51
|
Daoust L, Choi BSY, Lacroix S, Rodrigues Vilela V, Varin TV, Dudonné S, Pilon G, Roy D, Levy E, Desjardins Y, Chassaing B, Marette A. The postnatal window is critical for the development of sex-specific metabolic and gut microbiota outcomes in offspring. Gut Microbes 2022; 13:2004070. [PMID: 34812123 PMCID: PMC8632343 DOI: 10.1080/19490976.2021.2004070] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The Developmental Origins of Health and Disease (DOHaD) concept has been proposed to explain the influence of environmental conditions during critical developmental stages on the risk of diseases in adulthood. The aim of this study was to compare the impact of the prenatal vs. postnatal environment on the gut microbiota in dams during the preconception, gestation and lactation periods and their consequences on metabolic outcomes in offspring. Here we used the cross-fostering technique, e.g. the exchange of pups following birth to a foster dam, to decipher the metabolic effects of the intrauterine versus postnatal environmental exposures to a polyphenol-rich cranberry extract (CE). CE administration to high-fat high-sucrose (HFHS)-fed dams improved glucose homeostasis and reduced liver steatosis in association with a shift in the maternal gut microbiota composition. Unexpectedly, we observed that the postnatal environment contributed to metabolic outcomes in female offspring, as revealed by adverse effects on adiposity and glucose metabolism, while no effect was observed in male offspring. In addition to the strong sexual dimorphism, we found a significant influence of the nursing mother on the community structure of the gut microbiota based on α-diversity and β-diversity indices in offspring. Gut microbiota transplantation (GMT) experiments partly reproduced the observed phenotype in female offspring. Our data support the concept that the postnatal environment represents a critical window to influence future sex-dependent metabolic outcomes in offspring that are causally but partly linked with gut microbiome alterations.
Collapse
Affiliation(s)
- Laurence Daoust
- Quebec Heart and Lung Institute Research Center, Quebec, Montreal, Canada,Institute of Nutrition and Functional Food, Laval University, Quebec, Montreal, Canada
| | - Béatrice S.-Y. Choi
- Quebec Heart and Lung Institute Research Center, Quebec, Montreal, Canada,Institute of Nutrition and Functional Food, Laval University, Quebec, Montreal, Canada
| | - Sébastien Lacroix
- Institute of Nutrition and Functional Food, Laval University, Quebec, Montreal, Canada,Canada Research Excellence Chair in the Microbiome-Endocannabinoïdome Mediators Axis in Metabolic Health (Cerc-mend), Laval University, Quebec, Montreal, Canada
| | - Vanessa Rodrigues Vilela
- Quebec Heart and Lung Institute Research Center, Quebec, Montreal, Canada,Institute of Nutrition and Functional Food, Laval University, Quebec, Montreal, Canada
| | - Thibault Vincent Varin
- Quebec Heart and Lung Institute Research Center, Quebec, Montreal, Canada,Institute of Nutrition and Functional Food, Laval University, Quebec, Montreal, Canada
| | - Stéphanie Dudonné
- Institute of Nutrition and Functional Food, Laval University, Quebec, Montreal, Canada
| | - Geneviève Pilon
- Quebec Heart and Lung Institute Research Center, Quebec, Montreal, Canada,Institute of Nutrition and Functional Food, Laval University, Quebec, Montreal, Canada
| | - Denis Roy
- Institute of Nutrition and Functional Food, Laval University, Quebec, Montreal, Canada
| | - Emile Levy
- Institute of Nutrition and Functional Food, Laval University, Quebec, Montreal, Canada,Chu Sainte-Justine Research Center, Montreal University, Montreal, Canada
| | - Yves Desjardins
- Institute of Nutrition and Functional Food, Laval University, Quebec, Montreal, Canada
| | - Benoit Chassaing
- Inserm U1016, Team “Mucosal Microbiota in Chronic Inflammatory Diseases”, Cnrs Umr 8104, Université De Paris, Paris, France
| | - André Marette
- Quebec Heart and Lung Institute Research Center, Quebec, Montreal, Canada,Institute of Nutrition and Functional Food, Laval University, Quebec, Montreal, Canada,CONTACT André Marette Cardiology Axis of the Quebec Heart and Lung Institute, Laval University, QuébecG1V 0A6, Canada
| |
Collapse
|
52
|
Almeida JI, Tenreiro MF, Martinez-Santamaria L, Guerrero-Aspizua S, Gisbert JP, Alves PM, Serra M, Baptista PM. Hallmarks of the human intestinal microbiome on liver maturation and function. J Hepatol 2022; 76:694-725. [PMID: 34715263 DOI: 10.1016/j.jhep.2021.10.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/05/2021] [Accepted: 10/17/2021] [Indexed: 12/18/2022]
Abstract
As one of the most metabolically complex systems in the body, the liver ensures multi-organ homeostasis and ultimately sustains life. Nevertheless, during early postnatal development, the liver is highly immature and takes about 2 years to acquire and develop almost all of its functions. Different events occurring at the environmental and cellular levels are thought to mediate hepatic maturation and function postnatally. The crosstalk between the liver, the gut and its microbiome has been well appreciated in the context of liver disease, but recent evidence suggests that the latter could also be critical for hepatic function under physiological conditions. The gut-liver crosstalk is thought to be mediated by a rich repertoire of microbial metabolites that can participate in a myriad of biological processes in hepatic sinusoids, from energy metabolism to tissue regeneration. Studies on germ-free animals have revealed the gut microbiome as a critical contributor in early hepatic programming, and this influence extends throughout life, mediating liver function and body homeostasis. In this seminar, we describe the microbial molecules that have a known effect on the liver and discuss how the gut microbiome and the liver evolve throughout life. We also provide insights on current and future strategies to target the gut microbiome in the context of hepatology research.
Collapse
Affiliation(s)
- Joana I Almeida
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain; Instituto de Biologia Experimental e Tecnológica (iBET), Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), Oeiras, Portugal
| | - Miguel F Tenreiro
- Instituto de Biologia Experimental e Tecnológica (iBET), Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), Oeiras, Portugal
| | - Lucía Martinez-Santamaria
- Carlos III University of Madrid. Bioengineering and Aerospace Engineering, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER, ISCIII), Madrid, Spain; Instituto de Investigación Sanitaria Fundación Jiménez Díaz, Madrid, Spain
| | - Sara Guerrero-Aspizua
- Carlos III University of Madrid. Bioengineering and Aerospace Engineering, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER, ISCIII), Madrid, Spain
| | - Javier P Gisbert
- Gastroenterology Department. Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-IP), Universidad Autónoma de Madrid (UAM), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Paula M Alves
- Instituto de Biologia Experimental e Tecnológica (iBET), Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), Oeiras, Portugal
| | - Margarida Serra
- Instituto de Biologia Experimental e Tecnológica (iBET), Oeiras, Portugal; Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa (ITQB NOVA), Oeiras, Portugal
| | - Pedro M Baptista
- Instituto de Investigación Sanitaria Aragón (IIS Aragón), Zaragoza, Spain; Carlos III University of Madrid. Bioengineering and Aerospace Engineering, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain; Fundación ARAID, Zaragoza, Spain.
| |
Collapse
|
53
|
Thompson MD, Kang J, Faerber A, Hinrichs H, Özler O, Cowen J, Xie Y, Tarr PI, Davidson NO. Maternal obesogenic diet regulates offspring bile acid homeostasis and hepatic lipid metabolism via the gut microbiome in mice. Am J Physiol Gastrointest Liver Physiol 2022; 322:G295-G309. [PMID: 34984925 PMCID: PMC8816615 DOI: 10.1152/ajpgi.00247.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Mice exposed in gestation to maternal high-fat/high-sucrose (HF/HS) diet develop altered bile acid (BA) homeostasis. We hypothesized that these reflect an altered microbiome and asked if microbiota transplanted from HF/HS offspring change hepatic BA and lipid metabolism to determine the directionality of effect. Female mice were fed HF/HS or chow (CON) for 6 wk and bred with lean males. 16S sequencing was performed to compare taxa in offspring. Cecal microbiome transplantation (CMT) was performed from HF/HS or CON offspring into antibiotic-treated mice fed chow or high fructose. BA, lipid metabolic, and gene expression analyses were performed in recipient mice. Gut microbiomes from HF/HS offspring segregated from CON offspring, with increased Firmicutes to Bacteriodetes ratios and Verrucomicrobial abundance. After CMT was performed, HF/HS-recipient mice had larger BA pools, increased intrahepatic muricholic acid, and decreased deoxycholic acid species. HF/HS-recipient mice exhibited downregulated hepatic Mrp2, increased hepatic Oatp1b2, and decreased ileal Asbt mRNA expression. HF/HS-recipient mice exhibited decreased cecal butyrate and increased hepatic expression of Il6. HF/HS-recipient mice had larger livers and increased intrahepatic triglyceride versus CON-recipient mice after fructose feeding, with increased hepatic mRNA expression of lipogenic genes including Srebf1, Fabp1, Mogat1, and Mogat2. CMT from HF/HS offspring increased BA pool and shifted the composition of the intrahepatic BA pool. CMT from HF/HS donor offspring increased fructose-induced liver triglyceride accumulation. These findings support a causal role for vertical transfer of an altered microbiome in hepatic BA and lipid metabolism in HF/HS offspring.NEW & NOTEWORTHY We utilized a mouse model of maternal obesogenic diet exposure to evaluate the effect on offspring microbiome and bile acid homeostasis. We identified shifts in the offspring microbiome associated with changes in cecal bile acid levels. Transfer of the microbiome from maternal obesogenic diet-exposed offspring to microbiome-depleted mice altered bile acid homeostasis and increased fructose-induced hepatic steatosis.
Collapse
Affiliation(s)
- Michael D. Thompson
- 1Division of Endocrinology and Diabetes, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Jisue Kang
- 1Division of Endocrinology and Diabetes, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Austin Faerber
- 1Division of Endocrinology and Diabetes, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Holly Hinrichs
- 1Division of Endocrinology and Diabetes, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Oğuz Özler
- 1Division of Endocrinology and Diabetes, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Jamie Cowen
- 1Division of Endocrinology and Diabetes, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Yan Xie
- 2Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| | - Phillip I. Tarr
- 3Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri
| | - Nicholas O. Davidson
- 2Division of Gastroenterology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
54
|
Hsu CN, Yu HR, Chan JYH, Wu KLH, Lee WC, Tain YL. The Impact of Gut Microbiome on Maternal Fructose Intake-Induced Developmental Programming of Adult Disease. Nutrients 2022; 14:nu14051031. [PMID: 35268005 PMCID: PMC8912426 DOI: 10.3390/nu14051031] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 02/24/2022] [Accepted: 02/26/2022] [Indexed: 12/11/2022] Open
Abstract
Excessive or insufficient maternal nutrition can influence fetal development and the susceptibility of offspring to adult disease. As eating a fructose-rich diet is becoming more common, the effects of maternal fructose intake on offspring health is of increasing relevance. The gut is required to process fructose, and a high-fructose diet can alter the gut microbiome, resulting in gut dysbiosis and metabolic disorders. Current evidence from animal models has revealed that maternal fructose consumption causes various components of metabolic syndrome in adult offspring, while little is known about how gut microbiome is implicated in fructose-induced developmental programming and the consequential risks for developing chronic disease in offspring. This review will first summarize the current evidence supporting the link between fructose and developmental programming of adult diseases. This will be followed by presenting how gut microbiota links to common mechanisms underlying fructose-induced developmental programming. We also provide an overview of the reprogramming effects of gut microbiota-targeted therapy on fructose-induced developmental programming and how this approach may prevent adult-onset disease. Using gut microbiota-targeted therapy to prevent maternal fructose diet-induced developmental programming, we have the potential to mitigate the global burden of fructose-related disorders.
Collapse
Affiliation(s)
- Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Hong-Ren Yu
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan;
| | - Julie Y. H. Chan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (J.Y.H.C.); (K.L.H.W.)
| | - Kay L. H. Wu
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (J.Y.H.C.); (K.L.H.W.)
| | - Wei-Chia Lee
- Department of Urology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan;
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan;
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833, Taiwan; (J.Y.H.C.); (K.L.H.W.)
- Correspondence: ; Tel.: +886-975-056-995; Fax: +886-7733-8009
| |
Collapse
|
55
|
Souza AFP, Woyames J, Miranda RA, Oliveira LS, Caetano B, Martins IL, Souza MS, Andrade CBV, Bento-Bernardes T, Bloise FF, Fortunato RS, Trevenzoli IH, Souza LL, Pazos-Moura CC. Maternal Isocaloric High-Fat Diet Induces Liver Mitochondria Maladaptations and Homeostatic Disturbances Intensifying Mitochondria Damage in Response to Fructose Intake in Adult Male Rat Offspring. Mol Nutr Food Res 2022; 66:e2100514. [PMID: 35175665 DOI: 10.1002/mnfr.202100514] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 01/04/2022] [Indexed: 11/10/2022]
Abstract
SCOPE Perinatal maternal obesity and excessive fructose consumption have been associated with liver metabolic diseases. We investigated whether moderate maternal high-fat diet affects the liver mitochondria responses to fructose intake in adult offspring. METHODS AND RESULTS Wistar female rats received a standard diet (mSTD) or high-fat diet (mHFD) (9% and 28.6% fat, respectively), before mating until the end of lactation. Male offspring were fed standard diet from weaning to adulthood and received water or fructose-drinking water (15%) from 120 to 150 days old. Fructose induced liver mitochondrial ultrastructural alterations with higher intensity in mHFD offspring, accompanied by reduced autophagy markers. Isolated mitochondria respirometry showed unaltered ATP-coupled oxygen consumption with increased Atp5f1b mRNA only in mHFD offspring. Fructose increased basal respiration and encoding complex I-III mRNA, only in mSTD offspring. Uncoupled respiration was lower in mHFD mitochondria that were unable to exhibit fructose-induced increase Ucp2 mRNA. Fructose decreased antioxidative defense markers, increased unfolded protein response and insulin resistance only in mHFD offspring without fructose-induced hepatic lipid accumulation. CONCLUSION Mitochondrial dysfunction and homeostatic disturbances in response to fructose are early events evidencing the higher risk of fructose damage in the liver of adult offspring from dams fed an isocaloric moderate high-fat diet. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Aline F P Souza
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | - Juliana Woyames
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | - Rosiane A Miranda
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | - Lorraine S Oliveira
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | - Bruna Caetano
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | - Isabela L Martins
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | - Manuella S Souza
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | - Cherley B V Andrade
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | - Thais Bento-Bernardes
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | - Flavia F Bloise
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | - Rodrigo S Fortunato
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | - Isis H Trevenzoli
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | - Luana L Souza
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, RJ, Brazil
| | - Carmen C Pazos-Moura
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, RJ, Brazil
| |
Collapse
|
56
|
Valentini F, Rocchi G, Vespasiani-Gentilucci U, Guarino MPL, Altomare A, Carotti S. The Origins of NAFLD: The Potential Implication of Intrauterine Life and Early Postnatal Period. Cells 2022; 11:cells11030562. [PMID: 35159371 PMCID: PMC8834011 DOI: 10.3390/cells11030562] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 01/30/2022] [Accepted: 02/02/2022] [Indexed: 02/04/2023] Open
Abstract
Fetal life and the first few months after birth represent a plastic age, defined as a “window of opportunity”, as the organism is particularly susceptible to environmental pressures and has to adapt to environmental conditions. Several perturbations in pregnancy, such as excessive weight gain, obesity, gestational diabetes mellitus and an inadequate or high-fat diet, have been associated with long-term metabolic consequences in offspring, even without affecting birth weight. Moreover, great interest has also been focused on the relationship between the gut microbiome of early infants and health status in later life. Consistently, in various epidemiological studies, a condition of dysbiosis has been associated with an increased inflammatory response and metabolic alterations in the host, with important consequences on the intestinal and systemic health of the unborn child. This review aims to summarize the current knowledge on the origins of NAFLD, with particular attention to the potential implications of intrauterine life and the early postnatal period. Due to the well-known association between gut microbiota and the risk of NAFLD, a specific focus will be devoted to factors affecting early microbiota formation/composition.
