroids, thyroid hormones, and insulin-like growth factors, as crucial factors involved in physiological processes during pregnancy and lactation, and their role in developmental programming effects during fetal and early neonatal life. Also, we will consider epidemiological evidence and data from animal models of altered maternal hormonal environments and focus on the role of different tissues in the establishment of maternal and fetus/infant interaction. Finally, we will identify unresolved questions and discuss potential future research directions.
Collapse
Affiliation(s)
- Giselle Adriana Abruzzese
- Laboratorio de Fisio-patología ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Fiorella Campo Verde Arbocco
- Laboratorio de Hormonas y Biología del Cáncer, Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), CONICET, Mendoza, Argentina
- Laboratorio de Reproducción y Lactancia, IMBECU, CONICET, Mendoza, Argentina
- Facultad de Ciencias Médicas, Universidad de Mendoza, Mendoza, Argentina
| | - María José Ferrer
- Laboratorio de Fisio-patología ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Aimé Florencia Silva
- Laboratorio de Fisio-patología ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | - Alicia Beatriz Motta
- Laboratorio de Fisio-patología ovárica, Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Medicina, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| |
Collapse
|
10
|
Mo J, Liu X, Huang Y, He R, Zhang Y, Huang H. Developmental origins of adult diseases. MEDICAL REVIEW (BERLIN, GERMANY) 2022; 2:450-470. [PMID: 37724166 PMCID: PMC10388800 DOI: 10.1515/mr-2022-0027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 10/11/2022] [Indexed: 09/20/2023]
Abstract
The occurrence and mechanisms of developmental adult diseases have gradually attracted attention in recent years. Exposure of gametes and embryos to adverse environments, especially during plastic development, can alter the expression of certain tissue-specific genes, leading to increased susceptibility to certain diseases in adulthood, such as diabetes, cardiovascular disease, neuropsychiatric, and reproductive system diseases, etc. The occurrence of chronic disease in adulthood is partly due to genetic factors, and the remaining risk is partly due to environmental-dependent epigenetic information alteration, including DNA methylation, histone modifications, and noncoding RNAs. Changes in this epigenetic information potentially damage our health, which has also been supported by numerous epidemiological and animal studies in recent years. Environmental factors functionally affect embryo development through epimutation, transmitting diseases to offspring and even later generations. This review mainly elaborated on the concept of developmental origins of adult diseases, and revealed the epigenetic mechanisms underlying these events, discussed the theoretical basis for the prevention and treatment of related diseases.
Collapse
Affiliation(s)
- Jiaying Mo
- Department of Obstetrics and Gynecology, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang Province, China
- The Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Xuanqi Liu
- The Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Yutong Huang
- The Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Renke He
- Department of Obstetrics and Gynecology, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang Province, China
- The Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
| | - Yu Zhang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Hefeng Huang
- Department of Obstetrics and Gynecology, The Fourth Affiliated Hospital, International Institutes of Medicine, Zhejiang University School of Medicine, Yiwu, Zhejiang Province, China
- The Key Laboratory of Reproductive Genetics (Zhejiang University), Ministry of Education, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, China
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences (No. 2019RU056), Shanghai, China
| |
Collapse
|
11
|
Hatmal MM, Al-Hatamleh MAI, Olaimat AN, Alshaer W, Hasan H, Albakri KA, Alkhafaji E, Issa NN, Al-Holy MA, Abderrahman SM, Abdallah AM, Mohamud R. Immunomodulatory Properties of Human Breast Milk: MicroRNA Contents and Potential Epigenetic Effects. Biomedicines 2022; 10:1219. [PMID: 35740242 PMCID: PMC9219990 DOI: 10.3390/biomedicines10061219] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 05/15/2022] [Accepted: 05/17/2022] [Indexed: 02/07/2023] Open
Abstract
Infants who are exclusively breastfed in the first six months of age receive adequate nutrients, achieving optimal immune protection and growth. In addition to the known nutritional components of human breast milk (HBM), i.e., water, carbohydrates, fats and proteins, it is also a rich source of microRNAs, which impact epigenetic mechanisms. This comprehensive work presents an up-to-date overview of the immunomodulatory constituents of HBM, highlighting its content of circulating microRNAs. The epigenetic effects of HBM are discussed, especially those regulated by miRNAs. HBM contains more than 1400 microRNAs. The majority of these microRNAs originate from the lactating gland and are based on the remodeling of cells in the gland during breastfeeding. These miRNAs can affect epigenetic patterns by several mechanisms, including DNA methylation, histone modifications and RNA regulation, which could ultimately result in alterations in gene expressions. Therefore, the unique microRNA profile of HBM, including exosomal microRNAs, is implicated in the regulation of the genes responsible for a variety of immunological and physiological functions, such as FTO, INS, IGF1, NRF2, GLUT1 and FOXP3 genes. Hence, studying the HBM miRNA composition is important for improving the nutritional approaches for pregnancy and infant's early life and preventing diseases that could occur in the future. Interestingly, the composition of miRNAs in HBM is affected by multiple factors, including diet, environmental and genetic factors.
Collapse
Affiliation(s)
- Ma’mon M. Hatmal
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Mohammad A. I. Al-Hatamleh
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu 16150, Malaysia;
| | - Amin N. Olaimat
- Department of Clinical Nutrition and Dietetics, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan; (A.N.O.); (M.A.A.-H.)
| | - Walhan Alshaer
- Cell Therapy Center (CTC), The University of Jordan, Amman 11942, Jordan;
| | - Hanan Hasan
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman 11942, Jordan;
| | - Khaled A. Albakri
- Faculty of Medicine, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Enas Alkhafaji
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, The University of Jordan, Amman 11942, Jordan;
| | - Nada N. Issa
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Murad A. Al-Holy
- Department of Clinical Nutrition and Dietetics, Faculty of Applied Medical Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan; (A.N.O.); (M.A.A.-H.)
| | - Salim M. Abderrahman
- Department of Biology and Biotechnology, Faculty of Sciences, The Hashemite University, P.O. Box 330127, Zarqa 13133, Jordan;
| | - Atiyeh M. Abdallah
- Department of Biomedical Sciences, College of Health Sciences, QU Health, Qatar University, Doha 2713, Qatar;
| | - Rohimah Mohamud
- Department of Immunology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kota Bharu 16150, Malaysia;
| |
Collapse
|
12
|
Almeida JI, Tenreiro MF, Martinez-Santamaria L, Guerrero-Aspizua S, Gisbert JP, Alves PM, Serra M, Baptista PM. Hallmarks of the human intestinal microbiome on liver maturation and function. J Hepatol 2022; 76:694-725. [PMID: 34715263 DOI: 10.1016/j.jhep.2021.10.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/05/2021] [Accepted: 10/17/2021] [Indexed: 12/18/2022]
Abstract
As one of the most metabolically complex systems in the body, the liver ensures multi-organ homeostasis and ultimately sustains life. Nevertheless, during early postnatal development, the liver is highly immature and takes about 2 years to acquire and develop almost all of its functions. Different events occurring at the environmental and cellular levels are thought to mediate hepatic maturation and function postnatally. The crosstalk between the liver, the gut and its microbiome has been well appreciated in the context of liver disease, but recent evidence suggests that the latter could also be critical for hepatic function under physiological conditions. The gut-liver crosstalk is thought to be mediated by a rich repertoire of microbial metabolites that can participate in a myriad of biological processes in hepatic sinusoids, from energy metabolism to tissue regeneration. Studies on germ-free animals have revealed the gut microbiome as a critical contributor in early hepatic programming, and this influence extends throughout life, mediating liver function and body homeostasis. In this seminar, we describe the microbial molecules that have a known effect on the liver and discuss how the gut microbiome and the liver evolve throughout life. We also provide insights on current and future strategies to target the gut microbiome in the context of hepatology research.
Collapse
Affiliation(s)
- Joana I Almeida
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain; Instituto de Biologia Experimental e Tecnológica (iBET), Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), Oeiras, Portugal
| | - Miguel F Tenreiro
- Instituto de Biologia Experimental e Tecnológica (iBET), Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), Oeiras, Portugal
| | - Lucía Martinez-Santamaria
- Carlos III University of Madrid. Bioengineering and Aerospace Engineering, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER, ISCIII), Madrid, Spain; Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Madrid, Spain
| | - Sara Guerrero-Aspizua
- Carlos III University of Madrid. Bioengineering and Aerospace Engineering, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER, ISCIII), Madrid, Spain
| | - Javier P Gisbert
- Gastroenterology Department. Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid (UAM), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Paula M Alves
- Instituto de Biologia Experimental e Tecnológica (iBET), Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), Oeiras, Portugal
| | - Margarida Serra
- Instituto de Biologia Experimental e Tecnológica (iBET), Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), Oeiras, Portugal
| | - Pedro M Baptista
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain; Carlos III University of Madrid. Bioengineering and Aerospace Engineering, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain; Fundación ARAID, Zaragoza, Spain.
| |
Collapse
|
13
|
Bolte EE, Moorshead D, Aagaard KM. Maternal and early life exposures and their potential to influence development of the microbiome. Genome Med 2022; 14:4. [PMID: 35016706 PMCID: PMC8751292 DOI: 10.1186/s13073-021-01005-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 11/16/2021] [Indexed: 02/07/2023] Open
Abstract
At the dawn of the twentieth century, the medical care of mothers and children was largely relegated to family members and informally trained birth attendants. As the industrial era progressed, early and key public health observations among women and children linked the persistence of adverse health outcomes to poverty and poor nutrition. In the time hence, numerous studies connecting genetics ("nature") to public health and epidemiologic data on the role of the environment ("nurture") have yielded insights into the importance of early life exposures in relation to the occurrence of common diseases, such as diabetes, allergic and atopic disease, cardiovascular disease, and obesity. As a result of these parallel efforts in science, medicine, and public health, the developing brain, immune system, and metabolic physiology are now recognized as being particularly vulnerable to poor nutrition and stressful environments from the start of pregnancy to 3 years of age. In particular, compelling evidence arising from a diverse array of studies across mammalian lineages suggest that modifications to our metagenome and/or microbiome occur following certain environmental exposures during pregnancy and lactation, which in turn render risk of childhood and adult diseases. In this review, we will consider the evidence suggesting that development of the offspring microbiome may be vulnerable to maternal exposures, including an analysis of the data regarding the presence or absence of a low-biomass intrauterine microbiome.
Collapse
Affiliation(s)
- Erin E Bolte
- Translational Biology and Molecular Medicine Graduate Program, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, USA
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine and Texas Children's Hospital, Houston, USA
| | - David Moorshead
- Immunology & Microbiology Graduate Program, Baylor College of Medicine, Houston, USA
- Medical Scientist Training Program, Baylor College of Medicine, Houston, USA
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine and Texas Children's Hospital, Houston, USA
| | - Kjersti M Aagaard
- Translational Biology and Molecular Medicine Graduate Program, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA.
- Immunology & Microbiology Graduate Program, Baylor College of Medicine, Houston, USA.
- Medical Scientist Training Program, Baylor College of Medicine, Houston, USA.
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine and Texas Children's Hospital, Houston, USA.
- Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, USA.
- Department of Molecular & Cell Biology, Baylor College of Medicine, Houston, USA.
