1
|
Dali R, Langlet F. Tanycytes in the nexus of hypothalamic inflammation, appetite control, and obesity. Physiol Behav 2025; 296:114917. [PMID: 40222438 DOI: 10.1016/j.physbeh.2025.114917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/16/2025] [Accepted: 04/10/2025] [Indexed: 04/15/2025]
Abstract
Hypothalamic inflammation has been identified as a critical factor driving the development of obesity and associated metabolic disorders. This inflammation-related disruption of energy balance relies on alterations in metabolic cues sensing and hypothalamic cellular functions, together leading to overeating and weight gain. Within the hypothalamic cellular networks controlling energy balance, recent studies have highlighted the significance of glial dysfunction in these processes, suggesting that these cells could provide new avenues for weight loss therapies. Glia rapidly activates following the consumption of a high-fat diet, even after a very short exposure, and contributes to the disruption of the entire system through inflammatory crosstalk. This review explores recent progress in understanding the molecular interactions between glial cells and neurons in hypothalamic inflammation related to obesity, diabetes, and associated complications. Notably, it highlights specialized ependymal cells called tanycytes, whose role is still underestimated in hypothalamic inflammation, and examines the potential for targeting this cell type as a treatment strategy for metabolic disorders.
Collapse
Affiliation(s)
- Rafik Dali
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland; Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Fanny Langlet
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland; Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
2
|
Franklin Z, Hull C, Delibegovic M, Platt B. Pharmacological PTP1B inhibition rescues motor learning, neuroinflammation, and hyperglycaemia in a mouse model of Alzheimer's disease. Exp Neurol 2025; 385:115115. [PMID: 39672227 DOI: 10.1016/j.expneurol.2024.115115] [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: 09/10/2024] [Revised: 11/20/2024] [Accepted: 12/09/2024] [Indexed: 12/15/2024]
Abstract
BACKGROUND Patients with Alzheimer's Disease (AD) frequently suffer from comorbidities such as type 2 diabetes mellitus (T2DM), accompanied by shared common pathologies such as increased inflammation and impaired glucose homeostasis. Beta-secretase 1 (BACE1), the rate limiting enzyme in AD associated beta-amyloid (Aβ) production, is also implicated in metabolic dysfunction and can increase central and peripheral protein levels of protein tyrosine phosphatase 1B (PTP1B). PTP1B is a validated target in diabetes and obesity, and is a neuroinflammatory regulator involved in degenerative processes. This study investigated the effects of the PTP1B inhibitor, trodusquemine (MSI-1436) on the cognitive and metabolic phenotypes of the neuronal human BACE1 knock-in (PLB4) mouse, a co-morbidity model of AD and T2DM, and their wild-type (PLBWT) controls. METHODS Five-month-old male PLB4 and PLBWT mice received PTP1B inhibitor treatment (1 mg/kg intraperitoneal injection; 5 weeks). Activity and spatial habituation (Phenotyper), motor learning (RotaRod), glucose tolerance, and brain and liver molecular analyses were analysed following treatment. RESULTS Inhibition of PTP1B improved motor learning alongside glucose tolerance in PLB4 mice, without affecting body weight/adiposity. MSI-1436 treatment led to lower protein levels of amyloid precursor protein (APP), reduced astrogliosis and restoration of the endoplasmic chaperone immunoglobulin heavy chain binding protein (BIP) in the brain, alongside decreased insulin receptor substrate-1 (IRS1) and dipeptidyl peptidase-4 (DPP4) proteins in the liver. CONCLUSION We provide evidence that neuronal BACE1 contributes to neuroinflammation and hyperglycaemia in PLB4 mice, and this can be partially rescued by PTP1B inhibition. Targeting PTP1B may therefore offer an attractive therapeutic approach to ameliorate co-morbidity associated pathologies in AD and T2DM.
