1
|
Qi G, Tang H, Hu J, Kang S, Qin S. Potential role of tanycyte-derived neurogenesis in Alzheimer's disease. Neural Regen Res 2025; 20:1599-1612. [PMID: 38934388 DOI: 10.4103/nrr.nrr-d-23-01865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 04/17/2024] [Indexed: 06/28/2024] Open
Abstract
Tanycytes, specialized ependymal cells located in the hypothalamus, play a crucial role in the generation of new neurons that contribute to the neural circuits responsible for regulating the systemic energy balance. The precise coordination of the gene networks controlling neurogenesis in naive and mature tanycytes is essential for maintaining homeostasis in adulthood. However, our understanding of the molecular mechanisms and signaling pathways that govern the proliferation and differentiation of tanycytes into neurons remains limited. This article aims to review the recent advancements in research into the mechanisms and functions of tanycyte-derived neurogenesis. Studies employing lineage-tracing techniques have revealed that the neurogenesis specifically originating from tanycytes in the hypothalamus has a compensatory role in neuronal loss and helps maintain energy homeostasis during metabolic diseases. Intriguingly, metabolic disorders are considered early biomarkers of Alzheimer's disease. Furthermore, the neurogenic potential of tanycytes and the state of newborn neurons derived from tanycytes heavily depend on the maintenance of mild microenvironments, which may be disrupted in Alzheimer's disease due to the impaired blood-brain barrier function. However, the specific alterations and regulatory mechanisms governing tanycyte-derived neurogenesis in Alzheimer's disease remain unclear. Accumulating evidence suggests that tanycyte-derived neurogenesis might be impaired in Alzheimer's disease, exacerbating neurodegeneration. Confirming this hypothesis, however, poses a challenge because of the lack of long-term tracing and nucleus-specific analyses of newborn neurons in the hypothalamus of patients with Alzheimer's disease. Further research into the molecular mechanisms underlying tanycyte-derived neurogenesis holds promise for identifying small molecules capable of restoring tanycyte proliferation in neurodegenerative diseases. This line of investigation could provide valuable insights into potential therapeutic strategies for Alzheimer's disease and related conditions.
Collapse
Affiliation(s)
- Guibo Qi
- Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Han Tang
- Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Jianian Hu
- Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Siying Kang
- Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Song Qin
- Department of Anatomy, Histology, and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| |
Collapse
|
2
|
Lee TH, Nicolas JC, Quarta C. Molecular and functional mapping of the neuroendocrine hypothalamus: a new era begins. J Endocrinol Invest 2024; 47:2627-2648. [PMID: 38878127 DOI: 10.1007/s40618-024-02411-5] [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: 02/23/2024] [Accepted: 06/08/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Recent advances in neuroscience tools for single-cell molecular profiling of brain neurons have revealed an enormous spectrum of neuronal subpopulations within the neuroendocrine hypothalamus, highlighting the remarkable molecular and cellular heterogeneity of this brain area. RATIONALE Neuronal diversity in the hypothalamus reflects the high functional plasticity of this brain area, where multiple neuronal populations flexibly integrate a variety of physiological outputs, including energy balance, stress and fertility, through crosstalk mechanisms with peripheral hormones. Intrinsic functional heterogeneity is also observed within classically 'defined' subpopulations of neuroendocrine neurons, including subtypes with distinct neurochemical signatures, spatial organisation and responsiveness to hormonal cues. AIM The aim of this review is to critically evaluate past and current research on the functional diversity of hypothalamic neuroendocrine neurons and their plasticity. It focuses on how this neuronal plasticity in this brain area relates to metabolic control, feeding regulation and interactions with stress and fertility-related neural circuits. CONCLUSION Our analysis provides an original framework for improving our understanding of the hypothalamic regulation of hormone function and the development of neuroendocrine diseases.
Collapse
Affiliation(s)
- T H Lee
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, 33000, Bordeaux, France
| | - J-C Nicolas
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, 33000, Bordeaux, France
| | - C Quarta
- University of Bordeaux, INSERM, Neurocentre Magendie, U1215, 33000, Bordeaux, France.
| |
Collapse
|
3
|
Appenroth D, Cázarez-Márquez F. Seasonal food intake and energy balance: Neuronal and non-neuronal control mechanisms. Neuropharmacology 2024; 257:110050. [PMID: 38914372 DOI: 10.1016/j.neuropharm.2024.110050] [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: 02/26/2024] [Revised: 06/05/2024] [Accepted: 06/21/2024] [Indexed: 06/26/2024]
Abstract
Animals inhabiting temperate and high latitudes undergo drastic seasonal changes in energy storage, facilitated by changes in food intake and body mass. Those seasonal changes in the animal's biology are not mere consequences of environmental energy availability but are anticipatory responses to the energetic requirements of the upcoming season and are actively timed by tracking the annual progression in photoperiod. In this review, we discuss how photoperiod is used to control energy balance seasonally and how this is distinct from energy homeostasis. Most notably, we suggest that photoperiodic control of food intake and body mass does not originate from the arcuate nucleus, as for homeostatic appetite control, but is rather to be found in hypothalamic tanycytes. Tanycytes are specialized ependymal cells lining the third ventricle, which can sense metabolites from the cerebrospinal fluid (e.g. glucose) and can control access of circulating signals to the brain. They are also essential in conveying time-of-year information by integrating photoperiod and altering hypothalamic thyroid metabolism, a feature that is conserved in seasonal vertebrates and connects to seasonal breeding and metabolism. We also discuss how homeostatic feedback signals are handled during times of rapid energetic transitions. Studies on leptin in seasonal mammals suggest a seasonal shift in central sensitivity and blood-brain transport, which might be facilitated by tanycytes. This article is part of the Special Issue on "Food intake and feeding states".
Collapse
Affiliation(s)
- Daniel Appenroth
- Arctic Seasonal Timekeeping Initiative (ASTI), Arctic Chronobiology & Physiology, Arctic & Marine Biology, BFE, UiT - Arctic University of Norway, Tromsø, Norway.
| | - Fernando Cázarez-Márquez
- Arctic Seasonal Timekeeping Initiative (ASTI), Arctic Chronobiology & Physiology, Arctic & Marine Biology, BFE, UiT - Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
4
|
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
|
5
|
Sharif A, Prevot V. Astrogenesis in the hypothalamus: A life-long process contributing to the development and plasticity of neuroendocrine networks. Front Neuroendocrinol 2024; 75:101154. [PMID: 39226950 DOI: 10.1016/j.yfrne.2024.101154] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/05/2024]
Abstract
Astrocytes are now recognized as integral components of neural circuits, regulating their maturation, activity and plasticity. Neuroendocrinology has provided fertile ground for revealing the diverse strategies used by astrocytes to regulate the physiological and behavioural outcomes of neural circuit activity in response to internal and environmental inputs. However, the development of astrocytes in the hypothalamus has received much less attention than in other brain regions such as the cerebral cortex and spinal cord. In this review, we synthesize our current knowledge of astrogenesis in the hypothalamus across various life stages. A distinctive feature of hypothalamic astrogenesis is that it persists all throughout lifespan, and involves multiple cellular sources corresponding to radial glial cells during early development, followed by tanycytes, parenchymal progenitors and locally dividing astrocytes. Astrogenesis in the hypothalamus is closely coordinated with the maturation of hypothalamic neurons. This coordination is exemplified by recent findings in neurons producing gonadotropin-releasing hormone, which actively shape their astroglial environment during infancy to integrate functionally into their neural network and facilitate sexual maturation, a process vulnerable to endocrine disruption. While hypothalamic astrogenesis shares common principles with other brain regions, it also exhibits specific features in its dynamics and regulation, both at the inter- and intra-regional levels. These unique properties emphasize the importance of further exploration. Additionally, we discuss the experimental strategies used to assess astrogenesis in the hypothalamus and their potential bias and limitations. Understanding the mechanisms of hypothalamic astrogenesis throughout life will be crucial for comprehending the development and function of the hypothalamus under both physiological and pathological conditions.
Collapse
Affiliation(s)
- Ariane Sharif
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S 1172, FHU 1000 Days for Health, Lille, France.
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S 1172, FHU 1000 Days for Health, Lille, France.
| |
Collapse
|
6
|
Galichet C, Rizzoti K, Lovell-Badge R. Hypopituitarism in Sox3 null mutants correlates with altered NG2-glia in the median eminence and is influenced by aspirin and gut microbiota. PLoS Genet 2024; 20:e1011395. [PMID: 39325695 PMCID: PMC11426531 DOI: 10.1371/journal.pgen.1011395] [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: 01/15/2024] [Accepted: 08/13/2024] [Indexed: 09/28/2024] Open
Abstract
The median eminence (ME), located at the base of the hypothalamus, is an essential centre of information exchange between the brain and the pituitary. We and others previously showed that mutations and duplications affecting the transcription factor SOX3/Sox3 result in hypopituitarism, and this is likely of hypothalamic origin. We demonstrate here that the absence of Sox3 predominantly affects the ME with phenotypes that first occur in juvenile animals, despite the embryonic onset of SOX3 expression. In the pituitary, reduction in hormone levels correlates with a lack of endocrine cell maturation. In parallel, ME NG2-glia renewal and oligodendrocytic differentiation potential are affected. We further show that low-dose aspirin treatment, which is known to affect NG2-glia, or changes in gut microbiota, rescue both proliferative defects and hypopituitarism in Sox3 mutants. Our study highlights a central role of NG2-glia for ME function during a transitional period of post-natal development and indicates their sensitivity to extrinsic signals.
Collapse
Affiliation(s)
- Christophe Galichet
- Stem Cell Biology and Developmental Genetics Lab, The Francis Crick Institute, London, United Kingdom
- Neurobiological Research Facility, UCL Sainsbury Wellcome Centre for Neural Circuits and Behaviour, London, United Kingdom
| | - Karine Rizzoti
- Stem Cell Biology and Developmental Genetics Lab, The Francis Crick Institute, London, United Kingdom
| | - Robin Lovell-Badge
- Stem Cell Biology and Developmental Genetics Lab, The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
7
|
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
|
8
|
Brunner M, Lopez-Rodriguez D, Estrada-Meza J, Dali R, Rohrbach A, Deglise T, Messina A, Thorens B, Santoni F, Langlet F. Fasting induces metabolic switches and spatial redistributions of lipid processing and neuronal interactions in tanycytes. Nat Commun 2024; 15:6604. [PMID: 39098920 PMCID: PMC11298547 DOI: 10.1038/s41467-024-50913-w] [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: 08/02/2023] [Accepted: 07/25/2024] [Indexed: 08/06/2024] Open
Abstract
The ependyma lining the third ventricle (3V) in the mediobasal hypothalamus plays a crucial role in energy balance and glucose homeostasis. It is characterized by a high functional heterogeneity and plasticity, but the underlying molecular mechanisms governing its features are not fully understood. Here, 5481 hypothalamic ependymocytes were cataloged using FACS-assisted scRNAseq from fed, 12h-fasted, and 24h-fasted adult male mice. With standard clustering analysis, typical ependymal cells and β2-tanycytes appear sharply defined, but other subpopulations, β1- and α-tanycytes, display fuzzy boundaries with few or no specific markers. Pseudospatial approaches, based on the 3V neuroanatomical distribution, enable the identification of specific versus shared tanycyte markers and subgroup-specific versus general tanycyte functions. We show that fasting dynamically shifts gene expression patterns along the 3V, leading to a spatial redistribution of cell type-specific responses. Altogether, we show that changes in energy status induce metabolic and functional switches in tanycyte subpopulations, providing insights into molecular and functional diversity and plasticity within the tanycyte population.
Collapse
Affiliation(s)
- Maxime Brunner
- Service of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - David Lopez-Rodriguez
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Judith Estrada-Meza
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Rafik Dali
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Antoine Rohrbach
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Tamara Deglise
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Andrea Messina
- Service of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, Lausanne, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Bernard Thorens
- Center for Integrative Genomics, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Federico Santoni
- Service of Endocrinology, Diabetology, and Metabolism, Lausanne University Hospital, Lausanne, Switzerland.
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland.