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, USA.
| |
Collapse
|
14
|
Lisboa PC, Miranda RA, Souza LL, Moura EG. Can breastfeeding affect the rest of our life? Neuropharmacology 2021; 200:108821. [PMID: 34610290 DOI: 10.1016/j.neuropharm.2021.108821] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/17/2021] [Accepted: 09/30/2021] [Indexed: 12/31/2022]
Abstract
The breastfeeding period is one of the most important critical windows in our development, since milk, our first food after birth, contains several compounds, such as macronutrients, micronutrients, antibodies, growth factors and hormones that benefit human health. Indeed, nutritional, and environmental alterations during lactation, change the composition of breast milk and induce alterations in the child's development, such as obesity, leading to the metabolic dysfunctions, cardiovascular diseases and neurobehavioral disorders. This review is based on experimental animal models, most of them in rodents, and summarizes the impact of an adequate breast milk supply in view of the developmental origins of health and disease (DOHaD) concept, which has been proposed by researchers in the areas of epidemiology and basic science from around the world. Here, experimental advances in understanding the programming during breastfeeding were compiled with the purpose of generating knowledge about the genesis of chronic noncommunicable diseases and to guide the development of public policies to deal with and prevent the problems arising from this phenomenon. This review article is part of the special issue on "Cross talk between periphery and brain".
Collapse
Affiliation(s)
- Patricia C Lisboa
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Rosiane A Miranda
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Luana L Souza
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Egberto G Moura
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
15
|
Development of the circadian system and relevance of periodic signals for neonatal development. HANDBOOK OF CLINICAL NEUROLOGY 2021; 179:249-258. [PMID: 34225966 DOI: 10.1016/b978-0-12-819975-6.00015-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Circadian rhythms are generated endogenously with a period of approximately 24h. Studies carried out during the last decade indicate that the circadian system develops before birth, and that the suprachiasmatic nucleus, a structure that is considered the mammalian circadian clock, is present in primates from the middle of pregnancy. Recent evidence shows that the infants' circadian system is sensitive to light from very early stages of development; it has also been proposed that low-intensity lighting can regulate the developing clock. After birth there is a progressive maturation of the outputs of the circadian system with marked rhythms in sleep-wake phenomena and hormone secretion. These facts express the importance of circadian photic regulation in infants. Thus, the exposure of premature babies to light/dark cycles results in a rapid establishment of activity/rest patterns, which are in the light-dark cycle. With the continuous study of the development of the circadian system and the influence on human physiology and disease, it is anticipated that the application of circadian biology will become an increasingly important component in the perinatal care.
Collapse
|
16
|
Elsakr JM, Zhao SK, Ricciardi V, Dean TA, Takahashi DL, Sullivan E, Wesolowski SR, McCurdy CE, Kievit P, Friedman JE, Aagaard KM, Edwards DRV, Gannon M. Western-style diet consumption impairs maternal insulin sensitivity and glucose metabolism during pregnancy in a Japanese macaque model. Sci Rep 2021; 11:12977. [PMID: 34155315 PMCID: PMC8217225 DOI: 10.1038/s41598-021-92464-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 06/04/2021] [Indexed: 12/26/2022] Open
Abstract
The prevalence of maternal obesity is increasing in the United States. Offspring born to women with obesity or poor glycemic control have greater odds of becoming obese and developing metabolic disease later in life. Our group has utilized a macaque model to study the metabolic effects of consumption of a calorically-dense, Western-style diet (WSD; 36.3% fat) during pregnancy. Here, our objective was to characterize the effects of WSD and obesity, alone and together, on maternal glucose tolerance and insulin levels in dams during each pregnancy. Recognizing the collinearity of maternal measures, we adjusted for confounding factors including maternal age and parity. Based on intravenous glucose tolerance tests, dams consuming a WSD showed lower glucose area under the curve during first study pregnancies despite increased body fat percentage and increased insulin area under the curve. However, with (1) prolonged WSD feeding, (2) multiple diet switches, and/or (3) increasing age and parity, WSD was associated with increasingly higher insulin levels during glucose tolerance testing, indicative of insulin resistance. Our results suggest that prolonged or recurrent calorically-dense WSD and/or increased parity, rather than obesity per se, drive excess insulin resistance and metabolic dysfunction. These observations in a highly relevant species are likely of clinical and public health importance given the comparative ease of maternal dietary modifications relative to the low likelihood of successfully reversing obesity in the course of any given pregnancy.
Collapse
Affiliation(s)
- Joseph M Elsakr
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
| | - Sifang Kathy Zhao
- Division of Quantitative Sciences, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, 2525 West End Avenue, Suite 600, Nashville, TN, 37203-1738, USA
| | - Valerie Ricciardi
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, 2213 Garland Avenue, 7465 MRBIV, Nashville, TN, 37232-0475, USA
| | - Tyler A Dean
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Diana L Takahashi
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Elinor Sullivan
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, USA
| | | | - Carrie E McCurdy
- Department of Human Physiology, University of Oregon, Eugene, OR, 97403, USA
| | - Paul Kievit
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Jacob E Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Kjersti M Aagaard
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, and Molecular and Human Genetics, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, 77030, USA
| | - Digna R Velez Edwards
- Division of Quantitative Sciences, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, 2525 West End Avenue, Suite 600, Nashville, TN, 37203-1738, USA.
- Department of Biomedical Informatics, Department of Obstetrics and Gynecology, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Maureen Gannon
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA.
- Division of Diabetes, Endocrinology, and Metabolism, Department of Medicine, Vanderbilt University Medical Center, 2213 Garland Avenue, 7465 MRBIV, Nashville, TN, 37232-0475, USA.
- Department of Veterans Affairs Tennessee Valley, Nashville, TN, USA.
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA.
| |
Collapse
|
17
|
Carroll DT, Sassin AM, Aagaard KM, Gannon M. Developmental effects of in utero metformin exposure. TRENDS IN DEVELOPMENTAL BIOLOGY 2021; 14:1-17. [PMID: 36589485 PMCID: PMC9802655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
According to the Developmental Origins of Health and Disease (DOHaD) hypothesis, the intrauterine environment influences fetal programming and development, affecting offspring disease susceptibility in adulthood. In recent years, therapeutic use of the Type 2 diabetes drug metformin has expanded to the treatment of pre-diabetes, polycystic ovarian syndrome, and gestational diabetes. Because metformin both undergoes renal excretion and binds to receptors on the placenta, the fetus receives equivalent maternal dosing. Although no teratogenic nor short-term harmful fetal impact of metformin is known to occur, the effects of metformin exposure on longer-range offspring development have not yet been fully elucidated. This review encapsulates the (albeit limited) existing knowledge regarding the potential longer-term impact of intrauterine metformin exposure on the development of key organs including the liver, central nervous system, heart, gut, and endocrine pancreas in animal models and humans. We discuss molecular and cellular mechanisms that would be altered in response to treatment and describe the potential consequences of these developmental changes on postnatal health. Further studies regarding the influence of metformin exposure on fetal programming and adult metabolic health will provide necessary insight to its long-term risks, benefits, and limitations in order to guide decisions for use of metformin during pregnancy.
Collapse
Affiliation(s)
- Darian T. Carroll
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
| | - Alexa M. Sassin
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, and Departments of Molecular and Human Genetics, and Molecular and Cell Biology, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX
| | - Kjersti M. Aagaard
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, and Departments of Molecular and Human Genetics, and Molecular and Cell Biology, Baylor College of Medicine and Texas Children’s Hospital, Houston, TX
| | - Maureen Gannon
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN
- Department of Veterans Affairs Tennessee Valley, Nashville, TN
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
18
|
Banerjee S, Suter MA, Aagaard KM. Interactions between Environmental Exposures and the Microbiome: Implications for Fetal Programming. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2020; 13:39-48. [PMID: 33283070 PMCID: PMC7716732 DOI: 10.1016/j.coemr.2020.09.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Decades of population-based health outcomes data highlight the importance of understanding how environmental exposures in pregnancy affect maternal and neonatal outcomes. Animal model research and epidemiological studies have revealed that such exposures are able to alter fetal programming through stable changes in the epigenome, including altered DNA methylation patterns and histone modifications in the developing fetus and infant. It is similarly known that while microbes can biotransform environmental chemicals via conjugation and de-conjugation, specific exposures can also alter the community profile and function of the human microbiome. In this review, we consider how alterations to the maternal and or fetal/infant microbiome through environmental exposures could directly and indirectly alter fetal programming. We highlight two specific environmental exposures, cadmium (Cd) and polycyclic aromatic hydrocarbons (PAHs), and outline their effects on the developing fetus and the perinatal (maternal and fetal/infant) microbiome. We further consider how chemical exposures in the setting of natural disasters may be of particular importance to environmental health.
Collapse
Affiliation(s)
- Sohini Banerjee
- Baylor College of Medicine, Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine and Departments of Molecular & Human Genetics, Molecular & Cell Biology, and Molecular Physiology & Biophysics, 1 Baylor Plaza, Houston, TX 77030
| | - Melissa A. Suter
- Baylor College of Medicine, Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine and Departments of Molecular & Human Genetics, Molecular & Cell Biology, and Molecular Physiology & Biophysics, 1 Baylor Plaza, Houston, TX 77030
| | - Kjersti M. Aagaard
- Baylor College of Medicine, Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine and Departments of Molecular & Human Genetics, Molecular & Cell Biology, and Molecular Physiology & Biophysics, 1 Baylor Plaza, Houston, TX 77030
| |
Collapse
|
19
|
Huber HF, Jenkins SL, Li C, Nathanielsz PW. Strength of nonhuman primate studies of developmental programming: review of sample sizes, challenges, and steps for future work. J Dev Orig Health Dis 2020; 11:297-306. [PMID: 31566171 PMCID: PMC7103515 DOI: 10.1017/s2040174419000539] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Nonhuman primate (NHP) studies are crucial to biomedical research. NHPs are the species most similar to humans in lifespan, body size, and hormonal profiles. Planning research requires statistical power evaluation, which is difficult to perform when lacking directly relevant preliminary data. This is especially true for NHP developmental programming studies, which are scarce. We review the sample sizes reported, challenges, areas needing further work, and goals of NHP maternal nutritional programming studies. The literature search included 27 keywords, for example, maternal obesity, intrauterine growth restriction, maternal high-fat diet, and maternal nutrient reduction. Only fetal and postnatal offspring studies involving tissue collection or imaging were included. Twenty-eight studies investigated maternal over-nutrition and 33 under-nutrition; 23 involved macaques and 38 baboons. Analysis by sex was performed in 19; minimum group size ranged from 1 to 8 (mean 4.7 ± 0.52, median 4, mode 3) and maximum group size from 3 to 16 (8.3 ± 0.93, 8, 8). Sexes were pooled in 42 studies; minimum group size ranged from 2 to 16 (mean 5.3 ± 0.35, median 6, mode 6) and maximum group size from 4 to 26 (10.2 ± 0.92, 8, 8). A typical study with sex-based analyses had group size minimum 4 and maximum 8 per sex. Among studies with sexes pooled, minimum group size averaged 6 and maximum 8. All studies reported some significant differences between groups. Therefore, studies with group sizes 3-8 can detect significance between groups. To address deficiencies in the literature, goals include increasing age range, more frequently considering sex as a biological variable, expanding topics, replicating studies, exploring intergenerational effects, and examining interventions.