Collapse
Affiliation(s)
- Francesco Valentini
- Pediatric Unit, Sant’Andrea Hospital, Faculty of Medicine and Psychology, “Sapienza” University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy;
| | - Giulia Rocchi
- Unit of Food Science and Human Nutrition, Campus Biomedico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy;
| | - Umberto Vespasiani-Gentilucci
- Unit of Internal Medicine and Hepatology, Fondazione Policlinico Campus Biomedico of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy;
| | - Michele Pier Luca Guarino
- Gastroenterology Unit, Fondazione Policlinico Campus Biomedico of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy;
| | - Annamaria Altomare
- Gastroenterology Unit, Fondazione Policlinico Campus Biomedico of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy;
- Correspondence:
| | - Simone Carotti
- Unit of Microscopic and Ultrastructural Anatomy, Campus Biomedico University of Rome, Via Alvaro del Portillo 21, 00128 Rome, Italy;
| |
Collapse
|
57
|
Korsmo HW, Dave B, Trasino S, Saxena A, Liu J, Caviglia JM, Edwards K, Dembitzer M, Sheeraz S, Khaldi S, Jiang X. Maternal Choline Supplementation and High-Fat Feeding Interact to Influence DNA Methylation in Offspring in a Time-Specific Manner. Front Nutr 2022; 9:841787. [PMID: 35165655 PMCID: PMC8837519 DOI: 10.3389/fnut.2022.841787] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 01/07/2022] [Indexed: 11/18/2022] Open
Abstract
Maternal methyl donor supplementation during pregnancy has demonstrated lasting influence on offspring DNA methylation. However, it is unknown whether an adverse postnatal environment, such as high-fat (HF) feeding, overrides the influence of prenatal methyl donor supplementation on offspring epigenome. In this study, we examined whether maternal supplementation of choline (CS), a methyl donor, interacts with prenatal and postnatal HF feeding to alter global and site-specific DNA methylation in offspring. We fed wild-type C57BL/6J mouse dams a HF diet with or without CS throughout gestation. After weaning, the offspring were exposed to HF feeding for 6 weeks resembling a continued obesogenic environment. Our results suggest that maternal CS under the HF condition (HFCS) increased global DNA methylation and DNA methyltransferase 1 (Dnmt1) expression in both fetal liver and brain. However, during the postnatal period, HFCS offspring demonstrated lower global DNA methylation and Dnmt1 expression was unaltered in both the liver and visceral adipose tissue. Site-specific DNA methylation analysis during both fetal and postnatal periods demonstrated that HFCS offspring had higher methylation of CpGs in the promoter of Srebf1, a key mediator of de novo lipogenesis. In conclusion, the influence of maternal CS on offspring DNA methylation is specific to HF feeding status during prenatal and postnatal periods. Without continued CS during the postnatal period, global DNA methylation enhanced by prenatal CS in the offspring was overridden by postnatal HF feeding.
Collapse
Affiliation(s)
- Hunter W. Korsmo
- Department of Biochemistry, The Graduate Center, City University of New York (CUNY), New York, NY, United States
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY, United States
| | - Bhoomi Dave
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY, United States
| | - Steven Trasino
- School of Urban Public Health, Hunter College of the CUNY, New York, NY, United States
| | - Anjana Saxena
- Department of Biochemistry, The Graduate Center, City University of New York (CUNY), New York, NY, United States
- Department of Biology, Brooklyn College of the City University of New York, Brooklyn, NY, United States
| | - Jia Liu
- Advanced Science Research Center at the Graduate Center of the CUNY, New York, NY, United States
| | - Jorge Matias Caviglia
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY, United States
| | - Kaydine Edwards
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY, United States
| | - Moshe Dembitzer
- Department of Biology, Brooklyn College of the City University of New York, Brooklyn, NY, United States
| | - Shameera Sheeraz
- Department of Biology, Brooklyn College of the City University of New York, Brooklyn, NY, United States
| | - Sarah Khaldi
- Department of Biology, Brooklyn College of the City University of New York, Brooklyn, NY, United States
| | - Xinyin Jiang
- Department of Biochemistry, The Graduate Center, City University of New York (CUNY), New York, NY, United States
- Department of Health and Nutrition Sciences, Brooklyn College of the City University of New York, Brooklyn, NY, United States
| |
Collapse
|
58
|
Moeckli B, Delaune V, Prados J, Tihy M, Peloso A, Oldani G, Delmi T, Slits F, Gex Q, Rubbia-Brandt L, Goossens N, Lacotte S, Toso C. Impact of Maternal Obesity on Liver Disease in the Offspring: A Comprehensive Transcriptomic Analysis and Confirmation of Results in a Murine Model. Biomedicines 2022; 10:biomedicines10020294. [PMID: 35203502 PMCID: PMC8869223 DOI: 10.3390/biomedicines10020294] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/16/2022] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
The global obesity epidemic particularly affects women of reproductive age. Offspring of obese mothers suffer from an increased risk of liver disease but the molecular mechanisms involved remain unknown. We performed an integrative genomic analysis of datasets that investigated the impact of maternal obesity on the hepatic gene expression profile of the offspring in mice. Furthermore, we developed a murine model of maternal obesity and studied the development of liver disease and the gene expression profile of the top dysregulated genes by quantitative real-time polymerase chain reaction (qPCR). Our data are available for interactive exploration on our companion webpage. We identified five publicly available datasets relevant to our research question. Pathways involved in metabolism, the innate immune system, the clotting cascade, and the cell cycle were consistently dysregulated in the offspring of obese mothers. Concerning genes involved in the development of liver disease, Egfr, Vegfb, Wnt2,Pparg and six other genes were dysregulated in multiple independent datasets. In our own model, we observed a higher tendency towards the development of non-alcoholic liver disease (60 vs. 20%) and higher levels of alanine aminotransferase (41.0 vs. 12.5 IU/l, p = 0.008) in female offspring of obese mothers. Male offspring presented higher levels of liver fibrosis (2.4 vs. 0.6% relative surface area, p = 0.045). In a qPCR gene expression analysis of our own samples, we found Fgf21, Pparg, Ppard, and Casp6 to be dysregulated by maternal obesity. Maternal obesity represents a looming threat to the liver health of future generations. Our comprehensive transcriptomic analysis will help to better understand the mechanisms of the development of liver disease in the offspring of obese mothers and can give rise to further explorations.
Collapse
Affiliation(s)
- Beat Moeckli
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, Division of Visceral Surgery, University of Geneva, 1206 Geneva, Switzerland; (B.M.); (V.D.); (A.P.); (G.O.); (T.D.); (F.S.); (Q.G.); (C.T.)
- Department of Surgery, Division of Visceral Surgery, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Vaihere Delaune
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, Division of Visceral Surgery, University of Geneva, 1206 Geneva, Switzerland; (B.M.); (V.D.); (A.P.); (G.O.); (T.D.); (F.S.); (Q.G.); (C.T.)
- Department of Surgery, Division of Visceral Surgery, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Julien Prados
- Bioinformatics Support Platform, Services Communs de la Faculté, University of Geneva, 1206 Geneva, Switzerland;
| | - Matthieu Tihy
- Division of Clinical Pathology, Geneva University Hospitals, 1205 Geneva, Switzerland; (M.T.); (L.R.-B.)
| | - Andrea Peloso
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, Division of Visceral Surgery, University of Geneva, 1206 Geneva, Switzerland; (B.M.); (V.D.); (A.P.); (G.O.); (T.D.); (F.S.); (Q.G.); (C.T.)
- Department of Surgery, Division of Visceral Surgery, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Graziano Oldani
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, Division of Visceral Surgery, University of Geneva, 1206 Geneva, Switzerland; (B.M.); (V.D.); (A.P.); (G.O.); (T.D.); (F.S.); (Q.G.); (C.T.)
- Department of Surgery, Division of Visceral Surgery, Geneva University Hospitals, 1205 Geneva, Switzerland
| | - Thomas Delmi
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, Division of Visceral Surgery, University of Geneva, 1206 Geneva, Switzerland; (B.M.); (V.D.); (A.P.); (G.O.); (T.D.); (F.S.); (Q.G.); (C.T.)
| | - Florence Slits
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, Division of Visceral Surgery, University of Geneva, 1206 Geneva, Switzerland; (B.M.); (V.D.); (A.P.); (G.O.); (T.D.); (F.S.); (Q.G.); (C.T.)
| | - Quentin Gex
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, Division of Visceral Surgery, University of Geneva, 1206 Geneva, Switzerland; (B.M.); (V.D.); (A.P.); (G.O.); (T.D.); (F.S.); (Q.G.); (C.T.)
| | - Laura Rubbia-Brandt
- Division of Clinical Pathology, Geneva University Hospitals, 1205 Geneva, Switzerland; (M.T.); (L.R.-B.)
| | - Nicolas Goossens
- Division of Gastroenterology, Geneva University Hospitals, 1205 Geneva, Switzerland;
| | - Stéphanie Lacotte
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, Division of Visceral Surgery, University of Geneva, 1206 Geneva, Switzerland; (B.M.); (V.D.); (A.P.); (G.O.); (T.D.); (F.S.); (Q.G.); (C.T.)
- Correspondence:
| | - Christian Toso
- Hepatology and Transplantation Laboratory, Department of Surgery, Faculty of Medicine, Division of Visceral Surgery, University of Geneva, 1206 Geneva, Switzerland; (B.M.); (V.D.); (A.P.); (G.O.); (T.D.); (F.S.); (Q.G.); (C.T.)
- Department of Surgery, Division of Visceral Surgery, Geneva University Hospitals, 1205 Geneva, Switzerland
| |
Collapse
|
59
|
Dreisbach C, Morgan H, Cochran C, Gyamfi A, Henderson WA, Prescott S. Metabolic and Microbial Changes Associated With Diet and Obesity During Pregnancy: What Can We Learn From Animal Studies? Front Cell Infect Microbiol 2022; 11:795924. [PMID: 35118010 PMCID: PMC8804207 DOI: 10.3389/fcimb.2021.795924] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/13/2021] [Indexed: 12/22/2022] Open
Abstract
The intestinal microbiota changes throughout pregnancy and influences maternal metabolic adaptations to support fetal growth. Obesity induces alterations to the microbiota that include decreased microbial diversity and shifts in microbial composition, though specific species changes are inconsistent between published studies. In animal models, probiotics and exercise moderate maternal weight gain and partially correct the maternal microbiota. Supplemental Escherichia coli, however, exacerbate maternal obesity during the perinatal period, lending weight to the theory that inflammation-induced gut epithelial barrier leak influences metabolic dysregulation. Although birth weight is not always altered when offspring are exposed to an obesogenic diet during gestation, insulin resistance and lipid metabolism are impacted through adulthood in association with this exposure and can lead to increased body weight in adulthood. Postnatal offspring growth is accelerated in response to maternal overnutrition during lactation. Offspring microbiota, metabolism, and behavior are altered in response to early exposure to high fat and high sucrose diets. Consequences to this exposure include impaired glucose and insulin homeostasis, fatty liver, and neurobehavioral deficits that can be ameliorated by improving the microbial environment. In this mini review, we provide an overview of the use of translational animal models to understand the mechanisms associated with changes to the gastrointestinal microbiota due to maternal obesity and the microbial impact on the metabolic changes of pregnancy.
Collapse
Affiliation(s)
- Caitlin Dreisbach
- Data Science Institute, Columbia University, New York, NY, United States
| | - Hailey Morgan
- College of Nursing, University of South Florida, Tampa, FL, United States
| | - Caroline Cochran
- School of Nursing, Columbia University, New York, NY, United States
| | - Adwoa Gyamfi
- School of Medicine, University of Connecticut, Farmington, CT, United States
| | - Wendy Ann Henderson
- School of Medicine, University of Connecticut, Farmington, CT, United States
- School of Nursing, University of Connecticut, Storrs, CT, United States
| | - Stephanie Prescott
- College of Nursing, University of South Florida, Tampa, FL, United States
- *Correspondence: Stephanie Prescott,
| |
Collapse
|
60
|
Sugino KY, Mandala A, Janssen RC, Gurung S, Trammell M, Day MW, Brush RS, Papin JF, Dyer DW, Agbaga MP, Friedman JE, Castillo-Castrejon M, Jonscher KR, Myers DA. Western diet-induced shifts in the maternal microbiome are associated with altered microRNA expression in baboon placenta and fetal liver. FRONTIERS IN CLINICAL DIABETES AND HEALTHCARE 2022; 3:945768. [PMID: 36935840 PMCID: PMC10012127 DOI: 10.3389/fcdhc.2022.945768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Maternal consumption of a high-fat, Western-style diet (WD) disrupts the maternal/infant microbiome and contributes to developmental programming of the immune system and nonalcoholic fatty liver disease (NAFLD) in the offspring. Epigenetic changes, including non-coding miRNAs in the fetus and/or placenta may also underlie this risk. We previously showed that obese nonhuman primates fed a WD during pregnancy results in the loss of beneficial maternal gut microbes and dysregulation of cellular metabolism and mitochondrial dysfunction in the fetal liver, leading to a perturbed postnatal immune response with accelerated NAFLD in juvenile offspring. Here, we investigated associations between WD-induced maternal metabolic and microbiome changes, in the absence of obesity, and miRNA and gene expression changes in the placenta and fetal liver. After ~8-11 months of WD feeding, dams were similar in body weight but exhibited mild, systemic inflammation (elevated CRP and neutrophil count) and dyslipidemia (increased triglycerides and cholesterol) compared with dams fed a control diet. The maternal gut microbiome was mainly comprised of Lactobacillales and Clostridiales, with significantly decreased alpha diversity (P = 0.0163) in WD-fed dams but no community-wide differences (P = 0.26). At 0.9 gestation, mRNA expression of IL6 and TNF in maternal WD (mWD) exposed placentas trended higher, while increased triglycerides, expression of pro-inflammatory CCR2, and histological evidence for fibrosis were found in mWD-exposed fetal livers. In the mWD-exposed fetus, hepatic expression levels of miR-204-5p and miR-145-3p were significantly downregulated, whereas in mWD-exposed placentas, miR-182-5p and miR-183-5p were significantly decreased. Notably, miR-1285-3p expression in the liver and miR-183-5p in the placenta were significantly associated with inflammation and lipid synthesis pathway genes, respectively. Blautia and Ruminococcus were significantly associated with miR-122-5p in liver, while Coriobacteriaceae and Prevotellaceae were strongly associated with miR-1285-3p in the placenta; both miRNAs are implicated in pathways mediating postnatal growth and obesity. Our findings demonstrate that mWD shifts the maternal microbiome, lipid metabolism, and inflammation prior to obesity and are associated with epigenetic changes in the placenta and fetal liver. These changes may underlie inflammation, oxidative stress, and fibrosis patterns that drive NAFLD and metabolic disease risk in the next generation.