- Institute for Genetic and Biomedical Research (IRGB) - CNR, Monserrato, Italy.
| | - Fanny Langlet
- 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
|
9
|
Hazlerigg DG, Simonneaux V, Dardente H. Melatonin and Seasonal Synchrony in Mammals. J Pineal Res 2024; 76:e12996. [PMID: 39129720 DOI: 10.1111/jpi.12996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 05/27/2024] [Accepted: 07/16/2024] [Indexed: 08/13/2024]
Abstract
In mammals, seasonal opportunities and challenges are anticipated through programmed changes in physiology and behavior. Appropriate anticipatory timing depends on synchronization to the external solar year, achieved through the use of day length (photoperiod) as a synchronizing signal. In mammals, nocturnal production of melatonin by the pineal gland is the key hormonal mediator of photoperiodic change, exerting its effects via the hypothalamopituitary axis. In this review/perspective, we consider the key developments during the history of research into the seasonal synchronizer effect of melatonin, highlighting the role that the pars tuberalis-tanycyte module plays in this process. We go on to consider downstream pathways, which include discrete hypothalamic neuronal populations. Neurons that express the neuropeptides kisspeptin and (Arg)(Phe)-related peptide-3 (RFRP-3) govern seasonal reproductive function while neurons that express somatostatin may be involved in seasonal metabolic adaptations. Finally, we identify several outstanding questions, which need to be addressed to provide a much thorough understanding of the deep impact of melatonin upon seasonal synchronization.
Collapse
Affiliation(s)
- David G Hazlerigg
- Department of Arctic and Marine Biology, Arctic Chronobiology and Physiology Research Group, Arctic Seasonal Timekeeping Initiative (ASTI), UiT-The Arctic University of Norway, Tromsø, Norway
| | - Valérie Simonneaux
- Institute for Cellular and Integrative Neuroscience, University of Strasbourg, Strasbourg, France
| | | |
Collapse
|
10
|
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 2024:10.1038/s41574-024-01021-8. [PMID: 39054359 DOI: 10.1038/s41574-024-01021-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [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
|
11
|
Bai Y, Chen Q, Li Y. A single-cell transcriptomic study of heterogeneity in human embryonic tanycytes. Sci Rep 2024; 14:15384. [PMID: 38965316 PMCID: PMC11224400 DOI: 10.1038/s41598-024-66044-7] [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: 01/14/2024] [Accepted: 06/26/2024] [Indexed: 07/06/2024] Open
Abstract
Disruptions in energy homeostasis can lead to diseases like obesity and diabetes, affecting millions of people each year. Tanycytes, the adult stem cells in the hypothalamus, play crucial roles in assisting hypothalamic neurons in maintaining energy balance. Although tanycytes have been extensively studied in rodents, our understanding of human tanycytes remains limited. In this study, we utilized single-cell transcriptomics data to explore the heterogeneity of human embryonic tanycytes, investigate their gene regulatory networks, analyze their intercellular communication, and examine their developmental trajectory. Our analysis revealed the presence of two clusters of β tanycytes and three clusters of α tanycytes in our dataset. Surprisingly, human embryonic tanycytes displayed significant similarities to mouse tanycytes in terms of marker gene expression and transcription factor activities. Trajectory analysis indicated that α tanycytes were the first to be generated, giving rise to β tanycytes in a dorsal-ventral direction along the third ventricle. Furthermore, our CellChat analyses demonstrated that tanycytes generated earlier along the developmental lineages exhibited increased intercellular communication compared to those generated later. In summary, we have thoroughly characterized the heterogeneity of human embryonic tanycytes from various angles. We are confident that our findings will serve as a foundation for future research on human tanycytes.
Collapse
Affiliation(s)
- Yiguang Bai
- Department of Orthopaedics, The Second Clinical Institute of North Sichuan Medical College Nanchong, Nanchong Central Hospital, Nanchong, Sichuan, China.
- Nanchong Hospital of Beijing Anzhen Hospital Capital Medical University Sichuan, Beijing, China.
| | - Qiaoling Chen
- Department of Oncology, The Second Clinical Institute of North Sichuan Medical College Nanchong, Nanchong Central Hospital, Nanchong, Sichuan, China
- Nanchong Hospital of Beijing Anzhen Hospital Capital Medical University Sichuan, Beijing, China
| | - Yuan Li
- National Bioinformatics Infrastructure Sweden (NBIS), Science for Life Laboratory, Lund University, 223 87, Lund, Sweden.
- Department of Immunotechnology, Lund University, Medicon Village, 22387, Lund, Sweden.
- Human Neural Developmental Biology; BMC B11, Department of Experimental Medical Science Lund, Stem Cell Centre, Lund University, 22184, Lund, Sweden.
- Cell, Tissue & Organ Engineering Laboratory; BMC B11, Department of Clinical Sciences Lund, Stem Cell Centre, Lund University, 22184, Lund, Sweden.
| |
Collapse
|
12
|
Dardente H, Lomet D, Robert V, Lasserre O, Gonzalez AA, Mialhe X, Beltramo M. Photoperiod, but not progesterone, has a strong impact upon the transcriptome of the medio-basal hypothalamus in female goats and ewes. Mol Cell Endocrinol 2024; 588:112216. [PMID: 38556161 DOI: 10.1016/j.mce.2024.112216] [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: 11/23/2023] [Revised: 03/11/2024] [Accepted: 03/24/2024] [Indexed: 04/02/2024]
Abstract
Photoperiod is the main environmental driver of seasonal responses in organisms living at temperate and polar latitudes. Other external cues such as food and temperature, and internal cues including hormones, intervene to fine-tune phasing of physiological functions to the solar year. In mammals, the medio-basal hypothalamus (MBH) is the key integrator of these cues, which orchestrates a wide array of seasonal functions, including breeding. Here, using RNAseq and RT-qPCR, we demonstrate that molecular components of the photoperiodic response previously identified in ewes are broadly conserved in does (female goats, Capra hircus), with a common core of ∼50 genes. This core group can be defined as the "MBH seasonal trancriptome", which includes key players of the pars tuberalis-tanycytes neuroendocrine retrograde pathway that governs intra-MBH photoperiodic switches of triiodothyronine (T3) production (Tshb, Eya3, Dio2 and SlcO1c1), the two histone methyltransferases Suv39H2 and Ezh2 and the secreted protein Vmo1. Prior data in ewes revealed that T3 and estradiol (E2), both key hormones for the proper timing of seasonal breeding, differentially impact the MBH seasonal transcriptome, and identified cellular and molecular targets through which these hormones might act. In contrast, information regarding the potential impact of progesterone (P4) upon the MBH transcriptome was nonexistent. Here, we demonstrate that P4 has no discernible transcriptional impact in either does or ewes. Taken together, our data show that does and ewes possess a common core set of photoperiod-responsive genes in the MBH and conclusively demonstrate that P4 is not a key regulator of the MBH transcriptome.
Collapse
Affiliation(s)
- Hugues Dardente
- INRAE, CNRS, Université de Tours, PRC, 37380, Nouzilly, France.
| | - Didier Lomet
- INRAE, CNRS, Université de Tours, PRC, 37380, Nouzilly, France
| | - Vincent Robert
- INRAE, CNRS, Université de Tours, PRC, 37380, Nouzilly, France
| | | | - Anne-Alicia Gonzalez
- MGX-Montpellier GenomiX, Univ. Montpellier, CNRS, INSERM, 34094, Montpellier, France
| | - Xavier Mialhe
- MGX-Montpellier GenomiX, Univ. Montpellier, CNRS, INSERM, 34094, Montpellier, France
| | | |
Collapse
|
13
|
Baynat L, Yamamoto T, Tourdias T, Zhang B, Prevost V, Infante A, Klein A, Caid J, Cadart O, Dousset V, Gatta Cherifi B. Quantitative MRI Biomarkers Measure Changes in Targeted Brain Areas in Patients With Obesity. J Clin Endocrinol Metab 2024; 109:1850-1857. [PMID: 38195765 DOI: 10.1210/clinem/dgae014] [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: 08/10/2023] [Revised: 12/14/2023] [Accepted: 01/08/2024] [Indexed: 01/11/2024]
Abstract
CONTEXT Obesity is accompanied by damages to several tissues, including the brain. Pathological data and animal models have demonstrated an increased inflammatory reaction in hypothalamus and hippocampus. OBJECTIVE We tested whether we could observe such pathological modifications in vivo through quantitative magnetic resonance imaging (MRI) metrics. METHODS This prospective study was conducted between May 2019 and November 2022. The study was conducted in the Specialized Center for the Care of Obesity in a French University Hospital. Twenty-seven patients with obesity and 23 age and gender-paired normal-weight controls were prospectively recruited. All participants were examined using brain MRI. Anthropometric and biological data, eating behavior, anxiety, depression, and memory performance were assessed in both groups. The main outcome measure was brain MRI with the following parametric maps: quantitative susceptibility mapping (QSM), mean diffusivity (MD), fractional anisotropy (FA), magnetization transfer ratio map, and T2 relaxivity map. RESULTS In the hypothalamus, patients with obesity had higher FA and lower QSM than normal-weight controls. In the hippocampus, patients with obesity had higher FA and lower MD. There was no correlation between imaging biomarkers and eating behavior or anxiety. CONCLUSION Our findings are consistent with the presence of neuroinflammation in brain regions involved in food intake. In vivo brain biomarkers from quantitative MRI appear to provide an incremental information for the assessment of brain damages in patients with obesity.
Collapse
Affiliation(s)
- Louise Baynat
- University of Bordeaux, INSERM U1215, Neurocentre Magendie, 33000 Bordeaux, France
- CHU Bordeaux, Hôpital Haut Lévêque Service Endocrinologie, Diabétologie, Nutrition, 33600 Pessac, France
| | - Takayuki Yamamoto
- University of Bordeaux, INSERM U1215, Neurocentre Magendie, 33000 Bordeaux, France
| | - Thomas Tourdias
- University of Bordeaux, INSERM U1215, Neurocentre Magendie, 33000 Bordeaux, France
- CHU Bordeaux, Hôpital Pellegrin, Service de Neuroimagerie diagnostique et thérapeutique, 33000 Bordeaux, France
| | - Bei Zhang
- Magnetic Resonance, Canon Medical Systems Europe, 2718 Zoetermeer, Netherlands
| | - Valentin Prevost
- CT-MR Solution Planning Department, Canon Medical Systems Corporation, Tochigi, Japan
| | - Asael Infante
- CHU Bordeaux, Hôpital Haut Lévêque Service Endocrinologie, Diabétologie, Nutrition, 33600 Pessac, France
| | - Achille Klein
- CHU Bordeaux, Hôpital Haut Lévêque Service Endocrinologie, Diabétologie, Nutrition, 33600 Pessac, France
| | - Julien Caid
- CHU Bordeaux, Hôpital Haut Lévêque Service Endocrinologie, Diabétologie, Nutrition, 33600 Pessac, France
| | - Olivier Cadart
- Endocrinology, Centre Hospitalier d'Angoulême, Endocrinolology, Rond point Girac, 16000 Angouleme, France
| | - Vincent Dousset
- University of Bordeaux, INSERM U1215, Neurocentre Magendie, 33000 Bordeaux, France
- CHU Bordeaux, Hôpital Pellegrin, Service de Neuroimagerie diagnostique et thérapeutique, 33000 Bordeaux, France
| | - Blandine Gatta Cherifi
- University of Bordeaux, INSERM U1215, Neurocentre Magendie, 33000 Bordeaux, France
- CHU Bordeaux, Hôpital Haut Lévêque Service Endocrinologie, Diabétologie, Nutrition, 33600 Pessac, France
| |
Collapse
|
14
|
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
|
15
|
Benevento M, Alpár A, Gundacker A, Afjehi L, Balueva K, Hevesi Z, Hanics J, Rehman S, Pollak DD, Lubec G, Wulff P, Prevot V, Horvath TL, Harkany T. A brainstem-hypothalamus neuronal circuit reduces feeding upon heat exposure. Nature 2024; 628:826-834. [PMID: 38538787 PMCID: PMC11041654 DOI: 10.1038/s41586-024-07232-3] [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: 08/30/2022] [Accepted: 02/22/2024] [Indexed: 04/06/2024]
Abstract
Empirical evidence suggests that heat exposure reduces food intake. However, the neurocircuit architecture and the signalling mechanisms that form an associative interface between sensory and metabolic modalities remain unknown, despite primary thermoceptive neurons in the pontine parabrachial nucleus becoming well characterized1. Tanycytes are a specialized cell type along the wall of the third ventricle2 that bidirectionally transport hormones and signalling molecules between the brain's parenchyma and ventricular system3-8. Here we show that tanycytes are activated upon acute thermal challenge and are necessary to reduce food intake afterwards. Virus-mediated gene manipulation and circuit mapping showed that thermosensing glutamatergic neurons of the parabrachial nucleus innervate tanycytes either directly or through second-order hypothalamic neurons. Heat-dependent Fos expression in tanycytes suggested their ability to produce signalling molecules, including vascular endothelial growth factor A (VEGFA). Instead of discharging VEGFA into the cerebrospinal fluid for a systemic effect, VEGFA was released along the parenchymal processes of tanycytes in the arcuate nucleus. VEGFA then increased the spike threshold of Flt1-expressing dopamine and agouti-related peptide (Agrp)-containing neurons, thus priming net anorexigenic output. Indeed, both acute heat and the chemogenetic activation of glutamatergic parabrachial neurons at thermoneutrality reduced food intake for hours, in a manner that is sensitive to both Vegfa loss-of-function and blockage of vesicle-associated membrane protein 2 (VAMP2)-dependent exocytosis from tanycytes. Overall, we define a multimodal neurocircuit in which tanycytes link parabrachial sensory relay to the long-term enforcement of a metabolic code.
