1
|
Wei G, Shen FJ, Liu JL, Zhao JH, Yang FY, Feng RQ, Lu J, Zhang CY, Wang FW, Chen BD, Ding X, Yang JK. Uncoupling protein 1 deficiency leads to transcriptomic differences in livers of pregnancy female mice and aggravates hepatic steatosis. Arch Biochem Biophys 2025; 768:110395. [PMID: 40122441 DOI: 10.1016/j.abb.2025.110395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/24/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Pregnancy requires the coordination of metabolically active organs to support maternal nutrition and fetal growth. However, the metabolic cross-talk between adipose tissue and liver in females during pregnancy is still less clear. In this study, we evaluated the metabolic adaptations and phenotypes of liver in response to pregnancy-associated metabolic stress, particularly in the context of genetic ablation of Uncoupling protein 1 (Ucp1)-mediated catabolic circuit. Our results revealed that Ucp1 deficiency (UCP1 knockout, KO) mice during late pregnancy exhibited significantly deteriorated metabolic phenotypes, including hepatic steatosis and whole-body glucose and lipid homeostasis, as compared to Ucp1 deficiency or normal pregnancy mice. However, non-pregnant Ucp1 deficiency mice displayed nearly normal metabolic phenotypes and structure alterations similar to those of littermate controls. Moreover, transcriptomic analyses by RNA sequencing (RNA-seq) clearly revealed that Ucp1 deficiency led to a significant liver metabolic remodeling of differentially express genes (DEGs) before and especially during pregnancy. Consistently, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses demonstrated the potential altered functions and signaling pathways, including metabolic dysfunctions in ribosome, oxidative phosphorylation, etc. Importantly, as derived from trend analyses of DEGs, our results further revealed the distinct expression pattern of each subcluster, which coincided with potential biological functions and relevant signaling pathways. The findings in the present study might provide valuable insights into the molecular mechanism of metabolic dysfunction-associated fatty liver disease (MAFLD) during pregnancy. Additionally, our data may provide a novel animal model of MAFLD, thus facilitating its potential therapies. NEW & NOTEWORTHY: Genetic ablation of Ucp1 during pregnancy increases hepatic steatosis and deteriorated whole-body glucose and lipid homeostasis. Moreover, changes in hepatic gene expression are closely associated with metabolic dysfunctions in ribosome and oxidative phosphorylation. This work highlights the therapeutic potential of targeting UCP1- mediated catabolic circuit between adipose and liver during pregnancy, and the utility of RNA-seq analysis to reveal valuable information for the distinct expression pattern of each subcluster that contribute to pregnancy-dependent MASLD progression.
Collapse
Affiliation(s)
- Gang Wei
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China; Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang, 550025, China.
| | - Feng-Jie Shen
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Jun-Li Liu
- Neurology in the First Affiliated Hospital of XinXiang Medical University, Henan Institute of Neurology, Henan Joint International Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Weihui, 453100, Henan Province, China.
| | - Jian-Hua Zhao
- Neurology in the First Affiliated Hospital of XinXiang Medical University, Henan Institute of Neurology, Henan Joint International Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Weihui, 453100, Henan Province, China.
| | - Fang-Yuan Yang
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Ruo-Qi Feng
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Jing Lu
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Chen-Yang Zhang
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Feng-Wei Wang
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| | - Bei-Dong Chen
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100005, China.
| | - Xin Ding
- Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing Maternal and Child Health Care Hospital, Beijing, 100020, China.
| | - Jin-Kui Yang
- Beijing Key Laboratory of Diabetes Research and Care, Department of Endocrinology, Beijing Diabetes Institute, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China.
| |
Collapse
|
2
|
Wilsterman K, Bautista AI, Butler CE, Juergens MY, Larson AM. Evolution of Litter Size: Proximate and Ultimate Mechanisms. Integr Comp Biol 2024; 64:1643-1660. [PMID: 38802126 PMCID: PMC11659681 DOI: 10.1093/icb/icae052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
Relative reproductive success and failure are the ultimate determinants of Darwinian fitness. As such, reproductive traits and variations therein have an immediate and considerable impact on the evolutionary trajectory of lineages. Historically, significant attention has been paid to the ecological and evolutionary processes (ultimate factors) that shape the diversity and canalization of reproductive traits within groups to better our understanding of organismal diversity and population or species resilience. In contrast, the physiological systems that mediate variation within and among species (i.e., the proximate factors) in reproductive traits remain a significant black box. To date, there is comparatively little information about how proximate mechanisms constrain or promote evolutionary potential in reproductive traits. In this mini-review, we focus on litter size in Eutherian mammals as a trait with relatively well-defined diversity (litter sizes are well-described both within and across species) and for which some genetic determinants have been identified. We discuss both the ultimate and potential proximate determinants of litter size with special attention to the breadth of physiological traits that may act as "toggle" switches for evolution of litter size. We close with a brief discussion of the role that physiological plasticity may play in the evolution of litter size and lay out several forward-looking areas for future research.
Collapse
Affiliation(s)
- Kathryn Wilsterman
- Department of Biology, Colorado State University, Fort Collins, CO, 80521, USA
| | | | - Chloe E Butler
- Department of Biology, Colorado State University, Fort Collins, CO, 80521, USA
| | - Makenna Y Juergens
- Department of Biology, Colorado State University, Fort Collins, CO, 80521, USA
| | - Ashley M Larson
- Department of Biology, Colorado State University, Fort Collins, CO, 80521, USA
| |
Collapse
|
3
|
Pastor FM, de Melo Ocarino N, Silva JF, Reis AMS, Serakides R. Bone development in fetuses with intrauterine growth restriction caused by maternal endocrine-metabolic dysfunctions. Bone 2024; 186:117169. [PMID: 38880170 DOI: 10.1016/j.bone.2024.117169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/21/2024] [Accepted: 06/13/2024] [Indexed: 06/18/2024]
Abstract
Intrauterine growth restriction (IUGR) affects a large proportion of infants, particularly in underdeveloped countries. Among the main causes of IUGR, maternal endocrine-metabolic dysfunction is highlighted, either due to its high incidence or due to the severity of the immediate and mediated changes that these dysfunctions cause in the fetus and the mother. Although the effects of endocrine and metabolic disorders have been widely researched, there are still no reviews that bring together and summarize the effects of these conditions on bone development in cases of IUGR. Therefore, the present literature review was conducted with the aim of discussing bone changes observed in fetuses with IUGR caused by maternal endocrine-metabolic dysfunction. The main endocrine dysfunctions that occur with IUGR include maternal hyperthyroidism, hypothyroidism, and hypoparathyroidism. Diabetes mellitus, hypertensive disorders, and obesity are the most important maternal metabolic dysfunctions that compromise fetal growth. The bone changes reported in the fetus are, for the most part, due to damage to cell proliferation and differentiation, as well as failures in the synthesis and mineralization of the extracellular matrix, which results in shortening and fragility of the bones. Some maternal dysfunctions, such as hyperthyroidism, have been widely studied, whereas conditions such as hypoparathyroidism and gestational hypertensive disorders require further study regarding the mechanisms underlying the development of bone changes. Similarly, there is a gap in the literature regarding changes related to intramembranous ossification, as most published articles only describe changes in endochondral bone formation associated with IUGR. Furthermore, there is a need for more research aimed at elucidating the late postnatal changes that occur in the skeletons of individuals affected by IUGR and their possible relationships with adult diseases, such as osteoarthritis and osteoporosis.
Collapse
Affiliation(s)
- Felipe Martins Pastor
- Departamento de Cínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Natália de Melo Ocarino
- Departamento de Cínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Juneo Freitas Silva
- Centro de Microscopia Eletrônica, Departamento de Ciências Biológicas, Universidade Estadual de Santa Cruz, Rodovia Jorge Amado, Km 16, 45662-900 Ilhéus, Bahia, Brazil
| | - Amanda Maria Sena Reis
- Departamento de Patologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Rogéria Serakides
- Departamento de Cínica e Cirurgia Veterinárias, Escola de Veterinária, Universidade Federal de Minas Gerais, Avenida Antônio Carlos 6627, 31270-901 Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
4
|
Sousa D, Magalhães C, Matafome P, Pereira S. Adipose tissue-liver cross-talk: a route to hepatic dysfunction in pregnant women with obesity. Biosci Rep 2024; 44:BSR20231679. [PMID: 39083072 PMCID: PMC11327218 DOI: 10.1042/bsr20231679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 05/24/2024] [Accepted: 07/31/2024] [Indexed: 08/15/2024] Open
Abstract
Obesity during pregnancy has been escalating, becoming a huge problem that poses consequences not only for the health of the offspring but also for the maternal well-being. Women's adipose and hepatic tissue metabolism undergoes significant changes during the gestational period. During pregnancy, obesity is a primary instigator of steatosis, increasing the risk of non-alcholic fatty liver disease (NAFLD), now recognized under the updated nomenclature metabolic dysfunction-associated steatotic liver disease (MASLD). Pregnant women with obesity present higher levels of free fatty acids and glucose, reduction in insulin sensitivity, and adipose tissue endocrine dysregulation. Furthermore, obesity-induced modifications in clock genes and lipid-associated gene expression within adipose tissue disrupt crucial metabolic adaptations, potentially culminating in adipose tissue dysfunction. Thus, the liver experiences increased exposure to free fatty acids through the portal vein. Higher uptake of free fatty acids into the liver disrupts hepatic lipid oxidation while enhances lipogenesis, thereby predisposing to ectopic fat deposition within the liver. This review focuses on the obesity-induced changes during pregnancy in both liver and adipose tissue metabolism, elucidating how the metabolic crosstalk between these two organs can be dysregulated in pregnant women living with obesity.
Collapse
Affiliation(s)
- Diana Sousa
- Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
- Institute of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal
- Ph.D. Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - Carina C. Magalhães
- Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
| | - Paulo Matafome
- Coimbra Institute for Clinical and Biomedical Research (iCBR) and Institute of Physiology, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3004-561 Coimbra, Portugal
- Polytechnic University of Coimbra, Coimbra Health School, Rua 5 de Outubro—S. Martinho do Bispo, 3046-854 Coimbra, Portugal
| | - Susana P. Pereira
- CNC-UC—Center for Neuroscience and Cell Biology, University of Coimbra,3004-504 Coimbra, Portugal
- CIBB—Centre for Innovative Biomedicine and Biotechnology, University of Coimbra; 3004-517 Coimbra, Portugal
- Laboratory of Metabolism and Exercise (LaMetEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory of for Integrative and Translational Research in Population Health (ITR), Faculty of Sports, University of Porto, 4200-450 Porto, Portugal
| |
Collapse
|
5
|
He S, Guo Z, Zhou M, Wang H, Zhang Z, Shi M, Li X, Yang X, He L. Spatial-temporal proliferation of hepatocytes during pregnancy revealed by genetic lineage tracing. Cell Stem Cell 2023; 30:1549-1558.e5. [PMID: 37794588 DOI: 10.1016/j.stem.2023.09.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 08/04/2023] [Accepted: 09/05/2023] [Indexed: 10/06/2023]
Abstract
The maternal liver undergoes dramatic enlargement to adapt to the increased metabolic demands during pregnancy. However, the cellular sources for liver growth during pregnancy remain largely elusive. Here, we employed a proliferation recording system, ProTracer, to examine the spatial-temporal proliferation of hepatocytes during pregnancy. We discovered that during early to late pregnancy, hepatocyte proliferation initiated from zone 1, to zone 2, and lastly to zone 3, with the majority of new hepatocytes being generated in zone 2. Additionally, using single-cell RNA sequencing, we observed that Ccnd1 was highly enriched in zone 2 hepatocytes. We further applied dual-recombinase-mediated genetic lineage tracing to reveal that Ccnd1+ hepatocytes expanded preferentially during pregnancy. Moreover, we demonstrated that estrogen induces liver enlargement during pregnancy, which was abolished in Ccnd1 knockout mice. Our work revealed a unique spatial-temporal hepatocyte proliferation pattern during pregnancy, with Ccnd1+ hepatocytes in zone 2 serving as the major cellular source for hepatic enlargement.
