1
|
Jahan-Mihan A, Leftwich J, Berg K, Labyak C, Nodarse RR, Allen S, Griggs J. The Impact of Parental Preconception Nutrition, Body Weight, and Exercise Habits on Offspring Health Outcomes: A Narrative Review. Nutrients 2024; 16:4276. [PMID: 39770898 PMCID: PMC11678361 DOI: 10.3390/nu16244276] [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: 11/21/2024] [Revised: 12/06/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
An increasing number of studies highlight the critical role of both maternal and paternal nutrition and body weight before conception in shaping offspring health. Traditionally, research has focused on maternal factors, particularly in utero exposures, as key determinants of chronic disease development. However, emerging evidence underscores the significant influence of paternal preconception health on offspring metabolic outcomes. While maternal health remains vital, with preconception nutrition playing a pivotal role in fetal development, paternal obesity and poor nutrition are linked to increased risks of metabolic disorders, including type 2 diabetes and cardiovascular disease in children. This narrative review aims to synthesize recent findings on the effects of both maternal and paternal preconception health, emphasizing the need for integrated early interventions. The literature search utilized PubMed, UNF One Search, and Google Scholar, focusing on RCTs; cohort, retrospective, and animal studies; and systematic reviews, excluding non-English and non-peer-reviewed articles. The findings of this review indicate that paternal effects are mediated by epigenetic changes in sperm, such as DNA methylation and non-coding RNA, which influence gene expression in offspring. Nutrient imbalances during preconception in both parents can lead to low birth weight and increased metabolic disease risk, while deficiencies in folic acid, iron, iodine, and vitamin D are linked to developmental disorders. Additionally, maternal obesity elevates the risk of chronic diseases in children. Future research should prioritize human studies to explore the influence of parental nutrition, body weight, and lifestyle on offspring health, ensuring findings are applicable across diverse populations. By addressing both maternal and paternal factors, healthcare providers can better reduce the prevalence of metabolic syndrome and its associated risks in future generations.
Collapse
Affiliation(s)
- Alireza Jahan-Mihan
- Department of Nutrition and Dietetics, University of North Florida, 1 UNF Dr., Jacksonville, FL 32224, USA; (J.L.); (K.B.); (C.L.); (R.R.N.)
| | - Jamisha Leftwich
- Department of Nutrition and Dietetics, University of North Florida, 1 UNF Dr., Jacksonville, FL 32224, USA; (J.L.); (K.B.); (C.L.); (R.R.N.)
| | - Kristin Berg
- Department of Nutrition and Dietetics, University of North Florida, 1 UNF Dr., Jacksonville, FL 32224, USA; (J.L.); (K.B.); (C.L.); (R.R.N.)
| | - Corinne Labyak
- Department of Nutrition and Dietetics, University of North Florida, 1 UNF Dr., Jacksonville, FL 32224, USA; (J.L.); (K.B.); (C.L.); (R.R.N.)
| | - Reniel R. Nodarse
- Department of Nutrition and Dietetics, University of North Florida, 1 UNF Dr., Jacksonville, FL 32224, USA; (J.L.); (K.B.); (C.L.); (R.R.N.)
| | - Sarah Allen
- Greenleaf Behavioral Health, 2209 Pineview Dr., Valdosta, GA 31602, USA;
| | | |
Collapse
|
2
|
Nagagata BA, Brito G, Ornellas F, Mandarim-de-Lacerda CA, Aguila MB. Melatonin supplementation in obese mothers reduces hypothalamic inflammation and enhances thermogenesis in mice progeny. J Nutr Biochem 2024; 128:109625. [PMID: 38521130 DOI: 10.1016/j.jnutbio.2024.109625] [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: 01/18/2024] [Revised: 03/07/2024] [Accepted: 03/18/2024] [Indexed: 03/25/2024]
Abstract
Maternal obesity might induce obesity and metabolic alterations in the progeny. The study aimed to determine the effect of supplementing obese mothers with Mel (Mel) on thermogenesis and inflammation. C57BL/6 female mice (mothers) were fed from weaning to 12 weeks control diet (C, 17% kJ as fat) or a high-fat diet (HF, 49% kJ as fat) and then matted with male mice fed the control diet. Melatonin (10 mg/kg daily) was supplemented to mothers during gestation and lactation, forming the groups C, CMel, HF, and HFMel (n = 10/group). Twelve-week male offspring were studied (plasma biochemistry, immunohistochemistry, protein, and gene expressions at the hypothalamus - Hyp, subcutaneous white adipose tissue - sWAT, and interscapular brown adipose tissue - iBAT). Comparing HFMel vs. HF offspring, fat deposits and plasmatic proinflammatory markers decreased. Also, HFMel showed decreased Hyp proinflammatory markers and neuropeptide Y (anabolic) expression but improved proopiomelanocortin (catabolic) expression. Besides, HFMel sWAT adipocytes changed to a beige phenotype with-beta-3 adrenergic receptor and uncoupling protein-1 activation, concomitant with browning genes activation, triggering the iBAT thermogenic activity. In conclusion, compelling evidence indicated the beneficial effects of supplementing obese mothers with Mel on the health of their mature male offspring. Mel led to sWAT browning-related gene enhancement, increased iBAT thermogenis, and mitigated hypothalamic inflammation. Also, principal component analysis of the data significantly separated the untreated obese mother progeny from the progeny of treated obese mothers. If confirmed in humans, the findings encourage a future guideline recommending Mel supplementation during pregnancy and breastfeeding.
Collapse
Affiliation(s)
- Brenda A Nagagata
- Metabolism section, Laboratory of Morphometry, Metabolism and Cardiovascular Disease, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil; Nutrition section, Laboratory of Morphometry, Metabolism and Cardiovascular Disease, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gabrielle Brito
- Metabolism section, Laboratory of Morphometry, Metabolism and Cardiovascular Disease, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Carlos A Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
3
|
Spezani R, Marinho TS, Reis TS, Aguila MB, Mandarim-de-Lacerda CA. Cotadutide (GLP-1/Glucagon dual receptor agonist) modulates hypothalamic orexigenic and anorexigenic neuropeptides in obese mice. Peptides 2024; 173:171138. [PMID: 38147963 DOI: 10.1016/j.peptides.2023.171138] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/15/2023] [Accepted: 12/18/2023] [Indexed: 12/28/2023]
Abstract
The hypothalamic neuropeptides linked to appetite and satiety were investigated in obese mice treated with cotadutide (a dual receptor agonist of glucagon-like peptide 1 (GLP-1R)/Glucagon (GCGR)). Twelve-week-old male C57BL/6 mice were fed a control diet (C group, n = 20) or a high-fat diet (HF group, n = 20) for ten weeks. Each group was further divided, adding cotadutide treatment and forming groups C, CC, HF, and HFC for four additional weeks. The hypothalamic arcuate neurons were labeled by immunofluorescence, and protein expressions (Western blotting) for neuropeptide Y (NPY), proopiomelanocortin (POMC), agouti-related protein (AgRP), and cocaine- and amphetamine-regulated transcript (CART). Cotadutide enhanced POMC and CART neuropeptides and depressed NPY and AGRP neuropeptides. In addition, gene expressions (RT-qPCR) determined that Lepr (leptin receptor) and Calcr (calcitonin receptor) were diminished in HF compared to C but enhanced in CC compared to C and HFC compared to HF. Besides, Socs3 (suppressor of cytokine signaling 3) was decreased in HFC compared to HF, while Sst (somatostatin) was higher in HFC compared to HF; Tac1 (tachykinin 1) and Mc4r (melanocortin-4-receptor) were lower in HF compared to C but increased in HFC compared to HF. Also, Glp1r and Gcgr were higher in HFC compared to HF. In conclusion, the findings are compelling, demonstrating the effects of cotadutide on hypothalamic neuropeptides and hormone receptors of obese mice. Cotadutide modulates energy balance through the gut-brain axis and its associated signaling pathways. The study provides insights into the mechanisms underlying cotadutide's anti-obesity effects and its possible implications for obesity treatment.
