1
|
Aguiar DD, Wunderlich ALM, Stopa LRS, Guergolette RP, Martins AB, Souza CF, da Silva ACV, Bissochi IMT, Forcato S, Aquino ABO, Zaia DAM, Zaia CTBV, de Andrade FG, Kiss ACI, Gerardin DCC, Uchôa ET, Leite CM. Postnatal undernutrition increases estradiol plasma levels and sexual receptivity and disrupts the kisspeptin-GnRH pathway in adult female rats. Physiol Behav 2025; 292:114817. [PMID: 39855583 DOI: 10.1016/j.physbeh.2025.114817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 01/14/2025] [Accepted: 01/21/2025] [Indexed: 01/27/2025]
Abstract
Undernutrition has increased worldwide in recent years and it is known that environmental factors to which individuals are exposed in early life can result in metabolic and reproductive changes that remain in adult life. In this context, the litter size expansion is a classic model used to induce undernutrition early in development. Thus, this study aimed to evaluate the effects of neonatal undernutrition induced by the litter size expansion on metabolic and reproductive parameters of female rats. At birth, litter size was adjusted to large (LL - 16 pups) and normal (NL - 10 pups) litters. After weaning, the feed was offered ad libitum and the animals were euthanized from postnatal day 90, when in proestrus. Neonatal undernutrition resulted in lower body weight from weaning to adulthood, although food intake remained higher in the LL group in this period. These animals exhibited a delayed onset of puberty, demonstrated by a late first estrus, increased values of circulating estradiol, luteinizing hormone, follicle-stimulating hormone, and number of antral follicles in the ovaries, associated with higher sexual receptivity, without differences in maternal behavior. The LL group also exhibited decreased messenger RNA (mRNA) expression of kisspeptin and gonadotropin-releasing hormone (GnRH) in the preoptic area, without changes in the mRNA expression of GnRH receptor in the pituitary. These results demonstrate that moderate undernutrition in the lactational period promotes metabolic changes associated with impairments in the kisspeptin-GnRH pathway, without compromising maternal behavior and peripheral reproductive functions such as estrous cyclicity, sexual receptivity, and fertility.
Collapse
Affiliation(s)
- Danielly D Aguiar
- Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, PR, Brazil
| | - Ana L M Wunderlich
- Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, PR, Brazil
| | - Larissa R S Stopa
- Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, PR, Brazil
| | - Rhauany P Guergolette
- Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, PR, Brazil
| | - Andressa B Martins
- Multicenter Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, PR, Brazil
| | - Camila F Souza
- Multicenter Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, PR, Brazil
| | - Ana C V da Silva
- Department of Physiological Sciences, State University of Londrina, Londrina, PR, Brazil
| | - Isabella M T Bissochi
- Department of Physiological Sciences, State University of Londrina, Londrina, PR, Brazil
| | - Simone Forcato
- Multicenter Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, PR, Brazil
| | - Ana B O Aquino
- Department of Physiological Sciences, State University of Londrina, Londrina, PR, Brazil
| | - Dimas A M Zaia
- Department of Chemistry, State University of Londrina, Londrina, PR, Brazil
| | - Cássia T B V Zaia
- Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, PR, Brazil; Multicenter Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, PR, Brazil; Department of Physiological Sciences, State University of Londrina, Londrina, PR, Brazil
| | - Fabio G de Andrade
- Department of Histology, State University of Londrina, Londrina, PR, Brazil
| | - Ana C I Kiss
- Department of Structural and Functional Biology, Biosciences Institute, São Paulo State University, Botucatu, SP, Brazil
| | - Daniela C C Gerardin
- Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, PR, Brazil; Multicenter Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, PR, Brazil; Department of Physiological Sciences, State University of Londrina, Londrina, PR, Brazil
| | - Ernane T Uchôa
- Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, PR, Brazil; Multicenter Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, PR, Brazil; Department of Physiological Sciences, State University of Londrina, Londrina, PR, Brazil.
| | | |
Collapse
|
2
|
Guergolette RP, Aguiar DD, Stopa LRS, Souza CF, Wunderlich ALM, Zaia DAM, Zaia CTBV, Leite CM, Uchoa ET. Early maternal undernutrition induces sex-related metabolic changes in adult offspring. J Dev Orig Health Dis 2025; 16:e4. [PMID: 39834058 DOI: 10.1017/s2040174424000448] [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] [Indexed: 01/22/2025]
Abstract
Nutritional status during the developmental periods leads to predisposition to several diseases and comorbidities, highlighting metabolic and reproductive changes throughout adult life, and in the next generations. One of the experimental models used to induce undernutrition is litter size expansion, which decreases the availability of breast milk to pups and delays development. This work evaluated the effects of maternal undernutrition induced by litter size expansion, a maternal undernutrition preconception model, on the metabolic and reproductive alterations of the offspring. For this, metabolic and reproductive parameters were evaluated in male and female offspring of female rats reared in normal (NL - 10 pups: 5 males and 5 females) and large (LL - 16 pups: 8 males and 8 females) litters. Male and female offspring of LL mothers presented higher food intake than the offspring of NL mothers. Male offspring from undernourished females showed reduced body weight from lactation to adulthood, nasoanal distance in childhood, increased nasoanal distance, and decreased Lee index in adult life, while female offspring showed decreased nasoanal distance in childhood. The male offspring from LL mothers showed increased insulin plasma levels and glucose tolerance, and reduced triglycerides plasma levels, without changes in the female offspring. These results indicate that neonatal undernutrition in females predisposes their male and female offspring to develop metabolic alterations, without reproductive repercussions, and male offspring seems to be more susceptible to present these metabolic changes than females. Thus, there are sexual differences in the metabolic responses of the offspring elicited by maternal preconceptional undernutrition.
Collapse
Affiliation(s)
- Rhauany P Guergolette
- Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, Parana, Brazil
| | - Danielly D Aguiar
- Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, Parana, Brazil
| | - Larissa R S Stopa
- Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, Parana, Brazil
| | - Camila F Souza
- Multicenter Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, Parana, Brazil
| | - Ana Luiza M Wunderlich
- Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, Parana, Brazil
| | - Dimas A M Zaia
- Department of Chemistry, State University of Londrina, Londrina, Parana, Brazil
| | - Cássia Thais B V Zaia
- Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, Parana, Brazil
- Multicenter Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, Parana, Brazil
- Department of Physiological Sciences, State University of Londrina, Londrina, Parana, Brazil
| | | | - Ernane T Uchoa
- Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, Parana, Brazil
- Multicenter Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, Parana, Brazil
- Department of Physiological Sciences, State University of Londrina, Londrina, Parana, Brazil
| |
Collapse
|
3
|
Fortunato-Silva J, de Rezende LP, Ferreira-Neto ML, Bispo-da-Silva LB, Balbi APC. Intrauterine exposure to a high-fat diet, with different levels of lipids, and its gastrointestinal repercussions: a model of fetal programming in rats. J Dev Orig Health Dis 2024; 15:e33. [PMID: 39711030 DOI: 10.1017/s2040174424000382] [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] [Indexed: 12/24/2024]
Abstract
It is known that adverse stimuli, such as altered diets during pregnancy and lactation, can result in deleterious effects on the progeny. The aim of this study was to evaluate the possible gastrointestinal repercussions in the offspring of Wistar rats exposed to high-fat diets. Pregnant rats were divided into three groups: normolipidic diet (3.5% lipids), a diet containing 28% lipids, and a diet with 40% lipids. Body weight and food, water, daily caloric, and macronutrient intake were evaluated in the pregnant rats. Structural and functional gastrointestinal parameters were assessed in 30-day-old male pups. Depending on the lipid content of the maternal diet, the pups may exhibit gastric mucosal thickening, an increase in the relative weight of the small intestine, a reduction in the jejunal and ileal mucosa, and a decrease in the total thickness of the ileum. Additionally, there may be a reduction in the number of villi per area in these organs and a thinning of the muscular layer in the large intestine. The structural changes induced by the maternal high-fat diet seem to reduce the stomach's sensitivity to ethanol-induced ulcers, which is the only functional alteration observed. Therefore, the offspring of dams exposed to high-fat diets during pregnancy and lactation exhibits impaired gastrointestinal development, with alterations depending on dietary fat content and specific gastrointestinal regions. Structural changes did not always result in functional abnormalities and, in some cases, appeared protective. The long-term consequences of the observed morphological alterations require further investigation.
Collapse
Affiliation(s)
- Jéssica Fortunato-Silva
- Program in Applied Structural and Cellular Biology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Lívia Prometti de Rezende
- Program in Applied Structural and Cellular Biology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Marcos Luiz Ferreira-Neto
- Department of Physiology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Luiz Borges Bispo-da-Silva
- Department of Pharmacology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| | - Ana Paula Coelho Balbi
- Department of Physiology, Institute of Biomedical Sciences, Federal University of Uberlândia, Uberlândia, Brazil
| |
Collapse
|
4
|
Torres DB, Lopes A, Rodrigues AJ, Ventura-Silva AP, Sousa N, Gontijo JAR, Boer PA, Lopes MG. Early morphological and neurochemical changes of the bed nucleus of stria terminalis (BNST) in gestational protein-restricted male offspring. Nutr Neurosci 2024; 27:1250-1268. [PMID: 38576309 DOI: 10.1080/1028415x.2024.2320498] [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] [Indexed: 04/06/2024]
Abstract
BACKGROUND The bed nucleus of the stria terminalis (BNST) is a structure with a peculiar neurochemical composition involved in modulating anxietylike behavior and fear. AIM The present study investigated the effects on the BNST neurochemical composition and neuronal structure in critical moments of the postnatal period in gestational protein-restricted male rats' offspring. METHODS Dams were maintained during the pregnancy on isocaloric rodent laboratory chow with standard protein content [NP, 17%] or low protein content [LP, 6%]. BNST from male NP and age-matched LP offspring was studied using the isotropic fractionator method, Neuronal 3D reconstruction, dendritic-tree analysis, blotting analysis, and high-performance liquid chromatography. RESULTS Serum corticosterone levels were higher in male LP offspring than NP rats in 14-day-old offspring, without any difference in 7-day-old progeny. The BNST total cell number and anterodorsal BNST division volume in LP progeny were significantly reduced on the 14th postnatal day compared with NP offspring. The BNST HPLC analysis from 7 days-old LP revealed increased norepinephrine levels compared to NP progeny. The BNST blot analysis from 7-day-old LP revealed reduced levels of GR and BDNF associated with enhanced CRF1 expression compared to NP offspring. 14-day-old LP offspring showed reduced expression of MR and 5HT1A associated with decreased DOPAC and DOPA turnover levels relative to NP rats. In Conclusion, the BNST cellular and neurochemical changes may represent adaptation during development in response to elevated fetal exposure to maternal corticosteroid levels. In this way, gestational malnutrition alters the BNST content and structure and contributes to already-known behavioral changes.
Collapse
Affiliation(s)
- D B Torres
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Internal Medicine Department, School of Medicine, State University of Campinas, Campinas, Brazil
| | - A Lopes
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Internal Medicine Department, School of Medicine, State University of Campinas, Campinas, Brazil
| | - A J Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - A P Ventura-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - N Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - J A R Gontijo
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Internal Medicine Department, School of Medicine, State University of Campinas, Campinas, Brazil
| | - P A Boer
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Internal Medicine Department, School of Medicine, State University of Campinas, Campinas, Brazil
| | | |
Collapse
|
5
|
Menezes ACF, Wunderlich ALM, Luiz KG, Frigoli GF, Costa IRD, Stopa LRDS, Souza CF, Guergolette RP, Shishido PK, Aquino ABO, Forcato S, Gerardin DCC, Zaia CTBV, Uchoa ET, Fernandes GSA. Neonatal undernutrition induced by litter size expansion alters testicular parameters in adult Wistar rats. Br J Nutr 2024; 132:862-873. [PMID: 39391932 DOI: 10.1017/s0007114524002149] [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] [Indexed: 10/12/2024]
Abstract
Several models of maternal undernutrition reveal impairment of testicular development and compromise spermatogenesis in male offspring. The expansion of the litter size model, valuable for studying the impact of undernutrition on early development, has not yet been used to evaluate the consequences of early undernutrition in the adult male reproductive system. For this purpose, pups were raised in either normal litter (ten pups/dam) or large litter (LL; sixteen pups/dam). On postnatal day 90, sexual behaviour was evaluated or blood, adipose and reproductive tissues were collected for biochemical, histological and morphological analysis. Adult LL animals were lighter and thinner than controls. They showed increased food intake, but decrease of retroperitoneal white adipose tissue weight, glycaemia after oral glucose overload and plasma concentration of cholesterol. Reproductive organ weights were not altered by undernutrition, but histopathological analysis revealed an increased number of abnormal seminiferous tubules and number of immature spermatids in the tubular lumen of LL animals. These animals also showed reduction in total spermatic reserve and daily sperm production in the testes. Undernutrition decreased the number of Sertoli cells, and testosterone production was increased in the LL group. Mitochondrial activity of spermatozoa remained unchanged between experimental groups, suggesting no significant impact on the energy-related processes associated with sperm function. All animals from both experimental groups were considered sexually competent, with no significant difference in the parameters of sexual behaviour. We conclude that neonatal undernutrition induces histological and physiological testicular changes, without altering sperm quality and sexual behaviour of animals.
