1
|
Rasouli MA, Dumesic DA, Singhal V. Male infertility and obesity. Curr Opin Endocrinol Diabetes Obes 2024; 31:203-209. [PMID: 39253759 DOI: 10.1097/med.0000000000000883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
PURPOSE OF REVIEW The increasing rate of obesity is having an adverse impact on male reproduction. RECENT FINDINGS The negative effect of reactive oxygen species on male reproductive tissues and the age of onset of obesity are new areas of research on male infertility. SUMMARY This review highlights how obesity impairs male reproduction through complex mechanisms, including metabolic syndrome, lipotoxicity, sexual dysfunction, hormonal and adipokine alterations as well as epigenetic changes, and how new management strategies may improve the reproductive health of men throughout life.
Collapse
Affiliation(s)
| | | | - Vibha Singhal
- Division of Endocrinology, Department of Pediatrics, University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
2
|
Müller GA, Müller TD. A "poly-matter network" conception of biological inheritance. Genetica 2024; 152:211-230. [PMID: 39425866 PMCID: PMC11541361 DOI: 10.1007/s10709-024-00216-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 09/26/2024] [Indexed: 10/21/2024]
Abstract
Here we intend to shift the "DNA- and information-centric" conception of biological inheritance, with the accompanying exclusion of any non-DNA matter, to a "poly-matter network" framework which, in addition to DNA, considers the action of other cellular membranous constituents. These cellular structures, in particular organelles and plasma membranes, express "landscapes" of specific topologies at their surfaces, which may become altered in response to certain environmental factors. These so-called "membranous environmental landscapes" (MELs), which replicate by self-organization / autopoiesis rather than self-assembly, are transferred from donor to acceptor cells by various - vesicular and non-vesicular - mechanisms and exert novel features in the acceptor cells. The "DNA-centric" conception may be certainly explanatorily sufficient for the transfer of heritable phenotype variation to acceptor cells following the copying of DNA in donor cells and thereby for the phenomenon of biological inheritance of traits. However, it is not causally sufficient. With the observation of phenotype variation, as initially manifested during bacterial transformation, the impact of environmental factors, such as nutrition and stress, in the differential regulation of gene expression has been widely accepted and resulted in intense efforts to resolve the underlying epigenetic mechanisms. However, these are explained under a conceptual frame where the DNA (and associated proteins) are the only matter of inheritance. In contrast, it is our argumentation that inheritance can only be adequately understood as the transfer of DNA in concert with non-DNA matter in a "poly-matter network" conception. The adequate inclusion of the transfer of non-DNA matter is still a desideratum of future genetic research, which may pave the way for the experimental elucidation not only of how DNA and membrane matter act in concert to enable the inheritance of innate traits, but also whether they interact for that of acquired biological traits. Moreover, the "poly-matter network" conception may open new perspectives for an understanding of the pathogenesis of "common complex" diseases.
Collapse
Affiliation(s)
- Günter A Müller
- Institute of Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC) at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764, Oberschleissheim, Germany.
- Biology and Technology Studies Institute Munich (BITSIM), Lappenweg 16, 80939, Munich, Germany.
- Media, Culture and Society, Department of Media Studies, Faculty of Arts and Humanities, University Paderborn, Warburger Str. 100, 33098, Paderborn, Germany.
| | - Timo D Müller
- Institute of Diabetes and Obesity (IDO), Helmholtz Diabetes Center (HDC) at Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstädter Landstraße 1, 85764, Oberschleissheim, Germany
- Walther-Straub Institute of Pharmacology and Toxicology, Ludwig-Maximilians-University Munich, Munich, Germany
| |
Collapse
|
3
|
Larouche M, Bluteau O, Carrié A, Lauzière A, Khoury E, Brisson D, Gaudet D, Gallo A. Clinical Expression of Familial Hypercholesterolemia in Patients from France and French Canada Carrying Identical-by-Descent Pathogenic LDLR Gene Variants: A Proof-of-Concept Study. J Clin Med 2024; 13:5725. [PMID: 39407785 PMCID: PMC11477318 DOI: 10.3390/jcm13195725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/23/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Background: Studying patients carrying identical-by-descent (IBD) pathogenic gene variants allows us to control for the disease-causing genetic background and to more accurately document the impact of modifiers. Familial hypercholesterolemia (FH) is characterized by elevated low-density lipoprotein cholesterol (LDL-c) levels and premature atherosclerosis and is often caused by defects in the LDLR gene. There is a high prevalence of FH in French Canada as a result of a founder effect from France in the 17th century. Several FH patients currently living in French Canada (founder population) and in France (colonizing population) carry IBD FH-causing variants. The expression of FH is affected by environmental and genetic modifiers, and patients with IBD variants may present different characteristics. Methods: In this study, we compared FH clinical expression patients carrying IBD LDLR pathogenic variants living in France or Canada. Four IBD variants, namely c.259T>G p.(Trp87Gly), c.2000G>A p.(Cys667Tyr), c.682G>A p.(Glu228Lys), and c.1048C>T p.(Arg350*), were selected. Untreated plasma lipid profiles, the apolipoprotein E (APOE) genotype, cardiovascular risk factors, and the occurrence of symptomatic ASCVD were compared in 105 adult carriers (30 from France and 75 from French Canada). Results: All parameters were similar between the two populations, except for untreated total cholesterol (10.14 ± 1.89 mmol/L vs. 8.65 ± 1.84 mmol/L, p = 0.0006) and LDL-c concentrations (7.94 ± 1.86 mmol/L vs. 6.93 ± 1.78 mmol/L, p = 0.016), which were significantly higher in FH patients living in France, an observation that was revealed across all studied LDLR variants. Conclusions: This study illustrates that FH patients sharing IBD pathogenic LDLR variants that have evolved in different geographic, cultural, and socio-economic environments for hundreds of years differ in terms of cholesterol levels, highlighting the importance of better understanding the interplay between genetic and environmental modulators of FH expression.
Collapse
Affiliation(s)
- Miriam Larouche
- Departement of Medicine, Université de Montréal and ECOGENE-21, Chicoutimi, QC G7H 7K9, Canada; (M.L.); (A.L.); (E.K.); (D.B.)
| | - Olivier Bluteau
- Sorbonne-Université, Assistance Publique—Hôpitaux de Paris (APHP), INSERM Unité de Recherche sur les Maladies Cardiovasculaires et Métaboliques (UMRS) 1166, Hôpital Pitié-Salpêtrière, 75013 Paris, France; (O.B.); (A.C.); (A.G.)
| | - Alain Carrié
- Sorbonne-Université, Assistance Publique—Hôpitaux de Paris (APHP), INSERM Unité de Recherche sur les Maladies Cardiovasculaires et Métaboliques (UMRS) 1166, Hôpital Pitié-Salpêtrière, 75013 Paris, France; (O.B.); (A.C.); (A.G.)
| | - Alex Lauzière
- Departement of Medicine, Université de Montréal and ECOGENE-21, Chicoutimi, QC G7H 7K9, Canada; (M.L.); (A.L.); (E.K.); (D.B.)
| | - Etienne Khoury
- Departement of Medicine, Université de Montréal and ECOGENE-21, Chicoutimi, QC G7H 7K9, Canada; (M.L.); (A.L.); (E.K.); (D.B.)
| | - Diane Brisson
- Departement of Medicine, Université de Montréal and ECOGENE-21, Chicoutimi, QC G7H 7K9, Canada; (M.L.); (A.L.); (E.K.); (D.B.)
| | - Daniel Gaudet
- Departement of Medicine, Université de Montréal and ECOGENE-21, Chicoutimi, QC G7H 7K9, Canada; (M.L.); (A.L.); (E.K.); (D.B.)
| | - Antonio Gallo
- Sorbonne-Université, Assistance Publique—Hôpitaux de Paris (APHP), INSERM Unité de Recherche sur les Maladies Cardiovasculaires et Métaboliques (UMRS) 1166, Hôpital Pitié-Salpêtrière, 75013 Paris, France; (O.B.); (A.C.); (A.G.)
| |
Collapse
|
4
|
Zhu D, Zheng W, Kuang J, Wang Y, Deng X, Li X, Zhou W. Role of RIPK3 in lipid metabolism and postnatal overfeeding-induced metabolic disorders in mice. J Nutr Biochem 2024; 131:109688. [PMID: 38871059 DOI: 10.1016/j.jnutbio.2024.109688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 06/02/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024]
Abstract
Postnatal overfeeding can increase the long-term risk of metabolic disorders, such as obesity, but the underlying mechanisms remain unclear and treatment approaches are limited. Receptor-interacting protein kinase 3 (RIPK3) is associated with several metabolic diseases. We investigated the effects of RIPK3 on neonatal overfeeding-related metabolic disorders. On postnatal day 3, litter sizes were adjusted to 9-10 (normal litters, NL) or 2-3 (small litters, SL) mice per dam to mimic postnatal overfeeding. After weaning, NL and SL mouse were fed normal diet. We generated an adeno-associated virus (AAV) carrying short hairpin RNA (shRNA) against Ripk3 and an empty vector as a control. The NL and SL groups were treated intravenously with 1×1012 vector genome of AAV vectors at week 6. The SL group showed a higher body weight than the NL group from week 3 of age through adulthood. At weeks 6 and 13, the SL group exhibited impaired glucose and insulin tolerance, RIPK3 up-regulation, and lipid accumulation in liver and adipose tissues. In the SL group, the genes involved in lipid synthesis and lipolysis were increased, whereas fatty acid β-oxidation-related genes were weakened in adipose tissue and liver. At week 13, AAV-shRNA-Ripk3 ameliorated adipose tissue hypertrophy, hepatic steatosis, insulin resistance, and dysregulated lipid metabolism in the adipose tissue and liver of SL mice. These findings support a novel mechanism underlying the pathogenesis of postnatal overfeeding-related metabolic disorders and suggest potential therapeutic targets.
Collapse
Affiliation(s)
- Dandan Zhu
- Department of Child Health Care, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing 210008, China
| | - Wen Zheng
- Department of Child Health Care, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing 210008, China
| | - Jiasi Kuang
- Department of Child Health Care, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing 210008, China
| | - Yueshu Wang
- Department of Child Health Care, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing 210008, China
| | - Xueting Deng
- Medical Center for Digestive Diseases, Second Affiliated Hospital of Nanjing Medical University, 121 Jiangjiayuan Road, Nanjing 210011, China
| | - Xiaonan Li
- Department of Child Health Care, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing 210008, China.
| | - Wei Zhou
- Department of Child Health Care, Children's Hospital of Nanjing Medical University, 72 Guangzhou Road, Nanjing 210008, China.
| |
Collapse
|
5
|
Bontempo P, Capasso L, De Masi L, Nebbioso A, Rigano D. Therapeutic Potential of Natural Compounds Acting through Epigenetic Mechanisms in Cardiovascular Diseases: Current Findings and Future Directions. Nutrients 2024; 16:2399. [PMID: 39125279 PMCID: PMC11314203 DOI: 10.3390/nu16152399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/11/2024] [Accepted: 07/20/2024] [Indexed: 08/12/2024] Open
Abstract
Cardiovascular diseases (CVDs) remain a leading global cause of morbidity and mortality. These diseases have a multifaceted nature being influenced by a multitude of biochemical, genetic, environmental, and behavioral factors. Epigenetic modifications have a crucial role in the onset and progression of CVD. Epigenetics, which regulates gene activity without altering the DNA's primary structure, can modulate cardiovascular homeostasis through DNA methylation, histone modification, and non-coding RNA regulation. The effects of environmental stimuli on CVD are mediated by epigenetic changes, which can be reversible and, hence, are susceptible to pharmacological interventions. This represents an opportunity to prevent diseases by targeting harmful epigenetic modifications. Factors such as high-fat diets or nutrient deficiencies can influence epigenetic enzymes, affecting fetal growth, metabolism, oxidative stress, inflammation, and atherosclerosis. Recent studies have shown that plant-derived bioactive compounds can modulate epigenetic regulators and inflammatory responses, contributing to the cardioprotective effects of diets. Understanding these nutriepigenetic effects and their reversibility is crucial for developing effective interventions to combat CVD. This review delves into the general mechanisms of epigenetics, its regulatory roles in CVD, and the potential of epigenetics as a CVD therapeutic strategy. It also examines the role of epigenetic natural compounds (ENCs) in CVD and their potential as intervention tools for prevention and therapy.
Collapse
Affiliation(s)
- Paola Bontempo
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via L. De Crecchio 7, 80138 Naples, Italy; (L.C.); (A.N.)
| | - Lucia Capasso
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via L. De Crecchio 7, 80138 Naples, Italy; (L.C.); (A.N.)
| | - Luigi De Masi
- National Research Council (CNR), Institute of Biosciences and BioResources (IBBR), Via Università 133, 80055 Portici, Italy
| | - Angela Nebbioso
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via L. De Crecchio 7, 80138 Naples, Italy; (L.C.); (A.N.)
| | - Daniela Rigano
- Department of Pharmacy, University of Naples Federico II, Via Montesano 49, 80131 Naples, Italy;
| |
Collapse
|
6
|
Liang H, Zheng J, Sun Y. Prevalence and risk factors associated with circadian syndrome in community-dwelling middle-aged to older adults: Based on health ecology model. Sleep Med 2024; 119:210-213. [PMID: 38703604 DOI: 10.1016/j.sleep.2024.04.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 03/31/2024] [Accepted: 04/29/2024] [Indexed: 05/06/2024]
Abstract
OBJECTIVES To explore the prevalence and risk factors associated with circadian syndrome (CricS) in community-dwelling middle-aged to older adults. METHOD We performed a cross-sectional analysis of 13,516 participants from the China Health and Retirement Longitudinal Study (CHARLS). We used logistic regression to compute the odds ratios (OR) and 95 % confidence intervals (Cls), using covariates derived through the health ecology model. RESULTS The overall prevalence of CricS was 31.5 % (25.0 % males and 37.1 % females). With controlling all covariates, social isolation (OR 1.164, 95%CI 1.033-1.310), irritable mood (OR 1.689, 95%CI 1.488-1.917), fear responses (OR 1.546, 95%CI 1.262-1.894), chronic disease (OR 1.577, 95%CI 1.392-1.788), and financial debt (OR 0.806, 95%CI 0.657-0.990) were significantly correlated with increased CricS risk in males, whereas CricS syndrome was significantly associated with age (OR 1.285, 95%CI 1.214-1.361), married (OR 1.258, 95%CI 1.089-1.452), current drinkers (OR 0.835, 95%CI 0.716-0.974), social isolation (OR 1.175, 95%CI 1.065-1.296), irritable mood (OR 1.346, 95%CI 1.210-1.497), fear responses (OR 1.202, 95%CI 1.047-1.378), chronic disease (OR 1.363, 95%CI 1.225-1.517), chronic pain (OR 1.177, 95%CI 1.058-1.309), and universal basic income (OR 0.742, 95%CI 0.611-0.900) in females. CONCLUSION CricS is common in middle-aged to older adults, and health behavior factors have an important impact on CricS. The potential predictors identified for CricS should be further studied to prevent the occurrence of adverse health events in the presenium stage.