Collapse
Affiliation(s)
- Shun He
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310030, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou 310030, Zhejiang, China; Westlake Institute for Advanced Study, Hangzhou 310030, Zhejiang, China
| | - Zhihou Guo
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310030, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou 310030, Zhejiang, China; Westlake Institute for Advanced Study, Hangzhou 310030, Zhejiang, China
| | - Mingshan Zhou
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310030, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou 310030, Zhejiang, China; Westlake Institute for Advanced Study, Hangzhou 310030, Zhejiang, China
| | - Haichang Wang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310030, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou 310030, Zhejiang, China; Westlake Institute for Advanced Study, Hangzhou 310030, Zhejiang, China
| | - Zhuonan Zhang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310030, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou 310030, Zhejiang, China; Westlake Institute for Advanced Study, Hangzhou 310030, Zhejiang, China
| | - Mengyang Shi
- State Key Laboratory of Cell Biology, Center for Excellence in Molecular Cell Science, University of Chinese Academy of Sciences, Chinese Academic of Sciences, Shanghai 200031, China
| | - Xufeng Li
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, Zhejiang, China
| | - Xueying Yang
- Key Laboratory of Systems Health Science of Zhejiang Province, School of Life Science, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, Zhejiang, China
| | - Lingjuan He
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310030, Zhejiang, China; School of Life Sciences, Westlake University, Hangzhou 310030, Zhejiang, China; Westlake Institute for Advanced Study, Hangzhou 310030, Zhejiang, China.
| |
Collapse
|
6
|
Grilo LF, Martins JD, Diniz MS, Tocantins C, Cavallaro CH, Baldeiras I, Cunha-Oliveira T, Ford S, Nathanielsz PW, Oliveira PJ, Pereira SP. Maternal hepatic adaptations during obese pregnancy encompass lobe-specific mitochondrial alterations and oxidative stress. Clin Sci (Lond) 2023; 137:1347-1372. [PMID: 37565250 DOI: 10.1042/cs20230048] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 08/02/2023] [Accepted: 08/10/2023] [Indexed: 08/12/2023]
Abstract
Maternal obesity (MO) is rising worldwide, affecting half of all gestations, constituting a possible risk-factor for some pregnancy-associated liver diseases (PALD) and hepatic diseases. PALD occur in approximately 3% of pregnancies and are characterized by maternal hepatic oxidative stress (OS) and mitochondrial dysfunction. Maternal hepatic disease increases maternal and fetal morbidity and mortality. Understanding the role of MO on liver function and pathophysiology could be crucial for better understanding the altered pathways leading to PALD and liver disease, possibly paving the way to prevention and adequate management of disease. We investigated specific hepatic metabolic alterations in mitochondria and oxidative stress during MO at late-gestation. Maternal hepatic tissue was collected at 90% gestation in Control and MO ewes (fed 150% of recommended nutrition starting 60 days before conception). Maternal hepatic redox state, mitochondrial respiratory chain (MRC), and OS markers were investigated. MO decreased MRC complex-II activity and its subunits SDHA and SDHB protein expression, increased complex-I and complex-IV activities despite reduced complex-IV subunit mtCO1 protein expression, and increased ATP synthase ATP5A subunit. Hepatic MO-metabolic remodeling was characterized by decreased adenine nucleotide translocator 1 and 2 (ANT-1/2) and voltage-dependent anion channel (VDAC) protein expression and protein kinase A (PKA) activity (P<0.01), and augmented NAD+/NADH ratio due to reduced NADH levels (P<0.01). MO showed an altered redox state with increased OS, increased lipid peroxidation (P<0.01), decreased GSH/GSSG ratio (P=0.005), increased superoxide dismutase (P=0.03) and decreased catalase (P=0.03) antioxidant enzymatic activities, lower catalase, glutathione peroxidase (GPX)-4 and glutathione reductase protein expression (P<0.05), and increased GPX-1 abundance (P=0.03). MO-related hepatic changes were more evident in the right lobe, corroborated by the integrative data analysis. Hepatic tissue from obese pregnant ewes showed alterations in the redox state, consistent with OS and MRC and metabolism remodeling. These are hallmarks of PALD and hepatic disease, supporting MO as a risk-factor and highlighting OS and mitochondrial dysfunction as mechanisms responsible for liver disease predisposition.
Collapse
Affiliation(s)
- Luís F Grilo
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC-Biotech, Biocant Park, Cantanhede, Portugal
- Ph.D. Programme in Experimental Biology and Biomedicine (PDBEB), Institute for Interdisciplinary Research (IIIUC), University of Coimbra, Coimbra, Portugal
| | - João D Martins
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC-Biotech, Biocant Park, Cantanhede, Portugal
| | - Mariana S Diniz
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC-Biotech, Biocant Park, Cantanhede, Portugal
| | - Carolina Tocantins
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC-Biotech, Biocant Park, Cantanhede, Portugal
| | - Chiara H Cavallaro
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC-Biotech, Biocant Park, Cantanhede, Portugal
| | - Inês Baldeiras
- Neurological Clinic, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Teresa Cunha-Oliveira
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC-Biotech, Biocant Park, Cantanhede, Portugal
| | - Stephen Ford
- Department of Animal Science, University of Wyoming, Laramie, WY, U.S.A
| | | | - Paulo J Oliveira
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC-Biotech, Biocant Park, Cantanhede, Portugal
| | - Susana P Pereira
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC-Biotech, Biocant Park, Cantanhede, Portugal
- Laboratory of Metabolism and Exercise (LametEx), Research Centre in Physical Activity, Health and Leisure (CIAFEL), Laboratory for Integrative and Translational Research in Population Health (ITR), Faculty of Sport, University of Porto, Porto, Portugal
| |
Collapse
|
7
|
Wang L, Zhu J, Xie P, Gong D. Pigeon during the Breeding Cycle: Behaviors, Composition and Formation of Crop Milk, and Physiological Adaptation. Life (Basel) 2023; 13:1866. [PMID: 37763270 PMCID: PMC10533064 DOI: 10.3390/life13091866] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/21/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Pigeon is an important economic poultry species in many countries. As an altricial bird, its growth and development are largely reliant on pigeon milk produced by the crop tissue in the first week. During the breeding cycle, pigeons undergo a series of behavioral changes. Pigeon milk is generally characterized by having high concentrations of proteins and lipids, and a complicated regulatory network is involved in the milk formation. Hormones, especially prolactin, could promote the proliferation of crop epidermal cells and nutrient accumulation. The expression of target genes associated with these important biological processes in the crop epidermis is affected by non-coding RNAs. Meanwhile, signaling pathways, such as target of rapamycin (TOR), Janus kinase/signal transducer and activator of transcription proteins (JAK/STAT), protein kinase B (Akt), etc., influence the production of crop milk by either enhancing protein synthesis in crop cells or inducing apoptosis of crop epidermal cells. In order to adapt to the different breeding periods, pigeons are physiologically changed in their intestinal morphology and function and liver metabolism. This paper reviews the behaviors and physiological adaptations of pigeon during the breeding cycle, the composition of pigeon crop milk, and the mechanism of its formation, which is important for a better understanding of the physiology of altricial birds and the development of artificial crop milk.
Collapse
Affiliation(s)
- Liuxiong Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (L.W.); (J.Z.)
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huaian 223300, China
| | - Jianguo Zhu
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (L.W.); (J.Z.)
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huaian 223300, China
| | - Peng Xie
- Jiangsu Collaborative Innovation Center of Regional Modern Agriculture & Environmental Protection, Huaiyin Normal University, Huaian 223300, China
- Jiangsu Key Laboratory for Eco-Agricultural Biotechnology around Hongze Lake, Huaiyin Normal University, Huaian 223300, China
| | - Daoqing Gong
- College of Animal Science and Technology, Yangzhou University, Yangzhou 225009, China; (L.W.); (J.Z.)
| |
Collapse
|
8
|
Yung HW, Zhao X, Glover L, Burrin C, Pang PC, Jones CJ, Gill C, Duhig K, Olovsson M, Chappell LC, Haslam SM, Dell A, Burton GJ, Charnock-Jones DS. Perturbation of placental protein glycosylation by endoplasmic reticulum stress promotes maladaptation of maternal hepatic glucose metabolism. iScience 2023; 26:105911. [PMID: 36660474 PMCID: PMC9843443 DOI: 10.1016/j.isci.2022.105911] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/30/2022] [Accepted: 12/28/2022] [Indexed: 12/31/2022] Open
Abstract
Placental hormones orchestrate maternal metabolic adaptations to support pregnancy. We hypothesized that placental ER stress, which characterizes early-onset pre-eclampsia (ePE), compromises glycosylation, reducing hormone bioactivity and these maladaptations predispose the mother to metabolic disease in later life. We demonstrate ER stress reduces the complexity and sialylation of trophoblast protein N-glycosylation, while aberrant glycosylation of vascular endothelial growth factor reduced its bioactivity. ER stress alters the expression of 66 of the 146 genes annotated with "protein glycosylation" and reduces the expression of sialyltransferases. Using mouse placental explants, we show ER stress promotes the secretion of mis-glycosylated glycoproteins. Pregnant mice carrying placentas with junctional zone-specific ER stress have reduced blood glucose, anomalous hepatic glucose metabolism, increased cellular stress and elevated DNA methyltransferase 3A. Using pregnancy-specific glycoproteins as a readout, we also demonstrate aberrant glycosylation of placental proteins in women with ePE, thus providing a mechanistic link between ePE and subsequent maternal metabolic disorders.