Collapse
Affiliation(s)
- Ana Camila Ferreira Menezes
- State University of Londrina, Biological Sciences Centre, Department of General Biology, Laboratory of Toxicology and Metabolic Disorders of Reproduction, Londrina, Brazil
| | | | - Karen Gomes Luiz
- State University of Londrina, Biological Sciences Centre, Department of General Biology, Laboratory of Toxicology and Metabolic Disorders of Reproduction, Londrina, Brazil
| | - Giovanna Fachetti Frigoli
- State University of Londrina, Biological Sciences Centre, Department of General Biology, Laboratory of Toxicology and Metabolic Disorders of Reproduction, Londrina, Brazil
| | - Ivana Regina D Costa
- State University of Londrina, Biological Sciences Centre, Department of General Biology, Laboratory of Toxicology and Metabolic Disorders of Reproduction, Londrina, Brazil
| | | | - Camila Franciele Souza
- Multicentre Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, Brazil
| | | | - Polyana Keiko Shishido
- Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, Brazil
| | | | - Simone Forcato
- Multicentre Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, Brazil
| | - Daniela Cristina Ceccatto Gerardin
- Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, Brazil
- Multicentre Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, Brazil
- Department of Physiological Sciences, State University of Londrina, Londrina, Brazil
| | - Cássia Thaïs Bussamra Vieira Zaia
- Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, Brazil
- Multicentre Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, Brazil
- Department of Physiological Sciences, State University of Londrina, Londrina, Brazil
| | - Ernane Torres Uchoa
- Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, Brazil
- Multicentre Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, Brazil
- Department of Physiological Sciences, State University of Londrina, Londrina, Brazil
| | - Glaura Scantamburlo Alves Fernandes
- State University of Londrina, Biological Sciences Centre, Department of General Biology, Laboratory of Toxicology and Metabolic Disorders of Reproduction, Londrina, Brazil
- Postgraduate Program in Physiological Sciences, State University of Londrina, Londrina, Brazil
| |
Collapse
|
6
|
Ribeiro IT, Fioretto MN, Dos Santos SAA, Colombelli KT, Portela LMF, Niz Alvarez MV, de Magalhães Padilha P, Delgado AQ, Marques MVLSG, Bosqueiro JR, Seiva FRF, Barbisan LF, de Andrade Paes AM, Zambrano E, Justulin LA. Maternal protein restriction combined with postnatal sugar consumption alters liver proteomic profile and metabolic pathways in adult male offspring rats. Mol Cell Endocrinol 2024; 592:112316. [PMID: 38880278 DOI: 10.1016/j.mce.2024.112316] [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: 04/18/2024] [Revised: 06/13/2024] [Accepted: 06/14/2024] [Indexed: 06/18/2024]
Abstract
This study investigated the impact of maternal protein restriction (MPR) and early postnatal sugar consumption (SUG) on the liver health of adult male descendant rats. Male offspring of mothers fed a normal protein diet (NPD) or a low protein diet (LPD) were divided into four groups: Control (CTR), Sugar Control (CTR + SUG), LPD during gestation and lactation (GLLP), and LPD with sugar (GLLP + SUG). Sugar consumption (10% glucose diluted in water) began after weaning on day 21 (PND 21), and at 90 days (PND 90), rats were sacrificed for analysis. Sugar intake reduced food intake and increased water consumption in CTR + SUG and GLLP + SUG compared to CTR and GLLP. GLLP and GLLP + SUG groups showed lower body weight and total and retroperitoneal fat compared to CTR and CTR + SUG. CTR + SUG and GLLP + SUG groups exhibited hepatocyte vacuolization associated with increased hepatic glycogen content compared to CTR and GLLP. Hepatic catalase activity increased in GLLP compared to CTR. Proteomic analysis identified 223 differentially expressed proteins (DEPs) among experimental groups. While in the GLLP group, the DEPs enriched molecular pathways related to cellular stress, glycogen metabolic pathways were enriched in the GLLP + SUG and CTR + SUG groups. The association of sugar consumption amplifies the effects of MPR, deregulating molecular mechanisms related to metabolism and the antioxidant system.
Collapse
Affiliation(s)
- Isabelle Tenori Ribeiro
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil
| | - Matheus Naia Fioretto
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil
| | - Sérgio Alexandre Alcantara Dos Santos
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil; Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, USA
| | - Ketlin Thassiani Colombelli
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil
| | - Luiz Marcos Frediani Portela
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil
| | | | - Pedro de Magalhães Padilha
- Department of Chemical and Biological Sciences, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Aislan Quintiliano Delgado
- Department of Physical Education, Institute of Biosciences, Sao Paulo State University, Bauru, SP, Brazil
| | | | - José Roberto Bosqueiro
- Department of Physical Education, Institute of Biosciences, Sao Paulo State University, Bauru, SP, Brazil
| | - Fábio Rodrigues Ferreira Seiva
- Department of Chemical and Biological Sciences, Institute of Biosciences, Sao Paulo State University, Botucatu, SP, Brazil
| | - Luís Fernando Barbisan
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil
| | | | - Elena Zambrano
- Department Reproductive Biology, Salvador Zubirán National Institute of Medical Sciences and Nutrition, Mexico City, Mexico; Facultad de Química, Universidad Nacional Autónoma de Mexico, Mexico City, Mexico
| | - Luis Antonio Justulin
- UNESP- Sao Paulo State University, Department of Structural and Functional Biology, Institute of Biosciences, Botucatu, SP, Brazil.
| |
Collapse
|
7
|
Yuan R, Adlimoghaddam A, Zhu Y, Han X, Bartke A. Early Life Interventions: Impact on Aging and Longevity. Aging Dis 2024:AD.202.0516. [PMID: 39325935 DOI: 10.14336/ad.202.0516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 07/05/2024] [Indexed: 09/28/2024] Open
Abstract
Across mammals, lifespans vary remarkably, spanning over a hundredfold difference. Comparative studies consistently reveal a strong inverse relationship between developmental pace and lifespan, hinting at the potential for early-life interventions (ELIs) to influence aging and lifespan trajectories. Focusing on postnatal interventions in mice, this review explores how ELIs influence development, lifespan, and the underlying mechanisms. Previous ELI studies have employed a diverse array of approaches, including dietary modifications, manipulations of the somatotropic axis, and various chemical treatments. Notably, these interventions have demonstrated significant impacts on aging and lifespan in mice. The underlying mechanisms likely involve pathways related to mitochondrial function, mTOR and AMPK signaling, cellular senescence, and epigenetic alterations. Interestingly, ELI studies may serve as valuable models for investigating the complex regulatory mechanisms of development and aging, particularly regarding the interplay among somatic growth, sexual maturation, and lifespan. In addition, prior research has highlighted the intricacies of experimental design and data interpretation. Factors such as timing, sex-specific effects, administration methods, and animal husbandry practices must be carefully considered to ensure the reliability and reproducibility of results, as well as rigorous interpretation. Addressing these factors is essential for advancing our understanding of how development, aging, and lifespan are regulated, potentially opening avenues for interventions that promote healthy aging.
Collapse
Affiliation(s)
- Rong Yuan
- Division of Geriatrics Research, Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
| | - Aida Adlimoghaddam
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
- Department of Neurology, Center for Alzheimer's Research and Treatment, Neuroscience Institute, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
| | - Yun Zhu
- Division of Geriatrics Research, Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
| | - Xiuqi Han
- Division of Geriatrics Research, Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
- Department of Medical Microbiology, Immunology and Cell Biology, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
| | - Andrzej Bartke
- Division of Geriatrics Research, Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62702, USA
| |
Collapse
|
8
|
Ghasemi Z, Alizadeh Mogadam Masouleh A, Rashki Ghaleno L, Akbarinejad V, Rezazadeh Valojerdi M, Shahverdi A. Maternal nutrition and fetal imprinting of the male progeny. Anim Reprod Sci 2024; 265:107470. [PMID: 38657462 DOI: 10.1016/j.anireprosci.2024.107470] [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: 12/28/2023] [Revised: 03/28/2024] [Accepted: 03/30/2024] [Indexed: 04/26/2024]
Abstract
The global population as well as the demand for human food is rapidly growing worldwide, which necessitates improvement of efficiency in livestock operations. In this context, environmental factors during fetal and/or neonatal life have been observed to influence normal physical and physiological function of an individual during adulthood, and this phenomenon is called fetal or developmental programming. While numerous studies have reported the impact of maternal factors on development of the female progeny, limited information is available on the potential effects of fetal programming on reproductive function of the male offspring. Therefore, the objective for this review article was to focus on available literature regarding the impact of maternal factors, particularly maternal nutrition, on reproductive system of the male offspring. To this end, we highlighted developmental programming of the male offspring in domestic species (i.e., pig, cow and sheep) as well as laboratory species (i.e., mice and rat) during pregnancy and lactation. In this sense, we pointed out the effects of maternal nutrition on various functions of the male offspring including hypothalamic-pituitary axis, hormonal levels, testicular tissue and semen parameters.
Collapse
Affiliation(s)
- Zahrasadat Ghasemi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran; Animal Core Facility, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - AliReza Alizadeh Mogadam Masouleh
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran; Gyn-medicum, Center for Reproductive Medicine, Göttingen, Germany; Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany.
| | - Leila Rashki Ghaleno
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| | - Vahid Akbarinejad
- Department of Theriogenology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Mojtaba Rezazadeh Valojerdi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran; Department of Anatomy, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Abdolhossein Shahverdi
- Department of Embryology, Reproductive Biomedicine Research Center, Royan Institute for Reproductive Biomedicine, ACECR, Tehran, Iran
| |
Collapse
|
9
|
Saavedra LPJ, Piovan S, Moreira VM, Gonçalves GD, Ferreira ARO, Ribeiro MVG, Peres MNC, Almeida DL, Raposo SR, da Silva MC, Barbosa LF, de Freitas Mathias PC. Epigenetic programming for obesity and noncommunicable disease: From womb to tomb. Rev Endocr Metab Disord 2024; 25:309-324. [PMID: 38040983 DOI: 10.1007/s11154-023-09854-w] [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] [Accepted: 11/15/2023] [Indexed: 12/03/2023]
Abstract
Several epidemiological, clinical and experimental studies in recent decades have shown the relationship between exposure to stressors during development and health outcomes later in life. The characterization of these susceptible phases, such as preconception, gestation, lactation and adolescence, and the understanding of factors that influence the risk of an adult individual for developing obesity, metabolic and cardiovascular diseases, is the focus of the DOHaD (Developmental Origins of Health and Disease) research line. In this sense, advancements in molecular biology techniques have contributed significantly to the understanding of the mechanisms underlying the observed phenotypes, their morphological and physiological alterations, having as a main driving factor the epigenetic modifications and their consequent modulation of gene expression. The present narrative review aimed to characterize the different susceptible phases of development and associated epigenetic modifications, and their implication in the development of non-communicable diseases. Additionally, we provide useful insights into interventions during development to counteract or prevent long-term programming for disease susceptibility.
Collapse
Affiliation(s)
- Lucas Paulo Jacinto Saavedra
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Silvano Piovan
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Veridiana Mota Moreira
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Gessica Dutra Gonçalves
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Anna Rebeka Oliveira Ferreira
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Maiara Vanusa Guedes Ribeiro
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Maria Natália Chimirri Peres
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Douglas Lopes Almeida
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Scarlett Rodrigues Raposo
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Mariane Carneiro da Silva
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Letícia Ferreira Barbosa
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil
| | - Paulo Cezar de Freitas Mathias
- Department of Biotechnology, Genetics, and Cellular Biology, State University of Maringá, 5790 Av Colombo, Sala 19, Maringá, PR, 87020-900, Brazil.
| |
Collapse
|
10
|
Skowronski AA, Leibel RL, LeDuc CA. Neurodevelopmental Programming of Adiposity: Contributions to Obesity Risk. Endocr Rev 2024; 45:253-280. [PMID: 37971140 PMCID: PMC10911958 DOI: 10.1210/endrev/bnad031] [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: 06/07/2023] [Revised: 09/29/2023] [Accepted: 10/19/2023] [Indexed: 11/19/2023]
Abstract
This review analyzes the published evidence regarding maternal factors that influence the developmental programming of long-term adiposity in humans and animals via the central nervous system (CNS). We describe the physiological outcomes of perinatal underfeeding and overfeeding and explore potential mechanisms that may mediate the impact of such exposures on the development of feeding circuits within the CNS-including the influences of metabolic hormones and epigenetic changes. The perinatal environment, reflective of maternal nutritional status, contributes to the programming of offspring adiposity. The in utero and early postnatal periods represent critically sensitive developmental windows during which the hormonal and metabolic milieu affects the maturation of the hypothalamus. Maternal hyperglycemia is associated with increased transfer of glucose to the fetus driving fetal hyperinsulinemia. Elevated fetal insulin causes increased adiposity and consequently higher fetal circulating leptin concentration. Mechanistic studies in animal models indicate important roles of leptin and insulin in central and peripheral programming of adiposity, and suggest that optimal concentrations of these hormones are critical during early life. Additionally, the environmental milieu during development may be conveyed to progeny through epigenetic marks and these can potentially be vertically transmitted to subsequent generations. Thus, nutritional and metabolic/endocrine signals during perinatal development can have lifelong (and possibly multigenerational) impacts on offspring body weight regulation.
Collapse
Affiliation(s)
- Alicja A Skowronski
- Division of Molecular Genetics, Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Rudolph L Leibel
- Division of Molecular Genetics, Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Charles A LeDuc
- Division of Molecular Genetics, Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
11
|
Bokor S, Csölle I, Felső R, Vass RA, Funke S, Ertl T, Molnár D. Dietary nutrients during gestation cause obesity and related metabolic changes by altering DNA methylation in the offspring. Front Endocrinol (Lausanne) 2024; 15:1287255. [PMID: 38449848 PMCID: PMC10916691 DOI: 10.3389/fendo.2024.1287255] [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: 09/01/2023] [Accepted: 02/01/2024] [Indexed: 03/08/2024] Open
Abstract
Growing evidence shows that maternal nutrition from preconception until lactation has an important effect on the development of non-communicable diseases in the offspring. Biological responses to environmental stress during pregnancy, including undernutrition or overnutrition of various nutrients, are transmitted in part by DNA methylation. The aim of the present narrative review is to summarize literature data on altered DNA methylation patterns caused by maternal macronutrient or vitamin intake and its association with offspring's phenotype (obesity and related metabolic changes). With our literature search, we found evidence for the association between alterations in DNA methylation pattern of different genes caused by maternal under- or overnutrition of several nutrients (protein, fructose, fat, vitamin D, methyl-group donor nutrients) during 3 critical periods of programming (preconception, pregnancy, lactation) and the development of obesity or related metabolic changes (glucose, insulin, lipid, leptin, adiponectin levels, blood pressure, non-alcoholic fatty liver disease) in offspring. The review highlights that maternal consumption of several nutrients could individually affect the development of offspring's obesity and related metabolic changes via alterations in DNA methylation.
