1
|
Placzek M, Chinnaiya K, Kim DW, Blackshaw S. Control of tuberal hypothalamic development and its implications in metabolic disorders. Nat Rev Endocrinol 2025; 21:118-130. [PMID: 39313573 DOI: 10.1038/s41574-024-01036-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/29/2024] [Indexed: 09/25/2024]
Abstract
The tuberal hypothalamus regulates a range of crucial physiological processes, including energy homeostasis and metabolism. In this Review, we explore the intricate molecular mechanisms and signalling pathways that control the development of the tuberal hypothalamus, focusing on aspects that shape metabolic outcomes. Major developmental events are discussed in the context of their effect on the establishment of both functional hypothalamic neuronal circuits and brain-body interfaces that are pivotal to the control of metabolism. Emerging evidence indicates that aberrations in molecular pathways during tuberal hypothalamic development contribute to metabolic dysregulation. Understanding the molecular underpinnings of tuberal hypothalamic development provides a comprehensive view of neurodevelopmental processes and offers a promising avenue for future targeted interventions to prevent and treat metabolic disorders.
Collapse
Affiliation(s)
- Marysia Placzek
- School of Biosciences, University of Sheffield, Sheffield, UK.
- Bateson Centre, University of Sheffield, Sheffield, UK.
- Neuroscience Institute, University of Sheffield, Sheffield, UK.
| | | | - Dong Won Kim
- Danish Research Institute of Translational Neuroscience (DANDRITE), Nordic EMBL Partnership for Molecular Medicine, Aarhus University, Aarhus, Denmark
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
2
|
Chen B, de Launoit E, Meseguer D, Garcia Caceres C, Eichmann A, Renier N, Schneeberger M. The interactions between energy homeostasis and neurovascular plasticity. Nat Rev Endocrinol 2024; 20:749-759. [PMID: 39054359 DOI: 10.1038/s41574-024-01021-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/10/2024] [Indexed: 07/27/2024]
Abstract
Food intake and energy expenditure are sensed and processed by multiple brain centres to uphold energy homeostasis. Evidence from the past decade points to the brain vasculature as a new critical player in regulating energy balance that functions in close association with the local neuronal networks. Nutritional imbalances alter many properties of the neurovascular system (such as neurovascular coupling and blood-brain barrier permeability), thus suggesting a bidirectional link between the nutritional milieu and neurovascular health. Increasing numbers of people are consuming a Western diet (comprising ultra-processed food with high-fat and high-sugar content) and have a sedentary lifestyle, with these factors contributing to the current obesity epidemic. Emerging pharmacological interventions (for example, glucagon-like peptide 1 receptor agonists) successfully trigger weight loss. However, whether these approaches can reverse the detrimental effects of long-term exposure to the Western diet (such as neurovascular uncoupling, neuroinflammation and blood-brain barrier disruption) and maintain stable body weight in the long-term needs to be clarified in addition to possible adverse effects. Lifestyle interventions revert the nutritional trigger for obesity and positively affect our overall health, including the cardiovascular system. This Perspective examines how lifestyle interventions affect the neurovascular system and neuronal networks.
Collapse
Affiliation(s)
- Bandy Chen
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA.
| | - Elisa de Launoit
- Sorbonne Université, Institut Du Cerveau-Paris Brain Institute-ICM, Inserm U1127, CNRS UMR 7225, Paris, France
| | - David Meseguer
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
| | - Cristina Garcia Caceres
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Munich & German Center for Diabetes Research (DZD), Neuherberg, Germany
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Anne Eichmann
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA
- Cardiovascular Research Center, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- Paris Cardiovascular Research Center, Inserm U970, Université Paris, Paris, France
| | - Nicolas Renier
- Sorbonne Université, Institut Du Cerveau-Paris Brain Institute-ICM, Inserm U1127, CNRS UMR 7225, Paris, France
| | - Marc Schneeberger
- Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, USA.
- Wu Tsai Institute for Mind and Brain, Yale University, New Haven, CT, USA.
| |
Collapse
|
3
|
Yan R, Wei D, Varshneya A, Shan L, Dai B, Asencio HJ, Gollamudi A, Lin D. The multi-stage plasticity in the aggression circuit underlying the winner effect. Cell 2024; 187:6785-6803.e18. [PMID: 39406242 DOI: 10.1016/j.cell.2024.09.030] [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: 01/04/2024] [Revised: 07/17/2024] [Accepted: 09/18/2024] [Indexed: 10/23/2024]
Abstract
Winning increases the readiness to attack and the probability of winning, a widespread phenomenon known as the "winner effect." Here, we reveal a transition from target-specific to generalized aggression enhancement over 10 days of winning in male mice. This behavioral change is supported by three causally linked plasticity events in the ventrolateral part of the ventromedial hypothalamus (VMHvl), a critical node for aggression. Over 10 days of winning, VMHvl cells experience monotonic potentiation of long-range excitatory inputs, transient local connectivity strengthening, and a delayed excitability increase. Optogenetically coactivating the posterior amygdala (PA) terminals and VMHvl cells potentiates the PA-VMHvl pathway and triggers the same cascade of plasticity events observed during repeated winning. Optogenetically blocking PA-VMHvl synaptic potentiation eliminates all winning-induced plasticity. These results reveal the complex Hebbian synaptic and excitability plasticity in the aggression circuit during winning, ultimately leading to increased "aggressiveness" in repeated winners.
Collapse
Affiliation(s)
- Rongzhen Yan
- Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA.
| | - Dongyu Wei
- Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA
| | - Avni Varshneya
- Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA
| | - Lynn Shan
- Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA
| | - Bing Dai
- Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA
| | - Hector J Asencio
- Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA
| | - Aishwarya Gollamudi
- Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA
| | - Dayu Lin
- Neuroscience Institute, New York University Langone Medical Center, New York, NY 10016, USA; Department of Psychiatry, New York University Langone Medical Center, New York, NY 10016, USA; Department of Neuroscience and Physiology, New York University Langone Medical Center, New York, NY 10016, USA.
| |
Collapse
|
4
|
Qi G, Tang H, Gong P, Liu Y, He C, Hu J, Kang S, Chen L, Qin S. Sex-specific hypothalamic neuropathology and glucose metabolism in an amyloidosis transgenic mouse model of Alzheimer's disease. Cell Biosci 2024; 14:120. [PMID: 39272160 PMCID: PMC11395863 DOI: 10.1186/s13578-024-01295-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
BACKGROUND Amyloid toxicity and glucose metabolic disorders are key pathological features during the progression of Alzheimer's disease (AD). While the hypothalamus plays a crucial role in regulating systemic energy balance, the distribution of amyloid plaques in the preoptic, anterior, tuberal, and mammillary regions of the hypothalamus in AD mice, particularly across both sexes, remains largely unclear. Our ongoing research aims to explore hypothalamic neuropathology and glucose metabolic disturbances in a well-described APP/PS1 mouse model of AD. RESULTS Immunocytochemical staining revealed that Old-AD-Female mice exhibited a greater hypothalamic Amyloid β (Aβ) burden than their Old-AD-Male counterparts, with the mammillary bodies showing the most severe accumulation. Analysis of ionized calcium binding adaptor molecule 1 (IBA1) immunoreactivity and Iba1 mRNA indicated differential microgliosis based on sex, while tanycytic territory and ZO-1 tight junction protein expression remained stable in AD mice. Moreover, sex-specific peripheral glucose metabolic parameters (random and fasting blood glucose) seemed to be exacerbated by age. Old AD mice of both sexes exhibited limited hypothalamic activation (c-Fos + cells) in response to blood glucose fluctuations. Hypothalamic Glut 1 expression decreased in young but increased in old female AD mice compared with age-matched male AD mice. Pearson correlation analysis further supported a negative correlation between hypothalamic Aβ load and random blood glucose in old AD groups of both genders, shedding light on the mechanisms underlying this amyloidosis mouse model. CONCLUSION Aged APP/PS1 mice exhibit sex-specific hypothalamic neuropathology and differential glucose metabolism, highlighting distinct pathological mechanisms within each gender.
Collapse
Affiliation(s)
- Guibo Qi
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Han Tang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Pifang Gong
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Yitong Liu
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Chenzhao He
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Jianian Hu
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Siying Kang
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Liang Chen
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| | - Song Qin
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
5
|
Duquenne M, Deligia E, Folgueira C, Bourouh C, Caron E, Pfrieger F, Schwaninger M, Nogueiras R, Annicotte JS, Imbernon M, Prévot V. Tanycytic transcytosis inhibition disrupts energy balance, glucose homeostasis and cognitive function in male mice. Mol Metab 2024; 87:101996. [PMID: 39047908 PMCID: PMC11340606 DOI: 10.1016/j.molmet.2024.101996] [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: 05/10/2024] [Revised: 07/06/2024] [Accepted: 07/16/2024] [Indexed: 07/27/2024] Open
Abstract
OBJECTIVES In Western society, high-caloric diets rich in fats and sugars have fueled the obesity epidemic and its related disorders. Disruption of the body-brain communication, crucial for maintaining glucose and energy homeostasis, arises from both obesogenic and genetic factors, leading to metabolic disorders. Here, we investigate the role of hypothalamic tanycyte shuttles between the pituitary portal blood and the third ventricle cerebrospinal fluid in regulating energy balance. METHODS We inhibited vesicle-associated membrane proteins (VAMP1-3)-mediated release in tanycytes by expressing the botulinum neurotoxin type B light chain (BoNT/B) in a Cre-dependent manner in tanycytes. This was achieved by injecting either TAT-Cre in the third ventricle or an AAV1/2 expressing Cre under the control of the tanycyte-specific promoter iodothyronine deiodinase 2 into the lateral ventricle of adult male mice. RESULTS In male mice fed a standard diet, targeted expression of BoNT/B in adult tanycytes blocks leptin transport into the mediobasal hypothalamus and results in normal-weight central obesity, including increased food intake, abdominal fat deposition, and elevated leptin levels but no marked change in body weight. Furthermore, BoNT/B expression in adult tanycytes promotes fatty acid storage, leading to glucose intolerance and insulin resistance. Notably, these metabolic disturbances occur despite a compensatory increase in insulin secretion, observed both in response to exogenous glucose boluses in vivo and in isolated pancreatic islets. Intriguingly, these metabolic alterations are associated with impaired spatial memory in BoNT/B-expressing mice. CONCLUSIONS These findings underscore the central role of tanycytes in brain-periphery communication and highlight their potential implication in the age-related development of type 2 diabetes and cognitive decline. Our tanycytic BoNT/B mouse model provides a robust platform for studying how these conditions progress over time, from prediabetic states to full-blown metabolic and cognitive disorders, and the mechanistic contribution of tanycytes to their development. The recognition of the impact of tanycytic transcytosis on hormone transport opens new avenues for developing targeted therapies that could address both metabolic disorders and their associated cognitive comorbidities, which often emerge or worsen with advancing age.
Collapse
Affiliation(s)
- Manon Duquenne
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR_S1172, EGID, DISTALZ, Lille, France
| | - Eleonora Deligia
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR_S1172, EGID, DISTALZ, Lille, France
| | - Cintia Folgueira
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Cyril Bourouh
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1011-EGID, F-59000 Lille, France
| | - Emilie Caron
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR_S1172, EGID, DISTALZ, Lille, France
| | - Frank Pfrieger
- Centre National de la Recherche Scientifique, Universite de Strasbourg, Institut des Neurosciences Cellulaires et Integratives, 67000 Strasbourg, France
| | - Markus Schwaninger
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
| | - Ruben Nogueiras
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Jean-Sébastien Annicotte
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, F-59000 Lille, France
| | - Monica Imbernon
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR_S1172, EGID, DISTALZ, Lille, France.
| | - Vincent Prévot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR_S1172, EGID, DISTALZ, Lille, France.
| |
Collapse
|
6
|
Fernandois D, Rusidzé M, Mueller-Fielitz H, Sauve F, Deligia E, Silva MSB, Evrard F, Franco-García A, Mazur D, Martinez-Corral I, Jouy N, Rasika S, Maurage CA, Giacobini P, Nogueiras R, Dehouck B, Schwaninger M, Lenfant F, Prevot V. Estrogen receptor-α signaling in tanycytes lies at the crossroads of fertility and metabolism. Metabolism 2024; 158:155976. [PMID: 39019342 PMCID: PMC7616427 DOI: 10.1016/j.metabol.2024.155976] [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: 03/28/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024]
Abstract
BACKGROUND Estrogen secretion by the ovaries regulates the hypothalamic-pituitary-gonadal axis during the reproductive cycle, influencing gonadotropin-releasing hormone (GnRH) and luteinizing hormone (LH) secretion, and also plays a role in regulating metabolism. Here, we establish that hypothalamic tanycytes-specialized glia lining the floor and walls of the third ventricle-integrate estrogenic feedback signals from the gonads and couple reproduction with metabolism by relaying this information to orexigenic neuropeptide Y (NPY) neurons. METHODS Using mouse models, including mice floxed for Esr1 (encoding estrogen receptor alpha, ERα) and those with Cre-dependent expression of designer receptors exclusively activated by designer drugs (DREADDs), along with viral-mediated, pharmacological and indirect calorimetric approaches, we evaluated the role of tanycytes and tanycytic estrogen signaling in pulsatile LH secretion, cFos expression in NPY neurons, estrous cyclicity, body-weight changes and metabolic parameters in adult females. RESULTS In ovariectomized mice, chemogenetic activation of tanycytes significantly reduced LH pulsatile release, mimicking the effects of direct NPY neuron activation. In intact mice, tanycytes were crucial for the estrogen-mediated control of GnRH/LH release, with tanycytic ERα activation suppressing fasting-induced NPY neuron activation. Selective knockout of Esr1 in tanycytes altered estrous cyclicity and fertility in female mice and affected estrogen's ability to inhibit refeeding in fasting mice. The absence of ERα signaling in tanycytes increased Npy transcripts and body weight in intact mice and prevented the estrogen-mediated decrease in food intake as well as increase in energy expenditure and fatty acid oxidation in ovariectomized mice. CONCLUSIONS Our findings underscore the pivotal role of tanycytes in the neuroendocrine coupling of reproduction and metabolism, with potential implications for its age-related deregulation after menopause. SIGNIFICANCE STATEMENT Our investigation reveals that tanycytes, specialized glial cells in the brain, are key interpreters of estrogen signals for orexigenic NPY neurons in the hypothalamus. Disrupting tanycytic estrogen receptors not only alters fertility in female mice but also impairs the ability of estrogens to suppress appetite. This work thus sheds light on the critical role played by tanycytes in bridging the hormonal regulation of cyclic reproductive function and appetite/feeding behavior. This understanding may have potential implications for age-related metabolic deregulation after menopause.