Collapse
Affiliation(s)
- Hillary F. Huber
- Department of Animal Science, University of Wyoming, Laramie, WY, USA
| | - Susan L. Jenkins
- Department of Animal Science, University of Wyoming, Laramie, WY, USA
| | - Cun Li
- Department of Animal Science, University of Wyoming, Laramie, WY, USA
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| | - Peter W. Nathanielsz
- Department of Animal Science, University of Wyoming, Laramie, WY, USA
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX, USA
| |
Collapse
|
20
|
Valentine G, Prince A, Aagaard KM. The Neonatal Microbiome and Metagenomics: What Do We Know and What Is the Future? Neoreviews 2020; 20:e258-e271. [PMID: 31261078 DOI: 10.1542/neo.20-5-e258] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The human microbiota includes the trillions of microorganisms living in the human body whereas the human microbiome includes the genes and gene products of this microbiota. Bacteria were historically largely considered to be pathogens that inevitably led to human disease. However, because of advances in both cultivation-based methods and the advent of metagenomics, bacteria are now recognized to be largely beneficial commensal organisms and thus, key to normal and healthy human development. This relatively new area of medical research has elucidated insights into diseases such as inflammatory bowel disease and obesity, as well as metabolic and atopic disorders. However, much remains unknown about the complexity of microbe-microbe and microbe-host interactions. Future efforts aimed at answering key questions pertaining to the early establishment of the microbiome, alongside what defines its dysbiosis, will likely lead to long-term health and mitigation of disease. Here, we review the relevant literature pertaining to modulations in the perinatal and neonatal microbiome, the impact of environmental and maternal factors in shaping the neonatal microbiome, and future questions and directions in the exciting emerging arena of metagenomic medicine.
Collapse
Affiliation(s)
- Gregory Valentine
- Department of Pediatrics.,Division of Neonatology at Texas Children's Hospital, Houston, TX
| | - Amanda Prince
- Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine
| | - Kjersti M Aagaard
- Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine.,Center for Microbiome and Metagenomics Research, and Departments of.,Molecular & Human Genetics and.,Molecular & Cell Biology, Baylor College of Medicine, Houston, TX
| |
Collapse
|
21
|
Cleal JK, Bruce KD, Shearer JL, Thomas H, Plume J, Gregory L, Shepard JN, Spiers-Fitzgerald KL, Mani R, Lewis RM, Lillycrop KA, Hanson MA, Byrne CD, Cagampang FR. Maternal Obesity during Pregnancy Alters Daily Activity and Feeding Cycles, and Hypothalamic Clock Gene Expression in Adult Male Mouse Offspring. Int J Mol Sci 2019; 20:E5408. [PMID: 31671625 PMCID: PMC6862679 DOI: 10.3390/ijms20215408] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 10/12/2019] [Accepted: 10/28/2019] [Indexed: 02/07/2023] Open
Abstract
An obesogenic diet adversely affects the endogenous mammalian circadian clock, altering daily activity and metabolism, and resulting in obesity. We investigated whether an obese pregnancy can alter the molecular clock in the offspring hypothalamus, resulting in changes to their activity and feeding rhythms. Female mice were fed a control (C, 7% kcal fat) or high fat diet (HF, 45% kcal fat) before mating and throughout pregnancy. Male offspring were fed the C or HF diet postweaning, resulting in four offspring groups: C/C, C/HF, HF/C, and HF/HF. Daily activity and food intake were monitored, and at 15 weeks of age were killed at six time-points over 24 h. The clock genes Clock, Bmal1, Per2, and Cry2 in the suprachiasmatic nucleus (SCN) and appetite genes Npy and Pomc in the arcuate nucleus (ARC) were measured. Daily activity and feeding cycles in the HF/C, C/HF, and HF/HF offspring were altered, with increased feeding bouts and activity during the day and increased food intake but reduced activity at night. Gene expression patterns and levels of Clock, Bmal1, Per2, and Cry2 in the SCN and Npy and Pomc in the ARC were altered in HF diet-exposed offspring. The altered expression of hypothalamic molecular clock components and appetite genes, together with changes in activity and feeding rhythms, could be contributing to offspring obesity.
Collapse
Affiliation(s)
- Jane K Cleal
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Kimberley D Bruce
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Jasmin L Shearer
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Hugh Thomas
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Jack Plume
- Centre for Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton SO17 1BJ, UK.
| | - Louise Gregory
- Centre for Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton SO17 1BJ, UK.
| | - James N Shepard
- Centre for Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton SO17 1BJ, UK.
| | - Kerry L Spiers-Fitzgerald
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Ravi Mani
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Rohan M Lewis
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Karen A Lillycrop
- Centre for Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton SO17 1BJ, UK.
| | - Mark A Hanson
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Christopher D Byrne
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Felino R Cagampang
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| |
Collapse
|
22
|
Chu DM, Valentine GC, Seferovic MD, Aagaard KM. The Development of the Human Microbiome: Why Moms Matter. Gastroenterol Clin North Am 2019; 48:357-375. [PMID: 31383276 PMCID: PMC7261593 DOI: 10.1016/j.gtc.2019.04.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The human body is cohabitated with trillions of commensal bacteria that are essential for our health. However, certain bacteria can also cause diseases in the human host. Before the microbiome can be attributed to disease risk and pathogenesis, normal acquisition and development of the microbiome must be understood. Here, we explore the evidence surrounding in utero microbial exposures and the significant of this exposure in the proper development of the fetal and neonatal microbiome. We further explore the development of the fetal and neonatal microbiome and its relationship to preterm birth, feeding practices, and mode of delivery, and maternal diet.
Collapse
Affiliation(s)
| | | | | | - Kjersti M. Aagaard
- Corresponding author. Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine, Texas Children’s Hospital, 1 Baylor Plaza, Houston,TX 77030, USA.
| |
Collapse
|
23
|
Prince AL, Pace RM, Dean T, Takahashi D, Kievit P, Friedman JE, Aagaard KM. The development and ecology of the Japanese macaque gut microbiome from weaning to early adolescence in association with diet. Am J Primatol 2019; 81:e22980. [PMID: 31066111 DOI: 10.1002/ajp.22980] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 04/08/2019] [Accepted: 04/14/2019] [Indexed: 02/06/2023]
Abstract
Previously we have shown that the Japanese macaque gut microbiome differs not by obesity per se, but rather in association with high-fat diet (HFD) feeding. This held true for both pregnant dams, as well as their 1-year-old offspring, even when weaned onto a control diet. Here we aimed to examine the stability of the gut microbiome over time and in response to maternal and postweaning HFD feeding from 6 months of age, and at 1 and 3 years of age. In both cross-sectional and longitudinal specimens, we performed analysis of the V4 hypervariable region of the 16S rRNA gene on anus swabs collected from pregnant dams and their juveniles at age 6 months to 3 years (n = 55). Extracted microbial DNA was subjected to 16S-amplicon-based metagenomic sequencing on the Illumina MiSeq platform. We initially identified 272 unique bacterial genera, and multidimensional scaling revealed samples to cluster by age and diet exposures. Dirichlet multinomial mixture modeling of microbiota abundances enabled identification of two predominant enterotypes to which samples sorted, characterized primarily by Treponema abundance, or lack thereof. Approximating the time of initial weaning (6 months), the Japanese macaque offspring microbiome underwent a significant state type transition which stabilized from 1 to 3 years of age. However, we also found the low abundance Treponema enterotype to be strongly associated with HFD exposure, be it during gestation/lactation or in the postweaning interval. Examination of taxonomic co-occurrences revealed samples within the low Treponema cluster were relatively permissive (allowing for increased interactions between microbiota) whereas samples within the high Treponema cluster were relatively exclusionary (suggesting decreased interactions amongst microbiota). Taken together, these findings suggest that Treponemes are keystone species in the developing gut microbiome of the gut, and susceptible to HFD feeding in their relative abundance.
Collapse
Affiliation(s)
- Amanda L Prince
- Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine, Houston, Texas
| | - Ryan M Pace
- Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine, Houston, Texas
| | - Tyler Dean
- Divisions of Cardiometabolic Health and Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon
| | - Diana Takahashi
- Divisions of Cardiometabolic Health and Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon
| | - Paul Kievit
- Divisions of Cardiometabolic Health and Reproductive and Developmental Sciences, Oregon National Primate Research Center, Beaverton, Oregon
| | - Jacob E Friedman
- Department of Pediatrics-Neonatology, University of Colorado, Aurora, Colorado
| | - Kjersti M Aagaard
- Department of Obstetrics & Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine, Houston, Texas.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas.,Department of Molecular and Cell Biology, Bayor College of Medicine, Houston, Texas
| |
Collapse
|
24
|
Cerf ME. High Fat Programming and Cardiovascular Disease. MEDICINA (KAUNAS, LITHUANIA) 2018; 54:E86. [PMID: 30428585 PMCID: PMC6262472 DOI: 10.3390/medicina54050086] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/31/2018] [Accepted: 11/08/2018] [Indexed: 02/06/2023]
Abstract
Programming is triggered through events during critical developmental phases that alter offspring health outcomes. High fat programming is defined as the maintenance on a high fat diet during fetal and/or early postnatal life that induces metabolic and physiological alterations that compromise health. The maternal nutritional status, including the dietary fatty acid composition, during gestation and/or lactation, are key determinants of fetal and postnatal development. A maternal high fat diet and obesity during gestation compromises the maternal metabolic state and, through high fat programming, presents an unfavorable intrauterine milieu for fetal growth and development thereby conferring adverse cardiac outcomes to offspring. Stressors on the heart, such as a maternal high fat diet and obesity, alter the expression of cardiac-specific factors that alter cardiac structure and function. The proper nutritional balance, including the fatty acid balance, particularly during developmental windows, are critical for maintaining cardiac structure, preserving cardiac function and enhancing the cardiac response to metabolic challenges.
Collapse
Affiliation(s)
- Marlon E Cerf
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg 7505, South Africa.
- Division of Medical Physiology, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University of Stellenbosch, Tygerberg 7505, South Africa.
| |
Collapse
|
25
|
Friedman JE. Developmental Programming of Obesity and Diabetes in Mouse, Monkey, and Man in 2018: Where Are We Headed? Diabetes 2018; 67:2137-2151. [PMID: 30348820 PMCID: PMC6198344 DOI: 10.2337/dbi17-0011] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 08/21/2018] [Indexed: 12/11/2022]
Abstract
Childhood obesity and its comorbidities continue to accelerate across the globe. Two-thirds of pregnant women are obese/overweight, as are 20% of preschoolers. Gestational diabetes mellitus (GDM) is escalating, affecting up to 1 in 5 pregnant women. The field of developmental origins of health and disease has begun to move beyond associations to potential causal mechanisms for developmental programming. Evidence across species compellingly demonstrates that maternal obesity, diabetes, and Western-style diets create a long-lasting signature on multiple systems, including infant stem cells, the early immune system, and gut microbiota. Such exposures accelerate adipogenesis, disrupt mitochondrial metabolism, and impair energy sensing, affecting neurodevelopment, liver, pancreas, and skeletal muscle. Attempts to prevent developmental programming have met with very limited success. A challenging level of complexity is involved in how the host genome, metabolome, and microbiome throughout pregnancy and lactation increase the offspring's risk of metabolic diseases across the life span. Considerable gaps in knowledge include the timing of exposure(s) and permanence or plasticity of the response, encompassing effects from both maternal and paternal dysmetabolism. Basic, translational, and human intervention studies targeting pathways that connect diet, microbiota, and metabolism in mothers with obesity/GDM and their infants are a critical unmet need and present new challenges for disease prevention in the next generation.
