1
|
Ahmadi S, Ohkubo T. A Bird's-Eye Overview of Leptin and Female Reproduction -with Mammalian Comparisons. J Poult Sci 2025; 62:2025007. [PMID: 39916995 PMCID: PMC11794366 DOI: 10.2141/jpsa.2025007] [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: 10/23/2024] [Accepted: 01/10/2025] [Indexed: 02/09/2025] Open
Abstract
Leptin, a key regulator of reproductive physiology, influences various processes in vertebrates, including oocyte proliferation, embryogenesis, the onset of puberty, ovarian function, and follicle development. In mammals, leptin affects steroidogenesis, folliculogenesis, and hormonal regulation through the hypothalamic-pituitary-gonadal axis. Instead, in avian species, leptin-controlled mechanisms are poorly understood, because birds do not produce leptin in adipocytes. In birds, leptin is expressed in the brain, pituitary glands, and gonads, where it enhances ovarian function and egg-laying performance, particularly during feed deprivation. In this review, we discuss and summarize the recently discovered role of leptin in regulating ovarian function during different life stages in birds and compare it with its function in mammals.
Collapse
Affiliation(s)
- Sadequllah Ahmadi
- College of Agriculture, Ibaraki University, 3-21-1 Chuo, Ami, Ibaraki 300-0393, Japan
- Faculty of Animal Science, Afghanistan National Agricultural Sciences and Technology University (ANASTU), Kandahar 3801, Afghanistan
| | - Takeshi Ohkubo
- College of Agriculture, Ibaraki University, 3-21-1 Chuo, Ami, Ibaraki 300-0393, Japan
| |
Collapse
|
2
|
Laule C, Rahmouni K. Leptin and Associated Neural Pathways Underlying Obesity-Induced Hypertension. Compr Physiol 2025; 15:e8. [PMID: 40293220 PMCID: PMC12038170 DOI: 10.1002/cph4.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 10/24/2024] [Accepted: 11/05/2024] [Indexed: 04/30/2025]
Abstract
Obesity rates have surged to pandemic levels, placing tremendous burden on our society. This chronic and complex disease is related to the development of many life-threatening illnesses including cardiovascular diseases. Hypertension caused by obesity increases the risk for cardiovascular mortality and morbidity by promoting stroke, myocardial infarction, congestive heart failure, and end-stage renal disease. Overwhelming evidence supports neural origins for obesity-induced hypertension and pinpoints the adipose-derived hormone, leptin, and the sympathetic nervous system as major causal factors. Hyperleptinemia in obesity is associated with selective leptin resistance where leptin's renal sympathoexcitatory and pressor effects are preserved while the metabolic actions are impaired. Understanding the mechanisms driving this phenomenon is critical for developing effective therapeutics. This review describes the neural mechanisms of obesity-induced hypertension with a focus on the molecular and neuronal substrates of leptin action.
Collapse
Affiliation(s)
- Connor Laule
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Kamal Rahmouni
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Obesity Research and Education Initiative, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Iowa Neuroscience Institute, University of Iowa Carver College of Medicine, Iowa City, Iowa
- Veterans Affairs Health Care System, Iowa City, Iowa
| |
Collapse
|
3
|
Gusmao DO, de Sousa LMM, de Sousa ME, Rusew SJR, List EO, Kopchick JJ, Gomes AF, Campideli-Santana AC, Szawka RE, Donato J. Characterization and Regulation of the Neonatal Growth Hormone Surge. Endocrinology 2024; 165:bqae140. [PMID: 39446366 PMCID: PMC11544317 DOI: 10.1210/endocr/bqae140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Indexed: 11/09/2024]
Abstract
High neonatal growth hormone (GH) secretion has been described in several species. However, the neuroendocrine mechanisms behind this surge remain unknown. Thus, the pattern of postnatal GH secretion was investigated in mice and rats. Blood GH levels were very high on postnatal day (P)1 and progressively decreased until near zero by P17 in C57BL/6 mice without sex differences. This pattern was similar to that observed in rats, except that female rats showed higher GH levels on P1 than males. In comparison, follicle-stimulating hormone exhibited higher secretion in females during the first 3 weeks of life. Hypothalamic Sst mRNA and somatostatin neuroendocrine terminals in the median eminence were higher in P20/P21 mice than in newborns. Knockout mice for GH-releasing hormone (GHRH) receptor showed no GH surge, whereas knockdown mice for the Sst gene displayed increased neonatal GH peak. Leptin deficiency caused only minor effects on early-life GH secretion. GH receptor ablation in neurons or the entire body did not affect neonatal GH secretion, but the subsequent reduction in blood GH levels was attenuated or prevented by these genetic manipulations, respectively. This phenotype was also observed in knockout mice for the insulin-like growth factor-1 (IGF-1) receptor in GHRH neurons. Moreover, glucose-induced hyperglycemia overstimulated GH secretion in neonatal mice. In conclusion, GH surge in the first days of life is not regulated by negative feedback loops. However, neonatal GH secretion requires GHRH receptor, and is modulated by somatostatin and blood glucose levels, suggesting that this surge is controlled by hypothalamic-pituitary communication.
Collapse
MESH Headings
- Animals
- Female
- Growth Hormone/metabolism
- Growth Hormone/blood
- Animals, Newborn
- Male
- Mice, Knockout
- Mice, Inbred C57BL
- Somatostatin/metabolism
- Somatostatin/genetics
- Mice
- Receptor, IGF Type 1/metabolism
- Receptor, IGF Type 1/genetics
- Rats
- Receptors, Neuropeptide/genetics
- Receptors, Neuropeptide/metabolism
- Leptin/blood
- Leptin/metabolism
- Hypothalamus/metabolism
- Receptors, Pituitary Hormone-Regulating Hormone/genetics
- Receptors, Pituitary Hormone-Regulating Hormone/metabolism
- Growth Hormone-Releasing Hormone/metabolism
- Growth Hormone-Releasing Hormone/genetics
- Receptors, Somatotropin/genetics
- Receptors, Somatotropin/metabolism
- Follicle Stimulating Hormone/blood
- Follicle Stimulating Hormone/metabolism
- Insulin-Like Growth Factor I/metabolism
- Insulin-Like Growth Factor I/genetics
Collapse
Affiliation(s)
- Daniela O Gusmao
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, SP 05508-000, Brazil
| | - Ligia M M de Sousa
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, SP 05508-000, Brazil
| | - Maria E de Sousa
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, SP 05508-000, Brazil
| | - Stephanie J R Rusew
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, SP 05508-000, Brazil
| | - Edward O List
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - John J Kopchick
- Edison Biotechnology Institute and Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Andre F Gomes
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Ana C Campideli-Santana
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Raphael E Szawka
- Departamento de Fisiologia e Biofisica, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270-901, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de São Paulo, São Paulo, SP 05508-000, Brazil
| |
Collapse
|
4
|
Chen L, Liu LM, Guo M, Du Y, Chen YW, Xiong XY, Cheng Y. Altered leptin level in autism spectrum disorder and meta-analysis of adipokines. BMC Psychiatry 2024; 24:479. [PMID: 38951775 PMCID: PMC11218410 DOI: 10.1186/s12888-024-05936-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024] Open
Abstract
BACKGROUND Increasing evidence suggests that leptin is involved in the pathology of autism spectrum disorder (ASD). In this study, our objective was to investigate the levels of leptin in the blood of children with ASD and to examine the overall profile of adipokine markers in ASD through meta-analysis. METHODS Leptin concentrations were measured using an enzyme-linked immunosorbent assay (ELISA) kit, while adipokine profiling, including leptin, was performed via meta-analysis. Original reports that included measurements of peripheral adipokines in ASD patients and healthy controls (HCs) were collected from databases such as Web of Science, PubMed, and Cochrane Library. These studies were collected from September 2022 to September 2023 and followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Standardized mean differences were calculated using a random effects model for the meta-analysis. Additionally, we performed meta-regression and explored heterogeneity among studies. RESULTS Our findings revealed a significant increase in leptin levels in children with ASD compared to HCs (p = 0.0319). This result was consistent with the findings obtained from the meta-analysis (p < 0.001). Furthermore, progranulin concentrations were significantly reduced in children with ASD. However, for the other five adipokines analyzed, there were no significant differences observed between the children with ASD and HCs children. Heterogeneity was found among the studies, and the meta-regression analysis indicated that publication year and latitude might influence the results of the meta-analysis. CONCLUSIONS These findings provide compelling evidence that leptin levels are increased in children with ASD compared to healthy controls, suggesting a potential mechanism involving adipokines, particularly leptin, in the pathogenesis of ASD. These results contribute to a better understanding of the pathology of ASD and provide new insights for future investigations.
Collapse
Affiliation(s)
- Lei Chen
- Key Laboratory of Ethnomedicine of Ministry of Education, Center On Translational Neuroscience, School of Pharmacy, Minzu University of China, 27 South Zhongguancun Avenue, Beijing, 100081, China
| | - Li-Ming Liu
- Institute of National Security, Minzu University of China, Beijing, China
| | - Mei Guo
- Key Laboratory of Ethnomedicine of Ministry of Education, Center On Translational Neuroscience, School of Pharmacy, Minzu University of China, 27 South Zhongguancun Avenue, Beijing, 100081, China
| | - Yang Du
- Key Laboratory of Ethnomedicine of Ministry of Education, Center On Translational Neuroscience, School of Pharmacy, Minzu University of China, 27 South Zhongguancun Avenue, Beijing, 100081, China
| | - Yue-Wen Chen
- Chinese Academy of Sciences Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute, Shenzhen Key Laboratory of Translational Research for Brain Diseases, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen,, 518055, Guangdong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, HKUST Shenzhen Research Institute, Shenzhen, 518057, Guangdong, China
| | - Xi-Yue Xiong
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008, China.
| | - Yong Cheng
- Key Laboratory of Ethnomedicine of Ministry of Education, Center On Translational Neuroscience, School of Pharmacy, Minzu University of China, 27 South Zhongguancun Avenue, Beijing, 100081, China.
- Institute of National Security, Minzu University of China, Beijing, China.
- NHC Key Laboratory of Birth Defect for Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, 410008, China.
| |
Collapse
|
5
|
Paul B, Buchholz DR. Minireview: Glucocorticoid-Leptin Crosstalk: Role of Glucocorticoid-Leptin Counterregulation in Metabolic Homeostasis and Normal Development. Integr Comp Biol 2023; 63:1127-1139. [PMID: 37708034 DOI: 10.1093/icb/icad119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 08/16/2023] [Accepted: 08/30/2023] [Indexed: 09/16/2023] Open
Abstract
Glucocorticoids and leptin are two important hormones that regulate metabolic homeostasis by controlling appetite and energy expenditure in adult mammals. Also, glucocorticoids and leptin strongly counterregulate each other, such that chronic stress-induced glucocorticoids upregulate the production of leptin and leptin suppresses glucocorticoid production directly via action on endocrine organs and indirectly via action on food intake. Altered glucocorticoid or leptin levels during development can impair organ development and increase the risk of chronic diseases in adults, but there are limited studies depicting the significance of glucocorticoid-leptin interaction during development and its impact on developmental programming. In mammals, leptin-induced suppression of glucocorticoid production is critical during development, where leptin prevents stress-induced glucocorticoid production by inducing a period of short-hyporesponsiveness when the adrenal glands fail to respond to certain mild to moderate stressors. Conversely, reduced or absent leptin signaling increases glucocorticoid levels beyond what is appropriate for normal organogenesis. The counterregulatory interactions between leptin and glucocorticoids suggest the potential significant involvement of leptin in disorders that occur from stress during development.
Collapse
Affiliation(s)
- Bidisha Paul
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Daniel R Buchholz
- Department of Biological Sciences, University of Cincinnati, Cincinnati, OH 45221, USA
| |
Collapse
|
6
|
Ullah R, Shen Y, Zhou YD, Fu J. Perinatal metabolic inflammation in the hypothalamus impairs the development of homeostatic feeding circuitry. Metabolism 2023; 147:155677. [PMID: 37543245 DOI: 10.1016/j.metabol.2023.155677] [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: 05/22/2023] [Revised: 07/14/2023] [Accepted: 08/01/2023] [Indexed: 08/07/2023]
Abstract
Over the past few decades, there has been a global increase in childhood obesity. This rise in childhood obesity contributes to the susceptibility of impaired metabolism during both childhood and adulthood. The hypothalamus, specifically the arcuate nucleus (ARC), houses crucial neurons involved in regulating homeostatic feeding. These neurons include proopiomelanocortin (POMC) and agouti-related peptide (AGRP) secreting neurons. They play a vital role in sensing nutrients and metabolic hormones like insulin, leptin, and ghrelin. The neurogenesis of AGRP and POMC neurons completes at birth; however, axon development and synapse formation occur during the postnatal stages in rodents. Insulin, leptin, and ghrelin are the essential regulators of POMC and AGRP neurons. Maternal obesity and postnatal overfeeding or a high-fat diet (HFD) feeding cause metabolic inflammation, disrupted signaling of metabolic hormones, netrin-1, and neurogenic factors, neonatal obesity, and defective neuronal development in animal models; however, the mechanism is unclear. Within the hypothalamus and other brain areas, there exists a wide range of interconnected neuronal populations that regulate various aspects of feeding. However, this review aims to discuss how perinatal metabolic inflammation influences the development of POMC and AGRP neurons within the hypothalamus.