Collapse
Affiliation(s)
- Daniela Fernandois
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, F-59000 Lille, France
| | - Mariam Rusidzé
- Institute of Metabolic and Cardiovascular Diseases (I2MC) Equipe 4, Inserm U1297UPS, CHU, Toulouse, France
| | - Helge Mueller-Fielitz
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
| | - Florent Sauve
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, F-59000 Lille, France
| | - Eleonora Deligia
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, F-59000 Lille, France
| | - Mauro S B Silva
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, F-59000 Lille, France
| | - Florence Evrard
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, F-59000 Lille, France
| | - Aurelio Franco-García
- Group of Cellular and Molecular Pharmacology, Department of Pharmacology, CEIR Campus Mare Nostrum, University of Murcia, Spain, Instituto Murciano de Investigación Biosanitaria (IMIB), Pascual Parrilla, Murcia, Spain
| | - Daniele Mazur
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, F-59000 Lille, France
| | - Ines Martinez-Corral
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, F-59000 Lille, France
| | | | - S Rasika
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, F-59000 Lille, France
| | - Claude-Alain Maurage
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, F-59000 Lille, France
| | - Paolo Giacobini
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, F-59000 Lille, France
| | - Ruben Nogueiras
- CIMUS, Universidade de Santiago de Compostela-Instituto de Investigación Sanitaria, Santiago de Compostela 15782, Spain- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), 15706, Spain
| | - Benedicte Dehouck
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, F-59000 Lille, France
| | - Markus Schwaninger
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
| | - Francoise Lenfant
- Institute of Metabolic and Cardiovascular Diseases (I2MC) Equipe 4, Inserm U1297UPS, CHU, Toulouse, France
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, F-59000 Lille, France.
| |
Collapse
|
7
|
Modder M, Coomans CP, Saaltink DJ, Tersteeg MMH, Hoogduin J, Scholten L, Pronk ACM, Lalai RA, Boelen A, Kalsbeek A, Rensen PCN, Vreugdenhil E, Kooijman S. Doublecortin-like knockdown in mice attenuates obesity by stimulating energy expenditure in adipose tissue. Sci Rep 2024; 14:19517. [PMID: 39174821 PMCID: PMC11341836 DOI: 10.1038/s41598-024-70639-5] [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: 04/24/2024] [Accepted: 08/20/2024] [Indexed: 08/24/2024] Open
Abstract
Crosstalk between peripheral metabolic organs and the central nervous system is essential for body weight control. At the base of the hypothalamus, β-tanycytes surround the portal capillaries and function as gatekeepers to facilitate transfer of substances from the circulation into the cerebrospinal fluid and vice versa. Here, we investigated the role of the neuroplasticity gene doublecortin-like (DCL), highly expressed by β-tanycytes, in body weight control and whole-body energy metabolism. We demonstrated that DCL-knockdown through a doxycycline-inducible shRNA expression system prevents body weight gain by reducing adiposity in mice. DCL-knockdown slightly increased whole-body energy expenditure possibly as a result of elevated circulating thyroid hormones. In white adipose tissue (WAT) triglyceride uptake was increased while the average adipocyte cell size was reduced. At histological level we observed clear signs of browning, and thus increased thermogenesis in WAT. We found no indications for stimulated thermogenesis in brown adipose tissue (BAT). Altogether, we demonstrate an important, though subtle, role of tanycytic DCL in body weight control through regulation of energy expenditure, and specifically WAT browning. Elucidating mechanisms underlying the role of DCL in regulating brain-peripheral crosstalk further might identify new treatment targets for obesity.
Collapse
Affiliation(s)
- Melanie Modder
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Claudia P Coomans
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Dirk-Jan Saaltink
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Mayke M H Tersteeg
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Janna Hoogduin
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Leonie Scholten
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Amanda C M Pronk
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Reshma A Lalai
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Anita Boelen
- Endocrine Laboratory, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Andries Kalsbeek
- Endocrine Laboratory, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
- Department of Endocrinology and Metabolism, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
| | - Patrick C N Rensen
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Erno Vreugdenhil
- Laboratory for Neurophysiology, Department of Cell and Chemical Biology, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | - Sander Kooijman
- Division of Endocrinology, Department of Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands.
- Leiden University Medical Center, Albinusdreef 2, 2333ZA, Leiden, The Netherlands.
| |
Collapse
|
8
|
Yan R, Wei D, Varshneya A, Shan L, Asencio HJ, Lin D. The multi-stage plasticity in the aggression circuit underlying the winner effect. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.19.608611. [PMID: 39229201 PMCID: PMC11370333 DOI: 10.1101/2024.08.19.608611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Winning increases the readiness to attack and the probability of winning, a widespread phenomenon known as the "winner effect". Here, we reveal a transition from target-specific to generalized aggression enhancement over 10 days of winning in male mice, which is supported by three stages of plasticity in the ventrolateral part of the ventromedial hypothalamus (VMHvl), a critical node for aggression. Over 10-day winning, VMHvl cells experience monotonic potentiation of long-range excitatory inputs, a transient local connectivity strengthening, and a delayed excitability increase. These plasticity events are causally linked. Optogenetically coactivating the posterior amygdala (PA) terminals and VMHvl cells potentiates the PA-VMHvl pathway and triggers the cascade of plasticity events as those during repeated winning. Optogenetically blocking PA-VMHvl synaptic potentiation eliminates all winning-induced plasticity. These results reveal the complex Hebbian synaptic and excitability plasticity in the aggression circuit during winning that ultimately leads to an increase in "aggressiveness" in repeated winners.
Collapse
|
9
|
Liu D, Wang T, Zhao X, Chen J, Yang T, Shen Y, Zhou YD. Saturated fatty acids stimulate cytokine production in tanycytes via the PP2Ac-dependent signaling pathway. J Cereb Blood Flow Metab 2024; 44:985-999. [PMID: 38069840 PMCID: PMC11318396 DOI: 10.1177/0271678x231219115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 09/18/2023] [Accepted: 11/10/2023] [Indexed: 05/18/2024]
Abstract
The hypothalamic tanycytes are crucial for free fatty acids (FFAs) detection, storage, and transport within the central nervous system. They have been shown to effectively respond to fluctuations in circulating FFAs, thereby regulating energy homeostasis. However, the precise molecular mechanisms by which tanycytes modulate lipid utilization remain unclear. Here, we report that the catalytic subunit of protein phosphatase 2 A (PP2Ac), a serine/threonine phosphatase, is expressed in tanycytes and its accumulation and activation occur in response to high-fat diet consumption. In vitro, tanycytic PP2Ac responds to palmitic acid (PA) exposure and accumulates and is activated at an early stage in an AMPK-dependent manner. Furthermore, activated PP2Ac boosts hypoxia-inducible factor-1α (HIF-1α) accumulation, resulting in upregulation of an array of cytokines. Pretreatment with a PP2Ac inhibitor, LB100, prevented the PA-induced elevation of vascular endothelial growth factor (VEGF), fibroblast growth factor 1 (FGF1), hepatocyte growth factor (HGF), and dipeptidyl peptidase IV (DPPIV or CD26). Our results disclose a mechanism of lipid metabolism in tanycytes that involves the activation of PP2Ac and highlight the physiological significance of PP2Ac in hypothalamic tanycytes in response to overnutrition and efficacious treatment of obesity.
Collapse
Affiliation(s)
- Danyang Liu
- Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Brain Science and Brain Medicine, Hangzhou, China
- Nanhu Brain-computer Interface Institute, Hangzhou 311100, China
- Department of Ophthalmology of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, China
| | - Tao Wang
- Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Brain Science and Brain Medicine, Hangzhou, China
| | - Xingqi Zhao
- Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Brain Science and Brain Medicine, Hangzhou, China
| | - Juan Chen
- School of Mental Health, Bengbu Medical College, Bengbu, Anhui, China
| | - Tianqi Yang
- Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Brain Science and Brain Medicine, Hangzhou, China
| | - Yi Shen
- Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Brain Science and Brain Medicine, Hangzhou, China
- Department of Neurobiology and Department of General Intensive Care Unit of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu-Dong Zhou
- Institute of Neuroscience, NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, Zhejiang University School of Brain Science and Brain Medicine, Hangzhou, China
- Lingang Laboratory, Shanghai 200031, China
- Department of Ophthalmology of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Laboratory for Systems & Precision Medicine, Zhejiang University Medical Center, Hangzhou, China
| |
Collapse
|
10
|
Barahona MJ, Ferrada L, Vera M, Nualart F. Tanycytes release glucose using the glucose-6-phosphatase system during hypoglycemia to control hypothalamic energy balance. Mol Metab 2024; 84:101940. [PMID: 38641253 PMCID: PMC11060961 DOI: 10.1016/j.molmet.2024.101940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/09/2024] [Accepted: 04/09/2024] [Indexed: 04/21/2024] Open
Abstract
OBJECTIVE The liver releases glucose into the blood using the glucose-6-phosphatase (G6Pase) system, a multiprotein complex located in the endoplasmic reticulum (ER). Here, we show for the first time that the G6Pase system is also expressed in hypothalamic tanycytes, and it is required to regulate energy balance. METHODS Using automatized qRT-PCR and immunohistochemical analyses, we evaluated the expression of the G6Pase system. Fluorescent glucose analogue (2-NBDG) uptake was evaluated by 4D live-cell microscopy. Glucose release was tested using a glucose detection kit and high-content live-cell analysis instrument, Incucyte s3. In vivo G6pt knockdown in tanycytes was performed by AAV1-shG6PT-mCherry intracerebroventricular injection. Body weight gain, adipose tissue weight, food intake, glucose metabolism, c-Fos, and neuropeptide expression were evaluated at 4 weeks post-transduction. RESULTS Tanycytes sequester glucose-6-phosphate (G6P) into the ER through the G6Pase system and release glucose in hypoglycaemia via facilitative glucose transporters (GLUTs). Strikingly, in vivo tanycytic G6pt knockdown has a powerful peripheral anabolic effect observed through decreased body weight, white adipose tissue (WAT) tissue mass, and strong downregulation of lipogenesis genes. Selective deletion of G6pt in tanycytes also decreases food intake, c-Fos expression in the arcuate nucleus (ARC), and Npy mRNA expression in fasted mice. CONCLUSIONS The tanycyte-associated G6Pase system is a central mechanism involved in controlling metabolism and energy balance.
Collapse
Affiliation(s)
- María José Barahona
- Laboratory of Neurobiology and Stem Cells, NeuroCellT, Department of Cellular Biology, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile; Center for Advanced Microscopy CMA BIO BIO, University of Concepcion, Concepcion, Chile; Laboratory of Appetite Physiology (FIDELA), Faculty of Medicine and Sciences, University San Sebastián, Concepción Campus, Concepción, Chile
| | - Luciano Ferrada
- Center for Advanced Microscopy CMA BIO BIO, University of Concepcion, Concepcion, Chile
| | - Matías Vera
- Laboratory of Neurobiology and Stem Cells, NeuroCellT, Department of Cellular Biology, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile; Center for Advanced Microscopy CMA BIO BIO, University of Concepcion, Concepcion, Chile
| | - Francisco Nualart
- Laboratory of Neurobiology and Stem Cells, NeuroCellT, Department of Cellular Biology, Faculty of Biological Sciences, University of Concepcion, Concepcion, Chile; Center for Advanced Microscopy CMA BIO BIO, University of Concepcion, Concepcion, Chile.
| |
Collapse
|
11
|
Buller S, Blouet C. Brain access of incretins and incretin receptor agonists to their central targets relevant for appetite suppression and weight loss. Am J Physiol Endocrinol Metab 2024; 326:E472-E480. [PMID: 38381398 PMCID: PMC11193531 DOI: 10.1152/ajpendo.00250.2023] [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: 08/11/2023] [Revised: 01/05/2024] [Accepted: 02/13/2024] [Indexed: 02/22/2024]
Abstract
New incretin-based pharmacotherapies provide efficient and safe therapeutic options to curb appetite and produce weight loss in patients with obesity. Delivered systemically, these molecules produce pleiotropic metabolic benefits, but the target sites mediating their weight-suppressive action are located within the brain. Recent research has increased our understanding of the neural circuits and behavioral mechanisms involved in the anorectic and metabolic consequences of glucagon-like peptide 1 (GLP-1)-based weight loss strategies, yet little is known about how these drugs access their functional targets in the brain to produce sustained weight loss. The majority of brain cells expressing incretin receptors are located behind the blood-brain barrier, shielded from the circulation and fluctuations in the availability of peripheral signals, which is a major challenge for the development of CNS-targeted therapeutic peptides. GLP-1 receptor (GLP-1R) agonists with increased half-life and enhanced therapeutic benefit do not cross the blood-brain barrier, yet they manage to access discrete brain sites relevant to the regulation of energy homeostasis. In this review, we give a brief overview of the different routes for peptide hormones to access the brain. We then examine the evidence informing the routes employed by incretins and incretin receptor agonists to access brain targets relevant for their appetite and weight-suppressive actions. We highlight existing controversies and suggest future directions to further establish the functionally relevant access routes for GLP-1-based weight loss compounds, which might guide the development and selection of the future generation of incretin receptor polypharmacologies.
