1
|
Chavatte-Palmer P, Couturier-Tarrade A, Rousseau-Ralliard D. Intra-uterine programming of future fertility. Reprod Domest Anim 2024; 59:e14475. [PMID: 37942852 DOI: 10.1111/rda.14475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 09/05/2023] [Indexed: 11/10/2023]
Abstract
The developmental origins of health and disease (DOHaD) shows that a relationship exists between parental environment at large, foeto-placental development and the risk for the offspring to develop non-transmittable disease(s) in adulthood. This concept has been validated in both humans and livestock. In mammals, after fertilization and time spent free in the maternal reproductive tract, the embryo develops a placenta that, in close relationship with maternal endometrium, is the organ responsible for exchanges between dam and foetus. Any modification of the maternal environment can lead to adaptive mechanisms affecting placental morphology, blood flow, foetal-maternal exchanges (transporters) and/or endocrine function, ultimately modifying placental efficiency. Among deleterious environments, undernutrition, protein restriction, overnutrition, micronutrient deficiencies and food contaminants can be outlined. When placental adaptive capacities become insufficient, foetal growth and organ formation is no longer optimal, including foetal gonadal formation and maturation, which can affect subsequent offspring fertility. Since epigenetic mechanisms have been shown to be key to foetal programming, epigenetic modifications of the gametes may also occur, leading to inter-generational effects. After briefly describing normal gonadal development in domestic species and inter-species differences, this review highlights the current knowledge on intra-uterine programming of offspring fertility with a focus on domestic animals and underlines the importance to assess transgenerational effects on offspring fertility at a time when new breeding systems are developed to face the current climate changes.
Collapse
Affiliation(s)
- Pascale Chavatte-Palmer
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort, France
| | - Anne Couturier-Tarrade
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort, France
| | - Delphine Rousseau-Ralliard
- Université Paris-Saclay, UVSQ, INRAE, BREED, Jouy-en-Josas, France
- Ecole Nationale Vétérinaire d'Alfort, BREED, Maisons-Alfort, France
| |
Collapse
|
2
|
Skowronski AA, Leibel RL, LeDuc CA. Neurodevelopmental Programming of Adiposity: Contributions to Obesity Risk. Endocr Rev 2024; 45:253-280. [PMID: 37971140 PMCID: PMC10911958 DOI: 10.1210/endrev/bnad031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 09/29/2023] [Accepted: 10/19/2023] [Indexed: 11/19/2023]
Abstract
This review analyzes the published evidence regarding maternal factors that influence the developmental programming of long-term adiposity in humans and animals via the central nervous system (CNS). We describe the physiological outcomes of perinatal underfeeding and overfeeding and explore potential mechanisms that may mediate the impact of such exposures on the development of feeding circuits within the CNS-including the influences of metabolic hormones and epigenetic changes. The perinatal environment, reflective of maternal nutritional status, contributes to the programming of offspring adiposity. The in utero and early postnatal periods represent critically sensitive developmental windows during which the hormonal and metabolic milieu affects the maturation of the hypothalamus. Maternal hyperglycemia is associated with increased transfer of glucose to the fetus driving fetal hyperinsulinemia. Elevated fetal insulin causes increased adiposity and consequently higher fetal circulating leptin concentration. Mechanistic studies in animal models indicate important roles of leptin and insulin in central and peripheral programming of adiposity, and suggest that optimal concentrations of these hormones are critical during early life. Additionally, the environmental milieu during development may be conveyed to progeny through epigenetic marks and these can potentially be vertically transmitted to subsequent generations. Thus, nutritional and metabolic/endocrine signals during perinatal development can have lifelong (and possibly multigenerational) impacts on offspring body weight regulation.
Collapse
Affiliation(s)
- Alicja A Skowronski
- Division of Molecular Genetics, Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Rudolph L Leibel
- Division of Molecular Genetics, Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Charles A LeDuc
- Division of Molecular Genetics, Department of Pediatrics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY 10032, USA
- Naomi Berrie Diabetes Center, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
3
|
Higgins MBA, Glendining KA, Jasoni CL. The temporal and spatial pattern of leptin receptor-expressing cells in the developing mouse hypothalamus. J Neuroendocrinol 2024; 36:e13366. [PMID: 38279680 DOI: 10.1111/jne.13366] [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: 07/27/2023] [Revised: 12/10/2023] [Accepted: 12/30/2023] [Indexed: 01/28/2024]
Abstract
The arcuate nucleus is a crucial hypothalamic brain region involved in regulating body weight homeostasis. Neurons within the arcuate nucleus respond to peripheral metabolic signals, such as leptin, and relay these signals via neuronal projections to brain regions both within and outside the hypothalamus, ultimately causing changes in an animal's behaviour and physiology. There is a substantial amount of evidence to indicate that leptin is intimately involved with the postnatal development of arcuate nucleus melanocortin circuitry. Further, it is clear that leptin signalling directly in the arcuate nucleus is required for circuitry development. However, as leptin receptor long isoform (Leprb) mRNA is expressed in multiple nuclei within the developing hypothalamus, including the postsynaptic target regions of arcuate melanocortin projections, this raises the possibility that leptin also signals in these nuclei to promote circuitry development. Here, we used RT-qPCR and RNAscope® to reveal the spatio-temporal pattern of Leprb mRNA in the early postnatal mouse hypothalamus. We found that Leprb mRNA expression increased significantly in the arcuate nucleus, ventromedial nucleus and paraventricular nucleus of the hypothalamus from P8, in concert with the leptin surge. In the dorsomedial nucleus of the hypothalamus, increases in Leprb mRNA were slightly later, increasing significantly from P12. Using duplex RNAscope®, we found Leprb co-expressed with Sim1, Pou3f2, Mc4r and Bdnf in the paraventricular nucleus at P8. Together, these data suggest that leptin may signal in a subset of neurons postsynaptic to arcuate melanocortin neurons, as well as within the arcuate nucleus itself, to promote the formation of arcuate melanocortin circuitry during the early postnatal period.
Collapse
Affiliation(s)
- Matt B A Higgins
- Centre for Neuroendocrinology, Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Kelly A Glendining
- Centre for Neuroendocrinology, Department of Physiology, University of Otago, Dunedin, New Zealand
| | - Christine L Jasoni
- Centre for Neuroendocrinology, Department of Anatomy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
4
|
Di Gesù CM, Buffington SA. The early life exposome and autism risk: a role for the maternal microbiome? Gut Microbes 2024; 16:2385117. [PMID: 39120056 PMCID: PMC11318715 DOI: 10.1080/19490976.2024.2385117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 08/10/2024] Open
Abstract
Autism spectrum disorders (ASD) are highly heritable, heterogeneous neurodevelopmental disorders characterized by clinical presentation of atypical social, communicative, and repetitive behaviors. Over the past 25 years, hundreds of ASD risk genes have been identified. Many converge on key molecular pathways, from translational control to those regulating synaptic structure and function. Despite these advances, therapeutic approaches remain elusive. Emerging data unearthing the relationship between genetics, microbes, and immunity in ASD suggest an integrative physiology approach could be paramount to delivering therapeutic breakthroughs. Indeed, the advent of large-scale multi-OMIC data acquisition, analysis, and interpretation is yielding an increasingly mechanistic understanding of ASD and underlying risk factors, revealing how genetic susceptibility interacts with microbial genetics, metabolism, epigenetic (re)programming, and immunity to influence neurodevelopment and behavioral outcomes. It is now possible to foresee exciting advancements in the treatment of some forms of ASD that could markedly improve quality of life and productivity for autistic individuals. Here, we highlight recent work revealing how gene X maternal exposome interactions influence risk for ASD, with emphasis on the intrauterine environment and fetal neurodevelopment, host-microbe interactions, and the evolving therapeutic landscape for ASD.
Collapse
Affiliation(s)
- Claudia M. Di Gesù
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
| | - Shelly A. Buffington
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
5
|
Karlen-Amarante M, Bassi M, Barbosa RM, Sá JM, Menani JV, Colombari E, Zoccal DB, Colombari DSA. Maternal high-fat diet changes breathing pattern and causes excessive sympathetic discharge in juvenile offspring rat. Am J Physiol Lung Cell Mol Physiol 2023; 325:L662-L674. [PMID: 37786934 DOI: 10.1152/ajplung.00013.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 06/28/2023] [Accepted: 09/13/2023] [Indexed: 10/04/2023] Open
Abstract
Early life over-nutrition, as experienced in maternal obesity, is a risk factor for developing cardiorespiratory and metabolic diseases. Here we investigated the effects of high-fat diet (HFD) consumption on the breathing pattern and sympathetic discharge to blood vessels in juvenile offspring from dams fed with HFD (O-HFD). Adult female Holtzman rats were given a standard diet (SD) or HFD from 6 wk before gestation to weaning. At weaning (P21), the male offspring from SD dams (O-SD) and O-HFD received SD until the experimental day (P28-P45). Nerve recordings performed in decerebrated in situ preparations demonstrated that O-HFD animals presented abdominal expiratory hyperactivity under resting conditions and higher vasoconstrictor sympathetic activity levels. The latter was associated with blunted respiratory-related oscillations in sympathetic activity, especially in control animals. When exposed to elevated hypercapnia or hypoxia levels, the O-HFD animals mounted similar ventilatory and respiratory motor responses as the control animals. Hypercapnia and hypoxia exposure also increased sympathetic activity in both groups but did not reinstate the respiratory-sympathetic coupling in the O-HFD rats. In freely behaving conditions, O-HFD animals exhibited higher resting pulmonary ventilation and larger variability of arterial pressure levels than the O-SD animals due to augmented sympathetic modulation of blood vessel diameter. Maternal obesity modified the functioning of cardiorespiratory systems in offspring at a young age, inducing active expiration and sympathetic overactivity under resting conditions. These observations represent new evidence about pregnancy-related complications that lead to the development of respiratory distress and hypertension in children of obese mothers.NEW & NOTEWORTHY Maternal obesity is a risk factor for developing cardiorespiratory and metabolic diseases. This study highlights the changes on the breathing pattern and sympathetic discharge to blood vessels in juvenile offspring from dams fed with HFD. Maternal obesity modified the functioning of cardiorespiratory systems in offspring, inducing active expiration and sympathetic overactivity. These observations represent new evidence about pregnancy-related complications that lead to the development of respiratory distress and hypertension in children of obese mothers.
Collapse
Affiliation(s)
- Marlusa Karlen-Amarante
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| | - Mirian Bassi
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| | - Rafaela Moreira Barbosa
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| | - Jéssica Matheus Sá
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| | - José Vanderlei Menani
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| | - Eduardo Colombari
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| | - Daniel B Zoccal
- Department of Physiology and Pathology, School of Dentistry, São Paulo State University (UNESP), Araraquara, Brazil
| | | |
Collapse
|
6
|
Milà-Guasch M, Ramírez S, Llana SR, Fos-Domènech J, Dropmann LM, Pozo M, Eyre E, Gómez-Valadés AG, Obri A, Haddad-Tóvolli R, Claret M. Maternal emulsifier consumption programs offspring metabolic and neuropsychological health in mice. PLoS Biol 2023; 21:e3002171. [PMID: 37616199 PMCID: PMC10449393 DOI: 10.1371/journal.pbio.3002171] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 05/24/2023] [Indexed: 08/26/2023] Open
Abstract
Modern lifestyle is associated with a major consumption of ultra-processed foods (UPF) due to their practicality and palatability. The ingestion of emulsifiers, a main additive in UPFs, has been related to gut inflammation, microbiota dysbiosis, adiposity, and obesity. Maternal unbalanced nutritional habits during embryonic and perinatal stages perturb offspring's long-term metabolic health, thus increasing obesity and associated comorbidity risk. However, whether maternal emulsifier consumption influences developmental programming in the offspring remains unknown. Here, we show that, in mice, maternal consumption of dietary emulsifiers (1% carboxymethyl cellulose (CMC) and 1% P80 in drinking water), during gestation and lactation, perturbs the development of hypothalamic energy balance regulation centers of the progeny, leads to metabolic impairments, cognition deficits, and induces anxiety-like traits in a sex-specific manner. Our findings support the notion that maternal consumption of emulsifiers, common additives of UPFs, causes mild metabolic and neuropsychological malprogramming in the progeny. Our data call for nutritional advice during gestation.