Collapse
Affiliation(s)
- Szilvia Bokor
- Department of Paediatrics, Medical School, University of Pécs, Pécs, Hungary
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
| | - Ildikó Csölle
- Department of Paediatrics, Medical School, University of Pécs, Pécs, Hungary
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
- Doctoral School of Health Sciences, Faculty of Health Sciences, University of Pécs, Pécs, Hungary
| | - Regina Felső
- Department of Paediatrics, Medical School, University of Pécs, Pécs, Hungary
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
| | - Réka A. Vass
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
- Department of Obstetrics and Gynaecology, Medical School, University of Pécs, Pécs, Hungary
- Obstetrics and Gynecology, Magyar Imre Hospital Ajka, Ajka, Hungary
| | - Simone Funke
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
- Department of Obstetrics and Gynaecology, Medical School, University of Pécs, Pécs, Hungary
| | - Tibor Ertl
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
- Department of Obstetrics and Gynaecology, Medical School, University of Pécs, Pécs, Hungary
| | - Dénes Molnár
- Department of Paediatrics, Medical School, University of Pécs, Pécs, Hungary
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
| |
Collapse
|
12
|
Camargo AC, Constantino FB, Santos SA, Colombelli KT, Portela LM, Fioretto MN, Barata LA, Valente GT, Moreno CS, Justulin LA. Deregulation of ABCG1 early in life contributes to prostate carcinogenesis in maternally malnourished offspring rats. Mol Cell Endocrinol 2024; 580:112102. [PMID: 37972683 DOI: 10.1016/j.mce.2023.112102] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Revised: 10/12/2023] [Accepted: 10/24/2023] [Indexed: 11/19/2023]
Abstract
AIMS The developmental Origins of Health and Disease (DOHaD) concept has provided the framework to assess how early life experiences can shape health and disease throughout the life course. Using a model of maternal exposure to a low protein diet (LPD; 6% protein) during the gestational and lactational periods, we demonstrated changes in the ventral prostate (VP) transcriptomic landscape in young rats exposed to maternal malnutrition. Male offspring Sprague Dawley rats were submitted to maternal malnutrition during gestation and lactation, and they were weighed, and distance anogenital was measured, followed were euthanized by an overdose of anesthesia at 21 postnatal days. Next, the blood and the ventral prostate (VP) were collected and processed by morphological analysis, biochemical and molecular analyses. RNA-seq analysis identified 411 differentially expressed genes (DEGs) in the VP of maternally malnourished offspring compared to the control group. The molecular pathways enriched by these DEGs are related to cellular development, differentiation, and tissue morphogenesis, all of them involved in both normal prostate development and carcinogenesis. Abcg1 was commonly deregulated in young and old maternally malnourished offspring rats, as well in rodent models of prostate cancer (PCa) and in PCa patients. Our results described ABCG1 as a potential DOHaD gene associated with perturbation of prostate developmental biology with long-lasting effects on carcinogenesis in old offspring rats. A better understanding of these mechanisms may help with the discussion of preventive strategies against early life origins of non-communicable chronic diseases.
Collapse
Affiliation(s)
- Ana Cl Camargo
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, SP, Brazil; Laboratory of Human Genetics, Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Flávia B Constantino
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Sergio Aa Santos
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, SP, Brazil; Cancer Signaling and Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Ketlin T Colombelli
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Luiz Mf Portela
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Matheus N Fioretto
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Luísa A Barata
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Guilherme T Valente
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, SP, Brazil
| | - Carlos S Moreno
- Department of Pathology and Laboratory Medicine and Department of Biomedical Informatics, Emory University School of Medicine, USA
| | - Luis A Justulin
- Department of Structural and Functional Biology, Institute of Biosciences, Sao Paulo State University (UNESP), Botucatu, SP, Brazil.
| |
Collapse
|
13
|
Di Gesù CM, Buffington SA. The early life exposome and autism risk: a role for the maternal microbiome? Gut Microbes 2024; 16:2385117. [PMID: 39120056 PMCID: PMC11318715 DOI: 10.1080/19490976.2024.2385117] [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: 04/01/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 08/10/2024] Open
Abstract
Autism spectrum disorders (ASD) are highly heritable, heterogeneous neurodevelopmental disorders characterized by clinical presentation of atypical social, communicative, and repetitive behaviors. Over the past 25 years, hundreds of ASD risk genes have been identified. Many converge on key molecular pathways, from translational control to those regulating synaptic structure and function. Despite these advances, therapeutic approaches remain elusive. Emerging data unearthing the relationship between genetics, microbes, and immunity in ASD suggest an integrative physiology approach could be paramount to delivering therapeutic breakthroughs. Indeed, the advent of large-scale multi-OMIC data acquisition, analysis, and interpretation is yielding an increasingly mechanistic understanding of ASD and underlying risk factors, revealing how genetic susceptibility interacts with microbial genetics, metabolism, epigenetic (re)programming, and immunity to influence neurodevelopment and behavioral outcomes. It is now possible to foresee exciting advancements in the treatment of some forms of ASD that could markedly improve quality of life and productivity for autistic individuals. Here, we highlight recent work revealing how gene X maternal exposome interactions influence risk for ASD, with emphasis on the intrauterine environment and fetal neurodevelopment, host-microbe interactions, and the evolving therapeutic landscape for ASD.
Collapse
Affiliation(s)
- Claudia M. Di Gesù
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
| | - Shelly A. Buffington
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
14
|
Torres DB, Lopes A, Rodrigues AJ, Lopes MG, Ventura-Silva AP, Sousa N, Gontijo JAR, Boer PA. Gestational protein restriction alters early amygdala neurochemistry in male offspring. Nutr Neurosci 2023; 26:1103-1119. [PMID: 36331123 DOI: 10.1080/1028415x.2022.2131064] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND Gestational protein intake restriction-induced long-lasting harmful outcomes in the offspring's organs and systems. However, few studies have focused on this event's impact on the brain's structures and neurochemical compounds. AIM The present study investigated the effects on the amygdala neurochemical composition and neuronal structure in gestational protein-restricted male rats' offspring. METHODS Dams were maintained on isocaloric standard rodent laboratory chow with regular protein [NP, 17%] or low protein content [LP, 6%]. Total cells were quantified using the Isotropic fractionator method, Neuronal 3D reconstruction, and dendritic tree analysis using the Golgi-Cox technique. Western blot and high-performance liquid chromatography performed neurochemical studies. RESULTS The gestational low-protein feeding offspring showed a significant decrease in birth weight up to day 14, associated with unaltered brain weight in youth or adult progenies. The amygdala cell numbers were unchanged, and the dendrites length and dendritic ramifications 3D analysis in LP compared to age-matched NP progeny. However, the current study shows reduced amygdala content of norepinephrine, epinephrine, and dopamine in LP progeny. These offspring observed a significant reduction in the amygdala glucocorticoid (GR) and mineralocorticoid (MR) receptor protein levels. Also corticotrophin-releasing factor (CRF) amygdala protein content was reduced in 7 and 14-day-old LP rats. CONCLUSION The observed amygdala neurochemical changes may represent adaptation during embryonic development in response to elevated fetal exposure to maternal corticosteroid levels. In this way, gestational malnutrition stress can alter the amygdala's neurochemical content and may contribute to known behavioral changes induced by gestational protein restriction.
Collapse
Affiliation(s)
- Daniele B Torres
- Fetal Programming and Hydro-electrolyte Metabolism Laboratory, Internal Medicine Department, School of Medicine, State University of Campinas, Campinas, SP, Brazil
| | - Agnes Lopes
- Fetal Programming and Hydro-electrolyte Metabolism Laboratory, Internal Medicine Department, School of Medicine, State University of Campinas, Campinas, SP, Brazil
| | - Ana J Rodrigues
- Fetal Programming and Hydro-electrolyte Metabolism Laboratory, Internal Medicine Department, School of Medicine, State University of Campinas, Campinas, SP, Brazil
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Marcelo G Lopes
- Fetal Programming and Hydro-electrolyte Metabolism Laboratory, Internal Medicine Department, School of Medicine, State University of Campinas, Campinas, SP, Brazil
| | - Ana P Ventura-Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - José A R Gontijo
- Fetal Programming and Hydro-electrolyte Metabolism Laboratory, Internal Medicine Department, School of Medicine, State University of Campinas, Campinas, SP, Brazil
| | - Patricia A Boer
- Fetal Programming and Hydro-electrolyte Metabolism Laboratory, Internal Medicine Department, School of Medicine, State University of Campinas, Campinas, SP, Brazil
| |
Collapse
|
15
|
de Oliveira-Silva J, Lisboa PC, Lotufo-Denucci B, Fraga M, de Moura EG, Nunes FC, Ribeiro-Carvalho A, Filgueiras CC, Abreu-Villaça Y, Manhães AC. Maternal protein restriction during the lactation period disrupts the ontogenetic development of behavioral traits in male Wistar rat offspring. J Dev Orig Health Dis 2023:1-12. [PMID: 37185045 DOI: 10.1017/s2040174423000107] [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: 05/17/2023]
Abstract
Neonatal undernutrition in rats results in short- and long-term behavioral and hormonal alterations in the offspring. It is not clear, however, whether these effects are present since the original insult or if they develop at some specific age later in life. Here, we assessed the ontogenetic profile of behavioral parameters associated with anxiety, exploration and memory/learning of Wistar rat offspring that were subjected to protein malnutrition during lactation. Dams and respective litters were separated into two groups: (1) protein-restricted (PR), which received a hypoproteic chow (8% protein) from birth to weaning [postnatal day (PN) 21]; (2) control (C), which received normoproteic chow. Offspring's behaviors, corticosterone, catecholamines, T3 and T4 levels were assessed at PN21 (weaning), PN45 (adolescence), PN90 (young adulthood) or PN180 (adulthood). PR offspring showed an age-independent reduction in the levels of anxiety-like behaviors in the Elevated Plus Maze and better memory performance in the Radial Arm Water Maze. PR offspring showed peak exploratory activity in the Open Field earlier in life, at PN45, than C, which showed theirs at PN90. Corticosterone was reduced in PR offspring, particularly at young adulthood, while catecholamines were increased at weaning and adulthood. The current study shows that considerable age-dependent variations in the expression of the observed behaviors and hormonal levels exist from weaning to adulthood in rats, and that protein restriction during lactation has complex variable-dependent effects on the ontogenesis of the assessed parameters.
Collapse
Affiliation(s)
- Juliana de Oliveira-Silva
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Centro Biomédico, Universidade do Estado do Rio de Janeiro, Av. Prof. Manoel de Abreu 444, 5 andar - Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Patrícia C Lisboa
- Laboratório de Fisiologia Endócrina, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Centro Biomédico, Universidade do Estado do Rio de Janeiro, Av. Prof. Manoel de Abreu 444, 5 andar - Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Bruna Lotufo-Denucci
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Centro Biomédico, Universidade do Estado do Rio de Janeiro, Av. Prof. Manoel de Abreu 444, 5 andar - Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Mabel Fraga
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Centro Biomédico, Universidade do Estado do Rio de Janeiro, Av. Prof. Manoel de Abreu 444, 5 andar - Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Egberto G de Moura
- Laboratório de Fisiologia Endócrina, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Centro Biomédico, Universidade do Estado do Rio de Janeiro, Av. Prof. Manoel de Abreu 444, 5 andar - Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Fernanda C Nunes
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Centro Biomédico, Universidade do Estado do Rio de Janeiro, Av. Prof. Manoel de Abreu 444, 5 andar - Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Anderson Ribeiro-Carvalho
- Departamento de Ciências, Faculdade de Formação de Professores da Universidade do Estado do Rio de Janeiro, Rua Dr. Francisco Portela 1470 - Patronato, São Gonçalo, RJ, 24435-005, Brazil
| | - Cláudio C Filgueiras
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Centro Biomédico, Universidade do Estado do Rio de Janeiro, Av. Prof. Manoel de Abreu 444, 5 andar - Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Yael Abreu-Villaça
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Centro Biomédico, Universidade do Estado do Rio de Janeiro, Av. Prof. Manoel de Abreu 444, 5 andar - Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| | - Alex C Manhães
- Laboratório de Neurofisiologia, Departamento de Ciências Fisiológicas, Instituto de Biologia Roberto Alcantara Gomes, Centro Biomédico, Universidade do Estado do Rio de Janeiro, Av. Prof. Manoel de Abreu 444, 5 andar - Vila Isabel, Rio de Janeiro, RJ, 20550-170, Brazil
| |
Collapse
|
16
|
Sosa-Larios TC, Ortega-Márquez AL, Rodríguez-Aguilera JR, Vázquez-Martínez ER, Domínguez-López A, Morimoto S. A low-protein maternal diet during gestation affects the expression of key pancreatic β-cell genes and the methylation status of the regulatory region of the MafA gene in the offspring of Wistar rats. Front Vet Sci 2023; 10:1138564. [PMID: 36992977 PMCID: PMC10040775 DOI: 10.3389/fvets.2023.1138564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 02/21/2023] [Indexed: 03/16/2023] Open
Abstract
Maternal nutrition during gestation has important effects on gene expression-mediated metabolic programming in offspring. To evaluate the effect of a protein-restricted maternal diet during gestation, pancreatic islets from male progeny of Wistar rats were studied at postnatal days (PND) 36 (juveniles) and 90 (young adults). The expression of key genes involved in β-cell function and the DNA methylation pattern of the regulatory regions of two such genes, Pdx1 (pancreatic and duodenal homeobox 1) and MafA (musculoaponeurotic fibrosarcoma oncogene family, protein A), were investigated. Gene expression analysis in the pancreatic islets of restricted offspring showed significant differences compared with the control group at PND 36 (P < 0.05). The insulin 1 and 2 (Ins1 and Ins2), Glut2 (glucose transporter 2), Pdx1, MafA, and Atf2 (activating transcription factor 2), genes were upregulated, while glucokinase (Gck) and NeuroD1 (neuronal differentiation 1) were downregulated. Additionally, we studied whether the gene expression differences in Pdx1 and MafA between control and restricted offspring were associated with differential DNA methylation status in their regulatory regions. A decrease in the DNA methylation levels was found in the 5' flanking region between nucleotides −8118 to −7750 of the MafA regulatory region in restricted offspring compared with control pancreatic islets. In conclusion, low protein availability during gestation causes the upregulation of MafA gene expression in pancreatic β-cells in the male juvenile offspring at least in part through DNA hypomethylation. This process may contribute to developmental dysregulation of β-cell function and influence the long-term health of the offspring.
Collapse
Affiliation(s)
- Tonantzin C. Sosa-Larios
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición “Salvador Zubirán”, Mexico City, Mexico
| | - Ana L. Ortega-Márquez
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición “Salvador Zubirán”, Mexico City, Mexico
| | - Jesús R. Rodríguez-Aguilera
- Departamento de Biología Celular y Desarrollo, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Edgar R. Vázquez-Martínez
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Aaron Domínguez-López
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Sumiko Morimoto
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición “Salvador Zubirán”, Mexico City, Mexico
- *Correspondence: Sumiko Morimoto
| |
Collapse
|
17
|
Prenatal exposure to famine and the development of diabetes later in life: an age-period-cohort analysis of the China health and nutrition survey (CHNS) from 1997 to 2015. Eur J Nutr 2023; 62:941-950. [PMID: 36326864 DOI: 10.1007/s00394-022-03049-w] [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: 04/25/2022] [Accepted: 10/25/2022] [Indexed: 11/05/2022]
Abstract
PURPOSE Prenatal exposure to famine has been linked to increased diabetes risk in adulthood. However, one fundamental issue to be addressed is that the reported famine-diabetes relation may be confounded by the age differences between the exposed and non-exposed groups. We aimed to determine the association between prenatal exposure to the Chinese famine of 1959-1962 and risk of diabetes by applying age well-controlled strategies. METHODS Among 20,535 individuals born in 1955-1966 who participated in the China Health and Nutrition Survey from 1997 to 2015, we constructed age-matched exposed vs. non-exposed groups to investigate the role of prenatal exposure to the Chinese famine of 1959-1962 in relation to diabetes. We also built a hierarchical age-period-cohort (HAPC) model to specifically examine the relation of famine to diabetes risk independent of age. RESULTS Compared to the age-balanced men in the non-exposed group, the exposed men born in 1961 had a 154% increased risk of diabetes [odds ratio (OR) 2.54 (95% CI 1.07-6.03), P = 0.04). In the HAPC analysis, the predicted probabilities of diabetes peaked in the 1961-birth cohort of men [3.4% (95% CI 2.4%-5.0%)], as compared to the average probability of diabetes (reference) of 1.8% for men overall. Neither analytical strategy revealed any strong relation between famine exposure and diabetes risk in women. CONCLUSION Among the pre-defined Chinese famine period of 1959-1962, early-life exposure to famine was associated with increased diabetes risk in men but not in women, and these relations were independent of age.