Collapse
Affiliation(s)
- Jacob E Friedman
- Section of Neonatology, Department of Pediatrics; Department of Biochemistry & Molecular Genetics; Division of Endocrinology, Metabolism & Diabetes, Department of Medicine; and Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
26
|
Dorokhov VB, Puchkova AN, Arsen’ev GN, Slominsky PA, Dementienko VV, Sveshnikov DS, Putilov AA. Association of obesity in shift workers with the minor allele of a single-nucleotide polymorphism (rs4851377) in the largest circadian clock gene (NPAS2). BIOL RHYTHM RES 2018. [DOI: 10.1080/09291016.2018.1537558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Vladimir B. Dorokhov
- Laboratory of Sleep/Wake Neurobiology, the Institute of Higher Nervous Activity and Neurophysiology of the Russian Academy of Sciences, Moscow, Russia
| | - Alexandra N. Puchkova
- Laboratory of Sleep/Wake Neurobiology, the Institute of Higher Nervous Activity and Neurophysiology of the Russian Academy of Sciences, Moscow, Russia
| | - Gleb N. Arsen’ev
- Laboratory of Sleep/Wake Neurobiology, the Institute of Higher Nervous Activity and Neurophysiology of the Russian Academy of Sciences, Moscow, Russia
| | - Petr A. Slominsky
- Laboratory of Molecular Genetics of Hereditary Diseases, the Institute of Molecular Genetics of the Russian Academy of Sciences, Moscow, Russia
| | - Valeriy V. Dementienko
- Laboratory of Medical Electronics, Kotelnikov Institute of Radio Engineering and Electronics, Russian Academy of Science, Moscow, Russia
| | - Dmitry S. Sveshnikov
- Department of Normal Physiology, Medical Institute, Peoples’ Friendship University of Russia, Moscow, Russia
| | - Arcady A. Putilov
- Laboratory of Sleep/Wake Neurobiology, the Institute of Higher Nervous Activity and Neurophysiology of the Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
27
|
Relationships Between Perinatal Interventions, Maternal-Infant Microbiomes, and Neonatal Outcomes. Clin Perinatol 2018; 45:339-355. [PMID: 29747892 DOI: 10.1016/j.clp.2018.01.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The human microbiome acquires its vastness and diversity over a relatively short time period during development. Much is unknown, however, about the precise prenatal versus postnatal timing or its sources and determinants. Given early evidence of a role for influences during pregnancy and early neonatal and infant life on the microbiome and subsequent metabolic health, research investigating the development and shaping of the microbiome in the fetus and neonate is an important arena for study. This article reviews the relevant available literature and future questions on what shapes the microbiome during early development and mechanisms for doing so.
Collapse
|
28
|
Pace RM, Prince AL, Ma J, Belfort BDW, Harvey AS, Hu M, Baquero K, Blundell P, Takahashi D, Dean T, Kievit P, Sullivan EL, Friedman JE, Grove K, Aagaard KM. Modulations in the offspring gut microbiome are refractory to postnatal synbiotic supplementation among juvenile primates. BMC Microbiol 2018; 18:28. [PMID: 29621980 PMCID: PMC5887201 DOI: 10.1186/s12866-018-1169-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 03/19/2018] [Indexed: 02/07/2023] Open
Abstract
Background We and others have previously shown that alterations in the mammalian gut microbiome are associated with diet, notably early life exposure to a maternal high fat diet (HFD). Here, we aimed to further these studies by examining alterations in the gut microbiome of juvenile Japanese macaques (Macaca fuscata) that were exposed to a maternal HFD, weaned onto a control diet, and later supplemented with a synbiotic comprised of psyllium seed and Enterococcus and Lactobacillus species. Results Eighteen month old offspring (n = 7) of 36% HFD fed dams were fed a control (14% fat) diet post weaning, then were synbiotic supplemented for 75 days and longitudinal stool and serum samples were obtained. All stool samples were subjected to 16S rRNA metagenomic sequencing, and microbiome profiles and serum lipids and triglycerides were compared to untreated, healthy age matched and diet matched controls (n = 7). Overall, 16S-based metagenomic analysis revealed that supplementation exerted minimal alterations to the gut microbiome including transient increased abundance of Lactobacillus species and decreased abundance of few bacterial genera, including Faecalibacterium and Anaerovibrio. However, serum lipid analysis revealed significant decreases in triglycerides, cholesterol, and LDL (p < 0.05). Nevertheless, supplemented juveniles challenged 4 months later were not protected from HFD-induced gut dysbiosis. Conclusions Synbiotic supplementation is temporally associated with alterations in the gut microbiome and host lipid profiles of juvenile Japanese macaques that were previously exposed to a maternal HFD. Despite these presumptive temporal benefits, a protective effect against later HFD-challenge gut dysbiosis was not observed. Electronic supplementary material The online version of this article (10.1186/s12866-018-1169-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ryan M Pace
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Amanda L Prince
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jun Ma
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Benjamin D W Belfort
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Alexia S Harvey
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Min Hu
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Karalee Baquero
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97239, USA
| | - Peter Blundell
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97239, USA
| | - Diana Takahashi
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97239, USA
| | - Tyler Dean
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97239, USA
| | - Paul Kievit
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97239, USA
| | - Elinor L Sullivan
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97239, USA.,Biology Department, University of Portland, Portland, OR, 97203, USA
| | - Jacob E Friedman
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kevin Grove
- Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97239, USA
| | - Kjersti M Aagaard
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX, 77030, USA. .,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA. .,Oregon National Primate Research Center, Oregon Health & Science University, Beaverton, OR, 97239, USA.
| |
Collapse
|
29
|
Crew RC, Waddell BJ, Mark PJ. Obesity-induced changes in hepatic and placental clock gene networks in rat pregnancy†. Biol Reprod 2017; 98:75-88. [DOI: 10.1093/biolre/iox158] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Accepted: 11/24/2017] [Indexed: 12/13/2022] Open
|
30
|
Imam MU, Ismail M. The Impact of Traditional Food and Lifestyle Behavior on Epigenetic Burden of Chronic Disease. GLOBAL CHALLENGES (HOBOKEN, NJ) 2017; 1:1700043. [PMID: 31565292 PMCID: PMC6607231 DOI: 10.1002/gch2.201700043] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 07/12/2017] [Indexed: 05/11/2023]
Abstract
Noncommunicable chronic diseases (NCCDs) are the leading causes of morbidity and mortality globally. The mismatch between present day diets and ancestral genome is suggested to contribute to the NCCDs burden, which is promoted by traditional risk factors like unhealthy diets, physical inactivity, alcohol and tobacco. However, epigenetic evidence now suggests that cumulatively inherited epigenetic modifications may have made humans more prone to the effects of present day lifestyle factors. Perinatal starvation was widespread in the 19th century. This together with more recent events like increasing consumption of western and low fiber diets, smoking, harmful use of alcohol, physical inactivity, and environmental pollutants may have programed the human epigenome for higher NCCDs risk. In this review, on the basis of available epigenetic data it is hypothesized that transgenerational effects of lifestyle factors may be contributing to the current global burden of NCCDs. Thus, there is a need to reconsider prevention strategies so that the subsequent generations will not have to pay for our sins and those of our ancestors.
Collapse
Affiliation(s)
- Mustapha U. Imam
- Precision Nutrition Innovation InstituteCollege of Public HealthZhengzhou UniversityZhengzhou450001China
| | - Maznah Ismail
- Laboratory of Molecular BiomedicineInstitute of BioscienceUniversiti Putra MalaysiaSerdangSelangor43400Malaysia
| |
Collapse
|
31
|
Epigenetic effects of the pregnancy Mediterranean diet adherence on the offspring metabolic syndrome markers. J Physiol Biochem 2017; 73:495-510. [PMID: 28921259 DOI: 10.1007/s13105-017-0592-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 09/07/2017] [Indexed: 02/06/2023]
Abstract
Metabolic syndrome (MS) has a multifactorial and not yet fully clarified origin. Insulin resistance is a key element that connects all the accepted components of MS (obesity, dyslipemia, high blood pressure, and hyperglycemia). There is strong evidence that epigenetic changes during fetal development are key factors in the development of MS. These changes are induced by maternal nutrition, among different factors, affecting the intrauterine environment. The Mediterranean diet has been shown to be a healthy eating pattern that protects against the development of MS in adults. Similarly, the Mediterranean diet could have a similar action during pregnancy, protecting the fetus against the development of MS throughout life. This review assembles studies carried out, both in animals and humans, on the epigenetic modifications associated with the consumption, during pregnancy, of Mediterranean diet main components. The relationship between these modifications and the occurrence of factors involved in development of MS is also explained. In addition, the results of our group relating adherence to the Mediterranean diet with MS markers are discussed. The paper ends suggesting future actuation lines in order to increase knowledge on Mediterranean diet adherence as a prevention tool of MS development.
Collapse
|
32
|
Gene-by-environment interactions of the CLOCK, PEMT, and GHRELIN loci with average sleep duration in relation to obesity traits using a cohort of 643 New Zealand European children. Sleep Med 2017; 37:19-26. [DOI: 10.1016/j.sleep.2017.05.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 05/15/2017] [Accepted: 05/21/2017] [Indexed: 12/12/2022]
|
33
|
O'Neil DS, Stewart CJ, Chu DM, Goodspeed DM, Gonzalez-Rodriguez PJ, Shope CD, Aagaard KM. Conditional postnatal deletion of the neonatal murine hepatic circadian gene, Npas2, alters the gut microbiome following restricted feeding. Am J Obstet Gynecol 2017; 217:218.e1-218.e15. [PMID: 28373017 PMCID: PMC5545073 DOI: 10.1016/j.ajog.2017.03.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 03/03/2017] [Accepted: 03/23/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND We have recently shown in both non-human primates and in rodents that fetal and neonatal hepatic expression of the circadian transcription factor, Npas2, is modulated by a high fat maternal diet and plays a critical role in establishing life-long metabolic homeostasis. Similarly, we and others have also established the importance of the maternal and early postnatal diet on establishment of the early gut microbiome. OBJECTIVE We hypothesized that altered circadian gene expression solely in the neonatal liver would result in gut microbiome dysbiosis, especially with diet-induced metabolic stress (ie, restricted feeding). Using a murine model in which we conditionally knock out Npas2 in the neonatal liver, we aimed to determine the role of the circadian machinery in gut dysbiosis with restricted feeding. STUDY DESIGN We collected fecal samples from liver Npas2 conditional knockout (n = 11) and wild-type (n = 13) reproductive-aged mice before (study day 0) and after the restricted feeding study (study day 17). Extracted DNA was sequenced using the MiSeq Illumina platform using primers specific for the V4 region of the 16S ribosomal DNA gene. The resulting sequences were quality filtered, aligned, and assigned taxonomy. Principal coordinate analysis was performed on unweighted and weighted UniFrac distances between samples with a permutation analysis of variance to assess clustering significance between groups. Microbial taxa that significantly differ between groups of interest was determined using linear discriminate analysis effect size and randomForrest. RESULTS Principal coordinate analysis performed on weighted UniFrac distances between male conditional knockout and wild-type cohorts revealed that the gut microbiome of the mice did not differ by genotype at the start of the restricted feeding study but did differ by virtue of genotype at the end of the study (P = .001). Moreover, these differences could be at least partially attributed to restricted feeding-associated alterations in relative abundance of the Bacteroides genus, which has been implicated as crucial to establishing a healthy gut microbiome early in development. CONCLUSION Here we have provided an initial key insight into the interplay between neonatal establishment of the peripheral circadian clock in the liver and the ability of the gut microbiome to respond to dietary and metabolic stress. Because Npas2 expression in the liver is a target of maternal high-fat diet-induced metabolic perturbations during fetal development, we speculate that these findings have potential implications in the long-term metabolic health of their offspring.