Collapse
Affiliation(s)
- Sophie Buller
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| | - Clemence Blouet
- Medical Research Council (MRC) Metabolic Diseases Unit, Institute of Metabolic Science, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
12
|
Thornton P, Reader V, Digby Z, Smolak P, Lindsay N, Harrison D, Clarke N, Watt AP. Reversal of High Fat Diet-Induced Obesity, Systemic Inflammation, and Astrogliosis by the NLRP3 Inflammasome Inhibitors NT-0249 and NT-0796. J Pharmacol Exp Ther 2024; 388:813-826. [PMID: 38336379 DOI: 10.1124/jpet.123.002013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/14/2023] [Accepted: 01/02/2024] [Indexed: 02/12/2024] Open
Abstract
Systemic and cerebral inflammatory responses are implicated in the pathogenesis of obesity and associated metabolic impairment. While the NOD-, LRR-, and pyrin domain-containing protein 3 (NLRP3) inflammasome has been linked to obesity-associated inflammation, whether it contributes to the development or maintenance of obesity is unknown. We provide support for a direct role of saturated fatty acids, such as palmitic acid, as NLRP3 activating stimuli in obese states. To investigate whether NLRP3 activation contributes to the pathogenesis of diet-induced obesity (DIO) in mice, we tested two different clinical-stage NLRP3 inflammasome inhibitors. We demonstrate a contributory role of this key inflammasome to established obesity and associated systemic and cerebral inflammation. By comparing their effects to calorie restriction, we aimed to identify specific NLRP3-sensitive mechanisms contributing to obesity-induced inflammation (as opposed to be those regulated by weight loss per se). In addition, a direct comparison of an NLRP3 inhibitor to a glucagon like peptide-1 receptor agonist, semaglutide (Wegovy), in the DIO model allowed an appreciation of the relative efficacy of these two therapeutic strategies on obesity, its associated systemic inflammatory response, and cerebral gliosis. We show that two structurally distinct, NLRP3 inhibitors, NT-0249 and NT-0796, reverse obesity in the DIO mouse model and that brain exposure appears necessary for efficacy. In support of this, we show that DIO-driven hypothalamic glial fibrillary acidic protein expression is blocked by dosing with NT-0249/NT-0796. While matching weight loss driven by semaglutide or calorie restriction, remarkably, NLRP3 inhibition provided enhanced improvements in disease-relevant biomarkers of acute phase response, cardiovascular inflammation, and lipid metabolism. SIGNIFICANCE STATEMENT: Obesity is a global health concern that predisposes individuals to chronic disease such as diabetes and cardiovascular disease at least in part by promoting systemic inflammation. We report that in mice fed a high-fat, obesogenic diet, obesity is reversed by either of two inhibitors of the intracellular inflammatory mediator NLRP3. Furthermore, NLRP3 inhibition reduces both hypothalamic gliosis and circulating biomarkers of cardiovascular disease risk beyond what can be achieved by either the glucagon like peptide-1 agonist semaglutide or calorie restriction alone.
Collapse
Affiliation(s)
- Peter Thornton
- NodThera, Cambridge, United Kingdom (P.T., V.R., Z.D., N.L., D.H., N.C., A.P.W.) and Seattle, Washington (P.S.)
| | - Valérie Reader
- NodThera, Cambridge, United Kingdom (P.T., V.R., Z.D., N.L., D.H., N.C., A.P.W.) and Seattle, Washington (P.S.)
| | - Zsofia Digby
- NodThera, Cambridge, United Kingdom (P.T., V.R., Z.D., N.L., D.H., N.C., A.P.W.) and Seattle, Washington (P.S.)
| | - Pamela Smolak
- NodThera, Cambridge, United Kingdom (P.T., V.R., Z.D., N.L., D.H., N.C., A.P.W.) and Seattle, Washington (P.S.)
| | - Nicola Lindsay
- NodThera, Cambridge, United Kingdom (P.T., V.R., Z.D., N.L., D.H., N.C., A.P.W.) and Seattle, Washington (P.S.)
| | - David Harrison
- NodThera, Cambridge, United Kingdom (P.T., V.R., Z.D., N.L., D.H., N.C., A.P.W.) and Seattle, Washington (P.S.)
| | - Nick Clarke
- NodThera, Cambridge, United Kingdom (P.T., V.R., Z.D., N.L., D.H., N.C., A.P.W.) and Seattle, Washington (P.S.)
| | - Alan P Watt
- NodThera, Cambridge, United Kingdom (P.T., V.R., Z.D., N.L., D.H., N.C., A.P.W.) and Seattle, Washington (P.S.)
| |
Collapse
|
13
|
Jörgensen SK, Karnošová A, Mazzaferro S, Rowley O, Chen HJC, Robbins SJ, Christofides S, Merkle FT, Maletínská L, Petrik D. An analogue of the Prolactin Releasing Peptide reduces obesity and promotes adult neurogenesis. EMBO Rep 2024; 25:351-377. [PMID: 38177913 PMCID: PMC10897398 DOI: 10.1038/s44319-023-00016-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 11/02/2023] [Accepted: 11/17/2023] [Indexed: 01/06/2024] Open
Abstract
Hypothalamic Adult Neurogenesis (hAN) has been implicated in regulating energy homeostasis. Adult-generated neurons and adult Neural Stem Cells (aNSCs) in the hypothalamus control food intake and body weight. Conversely, diet-induced obesity (DIO) by high fat diets (HFD) exerts adverse influence on hAN. However, the effects of anti-obesity compounds on hAN are not known. To address this, we administered a lipidized analogue of an anti-obesity neuropeptide, Prolactin Releasing Peptide (PrRP), so-called LiPR, to mice. In the HFD context, LiPR rescued the survival of adult-born hypothalamic neurons and increased the number of aNSCs by reducing their activation. LiPR also rescued the reduction of immature hippocampal neurons and modulated calcium dynamics in iPSC-derived human neurons. In addition, some of these neurogenic effects were exerted by another anti-obesity compound, Liraglutide. These results show for the first time that anti-obesity neuropeptides influence adult neurogenesis and suggest that the neurogenic process can serve as a target of anti-obesity pharmacotherapy.
Collapse
Affiliation(s)
| | - Alena Karnošová
- First Faculty of Medicine, Charles University, Prague, 12108, Czech Republic
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, 16610, Czech Republic
| | - Simone Mazzaferro
- Wellcome-MRC Institute of Metabolic Science, Cambridge, CB2 0QQ, UK
- Wellcome-MRC Stem Cell Institute, Cambridge, CB2 0AW, UK
| | - Oliver Rowley
- School of Biosciences, Cardiff University, Cardiff, CF10 3AX, UK
| | - Hsiao-Jou Cortina Chen
- Wellcome-MRC Institute of Metabolic Science, Cambridge, CB2 0QQ, UK
- Wellcome-MRC Stem Cell Institute, Cambridge, CB2 0AW, UK
| | - Sarah J Robbins
- School of Biosciences, Cardiff University, Cardiff, CF10 3AX, UK
| | | | - Florian T Merkle
- Wellcome-MRC Institute of Metabolic Science, Cambridge, CB2 0QQ, UK
- Wellcome-MRC Stem Cell Institute, Cambridge, CB2 0AW, UK
| | - Lenka Maletínská
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Prague, 16610, Czech Republic
| | - David Petrik
- School of Biosciences, Cardiff University, Cardiff, CF10 3AX, UK.
| |
Collapse
|
14
|
Manuel J, Halbe E, Ewald AC, Hoff A, Jordan J, Tank J, Heusser K, Gerlach DA. Glucose-sensitive hypothalamic nuclei traced through functional magnetic resonance imaging. Front Neurosci 2023; 17:1297197. [PMID: 38146542 PMCID: PMC10749345 DOI: 10.3389/fnins.2023.1297197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 11/20/2023] [Indexed: 12/27/2023] Open
Abstract
Introduction Hypothalamic glucose-sensitive neural circuits, which regulate energy metabolism and can contribute to diseases such as obesity and type 2 diabetes, have been difficult to study in humans. We developed an approach to assess hypothalamic functional connectivity changes during glucose loading using functional magnetic resonance imaging (fMRI). Methods To do so, we conducted oral glucose tolerance tests while acquiring functional images before, and 10 and 45 min after glucose ingestion in a healthy male and cross-sectionally in 20 healthy participants on two different diets. Results At group level, 39 fMRI sessions were not sufficient to detect glucose-mediated connectivity changes. However, 10 repeated sessions in a single subject revealed significant intrinsic functional connectivity increases 45 min after glucose intake in the arcuate, paraventricular, and dorsomedial nuclei, as well as in the posterior hypothalamic area, median eminence, and mammillary bodies. Discussion Our methodology allowed to outline glucose-sensitive hypothalamic pathways in a single human being and holds promise in delineating individual pathophysiology mechanisms in patients with dysglycemia.
Collapse
Affiliation(s)
- Jorge Manuel
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
- Institute for Neuroradiology, Hannover Medical School, Hanover, Germany
| | - Eva Halbe
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
- Department of Psychiatry and Psychotherapy, University of Bonn, Bonn, Germany
| | - Ann Charlotte Ewald
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Alex Hoff
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Jens Jordan
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
- Medical Faculty, University of Cologne, Cologne, Germany
| | - Jens Tank
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Karsten Heusser
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Darius A. Gerlach
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| |
Collapse
|
15
|
Kannangara H, Cullen L, Miyashita S, Korkmaz F, Macdonald A, Gumerova A, Witztum R, Moldavski O, Sims S, Burgess J, Frolinger T, Latif R, Ginzburg Y, Lizneva D, Goosens K, Davies TF, Yuen T, Zaidi M, Ryu V. Emerging roles of brain tanycytes in regulating blood-hypothalamus barrier plasticity and energy homeostasis. Ann N Y Acad Sci 2023; 1525:61-69. [PMID: 37199228 PMCID: PMC10524199 DOI: 10.1111/nyas.15009] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Seasonal changes in food intake and adiposity in many animal species are triggered by changes in the photoperiod. These latter changes are faithfully transduced into a biochemical signal by melatonin secreted by the pineal gland. Seasonal variations, encoded by melatonin, are integrated by third ventricular tanycytes of the mediobasal hypothalamus through the detection of the thyroid-stimulating hormone (TSH) released from the pars tuberalis. The mediobasal hypothalamus is a critical brain region that maintains energy homeostasis by acting as an interface between the neural networks of the central nervous system and the periphery to control metabolic functions, including ingestive behavior, energy homeostasis, and reproduction. Among the cells involved in the regulation of energy balance and the blood-hypothalamus barrier (BHB) plasticity are tanycytes. Increasing evidence suggests that anterior pituitary hormones, specifically TSH, traditionally considered to have unitary functions in targeting single endocrine sites, display actions on multiple somatic tissues and central neurons. Notably, modulation of tanycytic TSH receptors seems critical for BHB plasticity in relation to energy homeostasis, but this needs to be proven.
Collapse
Affiliation(s)
- Hasni Kannangara
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Liam Cullen
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Sari Miyashita
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Funda Korkmaz
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Anne Macdonald
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Anisa Gumerova
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ronit Witztum
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ofer Moldavski
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Steven Sims
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Jocoll Burgess
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Tal Frolinger
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Rauf Latif
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Yelena Ginzburg
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Daria Lizneva
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ki Goosens
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Terry F. Davies
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Tony Yuen
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Mone Zaidi
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Vitaly Ryu
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
16
|
Infantes-López MI, Nieto-Quero A, Chaves-Peña P, Zambrana-Infantes E, Cifuentes M, Márquez J, Pedraza C, Pérez-Martín M. New insights into hypothalamic neurogenesis disruption after acute and intense stress: implications for microglia and inflammation. Front Neurosci 2023; 17:1190418. [PMID: 37425000 PMCID: PMC10327603 DOI: 10.3389/fnins.2023.1190418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/23/2023] [Indexed: 07/11/2023] Open
Abstract
In recent years, the hypothalamus has emerged as a new neurogenic area, capable of generating new neurons after development. Neurogenesis-dependent neuroplasticity seems to be critical to continuously adapt to internal and environmental changes. Stress is a potent environmental factor that can produce potent and enduring effects on brain structure and function. Acute and chronic stress is known to cause alterations in neurogenesis and microglia in classical adult neurogenic regions such as the hippocampus. The hypothalamus is one of the major brain regions implicated in homeostatic stress and emotional stress systems, but little is known about the effect of stress on the hypothalamus. Here, we studied the impact of acute and intense stress (water immersion and restrain stress, WIRS), which may be considered as an inducer of an animal model of posttraumatic stress disorder, on neurogenesis and neuroinflammation in the hypothalamus of adult male mice, focusing on three nuclei: PVN, VMN and ARC, and also in the periventricular area. Our data revealed that a unique stressor was sufficient to provoke a significant impact on hypothalamic neurogenesis by inducing a reduction in the proliferation and number of immature neurons identified as DCX+ cells. These differences were accompanied by marked microglial activation in the VMN and ARC, together with a concomitant increase in IL-6 levels, indicating that WIRS induced an inflammatory response. To investigate the possible molecular mechanisms responsible for neuroplastic and inflammatory changes, we tried to identify proteomic changes. The data revealed that WIRS induced changes in the hypothalamic proteome, modifying the abundance of three and four proteins after 1 h or 24 h of stress application, respectively. These changes were also accompanied by slight changes in the weight and food intake of the animals. These results are the first to show that even a short-term environmental stimulus such as acute and intense stress can have neuroplastic, inflammatory, functional and metabolic consequences on the adult hypothalamus.