Collapse
|
18
|
Mohammed S, Qadri SSYH, Molangiri A, Basak S, Rajkumar H. Gestational low dietary protein induces intrauterine inflammation and alters the programming of adiposity & insulin sensitivity in the adult offspring. J Nutr Biochem 2023; 116:109330. [PMID: 36967094 DOI: 10.1016/j.jnutbio.2023.109330] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 01/31/2023] [Accepted: 03/18/2023] [Indexed: 04/08/2023]
Abstract
Malnutrition associated with low dietary protein can induce gestational inflammation and sets a long-lasting metabolic impact on the offspring even after replenishment. The work investigated whether a low-protein diet (LPD) during pregnancy and lactation induces intrauterine inflammation and predisposes offspring to adiposity and insulin resistance in their adult life. Female Golden Syrian hamsters were fed LPD (10.0% energy from protein) or a control diet (CD, 20.0 % energy from protein) from preconception until lactation. All pups were switched to CD after lactation and continued until the end. Maternal LPD increased intrauterine inflammation by enhancing neutrophil infiltration, amniotic hsCRP, oxidative stress, and mRNA expression of NFκβ, IL8, COX2, and TGFβ in the chorioamniotic membrane (P<.05). The prepregnancy body weight, placental, and fetal weights, serum AST and ALT were decreased, while blood platelets, lymphocytes, insulin, and HDL were significantly increased in LPD-fed dams (P<.05). A postnatal switch to an adequate protein could not prevent hyperlipidemia in the 6-months LPD/CD offspring. The lipid profile and liver functions were restored over 10 months of protein feeding but failed to normalize fasting glucose and body fat accumulation compared to CD/CD. LPD/CD showed elevated GLUT4 expression & activated pIRS1 in the skeletal muscle and increased expression of IL6, IL1β, and p65-NFκB proteins in the liver (P<.05). In conclusion, present data suggest that maternal protein restriction may induce intrauterine inflammation and affect liver inflammation in the adult offspring by an influx of fats from adipose that may alter lipid metabolism and reduce insulin sensitivity in skeletal muscle.
Collapse
|
19
|
Tajonar K, Gonzalez-Ronquillo M, Relling A, Nordquist RE, Nawroth C, Vargas-Bello-Pérez E. Toward assessing the role of dietary fatty acids in lamb's neurological and cognitive development. Front Vet Sci 2023; 10:1081141. [PMID: 36865439 PMCID: PMC9971820 DOI: 10.3389/fvets.2023.1081141] [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: 10/26/2022] [Accepted: 01/13/2023] [Indexed: 02/16/2023] Open
Abstract
Understanding and measuring sheep cognition and behavior can provide us with measures to safeguard the welfare of these animals in production systems. Optimal neurological and cognitive development of lambs is important to equip individuals with the ability to better cope with environmental stressors. However, this development can be affected by nutrition with a special role from long-chain fatty acid supply from the dam to the fetus or in lamb's early life. Neurological development in lambs takes place primarily during the first two trimesters of gestation. Through late fetal and early postnatal life, the lamb brain has a high level of cholesterol synthesis. This rate declines rapidly at weaning and remains low throughout adulthood. The main polyunsaturated fatty acids (PUFA) in the brain are ω-6 arachidonic acid and ω-3 docosahexaenoic acid (DHA), which are elements of plasma membranes' phospholipids in neuronal cells. DHA is essential for keeping membrane integrity and is vital for normal development of the central nervous system (CNS), and its insufficiency can damage cerebral functions and the development of cognitive capacities. In sheep, there is evidence that supplying PUFA during gestation or after birth may be beneficial to lamb productive performance and expression of species-specific behaviors. The objective of this perspective is to discuss concepts of ruminant behavior and nutrition and reflect on future research directions that could help to improve our knowledge on how dietary fatty acids (FA) relate to optimal neurological and cognitive development in sheep.
Collapse
Affiliation(s)
- Karen Tajonar
- Departamento de Medicina y Zootecnia de Rumiantes, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Mexico City, Mexico,Department of Animal Sciences, School of Agriculture, Policy and Development, University of Reading, Reading, United Kingdom
| | - Manuel Gonzalez-Ronquillo
- Facultad de Medicina Veterinaria y Zootecnia, Universidad Autónoma del Estado de México, Toluca, Mexico
| | - Alejandro Relling
- Department of Animal Science, College of Food, Agricultural, and Environmental Sciences, The Ohio State University, Columbus, OH, United States
| | - Rebecca E. Nordquist
- Unit Animals in Science and Society, Department Population Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Christian Nawroth
- Institute of Behavioural Physiology, Research Institute for Farm Animal Biology (FBN), Dummerstorf, Germany,*Correspondence: Christian Nawroth ✉
| | - Einar Vargas-Bello-Pérez
- Department of Animal Sciences, School of Agriculture, Policy and Development, University of Reading, Reading, United Kingdom,Einar Vargas-Bello-Pérez ✉
| |
Collapse
|
20
|
Developmental Programming in Animal Models: Critical Evidence of Current Environmental Negative Changes. Reprod Sci 2023; 30:442-463. [PMID: 35697921 PMCID: PMC9191883 DOI: 10.1007/s43032-022-00999-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 06/02/2022] [Indexed: 11/23/2022]
Abstract
The Developmental Origins of Health and Disease (DOHaD) approach answers questions surrounding the early events suffered by the mother during reproductive stages that can either partially or permanently influence the developmental programming of children, predisposing them to be either healthy or exhibit negative health outcomes in adulthood. Globally, vulnerable populations tend to present high obesity rates, including among school-age children and women of reproductive age. In addition, adults suffer from high rates of diabetes, hypertension, cardiovascular, and other metabolic diseases. The increase in metabolic outcomes has been associated with the combination of maternal womb conditions and adult lifestyle-related factors such as malnutrition and obesity, smoking habits, and alcoholism. However, to date, "new environmental changes" have recently been considered negative factors of development, such as maternal sedentary lifestyle, lack of maternal attachment during lactation, overcrowding, smog, overurbanization, industrialization, noise pollution, and psychosocial stress experienced during the current SARS-CoV-2 pandemic. Therefore, it is important to recognize how all these factors impact offspring development during pregnancy and lactation, a period in which the subject cannot protect itself from these mechanisms. This review aims to introduce the importance of studying DOHaD, discuss classical programming studies, and address the importance of studying new emerging programming mechanisms, known as actual lifestyle factors, during pregnancy and lactation.
Collapse
|
21
|
Blasetti A, Quarta A, Guarino M, Cicolini I, Iannucci D, Giannini C, Chiarelli F. Role of Prenatal Nutrition in the Development of Insulin Resistance in Children. Nutrients 2022; 15:nu15010087. [PMID: 36615744 PMCID: PMC9824240 DOI: 10.3390/nu15010087] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/13/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022] Open
Abstract
Nutrition during the prenatal period is crucial for the development of insulin resistance (IR) and its consequences in children. The relationship between intrauterine environment, fetal nutrition and the onset of IR, type 2 diabetes (T2D), obesity and metabolic syndrome later in life has been confirmed in many studies. The intake of carbohydrates, protein, fat and micronutrients during pregnancy seems to damage fetal metabolism programming; indeed, epigenetic mechanisms change glucose-insulin metabolism. Intrauterine growth restriction (IUGR) induced by unbalanced nutrient intake during prenatal life cause fetal adipose tissue and pancreatic beta-cell dysfunction. In this review we have summarized and discussed the role of maternal nutrition in preventing insulin resistance in youth.
Collapse
|
22
|
Folguieri MS, Franco ATB, Vieira AS, Gontijo JAR, Boer PA. Transcriptome and morphological analysis on the heart in gestational protein-restricted aging male rat offspring. Front Cell Dev Biol 2022; 10:892322. [PMID: 36353510 PMCID: PMC9638007 DOI: 10.3389/fcell.2022.892322] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 10/10/2022] [Indexed: 09/08/2024] Open
Abstract
Background: Adverse factors that influence embryo/fetal development are correlated with increased risk of cardiovascular disease (CVD), type-2 diabetes, arterial hypertension, obesity, insulin resistance, impaired kidney development, psychiatric disorders, and enhanced susceptibility to oxidative stress and inflammatory processes in adulthood. Human and experimental studies have demonstrated a reciprocal relationship between birthweight and cardiovascular diseases, implying intrauterine adverse events in the onset of these abnormalities. In this way, it is plausible that confirmed functional and morphological heart changes caused by gestational protein restriction could be related to epigenetic effects anticipating cardiovascular disorders and reducing the survival time of these animals. Methods: Wistar rats were divided into two groups according to the protein diet content offered during the pregnancy: a normal protein diet (NP, 17%) or a Low-protein diet (LP, 6%). The arterial pressure was measured, and the cardiac mass, cardiomyocytes area, gene expression, collagen content, and immunostaining of proteins were performed in the cardiac tissue of male 62-weeks old NP compared to LP offspring. Results: In the current study, we showed a low birthweight followed by catch-up growth phenomena associated with high blood pressure development, increased heart collagen content, and cardiomyocyte area in 62-week-old LP offspring. mRNA sequencing analysis identified changes in the expression level of 137 genes, considering genes with a p-value < 0.05. No gene was. Significantly changed according to the adj-p-value. After gene-to-gene biological evaluation and relevance, the study demonstrated significant differences in genes linked to inflammatory activity, oxidative stress, apoptosis process, autophagy, hypertrophy, and fibrosis pathways resulting in heart function disorders. Conclusion: The present study suggests that gestational protein restriction leads to early cardiac diseases in the LP progeny. It is hypothesized that heart dysfunction is associated with fibrosis, myocyte hypertrophy, and multiple abnormal gene expression. Considering the above findings, it may suppose a close link between maternal protein restriction, specific gene expression, and progressive heart failure.
Collapse
Affiliation(s)
- Marina S. Folguieri
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Nucleus of Medicine and Experimental Surgery, Department of Internal Medicine, FCM, Campinas, Brazil
| | - Ana Teresa Barufi Franco
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Nucleus of Medicine and Experimental Surgery, Department of Internal Medicine, FCM, Campinas, Brazil
| | - André Schwambach Vieira
- Department of Structural and Functional Biology, Biology Institute, State University of Campinas (UNICAMP), Campinas, Brazil
| | - José Antonio Rocha Gontijo
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Nucleus of Medicine and Experimental Surgery, Department of Internal Medicine, FCM, Campinas, Brazil
| | - Patricia Aline Boer
- Fetal Programming and Hydroelectrolyte Metabolism Laboratory, Nucleus of Medicine and Experimental Surgery, Department of Internal Medicine, FCM, Campinas, Brazil
| |
Collapse
|
23
|
Chen D, Wang YY, Li SP, Zhao HM, Jiang FJ, Wu YX, Tong Y, Pang QF. Maternal propionate supplementation ameliorates glucose and lipid metabolic disturbance in hypoxia-induced fetal growth restriction. Food Funct 2022; 13:10724-10736. [PMID: 36177734 DOI: 10.1039/d2fo01481e] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Intrauterine growth restriction (IUGR), one of the major complications of pregnancy, is characterized by low birth weight and results in higher risks for long-term problems including developing metabolic and cardiovascular diseases. Short-chain fatty acids (SCFAs), especially propionate, have been reported to correct glucose and lipid disorders in metabolic diseases. We hypothesized that maternal propionate supplementation could prevent glucose and lipid metabolic disturbance in hypoxia-induced IUGR. Here, in our study, maternal hypoxia was induced from gestational day (GD) 11 to GD 17.5 to establish an IUGR mouse model. Maternal propionate treatment reversed reduced birth weight in male IUGR offspring. Hepatic transcriptomics demonstrated that SP treatment significantly lowered glucose and lipid metabolism-related genes (Scd1, G6pc, Pck1 and Fasl) in IUGR offspring. KOG enrichment analysis showed that propionate-induced down-regulated differential expressed genes (DEGs) mainly belonged to lipid transport and metabolism. KEGG enrichment results showed that the down-regulated DEGs were mostly enriched in PPAR and FoxO signaling pathways. We also found that maternal oral administration of SP decreased serum lipid content, attenuated hepatic insulin resistance and liver lipid accumulation, reduced hepatic key gene expressions of gluconeogenesis and lipogenesis, increased energy expenditure and improved liver function in 11-week-old male IUGR offspring. These results indicate that maternal propionate supplementation increases birth weight and corrects hepatic glucose and lipid metabolic disturbance and energy expenditure in male mice born with IUGR, which may provide a basis for using propionate to treat IUGR disease.
Collapse
Affiliation(s)
- Dan Chen
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, 1800 Lihu Avenue, Binhu District, Wuxi 214122, Jiangsu Province, China.
| | - Ying-Ying Wang
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, 1800 Lihu Avenue, Binhu District, Wuxi 214122, Jiangsu Province, China.
| | - Sheng-Peng Li
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, 1800 Lihu Avenue, Binhu District, Wuxi 214122, Jiangsu Province, China.
| | - Hui-Min Zhao
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, 1800 Lihu Avenue, Binhu District, Wuxi 214122, Jiangsu Province, China.
| | - Feng-Juan Jiang
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, 1800 Lihu Avenue, Binhu District, Wuxi 214122, Jiangsu Province, China.
| | - Ya-Xian Wu
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, 1800 Lihu Avenue, Binhu District, Wuxi 214122, Jiangsu Province, China.
| | - Ying Tong
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, 1800 Lihu Avenue, Binhu District, Wuxi 214122, Jiangsu Province, China.
| | - Qing-Feng Pang
- Department of Physiopathology, Wuxi School of Medicine, Jiangnan University, 1800 Lihu Avenue, Binhu District, Wuxi 214122, Jiangsu Province, China.
| |
Collapse
|
24
|
Simões-Alves AC, Arcoverde-Mello APFC, Campos JDO, Wanderley AG, Leandro CVG, da Costa-Silva JH, de Oliveira Nogueira Souza V. Cardiometabolic Effects of Postnatal High-Fat Diet Consumption in Offspring Exposed to Maternal Protein Restriction In Utero. Front Physiol 2022; 13:829920. [PMID: 35620602 PMCID: PMC9127546 DOI: 10.3389/fphys.2022.829920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 03/29/2022] [Indexed: 01/01/2023] Open
Abstract
In recent decades, the high incidence of infectious and parasitic diseases has been replaced by a high prevalence of chronic and degenerative diseases. Concomitantly, there have been profound changes in the behavior and eating habits of families around the world, characterizing a "nutritional transition" phenomenon, which refers to a shift in diet in response to modernization, urbanization, or economic development from undernutrition to the excessive consumption of hypercaloric and ultra-processed foods. Protein malnutrition that was a health problem in the first half of the 20th century has now been replaced by high-fat diets, especially diets high in saturated fat, predisposing consumers to overweight and obesity. This panorama points us to the alarming coexistence of both malnutrition and obesity in the same population. In this way, individuals whose mothers were undernourished early in pregnancy and then exposed to postnatal hyperlipidic nutrition have increased risk factors for developing metabolic dysfunction and cardiovascular diseases in adulthood. Thus, our major aim was to review the cardiometabolic effects resulting from postnatal hyperlipidic diets in protein-restricted subjects, as well as to examine the epigenetic repercussions occasioned by the nutritional transition.