Collapse
Affiliation(s)
- Haixu Liang
- School of Public Health, Xiamen University, Xiamen, China.
| | - Jiaqi Zheng
- School of Public Health, Shenyang Medical College, Shenyang, China.
| | - Yongkang Sun
- Traditional Chinese Medicine Department, Tianjin Baodi Hospital, Tianjin, China.
| |
Collapse
|
7
|
Dymek A, Oleksy Ł, Stolarczyk A, Bartosiewicz A. Choline-An Underappreciated Component of a Mother-to-Be's Diet. Nutrients 2024; 16:1767. [PMID: 38892700 PMCID: PMC11174651 DOI: 10.3390/nu16111767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 05/27/2024] [Accepted: 06/02/2024] [Indexed: 06/21/2024] Open
Abstract
The nutritional status of the mother-to-be has a key impact on the proper development of the fetus. Although all nutrients are important for the developing baby, recent research indicates the importance of adequate choline intake during the periconceptional period, pregnancy, and lactation. Choline plays a key role in the biosynthesis of cell membranes, supporting liver function, neurotransmission, brain development, and DNA and histone methylation. Choline participates in the formation of a child's nervous system, supports its cognitive development, and reduces the risk of neural tube defects. The human body is incapable of producing sufficient choline to meet its needs; therefore, it must be obtained from the diet. Current data indicate that most women in their reproductive years do not achieve the recommended daily intake of choline. The presented narrative review indicates the importance of educating mothers-to-be and thereby increasing their awareness of the effects of choline on maternal and child health, which can lead to a more aware and healthy pregnancy and proper child development.
Collapse
Affiliation(s)
- Agnieszka Dymek
- Students Scientific Club of Dietetics, Institute of Health Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland
| | - Łukasz Oleksy
- Department of Physiotherapy, Faculty of Health Sciences, Jagiellonian University Medical College, 31-008 Kraków, Poland
| | - Artur Stolarczyk
- Department of Orthopedics and Rehabilitation, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Anna Bartosiewicz
- Institute of Health Sciences, Medical College of Rzeszow University, 35-959 Rzeszow, Poland
| |
Collapse
|
8
|
Alves JGB, Alves LV. Early-life nutrition and adult-life outcomes. J Pediatr (Rio J) 2024; 100 Suppl 1:S4-S9. [PMID: 37813343 PMCID: PMC10960187 DOI: 10.1016/j.jped.2023.08.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 08/25/2023] [Indexed: 10/11/2023] Open
Abstract
OBJECTIVES To verify the association between early-life nutrition and chronic adult diseases. DATA SOURCES Medline, Embase, Cochrane Database, and Lilacs. SUMMARY OF FINDS The Developmental Origins of Health and Disease (DOHaD) hypothesis postulates that a mismatch between early-life circumstances and later-life situations may have an impact on chronic diseases. In this review, the authors emphasize the research supporting the impact of early nutrition on the origins of adult height, obesity and metabolic syndrome, type 2 diabetes mellitus, cardiovascular diseases, and reproductive outcomes. CONCLUSION Even though this is a new topic and there are still many research questions to be answered, there is strong evidence that both deficiency and excess nutrition in early life can cause epigenetic changes that have effects that last a lifetime and contribute to the development of chronic diseases. Public health efforts to protect adults from getting chronic diseases should focus on nutrition in the first 1000 days of life, from conception to the end of the second year of life.
Collapse
Affiliation(s)
| | - Lucas Victor Alves
- Department of Neuropediatrics, Instituto de Medicina Integral Prof. Fernando Figueira (IMIP), Recife, PE, Brazil
| |
Collapse
|
9
|
Yu G, Tam HCH, Huang C, Shi M, Lim CKP, Chan JCN, Ma RCW. Lessons and Applications of Omics Research in Diabetes Epidemiology. Curr Diab Rep 2024; 24:27-44. [PMID: 38294727 PMCID: PMC10874344 DOI: 10.1007/s11892-024-01533-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/04/2024] [Indexed: 02/01/2024]
Abstract
PURPOSE OF REVIEW Recent advances in genomic technology and molecular techniques have greatly facilitated the identification of disease biomarkers, advanced understanding of pathogenesis of different common diseases, and heralded the dawn of precision medicine. Much of these advances in the area of diabetes have been made possible through deep phenotyping of epidemiological cohorts, and analysis of the different omics data in relation to detailed clinical information. In this review, we aim to provide an overview on how omics research could be incorporated into the design of current and future epidemiological studies. RECENT FINDINGS We provide an up-to-date review of the current understanding in the area of genetic, epigenetic, proteomic and metabolomic markers for diabetes and related outcomes, including polygenic risk scores. We have drawn on key examples from the literature, as well as our own experience of conducting omics research using the Hong Kong Diabetes Register and Hong Kong Diabetes Biobank, as well as other cohorts, to illustrate the potential of omics research in diabetes. Recent studies highlight the opportunity, as well as potential benefit, to incorporate molecular profiling in the design and set-up of diabetes epidemiology studies, which can also advance understanding on the heterogeneity of diabetes. Learnings from these examples should facilitate other researchers to consider incorporating research on omics technologies into their work to advance the field and our understanding of diabetes and its related co-morbidities. Insights from these studies would be important for future development of precision medicine in diabetes.
Collapse
Affiliation(s)
- Gechang Yu
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, HKSAR, China
- Chinese University of Hong Kong- Shanghai Jiao Tong University Joint Research Centre in Diabetes Genomics and Precision Medicine, Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, HKSAR, China
- Laboratory for Molecular Epidemiology in Diabetes, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, HKSAR, China
| | - Henry C H Tam
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, HKSAR, China
- Chinese University of Hong Kong- Shanghai Jiao Tong University Joint Research Centre in Diabetes Genomics and Precision Medicine, Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, HKSAR, China
- Laboratory for Molecular Epidemiology in Diabetes, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, HKSAR, China
| | - Chuiguo Huang
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, HKSAR, China
- Chinese University of Hong Kong- Shanghai Jiao Tong University Joint Research Centre in Diabetes Genomics and Precision Medicine, Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, HKSAR, China
- Laboratory for Molecular Epidemiology in Diabetes, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, HKSAR, China
| | - Mai Shi
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, HKSAR, China
- Chinese University of Hong Kong- Shanghai Jiao Tong University Joint Research Centre in Diabetes Genomics and Precision Medicine, Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, HKSAR, China
- Laboratory for Molecular Epidemiology in Diabetes, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, HKSAR, China
| | - Cadmon K P Lim
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, HKSAR, China
- Chinese University of Hong Kong- Shanghai Jiao Tong University Joint Research Centre in Diabetes Genomics and Precision Medicine, Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, HKSAR, China
- Laboratory for Molecular Epidemiology in Diabetes, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, HKSAR, China
| | - Juliana C N Chan
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, HKSAR, China
- Chinese University of Hong Kong- Shanghai Jiao Tong University Joint Research Centre in Diabetes Genomics and Precision Medicine, Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, HKSAR, China
- Laboratory for Molecular Epidemiology in Diabetes, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, HKSAR, China
| | - Ronald C W Ma
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, HKSAR, China.
- Chinese University of Hong Kong- Shanghai Jiao Tong University Joint Research Centre in Diabetes Genomics and Precision Medicine, Hong Kong Institute of Diabetes and Obesity, The Chinese University of Hong Kong, Hong Kong, HKSAR, China.
- Laboratory for Molecular Epidemiology in Diabetes, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, HKSAR, China.
| |
Collapse
|
10
|
Tain YL, Hsu CN. Nutritional Approaches Targeting Gut Microbiota in Oxidative-Stress-Associated Metabolic Syndrome: Focus on Early Life Programming. Nutrients 2024; 16:683. [PMID: 38474810 DOI: 10.3390/nu16050683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/24/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Metabolic syndrome (MetS) denotes a constellation of risk factors associated with the development of cardiovascular disease, with its roots potentially traced back to early life. Given the pivotal role of oxidative stress and dysbiotic gut microbiota in MetS pathogenesis, comprehending their influence on MetS programming is crucial. Targeting these mechanisms during the early stages of life presents a promising avenue for preventing MetS later in life. This article begins by examining detrimental insults during early life that impact fetal programming, ultimately contributing to MetS in adulthood. Following that, we explore the role of oxidative stress and the dysregulation of gut microbiota in the initiation of MetS programming. The review also consolidates existing evidence on how gut-microbiota-targeted interventions can thwart oxidative-stress-associated MetS programming, encompassing approaches such as probiotics, prebiotics, postbiotics, and the modulation of bacterial metabolites. While animal studies demonstrate the favorable effects of gut-microbiota-targeted therapy in mitigating MetS programming, further clinical investigations are imperative to enhance our understanding of manipulating gut microbiota and oxidative stress for the prevention of MetS.
Collapse
Affiliation(s)
- You-Lin Tain
- Division of Pediatric Nephrology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chien-Ning Hsu
- Department of Pharmacy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan
- School of Pharmacy, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| |
Collapse
|
11
|
Faa G, Fanos V, Manchia M, Van Eyken P, Suri JS, Saba L. The fascinating theory of fetal programming of adult diseases: A review of the fundamentals of the Barker hypothesis. J Public Health Res 2024; 13:22799036241226817. [PMID: 38434579 PMCID: PMC10908242 DOI: 10.1177/22799036241226817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 12/27/2023] [Indexed: 03/05/2024] Open
Abstract
The theory of fetal programming of adult diseases was first proposed by David J.P. Barker in the eighties of the previous century, to explain the higher susceptibility of some people toward the development of ischemic heart disease. According to his hypothesis, poor maternal living conditions during gestation represent an important risk factor for the onset of atherosclerotic heart disease later in life. The analysis of the early phases of fetal development is a fundamental tool for the risk stratification of children and adults, allowing the identification of susceptible or resistant subjects to multiple diseases later in life. Here, we provide a narrative summary of the most relevant evidence supporting the Barker hypothesis in multiple fields of medicine, including neuropsychiatric disorders, such as Parkinson disease and Alzheimer disease, kidney failure, atherosclerosis, coronary heart disease, stroke, diabetes, cancer onset and progression, metabolic syndrome, and infectious diseases including COVID-19. Given the consensus on the role of body weight at birth as a practical indicator of the fetal nutritional status during gestation, every subject with a low birth weight should be considered an "at risk" subject for the development of multiple diseases later in life. The hypothesis of the "physiological regenerative medicine," able to improve fetal organs' development in the perinatal period is discussed, in the light of recent experimental data indicating Thymosin Beta-4 as a powerful growth promoter when administered to pregnant mothers before birth.
Collapse
Affiliation(s)
- Gavino Faa
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
- Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, USA
| | - Vassilios Fanos
- Unit of Neonatology and NICU Center, Department of Surgical Sciences, University of Cagliari, Cagliari, Italy
| | - Mirko Manchia
- Unit of Psychiatry, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Peter Van Eyken
- Department of Pathology, UZ Genk Regional Hospital, Genk, Belgium
| | - Jasjit S. Suri
- Stroke Diagnostic and Monitoring Division, Atheropoint, Roseville, CA, USA
| | - Luca Saba
- Unit of Radiology, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| |
Collapse
|
12
|
Kelleher CC, Kelly GE, Segurado R, Briody J, Sellers AM, McCalman J. Epidemiological transition: a historical analysis of immigration patterns by country of origin (1861-1986) related to circulatory system diseases and all-cause mortality in twentieth-century Australia. BMJ Open 2023; 13:e070996. [PMID: 38000816 PMCID: PMC10679994 DOI: 10.1136/bmjopen-2022-070996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Circulatory system disease (CSD) patterns vary over time and between countries, related to lifestyle risk factors, associated in turn with socioeconomic circumstances. Current global CSD epidemics in developing economies are similar in scale to those observed previously in the USA and Australasia. Australia exhibits an important macroeconomic phenomenon as a rapidly transitioning economy with high immigration throughout the nineteenth and twentieth centuries. We wished to examine how that historical immigration related to CSD patterns subsequently. METHODS AND SETTING We provide a novel empirical analysis employing census-derived place of birth by age bracket and sex from 1891 to 1986, in order to map patterns of immigration against CSD mortality rates from 1907 onwards. Age-specific generalised additive models for both CSD mortality in the general population, and all-cause mortality for the foreign-born (FB) only, from 1910 to 1980 were also devised for both males and females. RESULTS The percentage of FB fell from 32% in 1891 to 9.8% in 1947. Rates of CSD rose consistently, particularly from the 1940s onwards, peaked in the 1960s, then declined sharply in the 1980s and showed a strong period effect across age groups and genders. The main effects of age and census year and their interaction were highly statistically significant for CSD mortality for males (p<0.001, each term) and for females (p<0.001, each term). The main effect of age and year were statistically significant for all-cause mortality minus net migration rates for the FB females (each p<0.001), and for FB males, age (p<0.001) was significant. CONCLUSIONS We argue our empirical calculations, supported by historical and socioepidemiological evidence, employing immigration patterns as a proxy for epidemiological transition, affirm the life course hypothesis that both early life circumstances and later life lifestyle drive CSD patterns.
Collapse
Affiliation(s)
| | - Gabrielle E Kelly
- School of Mathematical Sciences, University College Dublin, Dublin, Ireland
| | - Ricardo Segurado
- CSTAR, School of Public Health, Physiotherapy and Population Science, University College Dublin, Dublin, Ireland
| | - Jonathan Briody
- Royal College of Surgeons in Ireland Division of Population Health Sciences, Dublin, Ireland
| | - Alexander M Sellers
- Royal Adelaide Hospital Cardiology Services, Adelaide, South Australia, Australia
| | | |
Collapse
|
13
|
Wu Y, Zeng Y, Ren Y, Yu J, Zhang Q, Xiao X. Insights into RNA N6-methyladenosine in Glucose and Lipid Metabolic Diseases and Their Therapeutic Strategies. Endocrinology 2023; 165:bqad170. [PMID: 37950364 DOI: 10.1210/endocr/bqad170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 10/20/2023] [Accepted: 11/07/2023] [Indexed: 11/12/2023]
Abstract
The incidence of glucose and lipid metabolism diseases, including type 2 diabetes, obesity, metabolic syndrome, and nonalcoholic fatty liver disease, is rising, which places an enormous burden on people around the world. However, the mechanism behind these disorders remains incompletely understood. N6-methyladenosine (m6A) is 1 type of posttranscriptional RNA modification, and research has shown that it plays a crucial role in several metabolic diseases. m6A methylation is reversibly and dynamically regulated by methyltransferases (writers), demethylases (erasers), and m6A binding proteins (readers). Dysregulation of RNA m6A modification is related to different metabolic processes. Targeting RNA m6A methylation is a potential treatment strategy for these chronic metabolic diseases. This review discusses studies on RNA m6A modification in metabolic diseases and existing therapeutic drugs, with the aim of providing a concise perspective on its potential applications in managing metabolic disorders.