Collapse
Affiliation(s)
- Marco Benevento
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Alán Alpár
- Department of Anatomy, Histology, and Embryology, Semmelweis University, Budapest, Hungary
- SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Semmelweis University, Budapest, Hungary
| | - Anna Gundacker
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Leila Afjehi
- Programme Proteomics, Paracelsus Medizinische Privatuniversität, Salzburg, Austria
| | - Kira Balueva
- Institute of Physiology, Christian Albrechts University, Kiel, Germany
| | - Zsofia Hevesi
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - János Hanics
- Department of Anatomy, Histology, and Embryology, Semmelweis University, Budapest, Hungary
- SE NAP Research Group of Experimental Neuroanatomy and Developmental Biology, Semmelweis University, Budapest, Hungary
| | - Sabah Rehman
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Daniela D Pollak
- Department of Neurophysiology and Neuropharmacology, Center for Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Gert Lubec
- Programme Proteomics, Paracelsus Medizinische Privatuniversität, Salzburg, Austria
| | - Peer Wulff
- Institute of Physiology, Christian Albrechts University, Kiel, Germany
| | - Vincent Prevot
- University of Lille, INSERM, CHU Lille, Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience and Cognition, UMR S1172, EGID, Lille, France
| | - Tamas L Horvath
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Tibor Harkany
- Department of Molecular Neurosciences, Center for Brain Research, Medical University of Vienna, Vienna, Austria.
- Department of Neuroscience, Karolinska Institutet, Solna, Sweden.
| |
Collapse
|
16
|
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
|
17
|
Zhou S, Makashova O, Chevillard PM, Josey V, Li B, Prager-Khoutorsky M. Constitutive cell proliferation and neurogenesis in the organum vasculosum lamina terminalis and subfornical organ of adult rats. J Neuroendocrinol 2024; 36:e13377. [PMID: 38418229 DOI: 10.1111/jne.13377] [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: 10/14/2023] [Revised: 02/08/2024] [Accepted: 02/09/2024] [Indexed: 03/01/2024]
Abstract
Neurogenesis continues throughout adulthood in the subventricular zone, hippocampal subgranular zone, and the hypothalamic median eminence (ME) and the adjacent medio-basal hypothalamus. The ME is one of the circumventricular organs (CVO), which are specialized brain areas characterized by an incomplete blood-brain barrier and, thus, are involved in mediating communication between the central nervous system and the periphery. Additional CVOs include the organum vasculosum laminae terminalis (OVLT) and the subfornical organs (SFO). Previous studies have demonstrated that the ME contains neural stem cells (NSCs) capable of generating new neurons and glia in the adult brain. However, it remains unclear whether the OVLT and SFO also contain proliferating cells, the identity of these cells, and their ability to differentiate into mature neurons. Here we show that glial and mural subtypes exhibit NSC characteristics, expressing the endogenous mitotic maker Ki67, and incorporating the exogenous mitotic marker BrdU in the OVLT and SFO of adult rats. Glial cells constitutively proliferating in the SFO comprise NG2 glia, while in the OVLT, both NG2 glia and tanycytes appear to constitute the NSC pool. Furthermore, pericytes, which are mural cells associated with capillaries, also contribute to the pool of cells constitutively proliferating in the OVLT and SFO of adult rats. In addition to these glial and mural cells, a fraction of NSCs containing proliferation markers Ki67 and BrdU also expresses the early postmitotic neuronal marker doublecortin, suggesting that these CVOs comprise newborn neurons. Notably, these neurons can differentiate and express the mature neuronal marker NeuN. These findings establish the sensory CVOs OVLT and SFO as additional neurogenic niches, where the generation of new neurons and glia persists in the adult brain.
Collapse
Affiliation(s)
- Suijian Zhou
- Department of Physiology, McIntyre Medical Sciences Building, McGill University, Montreal, Québec, Canada
| | - Olena Makashova
- Department of Physiology, McIntyre Medical Sciences Building, McGill University, Montreal, Québec, Canada
| | - Pierre-Marie Chevillard
- Department of Physiology, McIntyre Medical Sciences Building, McGill University, Montreal, Québec, Canada
| | - Vanessa Josey
- Department of Physiology, McIntyre Medical Sciences Building, McGill University, Montreal, Québec, Canada
| | - Banruo Li
- Department of Physiology, McIntyre Medical Sciences Building, McGill University, Montreal, Québec, Canada
| | - Masha Prager-Khoutorsky
- Department of Physiology, McIntyre Medical Sciences Building, McGill University, Montreal, Québec, Canada
| |
Collapse
|
18
|
Cotellessa L, Giacobini P. Role of Anti-Müllerian Hormone in the Central Regulation of Fertility. Semin Reprod Med 2024; 42:34-40. [PMID: 38608673 DOI: 10.1055/s-0044-1786050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/14/2024]
Abstract
In recent years, the expanding roles of anti-Müllerian hormone (AMH) in various aspects of reproductive health have attracted significant attention. Initially recognized for its classical role in male sexual differentiation, AMH is produced postnatally by the Sertoli cells in the male testes and by the granulosa cells in the female ovaries. Traditionally, it was believed to primarily influence gonadal development and function. However, research over the last decade has unveiled novel actions of AMH beyond the gonads, specifically all along the hypothalamic-pituitary-gonadal axis. This review will focus on the emerging roles of AMH within the hypothalamus and discusses its potential implications in reproductive physiology. Additionally, recent preclinical and clinical studies have suggested that elevated levels of AMH may disrupt the hypothalamic network regulating reproduction, which could contribute to the central pathophysiology of polycystic ovary syndrome. These findings underscore the intricate interplay between AMH and the neuroendocrine system, offering new avenues for understanding the mechanisms underlying fertility and reproductive disorders.
Collapse
Affiliation(s)
- Ludovica Cotellessa
- Inserm, CHU Lille, Unit 1172, Lille Neuroscience & Cognition (LilNCog), University of Lille, Lille, France
| | - Paolo Giacobini
- Inserm, CHU Lille, Unit 1172, Lille Neuroscience & Cognition (LilNCog), University of Lille, Lille, France
| |
Collapse
|
19
|
Lei Y, Liang X, Sun Y, Yao T, Gong H, Chen Z, Gao Y, Wang H, Wang R, Huang Y, Yang T, Yu M, Liu L, Yi CX, Wu QF, Kong X, Xu X, Liu S, Zhang Z, Liu T. Region-specific transcriptomic responses to obesity and diabetes in macaque hypothalamus. Cell Metab 2024; 36:438-453.e6. [PMID: 38325338 DOI: 10.1016/j.cmet.2024.01.003] [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: 06/08/2023] [Revised: 10/27/2023] [Accepted: 01/05/2024] [Indexed: 02/09/2024]
Abstract
The hypothalamus plays a crucial role in the progression of obesity and diabetes; however, its structural complexity and cellular heterogeneity impede targeted treatments. Here, we profiled the single-cell and spatial transcriptome of the hypothalamus in obese and sporadic type 2 diabetic macaques, revealing primate-specific distributions of clusters and genes as well as spatial region, cell-type-, and gene-feature-specific changes. The infundibular (INF) and paraventricular nuclei (PVN) are most susceptible to metabolic disruption, with the PVN being more sensitive to diabetes. In the INF, obesity results in reduced synaptic plasticity and energy sensing capability, whereas diabetes involves molecular reprogramming associated with impaired tanycytic barriers, activated microglia, and neuronal inflammatory response. In the PVN, cellular metabolism and neural activity are suppressed in diabetic macaques. Spatial transcriptomic data reveal microglia's preference for the parenchyma over the third ventricle in diabetes. Our findings provide a comprehensive view of molecular changes associated with obesity and diabetes.
Collapse
Affiliation(s)
- Ying Lei
- BGI-Research, Hangzhou 310012, China; BGI-Research, Shenzhen 518103, China
| | - Xian Liang
- State Key Laboratory of Genetic Engineering, Department of Endocrinology and Metabolism, Human Phenome Institute, Institute of Metabolism and Integrative Biology, and School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China; School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Yunong Sun
- BGI-Research, Hangzhou 310012, China; BGI-Research, Shenzhen 518103, China
| | - Ting Yao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University School of Medicine, Xi'an, Shanxi 710063, China
| | - Hongyu Gong
- School of Life Sciences, Institues of Biomedical Sciences, Inner Mongolia University, Hohhot 010000, China
| | - Zhenhua Chen
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuanqing Gao
- Jiangsu Provincial Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Hui Wang
- School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Ru Wang
- School of Kinesiology, Shanghai University of Sport, Shanghai 200438, China
| | - Yunqi Huang
- BGI-Research, Hangzhou 310012, China; BGI-Research, Shenzhen 518103, China
| | - Tao Yang
- China National GeneBank, BGI-Shenzhen, Shenzhen 518120, China
| | - Miao Yu
- School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Longqi Liu
- BGI-Research, Hangzhou 310012, China; BGI-Research, Shenzhen 518103, China
| | - Chun-Xia Yi
- Department of Endocrinology and Metabolism, Amsterdam University Medical Centers, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, the Netherlands
| | - Qing-Feng Wu
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Xingxing Kong
- School of Life Sciences, Fudan University, Shanghai 200438, China.
| | - Xun Xu
- BGI-Research, Hangzhou 310012, China; BGI-Research, Shenzhen 518103, China.
| | - Shiping Liu
- BGI-Research, Hangzhou 310012, China; BGI-Research, Shenzhen 518103, China.
| | - Zhi Zhang
- State Key Laboratory of Genetic Engineering, Department of Endocrinology and Metabolism, Human Phenome Institute, Institute of Metabolism and Integrative Biology, and School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China; School of Life Sciences, Fudan University, Shanghai 200438, China.
| | - Tiemin Liu
- State Key Laboratory of Genetic Engineering, Department of Endocrinology and Metabolism, Human Phenome Institute, Institute of Metabolism and Integrative Biology, and School of Life Sciences, Zhongshan Hospital, Fudan University, Shanghai 200438, China; School of Life Sciences, Fudan University, Shanghai 200438, China; School of Life Sciences, Institues of Biomedical Sciences, Inner Mongolia University, Hohhot 010000, China.
| |
Collapse
|
20
|
Anderson GM, Hill JW, Kaiser UB, Navarro VM, Ong KK, Perry JRB, Prevot V, Tena-Sempere M, Elias CF. Metabolic control of puberty: 60 years in the footsteps of Kennedy and Mitra's seminal work. Nat Rev Endocrinol 2024; 20:111-123. [PMID: 38049643 PMCID: PMC10843588 DOI: 10.1038/s41574-023-00919-z] [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: 10/19/2023] [Indexed: 12/06/2023]
Abstract
An individual's nutritional status has a powerful effect on sexual maturation. Puberty onset is delayed in response to chronic energy insufficiency and is advanced under energy abundance. The consequences of altered pubertal timing for human health are profound. Late puberty increases the chances of cardiometabolic, musculoskeletal and neurocognitive disorders, whereas early puberty is associated with increased risks of adult obesity, type 2 diabetes mellitus, cardiovascular diseases and various cancers, such as breast, endometrial and prostate cancer. Kennedy and Mitra's trailblazing studies, published in 1963 and using experimental models, were the first to demonstrate that nutrition is a key factor in puberty onset. Building on this work, the field has advanced substantially in the past decade, which is largely due to the impressive development of molecular tools for experimentation and population genetics. In this Review, we discuss the latest advances in basic and translational sciences underlying the nutritional and metabolic control of pubertal development, with a focus on perspectives and future directions.
Collapse
Affiliation(s)
- Greg M Anderson
- Centre for Neuroendocrinology, University of Otago, Dunedin, New Zealand
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Jennifer W Hill
- Department of Physiology and Pharmacology, University of Toledo, Toledo, OH, USA
- Center for Diabetes and Endocrine Research, University of Toledo, Toledo, OH, USA
| | - Ursula B Kaiser
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Victor M Navarro
- Division of Endocrinology, Diabetes, and Hypertension, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Ken K Ong
- Metabolic Research Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - John R B Perry
- Metabolic Research Laboratory, Wellcome-MRC Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
- MRC Epidemiology Unit, Wellcome-MRC Institute of Metabolic Science, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Vincent Prevot
- University of Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S 1172, Lille, France
- European Genomic Institute for Diabetes (EGID), Lille, France
| | - Manuel Tena-Sempere
- Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Cordoba, Spain.
- Department of Cell Biology, Physiology and Immunology, University of Cordoba, Cordoba, Spain.
- CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, Cordoba, Spain.
| | - Carol F Elias
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.
- Department of Obstetrics & Gynecology, University of Michigan, Ann Arbor, MI, USA.
- Caswell Diabetes Institute, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
21
|
Abstract
Traditional textbook physiology has ascribed unitary functions to hormones from the anterior and posterior pituitary gland, mainly in the regulation of effector hormone secretion from endocrine organs. However, the evolutionary biology of pituitary hormones and their receptors provides evidence for a broad range of functions in vertebrate physiology. Over the past decade, we and others have discovered that thyroid-stimulating hormone, follicle-stimulating hormone, adrenocorticotropic hormone, prolactin, oxytocin and arginine vasopressin act directly on somatic organs, including bone, adipose tissue and liver. New evidence also indicates that pituitary hormone receptors are expressed in brain regions, nuclei and subnuclei. These studies have prompted us to attribute the pathophysiology of certain human diseases, including osteoporosis, obesity and neurodegeneration, at least in part, to changes in pituitary hormone levels. This new information has identified actionable therapeutic targets for drug discovery.
Collapse
Affiliation(s)
- Mone Zaidi
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Departments of Medicine and Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Tony Yuen
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Departments of Medicine and Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Se-Min Kim
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Departments of Medicine and Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
22
|
Sauve F, Nampoothiri S, Clarke SA, Fernandois D, Ferreira Coêlho CF, Dewisme J, Mills EG, Ternier G, Cotellessa L, Iglesias-Garcia C, Mueller-Fielitz H, Lebouvier T, Perbet R, Florent V, Baroncini M, Sharif A, Ereño-Orbea J, Mercado-Gómez M, Palazon A, Mattot V, Pasquier F, Catteau-Jonard S, Martinez-Chantar M, Hrabovszky E, Jourdain M, Deplanque D, Morelli A, Guarnieri G, Storme L, Robil C, Trottein F, Nogueiras R, Schwaninger M, Pigny P, Poissy J, Chachlaki K, Maurage CA, Giacobini P, Dhillo W, Rasika S, Prevot V. Long-COVID cognitive impairments and reproductive hormone deficits in men may stem from GnRH neuronal death. EBioMedicine 2023; 96:104784. [PMID: 37713808 PMCID: PMC10507138 DOI: 10.1016/j.ebiom.2023.104784] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 08/02/2023] [Accepted: 08/21/2023] [Indexed: 09/17/2023] Open
Abstract
BACKGROUND We have recently demonstrated a causal link between loss of gonadotropin-releasing hormone (GnRH), the master molecule regulating reproduction, and cognitive deficits during pathological aging, including Down syndrome and Alzheimer's disease. Olfactory and cognitive alterations, which persist in some COVID-19 patients, and long-term hypotestosteronaemia in SARS-CoV-2-infected men are also reminiscent of the consequences of deficient GnRH, suggesting that GnRH system neuroinvasion could underlie certain post-COVID symptoms and thus lead to accelerated or exacerbated cognitive decline. METHODS We explored the hormonal profile of COVID-19 patients and targets of SARS-CoV-2 infection in post-mortem patient brains and human fetal tissue. FINDINGS We found that persistent hypotestosteronaemia in some men could indeed be of hypothalamic origin, favouring post-COVID cognitive or neurological symptoms, and that changes in testosterone levels and body weight over time were inversely correlated. Infection of olfactory sensory neurons and multifunctional hypothalamic glia called tanycytes highlighted at least two viable neuroinvasion routes. Furthermore, GnRH neurons themselves were dying in all patient brains studied, dramatically reducing GnRH expression. Human fetal olfactory and vomeronasal epithelia, from which GnRH neurons arise, and fetal GnRH neurons also appeared susceptible to infection. INTERPRETATION Putative GnRH neuron and tanycyte dysfunction following SARS-CoV-2 neuroinvasion could be responsible for serious reproductive, metabolic, and mental health consequences in long-COVID and lead to an increased risk of neurodevelopmental and neurodegenerative pathologies over time in all age groups. FUNDING European Research Council (ERC) grant agreements No 810331, No 725149, No 804236, the European Union Horizon 2020 research and innovation program No 847941, the Fondation pour la Recherche Médicale (FRM) and the Agence Nationale de la Recherche en Santé (ANRS) No ECTZ200878 Long Covid 2021 ANRS0167 SIGNAL, Agence Nationale de la recherche (ANR) grant agreements No ANR-19-CE16-0021-02, No ANR-11-LABEX-0009, No. ANR-10-LABEX-0046, No. ANR-16-IDEX-0004, Inserm Cross-Cutting Scientific Program HuDeCA, the CHU Lille Bonus H, the UK Medical Research Council (MRC) and National Institute of Health and care Research (NIHR).
Collapse
Affiliation(s)
- Florent Sauve
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France
| | - Sreekala Nampoothiri
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France
| | - Sophie A Clarke
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom
| | - Daniela Fernandois
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France
| | | | - Julie Dewisme
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France; CHU Lille, Department of Pathology, Centre Biologie Pathologie, France
| | - Edouard G Mills
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom
| | - Gaetan Ternier
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France
| | - Ludovica Cotellessa
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France
| | | | - Helge Mueller-Fielitz
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Thibaud Lebouvier
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France; CHU Lille, Department of Neurology, Memory Centre, Reference Centre for Early-Onset Alzheimer Disease and Related Disorders, Lille, France
| | - Romain Perbet
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France; CHU Lille, Department of Pathology, Centre Biologie Pathologie, France
| | - Vincent Florent
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France
| | - Marc Baroncini
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France
| | - Ariane Sharif
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France
| | - June Ereño-Orbea
- CIC bioGUNE, Basque Research and Technology Alliance (BRTACentro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain; Bizkaia Technology Park, Building 801A, 48160, Derio, Bizkaia, Spain
| | - Maria Mercado-Gómez
- CIC bioGUNE, Basque Research and Technology Alliance (BRTACentro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain; Bizkaia Technology Park, Building 801A, 48160, Derio, Bizkaia, Spain
| | - Asis Palazon
- CIC bioGUNE, Basque Research and Technology Alliance (BRTACentro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain; Bizkaia Technology Park, Building 801A, 48160, Derio, Bizkaia, Spain
| | - Virginie Mattot
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France
| | - Florence Pasquier
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France; CHU Lille, Department of Neurology, Memory Centre, Reference Centre for Early-Onset Alzheimer Disease and Related Disorders, Lille, France
| | - Sophie Catteau-Jonard
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France; CHU Lille, Department of Gynecology and Obstetrics, Jeanne de Flandres Hospital, F-59000, Lille, France
| | - Maria Martinez-Chantar
- CIC bioGUNE, Basque Research and Technology Alliance (BRTACentro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain; Bizkaia Technology Park, Building 801A, 48160, Derio, Bizkaia, Spain
| | - Erik Hrabovszky
- Laboratory of Reproductive Neurobiology, Institute of Experimental Medicine, Budapest, Hungary
| | - Mercé Jourdain
- Univ. Lille, Inserm, CHU Lille, Service de Médecine Intensive Réanimation, U1190, EGID, F-59000 Lille, France
| | - Dominique Deplanque
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France; University Lille, Inserm, CHU Lille, Centre d'investigation Clinique (CIC) 1403, F-59000, Lille, France; LICORNE Study Group, CHU Lille, Lille, France
| | - Annamaria Morelli
- Department of Experimental and Clinical Medicine, University of Florence, Italy
| | - Giulia Guarnieri
- Department of Experimental and Clinical Medicine, University of Florence, Italy
| | - Laurent Storme
- CHU Lille, Department of Neonatology, Hôpital Jeanne de Flandre, FHU 1000 Days for Health, F-59000, France
| | - Cyril Robil
- University Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - François Trottein
- University Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, U1019 - UMR 9017 - CIIL - Center for Infection and Immunity of Lille, F-59000 Lille, France
| | - Ruben Nogueiras
- CIMUS, Universidade de Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Pascal Pigny
- CHU Lille, Service de Biochimie et Hormonologie, Centre de Biologie Pathologie, Lille, France
| | - Julien Poissy
- LICORNE Study Group, CHU Lille, Lille, France; Univ. Lille, Inserm U1285, CHU Lille, Pôle de Réanimation, CNRS, UMR 8576 - UGSF - Unité de Glycobiologie Structurale et Fonctionnelle, F-59000, Lille, France
| | - Konstantina Chachlaki
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France
| | - Claude-Alain Maurage
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France; CHU Lille, Department of Pathology, Centre Biologie Pathologie, France; LICORNE Study Group, CHU Lille, Lille, France
| | - Paolo Giacobini
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France
| | - Waljit Dhillo
- Section of Endocrinology and Investigative Medicine, Imperial College London, London, United Kingdom; Department of Endocrinology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - S Rasika
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France.
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Lille Neuroscience & Cognition, UMR-S 1172, DistAlz, Lille, France.
| |
Collapse
|
23
|
Imoesi PI, Olarte-Sánchez CM, Croce L, Blaner WS, Morgan PJ, Heisler L, McCaffery P. Control by the brain of vitamin A homeostasis. iScience 2023; 26:107373. [PMID: 37599827 PMCID: PMC10432198 DOI: 10.1016/j.isci.2023.107373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 06/16/2023] [Accepted: 07/10/2023] [Indexed: 08/22/2023] Open
Abstract
Vitamin A is a micronutrient essential for vertebrate animals maintained in homeostatic balance in the body; however, little is known about the control of this balance. This study investigated whether the hypothalamus, a key integrative brain region, regulates vitamin A levels in the liver and circulation. Vitamin A in the form of retinol or retinoic acid was stereotactically injected into the 3rd ventricle of the rat brain. Alternatively, retinoids in the mouse hypothalamus were altered through retinol-binding protein 4 (Rbp4) gene knockdown. This led to rapid change in the liver proteins controlling vitamin A homeostasis as well as vitamin A itself in liver and the circulation. Prolonged disruption of Rbp4 in the region of the arcuate nucleus of the mouse hypothalamus altered retinol levels in the liver. This supports the concept that the brain may sense retinoids and influence whole-body vitamin A homeostasis with a possible "vitaminostatic" role.
Collapse
Affiliation(s)
- Peter I. Imoesi
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK
| | - Cristian M. Olarte-Sánchez
- Rowett Institute, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK
| | - Lorenzo Croce
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK
| | - William S. Blaner
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | - Peter J. Morgan
- Rowett Institute, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK
| | - Lora Heisler
- Rowett Institute, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK
| | - Peter McCaffery
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK
| |
Collapse
|
24
|
Jin K, Yao Z, van Velthoven CTJ, Kaplan ES, Glattfelder K, Barlow ST, Boyer G, Carey D, Casper T, Chakka AB, Chakrabarty R, Clark M, Departee M, Desierto M, Gary A, Gloe J, Goldy J, Guilford N, Guzman J, Hirschstein D, Lee C, Liang E, Pham T, Reding M, Ronellenfitch K, Ruiz A, Sevigny J, Shapovalova N, Shulga L, Sulc J, Torkelson A, Tung H, Levi B, Sunkin SM, Dee N, Esposito L, Smith K, Tasic B, Zeng H. Cell-type specific molecular signatures of aging revealed in a brain-wide transcriptomic cell-type atlas. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.26.550355. [PMID: 38168182 PMCID: PMC10760145 DOI: 10.1101/2023.07.26.550355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Biological aging can be defined as a gradual loss of homeostasis across various aspects of molecular and cellular function. Aging is a complex and dynamic process which influences distinct cell types in a myriad of ways. The cellular architecture of the mammalian brain is heterogeneous and diverse, making it challenging to identify precise areas and cell types of the brain that are more susceptible to aging than others. Here, we present a high-resolution single-cell RNA sequencing dataset containing ~1.2 million high-quality single-cell transcriptomic profiles of brain cells from young adult and aged mice across both sexes, including areas spanning the forebrain, midbrain, and hindbrain. We find age-associated gene expression signatures across nearly all 130+ neuronal and non-neuronal cell subclasses we identified. We detect the greatest gene expression changes in non-neuronal cell types, suggesting that different cell types in the brain vary in their susceptibility to aging. We identify specific, age-enriched clusters within specific glial, vascular, and immune cell types from both cortical and subcortical regions of the brain, and specific gene expression changes associated with cell senescence, inflammation, decrease in new myelination, and decreased vasculature integrity. We also identify genes with expression changes across multiple cell subclasses, pointing to certain mechanisms of aging that may occur across wide regions or broad cell types of the brain. Finally, we discover the greatest gene expression changes in cell types localized to the third ventricle of the hypothalamus, including tanycytes, ependymal cells, and Tbx3+ neurons found in the arcuate nucleus that are part of the neuronal circuits regulating food intake and energy homeostasis. These findings suggest that the area surrounding the third ventricle in the hypothalamus may be a hub for aging in the mouse brain. Overall, we reveal a dynamic landscape of cell-type-specific transcriptomic changes in the brain associated with normal aging that will serve as a foundation for the investigation of functional changes in the aging process and the interaction of aging and diseases.