Collapse
Affiliation(s)
- Aiany Cibelle Simões-Alves
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Department of Physical Education and Sport Sciences, Universidade Federal de Pernambuco UFPE, Vitória de Santo Antão, Brazil
| | - Ana Paula Fonseca Cabral Arcoverde-Mello
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Department of Physical Education and Sport Sciences, Universidade Federal de Pernambuco UFPE, Vitória de Santo Antão, Brazil
| | - Jéssica de Oliveira Campos
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Department of Physical Education and Sport Sciences, Universidade Federal de Pernambuco UFPE, Vitória de Santo Antão, Brazil
| | | | - Carol Virginia Gois Leandro
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Department of Physical Education and Sport Sciences, Universidade Federal de Pernambuco UFPE, Vitória de Santo Antão, Brazil
| | - João Henrique da Costa-Silva
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Department of Physical Education and Sport Sciences, Universidade Federal de Pernambuco UFPE, Vitória de Santo Antão, Brazil
| | - Viviane de Oliveira Nogueira Souza
- Laboratory of Nutrition, Physical Activity and Phenotypic Plasticity, Department of Physical Education and Sport Sciences, Universidade Federal de Pernambuco UFPE, Vitória de Santo Antão, Brazil
| |
Collapse
|
25
|
Serpente P, Zhang Y, Islimye E, Hart-Johnson S, Gould AP. Quantification of fetal organ sparing in maternal low-protein dietary models. Wellcome Open Res 2022; 6:218. [PMID: 35634534 PMCID: PMC9120932 DOI: 10.12688/wellcomeopenres.17124.2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/25/2022] [Indexed: 11/20/2022] Open
Abstract
Background: Maternal malnutrition can lead to fetal growth restriction. This is often associated with organ sparing and long-lasting physiological dysfunctions during adulthood, although the underlying mechanisms are not yet well understood. Methods: Low protein (LP) dietary models in C57BL/6J mice were used to investigate the proximal effects of maternal malnutrition on fetal organ weights and organ sparing at embryonic day 18.5 (E18.5). Results: Maternal 8% LP diet induced strikingly different degrees of fetal growth restriction in different animal facilities, but adjustment of dietary protein content allowed similar fetal body masses to be obtained. A maternal LP diet that restricted fetal body mass by 40% did not decrease fetal brain mass to the same extent, reflecting positive growth sparing of this organ. Under these conditions, fetal pancreas and liver mass decreased by 60-70%, indicative of negative organ sparing. A series of dietary swaps between LP and standard diets showed that the liver is capable of efficient catch-up growth from as late as E14.5 whereas, after E10.5, the pancreas is not. Conclusions: This study highlights that the reproducibility of LP fetal growth restriction studies between laboratories can be improved by careful calibration of maternal dietary protein content. LP diets that induce 30-40% restriction of prenatal growth provide a good model for fetal organ sparing. For the liver, recovery of growth following protein restriction is efficient throughout fetal development but, for the pancreas, transient LP exposures spanning the progenitor expansion phase lead to an irreversible fetal growth deficit.
Collapse
Affiliation(s)
- Patricia Serpente
- Laboratory of Physiology and Metabolism, The Francis Crick Institute, London, NW1 1AT, UK
- MRC National Institute for Medical Research, UK, Mill Hill, London, NW7 1AA, UK
| | - Ying Zhang
- Laboratory of Physiology and Metabolism, The Francis Crick Institute, London, NW1 1AT, UK
| | - Eva Islimye
- Laboratory of Physiology and Metabolism, The Francis Crick Institute, London, NW1 1AT, UK
| | - Sarah Hart-Johnson
- MRC National Institute for Medical Research, UK, Mill Hill, London, NW7 1AA, UK
- Biological Research Facility, The Francis Crick Institute, London, NW1 1AT, UK
| | - Alex P. Gould
- Laboratory of Physiology and Metabolism, The Francis Crick Institute, London, NW1 1AT, UK
- MRC National Institute for Medical Research, UK, Mill Hill, London, NW7 1AA, UK
| |
Collapse
|
26
|
Cavariani MM, de Mello Santos T, Chuffa LGDA, Pinheiro PFF, Scarano WR, Domeniconi RF. Maternal Protein Restriction Alters the Expression of Proteins Related to the Structure and Functioning of the Rat Offspring Epididymis in an Age-Dependent Manner. Front Cell Dev Biol 2022; 10:816637. [PMID: 35517501 PMCID: PMC9061959 DOI: 10.3389/fcell.2022.816637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 04/05/2022] [Indexed: 11/16/2022] Open
Abstract
Nutrition is an environmental factor able to activate physiological interactions between fetus and mother. Maternal protein restriction is able to alter sperm parameters associated with epididymal functions. Since correct development and functioning of the epididymides are fundamental for mammalian reproductive success, this study investigated the effects of maternal protein restriction on epididymal morphology and morphometry in rat offspring as well as on the expression of Src, Cldn-1, AR, ER, aromatase p450, and 5α-reductase in different stages of postnatal epididymal development. For this purpose, pregnant females were allocated to normal-protein (NP—17% protein) and low-protein (LP—6% protein) groups that received specific diets during gestation and lactation. After weaning, male offspring was provided only normal-protein diet until the ages of 21, 44, and 120 days, when they were euthanized and their epididymides collected. Maternal protein restriction decreased genital organs weight as well as crown-rump length and anogenital distance at all ages. Although the low-protein diet did not change the integrity of the epididymal epithelium, we observed decreases in tubular diameter, epithelial height and luminal diameter of the epididymal duct in 21-day-old LP animals. The maternal low-protein diet changed AR, ERα, ERβ, Src 416, and Src 527 expression in offspring epididymides in an age-dependent manner. Finally, maternal protein restriction increased Cldn-1 expression throughout the epididymides at all analyzed ages. Although some of these changes did not remain until adulthood, the insufficient supply of proteins in early life altered the structure and functioning of the epididymis in important periods of postnatal development.
Collapse
|
27
|
Vipin VA, Blesson CS, Yallampalli C. Maternal low protein diet and fetal programming of lean type 2 diabetes. World J Diabetes 2022; 13:185-202. [PMID: 35432755 PMCID: PMC8984567 DOI: 10.4239/wjd.v13.i3.185] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 12/30/2021] [Accepted: 02/10/2022] [Indexed: 02/06/2023] Open
Abstract
Maternal nutrition is found to be the key factor that determines fetal health in utero and metabolic health during adulthood. Metabolic diseases have been primarily attributed to impaired maternal nutrition during pregnancy, and impaired nutrition has been an immense issue across the globe. In recent years, type 2 diabetes (T2D) has reached epidemic proportion and is a severe public health problem in many countries. Although plenty of research has already been conducted to tackle T2D which is associated with obesity, little is known regarding the etiology and pathophysiology of lean T2D, a variant of T2D. Recent studies have focused on the effects of epigenetic variation on the contribution of in utero origins of lean T2D, although other mechanisms might also contribute to the pathology. Observational studies in humans and experiments in animals strongly suggest an association between maternal low protein diet and lean T2D phenotype. In addition, clear sex-specific disease prevalence was observed in different studies. Consequently, more research is essential for the understanding of the etiology and pathophysiology of lean T2D, which might help to develop better disease prevention and treatment strategies. This review examines the role of protein insufficiency in the maternal diet as the central driver of the developmental programming of lean T2D.
Collapse
Affiliation(s)
- Vidyadharan Alukkal Vipin
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX 77030, United States
| | - Chellakkan Selvanesan Blesson
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX 77030, United States
- Family Fertility Center, Texas Children's Hospital, Houston, TX 77030, United States
| | - Chandra Yallampalli
- Department of Obstetrics and Gynecology, Baylor College of Medicine, Houston, TX 77030, United States
| |
Collapse
|
28
|
Vargas-Rodríguez I, Reyes-Castro LA, Pacheco-López G, Lomas-Soria C, Zambrano E, Díaz-Ruíz A, Diaz-Cintra S. POSTNATAL EXPOSURE TO LIPOPOLYSACCHARIDE COMBINED WITH HIGH-FAT DIET CONSUMPTION INDUCES IMMUNE TOLERANCE WITHOUT PREVENTION IN SPATIAL WORKING MEMORY IMPAIRMENT. Behav Brain Res 2022; 423:113776. [PMID: 35120930 DOI: 10.1016/j.bbr.2022.113776] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 01/21/2022] [Accepted: 01/30/2022] [Indexed: 02/05/2023]
Abstract
High-fat diet (HFD) consumption has been related to metabolic alterations, such as obesity and cardiovascular problems, and has pronounced effects on brain plasticity and memory impairment. HFD exposure has a pro-inflammatory effect associated with microglial cell modifications in the hippocampus, a region involved in the working memory process. Immune tolerance can protect from inflammation in periphery induced by HFD consumption, when the immune response is desensitized in development period with lipopolysaccharide (LPS) exposure, maybe this previously state can change the course of the diseases associated to HFDs but is not known if can protect the hippocampus's inflammatory response. In the present study, male mice were injected with LPS (100μg.kg-1 body weight) on postnatal day 3 and fed with HFD for 16 weeks after weaning. Ours results indicated that postnatal exposure to LPS in the early postnatal developmental stage combined with HFD consumption prevented glycemia, insulin, HOMA-IR, microglial process, and increased pro-inflammatory cytokines mRNA expression, without changes in body weight gain and spatial working memory with respect vehicle + HFD group. These findings suggest that HFD consumption after postnatal LPS exposure induces hippocampal immune tolerance, without prevention in spatial working memory impairment on male mice.
Collapse
Affiliation(s)
- Isaac Vargas-Rodríguez
- Departamento de Neurobiología del Desarrollo y Neurofisiología. Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, C.P. 76230, México
| | - Luis Antonio Reyes-Castro
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición, Salvador Zubirán, México 14080
| | - Gustavo Pacheco-López
- División de Ciencias de Biológicas y de la Salud, Universidad Autónoma Metropolitana, Unidad Lerma, Estado de México, C.P. 52005, México
| | - Consuelo Lomas-Soria
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición, Salvador Zubirán, México 14080; CONACyT-Cátedras, Departamento de Biología de la Reproducción. Instituto Nacional de Ciencias Médicas y Nutrición, Salvador Zubirán, México 14080
| | - Elena Zambrano
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición, Salvador Zubirán, México 14080
| | - Araceli Díaz-Ruíz
- Departamento de Neuroquímica, Instituto Nacional de Neurologı́a y Neurocirugı́a, Manuel Velasco Suárez S.S.A, México, CP, 14269, México
| | - Sofía Diaz-Cintra
- Departamento de Neurobiología del Desarrollo y Neurofisiología. Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, C.P. 76230, México.
| |
Collapse
|
29
|
Intergenerational protein deficiency and adolescent reproductive function of subsequent female generations (F 1 and F 2) in rat model. Curr Res Physiol 2022; 5:16-24. [PMID: 35024624 PMCID: PMC8724923 DOI: 10.1016/j.crphys.2021.12.003] [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: 03/06/2021] [Revised: 12/02/2021] [Accepted: 12/20/2021] [Indexed: 11/30/2022] Open
Abstract
Background Efficient reproductive function is an important characteristic that has evolved through natural selection. Nutrition can modulate reproductive activities at different levels, and its effect on reproduction is deemed complex and less predictable. Objective This study aims at investigating the underlying effect of persistent dietary protein deficiency during early life on reproductive parameters of subsequent (F1 and F2) generations. Method Rats in group of four (4) were fed daily, different ration of protein diet (PD) formulated as: 21% protein diet, 10%protein diet, 5%protein diet and control diet (rat chow, containing 16–18% protein). They were fed ad libitum before mating, throughout gestation and lactation, and next generations were weaned to the maternal diet. Reproductive function analysis (which include; gestation and pubertal hormonal profiling, onset of puberty, oestrus cyclicity, sexual response) and morphometric analysis of the ovarian structure were carried out to assess associated consequences. Results There was significant reduction in the fertility index (Control; 85.8%., 21%PD; 88.43%., as compared to 10%PD; 65.9%., 5%PD; 35.78%.,) at F1, also recurring in F2 respectively as a consequence of altered reproductive function in the protein deficient models at P ≤ 0.05. Low protein diet posed suboptimal intrauterine condition, which was linked to increased prenatal morbidity and mortality (control; 11.3%., 21%PD; 3.3%., 10%PD; 27.4%., 5%PD; 32.9%), low birthweight (control; 5.29, 4.9 g., 21%PD; 5.5, 5.06 g., 10%PD; 4.05, 3.86 g., 5%PD; 2.7, 2.5 g) at F1 and F2 respectively, delayed onset of puberty (with average pubertal age set at: control; PND 36, 21%PD; PND 38 while 10%PD; PND 62., and 5%PD; PND 67), followed by induced cycle irregularity, altered follicular maturation and endocrine dysfunction, more severe in 5%PD. Conclusion Reproductive status of a female organism depends on the maintenance of ovarian structure and function that has been associated with the hypothalamic pituitary-gonadal axis, hormonal events and sexual maturity. There is therefore an association between persistent early life protein deficiency and reproductive response which mechanistically involves life-long changes in key ovarian cytoarchitecture and function. Intergenerational protein malnutrition exerts female hormonal dysregulation and irregular cyclicity at adolescence. It delayed pubertal attainment and reproductive performance that persists to the next–generation of rats. Perinatal protein deficiency also altered the ovarian morphology with an implication on fertility index across generations.