Collapse
Affiliation(s)
- Kameron Y. Sugino
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Ashok Mandala
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Rachel C. Janssen
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Sunam Gurung
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - MaJoi Trammell
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Michael W. Day
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Richard S. Brush
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - James F. Papin
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - David W. Dyer
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Martin-Paul Agbaga
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Jacob E. Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Marisol Castillo-Castrejon
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Karen R. Jonscher
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- CORRESPONDENCE: Karen R. Jonscher,
| | - Dean A. Myers
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| |
Collapse
|
61
|
Peral-Sanchez I, Hojeij B, Ojeda DA, Steegers-Theunissen RPM, Willaime-Morawek S. Epigenetics in the Uterine Environment: How Maternal Diet and ART May Influence the Epigenome in the Offspring with Long-Term Health Consequences. Genes (Basel) 2021; 13:31. [PMID: 35052371 PMCID: PMC8774448 DOI: 10.3390/genes13010031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 12/15/2021] [Accepted: 12/20/2021] [Indexed: 12/12/2022] Open
Abstract
The societal burden of non-communicable disease is closely linked with environmental exposures and lifestyle behaviours, including the adherence to a poor maternal diet from the earliest preimplantation period of the life course onwards. Epigenetic variations caused by a compromised maternal nutritional status can affect embryonic development. This review summarises the main epigenetic modifications in mammals, especially DNA methylation, histone modifications, and ncRNA. These epigenetic changes can compromise the health of the offspring later in life. We discuss different types of nutritional stressors in human and animal models, such as maternal undernutrition, seasonal diets, low-protein diet, high-fat diet, and synthetic folic acid supplement use, and how these nutritional exposures epigenetically affect target genes and their outcomes. In addition, we review the concept of thrifty genes during the preimplantation period, and some examples that relate to epigenetic change and diet. Finally, we discuss different examples of maternal diets, their effect on outcomes, and their relationship with assisted reproductive technology (ART), including their implications on epigenetic modifications.
Collapse
Affiliation(s)
- Irene Peral-Sanchez
- Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (D.A.O.); (S.W.-M.)
| | - Batoul Hojeij
- Department Obstetrics and Gynecology, Erasmus MC, University Medical Center, 3000 CA Rotterdam, The Netherlands; (B.H.); (R.P.M.S.-T.)
| | - Diego A. Ojeda
- Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK; (D.A.O.); (S.W.-M.)
| | - Régine P. M. Steegers-Theunissen
- Department Obstetrics and Gynecology, Erasmus MC, University Medical Center, 3000 CA Rotterdam, The Netherlands; (B.H.); (R.P.M.S.-T.)
| | | |
Collapse
|
62
|
Dai X, Wang C, Guo Z, Li Y, Liu T, Jin G, Wang S, Wang B, Jiang K, Cao H. Maternal sucralose exposure induces Paneth cell defects and exacerbates gut dysbiosis of progeny mice. Food Funct 2021; 12:12634-12646. [PMID: 34821899 DOI: 10.1039/d1fo02921e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Research has shown that maternal sucralose (MS) exposure alters the gut microbiota of offspring at weaning and predisposes the offspring to developing obesity, non-alcoholic fatty liver disease and metabolic syndrome later in life. However, the underlying mechanism remains unclear. Paneth cells are thought to critically influence the gut microbiota. This study aimed to investigate whether MS exposure induced Paneth cell defects and exacerbated gut dysbiosis of offspring. Female C57BL/6 mice were divided into the MS and control (water) groups during pregnancy and lactation. Progeny mice were fed a normal sucralose-free diet after weaning until adulthood. MS inhibited intestinal development and increased the expression of proinflammatory cytokines in the small intestines of 3-week-old progeny mice. MS increased the proportions of abnormal granule secretion by Paneth cells. The number of Paneth cells and mRNA expression of AMPs such as cryptdins and lysozyme were reduced in the MS group. MS disturbed the gut microbiota composition and diversity in the 3-week-old offspring mice. The relative abundances of pro-inflammatory bacteria, such as Desulfovibrionales, Helicobacter, Pasteurellales and Campylobacterales were significantly increased in the MS group, while anti-inflammatory bacteria, including Clostridium XI, were decreased. This dysbiosis continued into adulthood. These findings showed that MS exposure induced Paneth cell defects and exacerbated gut dysbiosis in offspring mice. Sucralose should be consumed with caution, especially during pregnancy and in early life.
Collapse
Affiliation(s)
- Xin Dai
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Chen Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Zixuan Guo
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Yun Li
- Department of Pharmacy, General Hospital, Tianjin Medical University, Tianjin, China
| | - Tianyu Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Ge Jin
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Sinan Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Kui Jiang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive Diseases, Tianjin, China.
| |
Collapse
|
63
|
Hsu CN, Hou CY, Hsu WH, Tain YL. Early-Life Origins of Metabolic Syndrome: Mechanisms and Preventive Aspects. Int J Mol Sci 2021; 22:11872. [PMID: 34769303 PMCID: PMC8584419 DOI: 10.3390/ijms222111872] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 10/31/2021] [Accepted: 10/31/2021] [Indexed: 02/07/2023] Open
Abstract
One of the leading global public-health burdens is metabolic syndrome (MetS), despite the many advances in pharmacotherapies. MetS, now known as "developmental origins of health and disease" (DOHaD), can have its origins in early life. Offspring MetS can be programmed by various adverse early-life conditions, such as nutrition imbalance, maternal conditions or diseases, maternal chemical exposure, and medication use. Conversely, early interventions have shown potential to revoke programming processes to prevent MetS of developmental origins, namely reprogramming. In this review, we summarize what is currently known about adverse environmental insults implicated in MetS of developmental origins, including the fundamental underlying mechanisms. We also describe animal models that have been developed to study the developmental programming of MetS. This review extends previous research reviews by addressing implementation of reprogramming strategies to prevent the programming of MetS. These mechanism-targeted strategies include antioxidants, melatonin, resveratrol, probiotics/prebiotics, and amino acids. Much work remains to be accomplished to determine the insults that could induce MetS, to identify the mechanisms behind MetS programming, and to develop potential reprogramming strategies for clinical translation.
Collapse
Affiliation(s)
- Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Chih-Yao Hou
- Department of Seafood Science, National Kaohsiung University of Science and Technology, Kaohsiung 811, Taiwan;
| | - Wei-Hsuan Hsu
- Department of Food Safety/Hygiene and Risk Management, College of Medicine, National Chen Kung University, Tainan 701, Taiwan;
| | - You-Lin Tain
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
| |
Collapse
|
64
|
Li Y, Pollock CA, Saad S. Aberrant DNA Methylation Mediates the Transgenerational Risk of Metabolic and Chronic Disease Due to Maternal Obesity and Overnutrition. Genes (Basel) 2021; 12:genes12111653. [PMID: 34828259 PMCID: PMC8624316 DOI: 10.3390/genes12111653] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/02/2021] [Accepted: 10/17/2021] [Indexed: 12/13/2022] Open
Abstract
Maternal obesity is a rapidly evolving universal epidemic leading to acute and long-term medical and obstetric health issues, including increased maternal risks of gestational diabetes, hypertension and pre-eclampsia, and the future risks for offspring's predisposition to metabolic diseases. Epigenetic modification, in particular DNA methylation, represents a mechanism whereby environmental effects impact on the phenotypic expression of human disease. Maternal obesity or overnutrition contributes to the alterations in DNA methylation during early life which, through fetal programming, can predispose the offspring to many metabolic and chronic diseases, such as non-alcoholic fatty liver disease, obesity, diabetes, and chronic kidney disease. This review aims to summarize findings from human and animal studies, which support the role of maternal obesity in fetal programing and the potential benefit of altering DNA methylation to limit maternal obesity related disease in the offspring.
Collapse
Affiliation(s)
- Yan Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China;
| | - Carol A. Pollock
- Kolling Institute of Medical Research, University of Sydney, Sydney, NSW 2065, Australia;
| | - Sonia Saad
- Kolling Institute of Medical Research, University of Sydney, Sydney, NSW 2065, Australia;
- Correspondence:
| |
Collapse
|
65
|
Dietary Patterns Influence Target Gene Expression through Emerging Epigenetic Mechanisms in Nonalcoholic Fatty Liver Disease. Biomedicines 2021; 9:biomedicines9091256. [PMID: 34572442 PMCID: PMC8468830 DOI: 10.3390/biomedicines9091256] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 12/12/2022] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) refers to the pathologic buildup of extra fat in the form of triglycerides in liver cells without excessive alcohol intake. NAFLD became the most common cause of chronic liver disease that is tightly associated with key aspects of metabolic disorders, including insulin resistance, obesity, diabetes, and metabolic syndrome. It is generally accepted that multiple mechanisms and pathways are involved in the pathogenesis of NAFLD. Heredity, sedentary lifestyle, westernized high sugar saturated fat diet, metabolic derangements, and gut microbiota, all may interact on a on genetically susceptible individual to cause the disease initiation and progression. While there is an unquestionable role for gene-diet interaction in the etiopathogenesis of NAFLD, it is increasingly apparent that epigenetic processes can orchestrate many aspects of this interaction and provide additional mechanistic insight. Exciting research demonstrated that epigenetic alterations in chromatin can influence gene expression chiefly at the transcriptional level in response to unbalanced diet, and therefore predispose an individual to NAFLD. Thus, further discoveries into molecular epigenetic mechanisms underlying the link between nutrition and aberrant hepatic gene expression can yield new insights into the pathogenesis of NAFLD, and allow innovative epigenetic-based strategies for its early prevention and targeted therapies. Herein, we outline the current knowledge of the interactive role of a high-fat high-calories diet and gene expression through DNA methylation and histone modifications on the pathogenesis of NAFLD. We also provide perspectives on the advancement of the epigenomics in the field and possible shortcomings and limitations ahead.
Collapse
|
66
|
Li J, Wan Y, Zheng Z, Zhang H, Li Y, Guo X, Li K, Li D. Maternal n-3 polyunsaturated fatty acids restructure gut microbiota of offspring mice and decrease their susceptibility to mammary gland cancer. Food Funct 2021; 12:8154-8168. [PMID: 34291263 DOI: 10.1039/d1fo00906k] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Our previous studies have revealed that a maternal diet rich in n-3 polyunsaturated fatty acids (PUFAs) is associated with decreased mammary cancer risk in offspring. However, the underlying mechanism remains unclear. The present study aimed to investigate the possible mechanism by which maternal n-3 PUFAs decrease the mammary cancer risk of offspring in terms of gut microbiota. C57BL/6 pregnant mice were fed a control standard chow (CON), fish oil supplemented diet (n-3 Sup-FO), flaxseed oil supplemented diet (n-3 Sup-FSO) or n-3 PUFA deficient diet (n-3 Def) (n = 10) throughout gestation and lactation. After weaning, all offspring were fed a AIN-93G diet. The tumor incidence and volume were significantly increased in n-3 Def offspring compared with the other groups. Maternal n-3 PUFA supplementation resulted in a significantly increased α-diversity of the gut microbiota in n-3 Sup-FO and n-3 Sup-FSO offspring compared with that in n-3 Def offspring. The relative abundances of Akkermansia, Lactobacillus and Mucispirillum observed in adult offspring of both the n-3 Sup-FO and n-3 Sup-FSO groups were higher than those observed in the control group, whereas the maternal n-3 Def diet was associated with decreased abundances of Lactobacillus, Bifidobacterium and Barnesiella in 7-week-old offspring. The levels of the pro-inflammatory factors IL-1β, IL-6 and TNF-α were significantly lower in n-3 PUFA supplemented offspring than in n-3 Def offspring. In addition, the abundance of Mucispirillum was positively associated with the concentration of the anti-inflammatory factor IL-10, whereas the abundances of Bifidobacterium and Akkermansia were negatively associated with IL-1β and IL-6, respectively. Based on the bacterial composition of the gut microbiota, metabolites were predicted and the results showed that arachidonic acid metabolism and the MAPK signaling pathways were more enriched, while the butyric acid metabolic pathway was less enriched in offspring of the n-3 Def group than in those of the other three groups. Our findings suggest that decreased pro-inflammatory factors and changed gut microbiota are associated with the protective effects of maternal n-3 PUFAs against offspring's mammary tumorigenesis.
Collapse
Affiliation(s)
- Jiaomei Li
- Institute of Nutrition and Health, Qingdao University, 308 Ningxia Road, Qingdao 266071, China.
| | | | | | | | | | | | | | | |
Collapse
|
67
|
Cardiovascular Diseases of Developmental Origins: Preventive Aspects of Gut Microbiota-Targeted Therapy. Nutrients 2021; 13:nu13072290. [PMID: 34371800 PMCID: PMC8308390 DOI: 10.3390/nu13072290] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/24/2021] [Accepted: 06/30/2021] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular diseases (CVDs) can originate from early life. Accumulating evidence suggests that gut microbiota in early life is linked to CVDs in later life. Gut microbiota-targeted therapy has gained significant importance in recent decades for its health-promoting role in the prevention (rather than just treatment) of CVDs. Thus far, available gut microbiota-based treatment modalities used as reprogramming interventions include probiotics, prebiotics, and postbiotics. The purpose of this review is, first, to highlight current studies that link dysbiotic gut microbiota to the developmental origins of CVD. This is followed by a summary of the connections between the gut microbiota and CVD behind cardiovascular programming, such as short chain fatty acids (SCFAs) and their receptors, trimethylamine-N-oxide (TMAO), uremic toxins, and aryl hydrocarbon receptor (AhR), and the renin-angiotensin system (RAS). This review also presents an overview of how gut microbiota-targeted reprogramming interventions can prevent the developmental origins of CVD from animal studies. Overall, this review reveals that recent advances in gut microbiota-targeted therapy might provide the answers to reduce the global burden of CVDs. Still, additional studies will be needed to put research findings into practice.