Collapse
Affiliation(s)
- Derek S O'Neil
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, Baylor College of Medicine, Houston, TX; Interdepartmental Graduate Program of Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX
| | - Christopher J Stewart
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX
| | - Derrick M Chu
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, Baylor College of Medicine, Houston, TX; Interdepartmental Graduate Program of Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX
| | - Danielle M Goodspeed
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, Baylor College of Medicine, Houston, TX
| | - Pablo J Gonzalez-Rodriguez
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, Baylor College of Medicine, Houston, TX
| | - Cynthia D Shope
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, Baylor College of Medicine, Houston, TX
| | - Kjersti M Aagaard
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, Baylor College of Medicine, Houston, TX; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX; Department of Molecular and Cell Biology, Baylor College of Medicine, Houston, TX; Department of Molecular and Cellular Physiology, Baylor College of Medicine, Houston, TX; Interdepartmental Graduate Program of Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX.
| |
Collapse
|
34
|
Crossland RF, Balasa A, Ramakrishnan R, Mahadevan SK, Fiorotto ML, Van den Veyver IB. Chronic Maternal Low-Protein Diet in Mice Affects Anxiety, Night-Time Energy Expenditure and Sleep Patterns, but Not Circadian Rhythm in Male Offspring. PLoS One 2017; 12:e0170127. [PMID: 28099477 PMCID: PMC5242516 DOI: 10.1371/journal.pone.0170127] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 12/29/2016] [Indexed: 12/14/2022] Open
Abstract
Offspring of murine dams chronically fed a protein-restricted diet have an increased risk for metabolic and neurobehavioral disorders. Previously we showed that adult offspring, developmentally exposed to a chronic maternal low-protein (MLP) diet, had lower body and hind-leg muscle weights and decreased liver enzyme serum levels. We conducted energy expenditure, neurobehavioral and circadian rhythm assays in male offspring to examine mechanisms for the body-weight phenotype and assess neurodevelopmental implications of MLP exposure. C57BL/6J dams were fed a protein restricted (8%protein, MLP) or a control protein (20% protein, C) diet from four weeks before mating until weaning of offspring. Male offspring were weaned to standard rodent diet (20% protein) and single-housed until 8–12 weeks of age. We examined body composition, food intake, energy expenditure, spontaneous rearing activity and sleep patterns and performed behavioral assays for anxiety (open field activity, elevated plus maze [EPM], light/dark exploration), depression (tail suspension and forced swim test), sociability (three-chamber), repetitive (marble burying), learning and memory (fear conditioning), and circadian behavior (wheel-running activity during light-dark and constant dark cycles). We also measured circadian gene expression in hypothalamus and liver at different Zeitgeber times (ZT). Male offspring from separate MLP exposed dams had significantly greater body fat (P = 0.03), less energy expenditure (P = 0.004), less rearing activity (P = 0.04) and a greater number of night-time rest/sleep bouts (P = 0.03) compared to control. MLP offspring displayed greater anxiety-like behavior in the EPM (P<0.01) but had no learning and memory deficit in fear-conditioning assay (P = 0.02). There was an effect of time on Per1, Per 2 and Clock circadian gene expression in the hypothalamus but not on circadian behavior. Thus, transplacental and early developmental exposure of dams to chronic MLP reduces food intake and energy expenditure, increases anxiety like behavior and disturbs sleep patterns but not circadian rhythm in adult male offspring.
Collapse
Affiliation(s)
- Randy F. Crossland
- Department of Obstetrics & Gynecology, Baylor College of Medicine, Houston, TX, United States of America
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States of America
| | - Alfred Balasa
- Department of Obstetrics & Gynecology, Baylor College of Medicine, Houston, TX, United States of America
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States of America
| | - Rajesh Ramakrishnan
- Department of Obstetrics & Gynecology, Baylor College of Medicine, Houston, TX, United States of America
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States of America
| | - Sangeetha K. Mahadevan
- Department of Obstetrics & Gynecology, Baylor College of Medicine, Houston, TX, United States of America
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States of America
- Interdepartmental Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, United States of America
| | - Marta L. Fiorotto
- USDA/Agricultural Research Service Children’s Nutrition Research Center, Houston, TX, United States of America
| | - Ignatia B. Van den Veyver
- Department of Obstetrics & Gynecology, Baylor College of Medicine, Houston, TX, United States of America
- Jan and Dan Duncan Neurological Research Institute at Texas Children’s Hospital, Houston, TX, United States of America
- Interdepartmental Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, United States of America
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States of America
- * E-mail:
| |
Collapse
|
35
|
Abstract
Recent human and animal studies investigating the roles of the genome, epigenome, and environmental cues have identified associations between offspring predisposition to life-long obesity/metabolic disease and epigenetic modifications such as DNA methylation. This review explores the mechanisms by which maternal exposures impair the health of not only the next generation but also potentially future generations of offspring.
Collapse
Affiliation(s)
- Kathleen Jaeger
- Washington University School of Medicine, St. Louis, Missouri
| | - Jessica L Saben
- Washington University School of Medicine, St. Louis, Missouri
| | - Kelle H Moley
- Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
36
|
Western diet in the perinatal period promotes dysautonomia in the offspring of adult rats. J Dev Orig Health Dis 2016; 8:216-225. [PMID: 27931267 DOI: 10.1017/s2040174416000623] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The present study investigated the impact of a western diet during gestation and lactation on the anthropometry, serum biochemical, blood pressure and cardiovascular autonomic control on the offspring. Male Wistar rats were divided into two groups according to their mother's diet received: control group (C: 18% calories of lipids) and westernized group (W: 32% calories of lipids). After weaning both groups received standard diet. On the 60th day of life, blood samples were collected for the analysis of fasting glucose and lipidogram. Cardiovascular parameters were measured on the same period. Autonomic nervous system modulation was evaluated by spectrum analysis of heart rate (HR) and systolic arterial pressure (SAP). The W increased glycemia (123±2 v. 155±2 mg/dl), low-density lipoprotein (15±1 v. 31±2 mg/dl), triglycerides (49±1 v. 85±2 mg/dl), total cholesterol (75±2 v. 86±2 mg/dl), and decreased high-density lipoprotein (50±4 v. 38±3 mg/dl), as well as increased body mass (209±4 v. 229±6 g) than C. Furthermore, the W showed higher SAP (130±4 v. 157±2 mmHg), HR (357±10 v. 428±14 bpm), sympathetic modulation to vessels (2.3±0.56 v. 6±0.84 mmHg2) and LF/HF ratio (0.15±0.01 v. 0.7±0.2) than C. These findings suggest that a western diet during pregnancy and lactation leads to overweight associated with autonomic misbalance and hypertension in adulthood.
Collapse
|
37
|
Gough EK, Moodie EE, Prendergast AJ, Ntozini R, Moulton LH, Humphrey JH, Manges AR. Linear growth trajectories in Zimbabwean infants. Am J Clin Nutr 2016; 104:1616-1627. [PMID: 27806980 PMCID: PMC5118730 DOI: 10.3945/ajcn.116.133538] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 09/14/2016] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Undernutrition in early life underlies 45% of child deaths globally. Stunting malnutrition (suboptimal linear growth) also has long-term negative effects on childhood development. Linear growth deficits accrue in the first 1000 d of life. Understanding the patterns and timing of linear growth faltering or recovery during this period is critical to inform interventions to improve infant nutritional status. OBJECTIVE We aimed to identify the pattern and determinants of linear growth trajectories from birth through 24 mo of age in a cohort of Zimbabwean infants. DESIGN We performed a secondary analysis of longitudinal data from a subset of 3338 HIV-unexposed infants in the Zimbabwe Vitamin A for Mothers and Babies trial. We used k-means clustering for longitudinal data to identify linear growth trajectories and multinomial logistic regression to identify covariates that were associated with each trajectory group. RESULTS For the entire population, the mean length-for-age z score declined from -0.6 to -1.4 between birth and 24 mo of age. Within the population, 4 growth patterns were identified that were each characterized by worsening linear growth restriction but varied in the timing and severity of growth declines. In our multivariable model, 1-U increments in maternal height and education and infant birth weight and length were associated with greater relative odds of membership in the least-growth restricted groups (A and B) and reduced odds of membership in the more-growth restricted groups (C and D). Male infant sex was associated with reduced odds of membership in groups A and B but with increased odds of membership in groups C and D. CONCLUSION In this population, all children were experiencing growth restriction but differences in magnitude were influenced by maternal height and education and infant sex, birth weight, and birth length, which suggest that key determinants of linear growth may already be established by the time of birth. This trial was registered at clinicaltrials.gov as NCT00198718.
Collapse
Affiliation(s)
- Ethan K Gough
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Canada
| | - Erica Em Moodie
- Department of Epidemiology, Biostatistics and Occupational Health, McGill University, Montreal, Canada
| | - Andrew J Prendergast
- Zvitambo Institute for Maternal Child Health Research, Harare, Zimbabwe.,Blizard Institute, Queen Mary University of London, London, United Kingdom
| | - Robert Ntozini
- Zvitambo Institute for Maternal Child Health Research, Harare, Zimbabwe
| | - Lawrence H Moulton
- Zvitambo Institute for Maternal Child Health Research, Harare, Zimbabwe.,Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; and
| | - Jean H Humphrey
- Zvitambo Institute for Maternal Child Health Research, Harare, Zimbabwe.,Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD; and
| | - Amee R Manges
- School of Population and Public Health, University of British Columbia, Vancouver, Canada
| |
Collapse
|
38
|
Frihauf JB, Fekete ÉM, Nagy TR, Levin BE, Zorrilla EP. Maternal Western diet increases adiposity even in male offspring of obesity-resistant rat dams: early endocrine risk markers. Am J Physiol Regul Integr Comp Physiol 2016; 311:R1045-R1059. [PMID: 27654396 PMCID: PMC5256983 DOI: 10.1152/ajpregu.00023.2016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Revised: 08/30/2016] [Accepted: 09/12/2016] [Indexed: 01/08/2023]
Abstract
Maternal overnutrition or associated complications putatively mediate the obesogenic effects of perinatal high-fat diet on developing offspring. Here, we tested the hypothesis that a Western diet developmental environment increases adiposity not only in male offspring from obesity-prone (DIO) mothers, but also in those from obesity-resistant (DR) dams, implicating a deleterious role for the Western diet per se. Selectively bred DIO and DR female rats were fed chow (17% kcal fat) or Western diet (32%) for 54 days before mating and, thereafter, through weaning. As intended, despite chow-like caloric intake, Western diet increased prepregnancy weight gain and circulating leptin levels in DIO, but not DR, dams. Yet, in both genotypes, maternal Western diet increased the weight and adiposity of preweanlings, as early as in DR offspring, and increased plasma leptin, insulin, and adiponectin of weanlings. Although body weight normalized with chow feeding during adolescence, young adult Western diet offspring subsequently showed decreased energy expenditure and, in DR offspring, decreased lipid utilization as a fuel substrate. By mid-adulthood, maternal Western diet DR offspring ate more chow, weighed more, and were fatter than controls. Thus, maternal Western diet covertly programmed increased adiposity in childhood and adulthood, disrupted relations of energy regulatory hormones with body fat, and decreased energy expenditure in offspring of lean, genetically obesity-resistant mothers. Maternal Western diet exposure alone, without maternal obesity or overnutrition, can promote offspring weight gain.