Collapse
Affiliation(s)
- Renata Spezani
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Institute of Biology, Biomedical Center, The University of the State of Rio de Janeiro, Brazil
| | - Thatiany Souza Marinho
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Institute of Biology, Biomedical Center, The University of the State of Rio de Janeiro, Brazil
| | - Thiago Santos Reis
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Institute of Biology, Biomedical Center, The University of the State of Rio de Janeiro, Brazil
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Institute of Biology, Biomedical Center, The University of the State of Rio de Janeiro, Brazil
| | - Carlos A Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism, and Cardiovascular Disease, Institute of Biology, Biomedical Center, The University of the State of Rio de Janeiro, Brazil.
| |
Collapse
|
4
|
González-Domínguez Á, Jurado-Sumariva L, Domínguez-Riscart J, Saez-Benito A, González-Domínguez R. Parental obesity predisposes to exacerbated metabolic and inflammatory disturbances in childhood obesity within the framework of an altered profile of trace elements. Nutr Diabetes 2024; 14:2. [PMID: 38238301 PMCID: PMC10796909 DOI: 10.1038/s41387-024-00258-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND Family history of obesity is known to increase the odds of developing childhood obesity in the offspring, but its influence in underlying molecular complications remains unexplored. SUBJECTS/METHODS Here, we investigated a population-based cohort comprising children with obesity, with and without parental obesity (PO+, N = 20; PO-, N = 29), and lean healthy children as controls (N = 30), from whom plasma and erythrocyte samples were collected to characterize their multi-elemental profile, inflammatory status, as well as carbohydrate and lipid metabolisms. RESULTS We found parental obesity to be associated with unhealthier outcomes in children, as reflected in increased blood insulin levels and reduced insulin sensitivity, unfavorable lipid profile, and pro-inflammatory milieu. This was accompanied by moderate alterations in the content of trace elements, including increased copper-to-zinc ratios and iron deficiency in circulation, as well as metal accumulation within erythrocytes. CONCLUSIONS Therefore, we hypothesize that family history of obesity could be an important risk factor in modulating the characteristic multi-elemental alterations behind childhood obesity, which in turn could predispose to boost related comorbidities and metabolic complications.
Collapse
Affiliation(s)
- Álvaro González-Domínguez
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, Universidad de Cádiz, 11009, Cádiz, Spain
| | - Lucía Jurado-Sumariva
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, Universidad de Cádiz, 11009, Cádiz, Spain
| | - Jesús Domínguez-Riscart
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, Universidad de Cádiz, 11009, Cádiz, Spain
- Unidad de Endocrinología Pediátrica y Diabetes, Servicio de Pediatría, Hospital Universitario Puerta del Mar, 11009, Cádiz, Spain
| | - Ana Saez-Benito
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, Universidad de Cádiz, 11009, Cádiz, Spain
- Unidad de Análisis Clínicos, Hospital Universitario Puerta del Mar, 11009, Cádiz, Spain
| | - Raúl González-Domínguez
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, Universidad de Cádiz, 11009, Cádiz, Spain.
| |
Collapse
|
5
|
Zhang J, Xiong YW, Tan LL, Zheng XM, Zhang YF, Ling Q, Zhang C, Zhu HL, Chang W, Wang H. Sperm Rhoa m6A modification mediates intergenerational transmission of paternally acquired hippocampal neuronal senescence and cognitive deficits after combined exposure to environmental cadmium and high-fat diet in mice. JOURNAL OF HAZARDOUS MATERIALS 2023; 458:131891. [PMID: 37354721 DOI: 10.1016/j.jhazmat.2023.131891] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 05/28/2023] [Accepted: 06/17/2023] [Indexed: 06/26/2023]
Abstract
Little is currently known about the effect and mechanism of combined paternal environmental cadmium (Cd) and high-fat diet (HFD) on offspring cognitive ability. Here, using in vivo model, we found that combined paternal environmental Cd and HFD caused hippocampal neuronal senescence and cognitive deficits in offspring. MeRIP-seq revealed m6A level of Rhoa, a regulatory gene of cellular senescence, was significantly increased in combined environmental Cd and HFD-treated paternal sperm. Interestingly, combined paternal environmental Cd and HFD markedly enhanced Rhoa mRNA, its m6A and reader protein IGF2BP1 in offspring hippocampus. STM2457, the inhibitor of m6A modification, markedly mitigated paternal exposure-caused the elevation of hippocampal Rhoa m6A, neuronal senescence and cognitive deficits in offspring. In vitro experiments, Rhoa siR significantly reversed mouse hippocampal neuronal senescence. Igf2bp1 siR obviously reduced the level and stability of Rhoa in aging mouse hippocampal neuronal cells. In conclusion, combined paternal environmental Cd and HFD induce offspring hippocampal neuronal senescence and cognitive deficits by promoting IGF2BP1-mediated Rhoa stabilization in offspring hippocampus via elevating Rhoa m6A in paternal sperm.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, China
| | - Yong-Wei Xiong
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, China
| | - Lu-Lu Tan
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Xin-Mei Zheng
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Yu-Feng Zhang
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Qing Ling
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Chao Zhang
- Teaching Center for Preventive Medicine, School of Public Health, Anhui Medical University, 81 Meishan Road, Hefei, Anhui, 230032, China
| | - Hua-Long Zhu
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, China
| | - Wei Chang
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China
| | - Hua Wang
- Department of Toxicology, School of Public Health, Anhui Medical University, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institutes, China; Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, China.
| |
Collapse
|
6
|
Moon RJ, D’Angelo S, Holroyd CR, Crozier SR, Godfrey KM, Davies JH, Cooper C, Harvey NC. Parent-Offspring Associations in Body Composition: Findings From the Southampton Women's Survey Prospective Cohort Study. J Clin Endocrinol Metab 2023; 108:e726-e733. [PMID: 36943299 PMCID: PMC10438875 DOI: 10.1210/clinem/dgad128] [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: 11/07/2022] [Indexed: 03/23/2023]
Abstract
CONTEXT Children born to parents who are overweight or obese have a high risk of adult obesity, but it is unclear if transgenerational associations relating to unfavorable body composition differ by parent. OBJECTIVE To examine differential mother-offspring and father-offspring associations in body composition in early childhood. METHODS A total of 240 mother-father-offspring trios from a prospective UK population-based pre-birth cohort (Southampton Women's Survey) were included for anthropometry and dual-energy x-ray absorptiometry assessment of whole-body-less-head body composition in the offspring at 3 different ages (4, 6-7, and 8-9 years) and in the mother and father at the 8- to 9-year offspring visit. Associations were assessed using linear regression adjusting for the other parent. RESULTS Positive associations between mother-daughter body mass index (BMI) and fat mass were observed at ages 6 to 7 (BMI: β = .29 SD/SD, 95% CI = .10, .48; fat mass β = .27 SD/SD, 95% CI = .05, .48) and 8 to 9 years (BMI: β = .33 SD/SD, 95% CI = .13, .54; fat mass β = .31 SD/SD, 95% CI = .12, .49), with similar associations at age 4 years but bounding the 95% CI. The mother-son, father-son, and father-daughter associations for BMI and fat mass were weaker at each of the ages studied. CONCLUSION A strong association between the fat mass of mothers and their daughters but not their sons was observed. In contrast, father-offspring body composition associations were not evident. The dimorphic parent-offspring effects suggest particular attention should be given to early prevention of unfavorable body composition in girls born to mothers with excess adiposity.