Collapse
Affiliation(s)
- Rahim Ullah
- Department of Endocrinology, Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, 310052, China; Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Brain Science and Brain Medicine, Hangzhou, China
| | - Yi Shen
- Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Brain Science and Brain Medicine, Hangzhou, China.
| | - Yu-Dong Zhou
- Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Brain Science and Brain Medicine, Hangzhou, China.
| | - Junfen Fu
- Department of Endocrinology, Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, Zhejiang, 310052, China.
| |
Collapse
|
7
|
Martinez ME, Wu Z, Hernandez A. Paternal developmental thyrotoxicosis disrupts neonatal leptin leading to increased adiposity and altered physiology of the melanocortin system. Front Endocrinol (Lausanne) 2023; 14:1210414. [PMID: 37560296 PMCID: PMC10407661 DOI: 10.3389/fendo.2023.1210414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 07/10/2023] [Indexed: 08/11/2023] Open
Abstract
Background The genetic code does not fully explain individual variability and inheritance of susceptibility to endocrine conditions, suggesting the contribution of epigenetic factors acting across generations. Methods We used a mouse model of developmental thyrotoxicosis (Dio3-/- mouse) to analyze endocrine outcomes in the adult offspring of Dio3-/- males using standard methods for body composition, and baseline and fasting hormonal and gene expression determinations in serum and tissues of relevance to the control of energy balance. Results Compared to controls, adult females with an exposed father (EF females) exhibited higher body weight and fat mass, but not lean mass, a phenotype that was much milder in EF males. After fasting, both EF females and males exhibited a more pronounced decrease in body weight than controls. EF females also showed markedly elevated serum leptin, increased white adipose tissue mRNA expression of leptin and mesoderm-specific transcript but decreased expression of type 2 deiodinase. EF females exhibited decreased serum ghrelin, which showed more pronounced post-fasting changes in EF females than in control females. EF female hypothalami also revealed significant decreases in the expression of pro-opiomelanocortin, agouti-related peptide, neuropeptide Y and melanocortin receptor 4. These markers also showed larger changes in response to fasting in EF females than in control females. Adult EF females showed no abnormalities in serum thyroid hormones, but pituitary expression of thyrotropin-releasing hormone receptor 1 and thyroid gland expression of thyroid-stimulating hormone receptor, thyroid peroxidase and iodotyrosine deiodinase were increased at baseline and showed differential regulation after fasting, with no increase in Trhr1 expression and more pronounced reductions in Tshr, Tpo and Iyd. In EF males, these abnormalities were generally milder. In addition, postnatal day 14 (P14) serum leptin was markedly reduced in EF pups. Discussion A paternal excess of thyroid hormone during development modifies the endocrine programming and energy balance in the offspring in a sexually dimorphic manner, with baseline and dynamic range alterations in the leptin-melanocortin system and thyroid gland, and consequences for adiposity phenotypes. We conclude that thyroid hormone overexposure may have important implications for the non-genetic, inherited etiology of endocrine and metabolic pathologies.
Collapse
Affiliation(s)
- Maria Elena Martinez
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, United States
| | - Zhaofei Wu
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, United States
| | - Arturo Hernandez
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, United States
- Graduate School for Biomedical Sciences and Engineering, University of Maine, Orono, ME, United States
- Department of Medicine, Tufts University School of Medicine, Boston, MA, United States
| |
Collapse
|
8
|
Frick JM, Eller OC, Foright RM, Levasseur BM, Yang X, Wang R, Winter MK, O'Neil MF, Morris EM, Thyfault JP, Christianson JA. High-fat/high-sucrose diet worsens metabolic outcomes and widespread hypersensitivity following early-life stress exposure in female mice. Am J Physiol Regul Integr Comp Physiol 2023; 324:R353-R367. [PMID: 36693166 PMCID: PMC9970659 DOI: 10.1152/ajpregu.00216.2022] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 12/21/2022] [Accepted: 01/13/2023] [Indexed: 01/25/2023]
Abstract
Exposure to stress early in life has been associated with adult-onset comorbidities such as chronic pain, metabolic dysregulation, obesity, and inactivity. We have established an early-life stress model using neonatal maternal separation (NMS) in mice, which displays evidence of increased body weight and adiposity, widespread mechanical allodynia, and hypothalamic-pituitary-adrenal axis dysregulation in male mice. Early-life stress and consumption of a Western-style diet contribute to the development of obesity; however, relatively few preclinical studies have been performed in female rodents, which are known to be protected against diet-induced obesity and metabolic dysfunction. In this study, we gave naïve and NMS female mice access to a high-fat/high-sucrose (HFS) diet beginning at 4 wk of age. Robust increases in body weight and fat were observed in HFS-fed NMS mice during the first 10 wk on the diet, driven partly by increased food intake. Female NMS mice on an HFS diet showed widespread mechanical hypersensitivity compared with either naïve mice on an HFS diet or NMS mice on a control diet. HFS diet-fed NMS mice also had impaired glucose tolerance and fasting hyperinsulinemia. Strikingly, female NMS mice on an HFS diet showed evidence of hepatic steatosis with increased triglyceride levels and altered glucocorticoid receptor levels and phosphorylation state. They also exhibited increased energy expenditure as observed via indirect calorimetry and expression of proinflammatory markers in perigonadal adipose. Altogether, our data suggest that early-life stress exposure increased the susceptibility of female mice to develop diet-induced metabolic dysfunction and pain-like behaviors.
Collapse
Affiliation(s)
- Jenna M Frick
- Department of Cell Biology and Physiology, School of Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Olivia C Eller
- Department of Cell Biology and Physiology, School of Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Rebecca M Foright
- Department of Cell Biology and Physiology, School of Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Brittni M Levasseur
- Department of Cell Biology and Physiology, School of Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Xiaofang Yang
- Department of Cell Biology and Physiology, School of Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Ruipeng Wang
- Department of Cell Biology and Physiology, School of Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Michelle K Winter
- Kansas Intellectual and Developmental Disabilities Research Association, School of Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Maura F O'Neil
- Department of Pathology and Laboratory Medicine, School of Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - E Matthew Morris
- Department of Cell Biology and Physiology, School of Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - John P Thyfault
- Department of Cell Biology and Physiology, School of Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States
- Research Service, Kansas City Veterans Affairs Medical Center, Kansas City, Kansas, United States
| | - Julie A Christianson
- Department of Cell Biology and Physiology, School of Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States
| |
Collapse
|
9
|
Plakkot B, Di Agostino A, Subramanian M. Implications of Hypothalamic Neural Stem Cells on Aging and Obesity-Associated Cardiovascular Diseases. Cells 2023; 12:cells12050769. [PMID: 36899905 PMCID: PMC10000584 DOI: 10.3390/cells12050769] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/14/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
The hypothalamus, one of the major regulatory centers in the brain, controls various homeostatic processes, and hypothalamic neural stem cells (htNSCs) have been observed to interfere with hypothalamic mechanisms regulating aging. NSCs play a pivotal role in the repair and regeneration of brain cells during neurodegenerative diseases and rejuvenate the brain tissue microenvironment. The hypothalamus was recently observed to be involved in neuroinflammation mediated by cellular senescence. Cellular senescence, or systemic aging, is characterized by a progressive irreversible state of cell cycle arrest that causes physiological dysregulation in the body and it is evident in many neuroinflammatory conditions, including obesity. Upregulation of neuroinflammation and oxidative stress due to senescence has the potential to alter the functioning of NSCs. Various studies have substantiated the chances of obesity inducing accelerated aging. Therefore, it is essential to explore the potential effects of htNSC dysregulation in obesity and underlying pathways to develop strategies to address obesity-induced comorbidities associated with brain aging. This review will summarize hypothalamic neurogenesis associated with obesity and prospective NSC-based regenerative therapy for the treatment of obesity-induced cardiovascular conditions.
Collapse
|
10
|
Langley-Evans SC. Early life programming of health and disease: the long-term consequences of obesity in pregnancy: a narrative review. J Hum Nutr Diet 2022; 35:816-832. [PMID: 35475555 PMCID: PMC9540012 DOI: 10.1111/jhn.13023] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/06/2022] [Indexed: 11/28/2022]
Abstract
The prevalence of overweight and obesity is rising in all parts of the world and among young women it presents a very clear danger during pregnancy. Women who are overweight or who gain excessive weight during pregnancy are at greater risk of complications in pregnancy and labour, and are more likely to lose their child to stillbirth, or themselves die during pregnancy. This narrative review considers the evidence that in addition to increasing risk of poor pregnancy outcomes, obesity has the capacity to programme fetuses to be at greater risk of cardiometabolic disorders later in life. An extensive body of evidence from prospective and retrospective cohorts, and record linkage studies demonstrates associations of maternal obesity and/or gestational diabetes with cardiovascular disease, type-1 and type-2 diabetes. Studies in animals suggest that these associations are underpinned by adaptations that occur in fetal life, which remodel the structures of major organs including the brain, kidney and pancreas. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Simon C Langley-Evans
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, LE12 5RD
| |
Collapse
|
11
|
Lustig RH, Collier D, Kassotis C, Roepke TA, Ji Kim M, Blanc E, Barouki R, Bansal A, Cave MC, Chatterjee S, Choudhury M, Gilbertson M, Lagadic-Gossmann D, Howard S, Lind L, Tomlinson CR, Vondracek J, Heindel JJ. Obesity I: Overview and molecular and biochemical mechanisms. Biochem Pharmacol 2022; 199:115012. [PMID: 35393120 PMCID: PMC9050949 DOI: 10.1016/j.bcp.2022.115012] [Citation(s) in RCA: 90] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 03/12/2022] [Accepted: 03/15/2022] [Indexed: 02/06/2023]
Abstract
Obesity is a chronic, relapsing condition characterized by excess body fat. Its prevalence has increased globally since the 1970s, and the number of obese and overweight people is now greater than those underweight. Obesity is a multifactorial condition, and as such, many components contribute to its development and pathogenesis. This is the first of three companion reviews that consider obesity. This review focuses on the genetics, viruses, insulin resistance, inflammation, gut microbiome, and circadian rhythms that promote obesity, along with hormones, growth factors, and organs and tissues that control its development. It shows that the regulation of energy balance (intake vs. expenditure) relies on the interplay of a variety of hormones from adipose tissue, gastrointestinal tract, pancreas, liver, and brain. It details how integrating central neurotransmitters and peripheral metabolic signals (e.g., leptin, insulin, ghrelin, peptide YY3-36) is essential for controlling energy homeostasis and feeding behavior. It describes the distinct types of adipocytes and how fat cell development is controlled by hormones and growth factors acting via a variety of receptors, including peroxisome proliferator-activated receptor-gamma, retinoid X, insulin, estrogen, androgen, glucocorticoid, thyroid hormone, liver X, constitutive androstane, pregnane X, farnesoid, and aryl hydrocarbon receptors. Finally, it demonstrates that obesity likely has origins in utero. Understanding these biochemical drivers of adiposity and metabolic dysfunction throughout the life cycle lends plausibility and credence to the "obesogen hypothesis" (i.e., the importance of environmental chemicals that disrupt these receptors to promote adiposity or alter metabolism), elucidated more fully in the two companion reviews.
Collapse
Affiliation(s)
- Robert H Lustig
- Division of Endocrinology, Department of Pediatrics, University of California, San Francisco, CA 94143, United States
| | - David Collier
- Brody School of Medicine, East Carolina University, Greenville, NC 27834, United States
| | - Christopher Kassotis
- Institute of Environmental Health Sciences and Department of Pharmacology, Wayne State University, Detroit, MI 48202, United States
| | - Troy A Roepke
- School of Environmental and Biological Sciences, Rutgers University, New Brunswick, NJ 08901, United States
| | - Min Ji Kim
- Department of Biochemistry and Toxicology, University of Paris, INSERM U1224 (T3S), 75006 Paris, France
| | - Etienne Blanc
- Department of Biochemistry and Toxicology, University of Paris, INSERM U1224 (T3S), 75006 Paris, France
| | - Robert Barouki
- Department of Biochemistry and Toxicology, University of Paris, INSERM U1224 (T3S), 75006 Paris, France
| | - Amita Bansal
- College of Health & Medicine, Australian National University, Canberra, Australia
| | - Matthew C Cave
- Division of Gastroenterology, Hepatology and Nutrition, University of Louisville, Louisville, KY 40402, United States
| | - Saurabh Chatterjee
- Environmental Health and Disease Laboratory, University of South Carolina, Columbia, SC 29208, United States
| | - Mahua Choudhury
- College of Pharmacy, Texas A&M University, College Station, TX 77843, United States
| | - Michael Gilbertson
- Occupational and Environmental Health Research Group, University of Stirling, Stirling, Scotland, United Kingdom
| | - Dominique Lagadic-Gossmann
- Research Institute for Environmental and Occupational Health, University of Rennes, INSERM, EHESP, Rennes, France
| | - Sarah Howard
- Healthy Environment and Endocrine Disruptor Strategies, Commonweal, Bolinas, CA 92924, United States
| | - Lars Lind
- Department of Medical Sciences, University of Uppsala, Uppsala, Sweden
| | - Craig R Tomlinson
- Norris Cotton Cancer Center, Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Lebanon, NH 03756, United States
| | - Jan Vondracek
- Department of Cytokinetics, Institute of Biophysics of the Czech Academy of Sciences, Brno, Czech Republic
| | - Jerrold J Heindel
- Healthy Environment and Endocrine Disruptor Strategies, Commonweal, Bolinas, CA 92924, United States.
| |
Collapse
|
12
|
Leptin enhances social motivation and reverses chronic unpredictable stress-induced social anhedonia during adolescence. Mol Psychiatry 2022; 27:4948-4958. [PMID: 36138127 PMCID: PMC9763124 DOI: 10.1038/s41380-022-01778-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/12/2022] [Accepted: 09/02/2022] [Indexed: 01/19/2023]
Abstract
Social anhedonia, a loss of interest and pleasure in social interactions, is a common symptom of major depression as well as other psychiatric disorders. Depression can occur at any age, but typically emerges in adolescence or early adulthood, which represents a sensitive period for social interaction that is vulnerable to stress. In this study, we evaluated social interaction reward using a conditioned place preference (CPP) paradigm in adolescent male and female mice. Adolescent mice of both sexes exhibited a preference for the social interaction-associated context. Chronic unpredictable stress (CUS) impaired the development of CPP for social interaction, mimicking social anhedonia in depressed adolescents. Conversely, administration of leptin, an adipocyte-derived hormone, enhanced social interaction-induced CPP in non-stressed control mice and reversed social anhedonia in CUS mice. By dissecting the motivational processes of social CPP into social approach and isolation avoidance components, we demonstrated that leptin treatment increased isolation aversion without overt social reward effect. Further mechanistic exploration revealed that leptin stimulated oxytocin gene transcription in the paraventricular nucleus of the hypothalamus, while oxytocin receptor blockade abolished the leptin-induced enhancement of socially-induced CPP. These results establish that chronic unpredictable stress can be used to study social anhedonia in adolescent mice and provide evidence that leptin modulates social motivation possibly via increasing oxytocin synthesis and oxytocin receptor activation.