Collapse
Affiliation(s)
- Hong Wa Yung
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Xiaohui Zhao
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Luke Glover
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Charlotte Burrin
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - Poh-Choo Pang
- Department of Life Sciences, Imperial College London, London, UK
| | - Carolyn J.P. Jones
- Maternal and Fetal Health Centre, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, UK
| | - Carolyn Gill
- Department of Women and Children’s Health, King’s College London, London, UK
| | - Kate Duhig
- Maternal and Fetal Health Centre, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, UK
- Department of Women and Children’s Health, King’s College London, London, UK
| | - Matts Olovsson
- Department of Women’s and Children’s Health, Uppsala University, Uppsala, Sweden
| | - Lucy C. Chappell
- Department of Women and Children’s Health, King’s College London, London, UK
| | - Stuart M. Haslam
- Department of Life Sciences, Imperial College London, London, UK
| | - Anne Dell
- Department of Life Sciences, Imperial College London, London, UK
| | - Graham J. Burton
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
| | - D. Stephen Charnock-Jones
- Centre for Trophoblast Research, University of Cambridge, Cambridge CB2 3EG, UK
- Department of Obstetrics and Gynaecology, University of Cambridge, Cambridge CB2 0SW, UK
| |
Collapse
|
9
|
Nambiar SM, Lee J, Yanum JA, Garcia V, Jiang H, Dai G. Maternal hepatocytes heterogeneously and dynamically exhibit developmental phenotypes partially via yes-associated protein 1 during pregnancy. Am J Physiol Gastrointest Liver Physiol 2023; 324:G38-G50. [PMID: 36283963 PMCID: PMC9799147 DOI: 10.1152/ajpgi.00197.2022] [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: 08/08/2022] [Revised: 10/11/2022] [Accepted: 10/21/2022] [Indexed: 02/08/2023]
Abstract
Pregnancy induces reprogramming of maternal physiology to support fetal development and growth. Maternal hepatocytes undergo hypertrophy and hyperplasia to drive maternal liver growth and alter their gene expression profiles simultaneously. This study aimed to further understand maternal hepatocyte adaptation to pregnancy. Timed pregnancies were generated in mice. In a nonpregnant state, most hepatocytes expressed Cd133, α-fetal protein (Afp) and epithelial cell adhesion molecule (Epcam) mRNAs, whereas overall, at the protein level, they exhibited a CD133-/AFP- phenotype; however, pericentral hepatocytes were EpCAM+. As pregnancy advanced, although most maternal hepatocytes retained Cd133, Afp, and Epcam mRNA expression, they generally displayed a phenotype of CD133+/AFP+, and EpCAM protein expression was switched from pericentral to periportal maternal hepatocytes. In addition, we found that the Hippo/yes-associated protein (YAP) pathway does not respond to pregnancy. Yap1 gene deletion specifically in maternal hepatocytes did not affect maternal liver growth or metabolic zonation. However, the absence of Yap1 gene eliminated CD133 protein expression without interfering with Cd133 transcript expression in maternal livers. We demonstrated that maternal hepatocytes acquire heterogeneous and dynamic developmental phenotypes, resembling fetal hepatocytes, partially via YAP1 through a posttranscriptional mechanism. Moreover, maternal liver is a new source of AFP. In addition, maternal liver grows and maintains its metabolic zonation independent of the Hippo/YAP1 pathway. Our findings revealed a novel and gestation-dependent phenotypic plasticity in adult hepatocytes.NEW & NOTEWORTHY We found that maternal hepatocytes exhibit developmental phenotypes in a temporal and spatial manner, similarly to fetal hepatocytes. They acquire this new property partially via yes-associated protein 1.
Collapse
Affiliation(s)
- Shashank Manohar Nambiar
- Department of Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana
| | - Joonyong Lee
- Department of Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana
| | - Jennifer Abla Yanum
- Department of Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana
| | - Veronica Garcia
- Department of Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana
| | - Huaizhou Jiang
- Department of Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana
| | - Guoli Dai
- Department of Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana
| |
Collapse
|
10
|
Monte AA, Mackenzie IA, Pattee J, Kaiser S, Willems E, Rumack B, Reynolds KM, Dart RC, Heard KJ. Genetic variants associated with ALT elevation from therapeutic acetaminophen. Clin Toxicol (Phila) 2022; 60:1198-1204. [PMID: 36102175 PMCID: PMC9701448 DOI: 10.1080/15563650.2022.2117053] [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: 06/07/2022] [Revised: 08/16/2022] [Accepted: 08/18/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Several studies have suggested genetic variants associated with acetaminophen induced liver injury (DILI) following overdose. Genetic variation associated with acetaminophen-induced alanine aminotransferase elevation during therapeutic dosing has not been examined. METHODS We performed genetic analyses on patients that ingested therapeutic doses of 4 grams of acetaminophen for up to 16 days. We examined 20 genes previously implicated in the metabolism of acetaminophen or the development of immune-mediated DILI using the Illumina Multi-Ethnic Global Array 2. Autosomes were aligned and imputed using TOPMed. A candidate gene region analysis was performed by testing each gene individually using linkage disequilibrium (LD) pruned variants with the adaptive sum of powered scores (aSPU) test from the aSPU R package. The highest measured ALT during therapy, the maximum ALT, was used as the outcome. RESULTS 192 subjects taking therapeutic APAP were included in the genetic analysis. 136 (70.8%) were female, 133 (69.2%) were Caucasian race, and the median age was 34 years (IQR: 26, 46). Age > 50 years was the only clinical factor associated with maximum ALT increase. Variants in SULT1E1, the gene responsible for Sulfotransferase Family 1E Member 1 enzyme production, were associated with maximum ALT. No single variant drove this association, but rather the association was due to the additive effects of numerous variants within the gene. No other genes were associated with maximum ALT increase in this cohort. CONCLUSION Acetaminophen induced ALT elevation at therapeutic doses was not associated with variation in most genes associated with acetaminophen metabolism or immune-induced DILI in this cohort. The role of SULT1E1 polymorphism in acetaminophen-induced elevated ALT needs further examination.
Collapse
Affiliation(s)
- Andrew A. Monte
- University of Colorado School of Medicine, Department of Emergency Medicine, Aurora, CO
- University of Colorado School of Medicine, Center for Bioinformatics & Personalized Medicine, Aurora, CO
- University of Colorado, Skaggs School of Pharmacy, Aurora, CO
- Rocky Mountain Poison & Drug Safety, Denver Health and Hospital Authority, Denver, CO
| | - Ian Arriaga Mackenzie
- Department of Biostatistics & Informatics, Colorado School of Public Health, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO
| | - Jack Pattee
- Department of Biostatistics & Informatics, Colorado School of Public Health, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO
| | - Sasha Kaiser
- Rocky Mountain Poison & Drug Safety, Denver Health and Hospital Authority, Denver, CO
| | - Emileigh Willems
- Department of Biostatistics & Informatics, Colorado School of Public Health, University of Colorado-Denver Anschutz Medical Campus, Aurora, CO
| | - Barry Rumack
- University of Colorado School of Medicine, Department of Emergency Medicine, Aurora, CO
- Rocky Mountain Poison & Drug Safety, Denver Health and Hospital Authority, Denver, CO
| | - Kate M. Reynolds
- Rocky Mountain Poison & Drug Safety, Denver Health and Hospital Authority, Denver, CO
| | - Richard C. Dart
- Rocky Mountain Poison & Drug Safety, Denver Health and Hospital Authority, Denver, CO
| | - Kennon J. Heard
- University of Colorado School of Medicine, Department of Emergency Medicine, Aurora, CO
- Rocky Mountain Poison & Drug Safety, Denver Health and Hospital Authority, Denver, CO
| |
Collapse
|
11
|
Liebmann M, Grupe K, Asuaje Pfeifer M, Rustenbeck I, Scherneck S. Differences in lipid metabolism in acquired versus preexisting glucose intolerance during gestation: role of free fatty acids and sphingosine-1-phosphate. Lipids Health Dis 2022; 21:99. [PMID: 36209101 PMCID: PMC9547403 DOI: 10.1186/s12944-022-01706-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 09/23/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The prevalence of gestational diabetes mellitus (GDM) is increasing worldwide. There is increasing evidence that GDM is a heterogeneous disease with different subtypes. An important question in this context is whether impaired glucose tolerance (IGT), which is a typical feature of the disease, may already be present before pregnancy and manifestation of the disease. The latter type resembles in its clinical manifestation prediabetes that has not yet manifested as type 2 diabetes (T2DM). Altered lipid metabolism plays a crucial role in the disorder's pathophysiology. The aim was to investigate the role of lipids which are relevant in diabetes-like phenotypes in these both models with different time of initial onset of IGT. METHODS Two rodent models reflecting different characteristics of human GDM were used to characterize changes in lipid metabolism occurring during gestation. Since the New Zealand obese (NZO)-mice already exhibit IGT before and during gestation, they served as a subtype model for GDM with preexisting IGT (preIGT) and were compared with C57BL/6 N mice with transient IGT acquired during gestation (aqIGT). While the latter model does not develop manifest diabetes even under metabolic stress conditions, the NZO mouse is prone to severe disease progression later in life. Metabolically healthy Naval Medical Research Institute (NMRI) mice served as controls. RESULTS In contrast to the aqIGT model, preIGT mice showed hyperlipidemia during gestation with elevated free fatty acids (FFA), triglycerides (TG), and increased atherogenic index. Interestingly, sphingomyelin (SM) concentrations in the liver decreased during gestation concomitantly with an increase in the sphingosine-1-phosphate (S1P) concentration in plasma. Further, preIGT mice showed impaired hepatic weight adjustment and alterations in hepatic FFA metabolism during gestation. This was accompanied by decreased expression of peroxisome proliferator-activated receptor alpha (PPARα) and lack of translocation of fatty acid translocase (FAT/CD36) to the hepatocellular plasma membrane. CONCLUSION The preIGT model showed impaired lipid metabolism both in plasma and liver, as well as features of insulin resistance consistent with increased S1P concentrations, and in these characteristics, the preIGT model differs from the common GDM subtype with aqIGT. Thus, concomitantly elevated plasma FFA and S1P concentrations, in addition to general shifts in sphingolipid fractions, could be an interesting signal that the metabolic disorder existed before gestation and that future pregnancies require more intensive monitoring to avoid complications. This graphical abstract was created with BioRender.com .