Collapse
Affiliation(s)
- Jennifer B Frihauf
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California
- Neurosciences Graduate Program, University of California, San Diego, La Jolla, California
| | - Éva M Fekete
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California
| | - Tim R Nagy
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, Alabama
| | - Barry E Levin
- Neurology Service, VA Medical Center, East Orange, New Jersey; and
- Department of Neurology, Rutgers, New Jersey Medical School, Newark, New Jersey
| | - Eric P Zorrilla
- Committee on the Neurobiology of Addictive Disorders, The Scripps Research Institute, La Jolla, California;
- Neurosciences Graduate Program, University of California, San Diego, La Jolla, California
| |
Collapse
|
39
|
Harris RA, Alcott CE, Sullivan EL, Takahashi D, McCurdy CE, Comstock S, Baquero K, Blundell P, Frias AE, Kahr M, Suter M, Wesolowski S, Friedman JE, Grove KL, Aagaard KM. Genomic Variants Associated with Resistance to High Fat Diet Induced Obesity in a Primate Model. Sci Rep 2016; 6:36123. [PMID: 27811965 PMCID: PMC5095882 DOI: 10.1038/srep36123] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 10/07/2016] [Indexed: 12/28/2022] Open
Abstract
Maternal obesity contributes to an increased risk of lifelong morbidity and mortality for both the mother and her offspring. In order to better understand the molecular mechanisms underlying these risks, we previously established and extensively characterized a primate model in Macaca fuscata (Japanese macaque). In prior studies we have demonstrated that a high fat, caloric dense maternal diet structures the offspring’s epigenome, metabolome, and intestinal microbiome. During the course of this work we have consistently observed that a 36% fat diet leads to obesity in the majority, but not all, of exposed dams. In the current study, we sought to identify the genomic loci rendering resistance to obesity despite chronic consumption of a high fat diet in macaque dams. Through extensive phenotyping together with exon capture array and targeted resequencing, we identified three novel single nucleotide polymorphisms (SNPs), two in apolipoprotein B (APOB) and one in phospholipase A2 (PLA2G4A) that significantly associated with persistent weight stability and insulin sensitivity in lean macaques. By application of explicit orthogonal modeling (NOIA), we estimated the polygenic and interactive nature of these loci against multiple metabolic traits and their measures (i.e., serum LDL levels) which collectively render an obesity resistant phenotype in our adult female dams.
Collapse
Affiliation(s)
- R Alan Harris
- Department of Obstetrics &Gynecology, Division of Maternal-Fetal Medicine at Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA.,Department of Molecular and Human Genetics at Baylor College of Medicine, Houston, TX, USA
| | - Callison E Alcott
- Developmental Biology Interdisciplinary Program at Baylor College of Medicine, Houston, TX, USA
| | - Elinor L Sullivan
- Oregon National Primate Research Center, Oregon Health &Science University (OHSU), Beaverton, OR, USA.,Department of Biology, University of Portland, USA
| | - Diana Takahashi
- Oregon National Primate Research Center, Oregon Health &Science University (OHSU), Beaverton, OR, USA
| | - Carrie E McCurdy
- Department of Human Physiology, University of Oregon, Eugene, OR, USA
| | - Sarah Comstock
- Department of Biology, Corban University, Salem, OR, USA
| | - Karalee Baquero
- Oregon National Primate Research Center, Oregon Health &Science University (OHSU), Beaverton, OR, USA
| | - Peter Blundell
- Oregon National Primate Research Center, Oregon Health &Science University (OHSU), Beaverton, OR, USA
| | - Antonio E Frias
- Oregon National Primate Research Center, Oregon Health &Science University (OHSU), Beaverton, OR, USA.,Department of Obstetrics &Gynecology, Division of Maternal-Fetal Medicine, OHSU, Portland, OR, USA
| | - Maike Kahr
- Department of Obstetrics &Gynecology, Division of Maternal-Fetal Medicine at Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - Melissa Suter
- Department of Obstetrics &Gynecology, Division of Maternal-Fetal Medicine at Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - Stephanie Wesolowski
- Departments of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Jacob E Friedman
- Departments of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Kevin L Grove
- Oregon National Primate Research Center, Oregon Health &Science University (OHSU), Beaverton, OR, USA
| | - Kjersti M Aagaard
- Department of Obstetrics &Gynecology, Division of Maternal-Fetal Medicine at Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA.,Department of Molecular and Human Genetics at Baylor College of Medicine, Houston, TX, USA.,Developmental Biology Interdisciplinary Program at Baylor College of Medicine, Houston, TX, USA.,Oregon National Primate Research Center, Oregon Health &Science University (OHSU), Beaverton, OR, USA.,Department of Molecular and Cell Biology at Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
40
|
McCurdy CE, Schenk S, Hetrick B, Houck J, Drew BG, Kaye S, Lashbrook M, Bergman BC, Takahashi DL, Dean TA, Nemkov T, Gertsman I, Hansen KC, Philp A, Hevener AL, Chicco AJ, Aagaard KM, Grove KL, Friedman JE. Maternal obesity reduces oxidative capacity in fetal skeletal muscle of Japanese macaques. JCI Insight 2016; 1:e86612. [PMID: 27734025 DOI: 10.1172/jci.insight.86612] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Maternal obesity is proposed to alter the programming of metabolic systems in the offspring, increasing the risk for developing metabolic diseases; however, the cellular mechanisms remain poorly understood. Here, we used a nonhuman primate model to examine the impact of a maternal Western-style diet (WSD) alone, or in combination with obesity (Ob/WSD), on fetal skeletal muscle metabolism studied in the early third trimester. We find that fetal muscle responds to Ob/WSD by upregulating fatty acid metabolism, mitochondrial complex activity, and metabolic switches (CPT-1, PDK4) that promote lipid utilization over glucose oxidation. Ob/WSD fetuses also had reduced mitochondrial content, diminished oxidative capacity, and lower mitochondrial efficiency in muscle. The decrease in oxidative capacity and glucose metabolism was persistent in primary myotubes from Ob/WSD fetuses despite no additional lipid-induced stress. Switching obese mothers to a healthy diet prior to pregnancy did not improve fetal muscle mitochondrial function. Lastly, while maternal WSD alone led only to intermediary changes in fetal muscle metabolism, it was sufficient to increase oxidative damage and cellular stress. Our findings suggest that maternal obesity or WSD, alone or in combination, leads to programmed decreases in oxidative metabolism in offspring muscle. These alterations may have important implications for future health.
Collapse
Affiliation(s)
- Carrie E McCurdy
- Department of Human Physiology, University of Oregon, Eugene, Oregon, USA.,Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Simon Schenk
- Department of Orthopaedic Surgery, University of California, San Diego, La Jolla, California, USA
| | - Byron Hetrick
- Department of Human Physiology, University of Oregon, Eugene, Oregon, USA
| | - Julie Houck
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Brian G Drew
- David Geffen School of Medicine, Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, University of California, Los Angeles, Los Angeles, California, USA.,Diabetes and Dyslipidaemia Laboratory, Baker IDI Heart and Diabetes Institute, Prahran, Victoria, Australia
| | - Spencer Kaye
- Departments of Health and Exercise Science and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Melanie Lashbrook
- Departments of Health and Exercise Science and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Bryan C Bergman
- Division of Endocrinology, Metabolism, and Diabetes, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Diana L Takahashi
- Division of Diabetes, Obesity and Metabolism, Oregon National Primate Research Center, Beaverton, Oregon, USA
| | - Tyler A Dean
- Division of Diabetes, Obesity and Metabolism, Oregon National Primate Research Center, Beaverton, Oregon, USA
| | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Ilya Gertsman
- Department of Pediatrics, University of California, San Diego, La Jolla, California, USA
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Andrew Philp
- School of Sport Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, B15 2TT, United Kingdom
| | - Andrea L Hevener
- David Geffen School of Medicine, Department of Medicine, Division of Endocrinology, Diabetes and Hypertension, University of California, Los Angeles, Los Angeles, California, USA
| | - Adam J Chicco
- Departments of Health and Exercise Science and Biomedical Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Kjersti M Aagaard
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Baylor College of Medicine and Texas Children's Hospital, Houston, Texas, USA
| | - Kevin L Grove
- Division of Diabetes, Obesity and Metabolism, Oregon National Primate Research Center, Beaverton, Oregon, USA.,Novo Nordisk Research Center, Seattle, Washington, USA
| | - Jacob E Friedman
- Department of Pediatrics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
41
|
Chu DM, Meyer KM, Prince AL, Aagaard KM. Impact of maternal nutrition in pregnancy and lactation on offspring gut microbial composition and function. Gut Microbes 2016; 7:459-470. [PMID: 27686144 PMCID: PMC5103658 DOI: 10.1080/19490976.2016.1241357] [Citation(s) in RCA: 130] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Evidence supporting the Developmental Origins of Health and Disease Hypothesis indicates that maternal nutrition in pregnancy has a significant impact on offspring disease risk later in life, likely by modulating developmental processes in utero. Gut microbiota have recently been explored as a potential mediating factor, as dietary components strongly influence microbiota abundance, function and its impact on host physiology. A growing body of evidence has additionally indicated that the intrauterine environment is not sterile as once presumed, indicating that maternal-fetal transmission of microbiota may occur during pregnancy. In this article, we will review the body of literature that supports this emerging hypothesis, as well as highlight the work in relevant animal models demonstrating associations with maternal gestational nutrition and the offspring gut microbiome that may influence offspring physiology and susceptibility to disease.