Collapse
Affiliation(s)
- Yifan Wu
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yuan Zeng
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yaolin Ren
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Jie Yu
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Qian Zhang
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Xinhua Xiao
- Key Laboratory of Endocrinology, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing 100730, China
| |
Collapse
|
14
|
Ren Z, Luo S, Cui J, Tang Y, Huang H, Ding G. Research Progress of Maternal Metabolism on Cardiac Development and Function in Offspring. Nutrients 2023; 15:3388. [PMID: 37571325 PMCID: PMC10420869 DOI: 10.3390/nu15153388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/18/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023] Open
Abstract
The developmental origin of health and disease (DOHaD) hypothesis refers to the adverse effects of suboptimal developmental environments during embryonic and early fetal stages on the long-term health of offspring. Intrauterine metabolic perturbations can profoundly impact organogenesis in offspring, particularly affecting cardiac development and giving rise to potential structural and functional abnormalities. In this discussion, we contemplate the existing understanding regarding the impact of maternal metabolic disorders, such as obesity, diabetes, or undernutrition, on the developmental and functional aspects of the offspring's heart. This influence has the potential to contribute to the susceptibility of offspring to cardiovascular health issues. Alteration in the nutritional milieu can influence mitochondrial function in the developing hearts of offspring, while also serving as signaling molecules that directly modulate gene expression. Moreover, metabolic disorders can exert influence on cardiac development-related genes epigenetically through DNA methylation, levels of histone modifications, microRNA expression, and other factors. However, the comprehensive understanding of the mechanistic underpinnings of these phenomena remains incomplete. Further investigations in this domain hold profound clinical significance, as they can contribute to the enhancement of public health and the prevention of cardiovascular diseases.
Collapse
Affiliation(s)
- Zhuoran Ren
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200001, China (H.H.)
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China
| | - Sisi Luo
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China
- Shanghai First Maternity and Infant Hospital, Shanghai 200126, China
| | - Jiajun Cui
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200001, China (H.H.)
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China
| | - Yunhui Tang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200001, China (H.H.)
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China
| | - Hefeng Huang
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200001, China (H.H.)
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China
| | - Guolian Ding
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai 200001, China (H.H.)
- Research Units of Embryo Original Diseases, Chinese Academy of Medical Sciences, Shanghai 200032, China
| |
Collapse
|
15
|
Oh J, Riek AE, Bauerle KT, Dusso A, McNerney KP, Barve RA, Darwech I, Sprague JE, Moynihan C, Zhang RM, Kutz G, Wang T, Xing X, Li D, Mrad M, Wigge NM, Castelblanco E, Collin A, Bambouskova M, Head RD, Sands MS, Bernal-Mizrachi C. Embryonic vitamin D deficiency programs hematopoietic stem cells to induce type 2 diabetes. Nat Commun 2023; 14:3278. [PMID: 37311757 PMCID: PMC10264405 DOI: 10.1038/s41467-023-38849-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 05/19/2023] [Indexed: 06/15/2023] Open
Abstract
Environmental factors may alter the fetal genome to cause metabolic diseases. It is unknown whether embryonic immune cell programming impacts the risk of type 2 diabetes in later life. We demonstrate that transplantation of fetal hematopoietic stem cells (HSCs) made vitamin D deficient in utero induce diabetes in vitamin D-sufficient mice. Vitamin D deficiency epigenetically suppresses Jarid2 expression and activates the Mef2/PGC1a pathway in HSCs, which persists in recipient bone marrow, resulting in adipose macrophage infiltration. These macrophages secrete miR106-5p, which promotes adipose insulin resistance by repressing PIK3 catalytic and regulatory subunits and down-regulating AKT signaling. Vitamin D-deficient monocytes from human cord blood have comparable Jarid2/Mef2/PGC1a expression changes and secrete miR-106b-5p, causing adipocyte insulin resistance. These findings suggest that vitamin D deficiency during development has epigenetic consequences impacting the systemic metabolic milieu.
Collapse
Affiliation(s)
- Jisu Oh
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Amy E Riek
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Kevin T Bauerle
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Medicine, VA Medical Center, St. Louis, MO, USA
| | - Adriana Dusso
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Kyle P McNerney
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Ruteja A Barve
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Isra Darwech
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Clare Moynihan
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Rong M Zhang
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Greta Kutz
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Ting Wang
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Xiaoyun Xing
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Daofeng Li
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Marguerite Mrad
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Nicholas M Wigge
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Alejandro Collin
- Instituto de Investigaciones en Ciencias de la Salud (INICSA), CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Monika Bambouskova
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Richard D Head
- Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA
| | - Mark S Sands
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Carlos Bernal-Mizrachi
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Medicine, VA Medical Center, St. Louis, MO, USA.
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
16
|
Ricci TA, Boonpattrawong N, Laher I, Devlin AM. Maternal nutrition and effects on offspring vascular function. Pflugers Arch 2023:10.1007/s00424-023-02807-x. [PMID: 37041303 DOI: 10.1007/s00424-023-02807-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/24/2023] [Accepted: 03/27/2023] [Indexed: 04/13/2023]
Abstract
Maternal nutrition during pregnancy may have profound effects on the developing fetus and impact risk for cardiovascular disease later in life. Here, we provide a narrative review on the impact of maternal diet during pregnancy on offspring vascular function. We review studies reporting effects of maternal micronutrient (folic acid, iron) intakes, high-fat diets, dietary energy restriction, and low protein intake on offspring endothelial function. We discuss the differences in study design and outcomes and potential underlying mechanisms contributing to the vascular phenotypes observed in the offspring. We further highlight key gaps in the literature and identify targets for future investigations.
Collapse
Affiliation(s)
- Taylor A Ricci
- Department of Pediatrics, The University of British Columbia, Vancouver, British Columbia, Canada
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Nicha Boonpattrawong
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Ismail Laher
- Department of Anesthesiology, Pharmacology, and Therapeutics, The University of British Columbia, Vancouver, British Columbia, Canada
| | - Angela M Devlin
- Department of Pediatrics, The University of British Columbia, Vancouver, British Columbia, Canada.
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.
| |
Collapse
|
17
|
Adeodato CSR, Soares-Lima SC, Batista PV, Fagundes MCN, Camuzi D, Tavares SJO, Pinto LFR, Scelza MFZ. Interleukin 6 and Interleukin 1β hypomethylation and overexpression are common features of apical periodontitis: a case-control study with gingival tissue as control. Arch Oral Biol 2023; 150:105694. [PMID: 37043986 DOI: 10.1016/j.archoralbio.2023.105694] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/20/2023] [Accepted: 04/04/2023] [Indexed: 04/08/2023]
Abstract
OBJECTIVES Apical periodontitis is a periradicular tissue disorder that usually arises from infection by microorganisms in the root canal system resulting in local bone resorption. This usually involves the dysregulation of inflammatory mediators, which can be mediated by epigenetic mechanisms. Thus, the objective of this study was to evaluate Interleukin 6 (IL6) and Interleukin 1β (IL1β) and DNA methylation and gene expression levels in apical periodontitis. METHODS Gene expression was analyzed in 60 participants using quantitative polymerase chain reaction, while the methylation levels of IL6 and IL1β promoters were analyzed in 72 patients using pyrosequencing. All statistical analyzes were performed using the GraphPad Prism software version 8.0. The p value was considered statistically significant when < 0.05. RESULTS A significantly higher IL6 and IL1β expression levels were observed in cases relative to controls (fold-changes of 27.4 and 11.43, respectively, and p < 0.0001). By comparing the same groups, lower promoter methylation levels were observed for both genes in cases (methylation percentage delta relative to controls of -24.57% and -16.02%, respectively, and p < 0.0001). A significant inverse correlation between gene expression and promoter methylation was observed for both IL6 (p = 0.0002) and IL1β (p = 0.001). Neither IL6 expression nor promoter methylation were significantly associated with cases' age, smoking history, alcohol consumption history or sex. For IL1β, alcoholic cases showed lower methylation level relative to non-alcoholic cases (p = 0.01), while females showed higher methylation levels relative to males (p = 0.03). CONCLUSIONS Our data suggest a role for DNA methylation in IL6 and IL1β upregulation in apical periodontitis.
Collapse
Affiliation(s)
- Caroline Sousa Ribeiro Adeodato
- Post-graduation Program in Dentistry of Fluminense Federal University (UFF), Mario Santos Braga Street, no 28, 24020-140 Niteroi, RJ, Brazil
| | - Sheila Coelho Soares-Lima
- Molecular Carcinogenesis Program of National Cancer Institute (INCA), André Cavalcante Street, no 37, 20231-050 Rio de Janeiro, Brazil
| | - Paula Vieira Batista
- Molecular Carcinogenesis Program of National Cancer Institute (INCA), André Cavalcante Street, no 37, 20231-050 Rio de Janeiro, Brazil
| | - Marina Chianello Nicolau Fagundes
- Molecular Carcinogenesis Program of National Cancer Institute (INCA), André Cavalcante Street, no 37, 20231-050 Rio de Janeiro, Brazil
| | - Diego Camuzi
- Molecular Carcinogenesis Program of National Cancer Institute (INCA), André Cavalcante Street, no 37, 20231-050 Rio de Janeiro, Brazil
| | - Sandro Junio Oliveira Tavares
- Post-graduation Program in Dentistry of Fluminense Federal University (UFF), Mario Santos Braga Street, no 28, 24020-140 Niteroi, RJ, Brazil
| | - Luis Felipe Ribeiro Pinto
- Molecular Carcinogenesis Program of National Cancer Institute (INCA), André Cavalcante Street, no 37, 20231-050 Rio de Janeiro, Brazil; Biochemistry Department, Biology Institute, State University of Rio de Janeiro, Boulevard 28 de Setembro, 87 - Vila Isabel, 20511-010 Rio de Janeiro, Brazil
| | - Miriam Fatima Zaccaro Scelza
- Endodontics Department, Faculty of Dentistry, Fluminense Federal University (UFF), Mario Santos Braga Street, no 28, 24020-140 Niteroi, RJ, Brazil.
| |
Collapse
|
18
|
Örkenby L, Skog S, Ekman H, Gozzo A, Kugelberg U, Ramesh R, Magadi S, Zambanini G, Nordin A, Cantú C, Nätt D, Öst A. Stress-sensitive dynamics of miRNAs and Elba1 in Drosophila embryogenesis. Mol Syst Biol 2023; 19:e11148. [PMID: 36938679 PMCID: PMC10167479 DOI: 10.15252/msb.202211148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 02/15/2023] [Accepted: 02/23/2023] [Indexed: 03/21/2023] Open
Abstract
Early-life stress can result in life-long effects that impact adult health and disease risk, but little is known about how such programming is established and maintained. Here, we show that such epigenetic memories can be initiated in the Drosophila embryo before the major wave of zygotic transcription, and higher-order chromatin structures are established. An early short heat shock results in elevated levels of maternal miRNA and reduced levels of a subgroup of zygotic genes in stage 5 embryos. Using a Dicer-1 mutant, we show that the stress-induced decrease in one of these genes, the insulator-binding factor Elba1, is dependent on functional miRNA biogenesis. Reduction in Elba1 correlates with the upregulation of early developmental genes and promotes a sustained weakening of heterochromatin in the adult fly as indicated by an increased expression of the PEV wm4h reporter. We propose that maternal miRNAs, retained in response to an early embryonic heat shock, shape the subsequent de novo heterochromatin establishment that occurs during early development via direct or indirect regulation of some of the earliest expressed genes, including Elba1.
Collapse
Affiliation(s)
- Lovisa Örkenby
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Signe Skog
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Helen Ekman
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Alessandro Gozzo
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Unn Kugelberg
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Rashmi Ramesh
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Srivathsa Magadi
- Division of Neurobiology (NEURO), Linköping University, Linköping, Sweden
| | - Gianluca Zambanini
- Division of Molecular Medicine and Virology (MMV), Linköping University, Linköping, Sweden
| | - Anna Nordin
- Division of Molecular Medicine and Virology (MMV), Linköping University, Linköping, Sweden
| | - Claudio Cantú
- Division of Molecular Medicine and Virology (MMV), Linköping University, Linköping, Sweden
| | - Daniel Nätt
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Anita Öst
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
19
|
Ribas-Aulinas F, Ribo S, Casas E, Mourin-Fernandez M, Ramon-Krauel M, Diaz R, Lerin C, Kalko SG, Vavouri T, Jimenez-Chillaron JC. Intergenerational Inheritance of Hepatic Steatosis in a Mouse Model of Childhood Obesity: Potential Involvement of Germ-Line microRNAs. Nutrients 2023; 15:nu15051241. [PMID: 36904241 PMCID: PMC10005268 DOI: 10.3390/nu15051241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/23/2023] [Accepted: 02/27/2023] [Indexed: 03/05/2023] Open
Abstract
Childhood obesity increases the risk of developing metabolic syndrome later in life. Moreover, metabolic dysfunction may be inherited into the following generation through non-genomic mechanisms, with epigenetics as a plausible candidate. The pathways involved in the development of metabolic dysfunction across generations in the context of childhood obesity remain largely unexplored. We have developed a mouse model of early adiposity by reducing litter size at birth (small litter group, SL: 4 pups/dam; control group, C: 8 pups/dam). Mice raised in small litters (SL) developed obesity, insulin resistance and hepatic steatosis with aging. Strikingly, the offspring of SL males (SL-F1) also developed hepatic steatosis. Paternal transmission of an environmentally induced phenotype strongly suggests epigenetic inheritance. We analyzed the hepatic transcriptome in C-F1 and SL-F1 mice to identify pathways involved in the development of hepatic steatosis. We found that the circadian rhythm and lipid metabolic process were the ontologies with highest significance in the liver of SL-F1 mice. We explored whether DNA methylation and small non-coding RNAs might be involved in mediating intergenerational effects. Sperm DNA methylation was largely altered in SL mice. However, these changes did not correlate with the hepatic transcriptome. Next, we analyzed small non-coding RNA content in the testes of mice from the parental generation. Two miRNAs (miR-457 and miR-201) appeared differentially expressed in the testes of SL-F0 mice. They are known to be expressed in mature spermatozoa, but not in oocytes nor early embryos, and they may regulate the transcription of lipogenic genes, but not clock genes, in hepatocytes. Hence, they are strong candidates to mediate the inheritance of adult hepatic steatosis in our murine model. In conclusion, litter size reduction leads to intergenerational effects through non-genomic mechanisms. In our model, DNA methylation does not seem to play a role on the circadian rhythm nor lipid genes. However, at least two paternal miRNAs might influence the expression of a few lipid-related genes in the first-generation offspring, F1.