Collapse
Affiliation(s)
- Kelly Jin
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Zizhen Yao
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | | | - Daniel Carey
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | - Max Departee
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Amanda Gary
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jessica Gloe
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jeff Goldy
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Changkyu Lee
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | | | | | - Josh Sevigny
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | - Josef Sulc
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Herman Tung
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Boaz Levi
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Nick Dee
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | | | | | - Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA, USA
| |
Collapse
|
25
|
Makrygianni EA, Chrousos GP. Neural Progenitor Cells and the Hypothalamus. Cells 2023; 12:1822. [PMID: 37508487 PMCID: PMC10378393 DOI: 10.3390/cells12141822] [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: 03/02/2023] [Revised: 05/22/2023] [Accepted: 06/02/2023] [Indexed: 07/30/2023] Open
Abstract
Neural progenitor cells (NPCs) are multipotent neural stem cells (NSCs) capable of self-renewing and differentiating into neurons, astrocytes and oligodendrocytes. In the postnatal/adult brain, NPCs are primarily located in the subventricular zone (SVZ) of the lateral ventricles (LVs) and subgranular zone (SGZ) of the hippocampal dentate gyrus (DG). There is evidence that NPCs are also present in the postnatal/adult hypothalamus, a highly conserved brain region involved in the regulation of core homeostatic processes, such as feeding, metabolism, reproduction, neuroendocrine integration and autonomic output. In the rodent postnatal/adult hypothalamus, NPCs mainly comprise different subtypes of tanycytes lining the wall of the 3rd ventricle. In the postnatal/adult human hypothalamus, the neurogenic niche is constituted by tanycytes at the floor of the 3rd ventricle, ependymal cells and ribbon cells (showing a gap-and-ribbon organization similar to that in the SVZ), as well as suprachiasmatic cells. We speculate that in the postnatal/adult human hypothalamus, neurogenesis occurs in a highly complex, exquisitely sophisticated neurogenic niche consisting of at least four subniches; this structure has a key role in the regulation of extrahypothalamic neurogenesis, and hypothalamic and extrahypothalamic neural circuits, partly through the release of neurotransmitters, neuropeptides, extracellular vesicles (EVs) and non-coding RNAs (ncRNAs).
Collapse
Affiliation(s)
- Evanthia A Makrygianni
- University Research Institute of Maternal and Child Health & Precision Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - George P Chrousos
- University Research Institute of Maternal and Child Health & Precision Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
26
|
Harris RBS. Low-dose peripheral leptin infusion produces selective activation of ventromedial hypothalamic and hindbrain STAT3. Am J Physiol Endocrinol Metab 2023; 325:E72-E82. [PMID: 37285599 PMCID: PMC10292972 DOI: 10.1152/ajpendo.00083.2023] [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: 03/20/2023] [Revised: 05/03/2023] [Accepted: 06/05/2023] [Indexed: 06/09/2023]
Abstract
Previous studies have shown that very low dose, acute, single peripheral leptin injections fully activate arcuate nucleus signal transducer and activator of transcription 3 (STAT3), but ventromedial hypothalamus (VMH) pSTAT3 continues to increase with higher doses of leptin that inhibit food intake. The lowest dose that inhibited intake increased circulating leptin 300-fold whereas food intake is inhibited by chronic peripheral leptin infusions that only double circulating leptin. This study examined whether the pattern of hypothalamic pSTAT3 was the same in leptin-infused rats as in leptin-injected rats. Male Sprague-Dawley rats received intraperitoneal infusions of 0, 5, 10, 20, or 40 µg leptin/day for 9 days. The highest dose of leptin increased serum leptin by 50-100%, inhibited food intake for 5 days, but inhibited weight gain and retroperitoneal fat mass for 9 days. Energy expenditure, respiratory exchange ratio, and brown fat temperature did not change. pSTAT3 was quantified in hypothalamic nuclei and the nucleus of the solitary tract (NTS) when food intake was inhibited and when it had returned to control levels. There was no effect of leptin on pSTAT3 in the medial or lateral arcuate nucleus or in the dorsomedial nucleus of the hypothalamus. VMH pSTAT3 was increased only at day 4 when food intake was inhibited, but NTS pSTAT3 was increased at both 4 and 9 days of infusion. These results suggest that activation of leptin VMH receptors contributes to the suppression of food intake, but that hindbrain receptors contribute to a sustained change in metabolism that maintains a reduced weight and fat mass.NEW & NOTEWORTHY Low-dose, chronic peripheral infusions of leptin produced an initial, transient inhibition of food intake that correlated with signal transducer and activator of transcription 3 (STAT3) activation in the ventromedial hypothalamus (VMH) and nucleus of the solitary tract (NTS). When intake normalized, but weight remained suppressed, the NTS was the only area that remained activated. These data suggest that leptin's primary function is to reduce body fat, that hypophagia is a means of achieving this and that different areas of the brain are responsible for the progressive response.
Collapse
Affiliation(s)
- Ruth B S Harris
- Center for Neuroinflammation and Cardiometabolic Disease, Georgia State University, Atlanta, Georgia, United States
| |
Collapse
|
27
|
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: 0] [Impact Index Per Article: 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
|
28
|
Pathak D, Sriram K. Neuron-astrocyte omnidirectional signaling in neurological health and disease. Front Mol Neurosci 2023; 16:1169320. [PMID: 37363320 PMCID: PMC10286832 DOI: 10.3389/fnmol.2023.1169320] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 05/09/2023] [Indexed: 06/28/2023] Open
Abstract
Astrocytes are an abundantly distributed population of glial cells in the central nervous system (CNS) that perform myriad functions in the normal and injured/diseased brain. Astrocytes exhibit heterogeneous phenotypes in response to various insults, a process known as astrocyte reactivity. The accuracy and precision of brain signaling are primarily based on interactions involving neurons, astrocytes, oligodendrocytes, microglia, pericytes, and dendritic cells within the CNS. Astrocytes have emerged as a critical entity within the brain because of their unique role in recycling neurotransmitters, actively modulating the ionic environment, regulating cholesterol and sphingolipid metabolism, and influencing cellular crosstalk in diverse neural injury conditions and neurodegenerative disorders. However, little is known about how an astrocyte functions in synapse formation, axon specification, neuroplasticity, neural homeostasis, neural network activity following dynamic surveillance, and CNS structure in neurological diseases. Interestingly, the tripartite synapse hypothesis came to light to fill some knowledge gaps that constitute an interaction of a subpopulation of astrocytes, neurons, and synapses. This review highlights astrocytes' role in health and neurological/neurodegenerative diseases arising from the omnidirectional signaling between astrocytes and neurons at the tripartite synapse. The review also recapitulates the disruption of the tripartite synapse with a focus on perturbations of the homeostatic astrocytic function as a key driver to modulate the molecular and physiological processes toward neurodegenerative diseases.
Collapse
|
29
|
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
|
30
|
Barbotin AL, Mimouni NEH, Kuchcinski G, Lopes R, Viard R, Rasika S, Mazur D, Silva MSB, Simon V, Boursier A, Pruvo JP, Yu Q, Candlish M, Boehm U, Bello FD, Medana C, Pigny P, Dewailly D, Prevot V, Catteau-Jonard S, Giacobini P. Hypothalamic neuroglial plasticity is regulated by anti-Müllerian hormone and disrupted in polycystic ovary syndrome. EBioMedicine 2023; 90:104535. [PMID: 37001236 PMCID: PMC10070524 DOI: 10.1016/j.ebiom.2023.104535] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 03/03/2023] [Accepted: 03/08/2023] [Indexed: 03/31/2023] Open
Abstract
BACKGROUND Polycystic ovary syndrome (PCOS) is the most common reproductive-endocrine disorder affecting between 5 and 18% of women worldwide. An elevated frequency of pulsatile luteinizing hormone (LH) secretion and higher serum levels of anti-Müllerian hormone (AMH) are frequently observed in women with PCOS. The origin of these abnormalities is, however, not well understood. METHODS We studied brain structure and function in women with and without PCOS using proton magnetic resonance spectroscopy (MRS) and diffusion tensor imaging combined with fiber tractography. Then, using a mouse model of PCOS, we investigated by electron microscopy whether AMH played a role on the regulation of hypothalamic structural plasticity. FINDINGS Increased AMH serum levels are associated with increased hypothalamic activity/axonal-glial signalling in PCOS patients. Furthermore, we demonstrate that AMH promotes profound micro-structural changes in the murine hypothalamic median eminence (ME), creating a permissive environment for GnRH secretion. These include the retraction of the processes of specialized AMH-sensitive ependymo-glial cells called tanycytes, allowing more GnRH neuron terminals to approach ME blood capillaries both during the run-up to ovulation and in a mouse model of PCOS. INTERPRETATION We uncovered a central function for AMH in the regulation of fertility by remodeling GnRH terminals and their tanycytic sheaths, and provided insights into the pivotal role of the brain in the establishment and maintenance of neuroendocrine dysfunction in PCOS. FUNDING INSERM (U1172), European Research Council (ERC) under the European Union's Horizon 2020 research and innovation program (grant agreement n° 725149), CHU de Lille, France (Bonus H).
Collapse
Affiliation(s)
- Anne-Laure Barbotin
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille F-59000, France; CHU Lille, Institut de Biologie de la Reproduction-Spermiologie-CECOS, Lille F-59000, France
| | - Nour El Houda Mimouni
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille F-59000, France
| | - Grégory Kuchcinski
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille F-59000, France; CHU Lille, Department of Neuroradiology, Lille F-59000, France
| | - Renaud Lopes
- CHU Lille, Department of Neuroradiology, Lille F-59000, France
| | - Romain Viard
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, US 41 - UAR 2014 - PLBS, Lille F-59000, France
| | - Sowmyalakshmi Rasika
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille F-59000, France
| | - Daniele Mazur
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille F-59000, France
| | - Mauro S B Silva
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille F-59000, France
| | - Virginie Simon
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille F-59000, France
| | - Angèle Boursier
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille F-59000, France; CHU Lille, Institut de Biologie de la Reproduction-Spermiologie-CECOS, Lille F-59000, France
| | | | - Qiang Yu
- Experimental Pharmacology, Center for Molecular Signalling (PZMS), Saarland University School of Medicine, Homburg 66123, Germany
| | - Michael Candlish
- Experimental Pharmacology, Center for Molecular Signalling (PZMS), Saarland University School of Medicine, Homburg 66123, Germany
| | - Ulrich Boehm
- Experimental Pharmacology, Center for Molecular Signalling (PZMS), Saarland University School of Medicine, Homburg 66123, Germany
| | - Federica Dal Bello
- Department of Molecular Biotechnology and Health Science, University of Turin, Turin 10125, Italy
| | - Claudio Medana
- Department of Molecular Biotechnology and Health Science, University of Turin, Turin 10125, Italy
| | - Pascal Pigny
- CHU Lille, Service de Biochimie et Hormonologie, Centre de Biologie Pathologie, Lille F-59000, France
| | - Didier Dewailly
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille F-59000, France
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille F-59000, France
| | - Sophie Catteau-Jonard
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille F-59000, France; CHU Lille, Service de Gynécologie Médicale, Hôpital Jeanne de Flandre, Lille F-59000, France
| | - Paolo Giacobini
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille F-59000, France.
| |
Collapse
|
31
|
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
|
32
|
Sewaybricker LE, Huang A, Chandrasekaran S, Melhorn SJ, Schur EA. The Significance of Hypothalamic Inflammation and Gliosis for the Pathogenesis of Obesity in Humans. Endocr Rev 2023; 44:281-296. [PMID: 36251886 DOI: 10.1210/endrev/bnac023] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 09/12/2022] [Indexed: 11/19/2022]
Abstract
Accumulated preclinical literature demonstrates that hypothalamic inflammation and gliosis are underlying causal components of diet-induced obesity in rodent models. This review summarizes and synthesizes available translational data to better understand the applicability of preclinical findings to human obesity and its comorbidities. The published literature in humans includes histopathologic analyses performed postmortem and in vivo neuroimaging studies measuring indirect markers of hypothalamic tissue microstructure. Both support the presence of hypothalamic inflammation and gliosis in children and adults with obesity. Findings predominantly point to tissue changes in the region of the arcuate nucleus of the hypothalamus, although findings of altered tissue characteristics in whole hypothalamus or other hypothalamic regions also emerged. Moreover, the severity of hypothalamic inflammation and gliosis has been related to comorbid conditions, including glucose intolerance, insulin resistance, type 2 diabetes, and low testosterone levels in men, independent of elevated body adiposity. Cross-sectional findings are augmented by a small number of prospective studies suggesting that a greater degree of hypothalamic inflammation and gliosis may predict adiposity gain and worsening insulin sensitivity in susceptible individuals. In conclusion, existing human studies corroborate a large preclinical literature demonstrating that hypothalamic neuroinflammatory responses play a role in obesity pathogenesis. Extensive or permanent hypothalamic tissue remodeling may negatively affect the function of neuroendocrine regulatory circuits and promote the development and maintenance of elevated body weight in obesity and/or comorbid endocrine disorders.