Collapse
Affiliation(s)
- María Inmaculada Infantes-López
- Departamento de Biología Celular, Genética y Fisiología, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma Bionand, Málaga, Spain
| | - Andrea Nieto-Quero
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma Bionand, Málaga, Spain
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Universidad de Málaga, Málaga, Spain
| | - Patricia Chaves-Peña
- Departamento de Biología Celular, Genética y Fisiología, Universidad de Málaga, Málaga, Spain
| | - Emma Zambrana-Infantes
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma Bionand, Málaga, Spain
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Universidad de Málaga, Málaga, Spain
| | - Manuel Cifuentes
- Departamento de Biología Celular, Genética y Fisiología, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma Bionand, Málaga, Spain
| | - Javier Márquez
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma Bionand, Málaga, Spain
- Departamento de Biología Molecular y Bioquímica, Canceromics Lab, Universidad de Málaga, Málaga, Spain
| | - Carmen Pedraza
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma Bionand, Málaga, Spain
- Departamento de Psicobiología y Metodología de las Ciencias del Comportamiento, Universidad de Málaga, Málaga, Spain
| | - Margarita Pérez-Martín
- Departamento de Biología Celular, Genética y Fisiología, Universidad de Málaga, Málaga, Spain
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina–IBIMA Plataforma Bionand, Málaga, Spain
| |
Collapse
|
17
|
Dali R, Estrada-Meza J, Langlet F. Tanycyte, the neuron whisperer. Physiol Behav 2023; 263:114108. [PMID: 36740135 DOI: 10.1016/j.physbeh.2023.114108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/23/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023]
Abstract
Reciprocal communication between neurons and glia is essential for normal brain functioning and adequate physiological functions, including energy balance. In vertebrates, the homeostatic process that adjusts food intake and energy expenditure in line with physiological requirements is tightly controlled by numerous neural cell types located within the hypothalamus and the brainstem and organized in complex networks. Within these neural networks, peculiar ependymoglial cells called tanycytes are nowadays recognized as multifunctional players in the physiological mechanisms of appetite control, partly by modulating orexigenic and anorexigenic neurons. Here, we review recent advances in tanycytes' impact on hypothalamic neuronal activity, emphasizing on arcuate neurons.
Collapse
Affiliation(s)
- Rafik Dali
- Department of biomedical sciences, University of Lausanne, 1005 Lausanne, Switzerland
| | - Judith Estrada-Meza
- Department of biomedical sciences, University of Lausanne, 1005 Lausanne, Switzerland
| | - Fanny Langlet
- Department of biomedical sciences, University of Lausanne, 1005 Lausanne, Switzerland.
| |
Collapse
|
18
|
Yu Q, Gamayun I, Wartenberg P, Zhang Q, Qiao S, Kusumakshi S, Candlish S, Götz V, Wen S, Das D, Wyatt A, Wahl V, Ectors F, Kattler K, Yildiz D, Prevot V, Schwaninger M, Ternier G, Giacobini P, Ciofi P, Müller TD, Boehm U. Bitter taste cells in the ventricular walls of the murine brain regulate glucose homeostasis. Nat Commun 2023; 14:1588. [PMID: 36949050 PMCID: PMC10033832 DOI: 10.1038/s41467-023-37099-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 03/02/2023] [Indexed: 03/24/2023] Open
Abstract
The median eminence (ME) is a circumventricular organ at the base of the brain that controls body homeostasis. Tanycytes are its specialized glial cells that constitute the ventricular walls and regulate different physiological states, however individual signaling pathways in these cells are incompletely understood. Here, we identify a functional tanycyte subpopulation that expresses key taste transduction genes including bitter taste receptors, the G protein gustducin and the gustatory ion channel TRPM5 (M5). M5 tanycytes have access to blood-borne cues via processes extended towards diaphragmed endothelial fenestrations in the ME and mediate bidirectional communication between the cerebrospinal fluid and blood. This subpopulation responds to metabolic signals including leptin and other hormonal cues and is transcriptionally reprogrammed upon fasting. Acute M5 tanycyte activation induces insulin secretion and acute diphtheria toxin-mediated M5 tanycyte depletion results in impaired glucose tolerance in diet-induced obese mice. We provide a cellular and molecular framework that defines how bitter taste cells in the ME integrate chemosensation with metabolism.
Collapse
Affiliation(s)
- Qiang Yu
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Igor Gamayun
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Philipp Wartenberg
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Qian Zhang
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Sen Qiao
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Soumya Kusumakshi
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Sarah Candlish
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Viktoria Götz
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Shuping Wen
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Debajyoti Das
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Amanda Wyatt
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Vanessa Wahl
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Fabien Ectors
- FARAH Mammalian Transgenics Platform, Liège University, Liège, Belgium
| | - Kathrin Kattler
- Department of Genetics, Saarland University, Saarbrücken, Germany
| | - Daniela Yildiz
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Postnatal Brain, Lille Neuroscience & Cognition, UMR-S1172, Lille, France
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism (CBBM), University of Lübeck, Lübeck, Germany
| | - Gaetan Ternier
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Postnatal Brain, Lille Neuroscience & Cognition, UMR-S1172, Lille, France
| | - Paolo Giacobini
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Postnatal Brain, Lille Neuroscience & Cognition, UMR-S1172, Lille, France
| | - Philippe Ciofi
- Neurocentre Magendie - INSERM Unit 1215, University of Bordeaux, Bordeaux, France
| | - Timo D Müller
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center, Helmholtz Zentrum München, Neuherberg, Germany; German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Ulrich Boehm
- Department of Pharmacology, Center for Molecular Signaling (PZMS), Saarland University School of Medicine, Homburg, Germany.
| |
Collapse
|
19
|
Chinnaiya K, Burbridge S, Jones A, Kim DW, Place E, Manning E, Groves I, Sun C, Towers M, Blackshaw S, Placzek M. A neuroepithelial wave of BMP signalling drives anteroposterior specification of the tuberal hypothalamus. eLife 2023; 12:e83133. [PMID: 36718990 PMCID: PMC9917434 DOI: 10.7554/elife.83133] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 01/29/2023] [Indexed: 02/01/2023] Open
Abstract
The tuberal hypothalamus controls life-supporting homeostatic processes, but despite its fundamental role, the cells and signalling pathways that specify this unique region of the central nervous system in embryogenesis are poorly characterised. Here, we combine experimental and bioinformatic approaches in the embryonic chick to show that the tuberal hypothalamus is progressively generated from hypothalamic floor plate-like cells. Fate-mapping studies show that a stream of tuberal progenitors develops in the anterior-ventral neural tube as a wave of neuroepithelial-derived BMP signalling sweeps from anterior to posterior through the hypothalamic floor plate. As later-specified posterior tuberal progenitors are generated, early specified anterior tuberal progenitors become progressively more distant from these BMP signals and differentiate into tuberal neurogenic cells. Gain- and loss-of-function experiments in vivo and ex vivo show that BMP signalling initiates tuberal progenitor specification, but must be eliminated for these to progress to anterior neurogenic progenitors. scRNA-Seq profiling shows that tuberal progenitors that are specified after the major period of anterior tuberal specification begin to upregulate genes that characterise radial glial cells. This study provides an integrated account of the development of the tuberal hypothalamus.
Collapse
Affiliation(s)
- Kavitha Chinnaiya
- School of BiosciencesUniversity of Sheffield, SheffieldUnited Kingdom
| | - Sarah Burbridge
- School of BiosciencesUniversity of Sheffield, SheffieldUnited Kingdom
| | - Aragorn Jones
- School of BiosciencesUniversity of Sheffield, SheffieldUnited Kingdom
| | - Dong Won Kim
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Elsie Place
- School of BiosciencesUniversity of Sheffield, SheffieldUnited Kingdom
| | - Elizabeth Manning
- School of BiosciencesUniversity of Sheffield, SheffieldUnited Kingdom
| | - Ian Groves
- School of BiosciencesUniversity of Sheffield, SheffieldUnited Kingdom
| | - Changyu Sun
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Matthew Towers
- School of BiosciencesUniversity of Sheffield, SheffieldUnited Kingdom
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Psychiatry and Behavioral Science, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Ophthalmology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Neurology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Institute for Cell Engineering, Johns Hopkins University School of MedicineBaltimoreUnited States
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Marysia Placzek
- School of BiosciencesUniversity of Sheffield, SheffieldUnited Kingdom
- Bateson Centre, University of SheffieldSheffieldUnited Kingdom
- Neuroscience Institute, University of SheffieldSheffieldUnited Kingdom
| |
Collapse
|
20
|
Huang X, Wang YJ, Xiang Y. Bidirectional communication between brain and visceral white adipose tissue: Its potential impact on Alzheimer's disease. EBioMedicine 2022; 84:104263. [PMID: 36122553 PMCID: PMC9490488 DOI: 10.1016/j.ebiom.2022.104263] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 08/21/2022] [Accepted: 08/24/2022] [Indexed: 11/20/2022] Open
Abstract
A variety of axes between brain and abdominal organs have been reported, but the interaction between brain and visceral white adipose tissue (vWAT) remains unclear. In this review, we summarized human studies on the association between brain and vWAT, and generalized their interaction and the underlying mechanisms according to animal and cell experiments. On that basis, we come up with the concept of the brain-vWAT axis (BVA). Furthermore, we analyzed the potential mechanisms of involvement of BVA in the pathogenesis of Alzheimer's disease (AD), including vWAT-derived fatty acids, immunological properties of vWAT, vWAT-derived retinoic acid and vWAT-regulated insulin resistance. The proposal of BVA may expand our understanding to some extent of how the vWAT impacts on brain health and diseases, and provide a novel approach to study the pathogenesis and treatment strategies of neurodegenerative disorders.
Collapse
|
21
|
Wang J, Beecher K, Chehrehasa F, Moody H. The limitations of investigating appetite through circuit manipulations: are we biting off more than we can chew? Rev Neurosci 2022; 34:295-311. [PMID: 36054842 DOI: 10.1515/revneuro-2022-0072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 07/09/2022] [Indexed: 11/15/2022]
Abstract
Disordered eating can underpin a number of debilitating and prevalent chronic diseases, such as obesity. Broader advances in psychopharmacology and biology have motivated some neuroscientists to address diet-induced obesity through reductionist, pre-clinical eating investigations on the rodent brain. Specifically, chemogenetic and optogenetic methods developed in the 21st century allow neuroscientists to perform in vivo, region-specific/projection-specific/promoter-specific circuit manipulations and immediately assess the impact of these manipulations on rodent feeding. These studies are able to rigorously conclude whether a specific neuronal population regulates feeding behaviour in the hope of eventually developing a mechanistic neuroanatomical map of appetite regulation. However, an artificially stimulated/inhibited rodent neuronal population that changes feeding behaviour does not necessarily represent a pharmacological target for treating eating disorders in humans. Chemogenetic/optogenetic findings must therefore be triangulated with the array of theories that contribute to our understanding of appetite. The objective of this review is to provide a wide-ranging discussion of the limitations of chemogenetic/optogenetic circuit manipulation experiments in rodents that are used to investigate appetite. Stepping into and outside of medical science epistemologies, this paper draws on philosophy of science, nutrition, addiction biology and neurophilosophy to prompt more integrative, transdisciplinary interpretations of chemogenetic/optogenetic appetite data. Through discussing the various technical and epistemological limitations of these data, we provide both an overview of chemogenetics and optogenetics accessible to non-neuroscientist obesity researchers, as well as a resource for neuroscientists to expand the number of lenses through which they interpret their circuit manipulation findings.
Collapse
Affiliation(s)
- Joshua Wang
- School of Clinical Sciences, Faculty of Health, Queensland University of Technology, 2 George Street, Brisbane 4000, QLD, Australia
| | - Kate Beecher
- UQ Centre for Clinical Research, Faculty of Medicine, University of Queensland, Building 71/918 Royal Brisbane and Women's Hospital Campus, Herston 4029, QLD, Australia
| | - Fatemeh Chehrehasa
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, 2 George Street, Brisbane 4000, QLD, Australia
| | - Hayley Moody
- Queensland University of Technology, 2 George Street, Brisbane 4000, QLD, Australia
| |
Collapse
|
22
|
Nampoothiri S, Nogueiras R, Schwaninger M, Prevot V. Glial cells as integrators of peripheral and central signals in the regulation of energy homeostasis. Nat Metab 2022; 4:813-825. [PMID: 35879459 PMCID: PMC7613794 DOI: 10.1038/s42255-022-00610-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 06/15/2022] [Indexed: 01/03/2023]
Abstract
Communication between the periphery and the brain is key for maintaining energy homeostasis. To do so, peripheral signals from the circulation reach the brain via the circumventricular organs (CVOs), which are characterized by fenestrated vessels lacking the protective blood-brain barrier (BBB). Glial cells, by virtue of their plasticity and their ideal location at the interface of blood vessels and neurons, participate in the integration and transmission of peripheral information to neuronal networks in the brain for the neuroendocrine control of whole-body metabolism. Metabolic diseases, such as obesity and type 2 diabetes, can disrupt the brain-to-periphery communication mediated by glial cells, highlighting the relevance of these cell types in the pathophysiology of such complications. An improved understanding of how glial cells integrate and respond to metabolic and humoral signals has become a priority for the discovery of promising therapeutic strategies to treat metabolic disorders. This Review highlights the role of glial cells in the exchange of metabolic signals between the periphery and the brain that are relevant for the regulation of whole-body energy homeostasis.
Collapse
Affiliation(s)
- Sreekala Nampoothiri
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, Lille, France
| | - Ruben Nogueiras
- Universidade de Santiago de Compostela-Instituto de Investigation Sanitaria, Santiago de Compostela, Spain
- CIBER Fisiopatologia de la Obesidad y Nutrition, Santiago de Compostela, Spain
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, EGID, DISTALZ, Lille, France.
| |
Collapse
|
23
|
Xie Y, Ma Y, Cai L, Jiang S, Li C. Reconsidering Meat Intake and Human Health: A Review of Current Research. Mol Nutr Food Res 2022; 66:e2101066. [PMID: 35199948 DOI: 10.1002/mnfr.202101066] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 01/13/2022] [Indexed: 12/19/2022]
Abstract
Meat consumption is gradually increasing and its impact on health has attracted widespread attention, resulting in epidemiological studies proposing a reduction in meat and processed meat intake. This review briefly summarizes recent advances in understanding the effects of meat or processed meat on human health, as well as the underlying mechanisms. Meat consumption varies widely among individuals, populations, and regions, with higher consumption in developed countries than in developing countries. However, increasing meat consumption may not be the main cause of increasing incidence of chronic disease, since the development of chronic disease is a complex physiological process that involves many factors, including excessive total energy intake and changes in food digestion processes, gut microbiota composition, and liver metabolism. In comparison, unhealthy dietary habits and a sedentary lifestyle with decreasing energy expenditure are factors more worthy of reflection. Meat and meat products provide high-value protein and many key essential micronutrients. In short, as long as excessive intake and overprocessing of meats are avoided, meat remains an indispensable source of nutrition for human health.