Collapse
|
68
|
Tokuhara D. Role of the Gut Microbiota in Regulating Non-alcoholic Fatty Liver Disease in Children and Adolescents. Front Nutr 2021; 8:700058. [PMID: 34250000 PMCID: PMC8267179 DOI: 10.3389/fnut.2021.700058] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Accepted: 06/04/2021] [Indexed: 01/18/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common form of chronic liver disease in children and adolescents. Although obesity is the leading cause of NAFLD, the etiologies of NAFLD are multifactorial (e.g., high-fat diet, a lack of exercise, gender, maternal obesity, the antibiotic use), and each of these factors leads to dysbiosis of the gut microbiota community. The gut microbiota is a key player in the development and regulation of the gut mucosal immune system as well as the regulation of both NAFLD and obesity. Dysbiosis of the gut microbiota promotes the development of NAFLD via alteration of gut-liver homeostasis, including disruption of the gut barrier, portal transport of bacterial endotoxin (lipopolysaccharide) to the liver, altered bile acid profiles, and decreased concentrations of short-chain fatty acids. In terms of prevention and treatment, conventional approaches (e.g., dietary and exercise interventions) against obesity and NAFLD have been confirmed to recover the dysbiosis and dysbiosis-mediated altered metabolism. In addition, increased understanding of the importance of gut microbiota-mediated homeostasis in the prevention of NAFLD suggests the potential effectiveness of gut microbiota-targeted preventive and therapeutic strategies (e.g., probiotics and fecal transplantation) against NAFLD in children and adolescents. This review comprehensively summarizes our current knowledge of the gut microbiota, focusing on its interaction with NAFLD and its potential therapeutic role in obese children and adolescents with this disorder.
Collapse
Affiliation(s)
- Daisuke Tokuhara
- Department of Pediatrics, Osaka City University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
69
|
El-Sayed A, Aleya L, Kamel M. The link among microbiota, epigenetics, and disease development. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:28926-28964. [PMID: 33860421 DOI: 10.1007/s11356-021-13862-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 04/06/2021] [Indexed: 06/12/2023]
Abstract
The microbiome is a community of various microorganisms that inhabit or live on the skin of humans/animals, sharing the body space with their hosts. It is a sort of complex ecosystem of trillions of commensals, symbiotic, and pathogenic microorganisms, including trillions of bacteria, archaea, protozoa, fungi, and viruses. The microbiota plays a role in the health and disease status of the host. Their number, species dominance, and viability are dynamic. Their long-term disturbance is usually accompanied by serious diseases such as metabolic disorders, cardiovascular diseases, or even cancer. While epigenetics is a term that refers to different stimuli that induce modifications in gene expression patterns without structural changes in the inherited DNA sequence, these changes can be reversible or even persist for several generations. Epigenetics can be described as cell memory that stores experience against internal and external factors. Results from multiple institutions have contributed to the role and close interaction of both microbiota and epigenetics in disease induction. Understanding the mechanisms of both players enables a better understanding of disease induction and development and also opens the horizon to revolutionary therapeutic approaches. The present review illustrates the roles of diet, microbiome, and epigenetics in the induction of several chronic diseases. In addition, it discusses the application of epigenetic data to develop diagnostic biomarkers and therapeutics and evaluate their safety for patients. Understanding the interaction among all these elements enables the development of innovative preventive/therapeutic approaches for disease control.
Collapse
Affiliation(s)
- Amr El-Sayed
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Lotfi Aleya
- Chrono-Environnement Laboratory, UMR CNRS 6249, Bourgogne Franche-Comté University, F-25030, Besançon Cedex, France
| | - Mohamed Kamel
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt.
| |
Collapse
|
70
|
Peng H, Xu H, Wu J, Li J, Zhou Y, Ding Z, Siwko SK, Yuan X, Schalinske KL, Alpini G, Zhang KK, Xie L. Maternal high-fat diet disrupted one-carbon metabolism in offspring, contributing to nonalcoholic fatty liver disease. Liver Int 2021; 41:1305-1319. [PMID: 33529448 PMCID: PMC8137550 DOI: 10.1111/liv.14811] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/12/2021] [Accepted: 01/28/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND & AIMS Pregnant women may transmit their metabolic phenotypes to their offspring, enhancing the risk for nonalcoholic fatty liver disease (NAFLD); however, the molecular mechanisms remain unclear. METHODS Prior to pregnancy female mice were fed either a maternal normal-fat diet (NF-group, "no effectors"), or a maternal high-fat diet (HF-group, "persistent effectors"), or were transitioned from a HF to a NF diet before pregnancy (H9N-group, "effectors removal"), followed by pregnancy and lactation, and then offspring were fed high-fat diets after weaning. Offspring livers were analysed by functional studies, as well as next-generation sequencing for gene expression profiles and DNA methylation changes. RESULTS The HF, but not the H9N offspring, displayed glucose intolerance and hepatic steatosis. The HF offspring also displayed a disruption of lipid homeostasis associated with an altered methionine cycle and abnormal one-carbon metabolism that caused DNA hypermethylation and L-carnitine depletion associated with deactivated AMPK signalling and decreased expression of PPAR-α and genes for fatty acid oxidation. These changes were not present in H9N offspring. In addition, we identified maternal HF diet-induced genes involved in one-carbon metabolism that were associated with DNA methylation modifications in HF offspring. Importantly, the DNA methylation modifications and their associated gene expression changes were reversed in H9N offspring livers. CONCLUSIONS Our results demonstrate for the first time that maternal HF diet disrupted the methionine cycle and one-carbon metabolism in offspring livers which further altered lipid homeostasis. CpG islands of specific genes involved in one-carbon metabolism modified by different maternal diets were identified.
Collapse
Affiliation(s)
- Hui Peng
- Department of Nutrition, Texas A&M University, College Station, TX,Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Huiting Xu
- Department of Pathology, University of North Dakota, Grand Forks, North Dakota,Hubei Cancer Hospital, Wuhan, Hubei, China
| | - Jie Wu
- Institute of Biosciences & Technology, Texas A&M University, Houston, TX
| | - Jiangyuan Li
- Department of Nutrition, Texas A&M University, College Station, TX,Department of Statistics, Texas A&M University, College Station, TX
| | - Yi Zhou
- Department of Nutrition, Texas A&M University, College Station, TX,Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zehuan Ding
- Department of Nutrition, Texas A&M University, College Station, TX
| | - Stefan K. Siwko
- Institute of Biosciences & Technology, Texas A&M University, Houston, TX
| | - Xianglin Yuan
- Tongji Hospital, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Kevin L. Schalinske
- Department of Food Science and Human Nutrition, Iowa State University, Ames, IA
| | - Gianfranco Alpini
- Richard L. Roudebush VA Medical Center, and Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Ke K. Zhang
- Department of Nutrition, Texas A&M University, College Station, TX,Institute of Biosciences & Technology, Texas A&M University, Houston, TX,Department of Pathology, University of North Dakota, Grand Forks, North Dakota,Co-corresponding author: These authors contributed equally to this work
| | - Linglin Xie
- Department of Nutrition, Texas A&M University, College Station, TX,Co-corresponding author: These authors contributed equally to this work
| |
Collapse
|
71
|
Kasper P, Breuer S, Hoffmann T, Vohlen C, Janoschek R, Schmitz L, Appel S, Fink G, Hünseler C, Quaas A, Demir M, Lang S, Steffen HM, Martin A, Schramm C, Bürger M, Mahabir E, Goeser T, Dötsch J, Hucklenbruch-Rother E, Bae-Gartz I. Maternal Exercise Mediates Hepatic Metabolic Programming via Activation of AMPK-PGC1α Axis in the Offspring of Obese Mothers. Cells 2021; 10:1247. [PMID: 34069390 PMCID: PMC8158724 DOI: 10.3390/cells10051247] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/11/2021] [Accepted: 05/11/2021] [Indexed: 12/13/2022] Open
Abstract
Maternal obesity is associated with an increased risk of hepatic metabolic dysfunction for both mother and offspring and targeted interventions to address this growing metabolic disease burden are urgently needed. This study investigates whether maternal exercise (ME) could reverse the detrimental effects of hepatic metabolic dysfunction in obese dams and their offspring while focusing on the AMP-activated protein kinase (AMPK), representing a key regulator of hepatic metabolism. In a mouse model of maternal western-style-diet (WSD)-induced obesity, we established an exercise intervention of voluntary wheel-running before and during pregnancy and analyzed its effects on hepatic energy metabolism during developmental organ programming. ME prevented WSD-induced hepatic steatosis in obese dams by alterations of key hepatic metabolic processes, including activation of hepatic ß-oxidation and inhibition of lipogenesis following increased AMPK and peroxisome-proliferator-activated-receptor-γ-coactivator-1α (PGC-1α)-signaling. Offspring of exercised dams exhibited a comparable hepatic metabolic signature to their mothers with increased AMPK-PGC1α-activity and beneficial changes in hepatic lipid metabolism and were protected from WSD-induced adipose tissue accumulation and hepatic steatosis in later life. In conclusion, this study demonstrates that ME provides a promising strategy to improve the metabolic health of both obese mothers and their offspring and highlights AMPK as a potential metabolic target for therapeutic interventions.
Collapse
Affiliation(s)
- Philipp Kasper
- Clinic for Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (P.K.); (S.L.); (H.-M.S.); (A.M.); (C.S.); (M.B.); (T.G.)
| | - Saida Breuer
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Thorben Hoffmann
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Christina Vohlen
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Ruth Janoschek
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Lisa Schmitz
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Sarah Appel
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Gregor Fink
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Christoph Hünseler
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Alexander Quaas
- Department of Pathology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany;
| | - Münevver Demir
- Charité Campus Mitte and Campus Virchow Clinic, Department of Hepatology and Gastroenterology, Charité University Medicine Berlin, D-13353 Berlin, Germany;
| | - Sonja Lang
- Clinic for Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (P.K.); (S.L.); (H.-M.S.); (A.M.); (C.S.); (M.B.); (T.G.)
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Hans-Michael Steffen
- Clinic for Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (P.K.); (S.L.); (H.-M.S.); (A.M.); (C.S.); (M.B.); (T.G.)
| | - Anna Martin
- Clinic for Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (P.K.); (S.L.); (H.-M.S.); (A.M.); (C.S.); (M.B.); (T.G.)
| | - Christoph Schramm
- Clinic for Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (P.K.); (S.L.); (H.-M.S.); (A.M.); (C.S.); (M.B.); (T.G.)
| | - Martin Bürger
- Clinic for Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (P.K.); (S.L.); (H.-M.S.); (A.M.); (C.S.); (M.B.); (T.G.)
| | - Esther Mahabir
- Comparative Medicine, Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, D-50937 Cologne, Germany;
| | - Tobias Goeser
- Clinic for Gastroenterology and Hepatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (P.K.); (S.L.); (H.-M.S.); (A.M.); (C.S.); (M.B.); (T.G.)
| | - Jörg Dötsch
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Eva Hucklenbruch-Rother
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| | - Inga Bae-Gartz
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, D-50937 Cologne, Germany; (S.B.); (T.H.); (C.V.); (R.J.); (L.S.); (S.A.); (G.F.); (C.H.); (J.D.); (E.H.-R.)
| |
Collapse
|
72
|
Pregnancy and lactation after Roux-en-Y gastric bypass worsen nonalcoholic fatty liver disease in obese rats and lead to differential programming of hepatic de novo lipogenesis in offspring. J Dev Orig Health Dis 2021; 13:263-273. [PMID: 33998431 DOI: 10.1017/s2040174421000271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Maternal obesity increases the risk of nonalcoholic fatty liver disease (NAFLD) in offspring. The Roux-en-Y gastric bypass (RYBG) is effective for achieving weight loss and ameliorates NAFLD. To determine whether these benefits are maintained after pregnancy and/or lactation, and whether they modulate hepatic morphofunction in the next generation, we evaluated hepatic lipid metabolism in Western diet (WD)-obese female rats that underwent RYGB and in their F1 offspring at adulthood. Female Wistar rats consumed a WD from 21 to 130 days of age, before being submitted to RYGB (WD-RYGB-F0) or SHAM (WD-SHAM-F0) operations. After 5 weeks, these females were mated with control male breeders, and the male and female F1 offspring were identified as WD-RYGB-F1 and WD-SHAM-F1. WD-RYGB-F0 dams exhibited lower serum lipids levels, but severe hepatic steatosis and pathological features of advanced liver injury. The hepatic proteins involved in lipogenesis were reduced in WD-RYGB-F0, as were the genes related to β-oxidation and bile acids (BAs). Although the female and male WD-RYGB-F1 groups did not exhibit hepatic steatosis, the livers of female WD-RYGB-F1 demonstrated higher amounts of lipogenic genes and proteins, while male WD-RYGB-F1 presented a similar downregulation of lipogenic factors to that seen in WD-RYGB-F0 dams. In contrast, maternal and offspring groups of both sexes displayed reductions in the expressions of genes involved in BAs physiology and gluconeogenesis. As such, RYGB aggravates NAFLD after pregnancy and lactation and induces a gender-dependent differential expression of the hepatic lipogenesis pathway in offspring, indicating that female WD-RYGB-F1 may be an increased risk of developing NAFLD.
Collapse
|
73
|
Early-life nutrition and metabolic disorders in later life: a new perspective on energy metabolism. Chin Med J (Engl) 2021; 133:1961-1970. [PMID: 32826460 PMCID: PMC7462214 DOI: 10.1097/cm9.0000000000000976] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Type 2 diabetes mellitus and metabolic disorders have become an epidemic globally. However, the pathogenesis remains largely unclear and the prevention and treatment are still limited. In addition to environmental factors during adulthood, early life is the critical developmental window with high tissue plasticity, which might be modified by external environmental cues. Substantial evidence has demonstrated the vital role of early-life nutrition in programming the metabolic disorders in later life. In this review, we aim to overview the concepts of fetal programming and investigate the effects of early-life nutrition on energy metabolism in later life and the potential epigenetic mechanism. The related studies published on PubMed database up to March 2020 were included. The results showed that both maternal overnutrition and undernutrition increased the riskes of metabolic disorders in offspring and epigenetic modifications, including DNA methylation, miRNAs, and histone modification, might be the vital mediators. The beneficial effects of early-life lifestyle modifications as well as dietary and nutritional interventions on these deleterious metabolic remolding were initially observed. Overall, characterizing the early-life malnutrition that reshapes metabolic disease trajectories may yield novel targets for early prevention and intervention and provide a new point of view to the energy metabolism.