Collapse
Affiliation(s)
- Zara Franklin
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, UK.
| | - Claire Hull
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| | - Mirela Delibegovic
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, UK; Aberdeen Cardiovascular and Diabetes Centre, Foresterhill, University of Aberdeen, Aberdeen AB25 2ZD, UK
| | - Bettina Platt
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, Foresterhill, University of Aberdeen, Aberdeen, AB25 2ZD, UK
| |
Collapse
|
3
|
Heinken A, Asara JM, Gnanaguru G, Singh C. Systemic regulation of retinal medium-chain fatty acid oxidation repletes TCA cycle flux in oxygen-induced retinopathy. Commun Biol 2025; 8:25. [PMID: 39789310 PMCID: PMC11718186 DOI: 10.1038/s42003-024-07394-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: 06/17/2024] [Accepted: 12/10/2024] [Indexed: 01/12/2025] Open
Abstract
Activation of anaplerosis takes away glutamine from the biosynthetic pathways to the energy-producing TCA cycle. Especially, induction of hyperoxia driven anaplerosis in neurovascular tissues such as the retina during early stages of development could deplete biosynthetic precursors from newly proliferating endothelial cells impeding physiological angiogenesis and leading to vasoobliteration. Using an oxygen-induced retinopathy (OIR) mouse model, we investigated the metabolic differences between OIR-resistant BALB/cByJ and OIR susceptible C57BL/6J strains at system levels to understand the molecular underpinnings that potentially contribute to hyperoxia-induced vascular abnormalities in the neural retina. Our systems level in vivo RNA-seq, proteomics, and lipidomic profiling and ex-vivo retinal explant studies show that the medium-chain fatty acids serves as an alternative source to feed the TCA cycle. Our findings strongly implicate that medium-chain fatty acids could suppress glutamine-fueled anaplerosis and ameliorate hyperoxia-induced vascular abnormalities in conditions such as retinopathy of prematurity.
Collapse
Affiliation(s)
- Almut Heinken
- Inserm UMRS 1256 NGERE, University of Lorraine, Nancy, France
| | - John M Asara
- Division of Signal Transduction/Mass Spectrometry Core, Beth Israel Deaconess Medical Center, Boston, MA, 02115, USA
- Department of Medicine, Harvard Medical School, Boston, MA, 02115, USA
| | - Gopalan Gnanaguru
- Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, 02111, USA
| | - Charandeep Singh
- Department of Ophthalmology, Tufts Medical Center, Tufts University School of Medicine, Boston, MA, 02111, USA.
- Division of Biochemical and Molecular Nutrition, Gerald J. and Dorothy R. Friedman School of Nutrition Science and Policy, Tufts University, Boston, MA, 02111, USA.
| |
Collapse
|
4
|
Zhang Y, Wang Y, Li Y, Pang J, Höhn A, Dong W, Gao R, Liu Y, Wang D, She Y, Guo R, Liu Z. Methionine restriction alleviates diabetes-associated cognitive impairment via activation of FGF21. Redox Biol 2024; 77:103390. [PMID: 39383602 PMCID: PMC11492615 DOI: 10.1016/j.redox.2024.103390] [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: 09/08/2024] [Revised: 09/28/2024] [Accepted: 10/06/2024] [Indexed: 10/11/2024] Open
Abstract
Glucose metabolism disturbances may result in diabetes-associated cognitive decline (DACI). Methionine restriction (MR) diet has emerged as a potential dietary strategy for managing glucose homeostasis. However, the effects and underlying mechanisms of MR on DACI have not been fully elucidated. Here, we found that a 13-week MR (0.17 % methionine, w/w) intervention starting at 8 weeks of age improved peripheral insulin sensitivity in male db/db mice, a model for type 2 diabetes. Notably, MR significantly improved working as well as long-term memory in db/db mice, accompanied by increased PSD-95 level and reduced neuroinflammatory factors, malondialdehyde (MDA), and 8-hydroxy-2'-deoxyguanosine (8-OHdG). We speculate that this effect may be mediated by MR activating hepatic fibroblast growth factor 21 (FGF21) and the brain FGFR1/AMPK/GLUT4 signaling pathway to enhance brain glucose metabolism. To further delineate the mechanism, we used intracerebroventricular injection of adeno-associated virus to specifically knock down FGFR1 in the brain to verify the role of FGFR1 in MR-mediated DACI. It was found that the positive effects of MR on DACI were offset, reflected in decreased cognitive function, impaired synaptic plasticity, upregulated neuroinflammation, and balanced enzymes regulating reactive oxygen species (Sod1, Sod2, Nox4). Of note, the FGFR1/AMPK/GLUT4 signaling pathway and brain glucose metabolism were inhibited. In summary, our study demonstrated that MR increased peripheral insulin sensitivity, activated brain FGFR1/AMPK/GLUT4 signaling through FGF21, maintained normal glucose metabolism and redox balance in the brain, and thereby alleviated DACI. These results provide new insights into the effects of MR diet on cognitive dysfunction caused by impaired brain energy metabolism.