Collapse
Affiliation(s)
| | - Sílvia Ribo
- Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues, 08950 Barcelona, Spain
| | - Eduard Casas
- Josep Carreras Leukemia Research Institute (IJC), 08916 Badalona, Spain
| | | | - Marta Ramon-Krauel
- Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues, 08950 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ruben Diaz
- Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues, 08950 Barcelona, Spain
| | - Carles Lerin
- Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues, 08950 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Susana G. Kalko
- Vall d’Hebron Research Institute (VHIR), 08035 Barcelona, Spain
| | - Tanya Vavouri
- Josep Carreras Leukemia Research Institute (IJC), 08916 Badalona, Spain
| | - Josep C. Jimenez-Chillaron
- Institut de Recerca Sant Joan de Déu (IRSJD), Esplugues, 08950 Barcelona, Spain
- School of Medicine, University of Barcelona, L’Hospitalet, 08907 Barcelona, Spain
- Correspondence: or ; Tel.: +34-934024267
| |
Collapse
|
20
|
Zordão OP, Campolim CM, Yariwake VY, Castro G, Ferreira CKDO, Santos A, Norberto S, Veras MM, Saad MJA, Saldiva PHN, Kim YB, Prada PO. Maternal exposure to air pollution alters energy balance transiently according to gender and changes gut microbiota. Front Endocrinol (Lausanne) 2023; 14:1069243. [PMID: 37082122 PMCID: PMC10112381 DOI: 10.3389/fendo.2023.1069243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 03/07/2023] [Indexed: 04/22/2023] Open
Abstract
Introduction The timing of maternal exposure to air pollution is crucial to define metabolic changes in the offspring. Here we aimed to determine the most critical period of maternal exposure to particulate matter (PM2.5) that impairs offspring's energy metabolism and gut microbiota composition. Methods Unexposed female and male C57BL/6J mice were mated. PM2.5 or filtered air (FA) exposure occurred only in gestation (PM2.5/FA) or lactation (FA/PM2.5). We studied the offspring of both genders. Results PM2.5 exposure during gestation increased body weight (BW) at birth and from weaning to young in male adulthood. Leptin levels, food intake, Agrp, and Npy levels in the hypothalamus were also increased in young male offspring. Ikbke, Tnf increased in male PM2.5/FA. Males from FA/PM2.5 group were protected from these phenotypes showing higher O2 consumption and Ucp1 in the brown adipose tissue. In female offspring, we did not see changes in BW at weaning. However, adult females from PM2.5/FA displayed higher BW and leptin levels, despite increased energy expenditure and thermogenesis. This group showed a slight increase in food intake. In female offspring from FA/PM2.5, BW, and leptin levels were elevated. This group displayed higher energy expenditure and a mild increase in food intake. To determine if maternal exposure to PM2.5 could affect the offspring's gut microbiota, we analyzed alpha diversity by Shannon and Simpson indexes and beta diversity by the Linear Discriminant Analysis (LDA) in offspring at 30 weeks. Unlike males, exposure during gestation led to higher adiposity and leptin maintenance in female offspring at this age. Gestation exposure was associated with decreased alpha diversity in the gut microbiota in both genders. Discussion Our data support that exposure to air pollution during gestation is more harmful to metabolism than exposure during lactation. Male offspring had an unfavorable metabolic phenotype at a young age. However, at an older age, only females kept more adiposity. Ultimately, our data highlight the importance of controlling air pollution, especially during gestation.
Collapse
Affiliation(s)
- Olivia Pizetta Zordão
- Department of Internal Medicine, School of Medical Science, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Clara Machado Campolim
- Department of Internal Medicine, School of Medical Science, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Victor Yuji Yariwake
- Laboratory of Environmental and Experimental Pathology, Department of Pathology, University of Sao Paulo School of Medicine, Sao Paulo, SP, Brazil
| | - Gisele Castro
- Department of Internal Medicine, School of Medical Science, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | | | - Andrey Santos
- Department of Internal Medicine, School of Medical Science, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Sónia Norberto
- Department of Internal Medicine, School of Medical Science, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Mariana Matera Veras
- Laboratory of Environmental and Experimental Pathology, Department of Pathology, University of Sao Paulo School of Medicine, Sao Paulo, SP, Brazil
| | - Mario Jose Abdalla Saad
- Department of Internal Medicine, School of Medical Science, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Paulo Hilário Nascimento Saldiva
- Laboratory of Environmental and Experimental Pathology, Department of Pathology, University of Sao Paulo School of Medicine, Sao Paulo, SP, Brazil
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Patricia Oliveira Prada
- Department of Internal Medicine, School of Medical Science, State University of Campinas (UNICAMP), Campinas, SP, Brazil
- School of Applied Sciences, State University of Campinas (UNICAMP), Limeira, SP, Brazil
- *Correspondence: Patricia Oliveira Prada, ;
| |
Collapse
|
21
|
Manfredini M, Breschi M, Fornasin A, Esposito M. Maternal nutritional status and offspring childlessness: Evidence from the late-nineteenth to early-twentieth centuries in a group of Italian populations. POPULATION STUDIES 2022; 76:477-493. [PMID: 35899492 DOI: 10.1080/00324728.2022.2099566] [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: 12/25/2022]
Abstract
The role of maternal nutrition in affecting offspring fertility, through alteration of foetal programming, has been demonstrated in animal-based experiments. However, results from human populations appear inconsistent and sometimes contradictory, likely because they have been based on single famine events. In this paper, we adopt a different approach. We combine official annual time series of daily nutrient availability with a sample of women's reproductive histories from the 1961 Italian Census to investigate the role of maternal nutritional status in pregnancy on offspring childlessness. The analysis therefore covers cohorts of females born between 1861 and 1939. Our results show a negative association between calorie availability in pregnancy and the odds of offspring childlessness, whereas no association is found between protein availability and offspring childlessness. The consequences of poor calorie intake were aggravated during the summer, likely due to the participation of pregnant women in physically demanding work.
Collapse
|
22
|
Andrawus M, Sharvit L, Atzmon G. Epigenetics and Pregnancy: Conditional Snapshot or Rolling Event. Int J Mol Sci 2022; 23:12698. [PMID: 36293556 PMCID: PMC9603966 DOI: 10.3390/ijms232012698] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/12/2022] [Accepted: 10/19/2022] [Indexed: 11/17/2022] Open
Abstract
Epigenetics modification such as DNA methylation can affect maternal health during the gestation period. Furthermore, pregnancy can drive a range of physiological and molecular changes that have the potential to contribute to pathological conditions. Pregnancy-related risk factors include multiple environmental, behavioral, and hereditary factors that can impact maternal DNA methylation with long-lasting consequences. Identification of the epigenetic patterns linked to poor pregnancy outcomes is crucial since changes in DNA methylation patterns can have long-term effects. In this review, we provide an overview of the epigenetic changes that influence pregnancy-related molecular programming such as gestational diabetes, immune response, and pre-eclampsia, in an effort to close the gap in current understanding regarding interactions between the environment, the genetics of the fetus, and the pregnant woman.
Collapse
Affiliation(s)
| | | | - Gil Atzmon
- Department of Human Biology, University of Haifa, Haifa 3498838, Israel
| |
Collapse
|
23
|
Sinzato YK, Paula VG, Gallego FQ, Moraes-Souza RQ, Corrente JE, Volpato GT, Damasceno DC. Maternal Diabetes and Postnatal High-Fat Diet on Pregnant Offspring. Front Cell Dev Biol 2022; 10:818621. [PMID: 35706903 PMCID: PMC9189289 DOI: 10.3389/fcell.2022.818621] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 04/12/2022] [Indexed: 12/22/2022] Open
Abstract
Maternal diabetes-induced fetal programming predisposes offspring to type 2 diabetes, cardiovascular disease, and obesity in adulthood. However, lifelong health and disease trajectories depend on several factors and nutrition is one of the main ones. We intend to understand the role of maternal diabetes-induced fetal programming and its association with a high-fat diet during lifelong in the female F1 generation focusing on reproductive outcomes and the possible changes in physiological systems during pregnancy as well as the repercussions on the F2 generation at birth. For this, we composed four groups: F1 female pups from control (OC) or from diabetic dams (OD) and fed with standard (SD) or high-fat diet from weaning to full-term pregnancy. During pregnancy, glucose intolerance and insulin sensitivity were evaluated. In a full-term pregnancy, the maternal blood and liver were collected to evaluate redox status markers. The maternal blood, placental tissue, and fetal blood (pool) were collected to evaluate adiponectin and leptin levels. Maternal reproductive parameters were evaluated as well. Maternal diabetes and high-fat diet consumption, in isolation, were both responsible for increased infertility rates and fasting glucose levels in the F1 generation and fetal growth restriction in the F2 generation. The association of both conditions showed, in addition to those, increased lipoperoxidation in maternal erythrocytes, regardless of the increased endogenous antioxidant enzyme activities, glucose intolerance, decreased number of implantation sites and live fetuses, decreased litter, fetal and placental weight, increased preimplantation losses, and increased fetal leptin serum levels. Thus, our findings show that fetal programming caused by maternal diabetes or lifelong high-fat diet consumption leads to similar repercussions in pregnant rats. In addition, the association of both conditions was responsible for glucose intolerance and oxidative stress in the first generation and increased fetal leptin levels in the second generation. Thus, our findings show both the F1 and F2 generations harmed health after maternal hyperglycemic intrauterine environment and exposure to a high-fat diet from weaning until the end of pregnancy.
Collapse
Affiliation(s)
- Yuri Karen Sinzato
- Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu, Brazil
| | - Verônyca Gonçalves Paula
- Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu, Brazil
| | - Franciane Quintanilha Gallego
- Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu, Brazil
| | - Rafaianne Q. Moraes-Souza
- Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu, Brazil
| | - José Eduardo Corrente
- Research Support Office, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu, Brazil
| | - Gustavo Tadeu Volpato
- Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso (UFMT), Barra do Garças, Brazil
| | - Débora Cristina Damasceno
- Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Botucatu Medical School, Sao Paulo State University (UNESP), Botucatu, Brazil
- *Correspondence: Débora Cristina Damasceno,
| |
Collapse
|
24
|
Freitas RGBDON, Vasques ACJ, Fernandes GDR, Ribeiro FB, Solar I, Barbosa MG, Pititto BDA, Geloneze B, Ferreira SRG. Associations of Blautia Genus With Early-Life Events and Later Phenotype in the NutriHS. Front Cell Infect Microbiol 2022; 12:838750. [PMID: 35646726 PMCID: PMC9134825 DOI: 10.3389/fcimb.2022.838750] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 04/11/2022] [Indexed: 12/15/2022] Open
Abstract
Introduction Early-life events are associated with the risk of obesity and comorbidities later in life. The gut microbiota—whose composition is influenced by genetics and environmental factors—could be involved. Since the microbiota affects metabolism and fat storage, early-life insults could contribute to the occurrence of obesity driven, in part, by microbiota composition. We examined associations of gut bacteria with early-life events, nutritional status, and body composition in the Nutritionist’s Health Study (NutriHS). Methods A cross-sectional study of 114 female participants examining early-life data, body composition, and biological samples was conducted. Fecal microbiota structure was determined targeting the V4 region of the 16S rRNA gene. Principal coordinates analysis (PCoA) and permutational multivariate analysis of variance (PERMANOVA) were used to test the impact of variables on microbial diversity. Profiles were identified using the Jensen-Shannon divergence matrix and Calinski–Harabasz index. Differential abundance between the categories of exclusive breastfeeding duration and nutritional status was tested using DESeq2. Results In the sample [median age 28 years and body mass index (BMI) 24.5 kg/m2], 2 microbiota profiles driven by the Blautia or Prevotella genus were identified. An estimated 9.1% of the variation was explained by the profiles (p < 0.001), 2.1% by nutritional status (p = 0.004), and 1.8% by exclusive breastfeeding (p = 0.012). The proportion of participants with BMI <25 kg/m2 and who were breastfed for at least 6 months was higher in the Blautia profile (p < 0.05). Conclusion Findings in a Blautia-driven profile of healthy women reinforce that early-life events play a role in defining gut microbiota composition, confirming the importance of exclusive breastfeeding for infant gut colonization in establishing a protective profile against adiposity-related outcomes in adulthood.
Collapse
Affiliation(s)
- Renata G. Borges de Oliveira Nascimento Freitas
- Department of Epidemiology, School of Public Health, University of São Paulo, São Paulo, Brazil
- Laboratory of Investigation in Metabolism and Diabetes, Gastrocentro, School of Medical Sciences, University of Campinas, Campinas, Brazil
| | - Ana Carolina J. Vasques
- Laboratory of Investigation in Metabolism and Diabetes, Gastrocentro, School of Medical Sciences, University of Campinas, Campinas, Brazil
- School of Applied Sciences, University of Campinas, Campinas, Brazil
| | | | - Francieli B. Ribeiro
- Laboratory of Investigation in Metabolism and Diabetes, Gastrocentro, School of Medical Sciences, University of Campinas, Campinas, Brazil
- School of Applied Sciences, University of Campinas, Campinas, Brazil
| | - Isabela Solar
- Laboratory of Investigation in Metabolism and Diabetes, Gastrocentro, School of Medical Sciences, University of Campinas, Campinas, Brazil
- School of Applied Sciences, University of Campinas, Campinas, Brazil
| | - Marina G. Barbosa
- School of Applied Sciences, University of Campinas, Campinas, Brazil
| | | | - Bruno Geloneze
- Laboratory of Investigation in Metabolism and Diabetes, Gastrocentro, School of Medical Sciences, University of Campinas, Campinas, Brazil
- Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Sandra Roberta G. Ferreira
- Department of Epidemiology, School of Public Health, University of São Paulo, São Paulo, Brazil
- *Correspondence: Sandra Roberta G. Ferreira,
| |
Collapse
|
25
|
Polverino A, Sorrentino P, Pesoli M, Mandolesi L. Nutrition and cognition across the lifetime: an overview on epigenetic mechanisms. AIMS Neurosci 2021; 8:448-476. [PMID: 34877399 PMCID: PMC8611190 DOI: 10.3934/neuroscience.2021024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 07/12/2021] [Indexed: 12/28/2022] Open
Abstract
The functioning of our brain depends on both genes and their interactions with environmental factors. The close link between genetics and environmental factors produces structural and functional cerebral changes early on in life. Understanding the weight of environmental factors in modulating neuroplasticity phenomena and cognitive functioning is relevant for potential interventions. Among these, nutrition plays a key role. In fact, the link between gut and brain (the gut-brain axis) is very close and begins in utero, since the Central Nervous System (CNS) and the Enteric Nervous System (ENS) originate from the same germ layer during the embryogenesis. Here, we investigate the epigenetic mechanisms induced by some nutrients on the cognitive functioning, which affect the cellular and molecular processes governing our cognitive functions. Furthermore, epigenetic phenomena can be positively affected by specific healthy nutrients from diet, with the possibility of preventing or modulating cognitive impairments. Specifically, we described the effects of several nutrients on diet-dependent epigenetic processes, in particular DNA methylation and histones post-translational modifications, and their potential role as therapeutic target, to describe how some forms of cognitive decline could be prevented or modulated from the early stages of life.