Collapse
Affiliation(s)
- Moritz Liebmann
- Institute of Pharmacology, Toxicology and Clinical Pharmacy, Technische Universität Braunschweig, D-38106, Braunschweig, Germany
| | - Katharina Grupe
- Institute of Pharmacology, Toxicology and Clinical Pharmacy, Technische Universität Braunschweig, D-38106, Braunschweig, Germany
| | - Melissa Asuaje Pfeifer
- Institute of Pharmacology, Toxicology and Clinical Pharmacy, Technische Universität Braunschweig, D-38106, Braunschweig, Germany
| | - Ingo Rustenbeck
- Institute of Pharmacology, Toxicology and Clinical Pharmacy, Technische Universität Braunschweig, D-38106, Braunschweig, Germany
| | - Stephan Scherneck
- Institute of Pharmacology, Toxicology and Clinical Pharmacy, Technische Universität Braunschweig, D-38106, Braunschweig, Germany.
| |
Collapse
|
12
|
Bowdridge EC, DeVallance E, Garner KL, Griffith JA, Schafner K, Seaman M, Engels KJ, Wix K, Batchelor TP, Goldsmith WT, Hussain S, Nurkiewicz TR. Nano-titanium dioxide inhalation exposure during gestation drives redox dysregulation and vascular dysfunction across generations. Part Fibre Toxicol 2022; 19:18. [PMID: 35260159 PMCID: PMC8905816 DOI: 10.1186/s12989-022-00457-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 02/24/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Pregnancy is associated with many rapid biological adaptations that support healthy development of the growing fetus. One of which is critical to fetal health and development is the coordination between maternal liver derived substrates and vascular delivery. This crucial adaptation can be potentially derailed by inhalation of toxicants. Engineered nanomaterials (ENM) are commonly used in household and industrial products as well as in medicinal applications. As such, the potential risk of exposure remains a concern, especially during pregnancy. We have previously reported that ENM inhalation leads to upregulation in the production of oxidative species. Therefore, we aimed to determine if F0 dam maternal nano-TiO2 inhalation exposure (exclusively) resulted in altered H2O2 production capacity and changes in downstream redox pathways in the F0 dams and subsequent F1 pups. Additionally, we investigated whether this persisted into adulthood within the F1 generation and how this impacted F1 gestational outcomes and F2 fetal health and development. We hypothesized that maternal nano-TiO2 inhalation exposure during gestation in the F0 dams would result in upregulated H2O2 production in the F0 dams as well as her F1 offspring. Additionally, this toxicological insult would result in gestational vascular dysfunction in the F1 dams yielding smaller F2 generation pups. RESULTS Our results indicate upregulation of hepatic H2O2 production capacity in F0 dams, F1 offspring at 8 weeks and F1 females at gestational day 20. H2O2 production capacity was accompanied by a twofold increase in phosphorylation of the redox sensitive transcription factor NF-κB. In cell culture, naïve hepatocytes exposed to F1-nano-TiO2 plasma increased H2O2 production. Overnight exposure of these hepatocytes to F1 plasma increased H2O2 production capacity in a partially NF-κB dependent manner. Pregnant F1- nano-TiO2 females exhibited estrogen disruption (12.12 ± 3.1 pg/ml vs. 29.81 ± 8.8 pg/ml sham-control) and vascular dysfunction similar to their directly exposed mothers. F1-nano-TiO2 uterine artery H2O2 production capacity was also elevated twofold. Dysfunctional gestational outcomes in the F1-nano-TiO2 dams resulted in smaller F1 (10.22 ± 0.6 pups vs. sham-controls 12.71 ± 0.96 pups) and F2 pups (4.93 ± 0.47 g vs. 5.78 ± 0.09 g sham-control pups), and fewer F1 male pups (4.38 ± 0.3 pups vs. 6.83 ± 0.84 sham-control pups). CONCLUSION In conclusion, this manuscript provides critical evidence of redox dysregulation across generations following maternal ENM inhalation. Furthermore, dysfunctional gestational outcomes are observed in the F1-nano-TiO2 generation and impact the development of F2 offspring. In total, this data provides strong initial evidence that maternal ENM exposure has robust biological impacts that persists in at least two generations.
Collapse
Affiliation(s)
- Elizabeth C. Bowdridge
- grid.268154.c0000 0001 2156 6140Department of Physiology and Pharmacology, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, West Virginia University, Morgantown, WV 26505-9229 USA ,grid.268154.c0000 0001 2156 6140Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV USA
| | - Evan DeVallance
- grid.268154.c0000 0001 2156 6140Department of Physiology and Pharmacology, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, West Virginia University, Morgantown, WV 26505-9229 USA ,grid.268154.c0000 0001 2156 6140Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV USA
| | - Krista L. Garner
- grid.268154.c0000 0001 2156 6140Department of Physiology and Pharmacology, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, West Virginia University, Morgantown, WV 26505-9229 USA ,grid.268154.c0000 0001 2156 6140Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV USA
| | - Julie A. Griffith
- grid.268154.c0000 0001 2156 6140Department of Physiology and Pharmacology, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, West Virginia University, Morgantown, WV 26505-9229 USA ,grid.268154.c0000 0001 2156 6140Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV USA
| | - Kallie Schafner
- grid.268154.c0000 0001 2156 6140Department of Physiology and Pharmacology, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, West Virginia University, Morgantown, WV 26505-9229 USA ,grid.268154.c0000 0001 2156 6140Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV USA
| | - Madison Seaman
- grid.268154.c0000 0001 2156 6140Department of Physiology and Pharmacology, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, West Virginia University, Morgantown, WV 26505-9229 USA
| | - Kevin J. Engels
- grid.268154.c0000 0001 2156 6140Department of Physiology and Pharmacology, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, West Virginia University, Morgantown, WV 26505-9229 USA
| | - Kimberley Wix
- grid.268154.c0000 0001 2156 6140Department of Physiology and Pharmacology, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, West Virginia University, Morgantown, WV 26505-9229 USA
| | - Thomas P. Batchelor
- grid.268154.c0000 0001 2156 6140Department of Physiology and Pharmacology, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, West Virginia University, Morgantown, WV 26505-9229 USA ,grid.268154.c0000 0001 2156 6140Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV USA
| | - William T. Goldsmith
- grid.268154.c0000 0001 2156 6140Department of Physiology and Pharmacology, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, West Virginia University, Morgantown, WV 26505-9229 USA ,grid.268154.c0000 0001 2156 6140Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV USA
| | - Salik Hussain
- grid.268154.c0000 0001 2156 6140Department of Physiology and Pharmacology, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, West Virginia University, Morgantown, WV 26505-9229 USA ,grid.268154.c0000 0001 2156 6140Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV USA
| | - Timothy R. Nurkiewicz
- grid.268154.c0000 0001 2156 6140Department of Physiology and Pharmacology, 64 Medical Center Drive, Robert C. Byrd Health Sciences Center, West Virginia University School of Medicine, West Virginia University, Morgantown, WV 26505-9229 USA ,grid.268154.c0000 0001 2156 6140Center for Inhalation Toxicology, West Virginia University School of Medicine, Morgantown, WV USA
| |
Collapse
|
13
|
Hsieh J, Molusky MM, McCabe KM, Fotakis P, Xiao T, Tascau L, Zeana-Schliep L, DaSilva-Jardine P, Tall AR. TTC39B destabilizes retinoblastoma protein promoting hepatic lipogenesis in a sex-specific fashion. J Hepatol 2022; 76:383-393. [PMID: 34600974 PMCID: PMC8766887 DOI: 10.1016/j.jhep.2021.09.021] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND & AIMS Molecular mechanisms underlying the different susceptibility of men and women to non-alcoholic fatty liver disease (NAFLD) are poorly understood. The TTC39B locus encodes a scaffolding protein, associates with gynecological disorders and its deletion protects mice from diet-induced steatohepatitis. This study aimed to elucidate the molecular mechanisms linking TTC39B (T39) to the expression of lipogenic genes and to explore sex-specific effects. METHODS Co-expression in HEK293A cells validated the novel T39/pRb interaction predicted by a protein-protein interaction algorithm. T39 was knocked down using an antisense oligonucleotide (ASO) in mice with dietary NAFLD and a genetic deficiency of pRb or its downstream effector E2F1, as well as in primary human hepatocytes. RESULTS T39 interacts with pRb via its C-terminal TPR domain and promotes its proteasomal degradation. In female mice, T39 deficiency reduces the mRNA of lipogenic genes, especially Pnpla3, in a pRb- and E2F1-dependent manner. In contrast, in male mice, T39 deficiency results in a much smaller reduction in lipogenic gene expression that is independent of pRb/E2F1. T39 also interacts with VAPB via an N-terminal FFAT motif and stabilizes the interaction of VAPB with SCAP. Ovariectomy abolishes the effect of T39 knockdown on the hepatic pRb/E2F1/Pnpla3 axis. In both sexes T39 knockdown reduces SCAP independently of pRb. In primary human hepatocytes, T39 knockdown reduces expression of PNPLA3 and other lipogenic genes in women but not men. CONCLUSIONS We have uncovered a conserved sexual dimorphism in the regulation of hepatic lipogenic genes, with effects of T39 mediated through pRb/E2F1 in females and VAPB/SCAP in both sexes. T39 inhibition could be a novel strategy to downregulate PNPLA3 and treat NAFLD in women. LAY SUMMARY In females, the protein TTC39B degrades a tumor suppressor in the liver to promote the synthesis of new fat and the expression of a major genetic risk factor for non-alcoholic fatty liver disease. TTC39B is a potential therapeutic target for non-alcoholic fatty liver disease, especially in women.
Collapse
Affiliation(s)
- Joanne Hsieh
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY, 10032, USA.
| | - Matthew M. Molusky
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY, 10032, USA
| | - Kristin M. McCabe
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY, 10032, USA
| | - Panagiotis Fotakis
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY, 10032, USA
| | - Tong Xiao
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY, 10032, USA
| | - Liana Tascau
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY, 10032, USA
| | - Lars Zeana-Schliep
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY, 10032, USA
| | | | - Alan R. Tall
- Division of Molecular Medicine, Department of Medicine, Columbia University, New York, NY, 10032, USA
| |
Collapse
|
14
|
Nuclear Receptors in Pregnancy and Outcomes: Clinical Perspective. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1390:3-19. [DOI: 10.1007/978-3-031-11836-4_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
15
|
Cho J, Tsugawa Y, Imai Y, Imai T. Chorionic gonadotropin stimulates maternal hepatocyte proliferation during pregnancy. Biochem Biophys Res Commun 2021; 579:110-115. [PMID: 34597993 DOI: 10.1016/j.bbrc.2021.09.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 09/17/2021] [Indexed: 12/22/2022]
Abstract
The liver increases its size during pregnancy to adapt to metabolic demand associated with pregnancy. Our previous study showed that proliferation of maternal hepatocytes are increased during pregnancy in mice and that estradiol (E2) is one of the candidate hormones responsible for maternal hepatocyte proliferation. Here, we discovered that chorionic gonadotropin (CG) induces maternal hepatocyte proliferation during pregnancy. CG administration was sufficient to stimulate hepatocyte proliferation in non-pregnant mice as well as in cell culture system. We conclude that CG stimulates proliferation in the early pregnancy of maternal hepatocytes. In contrast, estrogen stimulates hepatocyte proliferation in the late pregnancy.