Collapse
|
13
|
Marchlewicz E, McCabe C, Djuric Z, Hoenerhoff M, Barks J, Tang L, Song PX, Peterson K, Padmanabhan V, Dolinoy DC. Gestational exposure to high fat diets and bisphenol A alters metabolic outcomes in dams and offspring, but produces hepatic steatosis only in dams. CHEMOSPHERE 2022; 286:131645. [PMID: 34426127 PMCID: PMC8595757 DOI: 10.1016/j.chemosphere.2021.131645] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 06/29/2021] [Accepted: 07/21/2021] [Indexed: 05/07/2023]
Abstract
The prevalence of non-alcoholic fatty liver disease (NAFLD) is increasing worldwide. Perinatal development is a critical window for altered, lifelong health trajectory, and evidence supports the role of perinatal programming in chronic metabolic diseases. To examine the impact of diet and bisphenol A (BPA) on the developmental trajectory of NAFLD in offspring, we exposed dams from pre-gestation through lactation to a human-relevant dose of oral BPA coupled with intake of high fat Western or Mediterranean-style diets. We assessed hepatic steatosis by quantifying hepatic triglycerides (TGs) and metabolic health by measuring body weight, relative organ weights, and serum hormone levels in dams and offspring at postnatal day 10 (PND10) and 10-months of age. In dams, consumption of the Western or Mediterranean diet increased hepatic TGs 1.7-2.4-fold, independent of BPA intake. Among offspring, both perinatal diet and BPA exposure had a greater impact on metabolic outcomes than on hepatic steatosis. At PND10, serum leptin levels were elevated 2.6-4.8-fold in pups exposed to the Mediterranean diet, with a trend for sex-specific effects on body and organ weights. At 10-months, sex-specific increases in organ weight and hormone levels were observed in mice perinatally exposed to Western + BPA or Mediterranean + BPA. These findings suggest lifestage-specific interaction of perinatal exposures to experimental diets and BPA on offspring metabolic health without effects on NAFLD later in life. Importantly, alterations in dam phenotype by diet and BPA exposure appear to impact offspring health trajectory, emphasizing the need to define dam diet in assessing effects of environmental exposures on offspring health.
Collapse
Affiliation(s)
- Elizabeth Marchlewicz
- Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Carolyn McCabe
- Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Zora Djuric
- Department of Family Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Mark Hoenerhoff
- In Vivo Animal Core, Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - John Barks
- Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Lu Tang
- Department of Biostatistics, University of Pittsburgh, Pittsburg, PA, USA
| | - Peter X Song
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Karen Peterson
- Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA; Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Vasantha Padmanabhan
- Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA; Department of Pediatrics, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Obstetrics and Gynecology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Dana C Dolinoy
- Environmental Health Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA; Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA.
| |
Collapse
|
14
|
Vickers MH. Early life nutrition and neuroendocrine programming. Neuropharmacology 2021; 205:108921. [PMID: 34902348 DOI: 10.1016/j.neuropharm.2021.108921] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 12/06/2021] [Accepted: 12/08/2021] [Indexed: 12/26/2022]
Abstract
Alterations in the nutritional environment in early life can significantly increase the risk for obesity and a range of development of metabolic disorders in offspring in later life, effects that can be passed onto future generations. This process, termed development programming, provides the framework of the developmental origins of health and disease (DOHaD) paradigm. Early life nutritional compromise including undernutrition, overnutrition or specific macro/micronutrient deficiencies, results in a range of adverse health outcomes in offspring that can be further exacerbated by a poor postnatal nutritional environment. Although the mechanisms underlying programming remain poorly defined, a common feature across the phenotypes displayed in preclinical models is that of altered wiring of neuroendocrine circuits that regulate satiety and energy balance. As such, altered maternal nutritional exposures during critical early periods of developmental plasticity can result in aberrant hardwiring of these circuits with lasting adverse consequences for the offspring. There is also increasing evidence around the role of an altered epigenome and the gut-brain axis in mediating some of the central programming effects observed. Further, although such programming was once considered to result in a permanent change in developmental trajectory, there is evidence, at least from preclinical models, that programming can be reversed via targeted nutritional manipulations during early development. Further work is required at a mechanistic level to allow for identification for early markers of later disease risk, delineation of sex-specific effects and pathways to implementation of strategies aimed at breaking the transgenerational transmission of disease.
Collapse
Affiliation(s)
- M H Vickers
- Liggins Institute, University of Auckland, 85 Park Road, Grafton, Auckland, 1142, New Zealand.
| |
Collapse
|
15
|
Kabahizi A, Wallace B, Lieu L, Chau D, Dong Y, Hwang ES, Williams KW. Glucagon-like peptide-1 (GLP-1) signalling in the brain: From neural circuits and metabolism to therapeutics. Br J Pharmacol 2021; 179:600-624. [PMID: 34519026 PMCID: PMC8820188 DOI: 10.1111/bph.15682] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 08/23/2021] [Accepted: 08/27/2021] [Indexed: 12/18/2022] Open
Abstract
Glucagon‐like‐peptide‐1 (GLP‐1) derived from gut enteroendocrine cells and a discrete population of neurons in the caudal medulla acts through humoral and neural pathways to regulate satiety, gastric motility and pancreatic endocrine function. These physiological attributes contribute to GLP‐1 having a potent therapeutic action in glycaemic regulation and chronic weight management. In this review, we provide an overview of the neural circuits targeted by endogenous versus exogenous GLP‐1 and related drugs. We also highlight candidate subpopulations of neurons and cellular mechanisms responsible for the acute and chronic effects of GLP‐1 and GLP‐1 receptor agonists on energy balance and glucose metabolism. Finally, we present potential future directions to translate these findings towards the development of effective therapies for treatment of metabolic disease.
Collapse
Affiliation(s)
- Anita Kabahizi
- Department of Internal Medicine, Center for Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Briana Wallace
- Department of Internal Medicine, Center for Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Linh Lieu
- Department of Internal Medicine, Center for Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Dominic Chau
- Department of Internal Medicine, Center for Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Yanbin Dong
- Department of Internal Medicine, Center for Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Eun-Sang Hwang
- Department of Internal Medicine, Center for Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| | - Kevin W Williams
- Department of Internal Medicine, Center for Hypothalamic Research, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas, USA
| |
Collapse
|
16
|
Walczak K, Sieminska L. Obesity and Thyroid Axis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18189434. [PMID: 34574358 PMCID: PMC8467528 DOI: 10.3390/ijerph18189434] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/28/2021] [Accepted: 09/03/2021] [Indexed: 12/19/2022]
Abstract
Development of obesity is primarily the result of imbalance between energy intake and energy expenditure. Thyroid hormones influence energy expenditure by regulating cellular respiration and thermogenesis and by determining resting metabolic rate. Triiodothyronine influences lipid turnover in adipocytes and impacts appetite regulation through the central nervous system, mainly the hypothalamus. Thyroid-stimulating hormone may also influence thermogenesis, suppress appetite and regulate lipid storage through lipolysis and lipogenesis control. Subclinical hypothyroidism may induce changes in basal metabolic rate with subsequent increase in BMI, but obesity can also affect thyroid function via several mechanisms such as lipotoxicity and changes in adipokines and inflammatory cytokine secretion. The present study investigated the complex and mutual relationships between the thyroid axis and adiposity.
Collapse
Affiliation(s)
- Krzysztof Walczak
- Department of Thoracic Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 41-800 Zabrze, Poland;
| | - Lucyna Sieminska
- Department of Pathophysiology and Endocrinology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia in Katowice, 41-800 Zabrze, Poland
- Correspondence:
| |
Collapse
|
17
|
Desai M, Ross MG. Maternal-infant nutrition and development programming of offspring appetite and obesity. Nutr Rev 2021; 78:25-31. [PMID: 33196091 PMCID: PMC7667467 DOI: 10.1093/nutrit/nuaa121] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
In the United States and Mexico, the obesity epidemic represents a significant public health problem. Although obesity is often attributed to a Western-style, high-fat diet and decreased activity, there is now compelling evidence that this, in part, occurs because of the developmental programming effects resulting from exposure to maternal overnutrition. Human and animal studies demonstrate that maternal obesity and high-fat diet result in an increased risk for childhood and adult obesity. The potential programming effects of obesity have been partly attributed to hyperphagia, which occurs as a result of increased appetite with reduced satiety neuropeptides or neurons. However, depending on maternal nutritional status during the nursing period, the programmed hyperphagia and obesity can be exacerbated or prevented in offspring born to obese mothers. The underlying mechanism of this phenomenon likely involves the plasticity of the appetite regulatory center and thus presents an opportunity to modulate feeding and satiety regulation and break the obesity cycle.
Collapse
Affiliation(s)
- Mina Desai
- Department of Obstetrics and Gynecology, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, California, USA; and David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, USA
| | - Michael G Ross
- Department of Obstetrics and Gynecology, The Lundquist Institute at Harbor-UCLA Medical Center, Torrance, California, USA; and David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, California, USA
| |
Collapse
|
18
|
Koshko L, Debarba LK, Sacla M, de Lima JBM, Didyuk O, Fakhoury P, Sadagurski M. In Utero Maternal Benzene Exposure Predisposes to the Metabolic Imbalance in the Offspring. Toxicol Sci 2021; 180:252-261. [PMID: 33502539 DOI: 10.1093/toxsci/kfab010] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Environmental chemicals play a significant role in the development of metabolic disorders, especially when exposure occurs early in life. We have recently demonstrated that benzene exposure, at concentrations relevant to cigarette smoke, induces a severe metabolic imbalance in a sex-specific manner affecting male but not female mice. However, the roles of benzene in the development of aberrant metabolic outcomes following gestational exposure, remain largely unexplored. In this study, we exposed pregnant C57BL/6JB dams to benzene at 50 ppm or filtered air for 6 h/day from gestational day 0.5 (GD0.5) through GD21 and studied male and female offspring metabolic phenotypes in their adult life. While no changes in body weight or body composition were observed between groups, 4-month-old male and female offspring exhibited reduced parameters of energy homeostasis (VO2, VCO2, and heat production). However, only male offspring from benzene-exposed dams were glucose intolerant and insulin resistant at this age. By 6 months of age, both male and female offspring exhibited marked glucose intolerance however, only male offspring developed severe insulin resistance. This effect was accompanied by elevated insulin secretion and increased beta-cell mass only in male offspring. In support, Homeostatic Model Assessment for Insulin Resistance, the index of insulin resistance was elevated only in male but not in female offspring. Regardless, both male and female offspring exhibited a considerable increase in hepatic gene expression associated with inflammation and endoplasmic reticulum stress. Thus, gestational benzene exposure can predispose offspring to increased susceptibility to the metabolic imbalance in adulthood with differential sensitivity between sexes.
Collapse
Affiliation(s)
- Lisa Koshko
- Department of Biological Sciences, Integrative Biosciences Center (IBio), Wayne State University, Detroit, Michigan 48202, USA
| | - Lucas K Debarba
- Department of Biological Sciences, Integrative Biosciences Center (IBio), Wayne State University, Detroit, Michigan 48202, USA
| | - Mikaela Sacla
- Department of Biological Sciences, Integrative Biosciences Center (IBio), Wayne State University, Detroit, Michigan 48202, USA
| | - Juliana B M de Lima
- Department of Biological Sciences, Integrative Biosciences Center (IBio), Wayne State University, Detroit, Michigan 48202, USA
| | - Olesya Didyuk
- Department of Biological Sciences, Integrative Biosciences Center (IBio), Wayne State University, Detroit, Michigan 48202, USA
| | - Patrick Fakhoury
- Department of Biological Sciences, Integrative Biosciences Center (IBio), Wayne State University, Detroit, Michigan 48202, USA
| | - Marianna Sadagurski
- Department of Biological Sciences, Integrative Biosciences Center (IBio), Wayne State University, Detroit, Michigan 48202, USA
| |
Collapse
|
19
|
Abstract
A healthy nutritional state is required for all aspects of reproduction and is signaled by the adipokine leptin. Leptin acts in a relatively narrow concentration range: too much or too little will compromise fertility. The leptin signal timing is important to prepubertal development in both sexes. In the brain, leptin acts on ventral premammillary neurons which signal kisspeptin (Kiss1) neurons to stimulate gonadotropin releasing hormone (GnRH) neurons. Suppression of Kiss1 neurons occurs when agouti-related peptide neurons are activated by reduced leptin, because leptin normally suppresses these orexigenic neurons. In the pituitary, leptin stimulates production of GnRH receptors (GnRHRs) and follicle-stimulating hormone at midcycle, by activating pathways that derepress actions of the messenger ribonucleic acid translational regulatory protein Musashi. In females, rising estrogen stimulates a rise in serum leptin, which peaks at midcycle, synchronizing with nocturnal luteinizing hormone pulses. The normal range of serum leptin levels (10-20 ng/mL) along with gonadotropins and growth factors promote ovarian granulosa and theca cell functions and oocyte maturation. In males, the prepubertal rise in leptin promotes testicular development. However, a decline in leptin levels in prepubertal boys reflects inhibition of leptin secretion by rising androgens. In adult males, leptin levels are 10% to 50% of those in females, and high leptin inhibits testicular function. The obesity epidemic has elucidated leptin resistance pathways, with too much leptin in either sex leading to infertility. Under conditions of balanced nutrition, however, the secretion of leptin is timed and regulated within a narrow level range that optimizes its trophic effects.