Collapse
Affiliation(s)
- Arianna Polverino
- Institute of Diagnosis and Treatment Hermitage Capodimonte, Naples, Italy.,Department of Motor and Wellness Sciences, University of Naples "Parthenope", Naples, Italy
| | - Pierpaolo Sorrentino
- Institut de Neurosciences des Systèmes, Aix-Marseille University, Marseille, France.,Institute of Applied Sciences and Intelligent Systems, National Research Council, Pozzuoli, Italy
| | - Matteo Pesoli
- Department of Motor and Wellness Sciences, University of Naples "Parthenope", Naples, Italy
| | - Laura Mandolesi
- Department of Humanities Studies, University of Naples Federico II, Naples, Italy
| |
Collapse
|
26
|
Nutritional Approach to Prevention and Treatment of Cardiovascular Disease in Childhood. Nutrients 2021; 13:nu13072359. [PMID: 34371871 PMCID: PMC8308497 DOI: 10.3390/nu13072359] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 07/05/2021] [Accepted: 07/08/2021] [Indexed: 12/19/2022] Open
Abstract
Coronary Heart Disease (CHD) is a major mortality and morbidity cause in adulthood worldwide. The atherosclerotic process starts even before birth, progresses through childhood and, if not stopped, eventually leads to CHD. Therefore, it is important to start prevention from the earliest stages of life. CHD prevention can be performed at different interventional stages: primordial prevention is aimed at preventing risk factors, primary prevention is aimed at early identification and treatment of risk factors, secondary prevention is aimed at reducing the risk of further events in those patients who have already experienced a CHD event. In this context, CHD risk stratification is of utmost importance, in order to tailor the preventive and therapeutic approach. Nutritional intervention is the milestone treatment in pediatric patients at increased CHD risk. According to the Developmental Origin of Health and Disease theory, the origins of lifestyle-related disease is formed in the so called “first thousand days” from conception, when an insult, either positive or negative, can cause life-lasting consequences. Nutrition is a positive epigenetic factor: an adequate nutritional intervention in a developmental critical period can change the outcome from childhood into adulthood.
Collapse
|
27
|
Tovar R, Vargas A, Aranda J, Sánchez-Salido L, González-González L, Chowen JA, Rodríguez de Fonseca F, Suárez J, Rivera P. Analysis of Both Lipid Metabolism and Endocannabinoid Signaling Reveals a New Role for Hypothalamic Astrocytes in Maternal Caloric Restriction-Induced Perinatal Programming. Int J Mol Sci 2021; 22:ijms22126292. [PMID: 34208173 PMCID: PMC8230792 DOI: 10.3390/ijms22126292] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 12/29/2022] Open
Abstract
Maternal malnutrition in critical periods of development increases the risk of developing short- and long-term diseases in the offspring. The alterations induced by this nutritional programming in the hypothalamus of the offspring are of special relevance due to its role in energy homeostasis, especially in the endocannabinoid system (ECS), which is involved in metabolic functions. Since astrocytes are essential for neuronal energy efficiency and are implicated in brain endocannabinoid signaling, here we have used a rat model to investigate whether a moderate caloric restriction (R) spanning from two weeks prior to the start of gestation to its end induced changes in offspring hypothalamic (a) ECS, (b) lipid metabolism (LM) and/or (c) hypothalamic astrocytes. Monitorization was performed by analyzing both the gene and protein expression of proteins involved in LM and ECS signaling. Offspring born from caloric-restricted mothers presented hypothalamic alterations in both the main enzymes involved in LM and endocannabinoids synthesis/degradation. Furthermore, most of these changes were similar to those observed in hypothalamic offspring astrocytes in culture. In conclusion, a maternal low caloric intake altered LM and ECS in both the hypothalamus and its astrocytes, pointing to these glial cells as responsible for a large part of the alterations seen in the total hypothalamus and suggesting a high degree of involvement of astrocytes in nutritional programming.
Collapse
Affiliation(s)
- Rubén Tovar
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29010 Málaga, Spain; (R.T.); (A.V.); (J.A.); (L.S.-S.); (L.G.-G.); (F.R.d.F.)
- UGC Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Andalucia Tech, Facultad de Medicina, Universidad de Málaga, Campus de Teatinos s/n, 29071 Málaga, Spain
| | - Antonio Vargas
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29010 Málaga, Spain; (R.T.); (A.V.); (J.A.); (L.S.-S.); (L.G.-G.); (F.R.d.F.)
- UGC Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Jesús Aranda
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29010 Málaga, Spain; (R.T.); (A.V.); (J.A.); (L.S.-S.); (L.G.-G.); (F.R.d.F.)
- Andalucia Tech, Facultad de Medicina, Universidad de Málaga, Campus de Teatinos s/n, 29071 Málaga, Spain
| | - Lourdes Sánchez-Salido
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29010 Málaga, Spain; (R.T.); (A.V.); (J.A.); (L.S.-S.); (L.G.-G.); (F.R.d.F.)
- UGC Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Laura González-González
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29010 Málaga, Spain; (R.T.); (A.V.); (J.A.); (L.S.-S.); (L.G.-G.); (F.R.d.F.)
| | - Julie A. Chowen
- Department of Endocrinology, Instituto de Investigación Biomédica la Princesa, Fundación Investigación Biomédica del Hospital Infantil Universitario Niño Jesús, 28009 Madrid, Spain;
- CIBEROBN (Centro de Investigación Biomédica en Red Sobre Fisiopatología de la Obesidad y Nutrición), Instituto de Salud Carlos III, 28009 Madrid, Spain
- IMDEA Food Institute, CEI UAM + CSIC, 28009 Madrid, Spain
| | - Fernando Rodríguez de Fonseca
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29010 Málaga, Spain; (R.T.); (A.V.); (J.A.); (L.S.-S.); (L.G.-G.); (F.R.d.F.)
- UGC Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Juan Suárez
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29010 Málaga, Spain; (R.T.); (A.V.); (J.A.); (L.S.-S.); (L.G.-G.); (F.R.d.F.)
- Departamento de Anatomía Humana, Medicina Legal e Historia de la Ciencia, Universidad de Málaga, 29071 Málaga, Spain
- Correspondence: (J.S.); (P.R.); Tel.: +34-952614012 (J.S.); +34-952614012 (P.R.)
| | - Patricia Rivera
- Instituto de Investigación Biomédica de Málaga-IBIMA, 29010 Málaga, Spain; (R.T.); (A.V.); (J.A.); (L.S.-S.); (L.G.-G.); (F.R.d.F.)
- UGC Salud Mental, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
- Correspondence: (J.S.); (P.R.); Tel.: +34-952614012 (J.S.); +34-952614012 (P.R.)
| |
Collapse
|
28
|
Epigenetic Changes Induced by Maternal Factors during Fetal Life: Implication for Type 1 Diabetes. Genes (Basel) 2021; 12:genes12060887. [PMID: 34201206 PMCID: PMC8227197 DOI: 10.3390/genes12060887] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/06/2021] [Accepted: 06/07/2021] [Indexed: 02/07/2023] Open
Abstract
Organ-specific autoimmune diseases, such as type 1 diabetes, are believed to result from T-cell-mediated damage of the target tissue. The immune-mediated tissue injury, in turn, is known to depend on complex interactions between genetic and environmental factors. Nevertheless, the mechanisms whereby environmental factors contribute to the pathogenesis of autoimmune diseases remain elusive and represent a major untapped target to develop novel strategies for disease prevention. Given the impact of the early environment on the developing immune system, epigenetic changes induced by maternal factors during fetal life have been linked to a likelihood of developing an autoimmune disease later in life. In humans, DNA methylation is the epigenetic mechanism most extensively investigated. This review provides an overview of the critical role of DNA methylation changes induced by prenatal maternal conditions contributing to the increased risk of immune-mediated diseases on the offspring, with a particular focus on T1D. A deeper understanding of epigenetic alterations induced by environmental stressors during fetal life may be pivotal for developing targeted prevention strategies of type 1 diabetes by modifying the maternal environment.
Collapse
|
29
|
Affiliation(s)
- Pam Factor-Litvak
- Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York
| |
Collapse
|
30
|
Impact of the exposome on the development and function of pancreatic β-cells. Mol Aspects Med 2021; 87:100965. [PMID: 33965231 DOI: 10.1016/j.mam.2021.100965] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 04/13/2021] [Accepted: 04/15/2021] [Indexed: 11/22/2022]
Abstract
The development and plasticity of the endocrine pancreas responds to both the intrauterine and postnatal exposome in a constant attempt to predict and respond to alterations in nutritional availability and metabolic requirements. Both under- and over-nutrition in utero, or exposure to adverse environmental pollutants or maternal behaviors, can each lead to altered β-cell or function at birth, and a subsequent mismatch in pancreatic hormonal demands and secretory capacity postnatally. This can be further exacerbated by metabolic stress postnatally such as from obesity or pregnancy, resulting in an increased risk of gestational diabetes, type 2 diabetes, and even type 1 diabetes. This review will discuss evidence identifying the cellular pathways in early life whereby the plasticity of the endocrine pancreatic can become pathologically limited. By necessity, much of this evidence has been gained from animal models, although extrapolation to human fetal development is possible from the fetal growth trajectory and study of the newborn. Cellular limitations to plasticity include the balance between β-cell proliferation and apoptosis, the appearance of β-cell oxidative stress, impaired glucose-stimulated insulin secretion, and sensitivity to circulating cytokines and responsiveness to programmed death receptor-1. Evidence suggests that many of the cellular pathways responsible for limiting β-cell plasticity are related to paracrine interactions within the islets of Langerhans.
Collapse
|
31
|
Wang M, Ibeagha-Awemu EM. Impacts of Epigenetic Processes on the Health and Productivity of Livestock. Front Genet 2021; 11:613636. [PMID: 33708235 PMCID: PMC7942785 DOI: 10.3389/fgene.2020.613636] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/21/2020] [Indexed: 12/23/2022] Open
Abstract
The dynamic changes in the epigenome resulting from the intricate interactions of genetic and environmental factors play crucial roles in individual growth and development. Numerous studies in plants, rodents, and humans have provided evidence of the regulatory roles of epigenetic processes in health and disease. There is increasing pressure to increase livestock production in light of increasing food needs of an expanding human population and environment challenges, but there is limited related epigenetic data on livestock to complement genomic information and support advances in improvement breeding and health management. This review examines the recent discoveries on epigenetic processes due to DNA methylation, histone modification, and chromatin remodeling and their impacts on health and production traits in farm animals, including bovine, swine, sheep, goat, and poultry species. Most of the reports focused on epigenome profiling at the genome-wide or specific genic regions in response to developmental processes, environmental stressors, nutrition, and disease pathogens. The bulk of available data mainly characterized the epigenetic markers in tissues/organs or in relation to traits and detection of epigenetic regulatory mechanisms underlying livestock phenotype diversity. However, available data is inadequate to support gainful exploitation of epigenetic processes for improved animal health and productivity management. Increased research effort, which is vital to elucidate how epigenetic mechanisms affect the health and productivity of livestock, is currently limited due to several factors including lack of adequate analytical tools. In this review, we (1) summarize available evidence of the impacts of epigenetic processes on livestock production and health traits, (2) discuss the application of epigenetics data in livestock production, and (3) present gaps in livestock epigenetics research. Knowledge of the epigenetic factors influencing livestock health and productivity is vital for the management and improvement of livestock productivity.
Collapse
Affiliation(s)
- Mengqi Wang
- Agriculture and Agri-Food Canada, Sherbrooke Research and Development Centre, Sherbrooke, QC, Canada
- Department of Animal Science, Laval University, Quebec, QC, Canada
| | - Eveline M. Ibeagha-Awemu
- Agriculture and Agri-Food Canada, Sherbrooke Research and Development Centre, Sherbrooke, QC, Canada
| |
Collapse
|
32
|
Cruz-Carrillo G, Camacho-Morales A. Metabolic Flexibility Assists Reprograming of Central and Peripheral Innate Immunity During Neurodevelopment. Mol Neurobiol 2021; 58:703-718. [PMID: 33006752 DOI: 10.1007/s12035-020-02154-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/28/2020] [Indexed: 01/03/2023]
Abstract
Central innate immunity assists time-dependent neurodevelopment by recruiting and interacting with peripheral immune cells. Microglia are the major player of central innate immunity integrating peripheral signals arising from the circumventricular regions lacking the blood-brain barrier (BBB), via neural afferent pathways such as the vagal nerve and also by choroid plexus into the brain ventricles. Defective and/or unrestrained activation of central and peripheral immunity during embryonic development might set an aberrant connectome establishment and brain function, leading to major psychiatric disorders in postnatal stages. Molecular candidates leading to central and peripheral innate immune overactivation identified metabolic substrates and lipid species as major contributors of immunological priming, supporting the role of a metabolic flexibility node during trained immunity. Mechanistically, trained immunity is established by an epigenetic program including DNA methylation and histone acetylation, as the major molecular epigenetic signatures to set immune phenotypes. By definition, immunological training sets reprogramming of innate immune cells, enhancing or repressing immune responses towards a second challenge which potentially might contribute to neurodevelopment disorders. Notably, the innate immune training might be set during pregnancy by maternal immune activation stimuli. In this review, we integrate the most valuable scientific evidence supporting the role of metabolic cues assisting metabolic flexibility, leading to innate immune training during development and its effects on aberrant neurological phenotypes in the offspring. We also add reports supporting the role of methylation and histone acetylation signatures as a major epigenetic mechanism regulating immune training.
Collapse
Affiliation(s)
- Gabriela Cruz-Carrillo
- Departamento de Bioquímica. Facultad de Medicina,, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico
- Neurometabolism Unit, Center for Research and Development in Health Sciences, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, NL, Mexico
| | - Alberto Camacho-Morales
- Departamento de Bioquímica. Facultad de Medicina,, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, Mexico.