Collapse
Affiliation(s)
- Jaeyong Cho
- Department of Chemical Biology, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan.
| | - Yoji Tsugawa
- Department of Chemical Biology, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan.
| | - Yumi Imai
- Department of Internal Medicine, Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
| | - Takeshi Imai
- Department of Chemical Biology, National Center for Geriatrics and Gerontology, Obu, Aichi, 474-8511, Japan.
| |
Collapse
|
16
|
Gao M, Cai C, Han X, Wang L, Zhang W, Zhang L, Yang L. The early stage of pregnancy modulates toll-like receptor signaling in the ovine liver. JOURNAL OF APPLIED ANIMAL RESEARCH 2021. [DOI: 10.1080/09712119.2021.1990935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Meihong Gao
- Department of Animal Science, School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, People’s Republic of China
| | - Chunjiang Cai
- Department of Animal Science, School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, People’s Republic of China
| | - Xu Han
- Department of Animal Science, School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, People’s Republic of China
| | - Luyu Wang
- Department of Animal Science, School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, People’s Republic of China
| | - Weifeng Zhang
- Department of Animal Science, School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, People’s Republic of China
| | - Leying Zhang
- Department of Animal Science, School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, People’s Republic of China
| | - Ling Yang
- Department of Animal Science, School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, People’s Republic of China
| |
Collapse
|
17
|
Lee J, Garcia V, Nambiar SM, Jiang H, Dai G. Activation of Proneuronal Transcription Factor Ascl1 in Maternal Liver Ensures a Healthy Pregnancy. Cell Mol Gastroenterol Hepatol 2021; 13:35-55. [PMID: 34438112 PMCID: PMC8600092 DOI: 10.1016/j.jcmgh.2021.08.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 08/08/2021] [Accepted: 08/10/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND & AIMS Maternal liver shows robust adaptations to pregnancy to accommodate the metabolic needs of the developing and growing placenta and fetus by largely unknown mechanisms. We found that Ascl1, a gene encoding a basic helix-loop-helix transcription factor essential for neuronal development, is highly activated in maternal hepatocytes during the second half of gestation in mice. METHODS To investigate whether and how Ascl1 plays a pregnancy-dependent role, we deleted the Ascl1 gene specifically in maternal hepatocytes from midgestation until term. RESULTS As a result, we identified multiple Ascl1-dependent phenotypes. Maternal livers lacking Ascl1 showed aberrant hepatocyte structure, increased hepatocyte proliferation, enlarged hepatocyte size, reduced albumin production, and increased release of liver enzymes, indicating maternal liver dysfunction. Simultaneously, maternal pancreas and spleen and the placenta showed marked overgrowth; and the maternal ceca microbiome showed alterations in relative abundance of several bacterial subpopulations. Moreover, litters born from maternal hepatic Ascl1-deficient dams experienced abnormal postnatal growth after weaning, implying an adverse pregnancy outcome. Mechanistically, we found that maternal hepatocytes deficient for Ascl1 showed robust activation of insulin-like growth factor 2 expression, which may contribute to the Ascl1-dependent phenotypes widespread in maternal and uteroplacental compartments. CONCLUSIONS In summary, we show that maternal liver, via activating Ascl1 expression, modulates the adaptations of maternal organs and the growth of the placenta to maintain a healthy pregnancy. Our studies show that Ascl1 is a novel and critical regulator of the physiology of pregnancy.
Collapse
Affiliation(s)
- Joonyong Lee
- Department of Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana
| | - Veronica Garcia
- Department of Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana
| | - Shashank M Nambiar
- Department of Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana
| | - Huaizhou Jiang
- Department of Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana; School of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China.
| | - Guoli Dai
- Department of Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana.
| |
Collapse
|
18
|
Cao N, Cao L, Gao M, Wang H, Zhang L, Yang L. Changes in mRNA and protein levels of gonadotropin releasing hormone and receptor in ovine thymus, lymph node, spleen, and liver during early pregnancy. Domest Anim Endocrinol 2021; 76:106607. [PMID: 33582417 DOI: 10.1016/j.domaniend.2021.106607] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 01/17/2021] [Accepted: 01/17/2021] [Indexed: 12/18/2022]
Abstract
There exists maternal immunological modification in maternal immune organs during early pregnancy in mammals. Gonadotropin releasing hormone (GnRH) is widely distributed in vertebrate tissues, including immune organs. However, it is unclear that early pregnancy induces expression of GnRH and GnRH receptor (GnRHR) in ovine immune organs. The objective of this study was to explore the expression of GnRH and GnRHR in main immune organs (thymus, lymph node, spleen, and liver) during early pregnancy in sheep. Ovine thymus, lymph node, spleen and liver were sampled at day 16 of estrous cycle, and days 13, 16, and 25 of pregnancy. The expression of GnRH and GnRHR was detected through real-time quantitative PCR, Western blot and immunohistochemistry analysis. The results indicated that early pregnancy induced upregulation of mRNA and protein levels of GnRH and GnRHR in the maternal lymph node, spleen and liver, and mRNA and protein of GnRH in the maternal thymus, but mRNA and protein of GnRHR decreased in the maternal thymus during early pregnancy. In summary, the mRNA and protein levels of GnRH and GnRHR were changed in maternal thymus, lymph node, spleen and liver in a tissue specific manner during early pregnancy in sheep.
Collapse
Affiliation(s)
- N Cao
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan 056038, China
| | - L Cao
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan 056038, China
| | - M Gao
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan 056038, China
| | - H Wang
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan 056038, China
| | - L Zhang
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan 056038, China
| | - L Yang
- School of Life Sciences and Food Engineering, Hebei University of Engineering, Handan 056038, China.
| |
Collapse
|
19
|
Manna LB, Williamson C. Nuclear receptors, gestational metabolism and maternal metabolic disorders. Mol Aspects Med 2021; 78:100941. [PMID: 33455843 DOI: 10.1016/j.mam.2021.100941] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Accepted: 01/04/2021] [Indexed: 12/20/2022]
Abstract
Normal pregnancy is characterised by a gradual alteration in metabolism that results in elevated serum bile acids, dyslipidaemia and impaired glucose tolerance in the third trimester. Nuclear receptors play important roles in regulating metabolic pathways that influence alterations in these parameters. There is evidence for altered function of FXR and LXR in gestation; these nuclear receptors play an integral role in bile acid and lipid homeostasis. There is some evidence for influence of clock genes in late pregnancy metabolic changes, and this may be linked to alterations in placental gene expression and function, thereby influencing fetal growth. This article will review the current data from human studies and investigation of animal models to illustrate the role of nuclear receptors (namely LXR, FXR, PPARs and clock genes) in gestational alterations in metabolism and the ways this may influence susceptibility to metabolic disorders of pregnancy such as gestational diabetes mellitus and intrahepatic cholestasis of pregnancy.
Collapse
Affiliation(s)
- Luiza Borges Manna
- Division of Women and Children's Health, King's College London, London, United Kingdom
| | - Catherine Williamson
- Division of Women and Children's Health, King's College London, London, United Kingdom.
| |
Collapse
|
20
|
Garczyńska K, Tzschätzsch H, Kühl AA, Morr AS, Lilaj L, Häckel A, Schellenberger E, Berndt N, Holzhütter HG, Braun J, Sack I, Guo J. Changes in Liver Mechanical Properties and Water Diffusivity During Normal Pregnancy Are Driven by Cellular Hypertrophy. Front Physiol 2020; 11:605205. [PMID: 33329058 PMCID: PMC7719759 DOI: 10.3389/fphys.2020.605205] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Accepted: 10/29/2020] [Indexed: 12/27/2022] Open
Abstract
During pregnancy, the body’s hyperestrogenic state alters hepatic metabolism and synthesis. While biochemical changes related to liver function during normal pregnancy are well understood, pregnancy-associated alterations in biophysical properties of the liver remain elusive. In this study, we investigated 26 ex vivo fresh liver specimens harvested from pregnant and non-pregnant rats by diffusion-weighted imaging (DWI) and magnetic resonance elastography (MRE) in a 0.5-Tesla compact magnetic resonance imaging (MRI) scanner. Water diffusivity and viscoelastic parameters were compared with histological data and blood markers. We found livers from pregnant rats to have (i) significantly enlarged hepatocytes (26 ± 15%, p < 0.001), (ii) increased liver stiffness (12 ± 15%, p = 0.012), (iii) decreased viscosity (−23 ± 14%, p < 0.001), and (iv) increased water diffusivity (12 ± 11%, p < 0.001). In conclusion, increased stiffness and reduced viscosity of the liver during pregnancy are mainly attributable to hepatocyte enlargement. Hypertrophy of liver cells imposes fewer restrictions on intracellular water mobility, resulting in a higher hepatic water diffusion coefficient. Collectively, MRE and DWI have the potential to inform on structural liver changes associated with pregnancy in a clinical context.
Collapse
Affiliation(s)
- Karolina Garczyńska
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Heiko Tzschätzsch
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Anja A Kühl
- iPATH.Berlin Core Unit, Charitá - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Anna Sophie Morr
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ledia Lilaj
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Akvile Häckel
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Eyk Schellenberger
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Nikolaus Berndt
- Institute for Imaging Science and Computational Modelling in Cardiovascular Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Computational Systems Biochemistry Group, Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Hermann-Georg Holzhütter
- Computational Systems Biochemistry Group, Institute of Biochemistry, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jürgen Braun
- Institute of Medical Informatics, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Ingolf Sack
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jing Guo
- Department of Radiology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| |
Collapse
|
21
|
Yang L, Han X, Zhang L, Li N, Zhao Z, Bai J. Changes in expression of prostaglandin synthase in ovine liver during early pregnancy. CANADIAN JOURNAL OF ANIMAL SCIENCE 2020. [DOI: 10.1139/cjas-2019-0171] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Liver can function as part of the innate and adaptive immune systems. We hypothesize that prostaglandins participate in the regulation of hepatic immune function during early pregnancy in sheep. The objective of this study was to elucidate expression of prostaglandin synthase in ovine liver during early pregnancy. Ovine livers were sampled on day 16 of the estrous cycle, and days 13, 16, and 25 of pregnancy, and the expression of prostaglandin synthases, including prostaglandin-endoperoxide synthase 1 (PTGS1), PTGS2, prostaglandin E synthase (PTGES), and aldo-keto reductase family 1, member B1, a prostaglandin F synthase (PGFS), were detected by quantitative real-time polymerase chain reaction, Western blot, and immunohistochemistry analysis. There were increases in the expression of mRNA and the proteins of PTGS2, PTGES, and PGFS in the livers during early pregnancy, but PTGS1 was decreased in the pregnant ewes. The PGFS protein was limited to the hepatocytes and the endothelial cells of the proper hepatic arteries and hepatic portal veins. In summary, the upregulation of PTGS2, PTGES, and PGFS and downregulation of PTGS1 may be involved in the maternal hepatic immune adjustment during early pregnancy in sheep.
Collapse
Affiliation(s)
- Ling Yang
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, Hebei 056038, People’s Republic of China
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, Hebei 056038, People’s Republic of China
| | - Xu Han
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, Hebei 056038, People’s Republic of China
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, Hebei 056038, People’s Republic of China
| | - Leying Zhang
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, Hebei 056038, People’s Republic of China
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, Hebei 056038, People’s Republic of China
| | - Ning Li
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, Hebei 056038, People’s Republic of China
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, Hebei 056038, People’s Republic of China
| | - Zimo Zhao
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, Hebei 056038, People’s Republic of China
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, Hebei 056038, People’s Republic of China
| | - Jiachen Bai
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, Hebei 056038, People’s Republic of China
- College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, Hebei 056038, People’s Republic of China
| |
Collapse
|
22
|
Lee J, Garcia V, Nambiar SM, Jiang H, Dai G. Pregnancy facilitates maternal liver regeneration after partial hepatectomy. Am J Physiol Gastrointest Liver Physiol 2020; 318:G772-G780. [PMID: 32003603 PMCID: PMC7191459 DOI: 10.1152/ajpgi.00125.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Liver resection induces robust liver regrowth or regeneration to compensate for the lost tissue mass. In a clinical setting, pregnant women may need liver resection without terminating pregnancy in some cases. However, how pregnancy affects maternal liver regeneration remains elusive. We performed 70% partial hepatectomy (PH) in nonpregnant mice and gestation day 14 mice, and histologically and molecularly compared their liver regrowth during the next 4 days. We found that compared with the nonpregnant state, pregnancy altered the molecular programs driving hepatocyte replication, indicated by enhanced activities of epidermal growth factor receptor and STAT5A, reduced activities of cMet and p70S6K, decreased production of IL-6, TNFα, and hepatocyte growth factor, suppressed cyclin D1 expression, increased cyclin A1 expression, and early activated cyclin A2 expression. As a result, pregnancy allowed the remnant hepatocytes to enter the cell cycle at least 12 h earlier, increased hepatic fat accumulation, and enhanced hepatocyte mitosis. Consequently, pregnancy ameliorated maternal liver regeneration following PH. In addition, a report showed that maternal liver regrowth after PH is driven mainly by hepatocyte hypertrophy rather than hyperplasia during the second half of gestation in young adult mice. In contrast, we demonstrate that maternal liver relies mainly on hepatocyte hyperplasia instead of hypertrophy to restore the lost mass after PH. Overall, we demonstrate that pregnancy facilitates maternal liver regeneration likely via triggering an early onset of hepatocyte replication, accumulating excessive liver fat, and promoting hepatocyte mitosis. The results from our current studies enable us to gain more insights into how maternal liver regeneration progresses during gestation.NEW & NOTEWORTHY We demonstrate that pregnancy may generate positive effects on maternal liver regeneration following partial hepatectomy, which are manifested by early entry of the cell cycle of remnant hepatocytes, increased hepatic fat accumulation, enhanced hepatocyte mitosis, and overall accelerated liver regrowth.