Collapse
Affiliation(s)
- Rebecca J Moon
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton SO16 6YD, UK
- Paediatric Endocrinology, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Stefania D’Angelo
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton SO16 6YD, UK
| | - Christopher R Holroyd
- Department of Rheumatology, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Sarah R Crozier
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton SO16 6YD, UK
- NIHR Applied Research Collaboration Wessex, Southampton Science Park, Innovation Centre, Southampton, SO16 7NP, UK
| | - Keith M Godfrey
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton SO16 6YD, UK
- Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Justin H Davies
- Paediatric Endocrinology, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
- Faculty of Medicine, University of Southampton, Southampton SO16 6YD, UK
| | - Cyrus Cooper
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton SO16 6YD, UK
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
- NIHR Biomedical Research Centre, University of Oxford, Oxford OX4 2PG, UK
| | - Nicholas C Harvey
- MRC Lifecourse Epidemiology Centre, University of Southampton, Southampton SO16 6YD, UK
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| |
Collapse
|
7
|
Giovana Maciel Reis C, Rocha-Gomes A, Escobar Teixeira A, Gomes de Oliveira D, Mainy Oliveira Santiago C, Alves da Silva A, Regina Riul T, de Jesus Oliveira E. Short-term Cafeteria Diet Is Associated with Fat Mass Accumulation, Systemic and Amygdala Inflammation, and Anxiety-like Behavior in Adult Male Wistar Rats. Neuroscience 2023; 515:37-52. [PMID: 36773840 DOI: 10.1016/j.neuroscience.2023.02.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/31/2023] [Accepted: 02/03/2023] [Indexed: 02/12/2023]
Abstract
Obesity is linked to metabolic, hormonal and biochemical alterations, and is also a risk factor for behavioral disorders. Evidence suggests that these disorders may be related to the consumption of hypercaloric diets, fat mass accumulation and changes in inflammation and redox status. Although much is known about the chronic effects of hypercaloric diets on mental health, few studies have evaluated the consequences of short-term exposure of these diets on behavior. The aim of this study was to evaluate nutritional, behavioral (anxiety-like), inflammatory and redox status parameters in adult male Wistar rats exposed to short-term cafeteria diet. Adult Wistar male rats (90 days-old; n = 12/group) received, during 14 days, the diets: Control- standard diet; Simple Cafeteria Diet (SCD)- homogeneous cafeteria diet. Varied Cafeteria Diet (VCD)- cafeteria diet with rotation and variation. Nutritional analyzes and tests for anxiety-like behaviors were performed, in addition to inflammatory and redox status measurements in blood and amygdala. The SCD group showed higher fat energy intake, while the VCD group consumed more energy from carbohydrates. SCD and VCD showed higher fat mass accumulation, in addition to higher levels of TNFα, INFγ, TBARS and FRAP in the blood. Also, SCD and VCD groups reported high levels of TNFα in the amygdala. Regarding behavioral evaluations, SCD and VCD groups showed anxiogenesis in the elevated plus maze, light-dark box, and open field tests. Therefore, the two cafeteria diets induced obesity and systemic inflammation, which in turn, resulted in an increase in amygdala TNFα levels and anxiety-like behaviors in Wistar rats.
Collapse
Affiliation(s)
- Clarisse Giovana Maciel Reis
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina-MG, Brazil; Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição. Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina-MG, Brazil
| | - Arthur Rocha-Gomes
- Departamento de Farmacologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo (USP), Brazil
| | - Amanda Escobar Teixeira
- Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição. Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina-MG, Brazil
| | - Dalila Gomes de Oliveira
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina-MG, Brazil; Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição. Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina-MG, Brazil
| | - Camilla Mainy Oliveira Santiago
- Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição. Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina-MG, Brazil; Programa de Pós-Graduação em Ciências da Nutrição, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina-MG, Brazil
| | - Alexandre Alves da Silva
- Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição. Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina-MG, Brazil; Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina-MG, Brazil
| | - Tania Regina Riul
- Laboratório de Nutrição Experimental - LabNutrex - Departamento de Nutrição. Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina-MG, Brazil; Programa de Pós-Graduação em Ciências da Nutrição, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina-MG, Brazil
| | - Eduardo de Jesus Oliveira
- Programa de Pós-Graduação em Ciências Farmacêuticas, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina-MG, Brazil.
| |
Collapse
|
8
|
Rodrigues FDS, Jantsch J, Fraga GDF, Dias VS, Eller S, De Oliveira TF, Giovenardi M, Guedes RP. Cannabidiol treatment improves metabolic profile and decreases hypothalamic inflammation caused by maternal obesity. Front Nutr 2023; 10:1150189. [PMID: 36969815 PMCID: PMC10033544 DOI: 10.3389/fnut.2023.1150189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Accepted: 02/24/2023] [Indexed: 03/11/2023] Open
Abstract
IntroductionThe implications of maternal overnutrition on offspring metabolic and neuroimmune development are well-known. Increasing evidence now suggests that maternal obesity and poor dietary habits during pregnancy and lactation can increase the risk of central and peripheral metabolic dysregulation in the offspring, but the mechanisms are not sufficiently established. Furthermore, despite many studies addressing preventive measures targeted at the mother, very few propose practical approaches to treat the damages when they are already installed.MethodsHere we investigated the potential of cannabidiol (CBD) treatment to attenuate the effects of maternal obesity induced by a cafeteria diet on hypothalamic inflammation and the peripheral metabolic profile of the offspring in Wistar rats.ResultsWe have observed that maternal obesity induced a range of metabolic imbalances in the offspring in a sex-dependant manner, with higher deposition of visceral white adipose tissue, increased plasma fasting glucose and lipopolysaccharides (LPS) levels in both sexes, but the increase in serum cholesterol and triglycerides only occurred in females, while the increase in plasma insulin and the homeostatic model assessment index (HOMA-IR) was only observed in male offspring. We also found an overexpression of the pro-inflammatory cytokines tumor necrosis factor-alpha (TNFα), interleukin (IL) 6, and interleukin (IL) 1β in the hypothalamus, a trademark of neuroinflammation. Interestingly, the expression of GFAP, a marker for astrogliosis, was reduced in the offspring of obese mothers, indicating an adaptive mechanism to in utero neuroinflammation. Treatment with 50 mg/kg CBD oil by oral gavage was able to reduce white adipose tissue and revert insulin resistance in males, reduce plasma triglycerides in females, and attenuate plasma LPS levels and overexpression of TNFα and IL6 in the hypothalamus of both sexes.DiscussionTogether, these results indicate an intricate interplay between peripheral and central counterparts in both the pathogenicity of maternal obesity and the therapeutic effects of CBD. In this context, the impairment of internal hypothalamic circuitry caused by neuroinflammation runs in tandem with the disruptions of important metabolic processes, which can be attenuated by CBD treatment in both ends.
Collapse
Affiliation(s)
- Fernanda da Silva Rodrigues
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Jeferson Jantsch
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Gabriel de Farias Fraga
- Undergraduate Program in Biomedical Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Victor Silva Dias
- Undergraduate Program in Biomedical Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Sarah Eller
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Tiago Franco De Oliveira
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Márcia Giovenardi
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
| | - Renata Padilha Guedes
- Graduate Program in Biosciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
- Graduate Program in Health Sciences, Federal University of Health Sciences of Porto Alegre (UFCSPA), Porto Alegre, Rio Grande do Sul, Brazil
- *Correspondence: Renata Padilha Guedes,
| |
Collapse
|
9
|
Recent Advances in the Knowledge of the Mechanisms of Leptin Physiology and Actions in Neurological and Metabolic Pathologies. Int J Mol Sci 2023; 24:ijms24021422. [PMID: 36674935 PMCID: PMC9860943 DOI: 10.3390/ijms24021422] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/05/2023] [Accepted: 01/07/2023] [Indexed: 01/13/2023] Open
Abstract
Excess body weight is frequently associated with low-grade inflammation. Evidence indicates a relationship between obesity and cancer, as well as with other diseases, such as diabetes and non-alcoholic fatty liver disease, in which inflammation and the actions of various adipokines play a role in the pathological mechanisms involved in these disorders. Leptin is mainly produced by adipose tissue in proportion to fat stores, but it is also synthesized in other organs, where leptin receptors are expressed. This hormone performs numerous actions in the brain, mainly related to the control of energy homeostasis. It is also involved in neurogenesis and neuroprotection, and central leptin resistance is related to some neurological disorders, e.g., Parkinson's and Alzheimer's diseases. In peripheral tissues, leptin is implicated in the regulation of metabolism, as well as of bone density and muscle mass. All these actions can be affected by changes in leptin levels and the mechanisms associated with resistance to this hormone. This review will present recent advances in the molecular mechanisms of leptin action and their underlying roles in pathological situations, which may be of interest for revealing new approaches for the treatment of diseases where the actions of this adipokine might be compromised.