Collapse
Key Words
- (↑), Concentration Increase
- (↓), Concentration decrease
- Cycle irregularity
- E2, Estradiol/Estrogen
- F0, Parent
- F1, First filial generation
- F2, Second filial generation
- FSH, Follicle Stimulating Hormone
- Fertility index
- Follicle
- GD, Gestation Day
- IUGR, Intrauterine Growth Restriction
- LH, Leutenizing Hormone
- Ovarian-degeneration. ovarian function
- PD, Protein Diet
- PDD, Protein Deficient Diet
- PND, Postnatal Day
- PROG, Progesterone
- Protein deficiency
- Reproductive hormone
Collapse
|
30
|
Masiero BC, Calsa B, Oliveira CA, Moretti de Andrade TA, Esquisatto MAM, Catisti R. Morphofunctional and immunological cardiac evaluation of protein restriction on rat offspring. Ann Anat 2022; 241:151889. [DOI: 10.1016/j.aanat.2022.151889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 01/10/2022] [Accepted: 01/11/2022] [Indexed: 11/24/2022]
|
31
|
Grzęda E, Matuszewska J, Ziarniak K, Gertig-Kolasa A, Krzyśko- Pieczka I, Skowrońska B, Sliwowska JH. Animal Foetal Models of Obesity and Diabetes - From Laboratory to Clinical Settings. Front Endocrinol (Lausanne) 2022; 13:785674. [PMID: 35197931 PMCID: PMC8858803 DOI: 10.3389/fendo.2022.785674] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 01/11/2022] [Indexed: 12/26/2022] Open
Abstract
The prenatal period, during which a fully formed newborn capable of surviving outside its mother's body is built from a single cell, is critical for human development. It is also the time when the foetus is particularly vulnerable to environmental factors, which may modulate the course of its development. Both epidemiological and animal studies have shown that foetal programming of physiological systems may alter the growth and function of organs and lead to pathology in adulthood. Nutrition is a particularly important environmental factor for the pregnant mother as it affects the condition of offspring. Numerous studies have shown that an unbalanced maternal metabolic status (under- or overnutrition) may cause long-lasting physiological and behavioural alterations, resulting in metabolic disorders, such as obesity and type 2 diabetes (T2DM). Various diets are used in laboratory settings in order to induce maternal obesity and metabolic disorders, and to alter the offspring development. The most popular models are: high-fat, high-sugar, high-fat-high-sugar, and cafeteria diets. Maternal undernutrition models are also used, which results in metabolic problems in offspring. Similarly to animal data, human studies have shown the influence of mothers' diets on the development of children. There is a strong link between the maternal diet and the birth weight, metabolic state, changes in the cardiovascular and central nervous system of the offspring. The mechanisms linking impaired foetal development and adult diseases remain under discussion. Epigenetic mechanisms are believed to play a major role in prenatal programming. Additionally, sexually dimorphic effects on offspring are observed. Therefore, further research on both sexes is necessary.
Collapse
Affiliation(s)
- Emilia Grzęda
- Laboratory of Neurobiology, Faculty of Veterinary Medicine and Animal Science, Poznań University of Life Sciences, Poznań, Poland
| | - Julia Matuszewska
- Laboratory of Neurobiology, Faculty of Veterinary Medicine and Animal Science, Poznań University of Life Sciences, Poznań, Poland
| | - Kamil Ziarniak
- Laboratory of Neurobiology, Faculty of Veterinary Medicine and Animal Science, Poznań University of Life Sciences, Poznań, Poland
- Molecular and Cell Biology Unit, Poznań University of Medical Sciences, Poznań, Poland
| | - Anna Gertig-Kolasa
- Department of Paediatric Diabetes and Obesity, Poznań University of Medical Sciences, Poznań, Poland
| | - Izabela Krzyśko- Pieczka
- Department of Paediatric Diabetes and Obesity, Poznań University of Medical Sciences, Poznań, Poland
| | - Bogda Skowrońska
- Department of Paediatric Diabetes and Obesity, Poznań University of Medical Sciences, Poznań, Poland
| | - Joanna H. Sliwowska
- Laboratory of Neurobiology, Faculty of Veterinary Medicine and Animal Science, Poznań University of Life Sciences, Poznań, Poland
- *Correspondence: Joanna H. Sliwowska,
| |
Collapse
|
32
|
Cracco RC, Bussiman FDO, Polizel GHG, Furlan É, Garcia NP, Poit DAS, Pugliesi G, Santana MHDA. Effects of Maternal Nutrition on Female Offspring Weight Gain and Sexual Development. Front Genet 2021; 12:737382. [PMID: 34887899 PMCID: PMC8650139 DOI: 10.3389/fgene.2021.737382] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 10/04/2021] [Indexed: 11/29/2022] Open
Abstract
Maternal nutrition during pregnancy influences postnatal life of animals; nevertheless, few studies have investigated its effects on the productive performance and reproductive development of heifers. This study evaluated the performance, reproductive development, and correlation between reproduction × fat thickness and performance × ribeye area (REA) traits of heifers. We also performed an exploratory genomic association during the rearing period in heifers submitted to fetal programming. The study comprised 55 Nellore heifers born to dams exposed to one of the following nutritional planes: control, without protein-energy supplementation; PELT, protein-energy last trimester, protein-energy supplementation offered in the final third of pregnancy; and PEWG, protein-energy whole gestation, protein-energy supplementation upon pregnancy confirmation. Protein-energy supplementation occurred at the level of 0.3% live weight. After weaning, heifers were submitted to periodic evaluations of weight and body composition by ultrasonography. From 12 to 18 months, we evaluated the reproductive tract of heifers to monitor its development for sexual precocity and ovarian follicle population. The treatments had no effect (p > 0.05) on average daily gain; however, the weight of the animals showed a significant difference over time (p = 0.017). No differences were found between treatments for REA, backfat, and rump fat thickness, nor for puberty age, antral follicular count, and other traits related to reproductive tract development (p > 0.05). The correlation analysis between performance traits and REA showed high correlations (r > 0.37) between REA at weaning and year versus weight from weaning until yearling; however, no correlation was found for reproductive development traits versus fat thickness (p > 0.05). The exploratory genomic association study showed one single-nucleotide polymorphism (SNP) for each treatment on an intergenic region for control and PEWG, and the one for PELT on an intronic region of RAPGEF1 gene. Maternal nutrition affected only the weight of the animals throughout the rearing period.
Collapse
Affiliation(s)
- Roberta Cavalcante Cracco
- Department of Animal Science, College of Animal Science and Food Engineering - USP, Pirassununga, Brazil
| | | | | | - Édison Furlan
- Department of Animal Science, College of Animal Science and Food Engineering - USP, Pirassununga, Brazil
| | - Nara Pontes Garcia
- Departament of Veterinary Medicine, College of Animal Science and Food Engineering - USP, Pirassununga, Brazil
| | - Diego Angelo Schmidt Poit
- Department of Animal Reproduction, College of Veterinary Medicine and Animal Science - USP, Pirassununga, Brazil
| | - Guilherme Pugliesi
- Department of Animal Reproduction, College of Veterinary Medicine and Animal Science - USP, Pirassununga, Brazil
| | | |
Collapse
|
33
|
Lisboa PC, Miranda RA, Souza LL, Moura EG. Can breastfeeding affect the rest of our life? Neuropharmacology 2021; 200:108821. [PMID: 34610290 DOI: 10.1016/j.neuropharm.2021.108821] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 08/17/2021] [Accepted: 09/30/2021] [Indexed: 12/31/2022]
Abstract
The breastfeeding period is one of the most important critical windows in our development, since milk, our first food after birth, contains several compounds, such as macronutrients, micronutrients, antibodies, growth factors and hormones that benefit human health. Indeed, nutritional, and environmental alterations during lactation, change the composition of breast milk and induce alterations in the child's development, such as obesity, leading to the metabolic dysfunctions, cardiovascular diseases and neurobehavioral disorders. This review is based on experimental animal models, most of them in rodents, and summarizes the impact of an adequate breast milk supply in view of the developmental origins of health and disease (DOHaD) concept, which has been proposed by researchers in the areas of epidemiology and basic science from around the world. Here, experimental advances in understanding the programming during breastfeeding were compiled with the purpose of generating knowledge about the genesis of chronic noncommunicable diseases and to guide the development of public policies to deal with and prevent the problems arising from this phenomenon. This review article is part of the special issue on "Cross talk between periphery and brain".
Collapse
Affiliation(s)
- Patricia C Lisboa
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| | - Rosiane A Miranda
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Luana L Souza
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Egberto G Moura
- Laboratory of Endocrine Physiology, Department of Physiological Sciences, Roberto Alcantara Gomes Biology Institute, State University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
34
|
Chronic Effects of Maternal Low-Protein and Low-Quality Protein Diets on Body Composition, Glucose-Homeostasis and Metabolic Factors, Followed by Reversible Changes upon Rehabilitation in Adult Rat Offspring. Nutrients 2021; 13:nu13114129. [PMID: 34836384 PMCID: PMC8624605 DOI: 10.3390/nu13114129] [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: 09/14/2021] [Revised: 10/27/2021] [Accepted: 11/10/2021] [Indexed: 11/16/2022] Open
Abstract
Several studies suggest that the maternal protein content and source can affect the offspring's health. However, the chronic impact of maternal quality and quantity protein restriction, and reversible changes upon rehabilitation, if any, in the offspring, remains elusive. This study examined the effects of maternal low-quality protein (LQP) and low-protein (LP) intake from preconception to post-weaning, followed by rehabilitation from weaning, on body composition, glucose-homeostasis, and metabolic factors in rat offspring. Wistar rats were exposed to normal protein (NP; 20% casein), LQP (20% wheat gluten) or LP (8% casein) isocaloric diets for 7 weeks before pregnancy until lactation. After weaning, the offspring were exposed to five diets: NP, LQP, LQPR (LQP rehabilitated with NP), LP, and LPR (LP rehabilitated with NP) for 16 weeks. Body composition, glucose-homeostasis, lipids, and plasma hormones were investigated. The LQP and LP offspring had lower bodyweight, fat and lean mass, insulin and HOMA-IR than the NP. The LQP offspring had higher cholesterol, T3 and T4, and lower triacylglycerides and glucose, while these were unaltered in LP compared to NP. The majority of the above outcomes were reversed upon rehabilitation. These results suggest that the chronic exposure of rats to maternal LQP and LP diets induced differential adverse effects by influencing body composition and metabolism, which were reversed upon rehabilitation.
Collapse
|
35
|
Igbayilola Y, Morakinyo A, Iranloye B. Adverse effects of perinatal protein restriction on glucose homeostasis in offspring of Sprague-Dawley rats. SCIENTIFIC AFRICAN 2021. [DOI: 10.1016/j.sciaf.2021.e01036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
36
|
Foroutan A, Wishart DS, Fitzsimmons C. Exploring Biological Impacts of Prenatal Nutrition and Selection for Residual Feed Intake on Beef Cattle Using Omics Technologies: A Review. Front Genet 2021; 12:720268. [PMID: 34790219 PMCID: PMC8592258 DOI: 10.3389/fgene.2021.720268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 10/06/2021] [Indexed: 11/23/2022] Open
Abstract
Approximately 70% of the cost of beef production is impacted by dietary intake. Maximizing production efficiency of beef cattle requires not only genetic selection to maximize feed efficiency (i.e., residual feed intake (RFI)), but also adequate nutrition throughout all stages of growth and development to maximize efficiency of growth and reproductive capacity, even during gestation. RFI as a measure of feed efficiency in cattle has been recently accepted and used in the beef industry, but the effect of selection for RFI upon the dynamics of gestation has not been extensively studied, especially in the context of fluctuating energy supply to the dam and fetus. Nutrient restriction during gestation has been shown to negatively affect postnatal growth and development as well as fertility of beef cattle offspring. This, when combined with the genetic potential for RFI, may significantly affect energy partitioning in the offspring and subsequently important performance traits. In this review, we discuss: 1) the importance of RFI as a measure of feed efficiency and how it can affect other economic traits in beef cattle; 2) the influence of prenatal nutrition on physiological phenotypes in calves; 3) the benefits of investigating the interaction of genetic selection for RFI and prenatal nutrition; 4) how metabolomics, transcriptomics, and epigenomics have been employed to investigate the underlying biology associated with prenatal nutrition, RFI, or their interactions in beef cattle; and 5) how the integration of omics information is adding a level of deeper understanding of the genetic architecture of phenotypic traits in cattle.
Collapse
Affiliation(s)
- Aidin Foroutan
- Department of Pathology and Laboratory Medicine, Western University, London, ON, Canada
| | - David S. Wishart
- Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada
- Department of Computing Science, University of Alberta, Edmonton, AB, Canada
| | - Carolyn Fitzsimmons
- Department of Agricultural Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
- Agriculture and Agri-Food Canada, Edmonton, AB, Canada
| |
Collapse
|
37
|
Christians JK. The Placenta's Role in Sexually Dimorphic Fetal Growth Strategies. Reprod Sci 2021; 29:1895-1907. [PMID: 34699045 DOI: 10.1007/s43032-021-00780-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 10/19/2021] [Indexed: 12/27/2022]
Abstract
Fetal sex affects the risk of pregnancy complications and the long-term effects of prenatal environment on health. Some have hypothesized that growth strategies differ between the sexes, whereby males prioritize growth whereas females are more responsive to their environment. This review evaluates the role of the placenta in such strategies, focusing on (1) mechanisms underlying sexual dimorphism in gene expression, (2) the nature and extent of sexual dimorphism in placental gene expression, (3) sexually dimorphic responses to nutrient supply, and (4) sexual dimorphism in morphology and histopathology. The sex chromosomes contribute to sex differences in placental gene expression, and fetal hormones may play a role later in development. Sexually dimorphic placental gene expression may contribute to differences in the prevalence of complications such as preeclampsia, although this link is not clear. Placental responses to nutrient supply frequently show sexual dimorphism, but there is no consistent pattern where one sex is more responsive. There are sex differences in the prevalence of placental histopathologies, and placental changes in pregnancy complications, but also many similarities. Overall, no clear patterns support the hypothesis that females are more responsive to the maternal environment, or that males prioritize growth. While male fetuses are at greater risk of a variety of complications, total prenatal mortality is higher in females, such that males exposed to early insults may be more likely to survive and be observed in studies of adverse outcomes. Going forward, robust statistical approaches to test for sex-dependent effects must be more widely adopted to reduce the incidence of spurious results.