Collapse
Affiliation(s)
- Maria Milà-Guasch
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Sara Ramírez
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Sergio R. Llana
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Júlia Fos-Domènech
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Lea Maria Dropmann
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Macarena Pozo
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Elena Eyre
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Alicia G. Gómez-Valadés
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Arnaud Obri
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Roberta Haddad-Tóvolli
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Marc Claret
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
- School of Medicine, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
7
|
Koshko L, Scofield S, Debarba L, Stilgenbauer L, Fakhoury P, Jayarathne H, Perez-Mojica JE, Griggs E, Lempradl A, Sadagurski M. Prenatal benzene exposure in mice alters offspring hypothalamic development predisposing to metabolic disease in later life. CHEMOSPHERE 2023; 330:138738. [PMID: 37084897 PMCID: PMC10199724 DOI: 10.1016/j.chemosphere.2023.138738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 04/10/2023] [Accepted: 04/18/2023] [Indexed: 05/03/2023]
Abstract
Maternal exposure to environmental contaminants during pregnancy poses a significant threat to a developing fetus, as these substances can easily cross the placenta and disrupt the neurodevelopment of offspring. Specifically, the hypothalamus is essential in the regulation of metabolism, notably during critical windows of development. An abnormal hormonal and inflammatory milieu during development can trigger persistent changes in the function of hypothalamic circuits, leading to long-lasting effects on the body's energy homeostasis and metabolism. We recently demonstrated that gestational exposure to clinically relevant levels of benzene induces severe metabolic dysregulation in the offspring. Given the central role of the hypothalamus in metabolic control, we hypothesized that prenatal exposure to benzene impacts hypothalamic development, contributing to the adverse metabolic effects in the offspring. C57BL/6JB dams were exposed to benzene at 50 ppm in the inhalation chambers exclusively during pregnancy (from E0.5 to E19). Transcriptomic analysis of the exposed offspring at postnatal day 21 (P21) revealed hypothalamic changes in genes related to metabolic regulation, inflammation, and neurodevelopment exclusively in males. Moreover, the hypothalamus of prenatally benzene-exposed male offspring displayed alterations in orexigenic and anorexigenic projections, impairments in leptin signaling, and increased microgliosis. Additional exposure to benzene during lactation did not promote further microgliosis or astrogliosis in the offspring, while the high-fat diet (HFD) challenge in adulthood exacerbated glucose metabolism and hypothalamic inflammation in benzene-exposed offspring of both sexes. These findings reveal the persistent adverse effects of prenatal benzene exposure on hypothalamic circuits and neuroinflammation, predisposing the offspring to long-lasting metabolic health conditions.
Collapse
Affiliation(s)
- Lisa Koshko
- Department of Biological Sciences, Institute of Environmental Health Sciences, Integrative Biosciences Center (IBio), Wayne State University, Detroit, MI, USA
| | - Sydney Scofield
- Department of Biological Sciences, Institute of Environmental Health Sciences, Integrative Biosciences Center (IBio), Wayne State University, Detroit, MI, USA
| | - Lucas Debarba
- Department of Biological Sciences, Institute of Environmental Health Sciences, Integrative Biosciences Center (IBio), Wayne State University, Detroit, MI, USA
| | - Lukas Stilgenbauer
- Department of Biological Sciences, Institute of Environmental Health Sciences, Integrative Biosciences Center (IBio), Wayne State University, Detroit, MI, USA
| | - Patrick Fakhoury
- Department of Biological Sciences, Institute of Environmental Health Sciences, Integrative Biosciences Center (IBio), Wayne State University, Detroit, MI, USA
| | - Hashan Jayarathne
- Department of Biological Sciences, Institute of Environmental Health Sciences, Integrative Biosciences Center (IBio), Wayne State University, Detroit, MI, USA
| | | | - Ellen Griggs
- Van Andel Research Institute, Grand Rapids, MI, USA
| | | | - Marianna Sadagurski
- Department of Biological Sciences, Institute of Environmental Health Sciences, Integrative Biosciences Center (IBio), Wayne State University, Detroit, MI, USA.
| |
Collapse
|
8
|
Donato J. Programming of metabolism by adipokines during development. Nat Rev Endocrinol 2023:10.1038/s41574-023-00828-1. [PMID: 37055548 DOI: 10.1038/s41574-023-00828-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/16/2023] [Indexed: 04/15/2023]
Abstract
The intrauterine and early postnatal periods represent key developmental stages in which an organism is highly susceptible to being permanently influenced by maternal factors and nutritional status. Strong evidence indicates that either undernutrition or overnutrition during development can predispose individuals to disease later in life, especially type 2 diabetes mellitus and obesity, a concept known as metabolic programming. Adipose tissue produces important signalling molecules that control energy and glucose homeostasis, including leptin and adiponectin. In addition to their well-characterized metabolic effects in adults, adipokines have been associated with metabolic programming by affecting different aspects of development. Therefore, alterations in the secretion or signalling of adipokines, caused by nutritional insults in early life, might lead to metabolic diseases in adulthood. This Review summarizes and discusses the potential role of several adipokines in inducing metabolic programming through their effects during development. The identification of the endocrine factors that act in early life to permanently influence metabolism represents a key step in understanding the mechanisms behind metabolic programming. Thus, future strategies aiming to prevent and treat these metabolic diseases can be designed, taking into consideration the relationship between adipokines and the developmental origins of health and disease.
Collapse
Affiliation(s)
- Jose Donato
- Department of Physiology and Biophysics, Instituto de Ciencias Biomedicas, Universidade de Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
9
|
Lutz TA. Mammalian models of diabetes mellitus, with a focus on type 2 diabetes mellitus. Nat Rev Endocrinol 2023; 19:350-360. [PMID: 36941447 DOI: 10.1038/s41574-023-00818-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/21/2023] [Indexed: 03/23/2023]
Abstract
Although no single animal model replicates all aspects of diabetes mellitus in humans, animal models are essential for the study of energy balance and metabolism control as well as to investigate the reasons for their imbalance that could eventually lead to overt metabolic diseases such as type 2 diabetes mellitus. The most frequently used animal models in diabetes mellitus research are small rodents that harbour spontaneous genetic mutations or that can be manipulated genetically or by other means to influence their nutrient metabolism and nutrient handling. Non-rodent species, including pigs, cats and dogs, are also useful models in diabetes mellitus research. This Review will outline the advantages and disadvantages of selected animal models of diabetes mellitus to build a basis for their most appropriate use in biomedical research.
Collapse
Affiliation(s)
- Thomas A Lutz
- Institute of Veterinary Physiology, Vetsuisse Faculty University of Zurich, Zurich, Switzerland.
| |
Collapse
|
10
|
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
|
11
|
Haddad-Tóvolli R, Morari J, Barbizan R, Bóbbo VC, Carraro RS, Solon C, Dragano NR, Torsoni MA, Araujo EP, Velloso LA. Maternal obesity damages the median eminence blood-brain barrier structure and function in the progeny: the beneficial impact of cross-fostering by lean mothers. Am J Physiol Endocrinol Metab 2023; 324:E154-E166. [PMID: 36598900 DOI: 10.1152/ajpendo.00268.2022] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Maternal obesity is an important risk factor for obesity, cardiovascular, and metabolic diseases in the offspring. Studies have shown that it leads to hypothalamic inflammation in the progeny, affecting the function of neurons regulating food intake and energy expenditure. In adult mice fed a high-fat diet, one of the hypothalamic abnormalities that contribute to the development of obesity is the damage of the blood-brain barrier (BBB) at the median eminence-arcuate nucleus (ME-ARC) interface; however, how the hypothalamic BBB is affected in the offspring of obese mothers requires further investigation. Here, we used confocal and transmission electron microscopy, transcript expression analysis, glucose tolerance testing, and a cross-fostering intervention to determine the impact of maternal obesity and breastfeeding on BBB integrity at the ME-ARC interface. The offspring of obese mothers were born smaller; conversely, at weaning, they presented larger body mass and glucose intolerance. In addition, maternal obesity-induced structural and functional damage of the offspring's ME-ARC BBB. By a cross-fostering intervention, some of the defects in barrier integrity and metabolism seen during development in an obesogenic diet were recovered. The offspring of obese dams breastfed by lean dams presented a reduction of body mass and glucose intolerance as compared to the offspring continuously exposed to an obesogenic environment during intrauterine and perinatal life; this was accompanied by partial recovery of the anatomical structure of the ME-ARC interface, and by the normalization of transcript expression of genes coding for hypothalamic neurotransmitters involved in energy balance and BBB integrity. Thus, maternal obesity promotes structural and functional damage of the hypothalamic BBB, which is, in part, reverted by lactation by lean mothers.NEW & NOTEWORTHY Maternal dietary habits directly influence offspring health. In this study, we aimed at determining the impact of maternal obesity on BBB integrity. We show that DIO offspring presented a leakier ME-BBB, accompanied by changes in the expression of transcripts encoding for endothelial and tanycytic proteins, as well as of hypothalamic neuropeptides. Breastfeeding in lean dams was sufficient to protect the offspring from ME-BBB disruption, providing a preventive strategy of nutritional intervention during early life.
Collapse
Affiliation(s)
- Roberta Haddad-Tóvolli
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas, Campinas, Brazil
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Joseane Morari
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas, Campinas, Brazil
| | - Roberta Barbizan
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas, Campinas, Brazil
| | - Vanessa C Bóbbo
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas, Campinas, Brazil
| | - Rodrigo S Carraro
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas, Campinas, Brazil
- Center for Anatomy Studies, University San Francisco (USF), Bragança Paulista, Brazil
| | - Carina Solon
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas, Campinas, Brazil
| | - Nathalia R Dragano
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas, Campinas, Brazil
| | - Márcio A Torsoni
- Laboratory of Metabolic Disorders, Faculty of Applied Sciences, State University of Campinas, Campinas, Limeira, Brazil
| | - Eliana P Araujo
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas, Campinas, Brazil
- School of Nursing, University of Campinas, Campinas, Brazil
| | - Licio A Velloso
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas, Campinas, Brazil
| |
Collapse
|
12
|
Koshko L, Scofield S, Debarba L, Stilgenbauer L, Sacla M, Fakhoury P, Jayarathne H, Perez-Mojica JE, Griggs E, Lempradl A, Sadagurski M. Prenatal benzene exposure alters offspring hypothalamic development predisposing to metabolic disease in later life. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.05.522910. [PMID: 36711607 PMCID: PMC9881982 DOI: 10.1101/2023.01.05.522910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
The hypothalamus is essential in the regulation of metabolism, notably during critical windows of development. An abnormal hormonal and inflammatory milieu during development can trigger persistent changes in the function of hypothalamic circuits, leading to long-lasting effects on the body’s energy homeostasis and metabolism. We recently demonstrated that gestational exposure to benzene at smoking levels induces severe metabolic dysregulation in the offspring. Given the central role of the hypothalamus in metabolic control, we hypothesized that prenatal exposure to benzene impacts hypothalamic development, contributing to the adverse metabolic effects in the offspring. C57BL/6JB dams were exposed to benzene in the inhalation chambers exclusively during pregnancy (from E0.5 to E19). The transcriptome analysis of the offspring hypothalamus at postnatal day 21 (P21) revealed changes in genes related to metabolic regulation, inflammation, and neurodevelopment exclusively in benzene-exposed male offspring. Moreover, the hypothalamus of prenatally benzene-exposed male offspring displayed alterations in orexigenic and anorexigenic projections, impairments in leptin signaling, and increased microgliosis. Additional exposure to benzene during lactation did not promote further microgliosis or astrogliosis in the offspring, while the high-fat diet (HFD) challenge in adulthood exacerbated glucose metabolism and hypothalamic inflammation in benzene-exposed offspring of both sexes. These findings reveal the persistent impact of prenatal benzene exposure on hypothalamic circuits and neuroinflammation, predisposing the offspring to long-lasting metabolic health conditions.