Collapse
|
10
|
Ajackson M, Nagagata BA, Marcondes-de-Castro IA, Mandarim-de-Lacerda CA, Aguila MB. Adult mice offspring of obese mothers supplemented with melatonin show lessened liver steatosis, inflammation, oxidative stress, and endoplasmic reticulum stress. Life Sci 2023; 312:121253. [PMID: 36481166 DOI: 10.1016/j.lfs.2022.121253] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 11/23/2022] [Accepted: 11/26/2022] [Indexed: 12/12/2022]
Abstract
AIMS To investigate, in the liver of adult offspring, the possible effects of melatonin supplementation in the obese mother during pregnancy and lactation. MAIN METHODS C57BL/6 females were fed with a control (C) or a high-fat (HF) diet and supplemented with melatonin (Mel) during the pregnancy and lactation, forming the groups: C, CMel, HF, and HFMel. After weaning until three months old, the offspring only received the C diet. KEY FINDINGS The HF mothers and their offspring showed higher body weight (BW) than the C mothers and offspring. However, at 3-mo-old, BW was reduced in HFMel vs. HF offspring. Also, plasmatic and liver lipid markers increased in HF vs. C offspring but were reduced in HFMel vs. HF offspring. Liver lipid content was lessened in HFMel vs. HF offspring by 50 %. Also, lipid metabolism, pro-inflammatory and endoplasmic reticulum (ER) stress genes were higher expressed in HF vs. C offspring but reduced in HFMel vs. HF offspring. Contrarily, beta-oxidation and antioxidant enzyme genes were less expressed in HF vs. C offspring but improved in HFMel vs. HF offspring. Finally, AMPK/mTOR pathway genes, initially dysregulated in the HF, were restored in the HFMel offspring. SIGNIFICANCE The obese mother leads to liver alterations in the offspring. Current findings demonstrated the maternal melatonin supplementation during pregnancy and lactation in adult offspring's liver. Consequently, the effects were seen in mitigating the liver's AMPK/mTOR pathway genes, lipogenesis, beta-oxidation, inflammation, oxidative stress, and ER stress, preventing liver disease progression in the offspring.
Collapse
Affiliation(s)
- Matheus Ajackson
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Brenda A Nagagata
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ilitch A Marcondes-de-Castro
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlos A Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
11
|
Hameed M, Geerling E, Pinto AK, Miraj I, Weger-Lucarelli J. Immune response to arbovirus infection in obesity. Front Immunol 2022; 13:968582. [PMID: 36466818 PMCID: PMC9716109 DOI: 10.3389/fimmu.2022.968582] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 11/04/2022] [Indexed: 12/26/2023] Open
Abstract
Obesity is a global health problem that affects 650 million people worldwide and leads to diverse changes in host immunity. Individuals with obesity experience an increase in the size and the number of adipocytes, which function as an endocrine organ and release various adipocytokines such as leptin and adiponectin that exert wide ranging effects on other cells. In individuals with obesity, macrophages account for up to 40% of adipose tissue (AT) cells, three times more than in adipose tissue (10%) of healthy weight individuals and secrete several cytokines and chemokines such as interleukin (IL)-1β, chemokine C-C ligand (CCL)-2, IL-6, CCL5, and tumor necrosis factor (TNF)-α, leading to the development of inflammation. Overall, obesity-derived cytokines strongly affect immune responses and make patients with obesity more prone to severe symptoms than patients with a healthy weight. Several epidemiological studies reported a strong association between obesity and severe arthropod-borne virus (arbovirus) infections such as dengue virus (DENV), chikungunya virus (CHIKV), West Nile virus (WNV), and Sindbis virus (SINV). Recently, experimental investigations found that DENV, WNV, CHIKV and Mayaro virus (MAYV) infections cause worsened disease outcomes in infected diet induced obese (DIO) mice groups compared to infected healthy-weight animals. The mechanisms leading to higher susceptibility to severe infections in individuals with obesity remain unknown, though a better understanding of the causes will help scientists and clinicians develop host directed therapies to treat severe disease. In this review article, we summarize the effects of obesity on the host immune response in the context of arboviral infections. We have outlined that obesity makes the host more susceptible to infectious agents, likely by disrupting the functions of innate and adaptive immune cells. We have also discussed the immune response of DIO mouse models against some important arboviruses such as CHIKV, MAYV, DENV, and WNV. We can speculate that obesity-induced disruption of innate and adaptive immune cell function in arboviral infections ultimately affects the course of arboviral disease. Therefore, further studies are needed to explore the cellular and molecular aspects of immunity that are compromised in obesity during arboviral infections or vaccination, which will be helpful in developing specific therapeutic/prophylactic interventions to prevent immunopathology and disease progression in individuals with obesity.
Collapse
Affiliation(s)
- Muddassar Hameed
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| | - Elizabeth Geerling
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO, United States
| | - Amelia K. Pinto
- Department of Molecular Microbiology and Immunology, Saint Louis University, St. Louis, MO, United States
| | - Iqra Miraj
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, China
| | - James Weger-Lucarelli
- Department of Biomedical Sciences and Pathobiology, VA-MD Regional College of Veterinary Medicine, Virginia Tech, Blacksburg, VA, United States
| |
Collapse
|
12
|
da Silva RKB, de Vasconcelos DAA, da Silva AVE, da Silva RPB, de Oliveira Neto OB, Galindo LCM. Effects of maternal high-fat diet on the hypothalamic components related to food intake and energy expenditure in mice offspring. Life Sci 2022; 307:120880. [PMID: 35963301 DOI: 10.1016/j.lfs.2022.120880] [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: 06/26/2022] [Revised: 08/02/2022] [Accepted: 08/08/2022] [Indexed: 11/25/2022]
Abstract
Maternal exposure to a high-fat diet (HFD) during pregnancy and lactation has been related to changes in the hypothalamic circuits involved in the regulation of food intake. Furthermore, maternal HFD during the critical period of development can alter the offspring's metabolic programming with long-term repercussions. This study systematically reviewed the effects of HFD consumption during pre-pregnancy, pregnancy and/or lactation. The main outcomes evaluated were food intake; body weight; cellular or molecular aspects of peptides and hypothalamic receptors involved in the regulation of energy balance in mice. Two independent authors performed a search in the electronic databases Medline/PubMed, LILACS, Web of Science, EMBASE, SCOPUS and Sigle via Open Gray. Included were experimental studies of mice exposed to HFD during pregnancy and/or lactation that evaluated body composition, food intake, energy expenditure and hypothalamic components related to energy balance. Internal validity was assessed using the SYRCLE risk of bias. The Kappa index was measured to analyze the agreement between reviewers. The PRISMA statement was used to report this systematic review. Most studies demonstrated that there was a higher body weight, body fat deposits and food intake, as well as alterations in the expression of hypothalamic neuropeptides in offspring that consumed HFD. Therefore, the maternal diet can affect the phenotype and metabolism of the offspring, in addition to harming the hypothalamic circuits and favoring the orexigenic pathways.