Collapse
Affiliation(s)
- Gwen V Childs
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
- Correspondence: Gwen V. Childs, PhD, University of Arkansas for Medical Sciences, Little Rock, AR, USA. E-mail:
| | - Angela K Odle
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Melanie C MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Angus M MacNicol
- Department of Neurobiology and Developmental Sciences, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| |
Collapse
|
20
|
Gauda EB, Conde S, Bassi M, Zoccal DB, Almeida Colombari DS, Colombari E, Despotovic N. Leptin: Master Regulator of Biological Functions that Affects Breathing. Compr Physiol 2020; 10:1047-1083. [PMID: 32941688 DOI: 10.1002/cphy.c190031] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Obesity is a global epidemic in developed countries accounting for many of the metabolic and cardiorespiratory morbidities that occur in adults. These morbidities include type 2 diabetes, sleep-disordered breathing (SDB), obstructive sleep apnea, chronic intermittent hypoxia, and hypertension. Leptin, produced by adipocytes, is a master regulator of metabolism and of many other biological functions including central and peripheral circuits that control breathing. By binding to receptors on cells and neurons in the brainstem, hypothalamus, and carotid body, leptin links energy and metabolism to breathing. In this comprehensive article, we review the central and peripheral locations of leptin's actions that affect cardiorespiratory responses during health and disease, with a particular focus on obesity, SDB, and its effects during early development. Obesity-induced hyperleptinemia is associated with centrally mediated hypoventilation with decrease CO2 sensitivity. On the other hand, hyperleptinemia augments peripheral chemoreflexes to hypoxia and induces sympathoexcitation. Thus, "leptin resistance" in obesity is relative. We delineate the circuits responsible for these divergent effects, including signaling pathways. We review the unique effects of leptin during development on organogenesis, feeding behavior, and cardiorespiratory responses, and how undernutrition and overnutrition during critical periods of development can lead to cardiorespiratory comorbidities in adulthood. We conclude with suggestions for future directions to improve our understanding of leptin dysregulation and associated clinical diseases and possible therapeutic targets. Lastly, we briefly discuss the yin and the yang, specifically the contribution of relative adiponectin deficiency in adults with hyperleptinemia to the development of metabolic and cardiovascular disease. © 2020 American Physiological Society. Compr Physiol 10:1047-1083, 2020.
Collapse
Affiliation(s)
- Estelle B Gauda
- Division of Neonatology, Department of Pediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Silvia Conde
- CEDOC, NOVA Medical School, Faculdade de Ciências Médicas, Lisboa, Portugal
| | - Mirian Bassi
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Daniel B Zoccal
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Debora Simoes Almeida Colombari
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Eduardo Colombari
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, São Paulo, Brazil
| | - Nikola Despotovic
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
21
|
Effects of metabolic state on the regulation of melanocortin circuits. Physiol Behav 2020; 224:113039. [PMID: 32610101 PMCID: PMC7387173 DOI: 10.1016/j.physbeh.2020.113039] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 06/01/2020] [Accepted: 06/26/2020] [Indexed: 12/14/2022]
Abstract
Dysfunction in neurophysiological systems that regulate food intake and metabolism are at least partly responsible for obesity and related comorbidities. An important component of this process is the hypothalamic melanocortin system, where an imbalance can result in severe obesity and deficits in glucose metabolism. Exercise offers many health benefits related to cardiovascular improvements, hunger control, and blood glucose homeostasis. However, the molecular mechanism underlying the exercise-induced improvements to the melanocortin system remain undefined. Here, we review the role of the melanocortin system to sense hormonal, nutrient, and neuronal signals of energy status. This information is then relayed onto secondary neurons in order to regulate physiological parameters, which promote proper energy and glucose balance. We also provide an overview on the effects of physical exercise to induce biophysical changes in the melanocortin circuit which may regulate food intake, glucose metabolism and improve overall metabolic health.
Collapse
|
22
|
de Gortari P, Alcántara-Alonso V, Matamoros-Trejo G, Amaya MI, Alvarez-Salas E. Differential effects of leptin administration on feeding and HPT axis function in early-life overfed adult rats. Peptides 2020; 127:170285. [PMID: 32105809 DOI: 10.1016/j.peptides.2020.170285] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 01/29/2020] [Accepted: 02/22/2020] [Indexed: 12/18/2022]
Abstract
Early-life overfeeding (OF) disrupts neuroendocrine systems, energy homeostasis and food intake regulation inducing overeating and overweight in adults. Adult rats raised in small litters during lactation, display hyperphagia and overweight since weaning and exhibit a decrease in thyrotropin-releasing hormone (TRH) mRNA expression in hypothalamic paraventricular nucleus (PVN). This is counterintuitive because TRH expression should increase to activate the hypothalamic-pituitary-thyroid (HPT) axis and promote energy expenditure, thus, HPT axis seems inhibited in OF rats. Leptin, an adipocyte-synthesized hormone that stimulates hypothalamic TRH expression, enhances both TRH anorectic effects and HPT axis-induced metabolic rate. To evaluate hypothalamic resistance to the anorectic and HPT axis stimulatory actions of leptin, we injected leptin i.p. to ad libitum fed and to 48-h fasted adult control (reared in normal litters) and to small-litter reared (OF) male Wistar rats. Findings showed that HPT axis was still responsive to leptin, since PVN TRH mRNA levels, median eminence TRH release and T4 serum concentration increased in both, ad libitum and fasted OF rats after leptin administrations. Leptin was ineffective to reduce feeding of OF animals. By comparing leptin receptor (ObRb) expression changes between arcuate and PVN nuclei, we observed that arcuate ObRb was not modified in response to leptin administrations in OF rats, likely accounting for the differential effects in feeding and HPT axis function. Nevertheless, ObRb expression was modified by leptin in the PVN of OF rats to the same extent as controls; this supports the hormone's role as a therapeutic agent for early onset obesity in adults.
Collapse
Affiliation(s)
- P de Gortari
- Molecular Neurophysiology Laboratory, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñíz, Calz. México-Xochimilco No. 101, San Lorenzo Huipulco, CDMX, 14370, Mexico.
| | - V Alcántara-Alonso
- Molecular Neurophysiology Laboratory, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñíz, Calz. México-Xochimilco No. 101, San Lorenzo Huipulco, CDMX, 14370, Mexico
| | - G Matamoros-Trejo
- Molecular Neurophysiology Laboratory, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñíz, Calz. México-Xochimilco No. 101, San Lorenzo Huipulco, CDMX, 14370, Mexico
| | - M I Amaya
- Molecular Neurophysiology Laboratory, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñíz, Calz. México-Xochimilco No. 101, San Lorenzo Huipulco, CDMX, 14370, Mexico
| | - E Alvarez-Salas
- Molecular Neurophysiology Laboratory, Instituto Nacional de Psiquiatría Ramón de la Fuente Muñíz, Calz. México-Xochimilco No. 101, San Lorenzo Huipulco, CDMX, 14370, Mexico
| |
Collapse
|
23
|
Tavares MR, Lemes SF, de Fante T, Saenz de Miera C, Pavan ICB, Bezerra RMN, Prada PO, Torsoni MA, Elias CF, Simabuco FM. Modulation of hypothalamic S6K1 and S6K2 alters feeding behavior and systemic glucose metabolism. J Endocrinol 2020; 244:71-82. [PMID: 31557728 PMCID: PMC8010582 DOI: 10.1530/joe-19-0364] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 09/26/2019] [Indexed: 11/08/2022]
Abstract
The mTOR/S6Ks signaling is one of the intracellular pathways important for metabolic control, acting both peripherally and centrally. In the hypothalamus, mTOR/S6Ks axis mediates the action of leptin and insulin and can modulate the expression of neuropeptides. We analyzed the role of different S6Ks isoforms in the hypothalamic regulation of metabolism. We observed decreased food intake and decreased expression of agouti-related peptide (AgRP) following intracerebroventricular (icv) injections of adenoviral-mediated overexpression of three different S6Ks isoforms. Moreover, mice overexpressing p70-S6K1 in undefined periventricular hypothalamic neurons presented changes in glucose metabolism, as an increase in gluconeogenesis. To further evaluate the hypothalamic role of a less-studied S6K isoform, p54-S6K2, we used a Cre-LoxP approach to specifically overexpress it in AgRP neurons. Our findings demonstrate the potential participation of S6K2 in AgRP neurons regulating feeding behavior.
Collapse
Affiliation(s)
- Mariana Rosolen Tavares
- Multidisciplinary Laboratory of Food and Health (LABMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
- Laboratory of Metabolic Disorders (LABDIME), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Simone Ferreira Lemes
- Laboratory of Metabolic Disorders (LABDIME), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Thais de Fante
- Laboratory of Metabolic Disorders (LABDIME), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Cristina Saenz de Miera
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Isadora Carolina Betim Pavan
- Multidisciplinary Laboratory of Food and Health (LABMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
- Laboratory of Metabolic Disorders (LABDIME), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Rosangela Maria Neves Bezerra
- Multidisciplinary Laboratory of Food and Health (LABMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Patricia Oliveira Prada
- Laboratory of Molecular Research in Obesity (LABIMO), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Marcio Alberto Torsoni
- Laboratory of Metabolic Disorders (LABDIME), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| | - Carol Fuzeti Elias
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA
| | - Fernando Moreira Simabuco
- Multidisciplinary Laboratory of Food and Health (LABMAS), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
- Laboratory of Metabolic Disorders (LABDIME), School of Applied Sciences (FCA), University of Campinas (UNICAMP), Limeira, São Paulo, Brazil
| |
Collapse
|
24
|
Boyle CN, Le Foll C. Amylin and Leptin interaction: Role During Pregnancy, Lactation and Neonatal Development. Neuroscience 2019; 447:136-147. [PMID: 31846753 DOI: 10.1016/j.neuroscience.2019.11.034] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Revised: 11/08/2019] [Accepted: 11/21/2019] [Indexed: 01/04/2023]
Abstract
Amylin is co-secreted with insulin by pancreatic β-cells in response to a meal and produced by neurons in discrete hypothalamic brain areas. Leptin is proportionally secreted by the adipose tissue. Both hormones control food intake and energy homeostasis post-weaning in rodents. While amylin's main site of action is located in the area postrema (AP) and leptin's is located in the mediobasal hypothalamus, both hormones can also influence the other's signaling pathway; amylin has been shown enhance hypothalamic leptin signaling, and amylin signaling in the AP may rely on functional leptin receptors to modulate its effects. These two hormones also play major roles during other life periods. During pregnancy, leptin levels rise as a result of an increase in fat depot resulting in gestational leptin-resistance to prepare the maternal body for the metabolic needs during fetal development. The role of amylin is far less studied during pregnancy and lactation, though amylin levels seem to be elevated during pregnancy relative to insulin. Whether amylin and leptin interact during pregnancy and lactation remains to be assessed. Lastly, during brain development, amylin and leptin are major regulators of cell birth during embryogenesis and act as neurotrophic factors in the neonatal period. This review will highlight the role of amylin and leptin, and their possible interaction, during these dynamic time periods of pregnancy, lactation, and early development.
Collapse
Affiliation(s)
- Christina N Boyle
- Institute of Veterinary Physiology, University of Zurich, Winterthurerstrasse 260, 8057 Zurich, Switzerland.
| | - Christelle Le Foll
- Institute of Veterinary Physiology, University of Zurich, Winterthurerstrasse 260, 8057 Zurich, Switzerland.
| |
Collapse
|
25
|
Senn SS, Le Foll C, Whiting L, Tarasco E, Duffy S, Lutz TA, Boyle CN. Unsilencing of native LepRs in hypothalamic SF1 neurons does not rescue obese phenotype in LepR-deficient mice. Am J Physiol Regul Integr Comp Physiol 2019; 317:R451-R460. [PMID: 31314542 DOI: 10.1152/ajpregu.00111.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Leptin receptor (LepR) signaling in neurons of the ventromedial nucleus of the hypothalamus (VMH), specifically those expressing steroidogenic factor-1 (SF1), have been proposed to play a key role in controlling energy balance. By crossing LepR-silenced (LepRloxTB) mice with those expressing SF1-Cre, we unsilenced native LepR specifically in the VMH and tested whether SF1 neurons in the VMH are critical mediators of leptin's effect on energy homeostasis. LepRloxTB × SF1-Cre [knockout (KO)/Tg+] mice were metabolically phenotyped and compared with littermate controls that either expressed or were deficient in LepRs. Leptin-induced phosphorylated STAT3 was present in the VMH of KO/Tg+ mice and absent in other hypothalamic nuclei. VMH leptin signaling did not ameliorate obesity resulting from LepR deficiency in chow-fed mice. There was no change in food intake or energy expenditure when comparing complete LepR-null mice with KO/Tg+ mice, nor did KO/Tg+ mice show improved glucose tolerance. The presence of functional LepRs in the VMH mildly enhanced sensitivity to the pancreatic hormone amylin. When maintained on a high-fat diet (HFD), there was no reduction in diet-induced obesity in KO/Tg+ mice, but KO/Tg+ mice had improved glucose tolerance after 7 wk on an HFD compared with LepR-null mice. We conclude that LepR signaling in the VMH alone is not sufficient to correct metabolic dysfunction observed in LepR-null mice.
Collapse
Affiliation(s)
- Seraina S Senn
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Christelle Le Foll
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Lynda Whiting
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Erika Tarasco
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Sonya Duffy
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Thomas A Lutz
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland.,Zurich Centre for Integrative Human Physiology, University of Zurich, Zurich, Switzerland
| | - Christina Neuner Boyle
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
26
|
Leon Mercado L, Caron A, Wang Y, Burton M, Gautron L. Identification of Leptin Receptor-Expressing Cells in the Nodose Ganglion of Male Mice. Endocrinology 2019; 160:1307-1322. [PMID: 30907928 PMCID: PMC6482037 DOI: 10.1210/en.2019-00021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/18/2019] [Indexed: 12/29/2022]
Abstract
Leptin has been proposed to modulate viscerosensory information directly at the level of vagal afferents. In support of this view, broad expression for the leptin receptor (Lepr) has previously been reported in vagal afferents. However, the exact identity and distribution of leptin-sensitive vagal afferents has not been elucidated. Using quantitative PCR, we found that the whole mouse nodose ganglion was predominantly enriched in the short form of Lepr, rather than its long form. Consistent with this observation, the acute administration of leptin did not stimulate JAK-STAT signaling in the nodose ganglion. Using chromogenic in situ hybridization in wild-type mice and several reporter mouse models, we demonstrated that Lepr mRNA was restricted to nonneuronal cells in the epineurium and parenchyma of the nodose ganglion and a subset of vagal afferents, which accounted for only 3% of all neuronal profiles. Double labeling studies further established that Lepr-expressing vagal afferents were Nav1.8-negative fibers that did not supply the peritoneal cavity. Finally, double chromogenic in situ hybridization revealed that many Lepr-expressing neurons coexpressed the angiotensin 1a receptor (At1ar), which is a gene expressed in baroreceptors. Taken together, our data challenge the commonly held view that Lepr is broadly expressed in vagal afferents. Instead, our data suggest that leptin may exert a previously unrecognized role, mainly via its short form, as a direct modulator of a very small group of At1ar-positive vagal fibers.