Collapse
|
13
|
Neuroanatomical correlates of genetic risk for obesity in children. Transl Psychiatry 2023; 13:1. [PMID: 36596778 PMCID: PMC9810659 DOI: 10.1038/s41398-022-02301-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 01/04/2023] Open
Abstract
Obesity has a strong genetic component, with up to 20% of variance in body mass index (BMI) being accounted for by common polygenic variation. Most genetic polymorphisms associated with BMI are related to genes expressed in the central nervous system. At the same time, higher BMI is associated with neurocognitive changes. However, the direct link between genetics of obesity and neurobehavioral mechanisms related to weight gain is missing. Here, we use a large sample of participants (n > 4000) from the Adolescent Brain Cognitive Development cohort to investigate how genetic risk for obesity, expressed as polygenic risk score for BMI (BMI-PRS), is related to brain and behavioral measures in adolescents. In a series of analyses, we show that BMI-PRS is related to lower cortical volume and thickness in the frontal and temporal areas, relative to age-expected values. Relatedly, using structural equation modeling, we find that lower overall cortical volume is associated with higher impulsivity, which in turn is related to an increase in BMI 1 year later. In sum, our study shows that obesity might partially stem from genetic risk as expressed in brain changes in the frontal and temporal brain areas, and changes in impulsivity.
Collapse
|
14
|
Zordão OP, Campolim CM, Yariwake VY, Castro G, Ferreira CKDO, Santos A, Norberto S, Veras MM, Saad MJA, Saldiva PHN, Kim YB, Prada PO. Maternal exposure to air pollution alters energy balance transiently according to gender and changes gut microbiota. Front Endocrinol (Lausanne) 2023; 14:1069243. [PMID: 37082122 PMCID: PMC10112381 DOI: 10.3389/fendo.2023.1069243] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 03/07/2023] [Indexed: 04/22/2023] Open
Abstract
Introduction The timing of maternal exposure to air pollution is crucial to define metabolic changes in the offspring. Here we aimed to determine the most critical period of maternal exposure to particulate matter (PM2.5) that impairs offspring's energy metabolism and gut microbiota composition. Methods Unexposed female and male C57BL/6J mice were mated. PM2.5 or filtered air (FA) exposure occurred only in gestation (PM2.5/FA) or lactation (FA/PM2.5). We studied the offspring of both genders. Results PM2.5 exposure during gestation increased body weight (BW) at birth and from weaning to young in male adulthood. Leptin levels, food intake, Agrp, and Npy levels in the hypothalamus were also increased in young male offspring. Ikbke, Tnf increased in male PM2.5/FA. Males from FA/PM2.5 group were protected from these phenotypes showing higher O2 consumption and Ucp1 in the brown adipose tissue. In female offspring, we did not see changes in BW at weaning. However, adult females from PM2.5/FA displayed higher BW and leptin levels, despite increased energy expenditure and thermogenesis. This group showed a slight increase in food intake. In female offspring from FA/PM2.5, BW, and leptin levels were elevated. This group displayed higher energy expenditure and a mild increase in food intake. To determine if maternal exposure to PM2.5 could affect the offspring's gut microbiota, we analyzed alpha diversity by Shannon and Simpson indexes and beta diversity by the Linear Discriminant Analysis (LDA) in offspring at 30 weeks. Unlike males, exposure during gestation led to higher adiposity and leptin maintenance in female offspring at this age. Gestation exposure was associated with decreased alpha diversity in the gut microbiota in both genders. Discussion Our data support that exposure to air pollution during gestation is more harmful to metabolism than exposure during lactation. Male offspring had an unfavorable metabolic phenotype at a young age. However, at an older age, only females kept more adiposity. Ultimately, our data highlight the importance of controlling air pollution, especially during gestation.
Collapse
Affiliation(s)
- Olivia Pizetta Zordão
- Department of Internal Medicine, School of Medical Science, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Clara Machado Campolim
- Department of Internal Medicine, School of Medical Science, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Victor Yuji Yariwake
- Laboratory of Environmental and Experimental Pathology, Department of Pathology, University of Sao Paulo School of Medicine, Sao Paulo, SP, Brazil
| | - Gisele Castro
- Department of Internal Medicine, School of Medical Science, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | | | - Andrey Santos
- Department of Internal Medicine, School of Medical Science, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Sónia Norberto
- Department of Internal Medicine, School of Medical Science, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Mariana Matera Veras
- Laboratory of Environmental and Experimental Pathology, Department of Pathology, University of Sao Paulo School of Medicine, Sao Paulo, SP, Brazil
| | - Mario Jose Abdalla Saad
- Department of Internal Medicine, School of Medical Science, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Paulo Hilário Nascimento Saldiva
- Laboratory of Environmental and Experimental Pathology, Department of Pathology, University of Sao Paulo School of Medicine, Sao Paulo, SP, Brazil
| | - Young-Bum Kim
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Patricia Oliveira Prada
- Department of Internal Medicine, School of Medical Science, State University of Campinas (UNICAMP), Campinas, SP, Brazil
- School of Applied Sciences, State University of Campinas (UNICAMP), Limeira, SP, Brazil
- *Correspondence: Patricia Oliveira Prada, ;
| |
Collapse
|
15
|
Kulhanek D, Abrahante Llorens JE, Buckley L, Tkac I, Rao R, Paulsen ME. Female and male C57BL/6J offspring exposed to maternal obesogenic diet develop altered hypothalamic energy metabolism in adulthood. Am J Physiol Endocrinol Metab 2022; 323:E448-E466. [PMID: 36342228 PMCID: PMC9639756 DOI: 10.1152/ajpendo.00100.2022] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/12/2022] [Accepted: 09/12/2022] [Indexed: 11/22/2022]
Abstract
Maternal obesity is exceedingly common and strongly linked to offspring obesity and metabolic disease. Hypothalamic function is critical to obesity development. Hypothalamic mechanisms causing obesity following exposure to maternal obesity have not been elucidated. Therefore, we studied a cohort of C57BL/6J dams, treated with a control or high-fat-high-sugar diet, and their adult offspring to explore potential hypothalamic mechanisms to explain the link between maternal and offspring obesity. Dams treated with obesogenic diet were heavier with mild insulin resistance, which is reflective of the most common metabolic disease in pregnancy. Adult offspring exposed to maternal obesogenic diet had no change in body weight but significant increase in fat mass, decreased glucose tolerance, decreased insulin sensitivity, elevated plasma leptin, and elevated plasma thyroid-stimulating hormone. In addition, offspring exposed to maternal obesity had decreased energy intake and activity without change in basal metabolic rate. Hypothalamic neurochemical profile and transcriptome demonstrated decreased neuronal activity and inhibition of oxidative phosphorylation. Collectively, these results indicate that maternal obesity without diabetes is associated with adiposity and decreased hypothalamic energy production in offspring. We hypothesize that altered hypothalamic function significantly contributes to obesity development. Future studies focused on neuroprotective strategies aimed to improve hypothalamic function may decrease obesity development.NEW & NOTEWORTHY Offspring exposed to maternal diet-induced obesity demonstrate a phenotype consistent with energy excess. Contrary to previous studies, the observed energy phenotype was not associated with hyperphagia or decreased basal metabolic rate but rather decreased hypothalamic neuronal activity and energy production. This was supported by neurochemical changes in the hypothalamus as well as inhibition of hypothalamic oxidative phosphorylation pathway. These results highlight the potential for neuroprotective interventions in the prevention of obesity with fetal origins.
Collapse
Affiliation(s)
- Debra Kulhanek
- Division of Neonatology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota
| | | | - Lauren Buckley
- Division of Neonatology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Ivan Tkac
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Raghavendra Rao
- Division of Neonatology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota Medical School, Minneapolis, Minnesota
| | - Megan E Paulsen
- Division of Neonatology, Department of Pediatrics, University of Minnesota Medical School, Minneapolis, Minnesota
- Minnesota Institute for the Developing Brain, Minneapolis, Minnesota
| |
Collapse
|
16
|
Morales I. Brain regulation of hunger and motivation: The case for integrating homeostatic and hedonic concepts and its implications for obesity and addiction. Appetite 2022; 177:106146. [PMID: 35753443 DOI: 10.1016/j.appet.2022.106146] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 06/16/2022] [Accepted: 06/21/2022] [Indexed: 11/19/2022]
Abstract
Obesity and other eating disorders are marked by dysregulations to brain metabolic, hedonic, motivational, and sensory systems that control food intake. Classic approaches in hunger research have distinguished between hedonic and homeostatic processes, and have mostly treated these systems as independent. Hindbrain structures and a complex network of interconnected hypothalamic nuclei control metabolic processes, energy expenditure, and food intake while mesocorticolimbic structures are though to control hedonic and motivational processes associated with food reward. However, it is becoming increasingly clear that hedonic and homeostatic brain systems do not function in isolation, but rather interact as part of a larger network that regulates food intake. Incentive theories of motivation provide a useful route to explore these interactions. Adapting incentive theories of motivation can enable researchers to better how motivational systems dysfunction during disease. Obesity and addiction are associated with profound alterations to both hedonic and homeostatic brain systems that result in maladaptive patterns of consumption. A subset of individuals with obesity may experience pathological cravings for food due to incentive sensitization of brain systems that generate excessive 'wanting' to eat. Further progress in understanding how the brain regulates hunger and appetite may depend on merging traditional hedonic and homeostatic concepts of food reward and motivation.
Collapse
Affiliation(s)
- Ileana Morales
- Department of Psychology, University of Michigan, 530 Church Street, Ann Arbor, MI, 48109-1043, USA.
| |
Collapse
|
17
|
Asuquo EA, Nwodo OFC, Assumpta AC, Orizu UN, Oziamara ON, Solomon OA. FTO gene expression in diet-induced obesity is downregulated by Solanum fruit supplementation. Open Life Sci 2022; 17:641-658. [PMID: 35800074 PMCID: PMC9202533 DOI: 10.1515/biol-2022-0067] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 02/07/2022] [Accepted: 03/05/2022] [Indexed: 11/15/2022] Open
Abstract
The Fat Mass and Obesity-associated (FTO) gene has been shown to play an important role in developing obesity, manifesting in traits such as increased body mass index, increased waist-to-hip ratio, and the distribution of adipose tissues, which increases the susceptibility to various metabolic syndromes. In this study, we evaluated the impact of fruit-based diets of Solanum melongena (SMF) and Solanum aethiopicum fruits (SAF) on the FTO gene expression levels in a high-fat diet (HFD)-induced obese animals. Our results showed that the mRNA level of the FTO gene was downregulated in the hypothalamus, and white and brown adipose tissue following three and six weeks of treatment with SMF- and SAF-based diets in the HFD-induced obese animals. Additionally, the Solanum fruit supplementation exhibited a curative effect on obesity-associated abrasions on the white adipose tissue (WAT), hypothalamus, and liver. Our findings collectively suggest the anti-obesity potential of SMF and SAF via the downregulation of the FTO gene.