Collapse
Affiliation(s)
- Joonyong Lee
- 1Department of Biology, Center for Developmental and Regenerative Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana
| | - Veronica Garcia
- 1Department of Biology, Center for Developmental and Regenerative Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana
| | - Shashank Manohar Nambiar
- 1Department of Biology, Center for Developmental and Regenerative Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana
| | - Huaizhou Jiang
- 1Department of Biology, Center for Developmental and Regenerative Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana,2School of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Anhui, China
| | - Guoli Dai
- 1Department of Biology, Center for Developmental and Regenerative Biology, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana
| |
Collapse
|
23
|
Bai J, Zhang L, Zhao Z, Li N, Wang B, Yang L. Expression of melatonin receptors and CD4 in the ovine thymus, lymph node, spleen and liver during early pregnancy. Immunology 2020; 160:52-63. [PMID: 32052861 DOI: 10.1111/imm.13180] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 01/20/2020] [Accepted: 02/10/2020] [Indexed: 01/14/2023] Open
Abstract
As a pineal gland hormone, melatonin acts through its receptors to modulate the immune system. The immune system is composed of primary and secondary organs, and immune organs are adapted to the presence of the fetal alloantigen during pregnancy. However, it is unclear whether melatonin affects maternal immune organs during early pregnancy in sheep. In this study, the ovine thymus, lymph node, spleen and liver were sampled at day 16 of the oestrous cycle, and at days 13, 16 and 25 of pregnancy. The expression of melatonin receptor 1A (MT1), melatonin receptor 1B (MT2) and cluster of differentiation 4 (CD4) was detected by quantitative real-time polymerase chain reaction, Western blot and immunohistochemistry experiments. Our results showed that during early pregnancy there was an upregulation of MT1 mRNA and protein in the thymus, lymph node and liver, and there was a downregulation in the spleen. The expression of MT2 mRNA and protein was increased in the thymus but decreased in the spleen and liver, and there was no significant change in the lymph node during early pregnancy. CD4 protein was upregulated in the thymus, lymph node and liver, but there were no significant changes in the spleen during early pregnancy. In conclusion, early pregnancy induces tissue-specific expression of MT1, MT2 and CD4, which may be due to the different functions of the thymus, lymph node, spleen and liver. Further, melatonin is involved in immune regulation of the maternal thymus, lymph node, spleen and liver during early pregnancy in sheep.
Collapse
Affiliation(s)
- Jiachen Bai
- Department of Animal Science, College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Leying Zhang
- Department of Animal Science, College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Zimo Zhao
- Department of Animal Science, College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Ning Li
- Department of Animal Science, College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Bin Wang
- Department of Animal Science, College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Ling Yang
- Department of Animal Science, College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| |
Collapse
|
24
|
The Hepatobiliary System: An Overview of Normal Function and Diagnostic Testing in Pregnancy. Clin Obstet Gynecol 2019; 63:122-133. [PMID: 31770121 DOI: 10.1097/grf.0000000000000504] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Pregnancy is associated with physiological adaptions that affect every organ system. Changes in liver function in pregnancy have important effects on nutrient metabolism, protein synthesis, and the biotransformation of substances in preparation for excretion. A clear understanding of the anatomic and functional changes of the hepatobiliary system is necessary for the diagnosis and evaluation of disease, as well as understanding how these changes predispose women to pregnancy-specific hepatic conditions. In this review, the effect of gestational changes in hepatobiliary function on laboratory tests and the role of diagnostic imaging of the liver and gallbladder in pregnancy will be discussed.
Collapse
|
25
|
Yang L, Bai J, Zhao Z, Li N, Wang Y, Zhang L. Differential expression of T helper cytokines in the liver during early pregnancy in sheep. Anim Reprod 2019; 16:332-339. [PMID: 33224295 PMCID: PMC7673597 DOI: 10.21451/1984-3143-ar2018-0141] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Liver plays important roles in the innate and adaptive immunity, and contributes to the maternal immune adjustments during pregnancy in mice and rats. T helper 1 (Th1) and Th2 cytokines are related to immune response. However, expression of Th1 and Th2 cytokines in maternal livers is unclear during early pregnancy in sheep. In this study, livers were collected on day 16 of the estrous cycle and on days 13, 16 and 25 of pregnancy (n = 6 for each group) in ewes, and qRT-PCR, western blot and immunohistochemistry were used to analyze the expression of Th1 and Th2 cytokines in the livers. Our results showed that interferon-gamma (IFN-γ), interleukin (IL)-2, IL-4, IL-6 and IL-10 were downregulated, and IL-5 was upregulated in the livers during early pregnancy. Furthermore, there was no effect for early pregnancy on expression of TNF-β in the livers, and the IFN-γ protein was limited to the endothelial cells of the proper hepatic arteries and portal veins. In conclusion, early pregnancy exerted its effect on the liver to regulate the Th cytokines expression, but there was no evident shift from Th1 to Th2 cytokines, which may be necessary for the maternal hepatic immune adjustments during early pregnancy in sheep.
Collapse
Affiliation(s)
- Ling Yang
- Department of Animal Science, College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Jiachen Bai
- Department of Animal Science, College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Zimo Zhao
- Department of Animal Science, College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Ning Li
- Department of Animal Science, College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Yujiao Wang
- Department of Animal Science, College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| | - Leying Zhang
- Department of Animal Science, College of Life Sciences and Food Engineering, Hebei University of Engineering, Handan, China
| |
Collapse
|
26
|
Napso T, Yong HEJ, Lopez-Tello J, Sferruzzi-Perri AN. The Role of Placental Hormones in Mediating Maternal Adaptations to Support Pregnancy and Lactation. Front Physiol 2018; 9:1091. [PMID: 30174608 PMCID: PMC6108594 DOI: 10.3389/fphys.2018.01091] [Citation(s) in RCA: 271] [Impact Index Per Article: 38.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 07/23/2018] [Indexed: 12/12/2022] Open
Abstract
During pregnancy, the mother must adapt her body systems to support nutrient and oxygen supply for growth of the baby in utero and during the subsequent lactation. These include changes in the cardiovascular, pulmonary, immune and metabolic systems of the mother. Failure to appropriately adjust maternal physiology to the pregnant state may result in pregnancy complications, including gestational diabetes and abnormal birth weight, which can further lead to a range of medically significant complications for the mother and baby. The placenta, which forms the functional interface separating the maternal and fetal circulations, is important for mediating adaptations in maternal physiology. It secretes a plethora of hormones into the maternal circulation which modulate her physiology and transfers the oxygen and nutrients available to the fetus for growth. Among these placental hormones, the prolactin-growth hormone family, steroids and neuropeptides play critical roles in driving maternal physiological adaptations during pregnancy. This review examines the changes that occur in maternal physiology in response to pregnancy and the significance of placental hormone production in mediating such changes.
Collapse
Affiliation(s)
- Tina Napso
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| | - Hannah E J Yong
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| | - Jorge Lopez-Tello
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| | - Amanda N Sferruzzi-Perri
- Department of Physiology, Development and Neuroscience, Centre for Trophoblast Research, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
27
|
Price LR, Lillycrop KA, Irvine NA, Hanson MA, Burdge GC. Transcriptome-wide analysis suggests that temporal changes in the relative contributions of hyperplasia, hypertrophy and apoptosis underlie liver growth in pregnant mice. Biol Reprod 2018; 97:762-771. [PMID: 29091992 DOI: 10.1093/biolre/iox136] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Accepted: 10/27/2017] [Indexed: 12/18/2022] Open
Abstract
Maternal liver undergoes structural and metabolic changes during pregnancy to meet the demands of the developing fetus. In rodents, this involves increased liver weight, but the mechanism remains unclear. To address this, we analyzed the histology, gene expression, and DNA methylation of livers of nonpregnant and pregnant C57/BL6 mice. Gestational liver growth in pregnant mice was accompanied by increased hepatocyte area and lower cell density (days 14 and 18). Expression of cell proliferation markers was increased on days 14 and 18. A total of 115 genes were differentially expressed on day 14 and 123 genes on day 18 (79 on both days). Pathway analysis indicated that pregnancy involves progressive increase in cell proliferation and decreased apoptosis. This was confirmed using archived data from the FVB wild-type mouse liver transcriptome. Four differentially DNA methylated and two differentially DNA hydroxymethylated regions identified on days 14 and 18 by methylome-wide analysis, but were not associated with altered gene expression. Long interspersed nuclear element-1 hypomethylation on days 14 and 18 was accompanied by increased ten-eleven translocase-2 and decreased DNA methyltransferase 3a and 3b expression. These findings suggest that gestational liver growth involves increased mitosis and hypertrophy, and decreased apoptosis contingent on pregnancy stage. Such changes may involve repetitive sequence, but not gene specific, DNA methylation.