Collapse
Affiliation(s)
- Julian K Christians
- Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby, BC, V5A 1S6, Canada. .,Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, BC, Canada. .,British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada. .,Women's Health Research Institute, BC Women's Hospital and Health Centre, Vancouver, BC, Canada.
| |
Collapse
|
38
|
Zambrano E, Nathanielsz PW, Rodríguez-González GL. Developmental programming and ageing of male reproductive function. Eur J Clin Invest 2021; 51:e13637. [PMID: 34107063 DOI: 10.1111/eci.13637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/25/2021] [Accepted: 06/08/2021] [Indexed: 12/22/2022]
Abstract
Developmental programming predisposes offspring to metabolic, behavioural and reproductive dysfunction in adult life. Evidence is accumulating that ageing phenotype and longevity are in part developmentally programmed in each individual. Unfortunately, there are few studies addressing the effects of developmental programming by maternal nutrition on the rate of ageing of the male reproductive system. This review will discuss effects of foetal exposure to maternal environmental challenges on male offspring fertility and normal ageing of the male reproductive system. We focus on several key factors involved in reproductive ageing such as decreased hormone production, DNA fragmentation, oxidative stress, telomere shortening, epigenetics, maternal lifestyle and nutrition. There is compelling evidence that ageing of the male reproductive system is developmentally programmed. Both maternal over- or undernutrition accelerate ageing of male offspring reproductive function through similar mechanisms such as decreased serum testosterone levels, increase in oxidative stress biomarkers in both the testes and sperm and changes in sperm quality. Importantly, even in adult life, exercise in male offspring of obese mothers improves adverse effects of programming on reproductive function. Maternal consumption of a low-protein diet causes transgenerational effects in progeny via the paternal line. The seminal fluid has effects on the intrauterine environment. Programming by male factors may involve more than just the sperm. Improving knowledge on developmental programming ageing interactions will improve not only male health and life span but also the health of future generations by reducing programming via the paternal line.
Collapse
Affiliation(s)
- Elena Zambrano
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | | | - Guadalupe L Rodríguez-González
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| |
Collapse
|
39
|
Zambrano E, Lomas-Soria C, Nathanielsz PW. Rodent studies of developmental programming and ageing mechanisms: Special issue: In utero and early life programming of ageing and disease. Eur J Clin Invest 2021; 51:e13631. [PMID: 34061987 DOI: 10.1111/eci.13631] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/30/2021] [Accepted: 05/30/2021] [Indexed: 12/31/2022]
Abstract
Compelling evidence exists indicating that developmental programming influences ageing. Programming alters life-course phenotype in multiple organs, predisposing to diseases such as diabetes, obesity and cardiovascular disease that shorten lifespan. This review describes studies in rodents, the most commonly studied species, addressing interactions of programming challenges with ageing. We first consider ageing and programming of insulin function that has been clearly shown to decrease with age. It is important to evaluate ageing in pancreatic islets isolated from other systems. Studies discussed show premature pancreatic islet ageing resulting from both maternal under- and overnutrition. New ways to determine programming of adipose tissue and effects on fat storage are explored. Oxidative stress is a major factor that regulates ageing in tissues. Oxidative stress is discussed in relation to reproductive and cardiovascular ageing. Premature ageing is associated with both low and high glucocorticoid function. Both over and undernutrition have offspring sex-specific programming effects on life-course glucocorticoid concentrations. Evidence is provided that maternal age at conception affects offspring endocrine and metabolism ageing. Finally, the importance of matching foetal nutrition and energy availability with composition and energy content in the post-weaning diet is demonstrated. This mismatch can lead to a greatly shortened lifespan. General principles are discussed throughout. For example, sexual dimorphism of age-related outcomes can be marked. Accelerated ageing occurs early in life. Improving knowledge on programming ageing interactions will improve health span as well as lifespan. Finally, there are considerable similarities in outcomes programmed by maternal undernutrition and overnutrition.
Collapse
Affiliation(s)
- Elena Zambrano
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México
| | - Consuelo Lomas-Soria
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, México.,Reproductive Biology Department, CONACyT-Cátedras, Instituto Nacional de Ciencias Médicas y Nutrición SZ, Mexico City, México
| | - Peter W Nathanielsz
- Department of Animal Science, Texas Pregnancy and Life-course Health Center, University of Wyoming, Laramie, WY, USA
| |
Collapse
|
40
|
Serpente P, Zhang Y, Islimye E, Hart-Johnson S, Gould AP. Quantification of fetal organ sparing in maternal low-protein dietary models. Wellcome Open Res 2021; 6:218. [PMID: 35634534 PMCID: PMC9120932 DOI: 10.12688/wellcomeopenres.17124.1] [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] [Accepted: 08/20/2021] [Indexed: 11/08/2023] Open
Abstract
Background: Maternal malnutrition can lead to fetal growth restriction. This is often associated with organ sparing and long-lasting physiological dysfunctions during adulthood, although the underlying mechanisms are not yet well understood. Methods: Low protein (LP) dietary models in C57BL/6J mice were used to investigate the proximal effects of maternal malnutrition on fetal organ weights and organ sparing at embryonic day 18.5 (E18.5). Results: Maternal 8% LP diet induced strikingly different degrees of fetal growth restriction in different animal facilities, but adjustment of dietary protein content allowed similar fetal body masses to be obtained. A maternal LP diet that restricted fetal body mass by 40% did not decrease fetal brain mass to the same extent, reflecting positive growth sparing of this organ. Under these conditions, fetal pancreas and liver mass decreased by 60-70%, indicative of negative organ sparing. A series of dietary swaps between LP and standard diets showed that the liver is capable of efficient catch-up growth from as late as E14.5 whereas, after E10.5, the pancreas is not. Conclusions: This study highlights that the reproducibility of LP fetal growth restriction studies between laboratories can be improved by careful calibration of maternal dietary protein content. LP diets that induce 30-40% restriction of prenatal growth provide a good model for fetal organ sparing. For the liver, recovery of growth following protein restriction is efficient throughout fetal development but, for the pancreas, transient LP exposures spanning the progenitor expansion phase lead to an irreversible fetal growth deficit.
Collapse
Affiliation(s)
- Patricia Serpente
- Laboratory of Physiology and Metabolism, The Francis Crick Institute, London, NW1 1AT, UK
- MRC National Institute for Medical Research, UK, Mill Hill, London, NW7 1AA, UK
| | - Ying Zhang
- Laboratory of Physiology and Metabolism, The Francis Crick Institute, London, NW1 1AT, UK
| | - Eva Islimye
- Laboratory of Physiology and Metabolism, The Francis Crick Institute, London, NW1 1AT, UK
| | - Sarah Hart-Johnson
- MRC National Institute for Medical Research, UK, Mill Hill, London, NW7 1AA, UK
- Biological Research Facility, The Francis Crick Institute, London, NW1 1AT, UK
| | - Alex P. Gould
- Laboratory of Physiology and Metabolism, The Francis Crick Institute, London, NW1 1AT, UK
- MRC National Institute for Medical Research, UK, Mill Hill, London, NW7 1AA, UK
| |
Collapse
|
41
|
Castro-Rodríguez DC, Reyes-Castro LA, Vargas-Hernández L, Itani N, Nathanielsz PW, Taylor PD, Zambrano E. Maternal obesity (MO) programs morphological changes in aged rat offspring small intestine in a sex dependent manner: Effects of maternal resveratrol supplementation. Exp Gerontol 2021; 154:111511. [PMID: 34371097 DOI: 10.1016/j.exger.2021.111511] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 07/22/2021] [Accepted: 08/04/2021] [Indexed: 01/10/2023]
Abstract
Maternal obesity (MO) leads to offspring metabolic problems. The mechanisms involved are multifactorial. The small intestine plays an important role in the absorption of nutrients and is modified as we age. Few studies have explored MO programming effects on offspring (F1) small intestine morphology. The aim of this study was to investigate MO effects on old adult F1 intestinal morphology, and whether any F1 intestinal changes due to MO were modified by maternal resveratrol supplementation. From weaning throughout pregnancy and lactation, female Wistar rats (F0) ate standard chow (controls, C: 5%-fat) or high-fat diet (MO: 25%-fat). One month before mating at postnatal day (PND) 120 through lactation half of each group received 20 mg/kg/day of resveratrol orally (Cres or MOres). After weaning F1 were fed with chow diet until the end of the study at PND 650. Body weight, percent of fat, glucose, cholesterol and triglyceride serum concentrations were determined. F1 small intestinal samples were collected for histological analysis. Male F1 body weight was higher in MO and MOres compared with C and Cres. Female F1 body weight and percent of fat was higher in MO than C and MOres. Triglyceride concentrations were higher in MO and MOres male F1 compared with C and Cres. There were no differences among groups in female triglyceride concentrations. Male F1 duodenal villus height was smaller in MO compared with MOres. Female F1 duodenal and jejunal crypt depth was smaller in MO compared with C and was greater compared with MOres. Female F1 villus height in jejunum was greater in MO compared with MOres. In conclusion, exposure to the developmental challenge of MO changed the aged F1 intestinal morphological and metabolic profiles. Maternal resveratrol supplementation ameliorated these effects in an F1 sex dependent manner.
Collapse
Affiliation(s)
- Diana C Castro-Rodríguez
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico; CONACyT-Cátedras, Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Luis A Reyes-Castro
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Lilia Vargas-Hernández
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico; Instituto Mexicano del Seguro Social, Hospital de Ginecologia y Obstetricia No. 4 Luis Castelazo Ayala, Mexico
| | - Nozomi Itani
- Department of Women and Children's Health, School of Life Course Sciences, King's College London and King's Health Partners, London, UK
| | - Peter W Nathanielsz
- Wyoming Center for Pregnancy and Life Course Health Research, Department of Animal Science, University Wyoming, Laramie, WY, USA
| | - Paul D Taylor
- Department of Women and Children's Health, School of Life Course Sciences, King's College London and King's Health Partners, London, UK
| | - Elena Zambrano
- Departamento de Biología de la Reproducción, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| |
Collapse
|
42
|
Castro-Rodríguez DC, Reyes-Castro LA, Vega CC, Rodríguez-González GL, Yáñez-Fernández J, Zambrano E. Leuconostoc mesenteroides subsp. mesenteroides SD23 Prevents Metabolic Dysfunction Associated with High-Fat Diet-Induced Obesity in Male Mice. Probiotics Antimicrob Proteins 2021; 12:505-516. [PMID: 31129870 DOI: 10.1007/s12602-019-09556-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
High-fat diet (HFD) consumption induces obesity and increases blood glucose, insulin resistance, and metabolic disorders. Recent studies suggest that probiotics might be a novel approach to counteract these effects in the treatment of obesity. Here, we evaluated the effect of Leuconostoc mesenteroides subsp. mesenteroides SD23 on obesity-related metabolic dysfunction. In the present study, mice were randomly divided into four dietary groups: standard diet (C), HFD (OB), standard diet with L. mesenteroides SD23 (CP), and HFD with L. mesenteroides SD23 (OBP). Diets were maintained for 14 weeks. Animal weight was monitored and biochemical and histological analyses were performed after intervention. OB showed metabolic dysfunction, and increased the number of larger adipocytes compared to C. OB induced liver tumor necrosis factor-α (TNF-α) expression, increased cholesterol, leptin, and glucose levels compared to C. OBP reduced body weight, glucose, cholesterol, and leptin levels and improved glucose tolerance compared to OB. OBP also reduced liver steatosis, the number of larger adipocytes in adipose tissue, and reduced the villus height in the small intestine. OBP decreased expression of TNF-α and increased expression of IL-10 in liver. The parameters evaluated in the CP were similar to the C. This study provides novel evidence that dietary intervention with L. mesenteroides SD23 improves metabolic dysfunction related to obesity in HFD-fed mice.
Collapse
Affiliation(s)
- Diana C Castro-Rodríguez
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Bioprocess Department, Unidad Profesional Interdisciplinaria de Biotecnología (UPIBI), Instituto Politécnico Nacional (IPN), Mexico City, Mexico.,CONACyT-Cátedras, Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Luis A Reyes-Castro
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Claudia C Vega
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Guadalupe L Rodríguez-González
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Jorge Yáñez-Fernández
- Bioprocess Department, Unidad Profesional Interdisciplinaria de Biotecnología (UPIBI), Instituto Politécnico Nacional (IPN), Mexico City, Mexico
| | - Elena Zambrano
- Reproductive Biology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.
| |
Collapse
|
43
|
Savitsky B, Manor O, Lawrence G, Friedlander Y, Siscovick DS, Hochner H. Environmental mismatch and obesity in humans: The Jerusalem Perinatal Family Follow-Up Study. Int J Obes (Lond) 2021; 45:1404-1417. [PMID: 33762678 DOI: 10.1038/s41366-021-00802-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 03/04/2021] [Indexed: 11/09/2022]
Abstract
BACKGROUND According to the hypothesis of Gluckman and Hanson, mismatch between the developmental and postdevelopmental environments may lead to detrimental health impacts such as obesity. While several animal studies support the mismatch theory, there is a scarcity of evidence from human-based studies. OBJECTIVES Our study aims to examine whether a mismatch between the developmental and young-adult environments affect obesity in young adults of the Jerusalem Perinatal Family Follow-Up Study. METHODS Data from The Jerusalem Perinatal Family Follow-Up Study birth cohort was used to characterize early and late environments using offspring and parental sociodemographic and lifestyle information at birth, age 32 (n = 1140) and 42 (n = 404). Scores characterizing the early and late environments were constructed using factor analysis. To assess associations of mismatch with obesity, regression models were fitted using the first factor of each environment and adiposity measures at age 32 and 42. RESULTS Having a stable non-beneficial environment at birth and young-adulthood was most strongly associated with increased adiposity, while a stable beneficial environment was most favorable. The transition from a beneficial environment at birth to a less beneficial environment at young-adulthood was associated with higher obesity measures, including higher BMI (β = 0.979; 95% CI: 0.029, 1.929), waist circumference (β = 2.729; 95% CI: 0.317, 5.140) and waist-hip ratio (β = 0.017; 95% CI: 0.004, 0.029) compared with those experiencing a beneficial environment at both time points. Transition from a less beneficial environment at birth to a beneficial environment at adulthood was also associated with higher obesity measurements (BMI -β = 1.116; 95% CI: 0.085, 2.148; waist circumference -β = 2.736; 95% CI: 0.215, 5.256). CONCLUSIONS This study provides some support for the mismatch hypothesis. While there is indication that an accumulation of the effects of the non-beneficial environment has the strongest detrimental impact on obesity outcomes, our results also indicate that a mismatch between the developmental and later environments may result in maladaptation of the individual leading to obesity.