Collapse
Affiliation(s)
- Yunting Xie
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MARA, Jiangsu Innovative Center of Meat Production, Processing and Quality Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yafang Ma
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MARA, Jiangsu Innovative Center of Meat Production, Processing and Quality Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Linlin Cai
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MARA, Jiangsu Innovative Center of Meat Production, Processing and Quality Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Shuai Jiang
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MARA, Jiangsu Innovative Center of Meat Production, Processing and Quality Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Chunbao Li
- Key Laboratory of Meat Processing and Quality Control, MOE, Key Laboratory of Meat Processing, MARA, Jiangsu Innovative Center of Meat Production, Processing and Quality Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| |
Collapse
|
24
|
Moore A, Chinnaiya K, Kim DW, Brown S, Stewart I, Robins S, Dowsett GKC, Muir C, Travaglio M, Lewis JE, Ebling F, Blackshaw S, Furley A, Placzek M. Loss of Function of the Neural Cell Adhesion Molecule NrCAM Regulates Differentiation, Proliferation and Neurogenesis in Early Postnatal Hypothalamic Tanycytes. Front Neurosci 2022; 16:832961. [PMID: 35464310 PMCID: PMC9022636 DOI: 10.3389/fnins.2022.832961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 01/27/2022] [Indexed: 11/13/2022] Open
Abstract
Hypothalamic tanycytes are neural stem and progenitor cells, but little is known of how they are regulated. Here we provide evidence that the cell adhesion molecule, NrCAM, regulates tanycytes in the adult niche. NrCAM is strongly expressed in adult mouse tanycytes. Immunohistochemical and in situ hybridization analysis revealed that NrCAM loss of function leads to both a reduced number of tanycytes and reduced expression of tanycyte-specific cell markers, along with a small reduction in tyrosine hydroxylase-positive arcuate neurons. Similar analyses of NrCAM mutants at E16 identify few changes in gene expression or cell composition, indicating that NrCAM regulates tanycytes, rather than early embryonic hypothalamic development. Neurosphere and organotypic assays support the idea that NrCAM governs cellular homeostasis. Single-cell RNA sequencing (scRNA-Seq) shows that tanycyte-specific genes, including a number that are implicated in thyroid hormone metabolism, show reduced expression in the mutant mouse. However, the mild tanycyte depletion and loss of markers observed in NrCAM-deficient mice were associated with only a subtle metabolic phenotype.
Collapse
Affiliation(s)
- Alex Moore
- School of Biosciences, The University of Sheffield, Sheffield, United Kingdom
- Bateson Centre, The University of Sheffield, Sheffield, United Kingdom
- Neuroscience Institute, The University of Sheffield, Sheffield, United Kingdom
| | - Kavitha Chinnaiya
- School of Biosciences, The University of Sheffield, Sheffield, United Kingdom
- Bateson Centre, The University of Sheffield, Sheffield, United Kingdom
- Neuroscience Institute, The University of Sheffield, Sheffield, United Kingdom
| | - Dong Won Kim
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sarah Brown
- School of Biosciences, The University of Sheffield, Sheffield, United Kingdom
- Bateson Centre, The University of Sheffield, Sheffield, United Kingdom
- Neuroscience Institute, The University of Sheffield, Sheffield, United Kingdom
| | - Iain Stewart
- School of Biosciences, The University of Sheffield, Sheffield, United Kingdom
- Bateson Centre, The University of Sheffield, Sheffield, United Kingdom
- Neuroscience Institute, The University of Sheffield, Sheffield, United Kingdom
| | - Sarah Robins
- School of Biosciences, The University of Sheffield, Sheffield, United Kingdom
- Bateson Centre, The University of Sheffield, Sheffield, United Kingdom
- Neuroscience Institute, The University of Sheffield, Sheffield, United Kingdom
| | - Georgina K. C. Dowsett
- Wellcome Trust-Medical Research Council Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, United Kingdom
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Charlotte Muir
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Marco Travaglio
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Jo E. Lewis
- Wellcome Trust-Medical Research Council Institute of Metabolic Science-Metabolic Research Laboratories, University of Cambridge, Cambridge, United Kingdom
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Fran Ebling
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Andrew Furley
- School of Biosciences, The University of Sheffield, Sheffield, United Kingdom
- Bateson Centre, The University of Sheffield, Sheffield, United Kingdom
- Neuroscience Institute, The University of Sheffield, Sheffield, United Kingdom
| | - Marysia Placzek
- School of Biosciences, The University of Sheffield, Sheffield, United Kingdom
- Bateson Centre, The University of Sheffield, Sheffield, United Kingdom
- Neuroscience Institute, The University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
25
|
Nguyen TT, Hulme J, Vo TK, Van Vo G. The Potential Crosstalk Between the Brain and Visceral Adipose Tissue in Alzheimer's Development. Neurochem Res 2022; 47:1503-1512. [PMID: 35298764 DOI: 10.1007/s11064-022-03569-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 01/25/2022] [Accepted: 02/28/2022] [Indexed: 11/30/2022]
Abstract
The bidirectional communication between the brain and peripheral organs have been widely documented, but the impact of visceral adipose tissue (VAT) dysfunction and its relation to structural and functional brain changes have yet to be fully elucidated. This review initially examines the clinical evidence supporting associations between the brain and VAT before visiting the roles of the autonomic nervous system, fat and glucose metabolism, neuroinflammation, and metabolites. Finally, the possible effects and potential mechanisms of the brain-VAT axis on the pathogenesis of Alzheimer's disease are discussed, providing new insights regarding future prevention and therapeutic strategies.
Collapse
Affiliation(s)
- Thuy Trang Nguyen
- Faculty of Pharmacy, HUTECH University, Ho Chi Minh City, 700000, Vietnam
| | - John Hulme
- Department of BioNano Technology, Gachon University, Seongnam, 461-701, Republic of Korea.
| | - Tuong Kha Vo
- Vietnam Sports Hospital, Ministry of Culture, Sports and Tourism, Hanoi, 100000, Vietnam.,Department of Sports Medicine, University of Medicine and Pharmacy (VNU-UMP), Vietnam National University Hanoi, Hanoi, 100000, Vietnam
| | - Giau Van Vo
- Department of Biomedical Engineering, School of Medicine, Vietnam National University Ho Chi Minh City (VNU-HCM), Ho Chi Minh City, 700000, Vietnam. .,Research Center for Genetics and Reproductive Health (CGRH), School of Medicine, Vietnam National University Ho Chi Minh City (VNU-HCM), Ho Chi Minh City, 700000, Vietnam. .,Vietnam National University Ho Chi Minh City (VNU-HCM), Ho Chi Minh City, 700000, Vietnam.
| |
Collapse
|
26
|
Jais A, Brüning JC. Arcuate Nucleus-Dependent Regulation of Metabolism-Pathways to Obesity and Diabetes Mellitus. Endocr Rev 2022; 43:314-328. [PMID: 34490882 PMCID: PMC8905335 DOI: 10.1210/endrev/bnab025] [Citation(s) in RCA: 115] [Impact Index Per Article: 38.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Indexed: 01/12/2023]
Abstract
The central nervous system (CNS) receives information from afferent neurons, circulating hormones, and absorbed nutrients and integrates this information to orchestrate the actions of the neuroendocrine and autonomic nervous systems in maintaining systemic metabolic homeostasis. Particularly the arcuate nucleus of the hypothalamus (ARC) is of pivotal importance for primary sensing of adiposity signals, such as leptin and insulin, and circulating nutrients, such as glucose. Importantly, energy state-sensing neurons in the ARC not only regulate feeding but at the same time control multiple physiological functions, such as glucose homeostasis, blood pressure, and innate immune responses. These findings have defined them as master regulators, which adapt integrative physiology to the energy state of the organism. The disruption of this fine-tuned control leads to an imbalance between energy intake and expenditure as well as deregulation of peripheral metabolism. Improving our understanding of the cellular, molecular, and functional basis of this regulatory principle in the CNS could set the stage for developing novel therapeutic strategies for the treatment of obesity and metabolic syndrome. In this review, we summarize novel insights with a particular emphasis on ARC neurocircuitries regulating food intake and glucose homeostasis and sensing factors that inform the brain of the organismal energy status.
Collapse
Affiliation(s)
- Alexander Jais
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany.,Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany.,Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - Jens C Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany.,Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany.,Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany.,National Center for Diabetes Research (DZD), Neuherberg, Germany
| |
Collapse
|
27
|
Abstract
The role of central estrogen in cognitive, metabolic, and reproductive health has long fascinated the lay public and scientists alike. In the last two decades, insight into estrogen signaling in the brain and its impact on female physiology is beginning to catch up with the vast information already established for its actions on peripheral tissues. Using newer methods to manipulate estrogen signaling in hormone-sensitive brain regions, neuroscientists are now identifying the molecular pathways and neuronal subtypes required for controlling sex-dependent energy allocation. However, the immense cellular complexity of these hormone-sensitive brain regions makes it clear that more research is needed to fully appreciate how estrogen modulates neural circuits to regulate physiological and behavioral end points. Such insight is essential for understanding how natural or drug-induced hormone fluctuations across lifespan affect women's health.
Collapse
Affiliation(s)
- Holly A Ingraham
- Department of Cellular and Molecular Pharmacology, School of Medicine, Mission Bay, University of California, San Francisco, California, USA;
| | - Candice B Herber
- Department of Cellular and Molecular Pharmacology, School of Medicine, Mission Bay, University of California, San Francisco, California, USA;
| | - William C Krause
- Department of Cellular and Molecular Pharmacology, School of Medicine, Mission Bay, University of California, San Francisco, California, USA;
| |
Collapse
|
28
|
Maffei M, Giordano A. Leptin, the brain and energy homeostasis: From an apparently simple to a highly complex neuronal system. Rev Endocr Metab Disord 2022; 23:87-101. [PMID: 33822303 DOI: 10.1007/s11154-021-09636-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/11/2021] [Indexed: 12/14/2022]
Abstract
Leptin, produced and secreted by white adipose tissue in tight relationship with adipose mass, informs the brain about the status of the energy stores serving as the main peripheral signal for energy balance regulation through interaction with a multitude of highly interconnected neuronal populations. Most obese patients display resistance to the anorectic effect of the hormone. The present review unravels the multiple levels of complexity that trigger hypothalamic response to leptin with the objective of highlighting those critical hubs that, mainly in the hypothalamic arcuate nucleus, may undergo obesity-induced alterations and create an obstacle to leptin action. Several mechanisms underlying leptin resistance have been proposed, possibly representing useful targets to empower leptin effects. Among these, a special focus is herein dedicated to detail how leptin gains access into the brain and how neuronal plasticity may interfere with leptin function.
Collapse
Affiliation(s)
- Margherita Maffei
- Institute of Clinical Physiology, CNR, Via Moruzzi 1, 56124, Pisa, Italy.
- Obesity and Lipodystrophy Center, University Hospital of Pisa, Via Paradisa 2, 56124, Pisa, Italy.
| | - Antonio Giordano
- Department of Experimental and Clinical Medicine, Marche Polytechnic University, Via Tronto 10/A, 60020, Ancona, Italy.
| |
Collapse
|
29
|
Vohra MS, Benchoula K, Serpell CJ, Hwa WE. AgRP/NPY and POMC neurons in the arcuate nucleus and their potential role in treatment of obesity. Eur J Pharmacol 2022; 915:174611. [PMID: 34798121 DOI: 10.1016/j.ejphar.2021.174611] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Revised: 10/27/2021] [Accepted: 10/27/2021] [Indexed: 02/08/2023]
Abstract
Obesity is a major health crisis affecting over a third of the global population. This multifactorial disease is regulated via interoceptive neural circuits in the brain, whose alteration results in excessive body weight. Certain central neuronal populations in the brain are recognised as crucial nodes in energy homeostasis; in particular, the hypothalamic arcuate nucleus (ARC) region contains two peptide microcircuits that control energy balance with antagonistic functions: agouti-related peptide/neuropeptide-Y (AgRP/NPY) signals hunger and stimulates food intake; and pro-opiomelanocortin (POMC) signals satiety and reduces food intake. These neuronal peptides levels react to energy status and integrate signals from peripheral ghrelin, leptin, and insulin to regulate feeding and energy expenditure. To manage obesity comprehensively, it is crucial to understand cellular and molecular mechanisms of information processing in ARC neurons, since these regulate energy homeostasis. Importantly, a specific strategy focusing on ARC circuits needs to be devised to assist in treating obese patients and maintaining weight loss with minimal or no side effects. The aim of this review is to elucidate the recent developments in the study of AgRP-, NPY- and POMC-producing neurons, specific to their role in controlling metabolism. The impact of ghrelin, leptin, and insulin signalling via action of these neurons is also surveyed, since they also impact energy balance through this route. Lastly, we present key proteins, targeted genes, compounds, drugs, and therapies that actively work via these neurons and could potentially be used as therapeutic targets for treating obesity conditions.