Collapse
Affiliation(s)
- Luis Leon Mercado
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Alexandre Caron
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Yibing Wang
- Department of Biochemistry, Utah Southwestern Medical Center at Dallas, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Michael Burton
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, Texas
| | - Laurent Gautron
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
- Correspondence: Laurent Gautron, PhD, Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, Texas 75390. E-mail:
| |
Collapse
|
27
|
McGregor G, Harvey J. Leptin Regulation of Synaptic Function at Hippocampal TA-CA1 and SC-CA1 Synapses: Implications for Health and Disease. Neurochem Res 2019; 44:650-660. [PMID: 28819795 PMCID: PMC6420429 DOI: 10.1007/s11064-017-2362-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 07/05/2017] [Accepted: 07/21/2017] [Indexed: 12/16/2022]
Abstract
Growing evidence indicates that the endocrine hormone leptin regulates hippocampal synaptic function in addition to its established role as a hypothalamic satiety signal. Indeed, numerous studies show that leptin facilitates the cellular events that underlie hippocampal learning and memory including activity-dependent synaptic plasticity and glutamate receptor trafficking, indicating that leptin may be a potential cognitive enhancer. Although there has been extensive investigation into the modulatory role of leptin at hippocampal Schaffer collateral (SC)-CA1 synapses, recent evidence indicates that leptin also potently regulates excitatory synaptic transmission at the anatomically distinct temporoammonic (TA) input to hippocampal CA1 neurons. The cellular mechanisms underlying activity-dependent synaptic plasticity at TA-CA1 synapses differ from those at SC-CA1 synapses and the TA input is implicated in spatial and episodic memory formation. Furthermore, the TA input is an early target for neurodegeneration in Alzheimer's disease (AD) and aberrant leptin function is linked to AD. Here, we review the evidence that leptin regulates hippocampal synaptic function at both SC- and TA-CA1 synapses and discuss the consequences for neurodegenerative disorders like AD.
Collapse
Affiliation(s)
- Gemma McGregor
- Division of Neuroscience, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK
| | - Jenni Harvey
- Division of Neuroscience, School of Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK.
| |
Collapse
|
28
|
Quarta C, Fisette A, Xu Y, Colldén G, Legutko B, Tseng YT, Reim A, Wierer M, De Rosa MC, Klaus V, Rausch R, Thaker VV, Graf E, Strom TM, Poher AL, Gruber T, Le Thuc O, Cebrian-Serrano A, Kabra D, Bellocchio L, Woods SC, Pflugfelder GO, Nogueiras R, Zeltser L, Grunwald Kadow IC, Moon A, García-Cáceres C, Mann M, Treier M, Doege CA, Tschöp MH. Functional identity of hypothalamic melanocortin neurons depends on Tbx3. Nat Metab 2019; 1:222-235. [PMID: 32694784 PMCID: PMC8291379 DOI: 10.1038/s42255-018-0028-1] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 12/13/2018] [Indexed: 02/07/2023]
Abstract
Heterogeneous populations of hypothalamic neurons orchestrate energy balance via the release of specific signatures of neuropeptides. However, how specific intracellular machinery controls peptidergic identities and function of individual hypothalamic neurons remains largely unknown. The transcription factor T-box 3 (Tbx3) is expressed in hypothalamic neurons sensing and governing energy status, whereas human TBX3 haploinsufficiency has been linked with obesity. Here, we demonstrate that loss of Tbx3 function in hypothalamic neurons causes weight gain and other metabolic disturbances by disrupting both the peptidergic identity and plasticity of Pomc/Cart and Agrp/Npy neurons. These alterations are observed after loss of Tbx3 in both immature hypothalamic neurons and terminally differentiated mouse neurons. We further establish the importance of Tbx3 for body weight regulation in Drosophila melanogaster and show that TBX3 is implicated in the differentiation of human embryonic stem cells into hypothalamic Pomc neurons. Our data indicate that Tbx3 directs the terminal specification of neurons as functional components of the melanocortin system and is required for maintaining their peptidergic identity. In summary, we report the discovery of a key mechanistic process underlying the functional heterogeneity of hypothalamic neurons governing body weight and systemic metabolism.
Collapse
Affiliation(s)
- Carmelo Quarta
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, Bordeaux, France
- University of Bordeaux, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, Bordeaux, France
| | - Alexandre Fisette
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Yanjun Xu
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Division of Metabolic Diseases, Technische Universität München, Munich, Germany
| | - Gustav Colldén
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Beata Legutko
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Yu-Ting Tseng
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Alexander Reim
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Michael Wierer
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Maria Caterina De Rosa
- Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Department of Pediatrics, Columbia University, New York, NY, USA
| | - Valentina Klaus
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Division of Metabolic Diseases, Technische Universität München, Munich, Germany
| | - Rick Rausch
- Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Department of Pediatrics, Columbia University, New York, NY, USA
| | - Vidhu V Thaker
- Naomi Berrie Diabetes Center, Division of Molecular Genetics, Department of Pediatrics, Columbia University, New York, NY, USA
| | - Elisabeth Graf
- Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Tim M Strom
- Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Anne-Laure Poher
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Tim Gruber
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Ophélia Le Thuc
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Alberto Cebrian-Serrano
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Dhiraj Kabra
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Luigi Bellocchio
- INSERM U1215, NeuroCentre Magendie, Bordeaux, France
- Université de Bordeaux, NeuroCentre Magendie, Bordeaux, France
| | - Stephen C Woods
- University of Cincinnati College of Medicine, Department of Psychiatry and Behavioral Neuroscience, Metabolic Diseases Institute, Cincinnati, OH, USA
| | - Gert O Pflugfelder
- Institute of Developmental and Neurobiology. Johannes Gutenberg-University, Mainz, Germany
| | - Rubén Nogueiras
- Department of Physiology, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Lori Zeltser
- Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Ilona C Grunwald Kadow
- Technical University of Munich, School of Life Sciences, ZIEL - Institute for Food and Health, Freising, Germany
| | - Anne Moon
- Department of Molecular and Functional Genomics, Geisinger Clinic, Danville PA, USA
- Departments of Pediatrics and Human Genetics, University of Utah School of Medicine, Salt Lake City, UT, USA
| | - Cristina García-Cáceres
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Mathias Treier
- Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Claudia A Doege
- Naomi Berrie Diabetes Center, Columbia Stem Cell Initiative, Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Matthias H Tschöp
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Division of Metabolic Diseases, Technische Universität München, Munich, Germany.
| |
Collapse
|
29
|
Raghavan R, Zuckerman B, Hong X, Wang G, Ji Y, Paige D, DiBari J, Zhang C, Fallin MD, Wang X. Fetal and Infancy Growth Pattern, Cord and Early Childhood Plasma Leptin, and Development of Autism Spectrum Disorder in the Boston Birth Cohort. Autism Res 2018; 11:1416-1431. [PMID: 30248249 PMCID: PMC6320256 DOI: 10.1002/aur.2011] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 03/15/2018] [Accepted: 07/10/2018] [Indexed: 12/20/2022]
Abstract
Leptin is a proinflammatory cytokine that plays an important role in energy homeostasis. Emerging evidence suggests that leptin levels are altered in children with autism spectrum disorder (ASD); however, this has not been studied prospectively. Rapid growth during infancy and early childhood has been implicated in ASD, but the evidence is inconsistent. As leptin is involved in growth and is a potential risk factor for ASD, we explored the associations between (a) cord, early childhood leptin and ASD; and (b) birth weight for gestational age, early childhood weight gain, and ASD. We also assessed the mediating role of leptin in the relationship between weight gain during infancy and ASD. This study was conducted in a sample of 822 subjects from the Boston Birth Cohort. ASD was defined from diagnostic codes in electronic medical records. Extremely rapid weight gain during infancy was associated with a greater ASD risk and this persisted after adjusting for potential confounders (aOR: 3.11; 95% CI: 1.37, 7.07). Similarly, children that had higher plasma leptin levels, prior to ASD diagnosis, had an increased ASD risk in both unadjusted and adjusted models (aOR: 7.87; 95% CI: 2.06, 30.04). Further, early childhood leptin indirectly mediated the relationship between rapid weight gain and ASD. No associations were found between birth weight for gestational age, cord leptin and risk of ASD. Our findings provide a basis to further explore whether the combination of early life growth pattern and a biomarker such as leptin can predict ASD earlier. Autism Res 2018, 11: 1416-1431. © 2018 International Society for Autism Research, Wiley Periodicals, Inc. LAY SUMMARY: Is early life growth and a biomarker leptin related to ASD risk? To answer this question, we followed 822 children from birth and found that those who gained weight very quickly in infancy, had higher leptin levels in early childhood, had a greater chance of later ASD diagnosis. More research is needed to see if infant's weight gain pattern along with a biomarker (such as leptin) can be used to identify children with ASD sooner.
Collapse
Affiliation(s)
- Ramkripa Raghavan
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, 615 N. Wolfe St, Room E4132, Baltimore, MD 21205
| | - Barry Zuckerman
- Department of Pediatrics, Boston University School of Medicine and Boston Medical Center, 850 Harrison Ave, 3 Floor, Suite 324L, Boston, MA 02118
| | - Xiumei Hong
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, 615 N. Wolfe St, Room E4132, Baltimore, MD 21205
| | - Guoying Wang
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, 615 N. Wolfe St, Room E4132, Baltimore, MD 21205
| | - Yuelong Ji
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, 615 N. Wolfe St, Room E4132, Baltimore, MD 21205
| | - David Paige
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, 615 N. Wolfe St, Room E4132, Baltimore, MD 21205
| | - Jessica DiBari
- Office of Epidemiology and Research, Maternal & Child Health Bureau, Health Resources & Services Administration, 5600 Fishers Lane, 18N120, Rockville, MD 20857
| | - Cuilin Zhang
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, 615 N. Wolfe St, Room E4132, Baltimore, MD 21205
- Division of Intramural Population and Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, 6710 B, Rockledge Dr., Bethesda, MD 20817
| | - M. Daniele Fallin
- Wendy Klag Center for Autism and Developmental Disabilities & Department of Mental Health, Johns Hopkins University Bloomberg School of Public Health, 624 N. Broadway, HH 850, Baltimore, MD 21205
| | - Xiaobin Wang
- Center on the Early Life Origins of Disease, Department of Population, Family and Reproductive Health, Johns Hopkins University Bloomberg School of Public Health, 615 N. Wolfe St, Room E4132, Baltimore, MD 21205
- Division of General Pediatrics & Adolescent Medicine, Department of Pediatrics, Johns Hopkins University School of Medicine, 733 N Broadway, Baltimore, MD 21205
| |
Collapse
|
30
|
Ducsay CA, Goyal R, Pearce WJ, Wilson S, Hu XQ, Zhang L. Gestational Hypoxia and Developmental Plasticity. Physiol Rev 2018; 98:1241-1334. [PMID: 29717932 PMCID: PMC6088145 DOI: 10.1152/physrev.00043.2017] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Hypoxia is one of the most common and severe challenges to the maintenance of homeostasis. Oxygen sensing is a property of all tissues, and the response to hypoxia is multidimensional involving complicated intracellular networks concerned with the transduction of hypoxia-induced responses. Of all the stresses to which the fetus and newborn infant are subjected, perhaps the most important and clinically relevant is that of hypoxia. Hypoxia during gestation impacts both the mother and fetal development through interactions with an individual's genetic traits acquired over multiple generations by natural selection and changes in gene expression patterns by altering the epigenetic code. Changes in the epigenome determine "genomic plasticity," i.e., the ability of genes to be differentially expressed according to environmental cues. The genomic plasticity defined by epigenomic mechanisms including DNA methylation, histone modifications, and noncoding RNAs during development is the mechanistic substrate for phenotypic programming that determines physiological response and risk for healthy or deleterious outcomes. This review explores the impact of gestational hypoxia on maternal health and fetal development, and epigenetic mechanisms of developmental plasticity with emphasis on the uteroplacental circulation, heart development, cerebral circulation, pulmonary development, and the hypothalamic-pituitary-adrenal axis and adipose tissue. The complex molecular and epigenetic interactions that may impact an individual's physiology and developmental programming of health and disease later in life are discussed.
Collapse
Affiliation(s)
- Charles A. Ducsay
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Ravi Goyal
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - William J. Pearce
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Sean Wilson
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Xiang-Qun Hu
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| | - Lubo Zhang
- The Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, California
| |
Collapse
|
31
|
Lutz TA, Coester B, Whiting L, Dunn-Meynell AA, Boyle CN, Bouret SG, Levin BE, Le Foll C. Amylin Selectively Signals Onto POMC Neurons in the Arcuate Nucleus of the Hypothalamus. Diabetes 2018; 67:805-817. [PMID: 29467172 PMCID: PMC5910000 DOI: 10.2337/db17-1347] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 02/09/2018] [Indexed: 02/05/2023]
Abstract
Amylin phosphorylates ERK (p-ERK) in the area postrema to reduce eating and synergizes with leptin to phosphorylate STAT3 in the arcuate (ARC) and ventromedial (VMN) hypothalamic nuclei to reduce food intake and body weight. The current studies assessed potential amylin and amylin-leptin ARC/VMN interactions on ERK signaling and their roles in postnatal hypothalamic pathway development. In amylin knockout mice, the density of agouti-related protein (AgRP)-immunoreactive (IR) fibers in the hypothalamic paraventricular nucleus (PVN) was increased, while the density of α-melanocyte-stimulating hormone (αMSH) fibers was decreased. In mice deficient of the amylin receptor components RAMP1/3, both AgRP and αMSH-IR fiber densities were decreased, while only αMSH-IR fiber density was decreased in rats injected neonatally in the ARC/VMN with an adeno-associated virus short hairpin RNA against the amylin core receptor. Amylin induced p-ERK in ARC neurons, 60% of which was present in POMC-expressing neurons, with none in NPY neurons. An amylin-leptin interaction was shown by an additive effect on ARC ERK signaling in neonatal rats and a 44% decrease in amylin-induced p-ERK in the ARC of leptin receptor-deficient and of ob/ob mice. Together, these results suggest that amylin directly acts, through a p-ERK-mediated process, on POMC neurons to enhance ARC-PVN αMSH pathway development.