Collapse
|
74
|
Han S, Zhu F, Huang X, Yan P, Xu K, Shen F, Sun J, Yang Z, Jin G, Teng Y. Maternal obesity accelerated non-alcoholic fatty liver disease in offspring mice by reducing autophagy. Exp Ther Med 2021; 22:716. [PMID: 34007325 PMCID: PMC8120514 DOI: 10.3892/etm.2021.10148] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 03/02/2021] [Indexed: 12/14/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is a chronic liver disease characterized by an excessive accumulation of triacylglycerol in the liver. Autophagy is a lysosome-dependent degradation product recovery process, which widely occurs in eukaryotic cells, responsible for the vital maintenance of cellular energy balance. Previously published studies have demonstrated that autophagy is closely related to NAFLD occurrence and maternal obesity increases the susceptibility of offspring to non-alcoholic fatty liver disease, however, the underlying mechanism of this remains unclear. In the present study, NAFLD mouse models (offspring of an obese mother mouse via high-fat feeding) were generated, and the physiological indices of the liver were observed using total cholesterol, triglyceride, high-density lipoprotein and low-density lipoprotein serum assay kits. The morphological changes of the liver were also observed via HE, Masson and oil red O staining. Reverse transcription-quantitative-PCR and western blotting were performed to detect changes of autophagy-related genes in liver or fibrosis marker proteins (α-smooth muscle actin or TGF-β1). Changes in serum inflammatory cytokine IL-6 levels were determined via ELISA. The results of the present study demonstrated that the offspring of an obese mother were more likely to develop NALFD than the offspring of a chow-fed mother, due to their increased association with liver fibrosis. When feeding continued to 17 weeks, the worst cases of NAFLD were observed and the level of autophagy decreased significantly compared with the offspring of a normal weight mouse. In addition, after 17 weeks of feeding, compared with the offspring of a chow-fed mother, the offspring of an obese mouse mother had reduced adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK) phosphorylation levels and increased mammalian target of rapamycin (mTOR) phosphorylation levels. These results suggested that a reduced level of AMPK/mTOR mediated autophagy may be of vital importance for the increased susceptibility of offspring to NAFLD caused by maternal obesity. In conclusion, the current study provided a new direction for the treatment of NAFLD in offspring caused by maternal obesity.
Collapse
Affiliation(s)
- Shuguang Han
- Graduate School, Bengbu Medical College, Bengbu, Anhui 233000, P.R. China.,Department of Endocrinology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233000, P.R. China
| | - Feng Zhu
- Graduate School, Bengbu Medical College, Bengbu, Anhui 233000, P.R. China.,Department of Pediatrics, The Second Affiliated Hospital of Jiaxing University, Jiaxing Second Hospital, Jiaxing, Zhejiang 314000, P.R. China
| | - Xiaoxia Huang
- Department of Pediatrics, The Second Affiliated Hospital of Jiaxing University, Jiaxing Second Hospital, Jiaxing, Zhejiang 314000, P.R. China.,The Second Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310000, P.R. China
| | - Panpan Yan
- Graduate School, Bengbu Medical College, Bengbu, Anhui 233000, P.R. China.,Department of Pediatrics, The Second Affiliated Hospital of Jiaxing University, Jiaxing Second Hospital, Jiaxing, Zhejiang 314000, P.R. China
| | - Ke Xu
- Department of Pediatrics, The Second Affiliated Hospital of Jiaxing University, Jiaxing Second Hospital, Jiaxing, Zhejiang 314000, P.R. China
| | - Fangfang Shen
- Department of Pediatrics, The Second Affiliated Hospital of Jiaxing University, Jiaxing Second Hospital, Jiaxing, Zhejiang 314000, P.R. China
| | - Jiawen Sun
- Graduate School, Bengbu Medical College, Bengbu, Anhui 233000, P.R. China
| | - Zeyu Yang
- Graduate School, Bengbu Medical College, Bengbu, Anhui 233000, P.R. China
| | - Guoxi Jin
- Graduate School, Bengbu Medical College, Bengbu, Anhui 233000, P.R. China.,Department of Endocrinology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, Anhui 233000, P.R. China
| | - Yiqun Teng
- Graduate School, Bengbu Medical College, Bengbu, Anhui 233000, P.R. China.,Department of Pediatrics, The Second Affiliated Hospital of Jiaxing University, Jiaxing Second Hospital, Jiaxing, Zhejiang 314000, P.R. China
| |
Collapse
|
75
|
An update on the role of the microbiome in non-alcoholic fatty liver disease pathogenesis, diagnosis, and treatment. ACTA ACUST UNITED AC 2021; 18:270-280. [PMID: 33716495 DOI: 10.1007/s11938-020-00290-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The microbiome was originally postulated to contribute to the pathogenesis of NAFLD when the first studies of dysbiosis in NAFLD were reported. Since then, a number of studies have investigated this finding further, in order to discern whether the dysbiosis is the result of the metabolic dysregulation seen with NAFLD or a contributor to the pathogenesis of this condition.
Collapse
|
76
|
Saengnipanthkul S, Noh HL, Friedline RH, Suk S, Choi S, Acosta NK, Tran DA, Hu X, Inashima K, Kim AM, Lee KW, Kim JK. Maternal exposure to high-fat diet during pregnancy and lactation predisposes normal weight offspring mice to develop hepatic inflammation and insulin resistance. Physiol Rep 2021; 9:e14811. [PMID: 33769706 PMCID: PMC7995551 DOI: 10.14814/phy2.14811] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/01/2021] [Accepted: 03/04/2021] [Indexed: 11/24/2022] Open
Abstract
Increasing evidence shows a potential link between the perinatal nutrient environment and metabolic outcome in offspring. Here, we investigated the effects of maternal feeding of a high-fat diet (HFD) during the perinatal period on hepatic metabolism and inflammation in male offspring mice at weaning and in early adulthood. Female C57BL/6 J mice were fed HFD or normal chow (NC) for 4 weeks before mating and during pregnancy and lactation. The male offspring mice were weaned onto an NC diet, and metabolic and molecular experiments were performed in early adulthood. At postnatal day 21, male offspring mice from HFD-fed dams (Off-HFD) showed significant increases in whole body fat mass and fasting levels of glucose, insulin, and cholesterol compared to male offspring mice from NC-fed dams (Off-NC). The RT-qPCR analysis showed two- to fivefold increases in hepatic inflammatory markers (MCP-1, IL-1β, and F4/80) in Off-HFD mice. Hepatic expression of G6Pase and PEPCK was elevated by fivefold in the Off-HFD mice compared to the Off-NC mice. Hepatic expression of GLUT4, IRS-1, and PDK4, as well as lipid metabolic genes, CD36, SREBP1c, and SCD1 were increased in the Off-HFD mice compared to the Off-NC mice. In contrast, CPT1a mRNA levels were reduced by 60% in the Off-HFD mice. At postnatal day 70, despite comparable body weights to the Off-NC mice, Off-HFD mice developed hepatic inflammation with increased expression of MCP-1, CD68, F4/80, and CD36 compared to the Off-NC mice. Despite normal body weight, Off-HFD mice developed insulin resistance with defects in hepatic insulin action and insulin-stimulated glucose uptake in skeletal muscle and brown fat, and these metabolic effects were associated with hepatic inflammation in Off-HFD mice. Our findings indicate hidden, lasting effects of maternal exposure to HFD during pregnancy and lactation on metabolic homeostasis of normal weight offspring mice.
Collapse
Affiliation(s)
- Suchaorn Saengnipanthkul
- Division of NutritionDepartment of PediatricsFaculty of MedicineKhon Kaen UniversityKhon KaenThailand
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Hye Lim Noh
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Randall H. Friedline
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Sujin Suk
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Stephanie Choi
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Nicholas K. Acosta
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Duy A. Tran
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Xiaodi Hu
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Kunikazu Inashima
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Allison M. Kim
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
| | - Ki Won Lee
- Department of Agricultural BiotechnologyCollege of Agricultural and Life SciencesSeoul National UniversitySeoulSouth Korea
| | - Jason K. Kim
- Program in Molecular MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
- Division of Endocrinology, Metabolism, and DiabetesDepartment of MedicineUniversity of Massachusetts Medical SchoolWorcesterMAUSA
- Department of Agricultural BiotechnologyCollege of Agricultural and Life SciencesSeoul National UniversitySeoulSouth Korea
| |
Collapse
|
77
|
Sindi AS, Geddes DT, Wlodek ME, Muhlhausler BS, Payne MS, Stinson LF. Can we modulate the breastfed infant gut microbiota through maternal diet? FEMS Microbiol Rev 2021; 45:6133472. [PMID: 33571360 DOI: 10.1093/femsre/fuab011] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/09/2021] [Indexed: 12/11/2022] Open
Abstract
Initial colonisation of the infant gut is robustly influenced by regular ingestion of human milk, a substance that contains microbes, microbial metabolites, immune proteins, and oligosaccharides. Numerous factors have been identified as potential determinants of the human milk and infant gut microbiota, including maternal diet; however, there is limited data on the influence of maternal diet during lactation on either of these. Here, we review the processes thought to contribute to human milk and infant gut bacterial colonisation and provide a basis for considering the role of maternal dietary patterns during lactation in shaping infant gut microbial composition and function. Although only one observational study has directly investigated the influence of maternal diet during lactation on the infant gut microbiome, data from animal studies suggests that modulation of the maternal gut microbiota, via diet or probiotics, may influence the mammary or milk microbiota. Additionally, evidence from human studies suggests that the maternal diet during pregnancy may affect the gut microbiota of the breastfed infant. Together, there is a plausible hypothesis that maternal diet during lactation may influence the infant gut microbiota. If substantiated in further studies, this may present a potential window of opportunity for modulating the infant gut microbiome in early life.
Collapse
Affiliation(s)
- Azhar S Sindi
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia.,College of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Donna T Geddes
- School of Molecular Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Mary E Wlodek
- Department of Physiology, University of Melbourne, Melbourne, Victoria, Australia
| | - Beverly S Muhlhausler
- CSIRO, Adelaide, South Australia, Australia.,School of Agriculture, Food and Wine, The University of Adelaide, Adelaide, South Australia, Australia
| | - Matthew S Payne
- Division of Obstetrics and Gynaecology, The University of Western Australia, Perth, Western Australia, Australia
| | - Lisa F Stinson
- School of Molecular Sciences, The University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
78
|
Bennett H, Troutman TD, Sakai M, Glass CK. Epigenetic Regulation of Kupffer Cell Function in Health and Disease. Front Immunol 2021; 11:609618. [PMID: 33574817 PMCID: PMC7870864 DOI: 10.3389/fimmu.2020.609618] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 12/08/2020] [Indexed: 12/13/2022] Open
Abstract
Kupffer cells, the resident macrophages of the liver, comprise the largest pool of tissue macrophages in the body. Within the liver sinusoids Kupffer cells perform functions common across many tissue macrophages including response to tissue damage and antigen presentation. They also engage in specialized activities including iron scavenging and the uptake of opsonized particles from the portal blood. Here, we review recent studies of the epigenetic pathways that establish Kupffer cell identity and function. We describe a model by which liver-environment specific signals induce lineage determining transcription factors necessary for differentiation of Kupffer cells from bone-marrow derived monocytes. We conclude by discussing how these lineage determining transcription factors (LDTFs) drive Kupffer cell behavior during both homeostasis and disease, with particular focus on the relevance of Kupffer cell LDTF pathways in the setting of non-alcoholic fatty liver disease and non-alcoholic steatohepatitis.
Collapse
Affiliation(s)
- Hunter Bennett
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Ty D Troutman
- Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Mashito Sakai
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, United States.,Department of Biochemistry & Molecular Biology, Nippon Medical School, Tokyo, Japan
| | - Christopher K Glass
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, United States.,Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| |
Collapse
|
79
|
Chen JR, Zhao H, Wankhade UD, Chintapalli SV, Li C, Gai D, Shankar K, Zhan F, Lazarenko OP. GPR109A mediates the effects of hippuric acid on regulating osteoclastogenesis and bone resorption in mice. Commun Biol 2021; 4:53. [PMID: 33420329 PMCID: PMC7794563 DOI: 10.1038/s42003-020-01564-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 11/26/2020] [Indexed: 12/26/2022] Open
Abstract
The G protein-coupled receptor 109 A (GPR109A) is robustly expressed in osteoclastic precursor macrophages. Previous studies suggested that GPR109A mediates effects of diet-derived phenolic acids such as hippuric acid (HA) and 3-(3-hydroxyphenyl) propionic acid (3-3-PPA) on promoting bone formation. However, the role of GPR109A in metabolic bone homeostasis and osteoclast differentiation has not been investigated. Using densitometric, bone histologic and molecular signaling analytic methods, we uncovered that bone mass and strength were significantly higher in tibia and spine of standard rodent diet weaned 4-week-old and 6-month-old GPR109A gene deletion (GPR109A-/-) mice, compared to their wild type controls. Osteoclast numbers in bone and in ex vivo bone marrow cell cultures were significantly decreased in GPR109A-/- mice compared to wild type controls. In accordance with these data, CTX-1 in bone marrow plasma and gene expression of bone resorption markers (TNFα, TRAP, Cathepsin K) were significantly decreased in GPR109A-/- mice, while on the other hand, P1NP was increased in serum from both male and female GPR109A-/- mice compared to their respective controls. GPR109A deletion led to suppressed Wnt/β-catenin signaling in osteoclast precursors to inhibit osteoclast differentiation and activity. Indeed, HA and 3-3-PPA substantially inhibited RANKL-induced GPR109A expression and Wnt/β-catenin signaling in osteoclast precursors and osteoclast differentiation. Resultantly, HA significantly inhibited bone resorption and increased bone mass in wild type mice, but had no additional effects on bone in GPR109A-/- mice compared with their respective untreated control mice. These results suggest an important role for GPR109A during osteoclast differentiation and bone resorption mediating effects of HA and 3-3-PPA on inhibiting bone resorption during skeletal development.
Collapse
Affiliation(s)
- Jin-Ran Chen
- Arkansas Children's Nutrition Center, Little Rock, AR, 72202, USA.
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA.
| | - Haijun Zhao
- Arkansas Children's Nutrition Center, Little Rock, AR, 72202, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA
| | - Umesh D Wankhade
- Arkansas Children's Nutrition Center, Little Rock, AR, 72202, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA
| | - Sree V Chintapalli
- Arkansas Children's Nutrition Center, Little Rock, AR, 72202, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA
| | - Can Li
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA
| | - Dongzheng Gai
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA
| | - Kartik Shankar
- Arkansas Children's Nutrition Center, Little Rock, AR, 72202, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, 80045, USA
| | - Fenghuang Zhan
- Myeloma Center, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA
| | - Oxana P Lazarenko
- Arkansas Children's Nutrition Center, Little Rock, AR, 72202, USA
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, 72202, USA
| |
Collapse
|
80
|
Nutrients, Genetic Factors, and Their Interaction in Non-Alcoholic Fatty Liver Disease and Cardiovascular Disease. Int J Mol Sci 2020; 21:ijms21228761. [PMID: 33228237 PMCID: PMC7699550 DOI: 10.3390/ijms21228761] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/15/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in Western countries and expose patients to increased risk of hepatic and cardiovascular (CV) morbidity and mortality. Both environmental factors and genetic predisposition contribute to the risk. An inappropriate diet, rich in refined carbohydrates, especially fructose, and saturated fats, and poor in fibers, polyunsaturated fats, and vitamins is one of the main key factors, as well as the polymorphism of patatin-like phospholipase domain containing 3 (PNPLA3 gene) for NAFLD and the apolipoproteins and the peroxisome proliferator-activated receptor (PPAR) family for the cardiovascular damage. Beyond genetic influence, also epigenetics modifications are responsible for various clinical manifestations of both hepatic and CV disease. Interestingly, data are accumulating on the interplay between diet and genetic and epigenetic modifications, modulating pathogenetic pathways in NAFLD and CV disease. We report the main evidence from literature on the influence of both macro and micronutrients in NAFLD and CV damage and the role of genetics either alone or combined with diet in increasing the risk of developing both diseases. Understanding the interaction between metabolic alterations, genetics and diet are essential to treat the diseases and tailoring nutritional therapy to control NAFLD and CV risk.