- Neurometabolism Unit, Center for Research and Development in Health Sciences, Universidad Autónoma de Nuevo León, San Nicolás de los Garza, NL, Mexico.
| |
Collapse
|
33
|
Zhang W, Yang D, Yuan Y, Liu C, Chen H, Zhang Y, Wang Q, Petersen RB, Huang K, Zheng L. Muscular G9a Regulates Muscle-Liver-Fat Axis by Musclin Under Overnutrition in Female Mice. Diabetes 2020; 69:2642-2654. [PMID: 32994276 DOI: 10.2337/db20-0437] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 09/21/2020] [Indexed: 11/13/2022]
Abstract
Cross talk among different tissues and organs is a hotspot in metabolic research. Recent studies have revealed the regulatory roles of a number of myokines in metabolism. Here, we report that female mice lacking muscle-specific histone methylase G9a (Ehmt2 Ckmm knockout [KO] or Ehmt2 HSA KO) are resistant to high-fat diet (HFD)-induced obesity and hepatic steatosis. Furthermore, we identified a significantly upregulated circulating level of musclin, a myokine, in HFD-fed Ehmt2 Ckmm KO or Ehmt2 HSA KO female mice. Similarly, upregulated musclin was observed in mice injected with two structurally different inhibitors for G9a methylase activity: BIX01294 and A366. Moreover, injection of recombinant full-length musclin or its functional core domain inhibited the HFD-induced obesity and hepatic steatosis in wild-type female and male mice. Mechanistically, G9a methylase activity-dependently regulated muscular musclin level by binding to its promoter, also by regulating phosphorylated-FOXO1/FOXO1 levels in vivo and in vitro. Collectively, these data suggest a critical role for G9a in the muscle-liver-fat metabolic axis, at least for female mice. Musclin may serve as a potential therapeutic candidate for obesity and associated diseases.
Collapse
Affiliation(s)
- Wenquan Zhang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Dong Yang
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yangmian Yuan
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Chong Liu
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Hong Chen
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Zhang
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing Wang
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
| | - Robert B Petersen
- Foundational Sciences, Central Michigan University College of Medicine, Mount Pleasant, MI
| | - Kun Huang
- Tongji School of Pharmacy, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ling Zheng
- Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| |
Collapse
|
34
|
Defour M, Hooiveld GJEJ, van Weeghel M, Kersten S. Probing metabolic memory in the hepatic response to fasting. Physiol Genomics 2020; 52:602-617. [PMID: 33074794 DOI: 10.1152/physiolgenomics.00117.2020] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Tissues may respond differently to a particular stimulus if they have been previously exposed to that same stimulus. Here, we tested the hypothesis that a strong metabolic stimulus such as fasting may influence the hepatic response to a subsequent fast and thus elicit a memory effect. Overnight fasting in mice significantly increased plasma free fatty acids, glycerol, β-hydroxybutyrate, and liver triglycerides, and decreased plasma glucose, plasma triglycerides, and liver glycogen levels. In addition, fasting dramatically changed the liver transcriptome, upregulating genes involved in gluconeogenesis and in uptake, oxidation, storage, and mobilization of fatty acids, and downregulating genes involved in fatty acid synthesis, fatty acid elongation/desaturation, and cholesterol synthesis. Fasting also markedly impacted the liver metabolome, causing a decrease in the levels of numerous amino acids, glycolytic-intermediates, TCA cycle intermediates, and nucleotides. However, these fasting-induced changes were unaffected by two previous overnight fasts. Also, no significant effect was observed of prior fasting on glucose tolerance. Finally, analysis of the effect of fasting on the transcriptome in hepatocyte humanized mouse livers indicated modest similarity in gene regulation in mouse and human liver cells. In general, genes involved in metabolic pathways were upregulated or downregulated to a lesser extent in human liver cells than in mouse liver cells. In conclusion, we found that previous exposure to fasting in mice did not influence the hepatic response to a subsequent fast, arguing against the concept of metabolic memory in the liver. Our data provide a useful resource for the study of liver metabolism during fasting.
Collapse
Affiliation(s)
- Merel Defour
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Guido J E J Hooiveld
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| | - Michel van Weeghel
- Laboratory Genetic Metabolic Diseases, Amsterdam UMC, Amsterdam, The Netherlands
| | - Sander Kersten
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University, Wageningen, The Netherlands
| |
Collapse
|
35
|
The influence of TERC, TERT and ACYP2 genes polymorphisms on plasma telomerase concentration, telomeres length and T2DM. Gene 2020; 766:145127. [PMID: 32937184 DOI: 10.1016/j.gene.2020.145127] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 07/20/2020] [Accepted: 09/01/2020] [Indexed: 11/21/2022]
Abstract
Telomeres are duplex tandem repeats of DNA sequence 5'-TTAGGG-3' at chromosomal ends synthesized by telomerase enzyme (TE). Telomeres length (TL) shortening is associated with age and age-related disorders. Recently, we demonstrated marked leukocytes TL (LTL) shortening in T2DM. To set the relationship between the TE, LTL and T2DM, we analyzed samples from 212 Kuwaiti subjects, 112 patients withT2DM and 100 non-diabetic subjects. The plasma TE and fasting insulin were measured by ELISA, the LTL was estimated by qPCR and three SNPs of genes related to TL; TERC rs12696304 (C/G), TERT rs2736100 (C/A) and ACYP2 rs6713088 (C/G) were genotyped by rtPCR. Results revealed comparable TE levels and alleles/genotypes between the cases and controls with no influence of either on the LTL. Interestingly, although the plasma concentration of the TE was generally low, it was significantly influenced by the TERT and ACYP2 but not TERC polymorphisms. The CC genotype carriers of rs2736100 (C/A) had significantly higher plasma TE levels compared to CA and AA carriers, p 0.009 and p 0.047, respectively, and the A-allele was associated with low TE, p 0.018. Similarly, significantly higher TE levels were detected in CC carriers of ACYP2 rs6713088 (C/G) compared with GC carriers, p 0.002, and the G-allele was associated with low TE, p 0.009. Finally, the TERT and ACYP2 polymorphisms had an influence on blood glucose levels. In conclusion, the telomeres shortening in T2DM was not due to TE deficiency or gene polymorphisms, while the TE levels were significantly associated with the TERT and ACYP2 but not TERC polymorphisms.
Collapse
|
36
|
Terrin G, Coscia A, Boscarino G, Faccioli F, Di Chiara M, Greco C, Onestà E, Oliva S, Aloi M, Dito L, Cardilli V, Regoli D, De Curtis M. Long-term effects on growth of an energy-enhanced parenteral nutrition in preterm newborn: A quasi-experimental study. PLoS One 2020; 15:e0235540. [PMID: 32628715 PMCID: PMC7337335 DOI: 10.1371/journal.pone.0235540] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 06/18/2020] [Indexed: 12/13/2022] Open
Abstract
Aim To assess the best energy intake in Parenteral Nutrition (PN) for preterm newborns, considering both possible benefits for growth and risk of complications. Methods Quasi-experimental study comparing two cohorts of newborns, receiving Energy-Enhanced vs. Standard PN (Cohort A, from 1st January 2015 to 31 January 2016 and Cohort B from 1st February 2016 to 31 March 2017; respectively) after implementation of a change in the PN protocol. The primary outcome measure was growth at 24 months of life. The PN associated complications were also measured. Results We enrolled 132 newborns in two Cohorts, similar for prenatal and postnatal clinical characteristics. Although, body weight and length at 24 months of life were significantly higher (p<0.05) in the Cohort A (11.1, 95% CI 10.6 to 11.6 Kg; 85.0 95% CI 83.8 to 86.2 cm) compared with Cohort B (10.4, 95% CI 9.9 to 10.9 Kg; 81.3 95% CI 79.7 to 82.8 cm), body weight and length Z-Score in the first 24 months of life were similar between the two Cohorts. The rate of PN associated complications was very high in both study Cohorts (up to 98% of enrolments). Multivariate analysis showed that length at 24 months was significantly associated with receiving standard PN (cohort A) in the first week of life and on the energy intake in the first week of life. We also found a marginally insignificant association between Cohort A assignment and body weight at 24 months of life (p = 0.060). Conclusions Energy-enhanced PN in early life has not significant effects on long-term growth in preterm newborns. The high prevalence of PN associated complications, poses concerns about the utility of high energy intake recommended by current guidelines for PN.
Collapse
Affiliation(s)
- Gianluca Terrin
- Department of Maternal and Child Health Policlinico Umberto I, University La Sapienza, Rome, Italy
- * E-mail:
| | - Alessandra Coscia
- Neonatology Unit, Department of Public Health and Pediatrics, University of Turin, Turin, Italy
| | - Giovanni Boscarino
- Department of Maternal and Child Health Policlinico Umberto I, University La Sapienza, Rome, Italy
| | - Francesca Faccioli
- Department of Maternal and Child Health Policlinico Umberto I, University La Sapienza, Rome, Italy
| | - Maria Di Chiara
- Department of Maternal and Child Health Policlinico Umberto I, University La Sapienza, Rome, Italy
| | - Carla Greco
- Department of Maternal and Child Health Policlinico Umberto I, University La Sapienza, Rome, Italy
| | - Elisa Onestà
- Department of Maternal and Child Health Policlinico Umberto I, University La Sapienza, Rome, Italy
| | - Salvatore Oliva
- Department of Maternal and Child Health Policlinico Umberto I, University La Sapienza, Rome, Italy
| | - Marina Aloi
- Department of Maternal and Child Health Policlinico Umberto I, University La Sapienza, Rome, Italy
| | - Lucia Dito
- Department of Maternal and Child Health Policlinico Umberto I, University La Sapienza, Rome, Italy
| | - Viviana Cardilli
- Department of Maternal and Child Health Policlinico Umberto I, University La Sapienza, Rome, Italy
| | - Daniela Regoli
- Department of Maternal and Child Health Policlinico Umberto I, University La Sapienza, Rome, Italy
| | - Mario De Curtis
- Department of Maternal and Child Health Policlinico Umberto I, University La Sapienza, Rome, Italy
| |
Collapse
|
37
|
Shaito A, Hasan H, Habashy KJ, Fakih W, Abdelhady S, Ahmad F, Zibara K, Eid AH, El-Yazbi AF, Kobeissy FH. Western diet aggravates neuronal insult in post-traumatic brain injury: Proposed pathways for interplay. EBioMedicine 2020; 57:102829. [PMID: 32574954 PMCID: PMC7317220 DOI: 10.1016/j.ebiom.2020.102829] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/26/2020] [Accepted: 05/27/2020] [Indexed: 12/17/2022] Open
Abstract
Traumatic brain injury (TBI) is a global health burden and a major cause of disability and mortality. An early cascade of physical and structural damaging events starts immediately post-TBI. This primary injury event initiates a series of neuropathological molecular and biochemical secondary injury sequelae, that last much longer and involve disruption of cerebral metabolism, mitochondrial dysfunction, oxidative stress, neuroinflammation, and can lead to neuronal damage and death. Coupled to these events, recent studies have shown that lifestyle factors, including diet, constitute additional risk affecting TBI consequences and neuropathophysiological outcomes. There exists molecular cross-talk among the pathways involved in neuronal survival, neuroinflammation, and behavioral outcomes, that are shared among western diet (WD) intake and TBI pathophysiology. As such, poor dietary intake would be expected to exacerbate the secondary damage in TBI. Hence, the aim of this review is to discuss the pathophysiological consequences of WD that can lead to the exacerbation of TBI outcomes. We dissect the role of mitochondrial dysfunction, oxidative stress, neuroinflammation, and neuronal injury in this context. We show that currently available data conclude that intake of a diet saturated in fats, pre- or post-TBI, aggravates TBI, precludes recovery from brain trauma, and reduces the response to treatment.
Collapse
Affiliation(s)
- Abdullah Shaito
- Department of Biological and Chemical Sciences, Lebanese International University, Beirut, Lebanon and Faculty of Health Sciences, University of Balamand, Beirut, Lebanon
| | - Hiba Hasan
- Institute of Anatomy and Cell Biology, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | | | - Walaa Fakih
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Samar Abdelhady
- Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Fatima Ahmad
- Neuroscience Research Center, Faculty of Medicine, Lebanese University
| | - Kazem Zibara
- Biology Department, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Ali H Eid
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Department of Biomedical Sciences, College of Health Sciences, Doha, Qatar
| | - Ahmed F El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Egypt.
| | - Firas H Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon.
| |
Collapse
|
38
|
Rivera P, Tovar R, Ramírez-López MT, Navarro JA, Vargas A, Suárez J, de Fonseca FR. Sex-Specific Anxiety and Prefrontal Cortex Glutamatergic Dysregulation Are Long-Term Consequences of Pre-and Postnatal Exposure to Hypercaloric Diet in a Rat Model. Nutrients 2020; 12:nu12061829. [PMID: 32575416 PMCID: PMC7353464 DOI: 10.3390/nu12061829] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/14/2020] [Accepted: 06/16/2020] [Indexed: 12/16/2022] Open
Abstract
Both maternal and early life malnutrition can cause long-term behavioral changes in the offspring, which depends on the caloric availability and the timing of the exposure. Here we investigated in a rat model whether a high-caloric palatable diet given to the mother and/or to the offspring during the perinatal and/or postnatal period might dysregulate emotional behavior and prefrontal cortex function in the offspring at adult age. To this end, we examined both anxiety responses and the mRNA/protein expression of glutamatergic, GABAergic and endocannabinoid signaling pathways in the prefrontal cortex of adult offspring. Male animals born from mothers fed the palatable diet, and who continued with this diet after weaning, exhibited anxiety associated with an overexpression of the mRNA of Grin1, Gria1 and Grm5 glutamate receptors in the prefrontal cortex. In addition, these animals had a reduced expression of the endocannabinoid system, the main inhibitory retrograde input to glutamate synapses, reflected in a decrease of the Cnr1 receptor and the Nape-pld enzyme. In conclusion, a hypercaloric maternal diet induces sex-dependent anxiety, associated with alterations in both glutamatergic and cannabinoid signaling in the prefrontal cortex, which are accentuated with the continuation of the palatable diet during the life of the offspring.
Collapse
Affiliation(s)
- Patricia Rivera
- Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, 29010 Málaga, Spain; (R.T.); (J.A.N.); (A.V.); (J.S.)
- Correspondence: (P.R.); (F.R.d.F.); Tel.: +34-952-614-012 (P.R. & F.R.d.F.)
| | - Rubén Tovar
- Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, 29010 Málaga, Spain; (R.T.); (J.A.N.); (A.V.); (J.S.)
| | - María Teresa Ramírez-López
- Hospital Universitario de Getafe, Servicio de Ginecología y Obstetricia, 28905 Getafe, Spain;
- Departamento de Enfermería, Facultad de Enfermería, Fisioterapia y Podología, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Juan Antonio Navarro
- Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, 29010 Málaga, Spain; (R.T.); (J.A.N.); (A.V.); (J.S.)
| | - Antonio Vargas
- Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, 29010 Málaga, Spain; (R.T.); (J.A.N.); (A.V.); (J.S.)
| | - Juan Suárez
- Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, 29010 Málaga, Spain; (R.T.); (J.A.N.); (A.V.); (J.S.)
| | - Fernando Rodríguez de Fonseca
- Instituto de Investigación Biomédica de Málaga (IBIMA), Hospital Regional Universitario de Málaga, Universidad de Málaga, 29010 Málaga, Spain; (R.T.); (J.A.N.); (A.V.); (J.S.)