Collapse
Affiliation(s)
- Thomas A Lutz
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland
| | - Bernd Coester
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland
| | - Lynda Whiting
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland
| | | | - Christina N Boyle
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland
| | - Sebastien G Bouret
- Developmental Neuroscience Program, The Saban Research Institute, Children's Hospital Los Angeles, Department of Pediatrics, University of Southern California, Los Angeles, CA
- INSERM U1172, Jean-Pierre Aubert Research Center, Lille, France
| | - Barry E Levin
- Department of Neurology, Rutgers New Jersey Medical School, Newark, NJ
| | - Christelle Le Foll
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
32
|
Korgan AC, O'Leary E, King JL, Weaver ICG, Perrot TS. Effects of paternal high-fat diet and rearing environment on maternal investment and development of defensive responses in the offspring. Psychoneuroendocrinology 2018. [PMID: 29518693 DOI: 10.1016/j.psyneuen.2018.02.010] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Paternal preconception risk factors (e.g. stress, diet, drug use) correlate with metabolic dysfunction in offspring, which is often comorbid with depressive and anxiety-like phenotypes. Detection of these risk factors or deleterious phenotypes informs a female about prevailing ecological demands, in addition to potential adverse environment-induced phenotypes that may be disseminated to her offspring. We examined whether a F0 male rat's prior exposure to an obesogenic high-fat diet (HFD) influences a female's attraction towards a male, subsequent mother-infant interactions and the development of defensive (emotional) responses in the F1 offspring. Females displayed less interest in the HFD exposed F0 males relative to control diet-exposed F0 males. Dams that reared F1 offspring in larger, semi-naturalistic housing provided more licking and grooming and active arched-back-nursing behavior. However, some of these effects interacted with paternal experience. F0 HFD and maternal rearing environment revealed sex-dependent, between group differences in F1 offspring wean weight, juvenile social interactions and anxiety-like behavior in adolescence. Our results show for the first time in mammals that male exposure to HFD may contribute to stable behavioral variation among females in courtship, maternal care, even when the females are not directly exposed to a HFD, and anxiety-like behavior in F1 offspring. Furthermore, when offspring were exposed to a predatory threat, hypothalamic Crf gene regulation was influenced by early housing. These results, together with our previous findings, suggest that paternal experience and maternal rearing conditions can influence maternal behavior and development of defensive responses of offspring.
Collapse
Affiliation(s)
- Austin C Korgan
- Department of Psychology and Neuroscience, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada
| | - Elizabeth O'Leary
- Department of Psychology and Neuroscience, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada
| | - Jillian L King
- Department of Psychology and Neuroscience, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada
| | - Ian C G Weaver
- Department of Psychology and Neuroscience, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada; Department of Psychiatry, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada; Brain Repair Centre, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada
| | - Tara S Perrot
- Department of Psychology and Neuroscience, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada; Brain Repair Centre, Dalhousie University, Halifax B3H 4R2, Nova Scotia, Canada.
| |
Collapse
|
33
|
Bender MC, Hu C, Pelletier C, Denver RJ. To eat or not to eat: ontogeny of hypothalamic feeding controls and a role for leptin in modulating life-history transition in amphibian tadpoles. Proc Biol Sci 2018; 285:20172784. [PMID: 29593109 PMCID: PMC5897637 DOI: 10.1098/rspb.2017.2784] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 03/08/2018] [Indexed: 01/03/2023] Open
Abstract
Many animal life histories entail changing feeding ecology, but the molecular bases for these transitions are poorly understood. The amphibian tadpole is typically a growth and dispersal life-history stage. Tadpoles are primarily herbivorous, and they capitalize on growth opportunities to reach a minimum body size to initiate metamorphosis. During metamorphic climax, feeding declines, at which time the gastrointestinal (GI) tract remodels to accommodate the carnivorous diet of the adult frog. Here we show that anorexigenic hypothalamic feeding controls are absent in the tadpole, but develop during metamorphosis concurrent with the production of the satiety signal leptin. Before metamorphosis there is a large increase in leptin mRNA in fat tissue. Leptin receptor mRNA increased during metamorphosis in the preoptic area/hypothalamus, the key brain region involved with the control of food intake and metabolism. This corresponded with an increase in functional leptin receptor, as evidenced by induction of socs3 mRNA and phosphorylated STAT3 immunoreactivity, and suppression of feeding behaviour after injection of recombinant frog leptin. Furthermore, we found that immunoneutralization of leptin in tadpoles at metamorphic climax caused them to resume feeding. The absence of negative regulation of food intake in the tadpole allows the animal to maximize growth prior to metamorphosis. Maturation of leptin-responsive neural circuits suppresses feeding during metamorphosis to facilitate remodelling of the GI tract.
Collapse
Affiliation(s)
- Melissa Cui Bender
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1048, USA
| | - Caroline Hu
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1048, USA
| | - Chris Pelletier
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1048, USA
| | - Robert J Denver
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1048, USA
| |
Collapse
|
34
|
Pinos H, Carrillo B, Díaz F, Chowen JA, Collado P. Differential vulnerability to adverse nutritional conditions in male and female rats: Modulatory role of estradiol during development. Front Neuroendocrinol 2018; 48:13-22. [PMID: 28754628 DOI: 10.1016/j.yfrne.2017.07.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 07/07/2017] [Accepted: 07/23/2017] [Indexed: 01/21/2023]
Abstract
Many studies have shown the importance of an adequate nutritional environment during development to optimally establish the neurohormonal circuits that regulate feeding behavior. Under- or over-nutrition during early stages of life can lead to alterations in the physiology and brain networks that control food intake, resulting in a greater vulnerability to suffer maladjustments in energy metabolism in adulthood. These alterations produced by under- or over-nourishment during development differ between males and females, as does the modulatory action that estradiol exerts on the alterations produced by malnutrition. Estradiol regulates metabolism and brain metabolic circuits through the same transcription factor pathway, STAT3, that leptin and ghrelin use to program feeding circuits. Although more research is needed to disentangle the actual role of estradiol during development on the programming of feeding circuits, a synergistic role together with leptin and/or ghrelin might be hypothesized.
Collapse
Affiliation(s)
- Helena Pinos
- Departamento de Psicobiología, Universidad Nacional de Educación a Distancia (UNED), C/ Juan del Rosal n° 10, 28040 Madrid, Spain; Instituto Mixto de Investigación-Escuela Nacional de Sanidad (IMIENS), Spain
| | - Beatriz Carrillo
- Departamento de Psicobiología, Universidad Nacional de Educación a Distancia (UNED), C/ Juan del Rosal n° 10, 28040 Madrid, Spain; Instituto Mixto de Investigación-Escuela Nacional de Sanidad (IMIENS), Spain
| | - Francisca Díaz
- Departamento de Endocrinología, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Investigación Biomédica en Red (CIBER) de la Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Avda. Menéndez Pelayo, N° 65, 28009 Madrid, Spain
| | - Julie A Chowen
- Departamento de Endocrinología, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, Investigación Biomédica en Red (CIBER) de la Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Avda. Menéndez Pelayo, N° 65, 28009 Madrid, Spain
| | - Paloma Collado
- Departamento de Psicobiología, Universidad Nacional de Educación a Distancia (UNED), C/ Juan del Rosal n° 10, 28040 Madrid, Spain; Instituto Mixto de Investigación-Escuela Nacional de Sanidad (IMIENS), Spain.
| |
Collapse
|
35
|
Alterations in the programming of energy metabolism in adolescents with background exposure to dioxins, dl-PCBs and PBDEs. PLoS One 2017; 12:e0184006. [PMID: 28898241 PMCID: PMC5595283 DOI: 10.1371/journal.pone.0184006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 08/16/2017] [Indexed: 01/01/2023] Open
Abstract
Objectives Dioxins and PCBs are highly toxic and persistent environmental pollutants that are measurable in humans worldwide. These persistent organic pollutants are associated with a higher incidence of diabetes mellitus. We hypothesise that perinatal (background) exposure to industrial pollutants like dioxins also influences body mass development and energy metabolism in later life. Study design In The Netherlands, the perinatal exposure (prenatal exposure and postnatal lactational intake) to dioxins has been studied prospectively since 1987. Fasting glucose, insulin, HbA1c and leptin were analysed in 33 children of the original cohort of 60. BMI, glucose:insulin and BMI:leptin ratios were calculated. Prenatal exposure, lactational intake and current serum levels of dioxins (PCDD/F), dl-PCBs and PBDE concentrations were determined using (HR)GC-MS. Results Prenatal dioxin (PCDD/F) exposure was positively correlated to the glucose:insulin ratio (p = 0.024) and negatively correlated to the fasting insulin concentration (p = 0.017) in adolescence. Postnatal lactational PCDD/F intake was also negatively correlated to fasting insulin concentration (p = 0.028). Current serum levels of PCDD/Fs and total TEQ (dl-PCBs+PCDD/Fs) were positively correlated to the fasting serum glucose concentration (p = 0.015 and p = 0.037, respectively).No metabolic effects were seen in association with current serum levels of PBDEs. A positive correlation between the insulin and leptin concentrations (p = 0.034) was observed. No effects were found on leptin levels, BMI:leptin ratio, HbA1c levels or BMI. Discussion/Conclusion This study indicates that prenatal and lactational exposure influences glucose metabolism in adolescents, presumably through a negative effect on insulin secretion by pancreatic beta cells. Additionally, the very low recent background exposure to dioxins in puberty possibly has an effect on the glucose level.
Collapse
|
36
|
Reynolds CM, Perry JK, Vickers MH. Manipulation of the Growth Hormone-Insulin-Like Growth Factor (GH-IGF) Axis: A Treatment Strategy to Reverse the Effects of Early Life Developmental Programming. Int J Mol Sci 2017; 18:ijms18081729. [PMID: 28786951 PMCID: PMC5578119 DOI: 10.3390/ijms18081729] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 08/02/2017] [Accepted: 08/04/2017] [Indexed: 12/24/2022] Open
Abstract
Evidence from human clinical, epidemiological, and experimental animal models has clearly highlighted a link between the early life environment and an increased risk for a range of cardiometabolic disorders in later life. In particular, altered maternal nutrition, including both undernutrition and overnutrition, spanning exposure windows that cover the period from preconception through to early infancy, clearly highlight an increased risk for a range of disorders in offspring in later life. This process, preferentially termed “developmental programming” as part of the developmental origins of health and disease (DOHaD) framework, leads to phenotypic outcomes in offspring that closely resemble those of individuals with untreated growth hormone (GH) deficiency, including increased adiposity and cardiovascular disorders. As such, the use of GH as a potential intervention strategy to mitigate the effects of developmental malprogramming has received some attention in the DOHaD field. In particular, experimental animal models have shown that early GH treatment in the setting of poor maternal nutrition can partially rescue the programmed phenotype, albeit in a sex-specific manner. Although the mechanisms remain poorly defined, they include changes to endothelial function, an altered inflammasome, changes in adipogenesis and cardiovascular function, neuroendocrine effects, and changes in the epigenetic regulation of gene expression. Similarly, GH treatment to adult offspring, where an adverse metabolic phenotype is already manifest, has shown efficacy in reversing some of the metabolic disorders arising from a poor early life environment. Components of the GH-insulin-like growth factor (IGF)-IGF binding protein (GH-IGF-IGFBP) system, including insulin-like growth factor 1 (IGF-1), have also shown promise in ameliorating programmed metabolic disorders, potentially acting via epigenetic processes including changes in miRNA profiles and altered DNA methylation. However, as with the use of GH in the clinical setting of short stature and GH-deficiency, the benefits of treatment are also, in some cases, associated with potential unwanted side effects that need to be taken into account before effective translation as an intervention modality in the DOHaD context can be undertaken.
Collapse
Affiliation(s)
- Clare M Reynolds
- Liggins Institute, University of Auckland, Auckland 1142, New Zealand.
| | - Jo K Perry
- Liggins Institute, University of Auckland, Auckland 1142, New Zealand.
| | - Mark H Vickers
- Liggins Institute, University of Auckland, Auckland 1142, New Zealand.
| |
Collapse
|
37
|
Zhang P, Xu L, Guan H, Liu L, Liu J, Huang Z, Cao X, Liao Z, Xiao H, Li Y. Beraprost sodium, a prostacyclin analogue, reduces fructose-induced hepatocellular steatosis in mice and in vitro via the microRNA-200a and SIRT1 signaling pathway. Metabolism 2017; 73:9-21. [PMID: 28732575 DOI: 10.1016/j.metabol.2017.05.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 04/24/2017] [Accepted: 05/06/2017] [Indexed: 12/30/2022]
Abstract
PURPOSE To determine whether beraprost sodium, a prostacyclin analogue, could reduce hepatic lipid accumulation induced by fructose in mice and cultured human hepatocytes, and to investigate the expression of microRNAs and the sirtuin 1 (SIRT1) pathway. METHODS Male C57BL/6JNju mice were divided into three groups and fed one of the following diets: a normal diet, a high fructose diet, or a high fructose diet with beraprost sodium treatment. In addition, human-derived HepG2 cells were cultured and treated with fructose (25mmol/L) with or without beraprost sodium (10μmol/L) for 24h, and transfected with small interfering RNA (siRNA) against SIRT1, miR-200a mimic, or miR-200a inhibitor for 48h. The miRNA microarray analysis was performed on the HepG2 cells, and the expression profiles of miRNAs were analyzed using Gene Cluster 3.0 and verified using qPCR. RESULTS Beraprost sodium treatment attenuated hepatic steatosis, induced the transcription of genes involved in lipid metabolism in C57BL/6 mice (P<0.05), and increased the expression of hepatic SIRT1 and peroxisome proliferator activated receptor α (PPARα) in the cells treated with fructose. These effects were blocked in HepG2 cells after transfection with siRNA against SIRT1. MiR-200a was highly expressed during fructose treatment and was down regulated by beraprost sodium (P<0.05). A luciferase assay showed that miR-200a regulated SIRT1 by binding to the 3' UTR. Overexpression of miR-200a inhibited expression of hepatic SIRT1. CONCLUSIONS Our study demonstrated that SIRT1 pathway mediated the effects of beraprost sodium on attenuation of hepatic lipid disorders induced by fructose and revealed the primary role of miR-200a in the regulation of hepatic SIRT1 by beraprost sodium. Our findings suggested that SIRT1 might be a therapeutic target of fructose-related metabolism disorders.