Collapse
Affiliation(s)
- Yuyu Zhang
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Yajie Wang
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Yiju Li
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Jingxi Pang
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Annika Höhn
- German Institute of Human Nutrition (DIfE) Potsdam-Rehbruecke, Department of Molecular Toxicology, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany; German Center for Diabetes Research (DZD), 85764, Muenchen, Neuherberg, Germany
| | - Weixuan Dong
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Rui Gao
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yan Liu
- The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Da Wang
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Yongbo She
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Rui Guo
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China
| | - Zhigang Liu
- Laboratory of Functional Chemistry and Nutrition of Food, College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China; Northwest A&F University Shenzhen Research Institute, Shenzhen, Guangdong, 518000, China.
| |
Collapse
|
5
|
Sullivan AI, Jensen-Cody SO, Claflin KE, Vorhies KE, Flippo KH, Potthoff MJ. Characterization of FGF21 Sites of Production and Signaling in Mice. Endocrinology 2024; 165:bqae120. [PMID: 39253796 DOI: 10.1210/endocr/bqae120] [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: 02/15/2024] [Revised: 08/08/2024] [Accepted: 09/08/2024] [Indexed: 09/11/2024]
Abstract
Fibroblast growth factor (FGF) 21 is an endocrine hormone that signals to multiple tissues to regulate metabolism. FGF21 and another endocrine FGF, FGF15/19, signal to target tissues by binding to the co-receptor β-klotho (KLB), which then facilitates the interaction of these different FGFs with their preferred FGF receptor. KLB is expressed in multiple metabolic tissues, but the specific cell types and spatial distribution of these cells are not known. Furthermore, while circulating FGF21 is primarily produced by the liver, recent publications have indicated that brain-derived FGF21 impacts memory and learning. Here we use reporter mice to comprehensively assess KLB and FGF21 expression throughout the body. These data provide an important resource for guiding future studies to identify important peripheral and central targets of FGFs and to determine the significance of nonhepatic FGF21 production.
Collapse
Affiliation(s)
- Andrew I Sullivan
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Iowa Neurosciences Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Sharon O Jensen-Cody
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Iowa Neurosciences Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Kristin E Claflin
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Iowa Neurosciences Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Kai E Vorhies
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Iowa Neurosciences Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Kyle H Flippo
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Iowa Neurosciences Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Department of Veterans Affairs Medical Center, Iowa City, IA 52242, USA
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Matthew J Potthoff
- Department of Neuroscience and Pharmacology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Iowa Neurosciences Institute, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
- Department of Veterans Affairs Medical Center, Iowa City, IA 52242, USA
| |
Collapse
|
6
|
Nishizawa H, Matsumoto M, Yamanaka M, Irikura R, Nakajima K, Tada K, Nakayama Y, Konishi M, Itoh N, Funayama R, Nakayama K, Igarashi K. BACH1 inhibits senescence, obesity, and short lifespan by ferroptotic FGF21 secretion. Cell Rep 2024; 43:114403. [PMID: 38943639 DOI: 10.1016/j.celrep.2024.114403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 04/14/2024] [Accepted: 06/11/2024] [Indexed: 07/01/2024] Open
Abstract
Ferroptosis is a type of regulated cell death characterized by iron-dependent lipid peroxidation. A model cell system is constructed to induce ferroptosis by re-expressing the transcription factor BACH1, a potent ferroptosis inducer, in immortalized mouse embryonic fibroblasts (iMEFs). The transfer of the culture supernatant from ferroptotic iMEFs activates the proliferation of hepatoma cells and other fibroblasts and suppresses cellular senescence-like features. The BACH1-dependent secretion of the longevity factor FGF21 is increased in ferroptotic iMEFs. The anti-senescent effects of the culture supernatant from these iMEFs are abrogated by Fgf21 knockout. BACH1 activates the transcription of Fgf21 by promoting ferroptotic stress and increases FGF21 protein expression by suppressing its autophagic degradation through transcriptional Sqstm1 and Lamp2 repression. The BACH1-induced ferroptotic FGF21 secretion suppresses obesity in high-fat diet-fed mice and the short lifespan of progeria mice. The inhibition of these aging-related phenotypes can be physiologically significant regarding ferroptosis.