Collapse
Affiliation(s)
- Edeke Affiong Asuquo
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, 410001, Nsukka, Enugu State, Nigeria
| | | | - Anosike Chioma Assumpta
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, 410001, Nsukka, Enugu State, Nigeria
| | - Uchendu Nene Orizu
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, 410001, Nsukka, Enugu State, Nigeria
| | - Okoro Nkwachukwu Oziamara
- Department of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmaceutical Sciences, University of Nigeria, 410001, Nsukka, Enugu State, Nigeria
| | - Odiba Arome Solomon
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, 410001, Nsukka, Enugu State, Nigeria
- Department of Molecular Genetics and Biotechnology, Faculty of Biological Sciences, University of Nigeria, 410001, Nsukka, Enugu State, Nigeria
| |
Collapse
|
18
|
Mediators of Amylin Action in Metabolic Control. J Clin Med 2022; 11:jcm11082207. [PMID: 35456307 PMCID: PMC9025724 DOI: 10.3390/jcm11082207] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/08/2022] [Accepted: 04/13/2022] [Indexed: 02/06/2023] Open
Abstract
Amylin (also called islet amyloid polypeptide (IAPP)) is a pancreatic beta-cell hormone that is co-secreted with insulin in response to nutrient stimuli. The last 35 years of intensive research have shown that amylin exerts important physiological effects on metabolic control. Most importantly, amylin is a physiological control of meal-ending satiation, and it limits the rate of gastric emptying and reduces the secretion of pancreatic glucagon, in particular in postprandial states. The physiological effects of amylin and its analogs are mediated by direct brain activation, with the caudal hindbrain playing the most prominent role. The clarification of the structure of amylin receptors, consisting of the calcitonin core receptor plus receptor-activity modifying proteins, aided in the development of amylin analogs with a broad pharmacological profile. The general interest in amylin physiology and pharmacology was boosted by the finding that amylin is a sensitizer to the catabolic actions of leptin. Today, amylin derived analogs are considered to be among the most promising approaches for the pharmacotherapy against obesity. At least in conjunction with insulin, amylin analogs are also considered important treatment options in diabetic patients, so that new drugs may soon be added to the only currently approved compound pramlintide (Symlin®). This review provides a brief summary of the physiology of amylin’s mode of actions and its role in the control of the metabolism, in particular energy intake and glucose metabolism.
Collapse
|
19
|
Bouret SG. Developmental programming of hypothalamic melanocortin circuits. Exp Mol Med 2022; 54:403-413. [PMID: 35474338 PMCID: PMC9076880 DOI: 10.1038/s12276-021-00625-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 01/14/2023] Open
Abstract
The melanocortin system plays a critical role in the central regulation of food intake and energy balance. This system consists of neurons producing pro-opiomelanocortin (POMC), melanocortin receptors (MC4Rs), and the endogenous antagonist agouti-related peptide (AgRP). Pomc and Mc4r deficiency in rodents and humans causes early onset of obesity, whereas a loss of Agrp function is associated with leanness. Accumulating evidence shows that many chronic diseases, including obesity, might originate during early life. The melanocortin system develops during a relatively long period beginning during embryonic life with the birth of POMC and AgRP neurons and continuing postnatally with the assembly of their neuronal circuitry. The development of the melanocortin system requires the tight temporal regulation of molecular factors, such as transcription factors and axon guidance molecules, and cellular mechanisms, such as autophagy. It also involves a complex interplay of endocrine and nutritional factors. The disruption of one or more of these developmental factors can lead to abnormal maturation and function of the melanocortin system and has profound metabolic consequences later in life.
Collapse
Affiliation(s)
- Sebastien G Bouret
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition Research Center, UMR-S 1172, Lille, 59000, France.
- University of Lille, FHU 1,000 Days for Health, Lille, 59000, France.
| |
Collapse
|
20
|
Leuthardt AS, Bayer J, Monné Rodríguez JM, Boyle CN. Influence of High Energy Diet and Polygenic Predisposition for Obesity on Postpartum Health in Rat Dams. Front Physiol 2022; 12:772707. [PMID: 35222059 PMCID: PMC8867007 DOI: 10.3389/fphys.2021.772707] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 12/16/2021] [Indexed: 02/06/2023] Open
Abstract
It is estimated that 30% of pregnant women worldwide are overweight or obese, leading to adverse health effects for both mother and child. Women with obesity during pregnancy are at higher risk for developing both metabolic and mental disorders, such as diabetes and depression. Numerous studies have used rodent models of maternal obesity to understand its consequences on the offspring, yet characterization of changes in the dams is rare, and most rodent models rely solely on a high fat diet to induce maternal obesity, without regarding genetic propensity for obesity. Here we present the influence of both peripartum high energy diet (HE) and obesity-proneness on maternal health using selectively bred diet-resistant (DR) and diet-induced obese (DIO) rat dams. Outbred Sprague-Dawley rats were challenged with HE diet prior to mating and bred according to their propensity to gain weight. The original outbred breeding dams (F0) were maintained on low-fat chow during pregnancy and lactation. By comparison, the F1 dams consuming HE diet during pregnancy and lactation displayed higher gestational body weight gain (P < 0.01), and HE diet caused increased meal size and reduced meal frequency (P < 0.001). Sensitivity to the hormone amylin was preserved during pregnancy, regardless of diet. After several rounds of selective breeding, DIO and DR dams from generation F3 were provided chow or HE during pregnancy and lactation and assessed for their postpartum physiology and behaviors. We observed strong diet and phenotype effects on gestational weight gain, with DIO-HE dams gaining 119% more weight than DR-chow (P < 0.001). A high-resolution analysis of maternal behaviors did not detect main effects of diet or phenotype, but a subset of DIO dams showed delayed nursing behavior (P < 0.05). In generation F6/F7 dams, effects on gestational weight gain persisted (P < 0.01), and we observed a main effect of phenotype during a sucrose preference test (P < 0.05), with DIO-chow dams showing lower sucrose preference than DR controls (P < 0.05). Both DIO and DR dams consuming HE diet had hepatic steatosis (P < 0.001) and exhibited reduced leptin sensitivity in the arcuate nucleus (P < 0.001). These data demonstrate that both diet and genetic obesity-proneness have consequences on maternal health.
Collapse
Affiliation(s)
- Andrea S. Leuthardt
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Julia Bayer
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Josep M. Monné Rodríguez
- Laboratory for Animal Model Pathology (LAMP), Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Christina N. Boyle
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
- *Correspondence: Christina N. Boyle,
| |
Collapse
|
21
|
Maternal exercise and high-fat diet affect hypothalamic neural projections in rat offspring in a sex-specific manner. J Nutr Biochem 2022; 103:108958. [DOI: 10.1016/j.jnutbio.2022.108958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 10/20/2021] [Accepted: 01/05/2022] [Indexed: 11/23/2022]
|
22
|
Kovacs P, Hajnal A. Short-term high-fat diet consumption increases body weight and body adiposity and alters brain stem taste information processing in rats. Chem Senses 2022; 47:6673811. [PMID: 35997757 DOI: 10.1093/chemse/bjac020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Diet-induced obesity is known to develop whether exposed to a high-energy diet (HED) or a high-fat diet (HFD). However, it is still not clear whether the elevated energy content or the macronutrient imbalance is the key factor in early disease progression. Therefore, this study compared the short-term effects of 2 widely used rodent obesogenic diets, an HFD with 60 kcal% fat content and a carbohydrate-based HED, on the body weight, body fat content, glucose tolerance, and neuronal taste responses in rats. We found that only HFD induced an early significant body weight increase compared with the control normal diet (ND) group, starting on week 4, and resulting in a significantly elevated body adiposity compared with both the ND and HED groups. Oral glucose tolerance test revealed no difference across groups. Subsequently, we also found that HFD resulted in a significant body weight gain even under energy-restricted (isocaloric to ND) conditions. In vivo electrophysiological recordings revealed that only the ad libitum HFD and not the isocaloric-HFD altered the brain stem gustatory neural responses to oral taste stimulation. In conclusion, this study showed that increased fat intake might result in significant body weight gain even under isocaloric and metabolically healthy conditions and demonstrated changes in central taste processing in an early stage of dietary obesity. A better understanding of these initial physiological changes may offer new drug targets for preventing obesity.
Collapse
Affiliation(s)
- Peter Kovacs
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA
| | - Andras Hajnal
- Department of Neural and Behavioral Sciences, College of Medicine, The Pennsylvania State University, Hershey, PA 17033, USA
| |
Collapse
|
23
|
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
|
24
|
Freeman AK, Glendining KA, Jasoni CL. Developmental genes controlling neural circuit formation are expressed in the early postnatal hypothalamus and cellular lining of the third ventricle. J Neuroendocrinol 2021; 33:e13020. [PMID: 34423876 DOI: 10.1111/jne.13020] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 07/20/2021] [Accepted: 07/27/2021] [Indexed: 02/07/2023]
Abstract
The arcuate nucleus of the hypothalamus is central in the regulation of body weight homeostasis through its ability to sense peripheral metabolic signals and relay them, through neural circuits, to other brain areas, ultimately affecting physiological and behavioural changes. The early postnatal development of these neural circuits is critical for normal body weight homeostasis, such that perturbations during this critical period can lead to obesity. The role for peripheral regulators of body weight homeostasis, including leptin, insulin and ghrelin, in this postnatal development is well described, yet some of the fundamental processes underpinning axonal and dendritic growth remain unclear. Here, we hypothesised that molecules known to regulate axonal and dendritic growth processes in other areas of the developing brain would be expressed in the postnatal arcuate nucleus and/or target nuclei where they would function to mediate the development of this circuitry. Using state-of-the-art RNAscope® technology, we have revealed the expression patterns of genes encoding Dcc/Netrin-1, Robo1/Slit1 and Fzd5/Wnt5a receptor/ligand pairs in the early postnatal mouse hypothalamus. We found that individual genes had unique expression patterns across developmental time in the arcuate nucleus, paraventricular nucleus of the hypothalamus, ventromedial nucleus of the hypothalamus, dorsomedial nucleus of the hypothalamus, median eminence and, somewhat unexpectedly, the third ventricle epithelium. These observations indicate a number of new molecular players in the development of neural circuits regulating body weight homeostasis, as well as novel molecular markers of tanycyte heterogeneity.
Collapse
Affiliation(s)
- Alice Katherine Freeman
- Centre for Neuroendocrinology, Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Kelly A Glendining
- Centre for Neuroendocrinology, Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Christine L Jasoni
- Centre for Neuroendocrinology, Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
25
|
Deer EM, Phillips CL, Welch BA, Himel AR, Duncan BC, Spann RA, Grayson BE. Dysregulated appetitive leptin signaling in male rodent offspring from post-bariatric dams. Curr Res Physiol 2020; 3:50-58. [PMID: 33644768 PMCID: PMC7909366 DOI: 10.1016/j.crphys.2020.11.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Bariatric surgery produces significant positive benefits to recipients such as resolution of various obesity-related comorbidities, including improved reproductive function. Females of childbearing age seek bariatric surgical remedies to improve their chance of successful pregnancy; however, limited knowledge exists on the impact of surgical weight loss to subsequently born offspring. We previously reported that circulating leptin levels were reduced in pregnant females having previously received vertical sleeve gastrectomy (VSG) in comparison to control dams having received Sham surgery. Furthermore, the levels of leptin receptors in the VSG placenta were also reduced in comparison to controls in. These data suggest there may be a significant difference in leptin signaling during pregnancy that may produce an altered developmental environment for the offspring. Here, we investigate the adult offspring of dams having received VSG or Sham-VSG prior to pregnancy. Endogenous fasting plasma leptin levels were not different between Sham and VSG offspring. Fasting leptin receptor mRNA in the medial basal hypothalamus (MBH) was elevated in VSG offspring in comparison to Sham. Intraperitoneal administration of exogenous leptin produced reductions in acute food intake in male Sham offspring, but did not reduce food intake at any time point measured in male VSG offspring. Using Western blot, we identified elevated pSTAT3 and pSTAT3/STAT3 ratios in the MBH of post-VSG offspring in comparison to controls. Using immunohistochemistry, we found an increased number of pSTAT positive cells in the arcuate nucleus in the Sham offspring in comparison to VSG. In contrast, within the paraventricular and ventromedial hypothalamic nuclei the VSG offspring had elevated numbers of pSTAT-positive cells in comparison to controls. Collectively, these data support our hypothesis that leptin signaling is dysregulated in VSG offspring and may be partially responsible for the long-term impact of maternal bariatric surgery on the metabolic health of offspring. Pregnancies following bariatric surgeries have reduced circulating leptin and increased leptin receptors in placenta. Offspring of dams having previously received bariatric surgery have altered growth trajectories during postnatal life. The male offspring experience have elevated expression of hypothalamic leptin receptors. The bariatric offspring are non-responsive to the anorexigenic effects of exogenous leptin administration. pSTAT in the hypothalamus of post-bariatric offspring is elevated without a change in food intake. Leptin signaling may be responsible for long-term metabolic changes in post-bariatric offspring.