Collapse
Affiliation(s)
- Muhammad Sufyan Vohra
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Khaled Benchoula
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia
| | - Christopher J Serpell
- School of Physical Sciences, Ingram Building, University of Kent, Canterbury, Kent, CT2 7NH, United Kingdom
| | - Wong Eng Hwa
- School of Medicine, Faculty of Health and Medical Sciences, Taylor's University Lakeside Campus, 47500, Subang Jaya, Selangor Darul Ehsan, Malaysia.
| |
Collapse
|
30
|
Rodríguez-Cortés B, Hurtado-Alvarado G, Martínez-Gómez R, León-Mercado LA, Prager-Khoutorsky M, Buijs RM. Suprachiasmatic nucleus-mediated glucose entry into the arcuate nucleus determines the daily rhythm in blood glycemia. Curr Biol 2022; 32:796-805.e4. [PMID: 35030330 DOI: 10.1016/j.cub.2021.12.039] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/19/2021] [Accepted: 12/16/2021] [Indexed: 12/20/2022]
Abstract
Glycemia is maintained within very narrow boundaries with less than 5% variation at a given time of the day. However, over the circadian cycle, glycemia changes with almost 50% difference. How the suprachiasmatic nucleus, the biological clock, maintains these day-night variations with such tiny disparities remains obscure. We show that via vasopressin release at the beginning of the sleep phase, the suprachiasmatic nucleus increases the glucose transporter GLUT1 in tanycytes. Hereby GLUT1 promotes glucose entrance into the arcuate nucleus, thereby lowering peripheral glycemia. Conversely, blocking vasopressin activity or the GLUT1 transporter at the daily trough of glycemia increases circulating glucose levels usually seen at the peak of the rhythm. Thus, biological clock-controlled mechanisms promoting glucose entry into the arcuate nucleus explain why peripheral blood glucose is low before sleep onset.
Collapse
Affiliation(s)
- Betty Rodríguez-Cortés
- Department of Cellular Biology and Physiology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mario de la Cueva Circuit, Mexico City 04510, Mexico
| | - Gabriela Hurtado-Alvarado
- Department of Cellular Biology and Physiology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mario de la Cueva Circuit, Mexico City 04510, Mexico
| | - Ricardo Martínez-Gómez
- Department of Cellular Biology and Physiology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mario de la Cueva Circuit, Mexico City 04510, Mexico
| | - Luis A León-Mercado
- Department of Internal Medicine, Center for Hypothalamic Research, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | - Masha Prager-Khoutorsky
- Department of Physiology, McIntyre Medical Sciences Building, McGill University, 3655 Promenade Sir-William-Osler, Montréal, QC H3G 1Y6, Canada
| | - Ruud M Buijs
- Department of Cellular Biology and Physiology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Mario de la Cueva Circuit, Mexico City 04510, Mexico.
| |
Collapse
|
31
|
Butruille L, Batailler M, Cateau ML, Sharif A, Leysen V, Prévot V, Vaudin P, Pillon D, Migaud M. Selective Depletion of Adult GFAP-Expressing Tanycytes Leads to Hypogonadotropic Hypogonadism in Males. Front Endocrinol (Lausanne) 2022; 13:869019. [PMID: 35370973 PMCID: PMC8966543 DOI: 10.3389/fendo.2022.869019] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 02/21/2022] [Indexed: 01/22/2023] Open
Abstract
In adult mammals, neural stem cells are localized in three neurogenic regions, the subventricular zone of the lateral ventricle (SVZ), the subgranular zone of the dentate gyrus of the hippocampus (SGZ) and the hypothalamus. In the SVZ and the SGZ, neural stem/progenitor cells (NSPCs) express the glial fibrillary acidic protein (GFAP) and selective depletion of these NSPCs drastically decreases cell proliferation in vitro and in vivo. In the hypothalamus, GFAP is expressed by α-tanycytes, which are specialized radial glia-like cells in the wall of the third ventricle also recognized as NSPCs. To explore the role of these hypothalamic GFAP-positive tanycytes, we used transgenic mice expressing herpes simplex virus thymidine kinase (HSV-Tk) under the control of the mouse Gfap promoter and a 4-week intracerebroventricular infusion of the antiviral agent ganciclovir (GCV) which kills dividing cells expressing Tk. While GCV significantly reduced the number and growth of hypothalamus-derived neurospheres from adult transgenic mice in vitro, it causes hypogonadotropic hypogonadism in vivo. The selective death of dividing tanycytes expressing GFAP indeed results in a marked decrease in testosterone levels and testicular weight, as well as vacuolization of the seminiferous tubules and loss of spermatogenesis. Additionally, GCV-treated GFAP-Tk mice show impaired sexual behavior, but no alteration in food intake or body weight. Our results also show that the selective depletion of GFAP-expressing tanycytes leads to a sharp decrease in the number of gonadotropin-releasing hormone (GnRH)-immunoreactive neurons and a blunted LH secretion. Overall, our data show that GFAP-expressing tanycytes play a central role in the regulation of male reproductive function.
Collapse
Affiliation(s)
| | | | | | - Ariane Sharif
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neurosciences & Cognition, UMR-S1172, Lille, France
| | - Valérie Leysen
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neurosciences & Cognition, UMR-S1172, Lille, France
| | - Vincent Prévot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neurosciences & Cognition, UMR-S1172, Lille, France
| | - Pascal Vaudin
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| | - Delphine Pillon
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| | - Martine Migaud
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
- *Correspondence: Martine Migaud,
| |
Collapse
|
32
|
Clayton RW, Lovell-Badge R, Galichet C. The Properties and Functions of Glial Cell Types of the Hypothalamic Median Eminence. Front Endocrinol (Lausanne) 2022; 13:953995. [PMID: 35966104 PMCID: PMC9363565 DOI: 10.3389/fendo.2022.953995] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 06/17/2022] [Indexed: 02/06/2023] Open
Abstract
The median eminence (ME) is part of the neuroendocrine system (NES) that functions as a crucial interface between the hypothalamus and pituitary gland. The ME contains many non-neuronal cell types, including oligodendrocytes, oligodendrocyte precursor cells (OPCs), tanycytes, astrocytes, pericytes, microglia and other immune cells, which may be involved in the regulation of NES function. For example, in mice, ablation of tanycytes (a special class of ependymal glia with stem cell-like functions) results in weight gain, feeding, insulin insensitivity and increased visceral adipose, consistent with the demonstrated ability of these cells to sense and transport both glucose and leptin, and to differentiate into neurons that control feeding and metabolism in the hypothalamus. To give a further example, OPCs in the ME of mice have been shown to rapidly respond to dietary signals, in turn controlling composition of the extracellular matrix in the ME, derived from oligodendrocyte-lineage cells, which may contribute to the previously described role of these cells in actively maintaining leptin-receptor-expressing dendrites in the ME. In this review, we explore and discuss recent advances such as these, that have developed our understanding of how the various cell types of the ME contribute to its function in the NES as the interface between the hypothalamus and pituitary gland. We also highlight avenues of future research which promise to uncover additional functions of the ME and the glia, stem and progenitor cells it contains.
Collapse
|
33
|
Porniece Kumar M, Cremer AL, Klemm P, Steuernagel L, Sundaram S, Jais A, Hausen AC, Tao J, Secher A, Pedersen TÅ, Schwaninger M, Wunderlich FT, Lowell BB, Backes H, Brüning JC. Insulin signalling in tanycytes gates hypothalamic insulin uptake and regulation of AgRP neuron activity. Nat Metab 2021; 3:1662-1679. [PMID: 34931084 PMCID: PMC8688146 DOI: 10.1038/s42255-021-00499-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 10/26/2021] [Indexed: 12/02/2022]
Abstract
Insulin acts on neurons and glial cells to regulate systemic glucose metabolism and feeding. However, the mechanisms of insulin access in discrete brain regions are incompletely defined. Here we show that insulin receptors in tanycytes, but not in brain endothelial cells, are required to regulate insulin access to the hypothalamic arcuate nucleus. Mice lacking insulin receptors in tanycytes (IR∆Tan mice) exhibit systemic insulin resistance, while displaying normal food intake and energy expenditure. Tanycytic insulin receptors are also necessary for the orexigenic effects of ghrelin, but not for the anorexic effects of leptin. IR∆Tan mice exhibit increased agouti-related peptide (AgRP) neuronal activity, while displaying blunted AgRP neuronal adaptations to feeding-related stimuli. Lastly, a highly palatable food decreases tanycytic and arcuate nucleus insulin signalling to levels comparable to those seen in IR∆Tan mice. These changes are rooted in modifications of cellular stress responses and of mitochondrial protein quality control in tanycytes. Conclusively, we reveal a critical role of tanycyte insulin receptors in gating feeding-state-dependent regulation of AgRP neurons and systemic insulin sensitivity, and show that insulin resistance in tanycytes contributes to the pleiotropic manifestations of obesity-associated insulin resistance.
Collapse
Affiliation(s)
- Marta Porniece Kumar
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Anna Lena Cremer
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Paul Klemm
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Lukas Steuernagel
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Sivaraj Sundaram
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Alexander Jais
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
- Helmholtz Institute for Metabolic, Obesity and Vascular Research (HI-MAG) of the Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - A Christine Hausen
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Jenkang Tao
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Program in Neuroscience, Harvard Medical School, Boston, MA, USA
| | - Anna Secher
- Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
| | | | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - F Thomas Wunderlich
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Bradford B Lowell
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Program in Neuroscience, Harvard Medical School, Boston, MA, USA
| | - Heiko Backes
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Jens C Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Cologne, Germany.
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Cologne, Germany.
- National Center for Diabetes Research (DZD), Neuherberg, Germany.
| |
Collapse
|
34
|
Dou Z, Son JE, Hui CC. Irx3 and Irx5 - Novel Regulatory Factors of Postnatal Hypothalamic Neurogenesis. Front Neurosci 2021; 15:763856. [PMID: 34795556 PMCID: PMC8593166 DOI: 10.3389/fnins.2021.763856] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/07/2021] [Indexed: 12/27/2022] Open
Abstract
The hypothalamus is a brain region that exhibits highly conserved anatomy across vertebrate species and functions as a central regulatory hub for many physiological processes such as energy homeostasis and circadian rhythm. Neurons in the arcuate nucleus of the hypothalamus are largely responsible for sensing of peripheral signals such as leptin and insulin, and are critical for the regulation of food intake and energy expenditure. While these neurons are mainly born during embryogenesis, accumulating evidence have demonstrated that neurogenesis also occurs in postnatal-adult mouse hypothalamus, particularly in the first two postnatal weeks. This second wave of active neurogenesis contributes to the remodeling of hypothalamic neuronal populations and regulation of energy homeostasis including hypothalamic leptin sensing. Radial glia cell types, such as tanycytes, are known to act as neuronal progenitors in the postnatal mouse hypothalamus. Our recent study unveiled a previously unreported radial glia-like neural stem cell (RGL-NSC) population that actively contributes to neurogenesis in the postnatal mouse hypothalamus. We also identified Irx3 and Irx5, which encode Iroquois homeodomain-containing transcription factors, as genetic determinants regulating the neurogenic property of these RGL-NSCs. These findings are significant as IRX3 and IRX5 have been implicated in FTO-associated obesity in humans, illustrating the importance of postnatal hypothalamic neurogenesis in energy homeostasis and obesity. In this review, we summarize current knowledge regarding postnatal-adult hypothalamic neurogenesis and highlight recent findings on the radial glia-like cells that contribute to the remodeling of postnatal mouse hypothalamus. We will discuss characteristics of the RGL-NSCs and potential actions of Irx3 and Irx5 in the regulation of neural stem cells in the postnatal-adult mouse brain. Understanding the behavior and regulation of neural stem cells in the postnatal-adult hypothalamus will provide novel mechanistic insights in the control of hypothalamic remodeling and energy homeostasis.
Collapse
Affiliation(s)
- Zhengchao Dou
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Joe Eun Son
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Chi-chung Hui
- Program in Developmental & Stem Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
35
|
Surbhi, Wittmann G, Low MJ, Lechan RM. Adult-born proopiomelanocortin neurons derived from Rax-expressing precursors mitigate the metabolic effects of congenital hypothalamic proopiomelanocortin deficiency. Mol Metab 2021; 53:101312. [PMID: 34329773 PMCID: PMC8383116 DOI: 10.1016/j.molmet.2021.101312] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 07/12/2021] [Accepted: 07/25/2021] [Indexed: 12/19/2022] Open
Abstract
OBJECTIVE Proopiomelanocortin (POMC) neurons of the hypothalamic arcuate nucleus are essential regulators of energy balance. Selective loss of POMC production in these cells results in extreme obesity and metabolic comorbidities. Neurogenesis occurs in the adult hypothalamus, but it remains uncertain whether functional POMC neurons emerge in physiologically significant numbers during adulthood. Here, we tested whether Rax-expressing precursors generate POMC neurons in adult mice and rescue the metabolic phenotype caused by congenital hypothalamic POMC deficiency. METHODS Initially, we identified hypothalamic Rax-expressing cell types using wild-type and Rax-CreERT2:Ai34D mice. Then we generated compound Rax-CreERT2:ArcPomcloxTB/loxTB mice in which endogenous hypothalamic Pomc expression is silenced, but can be restored by tamoxifen administration selectively in neurons derived from Rax+ progenitors. The number of POMC neurons generated by Rax+ progenitors in adult mice and their axonal projections was determined. The metabolic effects of these neurons were assessed by measuring food intake, bodyweight, and body composition, along with glucose and insulin levels. RESULTS We found that Rax is expressed by tanycytes and a previously unrecognized cell type in the hypothalamic parenchyma of adult mice. Rax+ progenitors generated ~10% of the normal adult hypothalamic POMC neuron population within two weeks of tamoxifen treatment. The same rate and steady state of POMC neurogenesis persisted from young adult to aged mice. These new POMC neurons established terminal projections to brain regions that were involved in energy homeostasis. Mice with Rax+ progenitor-derived POMC neurons had reduced body fat mass, improved glucose tolerance, increased insulin sensitivity, and decreased bodyweight in proportion to the number of new POMC neurons. CONCLUSIONS These data demonstrate that Rax+ progenitors generate POMC neurons in sufficient numbers during adulthood to mitigate the metabolic abnormalities of hypothalamic POMC-deficient mice. The findings suggest that adult hypothalamic neurogenesis is a robust phenomenon in mice that can significantly impact energy homeostasis.