Collapse
Affiliation(s)
- Regina Katiuska Bezerra da Silva
- Post-Graduate Program in Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, 55608-680 Vitória de Santo Antão, PE, Brazil
| | - Diogo Antonio Alves de Vasconcelos
- Post-Graduate Program in Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, 55608-680 Vitória de Santo Antão, PE, Brazil; Department of Nutrition, Federal University of Pernambuco, 50670-901 Recife, PE, Brazil; Nutrition and Phenotypic Plasticity Study Unit, Department of Nutrition, Federal University of Pernambuco, 50670-901 Recife, PE, Brazil
| | | | - Roxana Patrícia Bezerra da Silva
- Post-Graduate Program in Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, 55608-680 Vitória de Santo Antão, PE, Brazil
| | | | - Lígia Cristina Monteiro Galindo
- Post-Graduate Program in Nutrition, Physical Activity and Phenotypic Plasticity, Federal University of Pernambuco, 55608-680 Vitória de Santo Antão, PE, Brazil; Department of Anatomy, Federal University of Pernambuco, 50670-901 Recife, PE, Brazil; Nutrition and Phenotypic Plasticity Study Unit, Department of Nutrition, Federal University of Pernambuco, 50670-901 Recife, PE, Brazil.
| |
Collapse
|
13
|
Santiago CMO, de Oliveira DG, Rocha‐Gomes A, Oliveira G, Bernardes EDO, Dias PL, Reis ÍG, Severiano CM, da Silva AA, Lessa MR, Dessimoni Pinto NAV, Riul TR. Unripe banana flour (
Musa cavendishii
) promotes increased hypothalamic antioxidant activity, reduced caloric intake, and abdominal fat accumulation in rats on a high‐fat diet. J Food Biochem 2022; 46:e14341. [DOI: 10.1111/jfbc.14341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 06/17/2022] [Accepted: 06/27/2022] [Indexed: 11/30/2022]
Affiliation(s)
- Camilla M. O. Santiago
- Programa de Pós‐Graduação em Ciências da Nutrição Universidade Federal dos Vales do Jequitinhonha e Mucuri Diamantina Minas Gerais Brazil
- Laboratório de Nutrição Experimental, LabNutrex, Departamento de Nutrição Universidade Federal dos Vales do Jequitinhonha e Mucuri Diamantina Minas Gerais Brazil
| | - Dalila G. de Oliveira
- Laboratório de Nutrição Experimental, LabNutrex, Departamento de Nutrição Universidade Federal dos Vales do Jequitinhonha e Mucuri Diamantina Minas Gerais Brazil
- Programa de Pós‐Graduação em Ciências Farmacêuticas Universidade Federal dos Vales do Jequitinhonha e Mucuri Diamantina Minas Gerais Brazil
| | - Arthur Rocha‐Gomes
- Laboratório de Nutrição Experimental, LabNutrex, Departamento de Nutrição Universidade Federal dos Vales do Jequitinhonha e Mucuri Diamantina Minas Gerais Brazil
| | - Gabriel A. Oliveira
- Laboratório de Nutrição Experimental, LabNutrex, Departamento de Nutrição Universidade Federal dos Vales do Jequitinhonha e Mucuri Diamantina Minas Gerais Brazil
| | - Eduardo de Oliveira Bernardes
- Laboratório de Nutrição Experimental, LabNutrex, Departamento de Nutrição Universidade Federal dos Vales do Jequitinhonha e Mucuri Diamantina Minas Gerais Brazil
| | - Patrick L. Dias
- Laboratório de Nutrição Experimental, LabNutrex, Departamento de Nutrição Universidade Federal dos Vales do Jequitinhonha e Mucuri Diamantina Minas Gerais Brazil
| | - Ítalo G. Reis
- Programa de Pós‐Graduação em Ciências da Nutrição Universidade Federal dos Vales do Jequitinhonha e Mucuri Diamantina Minas Gerais Brazil
- Laboratório de Nutrição Experimental, LabNutrex, Departamento de Nutrição Universidade Federal dos Vales do Jequitinhonha e Mucuri Diamantina Minas Gerais Brazil
| | - Cecília M. Severiano
- Laboratório de Nutrição Experimental, LabNutrex, Departamento de Nutrição Universidade Federal dos Vales do Jequitinhonha e Mucuri Diamantina Minas Gerais Brazil
| | - Alexandre A. da Silva
- Laboratório de Nutrição Experimental, LabNutrex, Departamento de Nutrição Universidade Federal dos Vales do Jequitinhonha e Mucuri Diamantina Minas Gerais Brazil
- Programa de Pós‐Graduação em Ciências da Saúde Universidade Federal dos Vales do Jequitinhonha e Mucuri Diamantina Minas Gerais Brazil
| | - Mayara R. Lessa
- Laboratório de Nutrição Experimental, LabNutrex, Departamento de Nutrição Universidade Federal dos Vales do Jequitinhonha e Mucuri Diamantina Minas Gerais Brazil
| | - Nisia A. V. Dessimoni Pinto
- Programa de Pós‐Graduação em Ciências da Nutrição Universidade Federal dos Vales do Jequitinhonha e Mucuri Diamantina Minas Gerais Brazil
| | - Tania R. Riul
- Programa de Pós‐Graduação em Ciências da Nutrição Universidade Federal dos Vales do Jequitinhonha e Mucuri Diamantina Minas Gerais Brazil
- Laboratório de Nutrição Experimental, LabNutrex, Departamento de Nutrição Universidade Federal dos Vales do Jequitinhonha e Mucuri Diamantina Minas Gerais Brazil
| |
Collapse
|
14
|
Gnawali A. Prematurity and the Risk of Development of Childhood Obesity: Piecing Together the Pathophysiological Puzzle. A Literature Review. Cureus 2021; 13:e20518. [PMID: 35070553 PMCID: PMC8765585 DOI: 10.7759/cureus.20518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/15/2021] [Indexed: 11/21/2022] Open
Abstract
One of the most devastating public health challenges in the twenty-first century is childhood obesity, and its prevalence is growing at a frightening rate. Premature infants have a greater likelihood of childhood obesity at age six to 16 compared to term infants. This study aims to explore the underlying mechanism of developing childhood obesity in this high-risk group. There are most likely multiple interconnected and supporting mechanisms that put this vulnerable population at risk of childhood obesity. Inflammation is a possible root cause. Prenatal causes included epigenetic changes as well as placental inflammation. Disturbances in hormonal pathways and elevated levels of serum bilirubin are possible explanations. Furthermore, preventable factors in the postnatal period were identified, such as weight gain and exclusive breastfeeding. The prevalence of childhood obesity in preterm infants is high; thus, it is essential to understand the pathophysiology and address any preventable factors to decrease this disease burden.