- Correspondence: (P.R.); (F.R.d.F.); Tel.: +34-952-614-012 (P.R. & F.R.d.F.)
| |
Collapse
|
39
|
Adeodato CSR, Alves GG, Botelho AMN, Caldas IP, Gonçalves FP, Pinto LFR, Lima SCS, Fagundes MCN, Masterson D, Scelza P, Scelza MFZ. Association of DNA sequence-independent genetic regulatory mechanisms with apical periodontitis: A scoping review. Arch Oral Biol 2020; 115:104737. [PMID: 32387743 DOI: 10.1016/j.archoralbio.2020.104737] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 04/20/2020] [Accepted: 04/21/2020] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Different studies in the last decade have proposed that gene expression alterations that are independent of the DNA sequence may also play an important role in periapical disease. The present study aimed to assess the available evidence supporting a relationship between these alterations and apical periodontitis through a scoping review. DESIGN Specific strategies were developed for different databases (MEDLINE via PubMed, Cochrane Library, Scopus, Web of Science, and Virtual Health Library) and a search performed by March 1st, 2019. The evidence sources were selected according to the eligibility criteria and underwent a critical appraisal of methodological quality. RESULTS The initial search retrieved 212 references, with eight eligible articles after the removal of replicates and application of exclusion criteria. Five studies identified altered DNA methylation on inflammatory response genes (FOXP3, CXCL3, FADD, MMP2, MMP9, IFNG, IL4, IL12) on AP patients. Three others identified the alterations on the expression of several microRNAs (miR-29b, 106b, 125b, 143, 146a, 155, 198) during AP. No evidence was identified regarding mechanisms of histone methylation, or of epigenetic heritability or stability. CONCLUSIONS There is available evidence for the involvement of different genetic regulatory mechanisms independent of changes in DNA sequence in the development or severity of apical periodontitis. However, due to methodological limitations, further research must be performed before novel therapies and diagnostic tools for AP may arise from these data.
Collapse
Affiliation(s)
- Caroline Sousa Ribeiro Adeodato
- Post-graduation Program in Dentistry of Fluminense Federal University (UFF), Mario Santos Braga Street, no 28, 24020-140, Niteroi, RJ, Brazil
| | - Gutemberg Gomes Alves
- Clinical Research Unit of the Antonio Pedro Hospital, Fluminense Federal University, Niteroi, RJ, Brazil
| | - Ana Maria Nunes Botelho
- Laboratory of Experimental Culture Cell (LECCel), Faculty of Dentistry, Fluminense Federal University (UFF) Mario Santos Braga Street, no 28, 24020-140, Niteroi, RJ, Brazil
| | - Isleine Portal Caldas
- Post-graduation Program in Dentistry of Fluminense Federal University (UFF), Mario Santos Braga Street, no 28, 24020-140, Niteroi, RJ, Brazil
| | - Fabiano Palmeira Gonçalves
- Post-graduation Program in Dentistry of Fluminense Federal University (UFF), Mario Santos Braga Street, no 28, 24020-140, Niteroi, RJ, Brazil
| | - Luis Felipe Ribeiro Pinto
- Molecular Carcinogenesis Program of National Cancer Institute (INCA), Andre Cavalcanti Street, no 37, 20231-050, Rio de Janeiro, Brazil
| | - Sheila Coelho Soares Lima
- Molecular Carcinogenesis Program of National Cancer Institute (INCA), Andre Cavalcanti Street, no 37, 20231-050, Rio de Janeiro, Brazil
| | - Marina Chianello Nicolau Fagundes
- Molecular Carcinogenesis Program of National Cancer Institute (INCA), Andre Cavalcanti Street, no 37, 20231-050, Rio de Janeiro, Brazil
| | - Daniele Masterson
- Central Library of the Health Science Center University Federal of Rio de Janeiro (UFRJ), Carlos Chagas Filho Avenue, no 373, 21940-902, Rio de Janeiro, Brazil
| | - Pantaleo Scelza
- Geriatric Dentistry Department, Faculty of Dentistry of Fluminense Federal University (UFF), Mario Santos Braga Street, no 28, 24020-140, Niteroi, RJ, Brazil
| | - Miriam Fátima Zaccaro Scelza
- Laboratory of Experimental Culture Cell (LECCel), Faculty of Dentistry, Fluminense Federal University (UFF) Mario Santos Braga Street, no 28, 24020-140, Niteroi, RJ, Brazil; Geriatric Dentistry Department, Faculty of Dentistry of Fluminense Federal University (UFF), Mario Santos Braga Street, no 28, 24020-140, Niteroi, RJ, Brazil; Endodontics Department, Faculty of Dentistry, Fluminense Federal University (UFF) Mario Santos Braga Street, no 28, 24020-140, Niteroi, RJ, Brazil.
| |
Collapse
|
40
|
Chirinos JA. Discerning the Age-Related Heterogeneity in Heart Failure With Preserved Ejection Fraction. J Am Coll Cardiol 2020; 74:613-616. [PMID: 31370951 DOI: 10.1016/j.jacc.2019.06.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 06/03/2019] [Indexed: 11/28/2022]
Affiliation(s)
- Julio A Chirinos
- Division of Cardiovascular Medicine, Hospital of the University of Pennsylvania and University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania.
| |
Collapse
|
41
|
Kato M, Natarajan R. Epigenetics and epigenomics in diabetic kidney disease and metabolic memory. Nat Rev Nephrol 2020; 15:327-345. [PMID: 30894700 DOI: 10.1038/s41581-019-0135-6] [Citation(s) in RCA: 334] [Impact Index Per Article: 66.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The development and progression of diabetic kidney disease (DKD), a highly prevalent complication of diabetes mellitus, are influenced by both genetic and environmental factors. DKD is an important contributor to the morbidity of patients with diabetes mellitus, indicating a clear need for an improved understanding of disease aetiology to inform the development of more efficacious treatments. DKD is characterized by an accumulation of extracellular matrix, hypertrophy and fibrosis in kidney glomerular and tubular cells. Increasing evidence shows that genes associated with these features of DKD are regulated not only by classical signalling pathways but also by epigenetic mechanisms involving chromatin histone modifications, DNA methylation and non-coding RNAs. These mechanisms can respond to changes in the environment and, importantly, might mediate the persistent long-term expression of DKD-related genes and phenotypes induced by prior glycaemic exposure despite subsequent glycaemic control, a phenomenon called metabolic memory. Detection of epigenetic events during the early stages of DKD could be valuable for timely diagnosis and prompt treatment to prevent progression to end-stage renal disease. Identification of epigenetic signatures of DKD via epigenome-wide association studies might also inform precision medicine approaches. Here, we highlight the emerging role of epigenetics and epigenomics in DKD and the translational potential of candidate epigenetic factors and non-coding RNAs as biomarkers and drug targets for DKD.
Collapse
Affiliation(s)
- Mitsuo Kato
- Department of Diabetes Complications and Metabolism, Diabetes Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA, USA.
| | - Rama Natarajan
- Department of Diabetes Complications and Metabolism, Diabetes Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, CA, USA.
| |
Collapse
|
42
|
DNA Methylation Changes are Associated with the Programming of White Adipose Tissue Browning Features by Resveratrol and Nicotinamide Riboside Neonatal Supplementations in Mice. Nutrients 2020; 12:nu12020461. [PMID: 32059412 PMCID: PMC7071331 DOI: 10.3390/nu12020461] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 02/06/2020] [Accepted: 02/08/2020] [Indexed: 12/22/2022] Open
Abstract
Neonatal supplementation with resveratrol (RSV) or nicotinamide riboside (NR) programs in male mice brown adipocyte-like features in white adipose tissue (WAT browning) together with improved metabolism in adulthood. We tested the involvement in this programming of long-term epigenetic changes in two browning-related genes that are overexpressed in WAT of supplemented mice, Slc27a1 and Prdm16. Suckling mice received orally the vehicle, RSV or NR from postnatal days 2-to-20. After weaning (d21) onto a chow diet, male mice were habituated to a normal-fat diet (NFD) starting d75, and split on d90 into continuation on the NFD or switching to a high-fat diet (HFD) until euthanization on d164. CpG methylation by bisulfite-sequencing was analyzed on inguinal WAT. Both treatments modified methylation marks in Slc27a1 and Prdm16 and the HFD-dependent dynamics of these marks in the adult WAT, with distinct and common effects. The treatments also affected gene expression of de novo DNA methylases in WAT of young animals (euthanized at d35 in independent experiments). Studies in 3T3-L1 adipocytes indicated the direct effects of RSV and NR on the DNA methylation machinery and favoring browning features. The results support epigenetic effects being involved in WAT programming by neonatal RSV or NR supplementation in male mice.
Collapse
|
43
|
Cheshmeh S, Nachvak SM, Rezvani N, Saber A. Effects of Breastfeeding and Formula Feeding on the Expression Level of FTO, CPT1A and PPAR-α Genes in Healthy Infants. Diabetes Metab Syndr Obes 2020; 13:2227-2237. [PMID: 32617012 PMCID: PMC7326192 DOI: 10.2147/dmso.s252122] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 06/12/2020] [Indexed: 12/16/2022] Open
Abstract
PURPOSE The study aimed to investigate the effect of breastfeeding, formula feeding and mix feeding (breastfed plus formula-fed) on the expression level of obesity-predisposing genes including fat mass and obesity-associated (FTO), carnitine palmitoyltransferase 1A (CPT1A), and peroxisome proliferator-activated receptor-α (PPAR-α) in 5- to 6-month-old infants. PATIENTS AND METHODS A total of 150 infants participated in this case-control study. All subjects were healthy infants aged 5-6 months that divided into 3 groups: breastfed, formula-fed, and mix-fed. The expression level of FTO, CPT1A, and PPAR-α genes in peripheral blood mononuclear cells (PBMC) was evaluated in each group using reverse transcription-polymerase chain reaction (RT-PCR) method. RESULTS Our findings showed that the current weight, height, and head circumference of infants in the formula feeding and mix feeding groups were significantly higher than those in the exclusive breastfeeding group. The expression level of FTO and CPT1A genes in formula-fed and mix-fed infants was significantly higher (p<0.001) than that in breastfed infants, while the expression level of PPAR-α gene was significantly lower (p<0.05). CONCLUSION Breastfeeding showed modulatory effects on the expression level of obesity-predisposing genes and can protect against obesity and subsequent non-communicable diseases. However, more investigations are required to explain the epigenetic effects of breast milk.
Collapse
Affiliation(s)
- Sahar Cheshmeh
- Department of Nutritional Sciences, School of Nutritional Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Student Research Committee, School of Nutritional Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyed Mostafa Nachvak
- Department of Nutritional Sciences, School of Nutritional Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Correspondence: Seyed Mostafa Nachvak Department of Nutritional Sciences, Faculty of Nutritional Sciences and Food Technologies, Kermanshah University of Medical Sciences, Isar Sq., Across from Farabi Hospital, P.O. Box 6719851351, Kermanshah, IranTel +98-8338395885Fax +98-83 37102002 Email
| | - Nayebali Rezvani
- Department of Clinical Biochemistry, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Nayebali Rezvani Department of Clinical Biochemistry, Kermanshah University of Medical Sciences, P.O. Box 67148-69914, Kermanshah, IranTel +98-9183391265 Email
| | - Amir Saber
- Department of Nutritional Sciences, School of Nutritional Sciences and Food Technology, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
44
|
Chavira-Suárez E, Ramírez-Mendieta AJ, Martínez-Gutiérrez S, Zárate-Segura P, Beltrán-Montoya J, Espinosa-Maldonado NC, de la Cerda-Ángeles JC, Vadillo-Ortega F. Influence of pre-pregnancy body mass index (p-BMI) and gestational weight gain (GWG) on DNA methylation and protein expression of obesogenic genes in umbilical vein. PLoS One 2019; 14:e0226010. [PMID: 31794592 PMCID: PMC6890247 DOI: 10.1371/journal.pone.0226010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 11/17/2019] [Indexed: 01/14/2023] Open
Abstract
Understanding the regulatory mechanisms that affect obesogenic genes expression in newborns is essential for early prevention efforts, but they remain unclear. Our study aimed to explore whether the maternal p-BMI and GWG were associated with regulatory single-locus DNA methylation in selected obesogenic genes. For this purpose, DNA methylation was assayed by Methylation-Sensitive High Resolution Melting (MS-HRM) technique and Sanger allele-bisulfite sequencing in fifty samples of umbilical vein to evaluate glucosamine-6-phosphate deaminase 2 (GNPDA2), peroxisome proliferator-activated receptor gamma coactivator 1 alpha (PGC1α), and leptin receptor (LEPR) genes. Correlations between DNA methylation levels and indicators of maternal nutritional status were carried out. Western blotting was used to evaluate protein expression in extracts of the same samples. Results indicated that GNPDA2 and PGC1α genes have the same level of DNA methylation in all samples; however, a differential DNA methylation of LEPR gene promoter was found, correlating it with GWG and this correlation is unaffected by maternal age or unhealthy habits. Furthermore, leptin receptor (Lep-Rb) was upregulated in samples that showed the lowest levels of DNA methylation. This study highlights the association between poor GWG and adjustments on obesogenic genes expression in newborn tissues with potential consequences for development of obesity in the future.
Collapse
Affiliation(s)
- Erika Chavira-Suárez
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Unidad de Vinculación Científica de la Facultad de Medicina, Universidad Nacional Autónoma de México en el Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Angélica Jazmín Ramírez-Mendieta
- Unidad de Vinculación Científica de la Facultad de Medicina, Universidad Nacional Autónoma de México en el Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Sofía Martínez-Gutiérrez
- Unidad de Vinculación Científica de la Facultad de Medicina, Universidad Nacional Autónoma de México en el Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Paola Zárate-Segura
- Laboratorio de Medicina Traslacional, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Jorge Beltrán-Montoya
- Instituto Nacional de Perinatología Isidro Espinosa de los Reyes, Mexico City, Mexico
| | | | | | - Felipe Vadillo-Ortega
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
45
|
Systemic endocrinopathies (thyroid conditions and diabetes): impact on postnatal life of the offspring. Fertil Steril 2019; 111:1076-1091. [PMID: 31155115 DOI: 10.1016/j.fertnstert.2019.04.039] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 04/25/2019] [Accepted: 04/26/2019] [Indexed: 12/22/2022]
Abstract
Fetal programming may influence childhood and adult life, determining the risk of specific diseases. During earlier stages of pregnancy, the transfer of maternal thyroid hormones to the fetus is vital for adequate neurologic development. The presence of severe maternal thyroid dysfunction, particularly severe iodine deficiency, is devastating, leading to irreversible neurologic sequelae. Moreover, mild maternal thyroid conditions, such as a mild-to-moderate iodine deficiency, may also lead to milder neurologic and behavioral conditions later during the life of the offspring. Maternal dysglycemia due to pregestational or gestational diabetes mellitus is another common situation in which fetal development encounters a hostile environment. Hyperglycemia in utero may trigger metabolic conditions in the offspring, including abnormalities of glucose tolerance and weight excess. Physicians assisting pregnant women have to be aware about these conditions, because they may go unnoticed if not properly screened. Because an early diagnosis and appropriate management may prevent most of the possible negative consequences for the progeny, the prevention, early diagnosis, and proper management of these endocrine conditions should be offered to all women undergoing pregnancy. Here, we comprehensively review the current evidence about the effects of maternal thyroid dysfunction and maternal dysglycemia on the cognitive function and carbohydrate metabolism in the offspring, two prevalent conditions of utmost importance for the child's health and development.