Collapse
Affiliation(s)
- Surbhi
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, 2800 Plymouth Rd, Ann Arbor, MI, 48109, USA.
| | - Gábor Wittmann
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Tufts Medical Center, 800 Washington St, Boston, MA, 02111, USA.
| | - Malcolm J Low
- Department of Molecular & Integrative Physiology, University of Michigan Medical School, 2800 Plymouth Rd, Ann Arbor, MI, 48109, USA.
| | - Ronald M Lechan
- Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Tufts Medical Center, 800 Washington St, Boston, MA, 02111, USA.
| |
Collapse
|
36
|
Voronkov DN, Stavrovskaya AV, Gushchina AS, Olshansky AS. Alterations in tanycytes and related cell populations of arcuate nucleus in streptozotocin-induced Alzheimer disease model. BULLETIN OF RUSSIAN STATE MEDICAL UNIVERSITY 2021. [DOI: 10.24075/brsmu.2021.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
It is assumed that dysfunction of tanycytes could be one of the components of pathogenesis of both Alzheimer disease and type 2 diabetes mellitus. The study was aimed to assess alterations in the tanycyte morphology in the Alzheimer disease model. The 3 mg/kg streptozotocin dose was injected in the lateral ventricles of Wistar rats in order to model the Alzheimer disease. Alterations in hypothalamic tanycytes were assessed 2 weeks, 4 weeks, 3 months and 6 months after administration of the toxin. Immunohistochemistry was used to identify the protein markers of tanycytes (vimentin, nestin), astrocytes (GFAP, glutamine synthetase) and neurons (HuC/D), as well as to assess cell proliferation (with the use of Ki67 protein) and mitochondrial alterations (mitochondrial complex IV, PGC1a). Administration of streptozotocin lead to β-amyloid accumulation in hypothalamus and ventricular enlargement (p < 0.001). Streptozotocin damaged both α1/α2 tanycytes and β1 tanycytes. The intensity of vimentin staining in α1/α2 tanycytes decreased by week 4 (p = 0.003), and in β1 tanycytes it decreased in three months (p < 0.001). The same trend was observed for nestin. The number of Ki67+ nuclei decreased (p < 0.05), and the expression of proteins associated with mitochondria changed. The density of hypothalamic tanycytes decreased by week 4 after administration of the toxin. Moreover, astrocyte activation was revealed. However, no prominent damage to both astrocytes and neurons was observed within four weeks after administration of streptozotocin. The revealed high tanycyte vulnerability to streptozotocin is in line with the hypothesis of the role of damage to hypothalamic structures in both local and systemic metabolic disorders occurring in the Alzheimer disease models.
Collapse
Affiliation(s)
- DN Voronkov
- Research Center of Neurology, Moscow, Russia
| | | | | | | |
Collapse
|
37
|
Abstract
Leptin for over 25 years has been a central theme in the study of appetite, obesity, and starvation. As the major site of leptin production is peripheral, and the site of action of greatest interest is the hypothalamus, how leptin accesses the central nervous system (CNS) and crosses the blood-brain barrier (BBB) has been of great interest. We review here the ongoing research that addresses fundamental questions such as the sites of leptin resistances in obesity and other conditions, the causes of resistances and their relations to one another, the three barrier sites of entry into the CNS, why recent studies using suprapharmacological doses cannot address these questions but give insight into nonsaturable entry of leptin into the CNS, and how that might be useful in using leptin therapeutically. The current status of the controversy of whether the short form of the leptin receptor acts as the BBB leptin transporter and how obesity may transform leptin transport is reviewed. Review of these and other topics summarizes in a new appreciation of what leptin may have actually evolved to do and what physiological role leptin resistance may play. © 2021 American Physiological Society. Compr Physiol 11:1-19, 2021.
Collapse
Affiliation(s)
- William A Banks
- Geriatric Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, Washington, USA.,Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, Washington, USA
| |
Collapse
|
38
|
Wang J, Beecher K. TSPO: an emerging role in appetite for a therapeutically promising biomarker. Open Biol 2021; 11:210173. [PMID: 34343461 PMCID: PMC8331234 DOI: 10.1098/rsob.210173] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
There is accumulating evidence that an obesogenic Western diet causes neuroinflammatory damage to the brain, which then promotes further appetitive behaviour. Neuroinflammation has been extensively studied by analysing the translocator protein of 18 kDa (TSPO), a protein that is upregulated in the inflamed brain following a damaging stimulus. As a result, there is a rich supply of TSPO-specific agonists, antagonists and positron emission tomography ligands. One TSPO ligand, etifoxine, is also currently used clinically for the treatment of anxiety with a minimal side-effect profile. Despite the neuroinflammatory pathogenesis of diet-induced obesity, and the translational potential of targeting TSPO, there is sparse literature characterizing the effect of TSPO on appetite. Therefore, in this review, the influence of TSPO on appetite is discussed. Three putative mechanisms for TSPO's appetite-modulatory effect are then characterized: the TSPO–allopregnanolone–GABAAR signalling axis, glucosensing in tanycytes and association with the synaptic protein RIM-BP1. We highlight that, in addition to its plethora of functions, TSPO is a regulator of appetite. This review ultimately suggests that the appetite-modulating function of TSPO should be further explored due to its potential therapeutic promise.
Collapse
Affiliation(s)
- Joshua Wang
- Addiction Neuroscience and Obesity Laboratory, School of Clinical Sciences, Faculty of Health, Translational Research Institute, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Kate Beecher
- Addiction Neuroscience and Obesity Laboratory, School of Clinical Sciences, Faculty of Health, Translational Research Institute, Queensland University of Technology, Brisbane, Queensland, Australia
| |
Collapse
|
39
|
High-Fat Diet Impairs Mouse Median Eminence: A Study by Transmission and Scanning Electron Microscopy Coupled with Raman Spectroscopy. Int J Mol Sci 2021; 22:ijms22158049. [PMID: 34360816 PMCID: PMC8347199 DOI: 10.3390/ijms22158049] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 12/28/2022] Open
Abstract
Hypothalamic dysfunction is an initial event following diet-induced obesity, primarily involving areas regulating energy balance such as arcuate nucleus (Arc) and median eminence (ME). To gain insights into the early hypothalamic diet-induced alterations, adult CD1 mice fed a high-fat diet (HFD) for 6 weeks were studied and compared with normo-fed controls. Transmission and scanning electron microscopy and histological staining were employed for morphological studies of the ME, while Raman spectroscopy was applied for the biochemical analysis of the Arc-ME complex. In HFD mice, ME β2-tanycytes, glial cells dedicated to blood-liquor crosstalk, exhibited remarkable ultrastructural anomalies, including altered alignment, reduced junctions, degenerating organelles, and higher content of lipid droplets, lysosomes, and autophagosomes. Degenerating tanycytes also displayed an electron transparent cytoplasm filled with numerous vesicles, and they were surrounded by dilated extracellular spaces extending up to the subependymal layer. Consistently, Raman spectroscopy analysis of the Arc-ME complex revealed higher glycogen, collagen, and lipid bands in HFD mice compared with controls, and there was also a higher band corresponding to the cyanide group in the former compared to the last. Collectively, these data show that ME β2-tanycytes exhibit early structural and chemical alterations due to HFD and reveal for the first-time hypothalamic cyanide presence following high dietary lipids consumption, which is a novel aspect with potential implications in the field of obesity.
Collapse
|
40
|
Rohrbach A, Caron E, Dali R, Brunner M, Pasquettaz R, Kolotuev I, Santoni F, Thorens B, Langlet F. Ablation of glucokinase-expressing tanycytes impacts energy balance and increases adiposity in mice. Mol Metab 2021; 53:101311. [PMID: 34325016 PMCID: PMC8379510 DOI: 10.1016/j.molmet.2021.101311] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 07/20/2021] [Indexed: 01/06/2023] Open
Abstract
Objectives Glucokinase (GCK) is critical for glucosensing. In rats, GCK is expressed in hypothalamic tanycytes and appears to play an essential role in feeding behavior. In this study, we investigated the distribution of GCK-expressing tanycytes in mice and their role in the regulation of energy balance. Methods In situ hybridization, reporter gene assay, and immunohistochemistry were used to assess GCK expression along the third ventricle in mice. To evaluate the impact of GCK-expressing tanycytes on arcuate neuron function and mouse physiology, Gck deletion along the ventricle was achieved using loxP/Cre recombinase technology in adult mice. Results GCK expression was low along the third ventricle, but detectable in tanycytes facing the ventromedial arcuate nucleus from bregma −1.5 to −2.2. Gck deletion induced the death of this tanycyte subgroup through the activation of the BAD signaling pathway. The ablation of GCK-expressing tanycytes affected different aspects of energy balance, leading to an increase in adiposity in mice. This phenotype was systematically associated with a defect in NPY neuron function. In contrast, the regulation of glucose homeostasis was mostly preserved, except for glucoprivic responses. Conclusions This study describes the role of GCK in tanycyte biology and highlights the impact of tanycyte loss on the regulation of energy balance. vmARH tanycytes express glucokinase. Glucokinase deletion in tanycytes induces cell death. Ablation of vmARH tanycytes alters energy balance and adiposity. Ablation of vmARH tanycytes alters NPY neuron function.
Collapse
Affiliation(s)
- Antoine Rohrbach
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland; Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Emilie Caron
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S, 1172, Lille, France
| | - Rafik Dali
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Maxime Brunner
- Service of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, Faculty of Biology and Medicine, 1011, Lausanne, Switzerland
| | - Roxane Pasquettaz
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Irina Kolotuev
- Electron Microscopy Facility, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Federico Santoni
- Service of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, Faculty of Biology and Medicine, 1011, Lausanne, Switzerland
| | - Bernard Thorens
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Fanny Langlet
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland; Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
41
|
Salgado M, García-Robles MÁ, Sáez JC. Purinergic signaling in tanycytes and its contribution to nutritional sensing. Purinergic Signal 2021; 17:607-618. [PMID: 34018139 DOI: 10.1007/s11302-021-09791-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 04/19/2021] [Indexed: 10/21/2022] Open
Abstract
Tanycytes are hypothalamic radial glial-like cells with an important role in the regulation of neuroendocrine axes and energy homeostasis. These cells have been implicated in glucose, amino acids, and fatty acid sensing in the hypothalamus of rodents, where they are strategically positioned. While their cell bodies contact the cerebrospinal fluid, their extensive processes contact neurons of the arcuate and ventromedial nuclei, protagonists in the regulation of food intake. A growing body of evidence has shown that purinergic signaling plays a relevant role in this homeostatic role of tanycytes, likely regulating the release of gliotransmitters that will modify the activity of satiety-controlling hypothalamic neurons. Connexin hemichannels have proven to be particularly relevant in these mechanisms since they are responsible for the release of ATP from tanycytes in response to nutritional signals. On the other hand, either ionotropic or metabotropic ATP receptors are involved in the generation of intracellular Ca2+ waves in response to hypothalamic nutrients, which can spread between glial cells and towards neighboring neurons. This review will summarize recent evidence that supports a nutrient sensor role for tanycytes, highlighting the participation of purinergic signaling in this process.
Collapse
Affiliation(s)
- Magdiel Salgado
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile.,Instituto de Neurociencias, Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - María Á García-Robles
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile.
| | - Juan C Sáez
- Instituto de Neurociencias, Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile.
| |
Collapse
|
42
|
Microglia-Neuron Crosstalk in Obesity: Melodious Interaction or Kiss of Death? Int J Mol Sci 2021; 22:ijms22105243. [PMID: 34063496 PMCID: PMC8155827 DOI: 10.3390/ijms22105243] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/11/2021] [Accepted: 05/13/2021] [Indexed: 12/11/2022] Open
Abstract
Diet-induced obesity can originate from the dysregulated activity of hypothalamic neuronal circuits, which are critical for the regulation of body weight and food intake. The exact mechanisms underlying such neuronal defects are not yet fully understood, but a maladaptive cross-talk between neurons and surrounding microglial is likely to be a contributing factor. Functional and anatomical connections between microglia and hypothalamic neuronal cells are at the core of how the brain orchestrates changes in the body's metabolic needs. However, such a melodious interaction may become maladaptive in response to prolonged diet-induced metabolic stress, thereby causing overfeeding, body weight gain, and systemic metabolic perturbations. From this perspective, we critically discuss emerging molecular and cellular underpinnings of microglia-neuron communication in the hypothalamic neuronal circuits implicated in energy balance regulation. We explore whether changes in this intercellular dialogue induced by metabolic stress may serve as a protective neuronal mechanism or contribute to disease establishment and progression. Our analysis provides a framework for future mechanistic studies that will facilitate progress into both the etiology and treatments of metabolic disorders.
Collapse
|
43
|
Abstract
Chronic metabolic diseases, including diabetes and obesity, have become a major global health threat of the twenty-first century. Maintaining glucose homeostasis is essential for survival in mammals. Complex and highly coordinated interactions between glucose-sensing mechanisms and multiple effector systems are essential for controlling glucose levels in the blood. The central nervous system (CNS) plays a crucial role in regulating glucose homeostasis. Growing evidence indicates that disruption of glucose sensing in selective CNS areas, such as the hypothalamus, is closely interlinked with the pathogenesis of obesity and type 2 diabetes mellitus. However, the underlying intracellular mechanisms of glucose sensing in the hypothalamus remain elusive. Here, we review the current literature on hypothalamic glucose-sensing mechanisms and discuss the impact of alterations of these mechanisms on the pathogenesis of diabetes.
Collapse
Affiliation(s)
- Nal Ae Yoon
- Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
- Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA
| | - Sabrina Diano
- Institute of Human Nutrition, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA.