Collapse
|
81
|
Mandala A, Janssen RC, Palle S, Short KR, Friedman JE. Pediatric Non-Alcoholic Fatty Liver Disease: Nutritional Origins and Potential Molecular Mechanisms. Nutrients 2020; 12:E3166. [PMID: 33081177 PMCID: PMC7602751 DOI: 10.3390/nu12103166] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 10/12/2020] [Accepted: 10/13/2020] [Indexed: 02/06/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the number one chronic liver disease worldwide and is estimated to affect nearly 40% of obese youth and up to 10% of the general pediatric population without any obvious signs or symptoms. Although the early stages of NAFLD are reversible with diet and lifestyle modifications, detecting such stages is hindered by a lack of non-invasive methods of risk assessment and diagnosis. This absence of non-invasive means of diagnosis is directly related to the scarcity of long-term prospective studies of pediatric NAFLD in children and adolescents. In the majority of pediatric NAFLD cases, the mechanisms driving the origin and rapid progression of NAFLD remain unknown. The progression from NAFLD to non-alcoholic steatohepatitis (NASH) in youth is associated with unique histological features and possible immune processes and metabolic pathways that may reflect different mechanisms compared with adults. Recent data suggest that circulating microRNAs (miRNAs) are important new biomarkers underlying pathways of liver injury. Several factors may contribute to pediatric NAFLD development, including high-sugar diets, in utero exposures via epigenetic alterations, changes in the neonatal microbiome, and altered immune system development and mitochondrial function. This review focuses on the unique aspects of pediatric NAFLD and how nutritional exposures impact the immune system, mitochondria, and liver/gastrointestinal metabolic health. These factors highlight the need for answers to how NAFLD develops in children and for early stage-specific interventions.
Collapse
Affiliation(s)
- Ashok Mandala
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (A.M.); (R.C.J.); (K.R.S.)
| | - Rachel C. Janssen
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (A.M.); (R.C.J.); (K.R.S.)
| | - Sirish Palle
- Department of Pediatrics, Section of Gastroenterology, Hepatology & Nutrition, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Kevin R. Short
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (A.M.); (R.C.J.); (K.R.S.)
- Department of Pediatrics, Section of Diabetes and Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Jacob E. Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA; (A.M.); (R.C.J.); (K.R.S.)
- Department of Pediatrics, Section of Diabetes and Endocrinology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
- Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
82
|
Guo W, Liu J, Hou S, Hu G, Ma H, Gong Q, Kan X, Ran X, Cao Y, Wang J, Fu S. The inflammatory environment mediated by a high-fat diet inhibited the development of mammary glands and destroyed the tight junction in pregnant mice. Food Funct 2020; 11:8193-8201. [PMID: 32966466 DOI: 10.1039/d0fo00609b] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Long-term intake of a high-fat diet seriously affects the health of pregnant women and leads to increased levels of inflammation in the mammary gland. Therefore, to further explore the effect of a high-fat diet on mammary gland development and the tight junction (TJ) during pregnancy, we placed mice into two groups: a high-fat diet group and a control group. We detected the expression of proteins related to fat synthesis in the mammary gland by western blotting. The results showed that a high-fat diet could lead to an increase in fat synthesis in the mammary gland. Then, the inflammatory levels and acinar cell morphology in the mammary gland were detected by ELISA and H&E staining. We also measured the levels of MAPK and NF-κB signal pathway-related proteins by western blotting. The results showed that a high-fat diet activated the MAPK and NF-κB signaling pathways and promoted the expression of inflammatory factors. Finally, the development of the mammary gland and the integrity of the TJ were determined by immunohistochemistry, immunofluorescence and western blotting. The results showed that a high-fat diet inhibited the development of the mammary gland and the expression of tight junction proteins (TJs). Our study showed that a high-fat diet could promote the expression of inflammatory factors by activating the MAPK and NF-κB signaling pathways and could reshape the microenvironment through extramammary inflammation. Finally, a high-fat diet inhibited the development of the mammary gland during pregnancy and destroyed the integrity of the TJ.
Collapse
Affiliation(s)
- Wenjin Guo
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Juxiong Liu
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Shuang Hou
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Guiqiu Hu
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - He Ma
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Qian Gong
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Xingchi Kan
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Xin Ran
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Yu Cao
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Jianfa Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Shoupeng Fu
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun 130062, China.
| |
Collapse
|
83
|
Thompson MD. Developmental Programming of NAFLD by Parental Obesity. Hepatol Commun 2020; 4:1392-1403. [PMID: 33024911 PMCID: PMC7527686 DOI: 10.1002/hep4.1578] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 07/08/2020] [Accepted: 07/08/2020] [Indexed: 12/11/2022] Open
Abstract
The surge of obesity across generations has become an increasingly relevant issue, with consequences for associated comorbidities in offspring. Data from longitudinal birth cohort studies support an association between maternal obesity and offspring nonalcoholic fatty liver disease (NAFLD), suggesting that perinatal obesity or obesogenic diet exposure reprograms offspring liver and increases NAFLD susceptibility. In preclinical models, offspring exposed to maternal obesogenic diet have increased hepatic steatosis after diet-induced obesity; however, the implications for later NAFLD development and progression are still unclear. Although some models show increased NAFLD incidence and progression in offspring, development of nonalcoholic steatohepatitis with fibrosis may be model dependent. Multigenerational programming of NAFLD phenotypes occurs after maternal obesogenic diet exposure; however, the mechanisms for such programming remain poorly understood. Likewise, emerging data on the role of paternal obesity in offspring NAFLD development reveal incomplete mechanisms. This review will explore the impact of parental obesity and obesogenic diet exposure on offspring NAFLD and areas for further investigation, including the impact of parental diet on disease progression, and consider potential interventions in preclinical models.
Collapse
Affiliation(s)
- Michael D. Thompson
- Division of Endocrinology and DiabetesDepartment of PediatricsWashington University School of MedicineSt. LouisMO
| |
Collapse
|
84
|
Sato S, Mukai Y. Modulation of Chronic Inflammation by Quercetin: The Beneficial Effects on Obesity. J Inflamm Res 2020; 13:421-431. [PMID: 32848440 PMCID: PMC7425105 DOI: 10.2147/jir.s228361] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/22/2020] [Indexed: 12/18/2022] Open
Abstract
Obesity has become a major risk factor for the development of chronic diseases such as insulin resistance, type 2 diabetes mellitus, and cardiovascular disease. Moreover, obesity induces chronic inflammation in adipose tissue, liver, skeletal muscle, and the vascular system. Quercetin is the major representative of the flavonoid subclass of flavonols, which is ubiquitously contained within natural plants such as green tea, and vegetables, including onions and apples. Researchers have focused greater attention to the beneficial physiological roles of quercetin, which has anti-oxidative, anti-inflammatory, and anti-fibrotic effects on insulin resistance and atherosclerosis in obesity-related diseases. Also, the anti-inflammatory effects of quercetin on intestinal microbiota have been demonstrated in obesity. In addition, there is increasing evidence that quercetin is associated with epigenetic activities in cancer, and in maternal undernutrition during gestation and lactation. In this review, we focus on the chemical properties of quercetin, its dietary sources in obesity, and its anti-inflammatory effects on insulin resistance, atherosclerosis, intestinal microbiota, and maternal under-nutrition with epigenetic activity.
Collapse
Affiliation(s)
- Shin Sato
- Department of Nutrition, Aomori University of Health and Welfare, Aomori 030-8505, Japan
| | - Yuuka Mukai
- School of Nutrition and Dietetics, Faculty of Health and Social Work, Kanagawa University of Human Services, Kanagawa 238-8522, Japan
| |
Collapse
|
85
|
Chen J, Lazarenko OP, Zhao H, Wankhade UD, Pedersen K, Watt J, Ronis MJJ. Nox4 Expression Is Not Required for OVX-Induced Osteoblast Senescence and Bone Loss in Mice. JBMR Plus 2020; 4:e10376. [PMID: 32803108 PMCID: PMC7422714 DOI: 10.1002/jbm4.10376] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 05/04/2020] [Accepted: 05/09/2020] [Indexed: 12/12/2022] Open
Abstract
Estrogen deficiency and aging play critical roles in the pathophysiology of bone as a result of increased oxidative stress. It has been suggested that prevention of NADPH oxidase- (Nox-) dependent accumulation of ROS may be an approach to potentially minimize bone loss caused by these conditions. Using ovariectomized (OVX) and Nox4 gene-deletion mouse models, we investigated the role of Nox4 in OVX-induced bone loss and osteoblast senescence signaling. Six-month-old WT C57Bl6 mice were allocated to a sham control group, OVX, and OVX plus E2 treatment group for 8 weeks. Decreased bone mass including BMD and BMC were found in the OVX group compared with the sham control (p < 0.05); E2 treatment completely reversed OVX-induced bone loss. Interestingly, the prevention of OVX-induced bone loss by E2 was associated with the elimination of increased senescence signaling in bone osteoblastic cells from the OVX group. E2 blunted OVX-induced p53 and p21 overexpression, but not p16 and Nox4 in bone. In addition, 8- and 11-month-old Nox4 KO female mice were OVX for 8 weeks. Significant bone loss and increased bone osteoblastic cell senescence signaling occurred not only in Nox4 KO OVX mice compared with sham-operated animals, but also in 11-month-old Nox4 KO sham mice compared with 8-month-old Nox4 KO sham mice (p < 0.05). These data suggest that Nox4-mediated ROS in bone osteoblastic cells may be dispensable for sex steroid deficiency-induced bone loss and senescence. © 2020 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Jin‐Ran Chen
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockARUSA
- Arkansas Children's Nutrition CenterLittle RockARUSA
| | - Oxana P Lazarenko
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockARUSA
- Arkansas Children's Nutrition CenterLittle RockARUSA
| | - Haijun Zhao
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockARUSA
- Arkansas Children's Nutrition CenterLittle RockARUSA
| | - Umesh D Wankhade
- Department of PediatricsUniversity of Arkansas for Medical SciencesLittle RockARUSA
- Arkansas Children's Nutrition CenterLittle RockARUSA
| | - Kim Pedersen
- Department of Pharmacology and Experimental TherapeuticsLouisiana State University Health Sciences CenterNew OrleansLAUSA
| | - James Watt
- Department of Pharmacology and Experimental TherapeuticsLouisiana State University Health Sciences CenterNew OrleansLAUSA
| | - Martin J J Ronis
- Department of Pharmacology and Experimental TherapeuticsLouisiana State University Health Sciences CenterNew OrleansLAUSA
| |
Collapse
|
86
|
Ghanemi A, Melouane A, Yoshioka M, St-Amand J. Exercise and High-Fat Diet in Obesity: Functional Genomics Perspectives of Two Energy Homeostasis Pillars. Genes (Basel) 2020; 11:genes11080875. [PMID: 32752100 PMCID: PMC7463441 DOI: 10.3390/genes11080875] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/24/2020] [Accepted: 07/28/2020] [Indexed: 12/15/2022] Open
Abstract
The heavy impact of obesity on both the population general health and the economy makes clarifying the underlying mechanisms, identifying pharmacological targets, and developing efficient therapies for obesity of high importance. The main struggle facing obesity research is that the underlying mechanistic pathways are yet to be fully revealed. This limits both our understanding of pathogenesis and therapeutic progress toward treating the obesity epidemic. The current anti-obesity approaches are mainly a controlled diet and exercise which could have limitations. For instance, the “classical” anti-obesity approach of exercise might not be practical for patients suffering from disabilities that prevent them from routine exercise. Therefore, therapeutic alternatives are urgently required. Within this context, pharmacological agents could be relatively efficient in association to an adequate diet that remains the most efficient approach in such situation. Herein, we put a spotlight on potential therapeutic targets for obesity identified following differential genes expression-based studies aiming to find genes that are differentially expressed under diverse conditions depending on physical activity and diet (mainly high-fat), two key factors influencing obesity development and prognosis. Such functional genomics approaches contribute to elucidate the molecular mechanisms that both control obesity development and switch the genetic, biochemical, and metabolic pathways toward a specific energy balance phenotype. It is important to clarify that by “gene-related pathways”, we refer to genes, the corresponding proteins and their potential receptors, the enzymes and molecules within both the cells in the intercellular space, that are related to the activation, the regulation, or the inactivation of the gene or its corresponding protein or pathways. We believe that this emerging area of functional genomics-related exploration will not only lead to novel mechanisms but also new applications and implications along with a new generation of treatments for obesity and the related metabolic disorders especially with the modern advances in pharmacological drug targeting and functional genomics techniques.
Collapse
Affiliation(s)
- Abdelaziz Ghanemi
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, QC G1V 0A6, Canada; (A.G.); (A.M.)
- Functional Genomics Laboratory, Endocrinology and Nephrology Axis, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada;
| | - Aicha Melouane
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, QC G1V 0A6, Canada; (A.G.); (A.M.)
- Functional Genomics Laboratory, Endocrinology and Nephrology Axis, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada;
| | - Mayumi Yoshioka
- Functional Genomics Laboratory, Endocrinology and Nephrology Axis, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada;
| | - Jonny St-Amand
- Department of Molecular Medicine, Faculty of Medicine, Laval University, Québec, QC G1V 0A6, Canada; (A.G.); (A.M.)