Collapse
|
15
|
César H, Sertorio MN, de Souza EA, Jamar G, Santamarina A, Jucá A, Casagrande BP, Pisani LP. Parental high-fat high-sugar diet programming and hypothalamus adipose tissue axis in male Wistar rats. Eur J Nutr 2021; 61:523-537. [PMID: 34657184 DOI: 10.1007/s00394-021-02690-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 09/28/2021] [Indexed: 01/04/2023]
Abstract
PURPOSE Maternal nutrition during early development and paternal nutrition pre-conception can programme offspring health status. Hypothalamus adipose axis is a target of developmental programming, and paternal and maternal high-fat, high-sugar diet (HFS) may be an important factor that predisposes offspring to develop obesity later in life. This study aims to investigate Wistar rats' maternal and paternal HFS differential contribution on the development, adiposity, and hypothalamic inflammation in male offspring from weaning until adulthood. METHODS Male progenitors were fed a control diet (CD) or HFS for 10 weeks before mating. After mating, dams were fed CD or HFS only during pregnancy and lactation. Forming the following male offspring groups: CD-maternal and paternal CD; MH-maternal HFS and paternal CD; PH-maternal CD and paternal HFS; PMH-maternal and paternal HFS. After weaning, male offspring were fed CD until adulthood. RESULTS Maternal HFS diet increased weight, visceral adiposity, and serum total cholesterol levels, and decreased hypothalamic weight in weanling male rats. In adult male offspring, maternal HFS increased weight, glucose levels, and hypothalamic NFκBp65. Paternal HFS diet lowered hypothalamic insulin receptor levels in weanling offspring and glucose and insulin levels in adult offspring. The combined effects of maternal and paternal HFS diets increased triacylglycerol, leptin levels, and hypothalamic inflammation in weanling rats, and increased visceral adiposity in adulthood. CONCLUSION Male offspring intake of CD diet after weaning reversed part of the effects of parental HFS diet during the perinatal period. However, maternal and paternal HFS diet affected adiposity and hypothalamic inflammation, which remained until adulthood.
Collapse
Affiliation(s)
- Helena César
- Programa de Pós-Graduação Interdisciplinar em Ciências da Saúde, Universidade Federal de São Paulo-UNIFESP, Santos, SP, Brazil
| | | | - Esther Alves de Souza
- Programa de Pós-Graduação em Nutrição, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Giovana Jamar
- Departamento de Biociências, Universidade Federal de São Paulo, Silva Jardim, 136. Laboratório 311, 3° andar, Vila Mathias, Santos, SP, 11015-020, Brazil
| | - Aline Santamarina
- Departamento de Biociências, Universidade Federal de São Paulo, Silva Jardim, 136. Laboratório 311, 3° andar, Vila Mathias, Santos, SP, 11015-020, Brazil
| | - Andrea Jucá
- Departamento de Biociências, Universidade Federal de São Paulo, Silva Jardim, 136. Laboratório 311, 3° andar, Vila Mathias, Santos, SP, 11015-020, Brazil
| | - Breno Picin Casagrande
- Departamento de Biociências, Universidade Federal de São Paulo, Silva Jardim, 136. Laboratório 311, 3° andar, Vila Mathias, Santos, SP, 11015-020, Brazil
| | - Luciana Pellegrini Pisani
- Departamento de Biociências, Universidade Federal de São Paulo, Silva Jardim, 136. Laboratório 311, 3° andar, Vila Mathias, Santos, SP, 11015-020, Brazil.
| |
Collapse
|
16
|
Callet T, Li H, Surget A, Terrier F, Sandres F, Lanuque A, Panserat S, Marandel L. No adverse effect of a maternal high carbohydrate diet on their offspring, in rainbow trout ( Oncorhynchus mykiss). PeerJ 2021; 9:e12102. [PMID: 34589301 PMCID: PMC8434805 DOI: 10.7717/peerj.12102] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 08/11/2021] [Indexed: 01/09/2023] Open
Abstract
In order to develop a sustainable salmonid aquaculture, it is essential to continue to reduce the use of the protein-rich fishmeal. One promising solution to do so is the use of plant-derived carbohydrates in diet destined to broodstock. However, in mammals, the reduction of protein content (replaced by carbohydrates) in parental diet is known to have strong adverse effects on offspring phenotypes and metabolism. For the first time, the effect of a paternal and a maternal high carbohydrate-low protein diet was assessed on progeny at long term in the rainbow trout. A 30% protein diminution in both males and females broodstock diet during 10 month and 5 months, respectively, did not trigger adverse consequences on their offspring. At the molecular level, offspring transcriptomes were not significantly altered, emphasizing no effect on metabolism. Tenuous differences in the biochemical composition of the liver and the viscera were observed. The recorded effects remained in the normal range of value and accordingly offspring growth were not negatively affected over the long term. Overall, we demonstrated here that a 30% protein diminution during gametogenesis is feasible, confirming the possibility to increase the proportion of plant-derived carbohydrates in female broodstock diets to replace fishmeal proteins.
Collapse
Affiliation(s)
- Therese Callet
- Institut National de la Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Saint-Pée-sur-Nivelle, France
| | - Hongyan Li
- Institut National de la Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Saint-Pée-sur-Nivelle, France.,State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Anne Surget
- Institut National de la Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Saint-Pée-sur-Nivelle, France
| | - Frederic Terrier
- Institut National de la Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Saint-Pée-sur-Nivelle, France
| | - Franck Sandres
- Institut National de la Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Saint-Pée-sur-Nivelle, France
| | - Anthony Lanuque
- Institut National de la Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Saint-Pée-sur-Nivelle, France
| | - Stephane Panserat
- Institut National de la Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Saint-Pée-sur-Nivelle, France
| | - Lucie Marandel
- Institut National de la Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Saint-Pée-sur-Nivelle, France
| |
Collapse
|
17
|
Zhu F, Zhang D, Shen F, Xu K, Huang X, Liu J, Zhang J, Teng Y. Maternal Socs3 knockdown attenuates postnatal obesity caused by an early life environment of maternal obesity and intrauterine overnutrition in progeny mice. IUBMB Life 2021; 73:1210-1221. [PMID: 34184397 DOI: 10.1002/iub.2526] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Revised: 03/29/2021] [Accepted: 06/20/2021] [Indexed: 11/07/2022]
Abstract
Pathological states in the early life environment of mammalian offspring, including maternal obesity and intrauterine overnutrition, can induce obesity and metabolic disorder later in life. Leptin resistance caused by upregulation of Socs3 in the hypothalamus of offspring was believed to be the main mechanism of this effect. In this study, obese mother (OM) and lean mother (LM) models were generated by feeding C57BL/6N female mice a high-fat diet or standard lean diet, respectively. Additionally, an obese mother with intervention (OMI) model was generated by injecting the high-fat diet group with Socs3-shRNA lentivirus during early pregnancy. The offspring of the groups was correspondingly named OM-F1 , LM-F1 , and OMI-F1 , representing progeny mouse models of different early life environments. The offspring were fed a high-fat diet to test their propensity for obesity. The body weight, food intake and fat accumulation were higher, while glucose intolerance and insulin resistance were worse in the OM-F1 group than LM-F1 group. By contrast, the obesity phenotype, hyperphagia and metabolic disorder were alleviated in the OMI-F1 group compared with the OM-F1 group. The mechanism was identified that downregulation of hypothalamic SOCS3 resulted in an increased level of p-STAT3 and p-JAK2, which ameliorated the leptin resistance and restored the lean expression of appetite regulatory genes (Pomc and Agrp) in hypothalamus of OMI-F1 group. Taken together, these results indicate that reducing maternal Socs3 expression during pregnancy can attenuate obesity caused by the early life environment in mice, which may inspire therapies that enable obese mothers to bear metabolically healthy children.