Collapse
Affiliation(s)
- Pengyuan Zhang
- The First Affiliated Hospital, Sun Yat-sen University, Department of Endocrinology, Guangzhou, 510080, China
| | - Lijuan Xu
- The First Affiliated Hospital, Sun Yat-sen University, Department of Endocrinology, Guangzhou, 510080, China
| | - Hongyu Guan
- The First Affiliated Hospital, Sun Yat-sen University, Department of Endocrinology, Guangzhou, 510080, China
| | - Liehua Liu
- The First Affiliated Hospital, Sun Yat-sen University, Department of Endocrinology, Guangzhou, 510080, China
| | - Juan Liu
- The First Affiliated Hospital, Sun Yat-sen University, Department of Endocrinology, Guangzhou, 510080, China
| | - Zhimin Huang
- The First Affiliated Hospital, Sun Yat-sen University, Department of Endocrinology, Guangzhou, 510080, China
| | - Xiaopei Cao
- The First Affiliated Hospital, Sun Yat-sen University, Department of Endocrinology, Guangzhou, 510080, China
| | - Zhihong Liao
- The First Affiliated Hospital, Sun Yat-sen University, Department of Endocrinology, Guangzhou, 510080, China
| | - Haipeng Xiao
- The First Affiliated Hospital, Sun Yat-sen University, Department of Endocrinology, Guangzhou, 510080, China
| | - Yanbing Li
- The First Affiliated Hospital, Sun Yat-sen University, Department of Endocrinology, Guangzhou, 510080, China.
| |
Collapse
|
38
|
Bender MC, Sifuentes CJ, Denver RJ. Leptin Induces Mitosis and Activates the Canonical Wnt/β-Catenin Signaling Pathway in Neurogenic Regions of Xenopus Tadpole Brain. Front Endocrinol (Lausanne) 2017; 8:99. [PMID: 28533765 PMCID: PMC5421298 DOI: 10.3389/fendo.2017.00099] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 04/20/2017] [Indexed: 12/16/2022] Open
Abstract
In addition to its well-known role as an adipostat in adult mammals, leptin has diverse physiological and developmental actions in vertebrates. Leptin has been shown to promote development of hypothalamic circuits and to induce mitosis in different brain areas of mammals. We investigated the ontogeny of leptin mRNA, leptin actions on cell proliferation in the brain, and gene expression in the preoptic area/hypothalamus of tadpoles of Xenopus laevis. The level of leptin mRNA was low in premetamorphic tadpoles, but increased strongly at the beginning of metamorphosis and peaked at metamorphic climax. This increase in leptin mRNA at the onset of metamorphosis correlated with increased cell proliferation in the neurogenic zones of tadpole brain. We found that intracerebroventricular (i.c.v.) injection of recombinant Xenopus leptin (rxLeptin) in premetamorphic tadpoles strongly increased cell proliferation in neurogenic zones throughout the tadpole brain. We conducted gene expression profiling of genes induced at 2 h following i.c.v. injection of rxLeptin. This analysis identified 2,322 genes induced and 1,493 genes repressed by rxLeptin. The most enriched Kyoto Encyclopedia of Genes and Genomes term was the canonical Wnt/β-catenin pathway. Using electroporation-mediated gene transfer into tadpole brain of a reporter vector responsive to the canonical Wnt/β-catenin signaling pathway, we found that i.c.v. rxLeptin injection activated Wnt/β-catenin-dependent transcriptional activity. Our findings show that leptin acts on the premetamorphic tadpole brain to induce cell proliferation, possibly acting via the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Melissa Cui Bender
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Christopher J. Sifuentes
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Robert J. Denver
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
39
|
Moody L, Chen H, Pan YX. Early-Life Nutritional Programming of Cognition-The Fundamental Role of Epigenetic Mechanisms in Mediating the Relation between Early-Life Environment and Learning and Memory Process. Adv Nutr 2017; 8:337-350. [PMID: 28298276 PMCID: PMC5347110 DOI: 10.3945/an.116.014209] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The perinatal period is a window of heightened plasticity that lays the groundwork for future anatomic, physiologic, and behavioral outcomes. During this time, maternal diet plays a pivotal role in the maturation of vital organs and the establishment of neuronal connections. However, when perinatal nutrition is either lacking in specific micro- and macronutrients or overloaded with excess calories, the consequences can be devastating and long lasting. The brain is particularly sensitive to perinatal insults, with several neurologic and psychiatric disorders having been linked to a poor in utero environment. Diseases characterized by learning and memory impairments, such as autism, schizophrenia, and Alzheimer disease, are hypothesized to be attributed in part to environmental factors, and evidence suggests that the etiology of these conditions may date back to very early life. In this review, we discuss the role of the early-life diet in shaping cognitive outcomes in offspring. We explore the endocrine and immune mechanisms responsible for these phenotypes and discuss how these systemic factors converge to change the brain's epigenetic landscape and regulate learning and memory across the lifespan. Through understanding the maternal programming of cognition, critical steps may be taken toward preventing and treating diseases that compromise learning and memory.
Collapse
Affiliation(s)
| | - Hong Chen
- Division of Nutritional Sciences,,Department of Food Science and Human Nutrition, and
| | - Yuan-Xiang Pan
- Division of Nutritional Sciences, .,Department of Food Science and Human Nutrition, and.,Illinois Informatics Institute, University of Illinois at Urbana-Champaign, Urbana, IL
| |
Collapse
|
40
|
Levin BE, Lutz TA. Amylin and Leptin: Co-Regulators of Energy Homeostasis and Neuronal Development. Trends Endocrinol Metab 2017; 28:153-164. [PMID: 27938937 DOI: 10.1016/j.tem.2016.11.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Revised: 11/03/2016] [Accepted: 11/07/2016] [Indexed: 12/18/2022]
Abstract
While the regulation of energy homeostasis by amylin is already well-characterized, emerging data suggest that amylin is also crucial for the development of neural pathways in the hypothalamus and caudal hindbrain (area postrema, AP; nucleus tractus solitarius, NTS). Exciting new findings demonstrate crucial amylin-leptin interactions in altering the activity of specific hypothalamic and AP neurons, and a role for amylin as a novel class of 'leptin sensitizers' which enhance leptin signaling in both leptin-sensitive and -resistant individuals, in part by stimulating IL-6 production by hypothalamic microglia. This review summarizes these findings and provides a hypothetical framework for future studies to elucidate the mechanisms by which amylin and leptin act individually and as co-conspirators to alter energy homeostasis and neuronal development.
Collapse
Affiliation(s)
- Barry E Levin
- Department of Neurology, Rutgers, New Jersey Medical School, Newark, NJ 07103, USA.
| | - Thomas A Lutz
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
41
|
Aiceles V, Gombar F, da Fonte Ramos C. Hormonal and testicular changes in rats submitted to congenital hypothyroidism in early life. Mol Cell Endocrinol 2017; 439:65-73. [PMID: 27793676 DOI: 10.1016/j.mce.2016.10.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 10/24/2016] [Accepted: 10/24/2016] [Indexed: 12/24/2022]
Abstract
The goal of this study was to evaluate the influence of hypothyroidism induced by MMI, during gestation (G) or gestation plus lactation (GL) on testis and its relation with leptin in rats. Six to eight pups were killed at 90 days of age. For statistical analysis One-way ANOVA followed by the Holm-Sìdak post hoc test was used. Hypothyroidism resulted in a significant reduction in LH, FSH and testosterone and an increase in leptin serum levels (p < 0.04). There was a significant decrease in StAR, AR, FSHR, LHR, pSTAT3 and SOCS3 (p < 0.04) protein expression and in the fertility parameters (p < 0.04). We can conclude that hypothyroidism is associated with reduction of steroidogenesis and spermatogenesis leading to a low fertility potential in these animals. This outcome could be a consequence of low pituitary stimulus and testicular response and probably are not related with leptin hormone since its signaling pathway is down-regulated in the testis.
Collapse
Affiliation(s)
- Veronica Aiceles
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Brazil
| | - Flavia Gombar
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Brazil
| | - Cristiane da Fonte Ramos
- Laboratory of Morphometry, Metabolism and Cardiovascular Disease, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Brazil.
| |
Collapse
|
42
|
Abstract
The brain is considered a major site for microRNA (miRNA) expression; as evidenced by several studies reporting microarray data of different brain substructures. The hypothalamus is among the brain regions that plays a crucial role in integrating signals from other brain nuclei as well as environmental, hormonal, metabolic and neuronal signals from the periphery in order to deliver an adequate response. The hypothalamus controls vital functions such as reproduction, energy homeostasis, water balance, circadian rhythm and stress. These functions need a high neuronal plasticity to adequately respond to physiological, environmental and psychological stimuli that could be limited to a specific temporal period during life or are cyclic events. In this context, miRNAs constitute major regulators and coordinators of gene expression. Indeed, in response to specific stimuli, changes in miRNA expression profiles finely tune specific mRNA targets to adequately fit to the immediate needs through mainly the modulation of neuronal plasticity.
Collapse
Affiliation(s)
- Mohammed Taouis
- Molecular Neuroendocrinology of Food Intake (NMPA), UMR 9197, University Paris-Sud, Orsay, France; NMPA, Neurosciences Paris Saclay Institute (NeuroPSI), Department Molecules & Circuits, CNRS UMR 9197, Orsay, France.
| |
Collapse
|
43
|
Janoschek R, Bae-Gartz I, Vohlen C, Alcázar MAA, Dinger K, Appel S, Dötsch J, Hucklenbruch-Rother E. Dietary intervention in obese dams protects male offspring from WAT induction of TRPV4, adiposity, and hyperinsulinemia. Obesity (Silver Spring) 2016; 24:1266-73. [PMID: 27106804 DOI: 10.1002/oby.21486] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Revised: 01/26/2016] [Accepted: 01/26/2016] [Indexed: 12/21/2022]
Abstract
OBJECTIVE One major risk factor for childhood overweight is maternal obesity. The underlying molecular mechanisms are ill-defined, and effective prevention strategies are missing. METHODS Diet-induced obese mouse dams were changed to standard chow during pregnancy and lactation as an intervention against predisposition for obesity and metabolic sequelea in the offspring. Expression of adipokines and TRPV4, a regulator of adipose oxidative metabolism, inflammation, and energy homeostasis, in offspring's white adipose tissue (WAT) was assessed. RESULTS Pathological effects on offspring's body weight, fat content, and serum insulin were fully reversed in intervention offspring on postnatal day 21. In WAT, a sixfold increase of Trpv4 mRNA expression in offspring consuming high-fat-containing diet was found, which was completely blunted in the intervention group. Simultaneously, WAT adipokine, interleukin-6, and peroxisome proliferator-activated receptor-γ mRNA and UCP1 protein expression were largely returned to control levels in intervention offspring. CONCLUSIONS Improvement of maternal nutrition offers a powerful strategy to improve offspring's metabolic health. Targeting TRPV4-linked aspects of WAT metabolic function during early development might be a promising approach to prevent long-term adverse metabolic effects of maternal high-fat nutrition.
Collapse
Affiliation(s)
- Ruth Janoschek
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Cologne, Germany
| | - Inga Bae-Gartz
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Cologne, Germany
| | - Christina Vohlen
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Cologne, Germany
| | | | - Katharina Dinger
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Cologne, Germany
| | - Sarah Appel
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Cologne, Germany
| | - Jörg Dötsch
- Department of Pediatrics and Adolescent Medicine, University of Cologne, Cologne, Germany
| | | |
Collapse
|
44
|
[Metabolic and cardiovascular consequences of suction-assisted lipectomy: Systematic review]. ANN CHIR PLAST ESTH 2016; 61:270-86. [PMID: 27114181 DOI: 10.1016/j.anplas.2016.03.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2016] [Accepted: 03/22/2016] [Indexed: 01/21/2023]
Abstract
BACKGROUND Suction-assisted lipectomy is one of the most frequent procedures in plastic surgery. The aim of this study was to investigate whether suction-assisted lipectomy causes changes in the carbohydrates and lipid metabolism and the potential effects on cardiovascular risk factors. METHODS We interrogated five databases: Medline, American College of Physicians Journal Club Database, Cochrane central register of controlled trials, Cochrane database of systematic reviews, Database of abstracts of reviews of effects. A systematic review of the literature was performed in order to compare results of randomized controlled trials and observational studies concerning changes in weight, metabolism, endocrinology, inflammatory markers and cardiovascular risk factors after suction-assisted lipectomy. All articles were assessed by criteria from Oxford Center For Evidence Based Medicine (OCEBM). RESULTS The search resulted in 40 articles: 12 experimental animal studies and 28 human studies. CONCLUSION Different metabolic parameters are affected by suction-assited lipectomy. First, all articles point out a decrease of body weight after suction-assisted lipectomy. Weight lost only affects fat mass without any change of lean mass. The potential compensatory growth of visceral fat seems to be counteracted by physical activity. Then, resting energy expenditure seems to be stable or decrease after the surgery. This reduction is significantly related to the decrease of leptin levels and also seems to be counteracted by physical activity. About adipocytokines, leptin level decreases after suction-assisted lipectomy while results are contradictory about adiponectin and resistin levels. However adiponectin seems to tend to increase after surgery. Inflammatory markers seem to increase within first hours after surgery. Then they seem to decrease or remain at the preoperative levels. Fasting insulin level decreases and is linked to the aspirated volume. So insulin sensitivity seems to be improved. Concerning lipid profil, it tends to remain the same or to be improved by suction-assisted lipectomy. In conclusion, regarding all the literature, there is still debate about metabolic effect of suction-assisted lipectomy. Prospective clinical studies are needed to confirm or invalidate some hypotheses. These studies must consider some potential biases as physical activity, diet and medical treatment modifications (statins).