Collapse
Affiliation(s)
| | - Alyssa Huang
- Department of Pediatrics, University of Washington, Division of Endocrinology and Diabetes, Seattle Children's Hospital, Seattle, WA 98015, USA
| | | | - Susan J Melhorn
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
| | - Ellen A Schur
- Department of Medicine, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
33
|
The Role of Serum Anti-Mullerian Hormone Measurement in the Diagnosis of Polycystic Ovary Syndrome. Diagnostics (Basel) 2023; 13:diagnostics13050907. [PMID: 36900051 PMCID: PMC10000702 DOI: 10.3390/diagnostics13050907] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/18/2023] [Accepted: 02/20/2023] [Indexed: 03/06/2023] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrinological disorder in women with significant reproductive, metabolic, and psychological health implications. The lack of a specific diagnostic test poses challenges in making the diagnosis of PCOS, resulting in underdiagnosis and undertreatment. Anti-Mullerian hormone (AMH) synthesized by the pre-antral and small antral ovarian follicles appears to play an important role in the pathophysiology of PCOS, and serum AMH levels are often elevated in women with PCOS. The aim of this review is to inform the possibility of utilizing anti-Mullerian hormone either as a diagnostic test for PCOS or as an alternative diagnostic criterion in place of polycystic ovarian morphology, hyperandrogenism, and oligo-anovulation. Increased levels of serum AMH correlate highly with PCOS, polycystic ovarian morphology, hyperandrogenism, and oligo/amenorrhea. Additionally, serum AMH has high diagnostic accuracy as an isolated marker for PCOS or as a replacement for polycystic ovarian morphology.
Collapse
|
34
|
Dong S, Hou B, Yang C, Li Y, Sun B, Guo Y, Deng M, Liu D, Liu G. Comparative Hypothalamic Transcriptome Analysis Reveals Crucial mRNAs, lncRNAs, and circRNAs Affecting Litter Size in Goats. Genes (Basel) 2023; 14:444. [PMID: 36833370 PMCID: PMC9956962 DOI: 10.3390/genes14020444] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Litter size is an important indicator to measure the reproductive performance of goats, which is affected by the reproductive function of animals. The hypothalamus, as the regulatory center of the endocrine system, plays an important role in the reproduction of female animals. Here, we performed high-throughput RNA sequencing using hypothalamic tissue from high-fecundity and low-fecundity Leizhou goats to explore critical functional genes associated with litter size. Differentially expressed mRNA, lncRNA, and circRNAs were screened using DESeq and were enriched, and then analyzed by Gene Ontology and Kyoto Encyclopedia of Gene and Genome. Results showed that some of these differentially expressed mRNAs could be enriched in reproductive processes, jak-STAT, prolactin signaling pathway, and other signaling pathways related to reproduction, such as SOCS3. Furthermore, the central proteins POSTN, MFAP5, and DCN from protein-protein interaction may regulate animal reproductive activity by affecting cell proliferation and apoptosis. lncRNA MSTRG.33887.2 as well as circRNAs chicirc_098002, chicirc_072583, and chicirc_053531 may be able to influence animal reproduction by participating in folate metabolism and energy metabolism homeostasis through their respective target genes. Our results expand the molecular mechanism of hypothalamic regulation on animal reproduction.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Guangbin Liu
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
35
|
Sufieva DA, Korzhevskii DE. Proliferative Markers and Neural Stem Cells Markers in Tanycytes of the Third Cerebral Ventricle in Rats. Bull Exp Biol Med 2023; 174:564-570. [PMID: 36894817 DOI: 10.1007/s10517-023-05748-8] [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: 09/06/2022] [Indexed: 03/11/2023]
Abstract
The proliferative properties of tanycyte subpopulations during postnatal development and aging were studied. Using immunohistochemical markers, we described the distribution of proliferative markers and markers of neural stem cells (NSC) in 4 tanycyte subpopulations (α1-, α2-, β1-, and β2-tanycytes). During the first postnatal week, all tanycyte subpopulations exhibit proliferative activity. During aging, β-tanycytes lose their proliferative activity and retain a limited set of NSC markers, whereas α-tanycytes maintain both the ability to proliferate and the properties of NSC throughout the entire postnatal development including aging. The data obtained significantly improve modern understanding of the proliferative potential of tanycytes and their subpopulation differences in early postnatal period and during aging.
Collapse
Affiliation(s)
- D A Sufieva
- Institute of Experimental Medicine, St. Petersburg, Russia.
| | | |
Collapse
|
36
|
Haddad-Tóvolli R, Morari J, Barbizan R, Bóbbo VC, Carraro RS, Solon C, Dragano NR, Torsoni MA, Araujo EP, Velloso LA. Maternal obesity damages the median eminence blood-brain barrier structure and function in the progeny: the beneficial impact of cross-fostering by lean mothers. Am J Physiol Endocrinol Metab 2023; 324:E154-E166. [PMID: 36598900 DOI: 10.1152/ajpendo.00268.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Maternal obesity is an important risk factor for obesity, cardiovascular, and metabolic diseases in the offspring. Studies have shown that it leads to hypothalamic inflammation in the progeny, affecting the function of neurons regulating food intake and energy expenditure. In adult mice fed a high-fat diet, one of the hypothalamic abnormalities that contribute to the development of obesity is the damage of the blood-brain barrier (BBB) at the median eminence-arcuate nucleus (ME-ARC) interface; however, how the hypothalamic BBB is affected in the offspring of obese mothers requires further investigation. Here, we used confocal and transmission electron microscopy, transcript expression analysis, glucose tolerance testing, and a cross-fostering intervention to determine the impact of maternal obesity and breastfeeding on BBB integrity at the ME-ARC interface. The offspring of obese mothers were born smaller; conversely, at weaning, they presented larger body mass and glucose intolerance. In addition, maternal obesity-induced structural and functional damage of the offspring's ME-ARC BBB. By a cross-fostering intervention, some of the defects in barrier integrity and metabolism seen during development in an obesogenic diet were recovered. The offspring of obese dams breastfed by lean dams presented a reduction of body mass and glucose intolerance as compared to the offspring continuously exposed to an obesogenic environment during intrauterine and perinatal life; this was accompanied by partial recovery of the anatomical structure of the ME-ARC interface, and by the normalization of transcript expression of genes coding for hypothalamic neurotransmitters involved in energy balance and BBB integrity. Thus, maternal obesity promotes structural and functional damage of the hypothalamic BBB, which is, in part, reverted by lactation by lean mothers.NEW & NOTEWORTHY Maternal dietary habits directly influence offspring health. In this study, we aimed at determining the impact of maternal obesity on BBB integrity. We show that DIO offspring presented a leakier ME-BBB, accompanied by changes in the expression of transcripts encoding for endothelial and tanycytic proteins, as well as of hypothalamic neuropeptides. Breastfeeding in lean dams was sufficient to protect the offspring from ME-BBB disruption, providing a preventive strategy of nutritional intervention during early life.
Collapse
Affiliation(s)
- Roberta Haddad-Tóvolli
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas, Campinas, Brazil
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Joseane Morari
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas, Campinas, Brazil
| | - Roberta Barbizan
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas, Campinas, Brazil
| | - Vanessa C Bóbbo
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas, Campinas, Brazil
| | - Rodrigo S Carraro
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas, Campinas, Brazil
- Center for Anatomy Studies, University San Francisco (USF), Bragança Paulista, Brazil
| | - Carina Solon
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas, Campinas, Brazil
| | - Nathalia R Dragano
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas, Campinas, Brazil
| | - Márcio A Torsoni
- Laboratory of Metabolic Disorders, Faculty of Applied Sciences, State University of Campinas, Campinas, Limeira, Brazil
| | - Eliana P Araujo
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas, Campinas, Brazil
- School of Nursing, University of Campinas, Campinas, Brazil
| | - Licio A Velloso
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas, Campinas, Brazil
| |
Collapse
|
37
|
Fong H, Kurrasch DM. Developmental and functional relationships between hypothalamic tanycytes and embryonic radial glia. Front Neurosci 2023; 16:1129414. [PMID: 36741057 PMCID: PMC9895379 DOI: 10.3389/fnins.2022.1129414] [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: 12/22/2022] [Accepted: 12/31/2022] [Indexed: 01/21/2023] Open
Abstract
The hypothalamus is a key regulator of several homeostatic processes, such as circadian rhythms, energy balance, thirst, and thermoregulation. Recently, the hypothalamic third ventricle has emerged as a site of postnatal neurogenesis and gliogenesis. This hypothalamic neural stem potential resides in a heterogeneous population of cells known as tanycytes, which, not unlike radial glia, line the floor and ventrolateral walls of the third ventricle and extend a long process into the hypothalamic parenchyma. Here, we will review historical and recent data regarding tanycyte biology across the lifespan, focusing on the developmental emergence of these diverse cells from embryonic radial glia and their eventual role contributing to a fascinating, but relatively poorly characterized, adult neural stem cell niche.
Collapse
Affiliation(s)
- Harmony Fong
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada,Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB, Canada,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Deborah M. Kurrasch
- Department of Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada,Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB, Canada,Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada,*Correspondence: Deborah M. Kurrasch,
| |
Collapse
|
38
|
Juvenile social isolation affects the structure of the tanycyte-vascular interface in the hypophyseal portal system of the adult mice. Neurochem Int 2023; 162:105439. [PMID: 36356785 DOI: 10.1016/j.neuint.2022.105439] [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: 06/06/2022] [Revised: 10/26/2022] [Accepted: 11/01/2022] [Indexed: 11/09/2022]
Abstract
Accumulating evidence indicates that social stress in the juvenile period affects hypothalamic-pituitary-adrenal (HPA) axis activity in adulthood. The biological mechanisms underlying this phenomenon remain unclear. We aimed to elucidate them by comparing adult mice that had experienced social isolation from postnatal day 21-35 (juvenile social isolation (JSI) group) with those reared normally (control group). JSI group mice showed an attenuated HPA response to acute swim stress, while the control group had a normal response to this stress. Activity levels of the paraventricular nucleus in both groups were comparable, as shown by c-Fos immunoreactivities and mRNA expression of c-Fos, Corticotropin-releasing factor (CRF), Glucocorticoid receptor, and Mineralocorticoid receptor. We found greater vascular coverage by tanycytic endfeet in the median eminence of the JSI group mice than in that of the control group mice under basal condition and after acute swim stress. Moreover, CRF content after acute swim stress was greater in the median eminence of the JSI group mice than in that of the control group mice. The attenuated HPA response to acute swim stress was specific to JSI group mice, but not to control group mice. Although a direct link awaits further experiments, tanycyte morphological changes in the median eminence could be related to the HPA response.
Collapse
|
39
|
Bakker W, Imbernon M, Salinas CG, Moro Chao DH, Hassouna R, Morel C, Martin C, Leger C, Denis RG, Castel J, Peter A, Heni M, Maetzler W, Nielsen HS, Duquenne M, Schwaninger M, Lundh S, Johan Hogendorf WF, Gangarossa G, Secher A, Hecksher-Sørensen J, Pedersen TÅ, Prevot V, Luquet S. Acute changes in systemic glycemia gate access and action of GLP-1R agonist on brain structures controlling energy homeostasis. Cell Rep 2022; 41:111698. [PMID: 36417883 PMCID: PMC9715912 DOI: 10.1016/j.celrep.2022.111698] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 08/19/2022] [Accepted: 10/20/2022] [Indexed: 11/23/2022] Open
Abstract
Therapies based on glucagon-like peptide-1 (GLP-1) long-acting analogs and insulin are often used in the treatment of metabolic diseases. Both insulin and GLP-1 receptors are expressed in metabolically relevant brain regions, suggesting a cooperative action. However, the mechanisms underlying the synergistic actions of insulin and GLP-1R agonists remain elusive. In this study, we show that insulin-induced hypoglycemia enhances GLP-1R agonists entry in hypothalamic and area, leading to enhanced whole-body fat oxidation. Mechanistically, this phenomenon relies on the release of tanycyctic vascular endothelial growth factor A, which is selectively impaired after calorie-rich diet exposure. In humans, low blood glucose also correlates with enhanced blood-to-brain passage of insulin, suggesting that blood glucose gates the passage other energy-related signals in the brain. This study implies that the preventing hyperglycemia is important to harnessing the full benefit of GLP-1R agonist entry in the brain and action onto lipid mobilization and body weight loss.