Collapse
Affiliation(s)
- Evangeline M Deer
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Charles L Phillips
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Bradley A Welch
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Alexandra R Himel
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Brittany C Duncan
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Redin A Spann
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Bernadette E Grayson
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
26
|
Li C, Xu JJ, Hu HT, Shi CY, Yu CJ, Sheng JZ, Wu YT, Huang HF. Amylin receptor insensitivity impairs hypothalamic POMC neuron differentiation in the male offspring of maternal high-fat diet-fed mice. Mol Metab 2020; 44:101135. [PMID: 33279727 PMCID: PMC7773963 DOI: 10.1016/j.molmet.2020.101135] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 11/29/2020] [Accepted: 11/29/2020] [Indexed: 11/26/2022] Open
Abstract
Objective Amylin was found to regulate glucose and lipid metabolism by acting on the arcuate nucleus of the hypothalamus (ARC). Maternal high-fat diet (HFD) induces sex-specific metabolic diseases mediated by the ARC in offspring. This study was performed to explore 1) the effect of maternal HFD-induced alterations in amylin on the differentiation of hypothalamic neurons and metabolic disorders in male offspring and 2) the specific molecular mechanism underlying the regulation of amylin and its receptor in response to maternal HFD. Methods Maternal HFD and gestational hyper-amylin mice models were established to explore the role of hypothalamic amylin and receptor activity-modifying protein 3 (Ramp3) in regulating offspring metabolism. RNA pull-down, mass spectrometry, RNA immunoprecipitation, and RNA decay assays were performed to investigate the mechanism underlying the influence of maternal HFD on Ramp3 deficiency in the fetal hypothalamus. Results Male offspring with maternal HFD grew heavier and developed metabolic disorders, whereas female offspring with maternal HFD showed a slight increase in body weight and did not develop metabolic disorders compared to those exposed to maternal normal chow diet (NCD). Male offspring exposed to a maternal HFD had hyperamylinemia from birth until adulthood, which was inconsistent with offspring exposed to maternal NCD. Hyperamylinemia in the maternal HFD-exposed male offspring might be attributed to amylin accumulation following Ramp3 deficiency in the fetal hypothalamus. After Ramp3 knockdown in hypothalamic neural stem cells (htNSCs), amylin was found to fail to promote the differentiation of anorexigenic alpha-melanocyte-stimulating hormone-proopiomelanocortin (α-MSH-POMC) neurons but not orexigenic agouti-related protein-neuropeptide Y (AgRP-Npy) neurons. An investigation of the mechanism involved showed that IGF2BP1 could specifically bind to Ramp3 in htNSCs and maintain its mRNA stability. Downregulation of IGF2BP1 in htNSCs in the HFD group could decrease Ramp3 expression and lead to an impairment of α-MSH-POMC neuron differentiation. Conclusions These findings suggest that gestational exposure to HFD decreases the expression of IGF2BP1 in the hypothalami of male offspring and destabilizes Ramp3 mRNA, which leads to amylin resistance. The subsequent impairment of POMC neuron differentiation induces sex-specific metabolic disorders in adulthood. Maternal HFD leads to Ramp3 deficiency in fetal hypothalami of male offspring. IGF2BP1 binds to Ramp3 in htNSCs specifically and maintains its mRNA stability. Maternal HFD decreases Ramp3 in htNSCs via downregulating IGF2BP1. Ramp3 deficiency induced by maternal HFD results in amylin resistance in htNSCs. Amylin resistance induced by Ramp3 deficiency impairs POMC neuron differentiation.
Collapse
Affiliation(s)
- Cheng Li
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China; Institute of Embryo-Fetal Original Adult Disease Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jing-Jing Xu
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China; Institute of Embryo-Fetal Original Adult Disease Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong-Tao Hu
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China; Institute of Embryo-Fetal Original Adult Disease Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chao-Yi Shi
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China; Institute of Embryo-Fetal Original Adult Disease Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chuan-Jin Yu
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China; Institute of Embryo-Fetal Original Adult Disease Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jian-Zhong Sheng
- Department of Pathology and Pathophysiology, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yan-Ting Wu
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China; Institute of Embryo-Fetal Original Adult Disease Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - He-Feng Huang
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China; Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China; Institute of Embryo-Fetal Original Adult Disease Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
27
|
Aylwin CF, Lomniczi A. Sirtuin (SIRT)-1: At the crossroads of puberty and metabolism. CURRENT OPINION IN ENDOCRINE AND METABOLIC RESEARCH 2020; 14:65-72. [PMID: 32905232 PMCID: PMC7467505 DOI: 10.1016/j.coemr.2020.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
In the arcuate nucleus (ARC) of the hypothalamus reside two neuronal systems in charge of regulating feeding control and reproductive development. The melanocortin system responds to metabolic fluctuations adjusting food intake, whereas kisspeptin neurons are in charge of the excitatory control of Gonadotropin Hormone Releasing Hormone (GnRH) neurons. While it is known that the melanocortin system regulates GnRH neuronal activity, it was recently demonstrated that kisspeptin neurons not only innervate melanocortin neurons, but also play an active role in the control of metabolism. These two neuronal systems are intricately interconnected forming loops of stimulation and inhibition according to metabolic status. Furthermore, intracellular and epigenetic pathways respond to external environmental signals by changing DNA conformation and gene expression. Here we review the role of Silent mating type Information Regulation 2 homologue 1 (Sirt1), a class III NAD+ dependent protein deacetylase, in the ARC control of pubertal development and feeding behavior.
Collapse
Affiliation(s)
- Carlos F Aylwin
- Division of Neuroscience, Oregon National Primate Research Center, OHSU, Beaverton, OR, USA
| | - Alejandro Lomniczi
- Division of Neuroscience, Oregon National Primate Research Center, OHSU, Beaverton, OR, USA
| |
Collapse
|
28
|
Huang KP, Goodson ML, Vang W, Li H, Page AJ, Raybould HE. Leptin signaling in vagal afferent neurons supports the absorption and storage of nutrients from high-fat diet. Int J Obes (Lond) 2020; 45:348-357. [PMID: 32917985 PMCID: PMC7854885 DOI: 10.1038/s41366-020-00678-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 07/30/2020] [Accepted: 09/03/2020] [Indexed: 12/18/2022]
Abstract
Objective: Activation of vagal afferent neurons (VAN) by postprandial gastrointestinal signals terminates feeding and facilitates nutrient digestion and absorption. Leptin modulates responsiveness of VAN to meal-related gastrointestinal signals. Rodents with high-fat diet (HF) feeding develop leptin resistance that impairs responsiveness of VAN. We hypothesized that lack of leptin signaling in VAN reduces responses to meal-related signals, which in turn decreases absorption of nutrients and energy storage from high-fat, calorically dense food. Methods: Mice with conditional deletion of the leptin receptor from VAN (Nav1.8-Cre/LepRfl/fl; KO) were used in this study. Six-week-old male mice were fed a 45% HF for 4 weeks; metabolic phenotype, food intake, and energy expenditure were measured. Absorption and storage of nutrients were investigated in the refed state. Results: After 4 weeks of HF feeding, KO mice gained less body weight and fat mass that WT controls, but this was not due to differences in food intake or energy expenditure. KO mice had reduced expression of carbohydrate transporters and absorption of carbohydrate in the jejunum. KO mice had fewer hepatic lipid droplets and decreased expression of de novo lipogenesis-associated enzymes and lipoproteins for endogenous lipoprotein pathway in liver, suggesting decreased long-term storage of carbohydrate in KO mice. Conclusions: Impairment of leptin signaling in VAN reduces responsiveness to gastrointestinal signals, which reduces intestinal absorption of carbohydrates and de novo lipogenesis resulting in reduced long-term energy storage. This study reveals a novel role of vagal afferents to support digestion and energy storage that may contribute to the effectiveness of vagal blockade to induce weight loss.
Collapse
Affiliation(s)
- Kuei-Pin Huang
- School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Michael L Goodson
- School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Wendie Vang
- School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Hui Li
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.,South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Amanda J Page
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.,South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - Helen E Raybould
- School of Veterinary Medicine, University of California Davis, Davis, CA, USA.
| |
Collapse
|
29
|
Glendining KA, Higgins MBA, Fisher LC, Jasoni CL. Maternal obesity modulates sexually dimorphic epigenetic regulation and expression of leptin receptor in offspring hippocampus. Brain Behav Immun 2020; 88:151-160. [PMID: 32173454 DOI: 10.1016/j.bbi.2020.03.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 03/07/2020] [Accepted: 03/08/2020] [Indexed: 12/20/2022] Open
Abstract
Maternal obesity during pregnancy is associated with a greater risk for obesity and neurodevelopmental deficits in offspring. This developmental programming of disease is proposed to involve neuroendocrine, inflammatory, and epigenetic factors during gestation that disrupt normal fetal brain development. The hormones leptin and insulin are each intrinsically linked to metabolism, inflammation, and neurodevelopment, which led us to hypothesise that maternal obesity may disrupt leptin or insulin receptor signalling in the developing brain of offspring. Using a C57BL/6 mouse model of high fat diet-induced maternal obesity (mHFD), we performed qPCR to examine leptin receptor (Lepr) and insulin receptor (Insr) gene expression in gestational day (GD) 17.5 fetal brain. We found a significant effect of maternal diet and offspring sex on Lepr regulation in the developing hippocampus, with increased Lepr expression in female mHFD offspring (p < 0.05) compared to controls. Maternal diet did not alter hippocampal Insr in the fetal brain, or Lepr or Insr in prefrontal cortex, amygdala, or hypothalamus of female or male offspring. Chromatin immunoprecipitation revealed decreased binding of histones possessing the repressive histone mark H3K9me3 at the Lepr promoter (p < 0.05) in hippocampus of female mHFD offspring compared to controls, but not in males. Sex-specific deregulation of Lepr could be reproduced in vitro by exposing female hippocampal neurons to the obesity related proinflammatory cytokine IL-6, but not IL-17a or IFNG. Our findings indicate that the obesity-related proinflammatory cytokine IL-6 during pregnancy leads to sexually dimorphic changes in the modifications of histones binding at the Lepr gene promoter, and concomitant changes to Lepr transcription in the developing hippocampus. This suggests that exposure of the fetus to metabolic inflammatory molecules can impact epigenetic regulation of gene expression in the developing hippocampus.
Collapse
Affiliation(s)
- K A Glendining
- Centre for Neuroendocrinology, Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - M B A Higgins
- Centre for Neuroendocrinology, Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - L C Fisher
- Centre for Neuroendocrinology, Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - C L Jasoni
- Centre for Neuroendocrinology, Department of Anatomy, University of Otago, Dunedin, New Zealand.
| |
Collapse
|
30
|
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
|
31
|
Foll CL, Lutz TA. Systemic and Central Amylin, Amylin Receptor Signaling, and Their Physiological and Pathophysiological Roles in Metabolism. Compr Physiol 2020; 10:811-837. [PMID: 32941692 DOI: 10.1002/cphy.c190034] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This article in the Neural and Endocrine Section of Comprehensive Physiology discusses the physiology and pathophysiology of the pancreatic hormone amylin. Shortly after its discovery in 1986, amylin has been shown to reduce food intake as a satiation signal to limit meal size. Amylin also affects food reward, sensitizes the brain to the catabolic actions of leptin, and may also play a prominent role in the development of certain brain areas that are involved in metabolic control. Amylin may act at different sites in the brain in addition to the area postrema (AP) in the caudal hindbrain. In particular, the sensitizing effect of amylin on leptin action may depend on a direct interaction in the hypothalamus. The concept of central pathways mediating amylin action became more complex after the discovery that amylin is also synthesized in certain hypothalamic areas but the interaction between central and peripheral amylin signaling remains currently unexplored. Amylin may also play a dominant pathophysiological role that is associated with the aggregation of monomeric amylin into larger, cytotoxic molecular entities. This aggregation in certain species may contribute to the development of type 2 diabetes mellitus but also cardiovascular disease. Amylin receptor pharmacology is complex because several distinct amylin receptor subtypes have been described, because other neuropeptides [e.g., calcitonin gene-related peptide (CGRP)] can also bind to amylin receptors, and because some components of the functional amylin receptor are also used for other G-protein coupled receptor (GPCR) systems. © 2020 American Physiological Society. Compr Physiol 10:811-837, 2020.