Collapse
Affiliation(s)
- Hironari Nishizawa
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan.
| | - Mitsuyo Matsumoto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan; Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Mie Yamanaka
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan; Gladstone Institute of Neurological Disease, Gladstone Institute, San Francisco, CA 94158, USA
| | - Riko Irikura
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Kazuma Nakajima
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Keisuke Tada
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan; Department of Pediatric Surgery, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Yoshiaki Nakayama
- Laboratory of Microbial Chemistry, Kobe Pharmaceutical University, Kobe, Hyogo 658-8558, Japan
| | - Morichika Konishi
- Laboratory of Microbial Chemistry, Kobe Pharmaceutical University, Kobe, Hyogo 658-8558, Japan
| | - Nobuyuki Itoh
- Graduate School of Pharmaceutical Sciences, Kyoto University, Sakyo, Kyoto 606-8501, Japan
| | - Ryo Funayama
- Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan; Department of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Keiko Nakayama
- Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan; Department of Cell Proliferation, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan; Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Sendai, Miyagi 980-8575, Japan.
| |
Collapse
|
7
|
Le Thuc O, García-Cáceres C. Obesity-induced inflammation: connecting the periphery to the brain. Nat Metab 2024; 6:1237-1252. [PMID: 38997442 DOI: 10.1038/s42255-024-01079-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Accepted: 06/11/2024] [Indexed: 07/14/2024]
Abstract
Obesity is often associated with a chronic, low-grade inflammatory state affecting the entire body. This sustained inflammatory state disrupts the coordinated communication between the periphery and the brain, which has a crucial role in maintaining homeostasis through humoural, nutrient-mediated, immune and nervous signalling pathways. The inflammatory changes induced by obesity specifically affect communication interfaces, including the blood-brain barrier, glymphatic system and meninges. Consequently, brain areas near the third ventricle, including the hypothalamus and other cognition-relevant regions, become susceptible to impairments, resulting in energy homeostasis dysregulation and an elevated risk of cognitive impairments such as Alzheimer's disease and dementia. This Review explores the intricate communication between the brain and the periphery, highlighting the effect of obesity-induced inflammation on brain function.
Collapse
Affiliation(s)
- Ophélia Le Thuc
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - Cristina García-Cáceres
- Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Zentrum München, Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
8
|
Li S, Zou T, Chen J, Li J, You J. Fibroblast growth factor 21: An emerging pleiotropic regulator of lipid metabolism and the metabolic network. Genes Dis 2024; 11:101064. [PMID: 38292170 PMCID: PMC10825286 DOI: 10.1016/j.gendis.2023.06.033] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 01/20/2023] [Accepted: 06/27/2023] [Indexed: 02/01/2024] Open
Abstract
Fibroblast growth factor 21 (FGF21) was originally identified as an important metabolic regulator which plays a crucial physiological role in regulating a variety of metabolic parameters through the metabolic network. As a novel multifunctional endocrine growth factor, the role of FGF21 in the metabolic network warrants extensive exploration. This insight was obtained from the observation that the FGF21-dependent mechanism that regulates lipid metabolism, glycogen transformation, and biological effectiveness occurs through the coordinated participation of the liver, adipose tissue, central nervous system, and sympathetic nerves. This review focuses on the role of FGF21-uncoupling protein 1 (UCP1) signaling in lipid metabolism and how FGF21 alleviates non-alcoholic fatty liver disease (NAFLD). Additionally, this review reveals the mechanism by which FGF21 governs glucolipid metabolism. Recent research on the role of FGF21 in the metabolic network has mostly focused on the crucial pathway of glucolipid metabolism. FGF21 has been shown to have multiple regulatory roles in the metabolic network. Since an adequate understanding of the concrete regulatory pathways of FGF21 in the metabolic network has not been attained, this review sheds new light on the metabolic mechanisms of FGF21, explores how FGF21 engages different tissues and organs, and lays a theoretical foundation for future in-depth research on FGF21-targeted treatment of metabolic diseases.