- Functional Genomics Laboratory, Endocrinology and Nephrology Axis, CHU de Québec-Université Laval Research Center, Québec, QC G1V 4G2, Canada;
- Correspondence: ; Tel.: +1-418-654-2296; Fax: +1-418-654-2761
| |
Collapse
|
87
|
Dai X, Guo Z, Chen D, Li L, Song X, Liu T, Jin G, Li Y, Liu Y, Ajiguli A, Yang C, Wang B, Cao H. Maternal sucralose intake alters gut microbiota of offspring and exacerbates hepatic steatosis in adulthood. Gut Microbes 2020; 11:1043-1063. [PMID: 32228300 PMCID: PMC7524393 DOI: 10.1080/19490976.2020.1738187] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Nonalcoholic fatty liver disease (NAFLD) is considered to be associated with diet and gut dysbiosis. Excessive sucralose can induce gut dysbiosis and negatively affect host health. Maternal diet shapes the microbial communities of neonate and this effect continues in later life. We aimed to investigate the effects of maternal sucralose (MS) intake on the susceptibility of offspring to hepatic steatosis in adulthood. METHODS C57BL/6 pregnant mice were randomized into MS group (MS during gestation and lactation) and maternal control (MC) group (MC diet). After weaning, all offspring were fed a control diet until 8 weeks of age, and then treated with a high-fat diet (HFD) for 4 weeks. The intestinal development, mucosal barrier function, and gut microbiota were assessed in the 3-week-old offspring. Moreover, the severity of hepatic steatosis, serum biochemistry, lipid metabolism, and gut microbiota was then assessed in the 12th week. RESULTS MS significantly inhibited intestinal development and disrupted barrier function in 3-week-old offspring. MS also induced intestinal low-grade inflammation, significantly changed the compositions and diversity of gut microbiota including reducing butyrate-producing bacteria and cecal butyrate production with down-regulation of GPR43. Mechanically, blocking GPR43 blunted the anti-inflammatory effect of one of the butyrate-producing bacteria, Clostridium butyricum in vitro. After HFD treatment, MS exacerbated hepatic steatosis, and disturbed fatty acid biosynthesis and metabolism, accompanied by inducing gut dysbiosis compared with MC group. CONCLUSIONS MS intake inhibits intestinal development, induces gut dysbiosis in offspring through down-regulation of GPR43, and exacerbates HFD-induced hepatic steatosis in adulthood.
Collapse
Affiliation(s)
- Xin Dai
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Zixuan Guo
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Danfeng Chen
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Lu Li
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Xueli Song
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Tianyu Liu
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Ge Jin
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China
| | - Yun Li
- Department of Pharmacy, General Hospital, Tianjin Medical University, Tianjin, China
| | - Yi Liu
- Department of Gastroenterology and Hepatology, Tianjin Third Central Hospital, Tianjin, China,Department of Gastroenterology and Hepatology, Hotan District People’s Hospital, Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Aihemaiti Ajiguli
- Department of Gastroenterology and Hepatology, Hotan District People’s Hospital, Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Cheng Yang
- Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine,Tianjin
| | - Bangmao Wang
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China,CONTACT Bangmao Wang Department of Gastroenterology and HepatologyGeneral Hospital, Tianjin Medical University, 154 Anshan Road, Heping District, China, Tianjin , 300052
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, Tianjin, China,Department of Gastroenterology and Hepatology, Hotan District People’s Hospital, Xinjiang Uygur Autonomous Region, Xinjiang, China,Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine,Tianjin,Hailong Cao Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical University, 154 Anshan Road, Heping District, Tianjin300052, China
| |
Collapse
|
88
|
Marzullo P, Di Renzo L, Pugliese G, De Siena M, Barrea L, Muscogiuri G, Colao A, Savastano S. From obesity through gut microbiota to cardiovascular diseases: a dangerous journey. INTERNATIONAL JOURNAL OF OBESITY SUPPLEMENTS 2020; 10:35-49. [PMID: 32714511 PMCID: PMC7371682 DOI: 10.1038/s41367-020-0017-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The co-existence of humans and gut microbiota started millions of years ago. Until now, a balance gradually developed between gut bacteria and their hosts. It is now recognized that gut microbiota are key to form adequate immune and metabolic functions and, more in general, for the maintenance of good health. Gut microbiota are established before birth under the influence of maternal nutrition and metabolic status, which can impact the future metabolic risk of the offspring in terms of obesity, diabetes, and cardiometabolic disorders during the lifespan. Obesity and diabetes are prone to disrupt the gut microbiota and alter the gut barrier permeability, leading to metabolic endotoxaemia with its detrimental consequences on health. Specific bacterial sequences are now viewed as peculiar signatures of the metabolic syndrome across life stages in each individual, and are linked to pathogenesis of cardiovascular diseases (CVDs) via metabolic products (metabolites) and immune modulation. These mechanisms have been linked, in association with abnormalities in microbial richness and diversity, to an increased risk of developing arterial hypertension, systemic inflammation, nonalcoholic fatty liver disease, coronary artery disease, chronic kidney disease, and heart failure. Emerging strategies for the manipulation of intestinal microbiota represent a promising therapeutic option for the prevention and treatment of CVD especially in individuals prone to CV events.
Collapse
Affiliation(s)
- Paolo Marzullo
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy
- Division of General Medicine, IRCCS Istituto Auxologico Italiano, 28923 Piancavallo, Verbania Italy
| | - Laura Di Renzo
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00136 Rome, Italy
| | - Gabriella Pugliese
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University, 80131 Naples, Italy
| | - Martina De Siena
- Division of Gastroenterology, Fondazione Policlinico Universitario A. Gemelli IRCCS—Università Cattolica del Sacro Cuore, Roma, Italy
- Digestive Endoscopy Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS—Università Cattolica del Sacro Cuore, Roma, Italy
| | - Luigi Barrea
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University, 80131 Naples, Italy
| | - Giovanna Muscogiuri
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University, 80131 Naples, Italy
| | - Annamaria Colao
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University, 80131 Naples, Italy
| | - Silvia Savastano
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University, 80131 Naples, Italy
| | - on behalf of Obesity Programs of nutrition, Education, Research and Assessment (OPERA) Group
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy
- Division of General Medicine, IRCCS Istituto Auxologico Italiano, 28923 Piancavallo, Verbania Italy
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00136 Rome, Italy
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University, 80131 Naples, Italy
- Division of Gastroenterology, Fondazione Policlinico Universitario A. Gemelli IRCCS—Università Cattolica del Sacro Cuore, Roma, Italy
- Digestive Endoscopy Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS—Università Cattolica del Sacro Cuore, Roma, Italy
| |
Collapse
|
89
|
Obri A, Serra D, Herrero L, Mera P. The role of epigenetics in the development of obesity. Biochem Pharmacol 2020; 177:113973. [DOI: 10.1016/j.bcp.2020.113973] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 04/08/2020] [Indexed: 12/13/2022]
|
90
|
Suarez-Trujillo A, Huff K, Ramires Ferreira C, Paschoal Sobreira TJ, Buhman KK, Casey T. High-fat-diet induced obesity increases the proportion of linoleic acyl residues in dam serum and milk and in suckling neonate circulation. Biol Reprod 2020; 103:736-749. [DOI: 10.1093/biolre/ioaa103] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 04/13/2020] [Accepted: 06/11/2020] [Indexed: 12/12/2022] Open
Abstract
AbstractMaternal obesity increases the risk of offspring to become obese and develop related pathologies. Exposure to maternal high-fat diet (HFD) only during lactation increases the risk of obesity-related diseases, suggesting that factors in milk affect long-term health. We hypothesized that prepregnancy obesity induced by HFD alters milk lipidome, and in turn, alterations may affect neonate serum lipidome. The objective of this study was to determine the effect of prepregnancy obesity induced by HFD on circulating lipids in dams and neonates and in milk. Female mice were fed an HFD (60% kcal fat) or control diet (CON, 10% kcal fat) beginning 4 weeks before breeding. On postnatal day 2 (PND2), pups were cross-fostered to create pup groups exposed to HFD during pregnancy, lactation, or both or exposed to CON. On PND12, dams were milked and then euthanized along with pups to collect blood. Serum and milk were processed for multiple reaction monitoring (MRM) lipidomics profiling to quantify the relative expression of lipid classes. Lipidome of HFD dam serum and milk had increased proportion of C18:2 free fatty acid and fatty acyl residues in all lipid classes. Lipidome of serum from pups exposed to maternal HFD during lactation was similarly affected. Thus, maternal HFD induced redistribution of fatty acyl residues in the dam’s circulation, which was associated with modification in milk and suckling neonate’s lipidome. Further studies are needed to determine if increased circulating levels of C18:2 in neonate affects development and predisposes offspring to obesity and metabolic syndrome.
Collapse
Affiliation(s)
| | - Katelyn Huff
- Animal Sciences Department, Purdue University, Indiana, USA
- Biological & Biomedical Sciences Program, University of North Carolina-Chapel Hill, North Carolina, USA
| | - Christina Ramires Ferreira
- Department of Chemistry, Purdue University, Indiana, USA
- Bindley Bioscience Center, Purdue University, Indiana, USA
| | | | | | - Theresa Casey
- Animal Sciences Department, Purdue University, Indiana, USA
| |
Collapse
|
91
|
Un Nisa K, Reza MI. Key Relevance of Epigenetic Programming of Adiponectin Gene in Pathogenesis of Metabolic Disorders. Endocr Metab Immune Disord Drug Targets 2020; 20:506-517. [DOI: 10.2174/1871530319666190801142637] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 06/20/2019] [Accepted: 06/20/2019] [Indexed: 12/20/2022]
Abstract
Background & Objective::
Significant health and social burdens have been created by the
growth of metabolic disorders like type 2 diabetes mellitus (T2DM), atherosclerosis, and non-alcoholic
steatohepatitis, worldwide. The number of the affected population is as yet rising, and it is assessed
that until 2030, 4−5 million individuals will acquire diabetes. A blend of environmental, genetic, epigenetic,
and other factors, such as diet, are accountable for the initiation and progression of metabolic
disorders. Several researches have shown strong relevance of adiponectin gene and metabolic disorders.
In this review, the potential influence of epigenetic mechanisms of adiponectin gene “ADIPOQ”
on increasing the risk of developing metabolic disorders and their potential in treating this major disorder
are discussed.
Results & Conclusion::
Various studies have postulated that a series of factors such as maternal High
fat diet (HFD), oxidative stress, pro-inflammatory mediators, sleep fragmentation throughout lifetime,
from gestation to old age, could accumulate epigenetic marks, including histone remodeling, DNA
methylation, and microRNAs (miRNAs) that, in turn, alter the expression of ADIPOQ gene and result
in hypoadiponectinemia which precipitates insulin resistance (IR) that in turn might induce or accelerate
the onset and development of metabolic disorder. A better understanding of global patterns of epigenetic
modifications and further their alterations in metabolic disorders will bestow better treatment
strategies design.
Collapse
Affiliation(s)
- Kaiser Un Nisa
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education & Research, SAS Nagar, India
| | - Mohammad Irshad Reza
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education & Research, SAS Nagar, India
| |
Collapse
|
92
|
A dietary intervention to improve the microbiome composition of pregnant women with Crohn's disease and their offspring: The MELODY (Modulating Early Life Microbiome through Dietary Intervention in Pregnancy) trial design. Contemp Clin Trials Commun 2020; 18:100573. [PMID: 32617430 PMCID: PMC7322804 DOI: 10.1016/j.conctc.2020.100573] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 04/13/2020] [Accepted: 04/26/2020] [Indexed: 12/18/2022] Open
Abstract
Crohn's disease (CD), a type of inflammatory bowel disease (IBD), is a chronic condition of the gastrointestinal tract that is caused by the loss of mucosal tolerance towards the commensal bacteria resulting in inflammatory responses. It has long been postulated that the gut microbiota, a complex and dynamic population of microorganisms, plays a key role in the pathogenesis of IBD. Maternal diagnosis of IBD has been identified as the greatest risk factor for IBD in offspring increasing the odds of developing the disease >4.5-fold. Moreover, babies born to mothers with IBD have demonstrated reduced gut bacterial diversity. There is accumulating evidence that the early life microbiota colonization is informed by maternal diet within the 3rd trimester of pregnancy. While babies born to mothers with IBD would pose an ideal cohort for intervention, no primary prevention measures are currently available. Therefore, we designed the MELODY (Modulating Early Life Microbiome through Dietary Intervention in Pregnancy) trial to test whether the IBD-AID™ dietary intervention during the last trimester of pregnancy can beneficially shift the microbiome of CD patients and their babies, thereby promoting a strong, effective immune system during a critical time of the immune system development. We will also test if favorable changes in the microbiome can lead to a reduced risk of postpartum CD relapse and lower mucosal inflammation in the offspring. This study will help create new opportunities to foster a healthy microbiome in the offspring at high risk of other immune-mediated diseases, potentially reducing their risk later in life.
Collapse
|
93
|
Meroni M, Longo M, Rustichelli A, Dongiovanni P. Nutrition and Genetics in NAFLD: The Perfect Binomium. Int J Mol Sci 2020; 21:ijms21082986. [PMID: 32340286 PMCID: PMC7215858 DOI: 10.3390/ijms21082986] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/19/2020] [Accepted: 04/21/2020] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) represents a global healthcare burden since it is epidemiologically related to obesity, type 2 diabetes (T2D) and Metabolic Syndrome (MetS). It embraces a wide spectrum of hepatic injuries, which include simple steatosis, nonalcoholic steatohepatitis (NASH), fibrosis, cirrhosis and hepatocellular carcinoma (HCC). The susceptibility to develop NAFLD is highly variable and it is influenced by several cues including environmental (i.e., dietary habits and physical activity) and inherited (i.e., genetic/epigenetic) risk factors. Nonetheless, even intestinal microbiota and its by-products play a crucial role in NAFLD pathophysiology. The interaction of dietary exposure with the genome is referred to as 'nutritional genomics,' which encompasses both 'nutrigenetics' and 'nutriepigenomics.' It is focused on revealing the biological mechanisms that entail both the acute and persistent genome-nutrient interactions that influence health and it may represent a promising field of study to improve both clinical and health nutrition practices. Thus, the premise of this review is to discuss the relevance of personalized nutritional advices as a novel therapeutic approach in NAFLD tailored management.
Collapse
Affiliation(s)
- Marica Meroni
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Pad. Granelli, via F Sforza 35, 20122 Milan, Italy; (M.M.); (M.L.); (A.R.)
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, 20122 Milano, Italy
| | - Miriam Longo
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Pad. Granelli, via F Sforza 35, 20122 Milan, Italy; (M.M.); (M.L.); (A.R.)
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, 20122 Milano, Italy
| | - Alice Rustichelli
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Pad. Granelli, via F Sforza 35, 20122 Milan, Italy; (M.M.); (M.L.); (A.R.)
| | - Paola Dongiovanni
- General Medicine and Metabolic Diseases, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Pad. Granelli, via F Sforza 35, 20122 Milan, Italy; (M.M.); (M.L.); (A.R.)
- Correspondence: ; Tel.: +39-02-5503-3467; Fax: +39-02-5503-4229
| |
Collapse
|
94
|
Franzago M, Santurbano D, Vitacolonna E, Stuppia L. Genes and Diet in the Prevention of Chronic Diseases in Future Generations. Int J Mol Sci 2020; 21:ijms21072633. [PMID: 32290086 PMCID: PMC7178197 DOI: 10.3390/ijms21072633] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 03/30/2020] [Accepted: 04/08/2020] [Indexed: 12/21/2022] Open
Abstract
Nutrition is a modifiable key factor that is able to interact with both the genome and epigenome to influence human health and fertility. In particular, specific genetic variants can influence the response to dietary components and nutrient requirements, and conversely, the diet itself is able to modulate gene expression. In this context and the era of precision medicine, nutrigenetic and nutrigenomic studies offer significant opportunities to improve the prevention of metabolic disturbances, such as Type 2 diabetes, gestational diabetes, hypertension, and cardiovascular diseases, even with transgenerational effects. The present review takes into account the interactions between diet, genes and human health, and provides an overview of the role of nutrigenetics, nutrigenomics and epigenetics in the prevention of non-communicable diseases. Moreover, we focus our attention on the mechanism of intergenerational or transgenerational transmission of the susceptibility to metabolic disturbances, and underline that the reversibility of epigenetic modifications through dietary intervention could counteract perturbations induced by lifestyle and environmental factors.