Collapse
Affiliation(s)
- Wineke Bakker
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, 75013 Paris, France,Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark,Corresponding author
| | - Monica Imbernon
- University Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, EGID, UMR-S 1172, 59000 Lille, France
| | - Casper Gravesen Salinas
- Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark,Image Analysis & Computer Graphics, Department of Applied Mathematics and Computer Science, Technical University of Denmark, Kgs. Lyngby, Denmark,Gubra ApS, Hørsholm Kongevej 11B, 2970 Hørsholm, Denmark
| | | | - Rim Hassouna
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, 75013 Paris, France
| | - Chloe Morel
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, 75013 Paris, France
| | - Claire Martin
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, 75013 Paris, France
| | - Caroline Leger
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, 75013 Paris, France
| | - Raphael G.P. Denis
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, 75013 Paris, France,Institut Cochin, Université Paris Cité, INSERM U1016, CNRS UMR 8104, 75014 Paris, France
| | - Julien Castel
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, 75013 Paris, France
| | - Andreas Peter
- Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany,German Center for Diabetes Research (DZD), Tübingen, Germany,Institute for Clinical Chemistry and Pathobiochemistry, Department for Diagnostic Laboratory Medicine, University Hospital of Tübingen, Tübingen, Germany
| | - Martin Heni
- Department of Internal Medicine IV, Division of Endocrinology, Diabetology and Nephrology, University of Tübingen, Tübingen, Germany,Institute of Diabetes Research and Metabolic Diseases (IDM) of the Helmholtz Center Munich at the University of Tübingen, Tübingen, Germany,German Center for Diabetes Research (DZD), Tübingen, Germany
| | - Walter Maetzler
- Department of Neurodegenerative Diseases, Center of Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany,German Center for Neurodegenerative Diseases, Tübingen, Germany,Department of Neurology, University of Kiel, Kiel, Germany
| | | | - Manon Duquenne
- University Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, EGID, UMR-S 1172, 59000 Lille, France
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Sofia Lundh
- Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
| | | | - Giuseppe Gangarossa
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, 75013 Paris, France
| | - Anna Secher
- Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark
| | - Jacob Hecksher-Sørensen
- Global Drug Discovery, Novo Nordisk A/S, Måløv, Denmark,Gubra ApS, Hørsholm Kongevej 11B, 2970 Hørsholm, Denmark
| | | | - Vincent Prevot
- University Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, EGID, UMR-S 1172, 59000 Lille, France
| | - Serge Luquet
- Université Paris Cité, CNRS, Unité de Biologie Fonctionnelle et Adaptative, 75013 Paris, France,Corresponding author
| |
Collapse
|
40
|
Dardente H, Lomet D, Desmarchais A, Téteau O, Lasserre O, Gonzalez AA, Dubois E, Beltramo M, Elis S. Impact of food restriction on the medio-basal hypothalamus of intact ewes as revealed by a large-scale transcriptomics study. J Neuroendocrinol 2022; 34:e13198. [PMID: 36168278 DOI: 10.1111/jne.13198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 08/25/2022] [Accepted: 08/28/2022] [Indexed: 11/27/2022]
Abstract
In mammals, the medio-basal hypothalamus (MBH) integrates photoperiodic and food-related cues to ensure timely phasing of physiological functions, including seasonal reproduction. The current human epidemics of obesity and associated reproductive disorders exemplifies the tight link between metabolism and reproduction. Yet, how food-related cues impact breeding at the level of the MBH remains unclear. In this respect, the sheep, which is a large diurnal mammal with a marked dual photoperiodic/metabolic control of seasonal breeding, is a relevant model. Here, we present a large-scale study in ewes (n = 120), which investigated the impact of food restriction (FRes) on the MBH transcriptome using unbiased RNAseq, followed by RT-qPCR. Few genes (~100) were impacted by FRes and the transcriptional impact was very modest (<2-fold increase or < 50% decrease for most genes). As anticipated, FRes increased expression of Npy/AgRP/LepR and decreased expression of Pomc/Cartpt, while Kiss1 expression was not impacted. Of particular interest, Eya3, Nmu and Dio2, genes involved in photoperiodic decoding within the MBH, were also affected by FRes. Finally, we also identified a handful of genes not known to be regulated by food-related cues (e.g., RNase6, HspA6, Arrdc2). In conclusion, our transcriptomics study provides insights into the impact of metabolism on the MBH in sheep, which may be relevant to human, and identifies possible molecular links between metabolism and (seasonal) reproduction.
Collapse
Affiliation(s)
- Hugues Dardente
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| | - Didier Lomet
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| | | | - Ophélie Téteau
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| | | | - Anne-Alicia Gonzalez
- MGX-Montpellier GenomiX, Université Montpellier, CNRS, INSERM, Montpellier, France
| | - Emeric Dubois
- MGX-Montpellier GenomiX, Université Montpellier, CNRS, INSERM, Montpellier, France
| | | | - Sébastien Elis
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| |
Collapse
|
41
|
Just N, Chevillard PM, Migaud M. Imaging and spectroscopic methods to investigate adult neurogenesis in vivo: New models and new avenues. Front Neurosci 2022; 16:933947. [PMID: 35992937 PMCID: PMC9389108 DOI: 10.3389/fnins.2022.933947] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 07/11/2022] [Indexed: 11/25/2022] Open
Abstract
Adult neurogenesis (AN) can be defined as the birth and development of new neurons in adulthood. Until the 1990s, AN was deemed not to happen after birth. Gradually, several groups demonstrated that specific zones of the brain of various species had a neurogenic potential. AN could be the key to treating a large range of neurodegenerative, neuropsychiatric, and metabolic diseases, with a better understanding of the mechanisms allowing for regeneration of new neurons. Despite this promising prospect, the existence of AN has not been validated in vivo in humans and therefore remains controversial. Moreover, the weight of AN-induced plasticity against other mechanisms of brain plasticity is not known, adding to the controversy. In this review, we would like to show that recent technical advances in brain MR imaging methods combined with improved models can resolve the debate.
Collapse
Affiliation(s)
- Nathalie Just
- Danish Research Centre for Magnetic Resonance, Center for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital Amager og Hvidovre, Hvidovre, Denmark
- Physiologie de la Reproduction et des Comportements, Centre INRAE Val de Loire, CNRS, IFCE, INRAE, and Université de Tours, Nouzilly, France
- *Correspondence: Nathalie Just
| | - Pierre-Marie Chevillard
- Physiologie de la Reproduction et des Comportements, Centre INRAE Val de Loire, CNRS, IFCE, INRAE, and Université de Tours, Nouzilly, France
| | - Martine Migaud
- Physiologie de la Reproduction et des Comportements, Centre INRAE Val de Loire, CNRS, IFCE, INRAE, and Université de Tours, Nouzilly, France
| |
Collapse
|
42
|
Tanycytes control hypothalamic liraglutide uptake and its anti-obesity actions. Cell Metab 2022; 34:1054-1063.e7. [PMID: 35716660 PMCID: PMC7613793 DOI: 10.1016/j.cmet.2022.06.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 08/08/2021] [Accepted: 06/01/2022] [Indexed: 11/24/2022]
Abstract
Liraglutide, an anti-diabetic drug and agonist of the glucagon-like peptide one receptor (GLP1R), has recently been approved to treat obesity in individuals with or without type 2 diabetes. Despite its extensive metabolic benefits, the mechanism and site of action of liraglutide remain unclear. Here, we demonstrate that liraglutide is shuttled to target cells in the mouse hypothalamus by specialized ependymoglial cells called tanycytes, bypassing the blood-brain barrier. Selectively silencing GLP1R in tanycytes or inhibiting tanycytic transcytosis by botulinum neurotoxin expression not only hampers liraglutide transport into the brain and its activation of target hypothalamic neurons, but also blocks its anti-obesity effects on food intake, body weight and fat mass, and fatty acid oxidation. Collectively, these striking data indicate that the liraglutide-induced activation of hypothalamic neurons and its downstream metabolic effects are mediated by its tanycytic transport into the mediobasal hypothalamus, strengthening the notion of tanycytes as key regulators of metabolic homeostasis.
Collapse
|
43
|
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: 7.5] [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
|
44
|
Pena-Leon V, Folgueira C, Barja-Fernández S, Pérez-Lois R, Da Silva Lima N, Martin M, Heras V, Martinez-Martinez S, Valero P, Iglesias C, Duquenne M, Al-Massadi O, Beiroa D, Souto Y, Fidalgo M, Sowmyalakshmi R, Guallar D, Cunarro J, Castelao C, Senra A, González-Saenz P, Vázquez-Cobela R, Leis R, Sabio G, Mueller-Fielitz H, Schwaninger M, López M, Tovar S, Casanueva FF, Valjent E, Diéguez C, Prevot V, Nogueiras R, Seoane LM. Prolonged breastfeeding protects from obesity by hypothalamic action of hepatic FGF21. Nat Metab 2022; 4:901-917. [PMID: 35879461 PMCID: PMC9314260 DOI: 10.1038/s42255-022-00602-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 06/08/2022] [Indexed: 12/25/2022]
Abstract
Early-life determinants are thought to be a major factor in the rapid increase of obesity. However, while maternal nutrition has been extensively studied, the effects of breastfeeding by the infant on the reprogramming of energy balance in childhood and throughout adulthood remain largely unknown. Here we show that delayed weaning in rat pups protects them against diet-induced obesity in adulthood, through enhanced brown adipose tissue thermogenesis and energy expenditure. In-depth metabolic phenotyping in this rat model as well as in transgenic mice reveals that the effects of prolonged suckling are mediated by increased hepatic fibroblast growth factor 21 (FGF21) production and tanycyte-controlled access to the hypothalamus in adulthood. Specifically, FGF21 activates GABA-containing neurons expressing dopamine receptor 2 in the lateral hypothalamic area and zona incerta. Prolonged breastfeeding thus constitutes a protective mechanism against obesity by affecting long-lasting physiological changes in liver-to-hypothalamus communication and hypothalamic metabolic regulation.
Collapse
Affiliation(s)
- Veronica Pena-Leon
- Endocrine Physiopathology Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Complejo Hospitalario Universitario de Santiago/SERGAS, Santiago de Compostela, Spain
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Cintia Folgueira
- Endocrine Physiopathology Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Complejo Hospitalario Universitario de Santiago/SERGAS, Santiago de Compostela, Spain
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
- CIBEROBN Physiopathology of Obesity and Nutrition, ISCIII, Santiago de Compostela, Spain
| | - Silvia Barja-Fernández
- Endocrine Physiopathology Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Complejo Hospitalario Universitario de Santiago/SERGAS, Santiago de Compostela, Spain
| | - Raquel Pérez-Lois
- Endocrine Physiopathology Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Complejo Hospitalario Universitario de Santiago/SERGAS, Santiago de Compostela, Spain
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Natália Da Silva Lima
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Marion Martin
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, Inserm, University of Lille, Lille, France
| | - Violeta Heras
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Sara Martinez-Martinez
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Paola Valero
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Cristina Iglesias
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Mannon Duquenne
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, Inserm, University of Lille, Lille, France
| | - Omar Al-Massadi
- Endocrine Physiopathology Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Complejo Hospitalario Universitario de Santiago/SERGAS, Santiago de Compostela, Spain
- CIBEROBN Physiopathology of Obesity and Nutrition, ISCIII, Santiago de Compostela, Spain
| | - Daniel Beiroa
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Yara Souto
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Miguel Fidalgo
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Rasika Sowmyalakshmi
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, Inserm, University of Lille, Lille, France
| | - Diana Guallar
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Juan Cunarro
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Cecilia Castelao
- Endocrine Physiopathology Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Complejo Hospitalario Universitario de Santiago/SERGAS, Santiago de Compostela, Spain
- CIBEROBN Physiopathology of Obesity and Nutrition, ISCIII, Santiago de Compostela, Spain
| | - Ana Senra
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
| | - Patricia González-Saenz
- Endocrine Physiopathology Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Complejo Hospitalario Universitario de Santiago/SERGAS, Santiago de Compostela, Spain
| | - Rocío Vázquez-Cobela
- Pediatrics Department, GI Pediatric Nutrition, Galicia Research Unit for Development, Growth and Human Nutrition, Complejo Hospitalario Universitario de Santiago, Santiago de Compostela, Spain
| | - Rosaura Leis
- Pediatrics Department, GI Pediatric Nutrition, Galicia Research Unit for Development, Growth and Human Nutrition, Complejo Hospitalario Universitario de Santiago, Santiago de Compostela, Spain
| | | | - Helge Mueller-Fielitz
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Miguel López
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
- CIBEROBN Physiopathology of Obesity and Nutrition, ISCIII, Santiago de Compostela, Spain
| | - Sulay Tovar
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
- CIBEROBN Physiopathology of Obesity and Nutrition, ISCIII, Santiago de Compostela, Spain
| | - Felipe F Casanueva
- CIBEROBN Physiopathology of Obesity and Nutrition, ISCIII, Santiago de Compostela, Spain
| | - Emmanuel Valjent
- IGF, University of Montpellier, CNRS, Inserm, Montpellier, France
| | - Carlos Diéguez
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain
- CIBEROBN Physiopathology of Obesity and Nutrition, ISCIII, Santiago de Compostela, Spain
| | - Vincent Prevot
- Laboratory of Development and Plasticity of the Neuroendocrine Brain, Inserm, University of Lille, Lille, France
| | - Rubén Nogueiras
- CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria de Santiago de Compostela, Santiago de Compostela, Spain.