Collapse
Affiliation(s)
- Leonie R Price
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Karen A Lillycrop
- Centre for Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton, UK
| | - Nicola A Irvine
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Mark A Hanson
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Graham C Burdge
- Academic Unit of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK
| |
Collapse
|
28
|
Wan XP, Xie P, Bu Z, Zou XT. Changes in hepatic glucose and lipid metabolism-related parameters in domestic pigeon (Columba livia) during incubation and chick rearing. J Anim Physiol Anim Nutr (Berl) 2017; 102:e558-e568. [PMID: 29024108 DOI: 10.1111/jpn.12796] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 07/13/2017] [Indexed: 11/28/2022]
Abstract
This study aimed to evaluate the hepatic glucose and lipid metabolism-related parameters of adult male and female White King pigeons (Columba livia) during incubation and chick rearing. At day 4 (I4), 10 (I10) and 17 (I17) of incubation and day 1 (R1), 7 (R7), 15 (R15) and 25 (R25) of chick rearing, livers were sampled from six pigeons for each sex. Glycogen and fat contents, activities of glycolytic enzymes (hexokinase, HK; 6-phosphofructokinase, 6-PFK), and genes expressions of key enzymes involved in glycolysis (pyruvate kinase, PK; glucokinase, GK), gluconeogenesis (phosphoenolpyruvate carboxykinase cytosolic, PCK1; fructose-1,6-bisphosphatase, FBP1; glucose-6-phosphatase, G6Pase), fatty acid synthesis (fatty acid synthase, FAS; acetyl-CoA carboxylase, ACC) and fatty acid β-oxidation (carnitine palmitoyltransferase 1, CPT1; acyl-CoA 1, ACO) were measured. In male and female pigeon livers, glycogen content and HK activity dramatically increased after I17 and after R1, respectively; expressions of FBP1 and G6Pase genes were maximized at R15; activity of 6-PFK and expressions of PK and CPT1 genes were highest at R7; fat content and expressions of FAS and ACC genes steeply increased from I10 to R1. In females, hepatic expressions of GK and PCK1 genes were greatest at R7 and I17, respectively; however, in males, both of them were maximized at R15. Hepatic expression of ACO gene was significantly enhanced at R1 compared to I17 and R7 in males, whereas it was notably up-regulated at I17 and R7 in females. Furthermore, expressions of PCK1, GK, FAS and ACC genes were in significant relation to fat content in the livers of female pigeons, while fat content in male pigeons was highly correlated with expression of PCK1, ACC, CPT1 and ACO genes. In conclusion, regulations of glucose and lipid metabolic processes were enhanced in parent pigeon livers from terminal phases of incubation to mid phase of chick rearing with sexual effects.
Collapse
Affiliation(s)
- X P Wan
- Feed Science Institute, College of Animal Science Zhejiang University, Hangzhou, China
| | - P Xie
- Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, China.,College of Life Science, Huaiyin Normal University, Huaian, China
| | - Z Bu
- Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, China
| | - X T Zou
- Feed Science Institute, College of Animal Science Zhejiang University, Hangzhou, China
| |
Collapse
|
29
|
Goddard ET, Hill RC, Nemkov T, D'Alessandro A, Hansen KC, Maller O, Mongoue-Tchokote S, Mori M, Partridge AH, Borges VF, Schedin P. The Rodent Liver Undergoes Weaning-Induced Involution and Supports Breast Cancer Metastasis. Cancer Discov 2017; 7:177-187. [PMID: 27974414 PMCID: PMC5459606 DOI: 10.1158/2159-8290.cd-16-0822] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 12/08/2016] [Accepted: 12/08/2016] [Indexed: 01/01/2023]
Abstract
Patients with postpartum breast cancer are at increased risk for metastasis compared with age-matched nulliparous or pregnant patients. Here, we address whether circulating tumor cells have a metastatic advantage in the postpartum host and find the postlactation rodent liver preferentially supports metastasis. Upon weaning, we observed liver weight loss, hepatocyte apoptosis, extracellular matrix remodeling including deposition of collagen and tenascin-C, and myeloid cell influx, data consistent with weaning-induced liver involution and establishment of a prometastatic microenvironment. Using intracardiac and intraportal metastasis models, we observed increased liver metastasis in post-weaning BALB/c mice compared with nulliparous controls. Human relevance is suggested by a ∼3-fold increase in liver metastasis in patients with postpartum breast cancer (n = 564) and by liver-specific tropism (n = 117). In sum, our data reveal a previously unknown biology of the rodent liver, weaning-induced liver involution, which may provide insight into the increased liver metastasis and poor prognosis of women diagnosed with postpartum breast cancer. SIGNIFICANCE We find that patients with postpartum breast cancer are at elevated risk for liver metastasis. We identify a previously unrecognized biology, namely weaning-induced liver involution, that establishes a prometastatic microenvironment, and which may account in part for the poor prognosis of patients with postpartum breast cancer. Cancer Discov; 7(2); 177-87. ©2016 AACR.This article is highlighted in the In This Issue feature, p. 115.
Collapse
Affiliation(s)
- Erica T Goddard
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon
| | - Ryan C Hill
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado
| | - Travis Nemkov
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, Colorado
| | - Ori Maller
- Department of Surgery, Center for Bioengineering and Tissue Regeneration, University of California, San Francisco, San Francisco, California
| | | | - Motomi Mori
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
- School of Public Health, Oregon Health & Science University, Portland, Oregon
| | - Ann H Partridge
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Virginia F Borges
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, Colorado.
- University of Colorado Cancer Center, Aurora, Colorado
- Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Pepper Schedin
- Department of Cell, Developmental and Cancer Biology, Oregon Health & Science University, Portland, Oregon.
- Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon
- Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
30
|
Cross JC. Adaptability and potential for treatment of placental functions to improve embryonic development and postnatal health. Reprod Fertil Dev 2017; 28:75-82. [PMID: 27062876 DOI: 10.1071/rd15342] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
For an organ that is so critical for life in eutherian mammals, the placenta hardly gets the attention that it deserves. The placenta does a series of remarkable things, including implanting the embryo in the uterus, negotiating with the mother for nutrients but also protecting her health during pregnancy, helping establish normal metabolic and cardiovascular function for life postnatally (developmental programming) and initiating changes that prepare the mother to care for and suckle her young after birth. Different lines of evidence in experimental animals suggest that the development and function of the placenta are adaptable. This means that some of the changes observed in pathological pregnancies may represent attempts to mitigate the impact of fetal growth and development. Key and emerging concepts are reviewed here concerning how we may view the placenta diagnostically and therapeutically in pregnancy complications, focusing on information from experimental studies in mice, sheep and cattle, as well as association studies from humans. Hundreds of different genes have been shown to underlie normal placental development and function, some of which have promise as tractable targets for intervention in pregnancies at risk for poor fetal growth.
Collapse
Affiliation(s)
- James C Cross
- Departments of Comparative Biology and Experimental Medicine, Biochemistry and Molecular Biology, Medical Genetics, and Obstetrics and Gynecology, University of Calgary, Calgary, Alberta T2N 4N1, Canada.
| |
Collapse
|
31
|
Kim J, Kim J, Kwon YH. Effects of disturbed liver growth and oxidative stress of high-fat diet-fed dams on cholesterol metabolism in offspring mice. Nutr Res Pract 2016; 10:386-92. [PMID: 27478544 PMCID: PMC4958640 DOI: 10.4162/nrp.2016.10.4.386] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 02/19/2016] [Accepted: 04/11/2016] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND/OBJECTIVES Changes in nutritional status during gestation and lactation have detrimental effects on offspring metabolism. Several animal studies have shown that maternal high-fat diet (HFD) can predispose the offspring to development of obesity and metabolic diseases, however the mechanisms underlying these transgenerational effects are poorly understood. Therefore, we examined the effect of maternal HFD consumption on metabolic phenotype and hepatic expression of involved genes in dams to determine whether any of these parameters were associated with the metabolic outcomes in the offspring. MATERIALS/METHODS Female C57BL/6 mice were fed a low-fat diet (LFD: 10% calories from fat) or a high-fat diet (HFD: 45% calories from fat) for three weeks before mating, and during pregnancy and lactation. Dams and their male offspring were studied at weaning. RESULTS Dams fed an HFD had significantly higher body and adipose tissue weights and higher serum triglyceride and cholesterol levels than dams fed an LFD. Hepatic lipid levels and mRNA levels of genes involved in lipid metabolism, including LXRα, SREBP-2, FXR, LDLR, and ABCG8 were significantly changed by maternal HFD intake. Significantly lower total liver DNA and protein contents were observed in dams fed an HFD, implicating the disturbed liver adaptation in the pregnancy-related metabolic demand. HFD feeding also induced significant oxidative stress in serum and liver of dams. Offspring of dams fed an HFD had significantly higher serum cholesterol levels, which were negatively correlated with liver weights of dams and positively correlated with hepatic lipid peroxide levels in dams. CONCLUSIONS Maternal HFD consumption induced metabolic dysfunction, including altered liver growth and oxidative stress in dams, which may contribute to the disturbed cholesterol homeostasis in the early life of male mice offspring.
Collapse
Affiliation(s)
- Juyoung Kim
- Department of Food and Nutrition, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea
| | - Juhae Kim
- Department of Food and Nutrition, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea
| | - Young Hye Kwon
- Department of Food and Nutrition, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea.; Research Institute of Human Ecology, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
32
|
A Transcriptomic Study of Maternal Thyroid Adaptation to Pregnancy in Rats. Int J Mol Sci 2015; 16:27339-49. [PMID: 26580608 PMCID: PMC4661888 DOI: 10.3390/ijms161126030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2015] [Revised: 11/03/2015] [Accepted: 11/10/2015] [Indexed: 01/22/2023] Open
Abstract
Thyroid disorders are relatively frequently observed in pregnant women. However, the impact of pregnancy on maternal thyroid has not been systematically evaluated. In the present study, using the rat as an animal model, we observed that the weight of maternal thyroid increased by about 18% in late pregnancy. To gain an insight into the molecular mechanisms, we took advantage of RNA-seq approaches to investigate global gene expression changes in the maternal thyroid. We identified a total of 615 differentially expressed genes, most of which (558 genes or 90.7%) were up-regulated in late pregnancy compared to the non-pregnant control. Gene ontology analysis showed that genes involved in cell cycle and metabolism were significantly enriched among up-regulated genes. Unexpectedly, pathway analysis revealed that expression levels for key components of the thyroid hormone synthesis pathway were not significantly altered. In addition, by examining of the promoter regions of up-regulated genes, we identified MAZ (MYC-associated zinc finger protein) and TFCP2 (transcription factor CP2) as two causal transcription factors. Our study contributes to an increase in the knowledge on the maternal thyroid adaptation to pregnancy.
Collapse
|
33
|
Rawn SM, Huang C, Hughes M, Shaykhutdinov R, Vogel HJ, Cross JC. Pregnancy Hyperglycemia in Prolactin Receptor Mutant, but Not Prolactin Mutant, Mice and Feeding-Responsive Regulation of Placental Lactogen Genes Implies Placental Control of Maternal Glucose Homeostasis. Biol Reprod 2015; 93:75. [PMID: 26269505 DOI: 10.1095/biolreprod.115.132431] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 07/27/2015] [Indexed: 12/23/2022] Open
Abstract
Pregnancy is often viewed as a conflict between the fetus and mother over metabolic resources. Insulin resistance occurs in mothers during pregnancy but does not normally lead to diabetes because of an increase in the number of the mother's pancreatic beta cells. In mice, this increase is dependent on prolactin (Prl) receptor signaling but the source of the ligand has been unclear. Pituitary-derived Prl is produced during the first half of pregnancy in mice but the placenta produces Prl-like hormones from implantation to term. Twenty-two separate mouse genes encode the placenta Prl-related hormones, making it challenging to assess their roles in knockout models. However, because at least four of them are thought to signal through the Prl receptor, we analyzed Prlr mutant mice and compared their phenotypes with those of Prl mutants. We found that whereas Prlr mutants develop hyperglycemia during gestation, Prl mutants do not. Serum metabolome analysis showed that Prlr mutants showed other changes consistent with diabetes. Despite the metabolic changes, fetal growth was normal in Prlr mutants. Of the four placenta-specific, Prl-related hormones that have been shown to interact with the Prlr, their gene expression localizes to different endocrine cell types. The Prl3d1 gene is expressed by trophoblast giant cells both in the labyrinth layer, sitting on the arterial side where maternal blood is highest in oxygen and nutrients, and in the junctional zone as maternal blood leaves the placenta. Expression increases during the night, though the increase in the labyrinth is circadian whereas it occurs only after feeding in the junctional zone. These data suggest that the placenta has a sophisticated endocrine system that regulates maternal glucose metabolism during pregnancy.