Collapse
Affiliation(s)
- Derrick M. Chu
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX,Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX,Medical Scientist Training Program, Baylor College of Medicine, Houston, TX
| | - Kristen M. Meyer
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX,Medical Scientist Training Program, Baylor College of Medicine, Houston, TX,Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX
| | - Amanda L. Prince
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX
| | - Kjersti M. Aagaard
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX,Interdepartmental Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX,Medical Scientist Training Program, Baylor College of Medicine, Houston, TX,Department of Molecular & Human Genetics, Baylor College of Medicine, Houston, TX,Department of Molecular & Cell Biology, Baylor College of Medicine, Houston, TX,Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX,CONTACT Kjersti M. Aagaard, MD PhD FACOG Associate Professor, Baylor College of Medicine, Division of Maternal-Fetal Medicine, One Baylor Plaza, Jones 314, Houston, TX, 77030
| |
Collapse
|
42
|
Gonzalez-Rodriguez P, Cantu J, O’Neil D, Seferovic MD, Goodspeed DM, Suter MA, Aagaard KM. Alterations in expression of imprinted genes from the H19/IGF2 loci in a multigenerational model of intrauterine growth restriction (IUGR). Am J Obstet Gynecol 2016; 214:625.e1-625.e11. [PMID: 26880735 DOI: 10.1016/j.ajog.2016.01.194] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2015] [Revised: 01/16/2016] [Accepted: 01/20/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND The H19/IGF2 imprinted loci have attracted recent attention because of their role in cellular differentiation and proliferation, heritable gene regulation, and in utero or early postnatal growth and development. Expression from the imprinted H19/IGF2 locus involves a complex interplay of 3 means of epigenetic regulation: proper establishment of DNA methylation, promoter occupancy of CTCF, and expression of microRNA-675. We have demonstrated previously in a multigenerational rat model of intrauterine growth restriction the epigenetic heritability of adult metabolic syndrome in a F2 generation. We have further demonstrated abrogation of the F2 adult metabolic syndrome phenotype with essential nutrient supplementation of intermediates along the 1-carbon pathway and shown that alterations in the metabolome precede the adult onset of metabolic syndrome. The upstream molecular and epigenomic mediators underlying these observations, however, have yet to be elucidated fully. OBJECTIVE In the current study, we sought to characterize the impact of the intrauterine growth-restricted lineage and essential nutrient supplementation on both levels and molecular mediators of H19 and IGF2 gene expression in the F2 generation. STUDY DESIGN F2 intrauterine growth-restricted and sham lineages were obtained by exposing P1 (grandmaternal) pregnant dams to bilateral uterine artery ligation or sham surgery at gestational day 19.5. F1 pups were allocated to the essential nutrient supplemented or control diet at postnatal day 21, and bred at 6-7 weeks of age. Hepatic tissues from the resultant F2 offspring at birth and at weaning (day 21) were obtained. Bisulfite modification and sequencing was employed for methylation analysis. H19 and IGF2 expression was measured by quantitative polymerase chain reaction. Promoter occupancy was quantified by the use of chromatin immunoprecipitation, or ChIP, against CTCF insulator proteins. RESULTS Growth-restricted F2 on control diet demonstrated significant down-regulation in H19 expression compared with sham lineage (0.7831 vs 1.287; P < .05); however, essential nutrient supplementation diet abrogates this difference (4.995 vs 5.100; P > .05). Conversely, Igf2 was up-regulated by essential nutrient supplemented diet on the sham lineage (2.0 fold, P = .01), an effect that was not observed in the growth restricted offspring. A significant differential methylation was observed in the promoter region of region H19 among the intrauterine growth-restricted lineage (18% vs 25%; P < .05) on a control diet, whereas the essential nutrient supplemented diet was alternately associated with hypermethylation in both lineages (sham: 50%; intrauterine growth restriction: 84%, P < .05). Consistent with essential nutrient supplementation impacting the epigenome, a decrease of CTCF promoter occupancy was observed in CTCF4 of the growth restricted lineage (2.45% vs 0.56%; P < .05) on the control diet, an effect that was repressed with essential nutrient supplementation. CONCLUSION Heritable growth restriction is associated with changes in H19 gene expression; these changes are reversible with diet supplementation to favorably impact adult metabolic syndrome.
Collapse
|
43
|
Bruce KD, Szczepankiewicz D, Sihota KK, Ravindraanandan M, Thomas H, Lillycrop KA, Burdge GC, Hanson MA, Byrne CD, Cagampang FR. Altered cellular redox status, sirtuin abundance and clock gene expression in a mouse model of developmentally primed NASH. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1861:584-93. [PMID: 27040510 PMCID: PMC4874946 DOI: 10.1016/j.bbalip.2016.03.026] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 03/07/2016] [Accepted: 03/25/2016] [Indexed: 02/06/2023]
Abstract
Background We have previously shown that high fat (HF) feeding during pregnancy primes the development of non-alcoholic steatohepatits (NASH) in the adult offspring. However, the underlying mechanisms are unclear. Aims Since the endogenous molecular clock can regulate hepatic lipid metabolism, we investigated whether exposure to a HF diet during development could alter hepatic clock gene expression and contribute to NASH onset in later life. Methods Female mice were fed either a control (C, 7% kcal fat) or HF (45% kcal fat) diet. Offspring were fed either a C or HF diet resulting in four offspring groups: C/C, C/HF, HF/C and HF/HF. NAFLD progression, cellular redox status, sirtuin expression (Sirt1, Sirt3), and the expression of core clock genes (Clock, Bmal1, Per2, Cry2) and clock-controlled genes involved in lipid metabolism (Rev-Erbα, Rev-Erbβ, RORα, and Srebp1c) were measured in offspring livers. Results Offspring fed a HF diet developed NAFLD. However HF fed offspring of mothers fed a HF diet developed NASH, coupled with significantly reduced NAD+/NADH (p < 0.05, HF/HF vs C/C), Sirt1 (p < 0.001, HF/HF vs C/C), Sirt3 (p < 0.01, HF/HF vs C/C), perturbed clock gene expression, and elevated expression of genes involved lipid metabolism, such as Srebp1c (p < 0.05, C/HF and HF/HF vs C/C). Conclusion Our results suggest that exposure to excess dietary fat during early and post-natal life increases the susceptibility to develop NASH in adulthood, involving altered cellular redox status, reduced sirtuin abundance, and desynchronized clock gene expression. Offspring of mothers fed a high fat diet show severe fatty liver in later life. HF feeding is associated with altered cellular redox status and reduced sirtuin gene expression. HF feeding desynchronises the expression of core clock genes and lipogenic transcription factors. Exposure to a HF diet during development causes changes in liver metabolism that precede severe fatty liver disease.
Collapse
Affiliation(s)
- Kimberley D Bruce
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK; University of Colorado Anschutz Medical Campus, Endocrinology, Metabolism and Diabetes, Aurora, USA.
| | - Dawid Szczepankiewicz
- Poznań University of Life Sciences, Department of Animal Physiology and Biochemistry, Poznań, Poland
| | - Kiran K Sihota
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Manoj Ravindraanandan
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Hugh Thomas
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Karen A Lillycrop
- Centre for Biological Sciences, Institute of Developmental Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton, UK
| | - Graham C Burdge
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Mark A Hanson
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Christopher D Byrne
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Felino R Cagampang
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| |
Collapse
|
44
|
Vansant G. Effect of Maternal and Paternal Nutrition on DNA Methylation in the Offspring: A Systematic Review of Human and Animal Studies. ACTA ACUST UNITED AC 2016. [DOI: 10.15406/aowmc.2016.04.00093] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
45
|
Nathanielsz PW, Yan J, Green R, Nijland M, Miller JW, Wu G, McDonald TJ, Caudill MA. Maternal obesity disrupts the methionine cycle in baboon pregnancy. Physiol Rep 2015; 3:e12564. [PMID: 26537341 PMCID: PMC4673623 DOI: 10.14814/phy2.12564] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Revised: 09/04/2015] [Accepted: 09/11/2015] [Indexed: 02/06/2023] Open
Abstract
Maternal intake of dietary methyl-micronutrients (e.g. folate, choline, betaine and vitamin B-12) during pregnancy is essential for normal maternal and fetal methionine metabolism, and is critical for important metabolic processes including those involved in developmental programming. Maternal obesity and nutrient excess during pregnancy influence developmental programming potentially predisposing adult offspring to a variety of chronic health problems. In the present study, we hypothesized that maternal obesity would dysregulate the maternal and fetal methionine cycle. To test this hypothesis, we developed a nulliparous baboon obesity model fed a high fat, high energy diet (HF-HED) prior to and during gestation, and examined methionine cycle biomarkers (e.g., circulating concentrations of homocysteine, methionine, choline, betaine, key amino acids, folate, and vitamin B-12). Animals were group housed allowing full physical activity and social interaction. Maternal prepregnancy percent body fat was 5% in controls and 19% in HF-HED mothers, while fetal weight was 16% lower in offspring of HF-HED mothers at term. Maternal and fetal homocysteine were higher, while maternal and fetal vitamin B-12 and betaine were lower in the HF-HED group. Elevations in circulating maternal folate were evident in the HF-HED group indicating impaired folate metabolism (methyl-trap) as a consequence of maternal vitamin B-12 depletion. Finally, fetal methionine, glycine, serine, and taurine were lower in the HF-HED fetuses. These data show that maternal obesity disturbs the methionine cycle in primate pregnancy, providing a mechanism for the epigenetic changes observed among obese pregnant women and suggesting diagnostic and therapeutic opportunities in human pregnancies complicated by obesity.
Collapse
Affiliation(s)
- Peter W Nathanielsz
- Center for Pregnancy and Newborn Research, Department OB/GYN, University of Texas Health Science Center San Antonio, San Antonio, Texas Texas Center for Pregnancy and Life Course Health, Southwest National Primate Research Institute Texas Biomedical Research Institute, San Antonio, Texas
| | - Jian Yan
- Division of Nutritional Sciences, Savage Hall, Ithaca, New York
| | - Ralph Green
- Department of Medical Pathology and Laboratory Medicine, University of California Davis Medical Center, Sacramento, California
| | - Mark Nijland
- Center for Pregnancy and Newborn Research, Department OB/GYN, University of Texas Health Science Center San Antonio, San Antonio, Texas
| | - Joshua W Miller
- Department of Medical Pathology and Laboratory Medicine, University of California Davis Medical Center, Sacramento, California Department of Nutritional Sciences, Rutgers University, New Brunswick, New Jersey
| | - Guoyao Wu
- Department of Animal Science and Center for Animal Biotechnology and Genomics, and Faculty of Nutrition, Texas A&M University, College Station, Texas
| | - Thomas J McDonald
- Center for Pregnancy and Newborn Research, Department OB/GYN, University of Texas Health Science Center San Antonio, San Antonio, Texas
| | - Marie A Caudill
- Division of Nutritional Sciences, Savage Hall, Ithaca, New York
| |
Collapse
|
46
|
Dearden L, Ozanne SE. Early life origins of metabolic disease: Developmental programming of hypothalamic pathways controlling energy homeostasis. Front Neuroendocrinol 2015; 39:3-16. [PMID: 26296796 DOI: 10.1016/j.yfrne.2015.08.001] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Revised: 08/07/2015] [Accepted: 08/17/2015] [Indexed: 12/30/2022]
Abstract
A wealth of animal and human studies demonstrate that perinatal exposure to adverse metabolic conditions - be it maternal obesity, diabetes or under-nutrition - results in predisposition of offspring to develop obesity later in life. This mechanism is a contributing factor to the exponential rise in obesity rates. Increased weight gain in offspring exposed to maternal obesity is usually associated with hyperphagia, implicating altered central regulation of energy homeostasis as an underlying cause. Perinatal development of the hypothalamus (a brain region key to metabolic regulation) is plastic and sensitive to metabolic signals during this critical time window. Recent research in non-human primate and rodent models has demonstrated that exposure to adverse maternal environments impairs the development of hypothalamic structure and consequently function, potentially underpinning metabolic phenotypes in later life. This review summarizes our current knowledge of how adverse perinatal environments program hypothalamic development and explores the mechanisms that could mediate these effects.