Collapse
Affiliation(s)
- Feng Zhu
- Department of Graduate, Bengbu Medical College, Bengbu, China
- College of Biological and Chemical Science and Engineering, Jiaxing University, Jiaxing, China
- Children's Medical Center, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Dawei Zhang
- College of Biological and Chemical Science and Engineering, Jiaxing University, Jiaxing, China
| | - Fangfang Shen
- Children's Medical Center, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Ke Xu
- Children's Medical Center, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Xin Huang
- College of Biological and Chemical Science and Engineering, Jiaxing University, Jiaxing, China
| | - Jue Liu
- Children's Medical Center, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jin Zhang
- College of Biological and Chemical Science and Engineering, Jiaxing University, Jiaxing, China
| | - Yiqun Teng
- Department of Graduate, Bengbu Medical College, Bengbu, China
- Children's Medical Center, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
18
|
Spezani R, da Silva RR, Martins FF, de Souza Marinho T, Aguila MB, Mandarim-de-Lacerda CA. Intermittent fasting, adipokines, insulin sensitivity, and hypothalamic neuropeptides in a dietary overload with high-fat or high-fructose diet in mice. J Nutr Biochem 2020; 83:108419. [PMID: 32580132 DOI: 10.1016/j.jnutbio.2020.108419] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 03/14/2020] [Accepted: 05/14/2020] [Indexed: 02/06/2023]
Abstract
The intermittent fasting (IF) might have benefits on metabolism and food intake. Twelve-week old C57BL/6 J mice were fed a control diet (C, 10% kcal fat), a high-fat diet (HF, 50% kcal fat) or a high-fructose diet (HFru, 50% kcal fructose) for 8 weeks, then half of the animals in each group underwent IF (24 h fed, 24 h fasting) for an additional 4 weeks. Although food intake on the fed day remained the same for all groups, all fasting groups showed a reduction in body mass compared to their counterparts. IF reduced total cholesterol, triacylglycerol, fasting glucose, fasting insulin resistance index, and plasma leptin, but increased plasma adiponectin. IF reduced Leptin gene expression in the HF-IF group, but increased proinflammatory markers in the hypothalamus, also in the C-IF group. Both groups HFru-IF and C-IF, showed alterations in the leptin signaling pathway (Leptin, OBRb, and SOCS3), mainly in the HFru-IF group, suggesting leptin resistance. NPY and POMC neuropeptides labeled the neurons of the hypothalamus by immunofluorescence, corroborating qualitatively other quantitative findings of the study. In conclusion, current results are convincing in demonstrating the IF effect on central regulation of food intake control, as shown by NPY and POMC neuropeptide expressions, resulting in a lower weight gain. Besides, IF improves glycemia, lipid metabolism, and consequently insulin and leptin resistance. However, there is increased expression of inflammatory markers in mouse hypothalamus challenged by the HF and HFru diets, which in the long term may induce adverse effects.
Collapse
Affiliation(s)
- Renata Spezani
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Centre, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renata Ribeiro da Silva
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Centre, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fabiane Ferreira Martins
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Centre, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Thatiany de Souza Marinho
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Centre, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcia Barbosa Aguila
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Centre, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlos A Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Centre, Institute of Biology, The University of the State of Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
19
|
Sex-linked changes and high cardiovascular risk markers in the mature progeny of father, mother, or both father and mother consuming a high-fructose diet. Nutrition 2019; 71:110612. [PMID: 31785517 DOI: 10.1016/j.nut.2019.110612] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 08/28/2019] [Accepted: 09/08/2019] [Indexed: 11/22/2022]
Abstract
OBJECTIVES The aim of this study was to observe the developmental origins of health and disease affecting offspring owing to the consumption of a diet containing high fructose by the father or mother or both, considering that progeny only received a control diet during postnatal life. METHODS Male (future father) and female (future mother) C57 BL/6 mice were fed a high-fructose diet (HFru; 45% energy) or a control diet (C) for 8 wk before mating until lactation. The offspring was termed according to sex, maternal diet (first acrostic), and paternal diet (second acrostic); and received a balanced control diet until 3-mo of age when they were sacrificed. Body mass (BM), plasmatic leptin, adiponectin, uric acid, and systolic blood pressure (BP) were measured in mature offspring. RESULTS Fasting glycemia and insulin were elevated in HFru fathers and mothers. Although there was no change in BM, fasting glycemia, or insulin of the offspring, those of HFru fathers, HFru mothers, and HFru fathers and mothers presented higher genital fat pad, leptin, uric acid, and BP, and lower adiponectin. The values of leptin and BP were maximized when both parents consumed a HFru diet. Also, there was sexual dimorphism in most of the variables, with the male offspring being affected to a greater extent than the females. CONCLUSIONS Consumption of a fructose-rich diet by the father, the mother, or both negatively affected the adipokines, BP, and uric acid concentrations of mature offspring, with males being more affected than females. It is significant to consider that high BP and plasmatic uric acid correspond to markers of elevated cardiovascular risk in the progeny.
Collapse
|
20
|
Associations of prenatal exposure to impaired glucose tolerance with eating in the absence of hunger in early adolescence. Int J Obes (Lond) 2019; 43:1903-1913. [PMID: 30622313 DOI: 10.1038/s41366-018-0296-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 10/25/2018] [Accepted: 11/30/2018] [Indexed: 01/25/2023]
Abstract
OBJECTIVE Exposure to impaired gestational glucose tolerance has been shown to have sex-specific associations with offspring obesity risk, perhaps by affecting the development of appetite regulation. We examined the extent to which prenatal exposure to impaired glucose tolerance was associated with eating in the absence of hunger (EAH) in early adolescent offspring, and in turn, whether EAH was cross-sectionally associated with body composition. METHODS We included data from 1097 adolescents participating in Project Viva, a pre-birth longitudinal cohort. We obtained the results of two-stage prenatal glycemic screening (50 g glucose challenge test, followed if abnormal by 100 g oral glucose tolerance test) at 26-28 weeks of gestation, and categorized mothers as having normal glucose tolerance, isolated hyperglycemia (IH, n = 92, 8.4%), impaired glucose tolerance (IGT, n = 36, 3.3%), or gestational diabetes mellitus (GDM, n = 52, 4.7%). At a median age of 13 years, offspring reported on two modified items of the Eating in the Absence of Hunger in Children and Adolescents questionnaire, we measured height and weight, and performed dual X-ray absorptiometry scans to assess fat and fat-free mass. We used multivariable linear regression analyses adjusted for sociodemographic and prenatal covariates, including maternal pre-pregnancy BMI. RESULTS On a ten-point scale, the mean (SD) EAH score was 4.4 points (SD = 1.5) in boys and 4.4 (SD = 1.4) in girls. In girls, prenatal exposure to both IH and IGT was associated with more EAH compared with normal glucose tolerance (e.g., for IH: 0.56 points, 95% CI: 0.17, 0.96), whereas in boys, prenatal exposure to IGT was associated with less EAH (-0.81 points, 95% CI: -1.41, -0.21). We did not observe an association between exposure to GDM and EAH, nor did we observe associations between EAH and body composition in early adolescence. CONCLUSIONS These findings suggest sex-specific associations of exposure to impaired gestational glucose tolerance with offspring EAH in early adolescence.
Collapse
|
21
|
|
22
|
Zhang YP, Huo YL, Fang ZQ, Wang XF, Li JD, Wang HP, Peng W, Johnson AK, Xue B. Maternal high-fat diet acts on the brain to induce baroreflex dysfunction and sensitization of angiotensin II-induced hypertension in adult offspring. Am J Physiol Heart Circ Physiol 2018; 314:H1061-H1069. [PMID: 29373045 PMCID: PMC6008148 DOI: 10.1152/ajpheart.00698.2017] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 01/22/2018] [Accepted: 01/22/2018] [Indexed: 12/12/2022]
Abstract
Accumulating evidence indicates that maternal high-fat diet (HFD) is associated with metabolic syndrome and cardiovascular disease in adult offspring. The present study tested the hypothesis that maternal HFD modulates the brain renin-angiotensin system (RAS), oxidative stress, and proinflammatory cytokines that alter angiotensin II (ANG II) and TNF-α actions and sensitize the ANG II-elicited hypertensive response in adult offspring. All offspring were cross fostered by dams on the same or opposite diet to yield the following four groups: offspring from normal-fat control diet-fed dams suckled by control diet-fed dams (OCC group) or by HFD-fed dams (OCH group) and offspring from HFD-fed dams fed a HFD suckled by control diet-fed dams (OHC group) or by HFD-fed dams (OHH group). RT-PCR analyses of the lamina terminalis and paraventricular nucleus indicated upregulation of mRNA expression of several RAS components, NADPH oxidase, and proinflammatory cytokines in 10-wk-old male offspring of dams fed a HFD during either pregnancy, lactation, or both (OHC, OCH, and OHH groups). These offspring also showed decreased cardiac baroreflex sensitivity and increased pressor responses to intracerebroventricular microinjection of either ANG II or TNF-α. Furthermore, chronic systemic infusion of ANG II resulted in enhanced upregulation of mRNA expression of RAS components, NADPH oxidase, and proinflammatory cytokines in the lamina terminalis and paraventricular nucleus and an augmented hypertensive response in the OHC, OCH, and OHH groups compared with the OCC group. The results suggest that maternal HFD blunts cardiac baroreflex function and enhances pressor responses to ANG II or proinflammatory cytokines through upregulation of the brain RAS, oxidative stress, and inflammation. NEW & NOTEWORTHY The results of our study indicate that a maternal high-fat diet during either pregnancy or lactation is sufficient for perinatal programming of sensitization for hypertension, which is associated with hyperreactivity of central cardiovascular nuclei that, in all likelihood, involves elevated expression of the renin-angiotensin system, NADPH oxidase, and proinflammatory cytokines. The present study demonstrates, for the first time, the central mechanism underlying maternal high-fat diet sensitization of the hypertensive response in adult offspring.