Collapse
Affiliation(s)
- Marica Franzago
- Department of Medicine and Aging, School of Medicine and Health Sciences, ‘G. d’Annunzio’ University of Chieti-Pescara, 66100 Chieti, Italy
- Center for Advanced Studies and Technology (CAST), ‘G. d’Annunzio’ University of Chieti-Pescara, 66100 Chieti, Italy
| | | | - Ester Vitacolonna
- Department of Medicine and Aging, School of Medicine and Health Sciences, ‘G. d’Annunzio’ University of Chieti-Pescara, 66100 Chieti, Italy
- Center for Advanced Studies and Technology (CAST), ‘G. d’Annunzio’ University of Chieti-Pescara, 66100 Chieti, Italy
- Correspondence:
| | - Liborio Stuppia
- Center for Advanced Studies and Technology (CAST), ‘G. d’Annunzio’ University of Chieti-Pescara, 66100 Chieti, Italy
- Department of Psychological, Health and Territorial Sciences, School of Medicine and Health Sciences, ‘G. d’Annunzio’ University of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
95
|
Nicholas LM, Ozanne SE. Early life programming in mice by maternal overnutrition: mechanistic insights and interventional approaches. Philos Trans R Soc Lond B Biol Sci 2020; 374:20180116. [PMID: 30966886 DOI: 10.1098/rstb.2018.0116] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Animal models have been indispensable in elucidating the potential causative mechanisms underlying the effects of maternal diet on offspring health. Of these, the mouse has been widely used to model maternal overnutrition and/or maternal obesity and to study its effects across one or more generations. This review discusses recent findings from mouse models, which resemble the human situation, i.e. overnutrition/obesity across pregnancy and lactation. It also highlights the importance of embryo transfer models in identifying critical developmental period(s) during which specific metabolic changes are programmed in the offspring. The mouse is also an excellent tool for maternal intervention studies aimed at elucidating the longer-term effects on the offspring and for defining possible maternal factors underling the programming of metabolic adversity in offspring. While knowledge of the mouse genome and the molecular tools available have allowed great progress to be made in the field, it is clear that we need to define if the effects on the offspring are mediated by maternal obesity per se or if specific components of the maternal metabolic environment are more important. We can then begin to identify at-risk offspring and to design more effective interventions for the mother and/or her child. This article is part of the theme issue 'Developing differences: early-life effects and evolutionary medicine'.
Collapse
Affiliation(s)
- Lisa M Nicholas
- Metabolic Research Laboratories, MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge , Addenbrooke's Hospital, Cambridge , UK
| | - Susan E Ozanne
- Metabolic Research Laboratories, MRC Metabolic Diseases Unit, Wellcome Trust-MRC Institute of Metabolic Science, University of Cambridge , Addenbrooke's Hospital, Cambridge , UK
| |
Collapse
|
96
|
Li T, Gong H, Yuan Q, Du M, Ren F, Mao X. Supplementation of polar lipids-enriched milk fat globule membrane in high-fat diet-fed rats during pregnancy and lactation promotes brown/beige adipocyte development and prevents obesity in male offspring. FASEB J 2020; 34:4619-4634. [PMID: 32020679 DOI: 10.1096/fj.201901867rrr] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 01/14/2020] [Accepted: 01/20/2020] [Indexed: 12/15/2022]
Abstract
Promoting brown adipose tissue (BAT) function or browning of white adipose tissue (WAT) provides a defense against obesity. The aim of the study was to investigate whether maternal polar lipids-enriched milk fat globule membrane (MFGM-PL) supplementation to high-fat diet (HFD) rats during pregnancy and lactation could promote brown/beige adipogenesis and protect against HFD-induced adiposity in offspring. Female SD rats were fed a HFD for 8 weeks to induce obesity and, then, fed a HFD during pregnancy and lactation with or without MFGM-PL. Male offspring were weaned at postnatal Day 21 and then fed a HFD for 9 weeks. MFGM-PL treatment to HFD dams decreased the body weight gain and WAT mass as well as lowered the serum levels of insulin and triglycerides in male offspring at weaning. MFGM-PL+HFD offspring showed promoted thermogenic function in BAT and inguinal WAT through the upregulation of UCP1 and other thermogenic genes. In adulthood, maternal MFGM-PL supplementation reduced adiposity and increased oxygen consumption, respiratory exchange ratio, and heat production in male offspring. The enhancement of energy expenditure was correlated with elevated BAT activity and inguinal WAT thermogenic program. In conclusion, maternal MFGM-PL treatment activated thermogenesis in offspring, which exerted long-term beneficial effects against HFD-induced obesity in later life.
Collapse
Affiliation(s)
- Tiange Li
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, Key Laboratory of Functional Dairy, China Agricultural University, Beijing, China
| | - Han Gong
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, Key Laboratory of Functional Dairy, China Agricultural University, Beijing, China
| | - Qichen Yuan
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, Key Laboratory of Functional Dairy, China Agricultural University, Beijing, China
| | - Min Du
- Department of Animal Sciences, Washington State University, Pullman, WA, USA
| | - Fazheng Ren
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, Key Laboratory of Functional Dairy, China Agricultural University, Beijing, China
| | - Xueying Mao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science & Nutritional Engineering, Key Laboratory of Functional Dairy, China Agricultural University, Beijing, China
| |
Collapse
|
97
|
Jonscher KR, Abrams J, Friedman JE. Maternal Diet Alters Trained Immunity in the Pathogenesis of Pediatric NAFLD. JOURNAL OF CELLULAR IMMUNOLOGY 2020; 2:315-325. [PMID: 33426540 PMCID: PMC7793570 DOI: 10.33696/immunology.2.061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Pediatric nonalcoholic fatty liver disease (NAFLD) affects 1 in 10 children in the US, increases risk of cirrhosis and transplantation in early adulthood, and shortens lifespan, even after transplantation. Exposure to maternal obesity and/or a diet high in fat, sugar and cholesterol is strongly associated with development of NAFLD in offspring. However, mechanisms by which "priming" of the immune system in early life increases susceptibility to NAFLD are poorly understood. Recent studies have focused on the role "non-reparative" macrophages play in accelerating inflammatory signals promoting fibrogenesis. In this Commentary, we review evidence that the pioneering gut bacteria colonizing the infant intestinal tract remodel the naïve immune system in the offspring. Epigenetic changes in hematopoietic stem and progenitor cells, induced by exposure to an obesogenic diet in utero, may skew lineage commitment of myeloid cells during gestation. Further, microbial dysbiosis in neonatal life contributes to training innate immune cell responsiveness in the gut, bone marrow, and liver, leading to developmental programming of pediatric NAFLD. Comprehensive understanding of how different gut bacteria and their byproducts shape development of the early innate immune system and microbiome will uncover early interventions to prevent NAFLD pathophysiology.
Collapse
Affiliation(s)
- Karen R. Jonscher
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, USA
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, USA
| | - Jesse Abrams
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, USA
| | - Jacob E. Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, USA
- Departments of Physiology and Pediatrics, University of Oklahoma Health Sciences Center, USA
| |
Collapse
|
98
|
Physical exercise and liver "fitness": Role of mitochondrial function and epigenetics-related mechanisms in non-alcoholic fatty liver disease. Mol Metab 2019; 32:1-14. [PMID: 32029220 PMCID: PMC6931125 DOI: 10.1016/j.molmet.2019.11.015] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/19/2019] [Accepted: 11/22/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Modern lifestyles, especially high-caloric intake and physical inactivity, contribute to the increased prevalence of non-alcoholic fatty liver disease (NAFLD), which becomes a significant health problem worldwide. Lifestyle changes, however, affect not only parental generation, but also their offspring, reinforcing the need for efficient preventive approaches to deal with this disease. This transgenerational influence of phenotypes dependent on parents (particularly maternal) behaviours may open additional research avenues. Despite persistent attempts to design an effective pharmacological therapy against NAFLD, physical activity, as a non-pharmacological approach, emerges as an exciting strategy. SCOPE OF REVIEW Here we briefly review the effect of physical exercise on liver mitochondria adaptations in NAFLD, highlighting the importance of mitochondrial metabolism and transgenerational and epigenetic mechanisms in liver diseases. MAJOR CONCLUSIONS A deeper look into cellular mechanisms sheds a light on possible effects of physical activity in the prevention and treatment of NAFLD through modulation of function and structure of particular organelles, namely mitochondria. Additionally, despite of increasing evidence regarding the contribution of epigenetic mechanisms in the pathogenesis of different diseases, the role of microRNAs, DNA methylation, and histone modification in NAFLD pathogenesis still needs to be elucidated.
Collapse
|
99
|
Zhang Y, Xu J, Wang X, Ren X, Liu Y. Changes of intestinal bacterial microbiota in coronary heart disease complicated with nonalcoholic fatty liver disease. BMC Genomics 2019; 20:862. [PMID: 31726978 PMCID: PMC6857132 DOI: 10.1186/s12864-019-6251-7] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Accepted: 10/31/2019] [Indexed: 02/07/2023] Open
Abstract
Background Previous study reported that patients who suffered coronary heart disease (CHD) complicated with non-alcoholic fatty liver disease (NAFLD) had worse cardiac function and clinical outcomes than patients with CHD only. Notably, the mechanism is still unclear. This study aimed to investigate the changes and roles of intestinal bacterial microbiota in CHD-NAFLD patients. Methods and results People were recruited and divided into three groups, including CHD patients (without NAFLD), CHD-NAFLD patients and healthy controls (HCs). Each group contained 24 people. Fecal samples and clinical information were carefully collected. The Illumina sequencing of 16S rRNA was applied to profile the overall structure of the fecal bacterial microbiota and the characteristics of the bacterial microbiota based on the Operational Taxonomic Units. In clinical information, the CHD-NAFLD patients showed an increase in BMI, uric acid and triglyceride. There was a significant reduction in the abundance of Parabacteroides and Collinsella in overall CHD patients (including CHD-NAFLD and CHD patients). The intestinal bacterial microbiota in CHD-NAFLD patients showed an increase in the abundance of Copococcus and Veillonella, and a reduction in the abundance of Parabacteroides, Bacteroides fragilis, Ruminococcus gnavus, Bacteroides dorei, and Bifidobacterium longum subsp infantis. Among them, the abundance of Ruminococcus gnavus and Bacteroides dorei was significantly lower than that in CHD patients. Additionally, BMI positively correlated with the abundance of Copococcus and negatively correlated with the abundance of Bifidobacterium longum subsp infantis. The abundance of Veillonella positively correlated with AST. The abundance of Bacteroides dorei negatively correlated with ALT and AST. It indicates that the abundance of intestinal microbiota was related to the changes in clinical indexes. Conclusions Changes of intestinal bacterial microbiota in CHD-NAFLD patients may be important factors affecting the degree of metabolic disorder, which may be one of the important reasons for the worse clinical outcome and disease progression in CHD-NAFLD patients than in CHD patients.
Collapse
Affiliation(s)
- Yiwen Zhang
- Department of Gastroenterology, Peking University People's Hospital, No.11 Xizhimen South Street, Xicheng District, Beijing, People's Republic of China.,Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, No.11 Xizhimen South Street, Xicheng, Beijing, People's Republic of China
| | - Jun Xu
- Department of Gastroenterology, Peking University People's Hospital, No.11 Xizhimen South Street, Xicheng District, Beijing, People's Republic of China.,Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, No.11 Xizhimen South Street, Xicheng, Beijing, People's Republic of China.,Institute of Clinical Molecular Biology & Central Laboratory, Peking University People's Hospital, No.11 Xizhimen South Street, Xicheng District, Beijing, People's Republic of China
| | - Xuemei Wang
- Department of Gastroenterology, Peking University People's Hospital, No.11 Xizhimen South Street, Xicheng District, Beijing, People's Republic of China.,Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, No.11 Xizhimen South Street, Xicheng, Beijing, People's Republic of China
| | - Xinhua Ren
- Department of Gastroenterology, Peking University People's Hospital, No.11 Xizhimen South Street, Xicheng District, Beijing, People's Republic of China.,Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, No.11 Xizhimen South Street, Xicheng, Beijing, People's Republic of China
| | - Yulan Liu
- Department of Gastroenterology, Peking University People's Hospital, No.11 Xizhimen South Street, Xicheng District, Beijing, People's Republic of China. .,Clinical Center of Immune-Mediated Digestive Diseases, Peking University People's Hospital, No.11 Xizhimen South Street, Xicheng, Beijing, People's Republic of China.
| |
Collapse
|
100
|
Paneth Cell Ablation Aggravates Pancreatic and Intestinal Injuries in a Rat Model of Acute Necrotizing Pancreatitis after Normal and High-Fat Diet. Mediators Inflamm 2019; 2019:8474523. [PMID: 31827383 PMCID: PMC6885277 DOI: 10.1155/2019/8474523] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/28/2019] [Accepted: 09/13/2019] [Indexed: 02/07/2023] Open
Abstract
We previously reported that acute necrotizing pancreatitis (ANP) after normal or high-fat diet is associated with a decreased number of Paneth cells in ileal crypts. Here, we ablated Paneth cells in a rat model of ANP after normal and high-fat diet to investigate the effects on disease symptoms. Adult male Sprague-Dawley rats received standard rat chow or a high-fat diet for 2 weeks, after which they were treated with dithizone to deplete Paneth cells. Six hours later, ANP was established by retrograde injection of sodium taurocholate into the biliopancreatic duct. Rats were sacrificed at 6, 12, and 24 h for assessment. We found dithizone aggravated ANP-associated pathological injuries to the pancreas and ileum in rats on high-fat or standard diets. Lysozyme expression in ileal crypts was decreased, while serum inflammatory cytokines (TNFα, IL-1β, and IL-17A) and intestinal permeability (serum DAO activity and D-lactate) were increased. Expression of tight junction proteins (claudin-1, zo-1, and occludin) was decreased. Using high-throughput 16S rRNA sequencing, we found dithizone reduced microbiota diversity and altered microbiota composition in rats on high-fat or standard diets. Dithizone decreased fecal short-chain fatty acids (SCFAs) in rats on high-fat or standard diets. Changes in intestinal microbiota correlated significantly with SCFAs, lysozyme, DAO activity, D-lactate, inflammatory cytokines, and pathological injury to the pancreas and ileum in rats on high-fat or standard diets. In conclusion, ablation of Paneth cells exacerbates pancreatic and intestinal injuries in ANP after normal and high-fat diet. These symptoms may be related to changes in the intestinal microbiota.
Collapse
|