Collapse
Affiliation(s)
- Laura Dearden
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Cambridge CB2 0QQ, United Kingdom
| | - Susan E Ozanne
- University of Cambridge Metabolic Research Laboratories, Wellcome Trust-MRC Institute of Metabolic Science, Box 289, Addenbrooke's Hospital, Cambridge CB2 0QQ, United Kingdom.
| |
Collapse
|
47
|
Abstract
Rapidly growing evidences link maternal lifestyle and prenatal factors with serious health consequences and diseases later in life. Extensive epidemiological studies have identified a number of factors such as diet, stress, gestational diabetes, exposure to tobacco and alcohol during gestation as influencing normal fetal development. In light of recent discoveries, epigenetic mechanisms such as alteration of DNA methylation, chromatin modifications and modulation of gene expression during gestation are believed to possibly account for various types of plasticity such as neural tube defects, autism spectrum disorder, congenital heart defects, oral clefts, allergies and cancer. The purpose of this article is to review a number of published studies to fill the gap in our understanding of how maternal lifestyle and intrauterine environment influence molecular modifications in the offspring, with an emphasis on epigenetic alterations. To support these associations, we highlighted laboratory studies of rodents and epidemiological studies of human based on sampling population cohorts.
Collapse
Affiliation(s)
- Subit Barua
- Structural Neurobiology Laboratory, Department of Developmental Biochemistry, New York State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY 10314, USA
| | | |
Collapse
|
48
|
Neri C, Edlow AG. Effects of Maternal Obesity on Fetal Programming: Molecular Approaches. Cold Spring Harb Perspect Med 2015; 6:a026591. [PMID: 26337113 DOI: 10.1101/cshperspect.a026591] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Maternal obesity has become a worldwide epidemic. Obesity and a high-fat diet have been shown to have deleterious effects on fetal programming, predisposing offspring to adverse cardiometabolic and neurodevelopmental outcomes. Although large epidemiological studies have shown an association between maternal obesity and adverse outcomes for offspring, the underlying mechanisms remain unclear. Molecular approaches have played a key role in elucidating the mechanistic underpinnings of fetal malprogramming in the setting of maternal obesity. These approaches include, among others, characterization of epigenetic modifications, microRNA expression, the gut microbiome, the transcriptome, and evaluation of specific mRNA expression via quantitative reverse transcription polmerase chain reaction (RT-qPCR) in fetuses and offspring of obese females. This work will review the data from animal models and human fluids/cells regarding the effects of maternal obesity on fetal and offspring neurodevelopment and cardiometabolic outcomes, with a particular focus on molecular approaches.
Collapse
Affiliation(s)
- Caterina Neri
- Department of Obstetrics and Gynecology, Università Cattolica del Sacro Cuore, Rome 00100, Italy
| | - Andrea G Edlow
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tufts Medical Center, Boston, Massachusetts 02111 Mother Infant Research Institute, Tufts Medical Center, Boston, Massachusetts 02111
| |
Collapse
|
49
|
van Dijk SJ, Tellam RL, Morrison JL, Muhlhausler BS, Molloy PL. Recent developments on the role of epigenetics in obesity and metabolic disease. Clin Epigenetics 2015; 7:66. [PMID: 27408648 PMCID: PMC4940755 DOI: 10.1186/s13148-015-0101-5] [Citation(s) in RCA: 135] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 06/29/2015] [Indexed: 12/20/2022] Open
Abstract
The increased prevalence of obesity and related comorbidities is a major public health problem. While genetic factors undoubtedly play a role in determining individual susceptibility to weight gain and obesity, the identified genetic variants only explain part of the variation. This has led to growing interest in understanding the potential role of epigenetics as a mediator of gene-environment interactions underlying the development of obesity and its associated comorbidities. Initial evidence in support of a role of epigenetics in obesity and type 2 diabetes mellitus (T2DM) was mainly provided by animal studies, which reported epigenetic changes in key metabolically important tissues following high-fat feeding and epigenetic differences between lean and obese animals and by human studies which showed epigenetic changes in obesity and T2DM candidate genes in obese/diabetic individuals. More recently, advances in epigenetic methodologies and the reduced cost of epigenome-wide association studies (EWAS) have led to a rapid expansion of studies in human populations. These studies have also reported epigenetic differences between obese/T2DM adults and healthy controls and epigenetic changes in association with nutritional, weight loss, and exercise interventions. There is also increasing evidence from both human and animal studies that the relationship between perinatal nutritional exposures and later risk of obesity and T2DM may be mediated by epigenetic changes in the offspring. The aim of this review is to summarize the most recent developments in this rapidly moving field, with a particular focus on human EWAS and studies investigating the impact of nutritional and lifestyle factors (both pre- and postnatal) on the epigenome and their relationship to metabolic health outcomes. The difficulties in distinguishing consequence from causality in these studies and the critical role of animal models for testing causal relationships and providing insight into underlying mechanisms are also addressed. In summary, the area of epigenetics and metabolic health has seen rapid developments in a short space of time. While the outcomes to date are promising, studies are ongoing, and the next decade promises to be a time of productive research into the complex interactions between the genome, epigenome, and environment as they relate to metabolic disease.
Collapse
Affiliation(s)
- Susan J van Dijk
- CSIRO Food and Nutrition Flagship, PO Box 52, North Ryde, NSW 1670 Australia
| | - Ross L Tellam
- CSIRO Agriculture Flagship, 306 Carmody Road, St Lucia, QLD 4067 Australia
| | - Janna L Morrison
- Early Origins of Adult Health Research Group, School of Pharmacy and Medical Sciences, Sansom Institute for Health Research, University of South Australia, GPO Box 2471, Adelaide, SA 5001 Australia
| | - Beverly S Muhlhausler
- FOODplus Research Centre, Waite Campus, The University of Adelaide, PMB 1, Glen Osmond, SA 5064 Australia.,Women's and Children's Health Research Institute, 72 King William Road, North Adelaide, SA 5006 Australia
| | - Peter L Molloy
- CSIRO Food and Nutrition Flagship, PO Box 52, North Ryde, NSW 1670 Australia
| |
Collapse
|
50
|
Oh KY, Roberts VHJ, Schabel MC, Grove KL, Woods M, Frias AE. Gadolinium Chelate Contrast Material in Pregnancy: Fetal Biodistribution in the Nonhuman Primate. Radiology 2015; 276:110-8. [PMID: 25763829 PMCID: PMC4485748 DOI: 10.1148/radiol.15141488] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
PURPOSE To determine the extent to which gadolinium chelate is found in nonhuman primate fetal tissues and amniotic fluid at 19-45 hours after intravenous injection of a weight-appropriate maternal dose of the contrast agent gadoteridol. MATERIALS AND METHODS Gravid Japanese macaques (n = 14) were maintained as approved by the institutional animal care and utilization committee. In the 3rd trimester of pregnancy, the macaques were injected with gadoteridol (0.1 mmol per kilogram of maternal weight). Fetuses were delivered by means of cesarean section within 24 hours of maternal injection (range, 19-21 hours; n = 11) or 45 hours after injection (n = 3). Gadolinium chelate levels in the placenta, fetal tissues, and amniotic fluid were obtained by using inductively coupled plasma mass spectrometry. The Wilcoxon rank sum test was used for quantitative comparisons. RESULTS Gadoteridol was present in the fetoplacental circulation at much lower quantities than in the mother. At both time points, the distribution of gadolinium chelate in the fetus was comparable to that expected in an adult. The highest concentration of the injected dose (ID) was found in the fetal kidney (0.0161% ID per gram in the 19-21-hour group). The majority of the in utero gadolinium chelate was found in the amniotic fluid and the placenta (mean, 0.1361% ID per organ ± 0.076 [standard deviation] and 0.0939% ID per organ ± 0.0494, respectively). Data acquired 45 hours after injection showed a significant decrease in the gadolinium chelate concentration in amniotic fluid compared with that in the 19-21-hour group (from 0.0017% to 0.0007% ID per gram; P = .01). CONCLUSION Amounts of gadolinium chelate in the fetal tissues and amniotic fluid were minimal compared with the maternal ID. This may impact future clinical studies on the safety of gadolinium contrast agent use in pregnancy.
Collapse
Affiliation(s)
- Karen Y. Oh
- From the Department of Radiology (K.Y.O.), Advanced Imaging Research Center (M.C.S., M.W.), and Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine (A.E.F.), Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, MC L340, Portland, OR 97239; Division of Diabetes, Obesity and Metabolism, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Ore (V.H.J.R., K.L.G., A.E.F.); and Department of Chemistry, Portland State University, Portland, Ore (M.W.)
| | - Victoria H. J. Roberts
- From the Department of Radiology (K.Y.O.), Advanced Imaging Research Center (M.C.S., M.W.), and Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine (A.E.F.), Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, MC L340, Portland, OR 97239; Division of Diabetes, Obesity and Metabolism, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Ore (V.H.J.R., K.L.G., A.E.F.); and Department of Chemistry, Portland State University, Portland, Ore (M.W.)
| | - Matthias C. Schabel
- From the Department of Radiology (K.Y.O.), Advanced Imaging Research Center (M.C.S., M.W.), and Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine (A.E.F.), Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, MC L340, Portland, OR 97239; Division of Diabetes, Obesity and Metabolism, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Ore (V.H.J.R., K.L.G., A.E.F.); and Department of Chemistry, Portland State University, Portland, Ore (M.W.)
| | - Kevin L. Grove
- From the Department of Radiology (K.Y.O.), Advanced Imaging Research Center (M.C.S., M.W.), and Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine (A.E.F.), Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, MC L340, Portland, OR 97239; Division of Diabetes, Obesity and Metabolism, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Ore (V.H.J.R., K.L.G., A.E.F.); and Department of Chemistry, Portland State University, Portland, Ore (M.W.)
| | - Mark Woods
- From the Department of Radiology (K.Y.O.), Advanced Imaging Research Center (M.C.S., M.W.), and Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine (A.E.F.), Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, MC L340, Portland, OR 97239; Division of Diabetes, Obesity and Metabolism, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Ore (V.H.J.R., K.L.G., A.E.F.); and Department of Chemistry, Portland State University, Portland, Ore (M.W.)
| | - Antonio E. Frias
- From the Department of Radiology (K.Y.O.), Advanced Imaging Research Center (M.C.S., M.W.), and Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine (A.E.F.), Oregon Health and Science University, 3181 SW Sam Jackson Park Rd, MC L340, Portland, OR 97239; Division of Diabetes, Obesity and Metabolism, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, Ore (V.H.J.R., K.L.G., A.E.F.); and Department of Chemistry, Portland State University, Portland, Ore (M.W.)
| |
Collapse
|