- Department of Molecular Pharmacology and Therapeutics, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
44
|
Yoo S, Kim J, Lyu P, Hoang TV, Ma A, Trinh V, Dai W, Jiang L, Leavey P, Duncan L, Won JK, Park SH, Qian J, Brown SP, Blackshaw S. Control of neurogenic competence in mammalian hypothalamic tanycytes. SCIENCE ADVANCES 2021; 7:eabg3777. [PMID: 34049878 PMCID: PMC8163082 DOI: 10.1126/sciadv.abg3777] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 04/09/2021] [Indexed: 05/07/2023]
Abstract
Hypothalamic tanycytes, radial glial cells that share many features with neuronal progenitors, can generate small numbers of neurons in the postnatal hypothalamus, but the identity of these neurons and the molecular mechanisms that control tanycyte-derived neurogenesis are unknown. In this study, we show that tanycyte-specific disruption of the NFI family of transcription factors (Nfia/b/x) robustly stimulates tanycyte proliferation and tanycyte-derived neurogenesis. Single-cell RNA sequencing (scRNA-seq) and single-cell assay for transposase-accessible chromatin sequencing (scATAC-seq) analysis reveals that NFI (nuclear factor I) factors repress Sonic hedgehog (Shh) and Wnt signaling in tanycytes and modulation of these pathways blocks proliferation and tanycyte-derived neurogenesis in Nfia/b/x-deficient mice. Nfia/b/x-deficient tanycytes give rise to multiple mediobasal hypothalamic neuronal subtypes that can mature, fire action potentials, receive synaptic inputs, and selectively respond to changes in internal states. These findings identify molecular mechanisms that control tanycyte-derived neurogenesis, which can potentially be targeted to selectively remodel the hypothalamic neural circuitry that controls homeostatic physiological processes.
Collapse
Affiliation(s)
- Sooyeon Yoo
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
- Department of Pathology, Seoul National University Hospital, 71 Daehak-ro, Jongno-gu 03082, Republic of Korea
| | - Juhyun Kim
- Department of Psychiatry and Behavioral Science, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Pin Lyu
- Department of Ophthalmology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Thanh V Hoang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Alex Ma
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Vickie Trinh
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Weina Dai
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Lizhi Jiang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Patrick Leavey
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Leighton Duncan
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jae-Kyung Won
- Department of Pathology, Seoul National University Hospital, 71 Daehak-ro, Jongno-gu 03082, Republic of Korea
| | - Sung-Hye Park
- Department of Pathology, Seoul National University Hospital, 71 Daehak-ro, Jongno-gu 03082, Republic of Korea
| | - Jiang Qian
- Department of Ophthalmology, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Solange P Brown
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21205, USA.
- Department of Ophthalmology, Johns Hopkins University, Baltimore, MD 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- Department of Neurology, Johns Hopkins University, Baltimore, MD 21205, USA
- Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| |
Collapse
|
45
|
Bolborea M, Langlet F. What is the physiological role of hypothalamic tanycytes in metabolism? Am J Physiol Regul Integr Comp Physiol 2021; 320:R994-R1003. [PMID: 33826442 DOI: 10.1152/ajpregu.00296.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In vertebrates, the energy balance process is tightly controlled by complex neural circuits that sense metabolic signals and adjust food intake and energy expenditure in line with the physiological requirements of optimal conditions. Within neural networks controlling energy balance, tanycytes are peculiar ependymoglial cells that are nowadays recognized as multifunctional players in the metabolic hypothalamus. However, the physiological function of hypothalamic tanycytes remains unclear, creating a number of ambiguities in the field. Here, we review data accumulated over the years that demonstrate the physiological function of tanycytes in the maintenance of metabolic homeostasis, opening up new research avenues. The presumed involvement of tanycytes in the pathophysiology of metabolic disorders and age-related neurodegenerative diseases will be finally discussed.
Collapse
Affiliation(s)
- Matei Bolborea
- Central and Peripheral Mechanisms of Neurodegeneration, INSERM U1118, Université de Strasbourg, Strasbourg, France.,School of Life Sciences, University of Warwick, Coventry, United Kingdom
| | - Fanny Langlet
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
46
|
Butruille L, Vancamp P, Demeneix BA, Remaud S. Thyroid hormone regulation of adult neural stem cell fate: A comparative analysis between rodents and primates. VITAMINS AND HORMONES 2021; 116:133-192. [PMID: 33752817 DOI: 10.1016/bs.vh.2021.02.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Thyroid hormone (TH) signaling, a highly conserved pathway across vertebrates, is crucial for brain development and function throughout life. In the adult mammalian brain, including that of humans, multipotent neural stem cells (NSCs) proliferate and generate neuronal and glial progenitors. The role of TH has been intensively investigated in the two main neurogenic niches of the adult mouse brain, the subventricular and the subgranular zone. A key finding is that T3, the biologically active form of THs, promotes NSC commitment toward a neuronal fate. In this review, we first discuss the roles of THs in the regulation of adult rodent neurogenesis, as well as how it relates to functional behavior, notably olfaction and cognition. Most research uncovering these roles of TH in adult neurogenesis was conducted in rodents, whose genetic background, brain structure and rate of neurogenesis are considerably different from that of humans. To bridge the phylogenetic gap, we also explore the similarities and divergences of TH-dependent adult neurogenesis in non-human primate models. Lastly, we examine how photoperiodic length changes TH homeostasis, and how that might affect adult neurogenesis in seasonal species to increase fitness. Several aspects by which TH acts on adult NSCs seem to be conserved among mammals, while we only start to uncover the molecular pathways, as well as how other in- and extrinsic factors are intertwined. A multispecies approach delivering more insights in the matter will pave the way for novel NSC-based therapies to combat neurological disorders.
Collapse
Affiliation(s)
- Lucile Butruille
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, Paris, France
| | - Pieter Vancamp
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, Paris, France
| | - Barbara A Demeneix
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, Paris, France
| | - Sylvie Remaud
- UMR 7221 Phyma, CNRS/Muséum National d'Histoire Naturelle, Paris, France.
| |
Collapse
|
47
|
Dardente H, Migaud M. Thyroid hormone and hypothalamic stem cells in seasonal functions. VITAMINS AND HORMONES 2021; 116:91-131. [PMID: 33752829 DOI: 10.1016/bs.vh.2021.02.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Seasonal rhythms are a pervasive feature of most living organisms, which underlie yearly timeliness in breeding, migration, hibernation or weight gain and loss. To achieve this, organisms have developed inner timing devices (circannual clocks) that endow them with the ability to predict then anticipate changes to come, usually using daylength as the proximate cue. In Vertebrates, daylength interpretation involves photoperiodic control of TSH production by the pars tuberalis (PT) of the pituitary, which governs a seasonal switch in thyroid hormone (TH) availability in the neighboring hypothalamus. Tanycytes, specialized glial cells lining the third ventricle (3V), are responsible for this TH output through the opposite, PT-TSH-driven, seasonal control of deiodinases 2/3 (Dio 2/3). Tanycytes comprise a photoperiod-sensitive stem cell niche and TH is known to play major roles in cell proliferation and differentiation, which suggests that seasonal control of tanycyte proliferation may be involved in the photoperiodic synchronization of seasonal rhythms. Here we review our current knowledge of the molecular and neuroendocrine pathway linking photoperiodic information to seasonal changes in physiological functions and discuss the potential implication of tanycytes, TH and cell proliferation in seasonal timing.
Collapse
Affiliation(s)
- Hugues Dardente
- PRC, INRAE, CNRS, IFCE, Université de Tours, Nouzilly, France.
| | - Martine Migaud
- PRC, INRAE, CNRS, IFCE, Université de Tours, Nouzilly, France
| |
Collapse
|
48
|
Bielefeld P, Abbink MR, Davidson AR, Reijner N, Abiega O, Lucassen PJ, Korosi A, Fitzsimons CP. Early life stress decreases cell proliferation and the number of putative adult neural stem cells in the adult hypothalamus. Stress 2021; 24:189-195. [PMID: 33494651 DOI: 10.1080/10253890.2021.1879787] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Stress is a potent environmental factor that can confer potent and enduring effects on brain structure and function. Exposure to stress during early life (ELS) has been linked to a wide range of consequences later in life. In particular, ELS exerts lasting effects on neurogenesis in the adult hippocampus, suggesting that ELS is a significant regulator of adult neural stem cell numbers and function. Here, we investigated the effect of ELS on cell proliferation and the numbers of neural stem/precursor cells in another neurogenic region: the hypothalamus of adult mice. We show that ELS has long-term suppressive effects on cell proliferation in the hypothalamic parenchyma and reduces the numbers of putative hypothalamic neural stem/precursor cells at 4 months of age. Specifically, ELS reduced the number of PCNA + cells present in hypothalamic areas surrounding the 3rd ventricle with a specific reduction in the proliferation of Sox2+/Nestin-GFP + putative stem cells present in the median eminence at the base of the 3rd ventricle. Furthermore, ELS reduced the total numbers of β-tanycytes lining the ventral 3rd ventricle, without affecting α-tanycyte numbers in more dorsal areas. These results are the first to indicate that ELS significantly reduces proliferation and β-tanycyte numbers in the adult hypothalamus, and may have (patho)physiological consequences for metabolic regulation or other hypothalamic functions in which β-tanycytes are involved.
Collapse
Affiliation(s)
- Pascal Bielefeld
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, The Netherlands
| | - Maralinde R Abbink
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, The Netherlands
| | - Anna R Davidson
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, The Netherlands
| | - Niels Reijner
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, The Netherlands
| | - Oihane Abiega
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, The Netherlands
| | - Paul J Lucassen
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, The Netherlands
| | - Aniko Korosi
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, The Netherlands
| | - Carlos P Fitzsimons
- Swammerdam Institute for Life Sciences, Faculty of Science, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
49
|
Rajaii F, Kim DW, Pan J, Mahoney NR, Eberhart CG, Qian J, Blackshaw S. Temperature and species-dependent regulation of browning in retrobulbar fat. Sci Rep 2021; 11:3094. [PMID: 33542375 PMCID: PMC7862600 DOI: 10.1038/s41598-021-82672-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/18/2021] [Indexed: 01/30/2023] Open
Abstract
Retrobulbar fat deposits surround the posterior retina and optic nerve head, but their function and origin are obscure. We report that mouse retrobulbar fat is a neural crest-derived tissue histologically and transcriptionally resembles interscapular brown fat. In contrast, human retrobulbar fat closely resembles white adipose tissue. Retrobulbar fat is also brown in other rodents, which are typically housed at temperatures below thermoneutrality, but is white in larger animals. We show that retrobulbar fat in mice housed at thermoneutral temperature show reduced expression of the brown fat marker Ucp1, and histological properties intermediate between white and brown fat. We conclude that retrobulbar fat can potentially serve as a site of active thermogenesis, that this capability is both temperature and species-dependent, and that this may facilitate regulation of intraocular temperature.
Collapse
Affiliation(s)
- Fatemeh Rajaii
- grid.21107.350000 0001 2171 9311Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Dong Won Kim
- grid.21107.350000 0001 2171 9311Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Jianbo Pan
- grid.21107.350000 0001 2171 9311Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Nicholas R. Mahoney
- grid.21107.350000 0001 2171 9311Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Charles G. Eberhart
- grid.21107.350000 0001 2171 9311Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD USA ,grid.21107.350000 0001 2171 9311Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Jiang Qian
- grid.21107.350000 0001 2171 9311Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD USA
| | - Seth Blackshaw
- grid.21107.350000 0001 2171 9311Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD USA ,grid.21107.350000 0001 2171 9311Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD USA ,grid.21107.350000 0001 2171 9311Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD USA ,grid.21107.350000 0001 2171 9311Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD USA ,grid.21107.350000 0001 2171 9311Kavli Neuroscience Discovery Institute, Johns Hopkins University School of Medicine, Baltimore, MD USA
| |
Collapse
|
50
|
de Souza Santos R, Gross AR, Sareen D. Hypothalamus and neuroendocrine diseases: The use of human-induced pluripotent stem cells for disease modeling. HANDBOOK OF CLINICAL NEUROLOGY 2021; 181:337-350. [PMID: 34238469 DOI: 10.1016/b978-0-12-820683-6.00025-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The hypothalamus, which is part of the brain of all vertebrate animals, is considered the link between the central nervous system (CNS) and (i) the endocrine system via the pituitary gland and (ii) with our organs via the autonomic nervous system. It synthesizes and releases neurohormones, which in turn stimulate or inhibit the secretion of other hormones within the CNS, and sends and receives signals to and from the peripheral nervous and endocrine systems. As the brain region responsible for energy homeostasis, the hypothalamus is the key regulator of thermoregulation, hunger and satiety, circadian rhythms, sleep and fatigue, memory and learning, arousal and reproductive cycling, blood pressure, and heart rate and thus orchestrates complex physiological responses in order to maintain metabolic homeostasis. These critical roles implicate the hypothalamus in neuroendocrine disorders such as obesity, diabetes, anorexia nervosa, bulimia, and others. In this chapter, we focus on the use of human-induced pluripotent stem cells (hiPSCs) and their differentiation into hypothalamic neurons in order to model neuroendocrine disorders such as extreme obesity in a dish. To do so, we discuss important steps of human hypothalamus development, neuroendocrine diseases related to the hypothalamus, multiple protocols to differentiate hiPSCs into hypothalamic neurons, and severe obesity modeling in vitro using hiPSCs-derived hypothalamic neurons.
Collapse
Affiliation(s)
- Roberta de Souza Santos
- Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States; Cedars-Sinai Biomanufacturing Center, West Hollywood, CA, United States
| | - Andrew R Gross
- Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States; Cedars-Sinai Biomanufacturing Center, West Hollywood, CA, United States
| | - Dhruv Sareen
- Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States; Cedars-Sinai Biomanufacturing Center, West Hollywood, CA, United States; Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States; iPSC Core, David and Janet Polak Foundation Stem Cell Core Laboratory, Cedars-Sinai Medical Center, Los Angeles, CA, United States.
| |
Collapse
|