Collapse
Affiliation(s)
- B Savitsky
- The Braun School of Public Health, The Hebrew University-Hadassah Medical Center, Jerusalem, Israel. .,Ashkelon Academic College, School of Health Sciences, Ashkelon, Israel.
| | - O Manor
- The Braun School of Public Health, The Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - G Lawrence
- The Braun School of Public Health, The Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | - Y Friedlander
- The Braun School of Public Health, The Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| | | | - H Hochner
- The Braun School of Public Health, The Hebrew University-Hadassah Medical Center, Jerusalem, Israel
| |
Collapse
|
44
|
Christians JK, Shergill HK, Albert AYK. Sex-dependent effects of prenatal food and protein restriction on offspring physiology in rats and mice: systematic review and meta-analyses. Biol Sex Differ 2021; 12:21. [PMID: 33563335 PMCID: PMC7871651 DOI: 10.1186/s13293-021-00365-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 01/31/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Males and females may experience different effects of early-life adversity on life-long health. One hypothesis is that male foetuses invest more in foetal growth and relatively less in placental growth, and that this makes them susceptible to poor nutrition in utero, particularly if nutrition is reduced part-way through gestation. OBJECTIVES Our objectives were to examine whether (1) food and/ or protein restriction in rats and mice has consistent sex-dependent effects, (2) sex-dependency differs between types of outcomes, and (3) males are more severely affected when restriction starts part-way through gestation. DATA SOURCES PubMed and Web of Science were searched to identify eligible studies. STUDY ELIGIBILITY CRITERIA Eligible studies described controlled experiments that restricted protein or food during gestation in rats or mice, examined physiological traits in offspring from manipulated pregnancies, and tested whether effects differed between males and females. RESULTS Our search identified 292 articles, of which the full texts of 72 were assessed, and 65 were included for further synthesis. A majority (50) used Wistar or Sprague-Dawley rats and so these were the primary focus. Among studies in which maternal diet was restricted for the duration of gestation, no type of trait was consistently more severely affected in one particular sex, although blood pressure was generally increased in both sexes. Meta-analysis found no difference between sexes in the effect of protein restriction throughout gestation on blood pressure. Among studies restricting food in the latter half of gestation only, there were again few consistent sex-dependent effects, although three studies found blood pressure was increased in males only. Meta-analysis found that food restriction in the second half of gestation increased adult blood pressure in both sexes, with a significantly greater effect in males. Birthweight was consistently reduced in both sexes, a result confirmed by meta-analysis. CONCLUSIONS We found little support for the hypotheses that males are more affected by food and protein restriction, or that effects are particularly severe if nutrition is reduced part-way through gestation. However, less than half of the studies tested for sex by maternal diet interactions to identify sex-dependent effects. As a result, many reported sex-specific effects may be false positives.
Collapse
Affiliation(s)
- Julian K Christians
- Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia, V5A 1S6, Canada. .,Centre for Cell Biology, Development and Disease, Simon Fraser University, Burnaby, Canada. .,British Columbia Children's Hospital Research Institute, Vancouver, BC, Canada. .,Women's Health Research Institute, BC Women's Hospital and Health Centre, Vancouver, British Columbia, Canada.
| | - Haroop K Shergill
- Department of Biological Sciences, Simon Fraser University, 8888 University Drive, Burnaby, British Columbia, V5A 1S6, Canada
| | - Arianne Y K Albert
- Women's Health Research Institute, BC Women's Hospital and Health Centre, Vancouver, British Columbia, Canada
| |
Collapse
|
45
|
Maternal High-Fiber Diet Protects Offspring against Type 2 Diabetes. Nutrients 2020; 13:nu13010094. [PMID: 33396680 PMCID: PMC7823372 DOI: 10.3390/nu13010094] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/27/2020] [Accepted: 12/28/2020] [Indexed: 02/08/2023] Open
Abstract
Previous studies have reported that maternal malnutrition is linked to increased risk of developing type 2 diabetes in adulthood. Although several diabetic risk factors associated with early-life environment have been identified, protective factors remain elusive. Here, we conducted a longitudinal study with 671 Nile rats whereby we examined the interplay between early-life environment (maternal diet) and later-life environment (offspring diet) using opposing diets that induce or prevent diet-induced diabetes. Specifically, we modulated the early-life environment throughout oogenesis, pregnancy, and nursing by feeding Nile rat dams a lifelong high-fiber diet to investigate whether the offspring are protected from type 2 diabetes. We found that exposure to a high-fiber maternal diet prior to weaning significantly lowered the risk of diet-induced diabetes in the offspring. Interestingly, offspring consuming a high-fiber diet after weaning did not develop diet-induced diabetes, even when exposed to a diabetogenic maternal diet. Here, we provide the first evidence that the protective effect of a high-fiber diet can be transmitted to the offspring through the maternal diet, which has important implications in diabetes prevention.
Collapse
|
46
|
Kim OY, Kim EM, Chung S. Impacts of Dietary Macronutrient Pattern on Adolescent Body Composition and Metabolic Risk: Current and Future Health Status-A Narrative Review. Nutrients 2020; 12:E3722. [PMID: 33276567 PMCID: PMC7761580 DOI: 10.3390/nu12123722] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/29/2020] [Accepted: 12/01/2020] [Indexed: 12/23/2022] Open
Abstract
Obesity, particularly in childhood and adolescence, is one of the serious public health problems worldwide. According to the World Health Organization, 10% of young people aged 5-17 years are obese, which is rapidly increasing around the world. Furthermore, approximately 80% of adolescents who become obese develop bodyweight-related health problems in adulthood. Eating habits and lifestyles play important roles in forming body composition and metabolic status. Changes in body composition in adolescence, the period in which secondary sex characteristics begin to develop, can alter hormonal and metabolic status, can consequently affect health status and the risk of developing chronic diseases in adulthood, and moreover may have an impact on probable body composition and metabolic status in the next generation. Here, we reviewed cross-sectional and interventional studies to analyze the role of dietary patterns focusing on macronutrient intake in growth, body composition, and metabolic changes in adolescents. These findings provide insights into optimal dietary guidelines for healthy growth with accretion of adequate body composition in adolescence, and provide an effective strategy for preventing and managing the risk of obesity-related metabolic disease in adulthood, with the additional benefit of providing potential benefits for the next generation's health.
Collapse
Affiliation(s)
- Oh Yoen Kim
- Department of Health Science, Graduate School, Dong-A University, Busan 49315, Korea;
- Department of Food Science and Nutrition, Dong-A University, Busan 49315, Korea
| | - Eun Mi Kim
- Dietetic Department, Kangbuk Samsung Hospital, Seoul 03181, Korea;
| | - Sochung Chung
- Department of Pediatrics, Konkuk University Medical Center, Konkuk University School of Medicine, Seoul 05030, Korea
| |
Collapse
|
47
|
Wu Y, Yin G, Wang P, Huang Z, Lin S. Effects of different diet-induced postnatal catch-up growth on glycolipid metabolism in intrauterine growth retardation male rats. Exp Ther Med 2020; 20:134. [PMID: 33082866 PMCID: PMC7560533 DOI: 10.3892/etm.2020.9263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 06/24/2020] [Indexed: 02/05/2023] Open
Abstract
A number of studies have reported the occurrence of long-term metabolic disorders in mammals following intrauterine growth retardation (IUGR). However, the effects of dietary patterns during IUGR have not been fully elucidated. The present study aimed to evaluate the effects of different dietary patterns during critical growth windows on metabolic outcomes in the offspring of rats with IUGR. Male offspring rats from mothers fed either a normal or low-protein diet were randomly assigned to one of the following groups: Normal diet throughout pregnancy, lactation and after weaning (CON); normal diet throughout pregnancy and high-fat diet throughout lactation and after weaning (N + H + H); low-protein diet throughout pregnancy and high-fat diet throughout lactation and after weaning (IUGR + H + H); low-protein diet throughout pregnancy and lactation and high-fat diet after weaning (IUGR + L + H); and low-protein diet throughout pregnancy and normal diet throughout lactation and after weaning. During lactation, the male offspring in the N + H + H group exhibited the fastest growth rate, whereas the slowest rate was in the IUGR + L + H group. Following weaning, all IUGR groups demonstrated significant catch-up growth. Abnormal insulin tolerance were observed in the N + H + H, IUGR + H + H and IUGR + L + H groups and insulin sensitivity was decreased in IUGR + L + H group. The triglycerides/high-density lipoprotein ratio in the IUGR + L + H group was significantly higher compared with in the other groups. The abdominal circumference, Lee's index and adipocyte diameter of IUGR groups were significantly increased compared with the CON group. High levels of leptin and interleukin-6 in adipose tissues, and low adiponectin were observed in the IUGR + L + H group. Different dietary patterns during specific growth windows showed numerous impacts on glycolipid metabolism in IUGR offspring. The present study elucidated the mechanisms and potential options for IUGR treatment and prevention.
Collapse
Affiliation(s)
- Yixi Wu
- Department of Endocrinology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, P.R. China
| | - Guoshu Yin
- Department of Endocrinology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, P.R. China
| | - Ping Wang
- Center of Reproductive Medicine, Shantou University Medical College, Shantou, Guangdong 515000, P.R. China
| | - Zhihua Huang
- Center of Reproductive Medicine, Shantou University Medical College, Shantou, Guangdong 515000, P.R. China
| | - Shaoda Lin
- Department of Endocrinology, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515000, P.R. China
- Correspondence to: Dr Shaoda Lin, Department of Endocrinology, The First Affiliated Hospital of Shantou University Medical College, 57 Changping Road, Shantou, Guangdong 515000, P.R. China
| |
Collapse
|
48
|
Maternal Malnutrition Affects Hepatic Metabolism through Decreased Hepatic Taurine Levels and Changes in HNF4A Methylation. Int J Mol Sci 2020; 21:ijms21239060. [PMID: 33260590 PMCID: PMC7729756 DOI: 10.3390/ijms21239060] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 11/22/2020] [Accepted: 11/25/2020] [Indexed: 12/16/2022] Open
Abstract
Fetal programming implies that the maternal diet during pregnancy affects the long-term health of offspring. Although maternal diet influences metabolic disorders and non-alcoholic fatty liver disease in offspring, the hepatic mechanisms related to metabolites are still unknown. Here, we investigated the maternal diet-related alterations in metabolites and the biological pathway in male offspring at three months of age. Pregnant rats were exposed to 50% food restriction during the prenatal period or a 45% high-fat diet during the prenatal and postnatal periods. The male offspring exposed to food restriction and high-fat diets had lower birth weights than controls, but had a catch-up growth spurt at three months of age. Hepatic taurine levels decreased in both groups compared to controls. The decreased hepatic taurine levels in offspring affected excessive lipid accumulation through changes in hepatocyte nuclear factor 4 A methylation. Moreover, the alteration of gluconeogenesis in offspring exposed to food restriction was observed to a similar extent as that of offspring exposed to a high fat diet. These results indicate that maternal diet affects the dysregulation in hepatic metabolism through changes in taurine levels and HNF4A methylation, and predisposes the offspring to Type 2 diabetes and non-alcoholic fatty liver disease in later life.
Collapse
|
49
|
Lang F, Rajaxavier J, Singh Y, Brucker SY, Salker MS. The Enigmatic Role of Serum & Glucocorticoid Inducible Kinase 1 in the Endometrium. Front Cell Dev Biol 2020; 8:556543. [PMID: 33195190 PMCID: PMC7609842 DOI: 10.3389/fcell.2020.556543] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 09/24/2020] [Indexed: 11/13/2022] Open
Abstract
The serum- and glucocorticoid-inducible kinase 1 (SGK1) is subject to genetic up-regulation by diverse stimulators including glucocorticoids, mineralocorticoids, dehydration, ischemia, radiation and hyperosmotic shock. To become active, the expressed kinase requires phosphorylation, which is accomplished by PI3K/PDK1 and mTOR dependent signaling. SGK1 enhances the expression/activity of various transport proteins including Na+/K+-ATPase as well as ion-, glucose-, and amino acid- carriers in the plasma membrane. SGK1 can further up-regulate diverse ion channels, such as Na+-, Ca2+-, K+- and Cl- channels. SGK1 regulates expression/activity of a wide variety of transcription factors (such as FKHRL1/Foxo3a, β-catenin, NFκB and p53). SGK1 thus contributes to the regulation of transport, glycolysis, angiogenesis, cell survival, immune regulation, cell migration, tissue fibrosis and tissue calcification. In this review we summarized the current findings that SGK1 plays a crucial function in the regulation of endometrial function. Specifically, it plays a dual role in the regulation of endometrial receptivity necessary for implantation and, subsequently in pregnancy maintenance. Furthermore, fetal programming of blood pressure regulation requires maternal SGK1. Underlying mechanisms are, however, still ill-defined and there is a substantial need for additional information to fully understand the role of SGK1 in the orchestration of embryo implantation, embryo survival and fetal programming.
Collapse
Affiliation(s)
- Florian Lang
- Department of Physiology, Eberhard-Karls University, Tübingen, Germany
| | - Janet Rajaxavier
- Research Institute of Women’s Health, Eberhard-Karls University, Tübingen, Germany
| | - Yogesh Singh
- Research Institute of Women’s Health, Eberhard-Karls University, Tübingen, Germany
- Institute of Medical Genetics and Applied Genomics, Eberhard-Karls University, Tübingen, Germany
| | - Sara Y. Brucker
- Research Institute of Women’s Health, Eberhard-Karls University, Tübingen, Germany
| | - Madhuri S. Salker
- Research Institute of Women’s Health, Eberhard-Karls University, Tübingen, Germany
| |
Collapse
|
50
|
De Guzman RM, Medina J, Saulsbery AI, Workman JL. Rotated nursing environment with underfeeding: A form of early-life adversity with sex- and age-dependent effects on coping behavior and hippocampal neurogenesis. Physiol Behav 2020; 225:113106. [PMID: 32717197 DOI: 10.1016/j.physbeh.2020.113106] [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: 01/28/2020] [Revised: 07/07/2020] [Accepted: 07/24/2020] [Indexed: 01/06/2023]
Abstract
We investigated how a unique form of early-life adversity (ELA), caused by rotated nursing environment to induce underfeeding, alters anxiety-like and stress-coping behaviors in male and female Sprague Dawley rats in adolescence and adulthood. Adult female rats underwent either thelectomy (thel; surgical removal of teats), sham surgery, or no surgery (control) before mating. Following parturition, litters were rotated between sham and thel rats every 12 h to generate a group of rats that experienced ELA (rotated housing, rotated mother, and 50% food restriction) from postnatal day 0 to 26. Control litters remained with their natal, nursing dams. Regardless of age and sex, ELA reduced activity in the periphery of the open field. ELA increased immobility in the forced swim test, particularly in adults. We used doublecortin immunohistochemistry to identify immature neurons in the hippocampus. ELA increased the number and density of immature neurons in the dentate gyrus of adolescent males (but not females) and reduced the density of immature neurons in adult males (but not females). This research indicates that a unique form of ELA alters stress-related passive coping and hippocampal neurogenesis in an age- and sex-dependent manner.
Collapse
Affiliation(s)
- Rose M De Guzman
- Department of Psychology, University at Albany, State University of New York, 1400 Washington Ave. Albany, NY 12222 United States
| | - Joanna Medina
- Department of Psychology, University at Albany, State University of New York, 1400 Washington Ave. Albany, NY 12222 United States
| | - Angela I Saulsbery
- Department of Psychology, University at Albany, State University of New York, 1400 Washington Ave. Albany, NY 12222 United States
| | - Joanna L Workman
- Department of Psychology, University at Albany, State University of New York, 1400 Washington Ave. Albany, NY 12222 United States; Center for Neuroscience Research, University at Albany, State University of New York, 1400 Washington Ave. Albany, NY 12222, United States.
| |
Collapse
|