Collapse
|
46
|
Georgia-Eirini D, Athina S, Wim VB, Christos K, Theodoros C. Natural Products from Mediterranean Diet: From Anti-hyperlipidemic Agents to Dietary Epigenetic Modulators. Curr Pharm Biotechnol 2019; 20:825-844. [DOI: 10.2174/1573407215666190628150921] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 09/23/2018] [Accepted: 06/03/2019] [Indexed: 01/05/2023]
Abstract
Background:
Cardiovascular Diseases (CVD) are, currently, the major contributor to global
mortality and will continue to dominate mortality rates in the future. Hyperlipidemia refers to the elevated
levels of lipids and cholesterol in the blood, and is also identified as dyslipidemia, manifesting in
the form of different disorders of lipoprotein metabolism. These abnormalities may lead to the development
of atherosclerosis, which can lead to coronary artery disease and stroke. In recent years, there
is a growing interest in the quest for alternative therapeutic treatments based on natural products, offering
better recovery and the avoidance of side effects. Recent technological advances have further improved
our understanding of the role of epigenetic mechanisms in hyperlipidemic disorders and dietary
prevention strategies.
Objective:
This is a comprehensive overview of the anti-hyperlipidemic effects of plant extracts, vegetables,
fruits and isolated compounds thereof, with a focus on natural products from the Mediterranean
region as well as the possible epigenetic changes in gene expression or cardiometabolic signaling
pathways.
Methods:
For the purpose of this study, we searched the PubMed, Scopus and Google Scholar databases
for eligible articles and publications over the last five years. The keywords included: “hyperlipidemia”,
“plant extract”, “herbs”, “natural products”, “vegetables”, “cholesterol” and others. We initially
included all relevant articles referring to in vitro studies, animal studies, Randomized Controlled
Trials (RCTs) and previous reviews.
Conclusion:
Many natural products found in the Mediterranean diet have been studied for the treatment
of hyperlipidemia. The antihyperlipidemic effect seems to be dose and/or consumption frequency
related, which highlights the fact that a healthy diet can only be effective in reversing disease markers
if it is consistent and within the framework of a healthy lifestyle. Finally, epigenetic biomarkers are increasingly
recognized as new lifestyle management tools to monitor a healthy dietary lifestyle for the
prevention of hyperlipidaemic disorders and comorbidities to promote a healthy life.
Collapse
Affiliation(s)
- Deligiannidou Georgia-Eirini
- Laboratory of Hygiene and Environmental Protection, Department of Medicine, Democritus University of Thrace, Alexandroupolis, 68100, Greece
| | - Sygkouna Athina
- Laboratory of Hygiene and Environmental Protection, Department of Medicine, Democritus University of Thrace, Alexandroupolis, 68100, Greece
| | - Vanden Berghe Wim
- Lab of Protein Science, Proteomics & Epigenetic Signaling (PPES), Department of Biomedical sciences, University Antwerp, 2610, Wilrijk, Belgium
| | - Kontogiorgis Christos
- Laboratory of Hygiene and Environmental Protection, Department of Medicine, Democritus University of Thrace, Alexandroupolis, 68100, Greece
| | - Constantinides Theodoros
- Laboratory of Hygiene and Environmental Protection, Department of Medicine, Democritus University of Thrace, Alexandroupolis, 68100, Greece
| |
Collapse
|
47
|
Séité S, Masagounder K, Heraud C, Véron V, Marandel L, Panserat S, Seiliez I. Early feeding of rainbow trout ( Oncorhynchus mykiss) with methionine-deficient diet over a 2 week period: consequences for liver mitochondria in juveniles. ACTA ACUST UNITED AC 2019; 222:jeb.203687. [PMID: 31488624 DOI: 10.1242/jeb.203687] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 08/28/2019] [Indexed: 12/13/2022]
Abstract
Methionine is a key factor in modulating the cellular availability of the main biological methyl donor S-adenosylmethionine (SAM), which is required for all biological methylation reactions including DNA and histone methylation. As such, it represents a potential critical factor in nutritional programming. Here, we investigated whether early methionine restriction at first feeding could have long-term programmed metabolic consequences in rainbow trout. For this purpose, trout fry were fed with either a control diet (C) or a methionine-deficient diet (MD) for 2 weeks from the first exogenous feeding. Next, fish were subjected to a 5 month growth trial with a standard diet followed by a 2 week challenge (with the MD or C diet) to test the programming effect of the early methionine restriction. The results showed that, whatever the dietary treatment of fry, the 2 week challenge with the MD diet led to a general mitochondrial defect associated with an increase in endoplasmic reticulum stress, mitophagy and apoptosis, highlighting the existence of complex cross-talk between these different functions. Moreover, for the first time, we also observed that fish fed the MD diet at the first meal later exhibited an increase in several critical factors of mitophagy, hinting that the early nutritional stimulus with methionine deficiency resulted in long-term programming of this cell function. Together, these data extend our understanding of the role of dietary methionine and emphasize the potential for this amino acid in the application of new feeding strategies, such as nutritional programming, to optimize the nutrition and health of farmed fish.
Collapse
Affiliation(s)
- Sarah Séité
- INRA, Université de Pau et des Pays de l'Adour, E2S UPPA, UMR 1419, Nutrition, Métabolisme, Aquaculture, Saint Pée sur Nivelle, F-64310, France.,Evonik Rexim, 80400 Ham, France.,Evonik Nutrition and Care GmbH, 63457 Hanau, Germany
| | | | - Cécile Heraud
- INRA, Université de Pau et des Pays de l'Adour, E2S UPPA, UMR 1419, Nutrition, Métabolisme, Aquaculture, Saint Pée sur Nivelle, F-64310, France
| | - Vincent Véron
- INRA, Université de Pau et des Pays de l'Adour, E2S UPPA, UMR 1419, Nutrition, Métabolisme, Aquaculture, Saint Pée sur Nivelle, F-64310, France
| | - Lucie Marandel
- INRA, Université de Pau et des Pays de l'Adour, E2S UPPA, UMR 1419, Nutrition, Métabolisme, Aquaculture, Saint Pée sur Nivelle, F-64310, France
| | - Stéphane Panserat
- INRA, Université de Pau et des Pays de l'Adour, E2S UPPA, UMR 1419, Nutrition, Métabolisme, Aquaculture, Saint Pée sur Nivelle, F-64310, France
| | - Iban Seiliez
- INRA, Université de Pau et des Pays de l'Adour, E2S UPPA, UMR 1419, Nutrition, Métabolisme, Aquaculture, Saint Pée sur Nivelle, F-64310, France
| |
Collapse
|
48
|
Greco EA, Lenzi A, Migliaccio S, Gessani S. Epigenetic Modifications Induced by Nutrients in Early Life Phases: Gender Differences in Metabolic Alteration in Adulthood. Front Genet 2019; 10:795. [PMID: 31572434 PMCID: PMC6749846 DOI: 10.3389/fgene.2019.00795] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 07/29/2019] [Indexed: 12/19/2022] Open
Abstract
Metabolic chronic diseases, also named noncommunicable diseases (NCDs), are considered multifactorial pathologies, which are dramatically increased during the last decades. Noncommunicable diseases such as cardiovascular diseases, obesity, diabetes mellitus, cancers, and chronic respiratory diseases markedly increase morbidity, mortality, and socioeconomic costs. Moreover, NCDs induce several and complex clinical manifestations that lead to a gradual deterioration of health status and quality of life of affected individuals. Multiple factors are involved in the development and progression of these diseases such as sedentary behavior, smoking, pollution, and unhealthy diet. Indeed, nutrition has a pivotal role in maintaining health, and dietary imbalances represent major determinants favoring chronic diseases through metabolic homeostasis alterations. In particular, it appears that specific nutrients and adequate nutrition are important in all periods of life, but they are essential during specific times in early life such as prenatal and postnatal phases. Indeed, epidemiologic and experimental studies report the deleterious effects of an incorrect nutrition on health status several decades later in life. During the last decade, a growing interest on the possible role of epigenetic mechanisms as link between nutritional imbalances and NCDs development has been observed. Finally, because of the pivotal role of the hormones in fat, carbohydrate, and protein metabolism regulation throughout life, it is expected that any hormonal modification of these processes can imbalance metabolism and fat storage. Therefore, a particular interest to several chemicals able to act as endocrine disruptors has been recently developed. In this review, we will provide an overview and discuss the epigenetic role of some specific nutrients and chemicals in the modulation of physiological and pathological mechanisms.
Collapse
Affiliation(s)
- Emanuela A Greco
- Section of Medical Pathophysiology, Endocrinology and Food Sciences, Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Andrea Lenzi
- Section of Medical Pathophysiology, Endocrinology and Food Sciences, Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | - Silvia Migliaccio
- Department of Movement, Human and Health Sciences, Foro Italico University of Rome, Rome, Italy
| | - Sandra Gessani
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
49
|
Balistreri CR, Garagnani P, Madonna R, Vaiserman A, Melino G. Developmental programming of adult haematopoiesis system. Ageing Res Rev 2019; 54:100918. [PMID: 31226498 DOI: 10.1016/j.arr.2019.100918] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 05/15/2019] [Accepted: 06/17/2019] [Indexed: 12/15/2022]
Abstract
The Barker hypothesis of 'foetal origin of adult diseases' has led to emphasize the concept of 'developmental programming', based on the crucial role of epigenetic factors. Accordingly, it has been demonstrated that parental adversity (before conception and during pregnancy) and foetal factors (i.e., hypoxia, malnutrition and placental insufficiency) permanently modify the physiological systems of the progeny, predisposing them to premature ageing and chronic disease during adulthood. Thus, an altered functionality of the endocrine, immune, nervous and cardiovascular systems is observed in the progeny. However, it remains to be understood whether the haematopoietic system itself also represents a portrait of foetal programming. Here, we provide evidence, reporting and discussing related theories, and results of studies described in the literature. In addition, we have outlined our opinions and suggest how it is possible to intervene to correct foetal mal-programming. Some pro-health interventions and recommendations are proposed, with the hope of guarantee the health of future generations and trying to combat the continuous increase in age-related diseases in human populations.
Collapse
|
50
|
Badran M, Yassin BA, Lin DTS, Kobor MS, Ayas N, Laher I. Gestational intermittent hypoxia induces endothelial dysfunction, reduces perivascular adiponectin and causes epigenetic changes in adult male offspring. J Physiol 2019; 597:5349-5364. [PMID: 31441069 DOI: 10.1113/jp277936] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 07/26/2019] [Indexed: 12/11/2022] Open
Abstract
KEY POINTS Obstructive sleep apnoea (OSA) is characterized by intermittent hypoxia, which causes oxidative stress and inflammation and increases the risk of cardiovascular disease. OSA during pregnancy causes adverse maternal and fetal outcomes. The effects of pre-existing OSA in pregnant women on cardiometabolic outcomes in the offspring are unknown. We evaluated basic metabolic parameters, as well as aortic vascular and perivascular adipose tissue (PVAT) function in response to adiponectin, and examined DNA methylation of adiponectin gene promoter in PVAT in 16-week-old adult offspring exposed to gestational intermittent hypoxia (GIH). GIH decreased body weights at week 1 in both male and female offspring, and caused subsequent increases in body weight and food consumption in male offspring only. Adult female offspring had normal levels of lipids, glucose and insulin, with no endothelial dysfunction. Adult male offspring exhibited dyslipidaemia, insulin resistance and hyperleptinaemia. Decreased endothelial-dependent vasodilatation, loss of anti-contractile activity of PVAT and low circulating PVAT adiponectin levels, as well as increased pro-inflammatory gene expression and DNA methylation of adiponectin gene promoter, occurred in adult male offspring. Our results suggest that male offspring of women with OSA could be at risk of developing cardiometabolic disease during adulthood. ABSTRACT Perturbations during pregnancy can program the offspring to develop cardiometabolic diseases later in life. Obstructive sleep apnoea (OSA) is a chronic condition that frequently affects pregnancies and leads to adverse fetal outcomes. We assessed the offspring of female mice experiencing gestational intermittent hypoxia (GIH), a hallmark of OSA, for changes in metabolic profiles, aortic nitric oxide (NO)-dependent relaxations, perivascular adipose tissue (PVAT) anti-contractile activities and the responses to adiponectin, and DNA methylation of the adiponectin gene promoter in PVAT tissue. Pregnant mouse dams were exposed to intermittent hypoxic cycles ( F I O 2 21-12%) for 18 days. GIH resulted in lower body weights of pups at week 1, followed by significant weight gain by week 16 of age in male but not female offspring. Plasma lipids, leptin and insulin resistance were higher in GIH male adult offspring. Endothelium-dependent relaxation in response to ACh and the anti-contractile activity of PVAT in the abdominal aorta was reduced in GIH adult male offspring. Incubation of arteries from GIH adult male offspring with adiponectin restored the anti-contractile activity of PVAT. Both circulating and PVAT tissue homogenate levels of adiponectin, as well as gene expression of adiponectin in PVAT, were lower in GIH male offspring, along with an increased gene expression of inflammatory cytokines. Pyrosequencing of adiponectin gene promoter in PVAT showed increased DNA methylation in GIH male offspring. Our results indicate that GIH leads to vascular disease in adult male offspring through PVAT dysfunction, which was associated with low adiponectin levels and epigenetic modifications on the adiponectin gene promoter.
Collapse
Affiliation(s)
- Mohammad Badran
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Bisher Abu Yassin
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - David Tse Shen Lin
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, and Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Michael S Kobor
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, and Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Najib Ayas
- Divisions of Critical Care and Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Sleep Disorders Program, UBC Hospital, Vancouver, BC, Canada.,Division of Critical Care Medicine, Providence Healthcare, Vancouver, BC, Canada
| | - Ismail Laher
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|