Collapse
Affiliation(s)
| | | | - Zhi-Qin Fang
- First Affiliated Hospital, Hebei North University, Zhangjiakou City, Hebei , China
| | | | | | | | | | - Alan Kim Johnson
- Department of Psychological and Brain Sciences, University of Iowa , Iowa City, Iowa
| | - Baojian Xue
- Department of Psychological and Brain Sciences, University of Iowa , Iowa City, Iowa
| |
Collapse
|
23
|
Liver metabolism in adult male mice offspring: consequences of a maternal, paternal or both maternal and paternal high-fructose diet. J Dev Orig Health Dis 2018; 9:450-459. [DOI: 10.1017/s2040174418000235] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
AbstractThe study aimed to evaluate the consequences of the consumption of a high-fructose diet (HFR; fructose was responsible for 45% of the energy from carbohydrates) by the mother, the father, or both on C57BL/6 adult male offspring. Non-consanguineous parents received the diet (HFR or control, C) from 8 weeks before mating until weaning (n=10 fathers and n=10 mothers on each diet). After weaning, only the C diet was offered to offspring. The groups were formed by one male randomly taken from each litter. The offspring groups were identified according to the mother’s diet (the first letter), then the father’s diet (the second letter), that is, C/C, C/HFR, HFR/C, HFR/HFR (n=10 per group). The parents exhibited the following characteristics: compared with those of the C group, the HFR parents had higher blood pressure (BP), enlarged liver, increased hepatic triacylglycerol content, hypercholesterolemia, hypertriglyceridemia, high plasma leptin and low adiponectin. The offspring exhibited the following characteristics: compared with the C/C group, the HFR/HFR group had high BP. The C/HFR, HFR/C and HFR/HFR showed elevated uric acid and leptin levels and diminished adiponectin. The HFR/HFR group showed liver inflammation (increased NFκB, SOCS3, JNK, TNF-α, IL1-β and IL6 levels). Likewise, SREBP-1c and FAS were upregulated. In conclusion, the consumption of a HFR by the mother and/or father is associated with adverse effects on liver metabolism in adult male offspring. When both mother and father are fed a HFR, the adverse effects on the offspring are more severe.
Collapse
|
24
|
Zhang WX, Li YP, Fan J, Chen HJ, Li GL, Ouyang YQ, Yan YE. Perinatal nicotine exposure increases obesity susceptibility by peripheral leptin resistance in adult female rat offspring. Toxicol Lett 2018; 283:91-99. [DOI: 10.1016/j.toxlet.2017.11.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 11/09/2017] [Accepted: 11/13/2017] [Indexed: 12/11/2022]
|
25
|
Ornellas F, Carapeto PV, Mandarim‐de‐Lacerda CA, Aguila MB. Obese fathers lead to an altered metabolism and obesity in their children in adulthood: review of experimental and human studies. JORNAL DE PEDIATRIA (VERSÃO EM PORTUGUÊS) 2017. [DOI: 10.1016/j.jpedp.2017.08.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
26
|
Ornellas F, Carapeto PV, Mandarim-de-Lacerda CA, Aguila MB. Obese fathers lead to an altered metabolism and obesity in their children in adulthood: review of experimental and human studies. J Pediatr (Rio J) 2017; 93:551-559. [PMID: 28822233 DOI: 10.1016/j.jped.2017.02.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Accepted: 01/18/2017] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE To discuss the recent literature on paternal obesity, focusing on the possible mechanisms of transmission of the phenotypes from the father to the children. SOURCES A non-systematic review in the PubMed database found few publications in which paternal obesity was implicated in the adverse transmission of characteristics to offspring. Specific articles on epigenetics were also evaluated. As the subject is recent and still controversial, all articles were considered regardless of year of publication. SUMMARY OF FINDINGS Studies in humans and animals have established that paternal obesity impairs their hormones, metabolism, and sperm function, which can be transmitted to their offspring. In humans, paternal obesity results in insulin resistance/type 2 diabetes and increased levels of cortisol in umbilical cord blood, which increases the risk factors for cardiovascular disease. Notably, there is an association between body fat in parents and the prevalence of obesity in their daughters. In animals, paternal obesity led to offspring alterations on glucose-insulin homeostasis, hepatic lipogenesis, hypothalamus/feeding behavior, kidney of the offspring; it also impairs the reproductive potential of male offspring with sperm oxidative stress and mitochondrial dysfunction. An explanation for these observations (human and animal) is epigenetics, considered the primary tool for the transmission of phenotypes from the father to offspring, such as DNA methylation, histone modifications, and non-coding RNA. CONCLUSIONS Paternal obesity can induce programmed phenotypes in offspring through epigenetics. Therefore, it can be considered a public health problem, affecting the children's future life.
Collapse
Affiliation(s)
- Fernanda Ornellas
- Universidade do Estado do Rio de Janeiro (UERJ), Centro Biomédico, Laboratório de Morfometria, Metabolismo e Doenças Cardiovasculares, Rio de Janeiro, RJ, Brazil
| | - Priscila V Carapeto
- Universidade do Estado do Rio de Janeiro (UERJ), Centro Biomédico, Laboratório de Morfometria, Metabolismo e Doenças Cardiovasculares, Rio de Janeiro, RJ, Brazil
| | - Carlos A Mandarim-de-Lacerda
- Universidade do Estado do Rio de Janeiro (UERJ), Centro Biomédico, Laboratório de Morfometria, Metabolismo e Doenças Cardiovasculares, Rio de Janeiro, RJ, Brazil.
| | - Marcia B Aguila
- Universidade do Estado do Rio de Janeiro (UERJ), Centro Biomédico, Laboratório de Morfometria, Metabolismo e Doenças Cardiovasculares, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
27
|
Father’s obesity programs the adipose tissue in the offspring via the local renin–angiotensin system and MAPKs pathways, especially in adult male mice. Eur J Nutr 2017; 57:1901-1912. [DOI: 10.1007/s00394-017-1473-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 05/16/2017] [Indexed: 12/31/2022]
|
28
|
Singer K, Lumeng CN. The initiation of metabolic inflammation in childhood obesity. J Clin Invest 2017; 127:65-73. [PMID: 28045405 DOI: 10.1172/jci88882] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
An understanding of the events that initiate metabolic inflammation (metainflammation) can support the identification of targets for preventing metabolic disease and its negative effects on health. There is ample evidence demonstrating that the initiating events in obesity-induced inflammation start early in childhood. This has significant implications on our understanding of how early life events in childhood influence adult disease. In this Review we frame the initiating events of metainflammation in the context of child development and discuss what this reveals about the mechanisms by which this unique form of chronic inflammation is initiated and sustained into adulthood.
Collapse
|