Collapse
Affiliation(s)
- Christelle Le Foll
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland
| | - Thomas A Lutz
- Institute of Veterinary Physiology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
32
|
Coester B, Koester-Hegmann C, Lutz TA, Le Foll C. Amylin/Calcitonin Receptor-Mediated Signaling in POMC Neurons Influences Energy Balance and Locomotor Activity in Chow-Fed Male Mice. Diabetes 2020; 69:1110-1125. [PMID: 32152204 DOI: 10.2337/db19-0849] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 02/28/2020] [Indexed: 11/13/2022]
Abstract
Amylin, a pancreatic hormone and neuropeptide, acts principally in the hindbrain to decrease food intake and has recently been shown to act as a neurotrophic factor to control the development of area postrema → nucleus of the solitary tract and arcuate hypothalamic nucleus → paraventricular nucleus axonal fiber outgrowth. Amylin is also able to activate ERK signaling specifically in POMC neurons independently of leptin. For investigation of the physiological role of amylin signaling in POMC neurons, the core component of the amylin receptor, calcitonin receptor (CTR), was depleted from POMC neurons using an inducible mouse model. The loss of CTR in POMC neurons leads to increased body weight gain, increased adiposity, and glucose intolerance in male knockout mice, characterized by decreased energy expenditure (EE) and decreased expression of uncoupling protein 1 (UCP1) in brown adipose tissue. Furthermore, a decreased spontaneous locomotor activity and absent thermogenic reaction to the application of the amylin receptor agonist were observed in male and female mice. Together, these results show a significant physiological impact of amylin/calcitonin signaling in CTR-POMC neurons on energy metabolism and demonstrate the need for sex-specific approaches in obesity research and potentially treatment.
Collapse
Affiliation(s)
- Bernd Coester
- 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
| | - Christelle Le Foll
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| |
Collapse
|
33
|
Haddad-Tóvolli R, Altirriba J, Obri A, Sánchez EE, Chivite I, Milà-Guasch M, Ramírez S, Gómez-Valadés AG, Pozo M, Burguet J, Velloso LA, Claret M. Pro-opiomelanocortin (POMC) neuron translatome signatures underlying obesogenic gestational malprogramming in mice. Mol Metab 2020; 36:100963. [PMID: 32283518 PMCID: PMC7152705 DOI: 10.1016/j.molmet.2020.02.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 02/07/2020] [Accepted: 02/10/2020] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE Maternal unbalanced nutritional habits during embryonic development and perinatal stages perturb hypothalamic neuronal programming of the offspring, thus increasing obesity-associated diabetes risk. However, the underlying molecular mechanisms remain largely unknown. In this study we sought to determine the translatomic signatures associated with pro-opiomelanocortin (POMC) neuron malprogramming in maternal obesogenic conditions. METHODS We used the RiboTag mouse model to specifically profile the translatome of POMC neurons during neonatal (P0) and perinatal (P21) life and its neuroanatomical, functional, and physiological consequences. RESULTS Maternal high-fat diet (HFD) exposure did not interfere with offspring's hypothalamic POMC neuron specification, but significantly impaired their spatial distribution and axonal extension to target areas. Importantly, we established POMC neuron-specific translatome signatures accounting for aberrant neuronal development and axonal growth. These anatomical and molecular alterations caused metabolic dysfunction in early life and adulthood. CONCLUSIONS Our study provides fundamental insights on the molecular mechanisms underlying POMC neuron malprogramming in obesogenic contexts.
Collapse
Affiliation(s)
- Roberta Haddad-Tóvolli
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Jordi Altirriba
- Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| | - Arnaud Obri
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Elena Eyre Sánchez
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Iñigo Chivite
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Maria Milà-Guasch
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Sara Ramírez
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Alicia G Gómez-Valadés
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Macarena Pozo
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Jasmine Burguet
- Institut Jean-Pierre Bourgin, INRAE, AgroParisTech, Université Paris-Saclay, 78000, Versailles, France.
| | - Licio A Velloso
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas (UNICAMP), Brazil.
| | - Marc Claret
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain; School of Medicine, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
34
|
Wasinski F, Furigo IC, Teixeira PDS, Ramos-Lobo AM, Peroni CN, Bartolini P, List EO, Kopchick JJ, Donato J. Growth Hormone Receptor Deletion Reduces the Density of Axonal Projections from Hypothalamic Arcuate Nucleus Neurons. Neuroscience 2020; 434:136-147. [PMID: 32229232 DOI: 10.1016/j.neuroscience.2020.03.037] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 03/20/2020] [Accepted: 03/23/2020] [Indexed: 12/18/2022]
Abstract
The arcuate nucleus (ARH) is an important hypothalamic area for the homeostatic control of feeding and other metabolic functions. In the ARH, proopiomelanocortin- (POMC) and agouti-related peptide (AgRP)-expressing neurons play a key role in the central regulation of metabolism. These neurons are influenced by circulating factors, such as leptin and growth hormone (GH). The objective of the present study was to determine whether a direct action of GH on ARH neurons regulates the density of POMC and AgRP axonal projections to major postsynaptic targets. We studied POMC and AgRP axonal projections to the hypothalamic paraventricular (PVH), lateral (LHA) and dorsomedial (DMH) nuclei in leptin receptor (LepR)-deficient mice (Leprdb/db), GH-deficient mice (Ghrhrlit/lit) and in mice carrying specific ablations of GH receptor (GHR) either in LepR- or AgRP-expressing cells. Leprdb/db mice presented reduction in the density of POMC innervation to the PVH compared to wild-type and Ghrhrlit/lit mice. Additionally, both Leprdb/db and Ghrhrlit/lit mice showed reduced AgRP fiber density in the PVH, LHA and DMH. LepR GHR knockout mice showed decreased density of POMC innervation in the PVH and DMH, compared to control mice, whereas a reduction in the density of AgRP innervation was observed in all areas analyzed. Conversely, AgRP-specific ablation of GHR led to a significant reduction in AgRP projections to the PVH, LHA and DMH, without affecting POMC innervation. Our findings indicate that GH has direct trophic effects on the formation of POMC and AgRP axonal projections and provide additional evidence that GH regulates hypothalamic neurocircuits controlling energy homeostasis.
Collapse
Affiliation(s)
- Frederick Wasinski
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP 05508-000, Brazil
| | - Isadora C Furigo
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP 05508-000, Brazil
| | - Pryscila D S Teixeira
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP 05508-000, Brazil
| | - Angela M Ramos-Lobo
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP 05508-000, Brazil
| | - Cibele N Peroni
- Centro de Biotecnologia, Instituto de Pesquisas Energéticas e Nucleares (IPEN-CNEN), São Paulo, SP 05508-900, Brazil
| | - Paolo Bartolini
- Centro de Biotecnologia, Instituto de Pesquisas Energéticas e Nucleares (IPEN-CNEN), São Paulo, SP 05508-900, 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
| | - Jose Donato
- Departamento de Fisiologia e Biofísica, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP 05508-000, Brazil.
| |
Collapse
|
35
|
Park S, Jang A, Bouret SG. Maternal obesity-induced endoplasmic reticulum stress causes metabolic alterations and abnormal hypothalamic development in the offspring. PLoS Biol 2020; 18:e3000296. [PMID: 32163401 PMCID: PMC7067374 DOI: 10.1371/journal.pbio.3000296] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 02/05/2020] [Indexed: 01/19/2023] Open
Abstract
The steady increase in the prevalence of obesity and associated type II diabetes mellitus is a major health concern, particularly among children. Maternal obesity represents a risk factor that contributes to metabolic perturbations in the offspring. Endoplasmic reticulum (ER) stress has emerged as a critical mechanism involved in leptin resistance and type 2 diabetes in adult individuals. Here, we used a mouse model of maternal obesity to investigate the importance of early life ER stress in the nutritional programming of this metabolic disease. Offspring of obese dams developed glucose intolerance and displayed increased body weight, adiposity, and food intake. Moreover, maternal obesity disrupted the development of melanocortin circuits associated with neonatal hyperleptinemia and leptin resistance. ER stress-related genes were up-regulated in the hypothalamus of neonates born to obese mothers. Neonatal treatment with the ER stress-relieving drug tauroursodeoxycholic acid improved metabolic and neurodevelopmental deficits and reversed leptin resistance in the offspring of obese dams.
Collapse
Affiliation(s)
- Soyoung Park
- The Saban Research Institute, Developmental Neuroscience Program, Children’s Hospital Los Angeles, Los Angeles, California, United States of America
| | - Alice Jang
- The Saban Research Institute, Developmental Neuroscience Program, Children’s Hospital Los Angeles, Los Angeles, California, United States of America
| | - Sebastien G. Bouret
- Inserm, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Centre, Lille, France
- University of Lille, FHU 1,000 Days for Health, Lille, France
| |
Collapse
|
36
|
Méquinion M, Foldi CJ, Andrews ZB. The Ghrelin-AgRP Neuron Nexus in Anorexia Nervosa: Implications for Metabolic and Behavioral Adaptations. Front Nutr 2020; 6:190. [PMID: 31998738 PMCID: PMC6962137 DOI: 10.3389/fnut.2019.00190] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 12/11/2019] [Indexed: 11/13/2022] Open
Abstract
Anorexia Nervosa (AN) is viewed as primarily a psychiatric disorder owing to the considerable behavioral and genetic overlap with mood disorders and other psychiatric traits. However, the recent reconceptualization of AN as one of both psychiatric and metabolic etiology suggests that metabolic circuits conveying hunger, or sensitive to signals of hunger, may be a critical nexus linking metabolic dysfunction to mood disturbances. Within the brain, hunger is primarily percieved by Agouti-related (AgRP) neurons and hunger increases plasma concentrations of the hormone ghrelin, which targets ghrelin receptors on AgRP neurons to facilitate metabolic adaptations to low energy availability. However, beyond the fundamental role in maintaining hunger signaling, AgRP neurons regulate a diverse range of behaviors such as motivation, locomotor activity, negative reinforcement, anxiety, and obsession and a key factor involved in the manifestation of these behavioral changes in response to activation is the presence or absence of food availability. These changes can be considered adaptive in that they promote affective food-seeking strategies in environments with limited food availability. However, it also suggests that these neurons, so well-studied for their metabolic control, shape mood-related behaviors in a context-dependent manner and dysfunctional control leads not only to metabolic problems but also potentially mood-related problems. The purpose of this review is to underline the potential role of AgRP neurons and ghrelin signaling in both the metabolic and behavioral changes observed in anorexia nervosa. We aim to highlight the most recent studies on AgRP neurons and ghrelin signaling and integrate their metabolic and behavioral roles in normal function and highlight how dysfunction may contribute to the development of AN.
Collapse
Affiliation(s)
| | | | - Zane B. Andrews
- Monash Biomedicine Discovery Institute and Department of Physiology, Monash University, Clayton, VIC, Australia
| |
Collapse
|
37
|
Abstract
Many animal models that are currently used in appetite and obesity research share at least some main features of human obesity and its comorbidities. Hence, even though no animal model replicates all aspects of "common" human obesity, animal models are imperative in studying the control of energy balance and reasons for its imbalance that may eventually lead to overt obesity. The most frequently used animal models are small rodents that may be based on mutations or manipulations of individual or several genes and on the exposure to obesogenic diets or other manipulations that predispose the animals to gaining or maintaining excessive weight. Characteristics include hyperphagia or changes in energy metabolism and at least in some models the frequent comorbidities of obesity, like hyperglycemia, insulin resistance, or diabetes-like syndromes. Some of the most frequently used animal models of obesity research involve animals with monogenic mutations of the leptin pathway which in fact are useful to study specific mechanistic aspects of eating controls, but typically do not recapitulate "common" obesity in the human population. Hence, this review will mention advantages and disadvantages of respective animal models in order to build a basis for the most appropriate use in biomedical research.