- CIBEROBN Physiopathology of Obesity and Nutrition, ISCIII, Santiago de Compostela, Spain.
- Galician Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, Spain.
| | - Luisa M Seoane
- Endocrine Physiopathology Group, Instituto de Investigación Sanitaria de Santiago de Compostela, Complejo Hospitalario Universitario de Santiago/SERGAS, Santiago de Compostela, Spain.
- CIBEROBN Physiopathology of Obesity and Nutrition, ISCIII, Santiago de Compostela, Spain.
| |
Collapse
|
45
|
Desroziers E. Unusual suspects: Glial cells in fertility regulation and their suspected role in polycystic ovary syndrome. J Neuroendocrinol 2022; 34:e13136. [PMID: 35445462 PMCID: PMC9489003 DOI: 10.1111/jne.13136] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 11/28/2022]
Abstract
Gonadotropin-releasing-hormone (GnRH) neurons sitting within the hypothalamus control the production of gametes and sex steroids by the gonads, therefore ensuring survival of species. As orchestrators of reproductive function, GnRH neurons integrate information from external and internal cues. This occurs through an extensively studied neuronal network known as the "GnRH neuronal network." However, the brain is not simply composed of neurons. Evidence suggests a role for glial cells in controlling GnRH neuron activity, secretion and fertility outcomes, although numerous questions remain. Glial cells have historically been seen as support cells for neurons. This idea has been challenged by the discovery that some neurological diseases originate from glial dysfunction. The prevalence of infertility disorders is increasing worldwide, with one in four couples being affected; therefore, it remains essential to understand the mechanisms by which the brain controls fertility. The "GnRH glial network" could be a major player in infertility disorders and represent a potential therapeutic target. In polycystic ovary syndrome (PCOS), the most common infertility disorder of reproductive aged women worldwide, the brain is considered a prime suspect. Recent studies have demonstrated pathological neuronal wiring of the "GnRH neuronal network" in PCOS-like animal models. However, the role of the "GnRH glial network" remains to be elucidated. In this review, I aim to propose glial cells as unusual suspects in infertility disorders such as PCOS. In the first part, I state our current knowledge about the role of glia in the regulation of GnRH neurons and fertility. In the second part, based on our recent findings, I discuss how glial cells could be implicated in PCOS pathology.
Collapse
Affiliation(s)
- Elodie Desroziers
- Department of Physiology, Centre for NeuroendocrinologyUniversity of OtagoDunedinNew Zealand
- Sorbonne Université, CNRS, INSERM, Neuroscience Paris Seine – Institut de Biologie Paris Seine, Neuroplasticity of Reproductive Behaviours TeamParisFrance
| |
Collapse
|
46
|
Hypothalamic remodeling of thyroid hormone signaling during hibernation in the arctic ground squirrel. Commun Biol 2022; 5:492. [PMID: 35606540 PMCID: PMC9126913 DOI: 10.1038/s42003-022-03431-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 04/29/2022] [Indexed: 12/16/2022] Open
Abstract
Hibernation involves prolonged intervals of profound metabolic suppression periodically interrupted by brief arousals to euthermy, the function of which is unknown. Annual cycles in mammals are timed by a photoperiodically-regulated thyroid-hormone-dependent mechanism in hypothalamic tanycytes, driven by thyrotropin (TSH) in the pars tuberalis (PT), which regulates local TH-converting deiodinases and triggers remodeling of neuroendocrine pathways. We demonstrate that over the course of hibernation in continuous darkness, arctic ground squirrels (Urocitellus parryii) up-regulate the retrograde TSH/Deiodinase/TH pathway, remodel hypothalamic tanycytes, and activate the reproductive axis. Forcing the premature termination of hibernation by warming animals induced hypothalamic deiodinase expression and the accumulation of secretory granules in PT thyrotrophs and pituitary gonadotrophs, but did not further activate the reproductive axis. We suggest that periodic arousals may allow for the transient activation of hypothalamic thyroid hormone signaling, cellular remodeling, and re-programming of brain circuits in preparation for the short Arctic summer. Arctic ground squirrels hibernating in darkness activate the pars tuberalis - hypothalamus thyroid hormone signaling pathway, remodel hypothalamic tanycytes, and activate the reproductive axis.
Collapse
|
47
|
Dardente H, Simonneaux V. GnRH and the photoperiodic control of seasonal reproduction: Delegating the task to kisspeptin and RFRP-3. J Neuroendocrinol 2022; 34:e13124. [PMID: 35384117 DOI: 10.1111/jne.13124] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 02/22/2022] [Accepted: 03/03/2022] [Indexed: 10/18/2022]
Abstract
Synchronization of mammalian breeding activity to the annual change of photoperiod and environmental conditions is of the utmost importance for individual survival and species perpetuation. Subsequent to the early 1960s, when the central role of melatonin in this adaptive process was demonstrated, our comprehension of the mechanisms through which light regulates gonadal activity has increased considerably. The current model for the photoperiodic neuroendocrine system points to pivotal roles for the melatonin-sensitive pars tuberalis (PT) and its seasonally-regulated production of thyroid-stimulating hormone (TSH), as well as for TSH-sensitive hypothalamic tanycytes, radial glia-like cells located in the basal part of the third ventricle. Tanycytes respond to TSH through increased expression of thyroid hormone (TH) deiodinase 2 (Dio2), which leads to heightened production of intrahypothalamic triiodothyronine (T3) during longer days of spring and summer. There is strong evidence that this local, long-day driven, increase in T3 links melatonin input at the PT to gonadotropin-releasing hormone (GnRH) output, to align breeding with the seasons. The mechanism(s) through which T3 impinges upon GnRH remain(s) unclear. However, two distinct neuronal populations of the medio-basal hypothalamus, which express the (Arg)(Phe)-amide peptides kisspeptin and RFamide-related peptide-3, appear to be well-positioned to relay this seasonal T3 message towards GnRH neurons. Here, we summarize our current understanding of the cellular, molecular and neuroendocrine players, which keep track of photoperiod and ultimately govern GnRH output and seasonal breeding.
Collapse
Affiliation(s)
- Hugues Dardente
- CNRS, IFCE, INRAE, Université de Tours, PRC, Nouzilly, France
| | - Valérie Simonneaux
- Institute for Cellular and Integrative Neuroscience, University of Strasbourg, Strasbourg, France
| |
Collapse
|
48
|
Prevot V, Sharif A. The polygamous GnRH neuron: Astrocytic and tanycytic communication with a neuroendocrine neuronal population. J Neuroendocrinol 2022; 34:e13104. [PMID: 35233849 DOI: 10.1111/jne.13104] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 01/12/2022] [Accepted: 01/30/2022] [Indexed: 11/28/2022]
Abstract
To ensure the survival of the species, hypothalamic neuroendocrine circuits controlling fertility, which converge onto neurons producing gonadotropin-releasing hormone (GnRH), must respond to fluctuating physiological conditions by undergoing rapid and reversible structural and functional changes. However, GnRH neurons do not act alone, but through reciprocal interactions with multiple hypothalamic cell populations, including several glial and endothelial cell types. For instance, it has long been known that in the hypothalamic median eminence, where GnRH axons terminate and release their neurohormone into the pituitary portal blood circulation, morphological plasticity displayed by distal processes of tanycytes modifies their relationship with adjacent neurons as well as the spatial properties of the neurohemal junction. These alterations not only regulate the capacity of GnRH neurons to release their neurohormone, but also the activation of discrete non-neuronal pathways that mediate feedback by peripheral hormones onto the hypothalamus. Additionally, a recent breakthrough has demonstrated that GnRH neurons themselves orchestrate the establishment of their neuroendocrine circuitry during postnatal development by recruiting an entourage of newborn astrocytes that escort them into adulthood and, via signalling through gliotransmitters such as prostaglandin E2, modulate their activity and GnRH release. Intriguingly, several environmental and behavioural toxins perturb these neuron-glia interactions and consequently, reproductive maturation and fertility. Deciphering the communication between GnRH neurons and other neural cell types constituting hypothalamic neuroendocrine circuits is thus critical both to understanding physiological processes such as puberty, oestrous cyclicity and aging, and to developing novel therapeutic strategies for dysfunctions of these processes, including the effects of endocrine disruptors.
Collapse
Affiliation(s)
- Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, FHU 1000 Days for Health, Lille, France
| | - Ariane Sharif
- Univ. Lille, Inserm, CHU Lille, Laboratory of Development and Plasticity of the Neuroendocrine Brain, Lille Neuroscience & Cognition, UMR-S1172, FHU 1000 Days for Health, Lille, France
| |
Collapse
|
49
|
Socol CT, Chira A, Martinez-Sanchez MA, Nuñez-Sanchez MA, Maerescu CM, Mierlita D, Rusu AV, Ruiz-Alcaraz AJ, Trif M, Ramos-Molina B. Leptin Signaling in Obesity and Colorectal Cancer. Int J Mol Sci 2022; 23:4713. [PMID: 35563103 PMCID: PMC9102849 DOI: 10.3390/ijms23094713] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 04/19/2022] [Accepted: 04/22/2022] [Indexed: 12/05/2022] Open
Abstract
Obesity and colorectal cancer (CRC) are among the leading diseases causing deaths in the world, showing a complex multifactorial pathology. Obesity is considered a risk factor in CRC development through inflammation, metabolic, and signaling processes. Leptin is one of the most important adipokines related to obesity and an important proinflammatory marker, mainly expressed in adipose tissue, with many genetic variation profiles, many related influencing factors, and various functions that have been ascribed but not yet fully understood and elucidated, the most important ones being related to energy metabolism, as well as endocrine and immune systems. Aberrant signaling and genetic variations of leptin are correlated with obesity and CRC, with the genetic causality showing both inherited and acquired events, in addition to lifestyle and environmental risk factors; these might also be related to specific pathogenic pathways at different time points. Moreover, mutation gain is a crucial factor enabling the genetic process of CRC. Currently, the inconsistent and insufficient data related to leptin's relationship with obesity and CRC indicate the necessity of further related studies. This review summarizes the current knowledge on leptin genetics and its potential relationship with the main pathogenic pathways of obesity and CRC, in an attempt to understand the molecular mechanisms of these associations, in the context of inconsistent and contradictory data. The understanding of these mechanisms linking obesity and CRC could help to develop novel therapeutic targets and prevention strategies, resulting in a better prognosis and management of these diseases.
Collapse
Affiliation(s)
| | - Alexandra Chira
- 2nd Medical Clinic, Department of Internal Medicine, “Iuliu Hatieganu” University of Medicine and Pharmacy, 400006 Cluj-Napoca, Romania;
| | - Maria Antonia Martinez-Sanchez
- Obesity and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB), 30120 Murcia, Spain; (M.A.M.-S.); (M.A.N.-S.)
| | - Maria Angeles Nuñez-Sanchez
- Obesity and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB), 30120 Murcia, Spain; (M.A.M.-S.); (M.A.N.-S.)
| | | | - Daniel Mierlita
- Department of Nutrition, University of Oradea, 410048 Oradea, Romania;
| | - Alexandru Vasile Rusu
- Life Science Institute, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, 400372 Cluj-Napoca, Romania;
- Faculty of Animal Science and Biotechnology, University of Agricultural Sciences and Veterinary Medicine Cluj-Napoca, 400372 Cluj-Napoca, Romania
| | - Antonio Jose Ruiz-Alcaraz
- Department of Biochemistry and Molecular B and Immunology, Faculty of Medicine, University of Murcia, 30100 Murcia, Spain;
| | - Monica Trif
- Department of Food Research, Centiv GmbH, 28857 Syke, Germany;
| | - Bruno Ramos-Molina
- Obesity and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB), 30120 Murcia, Spain; (M.A.M.-S.); (M.A.N.-S.)
| |
Collapse
|
50
|
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
|