Collapse
|
45
|
Mela V, Díaz F, Vázquez MJ, Argente J, Tena-Sempere M, Viveros MP, Chowen JA. Interaction between neonatal maternal deprivation and serum leptin levels on metabolism, pubertal development, and sexual behavior in male and female rats. Biol Sex Differ 2016; 7:2. [PMID: 26759712 PMCID: PMC4710050 DOI: 10.1186/s13293-015-0054-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Accepted: 12/23/2015] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Maternal deprivation (MD) during neonatal life can have long-term effects on metabolism and behavior, with males and females responding differently. We previously reported that MD during 24 h at postnatal day (PND) 9 blocks the physiological neonatal leptin surge in both sexes. It is known that modifications in neonatal leptin levels can affect metabolism in adulthood. Thus, we hypothesized that at least some of the long-term metabolic changes that occur in response to MD are due to the decline in serum leptin during this critical period of development. Hence, we predicted that treatment with leptin during MD would normalize these metabolic changes, with this response also differing between the sexes. METHODS MD was carried-out in Wistar rats for 24 h on PND9. Control and MD rats of both sexes were treated from PND 9 to 13 with leptin (3 mg/kg/day sc) or vehicle. Weight gain, food intake, glucose tolerance, and pubertal onset were monitored. Sexual behavior was analyzed in males. Rats were killed at PND90, and serum hormones and hypothalamic neuropeptides involved in metabolic control and reproduction were measured. Results were analyzed by three-way analysis of covariance using sex, MD, and leptin treatment as factors and litter as the covariate and employing repeated measures where appropriate. RESULTS In males, MD advanced the external signs of puberty and increased serum insulin and triglyceride levels and hypothalamic proopiomelanocortin mRNA levels at PND90. Neonatal leptin treatment normalized these effects. In contrast, MD decreased circulating triglycerides, as well as estradiol levels, in females at PND90 and these changes were also normalized by neonatal leptin treatment. Neonatal leptin treatment also had long-term effects in control rats as it advanced the external signs of puberty in control males, but delayed them in females. Neonatal leptin treatment increased serum insulin and hypothalamic mRNA levels of the leptin receptor and cocaine- and amphetamine-regulated transcript in control males and increased orexin mRNA levels in controls of both sexes. Although pubertal onset in males was advanced by either MD or neonatal leptin treatment in males and delayed by leptin treatment in females, the mRNA levels of hypothalamic neuropeptides and receptors related to reproduction were not affected by MD or neonatal leptin treatment in either sex at PND90. CONCLUSIONS These findings indicate that some of the long-term changes in metabolic and reproductive parameters induced by MD, such as advanced pubertal onset and increased hypothalamic proopiomelanocortin (POMC) expression, hyperinsulinemia, and hypertriglyceridemia in adult males and decreased serum triglyceride and estradiol levels in females, are most likely due to the decrease in leptin levels during the period of MD.
Collapse
Affiliation(s)
- Virginia Mela
- Department of Physiology (Animal Physiology II), Faculty of Biology. Complutense University Madrid, Madrid, Spain
| | - Francisca Díaz
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, Avenida Menéndez Pelayo, 65, Madrid, 28009 Spain ; CIBEROBN, Instituto Carlos III Madrid, Madrid, Spain
| | - María Jesús Vázquez
- CIBEROBN, Instituto Carlos III Madrid, Madrid, Spain ; Department of Cell Biology, Physiology and Immunology, University of Cordoba & Instituto Maimónides de Investigación Biomédica (IMIBIC), Hospital Universitario Reina Sofia, Córdoba, Spain
| | - Jesús Argente
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, Avenida Menéndez Pelayo, 65, Madrid, 28009 Spain ; CIBEROBN, Instituto Carlos III Madrid, Madrid, Spain ; Department of Pediatrics, Universidad Autónoma de Madrid, Madrid, Spain
| | - Manuel Tena-Sempere
- CIBEROBN, Instituto Carlos III Madrid, Madrid, Spain ; Department of Cell Biology, Physiology and Immunology, University of Cordoba & Instituto Maimónides de Investigación Biomédica (IMIBIC), Hospital Universitario Reina Sofia, Córdoba, Spain
| | - Maria-Paz Viveros
- Department of Physiology (Animal Physiology II), Faculty of Biology. Complutense University Madrid, Madrid, Spain
| | - Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, Avenida Menéndez Pelayo, 65, Madrid, 28009 Spain ; CIBEROBN, Instituto Carlos III Madrid, Madrid, Spain
| |
Collapse
|
46
|
Cannon AM, Kakulas F, Hepworth AR, Lai CT, Hartmann PE, Geddes DT. The Effects of Leptin on Breastfeeding Behaviour. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2015; 12:12340-55. [PMID: 26437426 PMCID: PMC4626972 DOI: 10.3390/ijerph121012340] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 09/07/2015] [Accepted: 09/23/2015] [Indexed: 11/27/2022]
Abstract
Breastfed infants have a reduced risk of becoming overweight and/or obese later in life. This protective effect has been partly attributed to leptin present in breastmilk. This study investigated 24-h variations of skim milk leptin and its relationship with breastmilk macronutrients and infant breastfeeding patterns. Exclusive breastfeeding mothers of term singletons (n = 19; age 10 ± 5 weeks) collected pre- and post-feed breastmilk samples for every breastfeed over a 24-h period and test-weighed their infants to determine milk intake at every breastfeed over a 24-h period. Samples (n = 454) were analysed for leptin, protein, lactose and fat content. Skim milk leptin concentration did not change with feeding (p = 0.184). However, larger feed volumes (>105 g) were associated with a decrease in post-feed leptin levels (p = 0.009). There was no relationship between the change in leptin levels and change in protein (p = 0.313) or lactose levels (p = 0.587) between pre- and post-feed milk, but there was a trend for a positive association with changes in milk fat content (p = 0.056). Leptin concentration significantly increased at night (p < 0.001) indicating a possible 24-h pattern. Leptin dose (ng) was not associated with the time between feeds (p = 0.232). Further research should include analysis of whole breastmilk and other breastmilk fractions to extend these findings.
Collapse
Affiliation(s)
- Anna M Cannon
- School of Chemistry and Biochemistry, Faculty of Science, The University of Western Australia, Perth, Western Australia 6009, Australia.
| | - Foteini Kakulas
- School of Chemistry and Biochemistry, Faculty of Science, The University of Western Australia, Perth, Western Australia 6009, Australia.
| | - Anna R Hepworth
- School of Chemistry and Biochemistry, Faculty of Science, The University of Western Australia, Perth, Western Australia 6009, Australia.
| | - Ching Tat Lai
- School of Chemistry and Biochemistry, Faculty of Science, The University of Western Australia, Perth, Western Australia 6009, Australia.
| | - Peter E Hartmann
- School of Chemistry and Biochemistry, Faculty of Science, The University of Western Australia, Perth, Western Australia 6009, Australia.
| | - Donna T Geddes
- School of Chemistry and Biochemistry, Faculty of Science, The University of Western Australia, Perth, Western Australia 6009, Australia.
| |
Collapse
|
47
|
Mela V, Díaz F, Borcel E, Argente J, Chowen JA, Viveros MP. Long Term Hippocampal and Cortical Changes Induced by Maternal Deprivation and Neonatal Leptin Treatment in Male and Female Rats. PLoS One 2015; 10:e0137283. [PMID: 26382238 PMCID: PMC4575204 DOI: 10.1371/journal.pone.0137283] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Accepted: 08/14/2015] [Indexed: 01/17/2023] Open
Abstract
Maternal deprivation (MD) during neonatal life has diverse long-term behavioral effects and alters the development of the hippocampus and frontal cortex, with several of these effects being sexually dimorphic. MD animals show a marked reduction in their circulating leptin levels, not only during the MD period, but also several days later (PND 13). A neonatal leptin surge occurs in rodents (beginning around PND 5 and peaking between PND 9 and 10) that has an important neurotrophic role. We hypothesized that the deficient neonatal leptin signaling of MD rats could be involved in the altered development of their hippocampus and frontal cortex. Accordingly, a neonatal leptin treatment in MD rats would at least in part counteract their neurobehavioural alterations. MD was carried out in Wistar rats for 24 h on PND 9. Male and female MD and control rats were treated from PND 9 to 13 with rat leptin (3 mg/kg/day sc) or vehicle. In adulthood, the animals were submitted to the open field, novel object memory test and the elevated plus maze test of anxiety. Neuronal and glial population markers, components of the glutamatergic and cannabinoid systems and diverse synaptic plasticity markers were evaluated by PCR and/or western blotting. Main results include: 1) In some of the parameters analyzed, neonatal leptin treatment reversed the effects of MD (eg., mRNA expression of hippocampal IGF1 and protein expression of GFAP and vimentin) partially confirming our hypothesis; 2) The neonatal leptin treatment, per se, exerted a number of behavioral (increased anxiety) and neural effects (eg., expression of the following proteins: NG2, NeuN, PSD95, NCAM, synaptophysin). Most of these effects were sex dependent. An adequate neonatal leptin level (avoiding excess and deficiency) appears to be necessary for its correct neuro-programing effect.
Collapse
Affiliation(s)
- Virginia Mela
- Department of Physiology (Anim Physiol II), Faculty of Biology, Complutense University Madrid, Madrid, Spain
| | - Francisca Díaz
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa & CIBEROBN Instituto Carlos III, Madrid, Spain
| | - Erika Borcel
- Brain Mind Institute and School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Jesús Argente
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa & CIBEROBN Instituto Carlos III, Madrid, Spain
| | - Julie A. Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa & CIBEROBN Instituto Carlos III, Madrid, Spain
| | - Maria-Paz Viveros
- Department of Physiology (Anim Physiol II), Faculty of Biology, Complutense University Madrid, Madrid, Spain
- * E-mail:
| |
Collapse
|
48
|
Neri C, Edlow AG. Effects of Maternal Obesity on Fetal Programming: Molecular Approaches. Cold Spring Harb Perspect Med 2015; 6:a026591. [PMID: 26337113 DOI: 10.1101/cshperspect.a026591] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Maternal obesity has become a worldwide epidemic. Obesity and a high-fat diet have been shown to have deleterious effects on fetal programming, predisposing offspring to adverse cardiometabolic and neurodevelopmental outcomes. Although large epidemiological studies have shown an association between maternal obesity and adverse outcomes for offspring, the underlying mechanisms remain unclear. Molecular approaches have played a key role in elucidating the mechanistic underpinnings of fetal malprogramming in the setting of maternal obesity. These approaches include, among others, characterization of epigenetic modifications, microRNA expression, the gut microbiome, the transcriptome, and evaluation of specific mRNA expression via quantitative reverse transcription polmerase chain reaction (RT-qPCR) in fetuses and offspring of obese females. This work will review the data from animal models and human fluids/cells regarding the effects of maternal obesity on fetal and offspring neurodevelopment and cardiometabolic outcomes, with a particular focus on molecular approaches.
Collapse
Affiliation(s)
- Caterina Neri
- Department of Obstetrics and Gynecology, Università Cattolica del Sacro Cuore, Rome 00100, Italy
| | - Andrea G Edlow
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, Tufts Medical Center, Boston, Massachusetts 02111 Mother Infant Research Institute, Tufts Medical Center, Boston, Massachusetts 02111
| |
Collapse
|
49
|
Strandvik B. Perinatal programming by diets with essential fatty acid deficient/high saturated fatty acids or different n‐6/n‐3 ratios for diseases in adulthood. EUR J LIPID SCI TECH 2015. [DOI: 10.1002/ejlt.201400516] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Birgitta Strandvik
- Department of Biosciences and NutritionKarolinska InstitutetStockholmSweden
| |
Collapse
|
50
|
Docherty JEB, Manno JE, McDermott JE, DiAngelo JR. Mio acts in the Drosophila brain to control nutrient storage and feeding. Gene 2015; 568:190-5. [PMID: 26024590 DOI: 10.1016/j.gene.2015.05.055] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 04/01/2015] [Accepted: 05/21/2015] [Indexed: 12/20/2022]
Abstract
Animals recognize the availability of nutrients and regulate the intake and storage of these nutrients accordingly. However, the molecular mechanisms underlying nutrient sensing and subsequent changes in behavior and metabolism are not fully understood. Mlx interactor (Mio), the Drosophila homolog of carbohydrate response element binding protein (ChREBP), functions as a transcription factor in the fat body of the fly to control triglyceride storage as well as feeding, suggesting that Mio may act in a nutrient-sensing pathway to coordinate food consumption and metabolism. Here, we show that Mio functions in neurons in Drosophila to regulate feeding and nutrient storage. Pan-neuronal disruption of Mio function leads to increased triglyceride and glycogen storage, and this phenotype is not due to increased food consumption. Interestingly, targeted disruption of Mio specifically in the insulin-producing cells (IPCs) has little effect on nutrient storage, but increases food consumption suggesting that Mio acts in these neurons to control feeding behavior. Since Mio is a transcription factor, one possible way Mio may act in the IPCs to control feeding is through regulating the expression of Drosophila insulin-like peptides (dilps) or drosulfakinin (dsk), neuropeptides produced in the IPCs. Consistent with this hypothesis, IPC-specific knockdown of Mio leads to an increase in dilp3 expression, while not affecting dilp2, 5 or dsk levels. Together, this study indicates a new function for Mio in the Drosophila brain and specifically in the IPCs, controlling neuropeptide gene expression, feeding and metabolism in accordance with nutrient availability.
Collapse
Affiliation(s)
| | - Joseph E Manno
- Department of Biology, Hofstra University, Hempstead, NY 11549, USA
| | | | - Justin R DiAngelo
- Department of Biology, Hofstra University, Hempstead, NY 11549, USA; Hofstra North Shore-LIJ School of Medicine at Hofstra University, Hempstead, NY 11549, USA.
| |
Collapse
|