Collapse
Affiliation(s)
- Saara M Rawn
- Department of Comparative Biology & Experimental Medicine, University of Calgary, Calgary, Alberta, Canada Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, Alberta, Canada
| | - Carol Huang
- Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada
| | - Martha Hughes
- Department of Comparative Biology & Experimental Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Rustem Shaykhutdinov
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - Hans J Vogel
- Department of Biological Sciences, University of Calgary, Calgary, Alberta, Canada
| | - James C Cross
- Department of Comparative Biology & Experimental Medicine, University of Calgary, Calgary, Alberta, Canada Department of Biochemistry & Molecular Biology, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
34
|
Oca FGGMD, López-González MDL, Escobar-Wilches DC, Chavira-Ramírez R, Sierra-Santoyo A. Vinclozolin modulates hepatic cytochrome P450 isoforms during pregnancy. Reprod Toxicol 2015; 53:119-26. [DOI: 10.1016/j.reprotox.2015.04.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2014] [Revised: 04/16/2015] [Accepted: 04/20/2015] [Indexed: 12/15/2022]
|
35
|
Casey T, Patel OV, Plaut K. Transcriptomes reveal alterations in gravity impact circadian clocks and activate mechanotransduction pathways with adaptation through epigenetic change. Physiol Genomics 2015; 47:113-28. [PMID: 25649141 DOI: 10.1152/physiolgenomics.00117.2014] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 02/03/2015] [Indexed: 12/21/2022] Open
Abstract
Few studies have investigated the impact of alterations in gravity on mammalian transcriptomes. Here, we describe the impact of spaceflight on mammary transcriptome of late pregnant rats and the effect of hypergravity exposure on mammary, liver, and adipose transcriptomes in late pregnancy and at the onset of lactation. RNA was isolated from mammary collected on pregnancy day 20 from rats exposed to spaceflight from days 11 to 20 of gestation. To measure the impact of hypergravity on mammary, liver, and adipose transcriptomes we isolated RNA from tissues collected on P20 and lactation day 1 from rats exposed to hypergravity beginning on pregnancy day 9. Gene expression was measured with Affymetrix GeneChips. Microarray analysis of variance revealed alterations in gravity affected the expression of genes that regulate circadian clocks and activate mechanotransduction pathways. Changes in these systems may explain global gene expression changes in immune response, metabolism, and cell proliferation. Expression of genes that modify chromatin structure and methylation was affected, suggesting adaptation to gravity alterations may proceed through epigenetic change. Altered gravity experiments offer insights into the role of forces omnipresent on Earth that shape genomes in heritable ways. Our study is the first to analyze the impact of alterations in gravity on transcriptomes of pregnant and lactating mammals. Findings provide insight into systems that sense gravity and the way in which they affect phenotype, as well as the possibility of sustaining life beyond Earth's orbit.
Collapse
Affiliation(s)
- Theresa Casey
- Department of Animal Science, Purdue University, West Lafayette, Indiana; and
| | - Osman V Patel
- Department of Cell and Molecular Biology, Grand Valley State University, Allendale, Michigan
| | - Karen Plaut
- Department of Animal Science, Purdue University, West Lafayette, Indiana; and
| |
Collapse
|
36
|
Vaughan OR, Fisher HM, Dionelis KN, Jeffreys EC, Higgins JS, Musial B, Sferruzzi-Perri AN, Fowden AL. Corticosterone alters materno-fetal glucose partitioning and insulin signalling in pregnant mice. J Physiol 2015; 593:1307-21. [PMID: 25625347 DOI: 10.1113/jphysiol.2014.287177] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 12/24/2014] [Indexed: 12/17/2022] Open
Abstract
Glucocorticoids affect glucose metabolism in adults and fetuses, although their effects on materno-fetal glucose partitioning remain unknown. The present study measured maternal hepatic glucose handling and placental glucose transport together with insulin signalling in these tissues in mice drinking corticosterone either from day (D) 11 to D16 or D14 to D19 of pregnancy (term = D21). On the final day of administration, corticosterone-treated mice were hyperinsulinaemic (P < 0.05) but normoglycaemic compared to untreated controls. In maternal liver, there was no change in glycogen content or glucose 6-phosphatase activity but increased Slc2a2 glucose transporter expression in corticosterone-treated mice, on D16 only (P < 0.05). On D19, but not D16, transplacental (3) H-methyl-d-glucose clearance was reduced by 33% in corticosterone-treated dams (P < 0.05). However, when corticosterone-treated animals were pair-fed to control intake, aiming to prevent the corticosterone-induced increase in food consumption, (3) H-methyl-d-glucose clearance was similar to the controls. Depending upon gestational age, corticosterone treatment increased phosphorylation of the insulin-signalling proteins, protein kinase B (Akt) and glycogen synthase-kinase 3β, in maternal liver (P < 0.05) but not placenta (P > 0.05). Insulin receptor and insulin-like growth factor type I receptor abundance did not differ with treatment in either tissue. Corticosterone upregulated the stress-inducible mechanistic target of rapamycin (mTOR) suppressor, Redd1, in liver (D16 and D19) and placenta (D19), in ad libitum fed animals (P < 0.05). Concomitantly, hepatic protein content and placental weight were reduced on D19 (P < 0.05), in association with altered abundance and/or phosphorylation of signalling proteins downstream of mTOR. Taken together, the data indicate that maternal glucocorticoid excess reduces fetal growth partially by altering placental glucose transport and mTOR signalling.
Collapse
Affiliation(s)
- O R Vaughan
- Centre for Trophoblast Research, Department of Physiology, Development and Neuroscience, University of Cambridge, Cambridge, CB2 3EG, UK
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Zou Y, Hu M, Bao Q, Chan JY, Dai G. Nrf2 participates in regulating maternal hepatic adaptations to pregnancy. J Cell Sci 2013; 126:1618-25. [PMID: 23418358 DOI: 10.1242/jcs.118109] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Pregnancy induces widespread adaptive responses in maternal organ systems including the liver. The maternal liver exhibits significant growth by increasing the number and size of hepatocytes, by largely unknown mechanisms. Nrf2 mediates cellular defense against oxidative stress and inflammation and also regulates liver regeneration. To determine whether Nrf2 is involved in the regulation of maternal hepatic adaptations to pregnancy, we assessed the proliferation and size of maternal hepatocytes and the associated molecular events in wild-type and Nrf2-null mice at various stages of gestation. We found that wild-type maternal hepatocytes underwent proliferation and size reduction during the first half, and size increase without overt replication during the second half, of pregnancy. Although pregnancy decreased Nrf2 activity in the maternal liver, Nrf2 deficiency caused a delay in maternal hepatocyte proliferation, concomitant with dysregulation of the activation of Cyclin D1, E1, and, more significantly, A2. Remarkably, as a result of Nrf2 absence, the maternal hepatocytes were largely prevented from reducing their sizes during the first half of pregnancy, which was associated with an increase in mTOR activation. During the second half of pregnancy, maternal hepatocytes of both genotypes showed continuous volume increase accompanied by persistent activation of mTOR. However, the lack of Nrf2 resulted in dysregulation of the activation of the mTOR upstream regulator AKT1 and the mTOR target p70SK6 and thus disruption of the AKT1/mTOR/p70S6K pathway, which is known to control cell size. This suggests an mTOR-dependent and AKT1- and p70S6K-independent compensatory mechanism when Nrf2 is deficient. In summary, our study demonstrates that Nrf2 is required for normal maternal hepatic adjustments to pregnancy by ensuring proper regulation of the number and size of maternal hepatocytes.
Collapse
Affiliation(s)
- Yuhong Zou
- Department of Biology, School of Science, Center for Regenerative Biology and Medicine, Indiana University-Purdue University, Indianapolis, IN 46202, USA
| | | | | | | | | |
Collapse
|
38
|
Dai G, Bustamante JJ, Zou Y, Myronovych A, Bao Q, Kumar S, Soares MJ. Maternal hepatic growth response to pregnancy in the mouse. Exp Biol Med (Maywood) 2011; 236:1322-32. [PMID: 21969712 DOI: 10.1258/ebm.2011.011076] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Pregnancy is characterized by physiological adjustments in the maternal compartment. In this investigation, the influence of pregnancy on maternal liver was examined in CD-1 mice. Dramatic changes were observed in the size of the maternal liver during pregnancy. Livers doubled in weight from the non-pregnant state to day 18 of pregnancy. The pregnancy-induced hepatomegaly was a physiological event of liver growth confirmed by DNA content increase and detection of hepatocyte hyperplasia and hypertrophy. Growth of the liver was initiated following implantation and peaked at parturition. The expression and/or activities of key genes known to regulate liver regeneration, a phenomenon of liver growth compensatory to liver mass loss, were investigated. The results showed that pregnancy-dependent liver growth was associated with interleukin (IL)-6, tumor necrosis factor α, c-Jun and IL-1β, but independent of hepatocyte growth factor, fibroblast growth factor 1, tumor necrosis factor receptor 1, constitutive androstane receptor and pregnane X receptor. Furthermore, maternal liver growth was associated with the activation of hepatic signal transducer and activator of transcription 3, β-catenin and epidermal growth factor receptor, but pregnancy did not activate hepatic c-Met. The findings suggest that the molecular mechanisms regulating pregnancy-induced liver growth and injury-induced liver regeneration exhibit overlapping features but are not identical. In summary, the liver of the mouse adapts to the demands of pregnancy via a dramatic growth response driven by hepatocyte proliferation and size increase.
Collapse
Affiliation(s)
- Guoli Dai
- Department of Biology, Center for Regenerative Biology and Medicine, School of Science, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Papacleovoulou G, Abu-Hayyeh S, Williamson C. Nuclear receptor-driven alterations in bile acid and lipid metabolic pathways during gestation. Biochim Biophys Acta Mol Basis Dis 2010; 1812:879-87. [PMID: 21073948 DOI: 10.1016/j.bbadis.2010.11.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2010] [Revised: 11/02/2010] [Accepted: 11/03/2010] [Indexed: 12/27/2022]
Abstract
Nuclear receptor signalling is essential for physiological processes such as metabolism, development, and reproduction. Alterations in the endocrine state that naturally occur during pregnancy result in maternal adaptations to support the feto-placental unit. A series of studies have shown that nuclear receptor signalling is involved in maternal adaptations of bile acid, cholesterol, and lipid homeostasis pathways to ensure maintenance of the nutritional demands of the fetus. We discuss regulation of hepatic nuclear receptors and their target genes in pregnancy and their impact on the development of disorders such as intrahepatic cholestasis of pregnancy and oestrogen-induced hepatotoxicity. This article is part of a Special Issue entitled: Translating nuclear receptors from health to disease.
Collapse
Affiliation(s)
- Georgia Papacleovoulou
- Imperial College London, Maternal and Fetal Disease Group, Institute of Reproductive and Developmental Biology, Du Cane Road, London W12 0NN, UK
| | | | | |
Collapse
|