Collapse
Affiliation(s)
- Thomas A Lutz
- Institute of Veterinary Physiology, Vetsuisse Faculty University of Zurich, Zurich, Switzerland.
| |
Collapse
|
38
|
Wang N, Lv B, Guan L, Qiao H, Sun B, Luo X, Jia R, Chen K, Yan J. Maternal low protein exposure alters glucose tolerance and intestinal nutrient-responsive receptors and transporters expression of rat offspring. Life Sci 2019; 243:117216. [PMID: 31884096 DOI: 10.1016/j.lfs.2019.117216] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 12/09/2019] [Accepted: 12/20/2019] [Indexed: 12/25/2022]
Abstract
AIMS Maternal protein malnutrition during perinatal period has long-term consequences on the offspring's metabolic phenotype. Here we determined the effects of maternal protein-restricted (PR) diet on offspring's metabolism in 3- and 12-week-old. MAIN METHODS Sprague-Dawley rats were fed with standard chow diet or PR diet during pregnancy and lactation. Food intake and body weight of offspring were measured weekly. The oral glucose tolerance tests were underwent, the pancreases were collected for histochemical staining, and the duodenum, jejunum and ileum were collected for gene and protein expression analysis in 3- and 12-week-old offspring. KEY FINDINGS PR offspring had significant lower body weight and persisted till 12-week-old. From 3- to 12-week-old, PR offspring presented considerably impaired glucose tolerance, while no marked change was shown in control rats. Additionally, the average islet size of PR offspring decreased significantly in 12-week-old. The mRNA and protein expression of nutrient-responsive receptors and transporters T1R3, SGLT1 and GLUT2 increased significantly in the intestine of 3-week-old PR offspring. And from 3- and 12-week-old, the increase tendency of expression subdued. SIGNIFICANCE These results suggest that maternal PR diet during critical developmental windows influences offspring metabolism, which may be subdued partially, but not be reversed completely by chow diet after weaning.
Collapse
Affiliation(s)
- Nan Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Bo Lv
- School of Humanities, Xidian University, Xi'an, Shaanxi 710126, China
| | - Limin Guan
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Xi'an Jiaotong University College of Stomatology, Xi'an, Shaanxi 710000, China
| | - Hu Qiao
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Xi'an Jiaotong University College of Stomatology, Xi'an, Shaanxi 710000, China
| | - Bo Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Xiao Luo
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China
| | - Ru Jia
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Xi'an Jiaotong University College of Stomatology, Xi'an, Shaanxi 710000, China
| | - Ke Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China.
| | - Jianqun Yan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
39
|
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
|
40
|
Control-theory models of body-weight regulation and body-weight-regulatory appetite. Appetite 2019; 144:104440. [PMID: 31494154 DOI: 10.1016/j.appet.2019.104440] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 08/08/2019] [Accepted: 09/02/2019] [Indexed: 12/24/2022]
Abstract
Human body weight (BW), or some variable related to it, is physiologically regulated. That is, negative feedback from changes in BW elicits compensatory influences on appetite, which may be called BW-regulatory appetite, and a component of energy expenditure (EE) called adaptive thermogenesis (AdEE). BW-regulatory appetite is of general significance because it appears to be related to a variety of aspects of human appetite beyond just energy intake. BW regulation, BW-regulatory appetite and AdEE are frequently discussed using concepts derived from control theory, which is the mathematical description of dynamic systems involving negative feedback. The aim of this review is to critically assess these discussions. Two general types of negative-feedback control have been invoked to describe BW regulation, set-point control and simple negative-feedback control, often called settling-point control in the BW literature. The distinguishing feature of set-point systems is the existence of an externally controlled target level of regulation, the set point. The performance of almost any negative-feedback regulatory system, however, can be modeled on the basis of feedback gain without including a set point. In both set-point and simple negative-feedback models of BW regulation, the precision of regulation is usually determined mainly by feedback gain, which refers to the transformations of feedback into compensatory changes in BW-regulatory appetite and AdEE. Stable BW most probably represents equilibria shaped by feedback gain and tonic open-loop challenges, especially obesogenic environments. Data indicate that simple negative-feedback control accurately models human BW regulation and that the set-point concept is superfluous unless its neuroendocrine representation is found in the brain. Additional research aimed at testing control-theory models in humans and non-human animals is warranted.
Collapse
|
41
|
Grabauskas G, Wu X, Zhou S, Li J, Gao J, Owyang C. High-fat diet-induced vagal afferent dysfunction via upregulation of 2-pore domain potassium TRESK channel. JCI Insight 2019; 4:130402. [PMID: 31484832 PMCID: PMC6777907 DOI: 10.1172/jci.insight.130402] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 07/30/2019] [Indexed: 12/17/2022] Open
Abstract
Research shows that rats and humans on a high-fat diet (HFD) are less sensitive to satiety signals known to act via vagal afferent pathways. We hypothesize that HFD causes an upregulation of 2-pore domain potassium channels, resulting in hyperpolarization of nodose ganglia (NG) and decreased vagal response to satiety signals, which contribute to hyperphagia. We show that a 2-week HFD caused an upregulation of 2-pore domain TWIK-related spinal cord K+ (TRESK) and TWIK-related acid-sensitive K+ 1 (TASK1) channels by 330% ± 50% and 60% ± 20%, respectively, in NG. Patch-clamp studies of isolated NG neurons demonstrated a decrease in excitability. In vivo single-unit NG recordings showed that a 2-week HFD led to a 55% reduction in firing frequency in response to CCK-8 or leptin stimulation. NG electroporation with TRESK siRNA restored NG responsiveness to CCK-8 and leptin. Rats fed a 2-week HFD consumed ~40% more calories compared with controls. Silencing NG TRESK but not TASK1 channel expression in HFD-fed rats restored normal calorie consumption. In conclusion, HFD caused upregulation of TRESK channels, resulting in NG hyperpolarization and decreased vagal responsiveness to satiety signals. This finding provides a pharmacological target to prevent or treat HFD-induced hyperphagia.
Collapse
|
42
|
Sadagurski M, Debarba LK, Werneck-de-Castro JP, Ali Awada A, Baker TA, Bernal-Mizrachi E. Sexual dimorphism in hypothalamic inflammation in the offspring of dams exposed to a diet rich in high fat and branched-chain amino acids. Am J Physiol Endocrinol Metab 2019; 317:E526-E534. [PMID: 31361548 PMCID: PMC6766606 DOI: 10.1152/ajpendo.00183.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Branched-chain amino acid (BCAAs: leucine, isoleucine, and valine) contribute to the development of obesity-associated insulin resistance in the context of consumption of a high-fat diet (HFD) in humans and rodents. Maternal diet is a major determinant of offspring health, and there is strong evidence that maternal HFD alters hypothalamic developmental programming and disrupts offspring energy homeostasis in rodents. In this study, we exposed pregnant and lactating C57BL/6JB female mice to either HFD, HFD with supplemented BCAA (HFD+BCAA), or standard diet (SC), and we studied offspring metabolic phenotypes. Both maternal HFD and HFD supplemented with BCAA had similar effect rendering the offspring metabolic imbalance and impairing their ability to cope with HFD when challenged during aging. The metabolic effects of HFD challenge were more profound in females, worsening female offspring ability to cope with an HFD challenge by activating hypothalamic inflammation in aging. Moreover, the sex differences in hypothalamic estrogen receptor α (ER-α) expression levels were lost in female offspring upon HFD challenge, supporting a link between ER-α levels and hypothalamic inflammation in offspring and highlighting the programming potential of hypothalamic inflammatory responses and maternal nutrition.
Collapse
Affiliation(s)
- Marianna Sadagurski
- Department of Biological Sciences, Integrative Biosciences Center, Wayne State University, Detroit, Michigan
| | - Lucas Kniess Debarba
- Department of Biological Sciences, Integrative Biosciences Center, Wayne State University, Detroit, Michigan
| | - Joao Pedro Werneck-de-Castro
- Division of Endocrinology, Diabetes and Metabolism, Diabetes Research Institute, University of Miami, Miller School of Medicine, Miami, Florida
| | - Abear Ali Awada
- Department of Biological Sciences, Integrative Biosciences Center, Wayne State University, Detroit, Michigan
| | - Tess A Baker
- Division of Endocrinology, Diabetes and Metabolism, Diabetes Research Institute, University of Miami, Miller School of Medicine, Miami, Florida
| | - Ernesto Bernal-Mizrachi
- Division of Endocrinology, Diabetes and Metabolism, Diabetes Research Institute, University of Miami, Miller School of Medicine, Miami, Florida
- Miami Veterans Affairs Health Care System, Miami, Florida
| |
Collapse
|
43
|
Feeding circuit development and early-life influences on future feeding behaviour. Nat Rev Neurosci 2019; 19:302-316. [PMID: 29662204 DOI: 10.1038/nrn.2018.23] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A wide range of maternal exposures - undernutrition, obesity, diabetes, stress and infection - are associated with an increased risk of metabolic disease in offspring. Developmental influences can cause persistent structural changes in hypothalamic circuits regulating food intake in the service of energy balance. The physiological relevance of these alterations has been called into question because maternal impacts on daily caloric intake do not persist to adulthood. Recent behavioural and epidemiological studies in humans provide evidence that the relative contribution of appetitive traits related to satiety, reward and the emotional aspects of food intake regulation changes across the lifespan. This Opinion article outlines a neurodevelopmental framework to explore the possibility that crosstalk between developing circuits regulating different modalities of food intake shapes future behavioural responses to environmental challenges.
Collapse
|
44
|
Obri A, Claret M. The role of epigenetics in hypothalamic energy balance control: implications for obesity. Cell Stress 2019; 3:208-220. [PMID: 31309172 PMCID: PMC6612891 DOI: 10.15698/cst2019.07.191] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Despite enormous social and scientific efforts, obesity rates continue to increase worldwide. While genetic factors contribute to obesity development, genetics alone cannot explain the current epidemic. Obesity is essentially the consequence of complex genetic-environmental interactions. Evidence suggests that contemporary lifestyles trigger epigenetic changes, which can dysregulate energy balance and thus contribute to obesity. The hypothalamus plays a pivotal role in the regulation of body weight, through a sophisticated network of neuronal systems. Alterations in the activity of these neuronal pathways have been implicated in the pathophysiology of obesity. Here, we review the current knowledge on the central control of energy balance with a focus on recent studies linking epigenetic mechanisms in the hypothalamus to the development of obesity and metabolic disorders.
Collapse
Affiliation(s)
- Arnaud Obri
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Marc Claret
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.,Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 08036 Barcelona, Spain
| |
Collapse
|
45
|
Andreoli MF, Donato J, Cakir I, Perello M. Leptin resensitisation: a reversion of leptin-resistant states. J Endocrinol 2019; 241:R81-R96. [PMID: 30959481 DOI: 10.1530/joe-18-0606] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 04/02/2019] [Indexed: 12/30/2022]
Abstract
Leptin resistance refers to states in which leptin fails to promote its anticipated effects, frequently coexisting with hyperleptinaemia. Leptin resistance is closely associated with obesity and also observed in physiological situations such as pregnancy and in seasonal animals. Leptin resensitisation refers to the reversion of leptin-resistant states and is associated with improvement in endocrine and metabolic disturbances commonly observed in obesity and a sustained decrease of plasma leptin levels, possibly below a critical threshold level. In obesity, leptin resensitisation can be achieved with treatments that reduce body adiposity and leptinaemia, or with some pharmacological compounds, while physiological leptin resistance reverts spontaneously. The restoration of leptin sensitivity could be a useful strategy to treat obesity, maintain weight loss and/or reduce the recidivism rate for weight regain after dieting. This review provides an update and discussion about reversion of leptin-resistant states and modulation of the molecular mechanisms involved in each situation.
Collapse
Affiliation(s)
- María F Andreoli
- Laboratory of Experimental Neurodevelopment, Institute of Development and Paediatric Research (IDIP), La Plata Children's Hospital and Scientific Research Commission, Province of Buenos Aires (CIC-PBA), La Plata, Buenos Aires, Argentina
- Argentine Research Council (CONICET), La Plata, Buenos Aires, Argentina
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, São Paulo, Brazil
| | - Isin Cakir
- Life Sciences Institute, University of Michigan, Ann Arbor, Michigan, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Mario Perello
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology (IMBICE, Argentine Research Council (CONICET), National University of La Plata and Scientific Research Commission, Province of Buenos Aires (CIC-PBA)), La Plata, Buenos Aires, Argentina
| |
Collapse
|
46
|
Cordner ZA, Khambadkone SG, Boersma GJ, Song L, Summers TN, Moran TH, Tamashiro KLK. Maternal high-fat diet results in cognitive impairment and hippocampal gene expression changes in rat offspring. Exp Neurol 2019; 318:92-100. [PMID: 31051155 DOI: 10.1016/j.expneurol.2019.04.018] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 04/13/2019] [Accepted: 04/29/2019] [Indexed: 01/01/2023]
Abstract
Consumption of a high-fat diet has long been known to increase risk for obesity, diabetes, and the metabolic syndrome. Further evidence strongly suggests that these same metabolic disorders are associated with an increased risk of cognitive impairment later in life. Now faced with an expanding global burden of obesity and increasing prevalence of dementia due to an aging population, understanding the effects of high-fat diet consumption on cognition is of increasingly critical importance. Further, the developmental origins of many adult onset neuropsychiatric disorders have become increasingly clear, indicating a need to investigate effects of various risk factors, including diet, across the lifespan. Here, we use a rat model to assess the effects of maternal diet during pregnancy and lactation on cognition and hippocampal gene expression of offspring. Behaviorally, adult male offspring of high-fat fed dams had impaired object recognition memory and impaired spatial memory compared to offspring of chow-fed dams. In hippocampus, we found decreased expression of Insr, Lepr, and Slc2a1 (GLUT1) among offspring of high-fat fed dams at postnatal day 21. The decreased expression of Insr and Lepr persisted at postnatal day 150. Together, these data provide additional evidence to suggest that maternal exposure to high-fat diet during pregnancy and lactation can have lasting effects on the brain, behavior, and cognition on adult offspring.
Collapse
Affiliation(s)
- Zachary A Cordner
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA
| | - Seva G Khambadkone
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA; Cellular & Molecular Medicine Graduate Program, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA
| | - Gretha J Boersma
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA
| | - Lin Song
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA
| | - Tyler N Summers
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA
| | - Timothy H Moran
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA; Cellular & Molecular Medicine Graduate Program, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA
| | - Kellie L K Tamashiro
- Department of Psychiatry & Behavioral Sciences, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA; Cellular & Molecular Medicine Graduate Program, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross 618, Baltimore, MD 21205, USA.
| |
Collapse
|
47
|
Lutz TA, Le Foll C. Endogenous amylin contributes to birth of microglial cells in arcuate nucleus of hypothalamus and area postrema during fetal development. Am J Physiol Regul Integr Comp Physiol 2019; 316:R791-R801. [PMID: 30943041 DOI: 10.1152/ajpregu.00004.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Amylin acts in the area postrema (AP) and arcuate nucleus (ARC) to control food intake. Amylin also increases axonal fiber outgrowth from the AP→nucleus tractus solitarius and from ARC→hypothalamic paraventricular nucleus. More recently, exogenous amylin infusion for 4 wk was shown to increase neurogenesis in adult rats in the AP. Furthermore, amylin has been shown to enhance leptin signaling in the ARC and ventromedial nucleus of the hypothalamus (VMN). Thus, we hypothesized that endogenous amylin could be a critical factor in regulating cell birth in the ARC and AP and that amylin could also be involved in the birth of leptin-sensitive neurons. Amylin+/- dams were injected with BrdU at embryonic day 12 and at postnatal day 2; BrdU+ cells were quantified in wild-type (WT) and amylin knockout (KO) mice. The number of BrdU+HuC/D+ neurons was similar in ARC and AP, but the number of BrdU+Iba1+ microglia was significantly decreased in both nuclei. Five-week-old WT and KO littermates were injected with leptin to test whether amylin is involved in the birth of leptin-sensitive neurons. Although there was no difference in the number of BrdU+c-Fos+ neurons in the ARC and dorsomedial nucleus, an increase in BrdU+c-Fos+ neurons was seen in VMN and lateral hypothalamus (LH) in amylin KO mice. In conclusion, these data suggest that during fetal development, endogenous amylin favors the birth of microglial cells in the ARC and AP and that it decreases the birth of leptin-sensitive neurons in the VMN and LH.
Collapse
Affiliation(s)
- Thomas A Lutz
- 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
| |
Collapse
|
48
|
Roberts BL, Bennett CM, Carroll JM, Lindsley SR, Kievit P. Early overnutrition alters synaptic signaling and induces leptin resistance in arcuate proopiomelanocortin neurons. Physiol Behav 2019; 206:166-174. [PMID: 30951750 DOI: 10.1016/j.physbeh.2019.04.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 03/08/2019] [Accepted: 04/01/2019] [Indexed: 01/17/2023]
Abstract
Early overnutrition disrupts leptin sensitivity and the development of hypothalamic pathways involved in the regulation of metabolism and feeding behavior. While previous studies have largely focused on the development of neuronal projections, few studies have examined the impact of early nutrition on hypothalamic synaptic physiology. In this study we characterized the synaptic development of proopiomelanocortin (POMC) neurons in the arcuate nucleus of the hypothalamus (ARH), their sensitivity to leptin, and the impact of early overnutrition on the development of these neurons. Electrophysiology recordings were performed in mouse ARH brain slices containing POMC-EGFP neurons from postnatal age (P) 7-9 through adulthood. We determined that pre- and postsynaptic components of inhibitory inputs increased throughout the first 3 weeks of the postnatal period, which coincided with a decreased membrane potential in POMC neurons. We then examined whether chronic postnatal overnutrition (CPO) altered these synaptic connections. CPO mice exhibited increased body weight and circulating leptin levels, as described previously. POMC neurons in CPO mice had an increase in post-synaptic inhibitory currents compared to controls at 2 weeks of age, but this effect reversed by the third week. In control mice we observed heterogenous effects of leptin on POMC neurons in early life that transitioned to predominantly stimulatory actions in adulthood. However, postnatal overfeeding resulted in POMC neurons becoming leptin-resistant which persisted into adulthood. These studies suggest that postnatal overfeeding alters the postsynaptic development of POMC neurons and induces long-lasting leptin resistance in ARH-POMC neurons.
Collapse
Affiliation(s)
- Brandon L Roberts
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, OR 97006, USA
| | - Camdin M Bennett
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, OR 97006, USA
| | - Julie M Carroll
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, OR 97006, USA
| | - Sarah R Lindsley
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, OR 97006, USA
| | - Paul Kievit
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, OR 97006, USA.
| |
Collapse
|
49
|
Ramos-Lobo AM, Furigo IC, Teixeira PDS, Zampieri TT, Wasinski F, Buonfiglio DC, Donato J. Maternal metabolic adaptations are necessary for normal offspring growth and brain development. Physiol Rep 2019. [PMID: 29536670 PMCID: PMC5849578 DOI: 10.14814/phy2.13643] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Several metabolic adaptations emerge during pregnancy and continue through lactation, including increases in food intake and body weight, as well as insulin and leptin resistance. These maternal adaptations are thought to play a role in offspring viability and success. Using a model of attenuated maternal metabolic adaptations induced by ablation of the Socs3 gene in leptin receptor expressing cells (SOCS3 KO mice), our study aimed to investigate whether maternal metabolic changes are required for normal offspring development, and if their absence causes metabolic imbalances in adulthood. The litters were subjected to a cross‐fostering experimental design to distinguish the prenatal and postnatal effects caused by maternal metabolic adaptations. Males either born or raised by SOCS3 KO mice showed reduced body weight until 8 weeks of life. Both adult males and females born or raised by SOCS3 KO mice also had lower body adiposity. Despite that, no significant changes in energy expenditure, glucose tolerance or insulin resistance were observed. However, males either born or raised by SOCS3 KO mice showed reduced brain mass in adulthood. Furthermore, animals born from SOCS3 KO mice also had lower proopiomelanocortin fiber density in the paraventricular nucleus of the hypothalamus. In conclusion, these findings indicate that the commonly observed metabolic changes in pregnancy and lactation are necessary for normal offspring growth and brain development.
Collapse
Affiliation(s)
- Angela M Ramos-Lobo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Isadora C Furigo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Pryscila D S Teixeira
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Thais T Zampieri
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Frederick Wasinski
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Daniella C Buonfiglio
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
50
|
Mirzadeh Z, Alonge KM, Cabrales E, Herranz-Pérez V, Scarlett JM, Brown JM, Hassouna R, Matsen ME, Nguyen HT, Garcia-Verdugo JM, Zeltser LM, Schwartz MW. Perineuronal Net Formation during the Critical Period for Neuronal Maturation in the Hypothalamic Arcuate Nucleus. Nat Metab 2019; 1:212-221. [PMID: 31245789 PMCID: PMC6594569 DOI: 10.1038/s42255-018-0029-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In leptin-deficient ob/ob mice, obesity and diabetes are associated with abnormal development of neurocircuits in the hypothalamic arcuate nucleus (ARC)1, a critical brain area for energy and glucose homeostasis2,3. As this developmental defect can be remedied by systemic leptin administration, but only if given before postnatal day 28, a critical period (CP) for leptin-dependent development of ARC neurocircuits has been proposed4. In other brain areas, CP closure coincides with the appearance of perineuronal nets (PNNs), extracellular matrix specializations that restrict the plasticity of neurons that they enmesh5. Here we report that in humans as well as rodents, subsets of neurons in the mediobasal aspect of the ARC are enmeshed by PNN-like structures. In mice, these neurons are densely-packed into a continuous ring that encircles the junction of the ARC and median eminence, which facilitates exposure of ARC neurons to the circulation. Most of the enmeshed neurons are both GABAergic and leptin receptor-positive, including a majority of Agrp neurons. Postnatal formation of the PNN-like structures coincides precisely with closure of the CP for Agrp neuron maturation and is dependent on input from circulating leptin, as postnatal ob/ob mice have reduced ARC PNN-like material that is restored by leptin administration during the CP. We conclude that neurons crucial to metabolic homeostasis are enmeshed by PNN-like structures and organized into a densely packed cluster situated circumferentially at the ARC-ME junction, where metabolically-relevant humoral signals are sensed.
Collapse
Affiliation(s)
- Zaman Mirzadeh
- Department of Neurosurgery, Barrow Neurological Institute, Phoenix, AZ, USA.
| | - Kimberly M Alonge
- University of Washington Medicine Diabetes Institute, School of Medicine, University of Washington, Seattle, WA, USA
| | - Elaine Cabrales
- Department of Neurosurgery, Barrow Neurological Institute, Phoenix, AZ, USA
| | - Vicente Herranz-Pérez
- Laboratory of Comparative Neurobiology, Instituto Cavanilles, CIBERNED, Universidad de Valencia, Valencia, Spain
- Predepartmental Unit of Medicine, Faculty of Health Sciences, Universitat Jaume I, Castellón de la Plana, Spain
| | - Jarrad M Scarlett
- University of Washington Medicine Diabetes Institute, School of Medicine, University of Washington, Seattle, WA, USA
- Department of Pediatric Gastroenterology and Hepatology, Seattle Children's Hospital, Seattle, WA, USA
| | - Jenny M Brown
- University of Washington Medicine Diabetes Institute, School of Medicine, University of Washington, Seattle, WA, USA
| | - Rim Hassouna
- Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, NY, USA
| | - Miles E Matsen
- University of Washington Medicine Diabetes Institute, School of Medicine, University of Washington, Seattle, WA, USA
| | - Hong T Nguyen
- University of Washington Medicine Diabetes Institute, School of Medicine, University of Washington, Seattle, WA, USA
| | - Jose Manuel Garcia-Verdugo
- Laboratory of Comparative Neurobiology, Instituto Cavanilles, CIBERNED, Universidad de Valencia, Valencia, Spain
| | - Lori M Zeltser
- Naomi Berrie Diabetes Center, Columbia University Medical Center, New York, NY, USA
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA
| | - Michael W Schwartz
- University of Washington Medicine Diabetes Institute, School of Medicine, University of Washington, Seattle, WA, USA.
| |
Collapse
|