Collapse
Affiliation(s)
| | | | - Jun Chen
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China
| | - Jiaming Li
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China
| | - Jinming You
- Jiangxi Province Key Laboratory of Animal Nutrition, Jiangxi Agricultural University, Nanchang, Jiangxi 330045, China
| |
Collapse
|
9
|
Jackson TC, Herrmann JR, Fink EL, Au AK, Kochanek PM. Harnessing the Promise of the Cold Stress Response for Acute Brain Injury and Critical Illness in Infants and Children. Pediatr Crit Care Med 2024; 25:259-270. [PMID: 38085024 PMCID: PMC10932834 DOI: 10.1097/pcc.0000000000003424] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Affiliation(s)
- Travis C. Jackson
- Department of Molecular Pharmacology & Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL
| | - Jeremy R. Herrmann
- Department of Anesthesiology and Critical Care Medicine, Perelman School of Medicine, University of Pennsylvania, Children’s Hospital of Philadelphia, Philadelphia, PA
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
| | - Ericka L. Fink
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Pediatrics, University of Pittsburgh School of Medicine, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA
| | - Alicia K. Au
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Pediatrics, University of Pittsburgh School of Medicine, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA
| | - Patrick M. Kochanek
- Safar Center for Resuscitation Research, Department of Critical Care Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA
- Department of Pediatrics, University of Pittsburgh School of Medicine, UPMC Children’s Hospital of Pittsburgh, Pittsburgh, PA
| |
Collapse
|
10
|
Zangerolamo L, Carvalho M, Velloso LA, Barbosa HCL. Endocrine FGFs and their signaling in the brain: Relevance for energy homeostasis. Eur J Pharmacol 2024; 963:176248. [PMID: 38056616 DOI: 10.1016/j.ejphar.2023.176248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 11/10/2023] [Accepted: 11/30/2023] [Indexed: 12/08/2023]
Abstract
Since their discovery in 2000, there has been a continuous expansion of studies investigating the physiology, biochemistry, and pharmacology of endocrine fibroblast growth factors (FGFs). FGF19, FGF21, and FGF23 comprise a subfamily with attributes that distinguish them from typical FGFs, as they can act as hormones and are, therefore, referred to as endocrine FGFs. As they participate in a broad cross-organ endocrine signaling axis, endocrine FGFs are crucial lipidic, glycemic, and energetic metabolism regulators during energy availability fluctuations. They function as powerful metabolic signals in physiological responses induced by metabolic diseases, like type 2 diabetes and obesity. Pharmacologically, FGF19 and FGF21 cause body weight loss and ameliorate glucose homeostasis and energy expenditure in rodents and humans. In contrast, FGF23 expression in mice and humans has been linked with insulin resistance and obesity. Here, we discuss emerging concepts in endocrine FGF signaling in the brain and critically assess their putative role as therapeutic targets for treating metabolic disorders.
Collapse
Affiliation(s)
- Lucas Zangerolamo
- Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Sao Paulo, Brazil
| | - Marina Carvalho
- Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Sao Paulo, Brazil
| | - Licio A Velloso
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Sao Paulo, Brazil
| | - Helena C L Barbosa
- Obesity and Comorbidities Research Center, University of Campinas, UNICAMP, Campinas, Sao Paulo, Brazil.
| |
Collapse
|
11
|
Singh C. Systems levels analysis of lipid metabolism in oxygen-induced retinopathy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.21.568200. [PMID: 38045301 PMCID: PMC10690220 DOI: 10.1101/2023.11.21.568200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Hyperoxia induces glutamine-fueled anaplerosis in the Muller cells, endothelial cells, and retinal explants. Anaplerosis takes away glutamine from the biosynthetic pathway to the energy-producing TCA cycle. This process depletes biosynthetic precursors from newly proliferating endothelial cells. The induction of anaplerosis in the hyperoxic retina is a compensatory response, either to decreased glycolysis or decreased flux from glycolysis to the TCA cycle. We hypothesized that by providing substrates that feed into TCA, we could reverse or prevent glutamine-fueled anaplerosis, thereby abating the glutamine wastage for energy generation. Using an oxygen-induced retinopathy (OIR) mouse model, we first compared the difference in fatty acid metabolism between OIR-resistant BALB/cByJ and OIR susceptible C57BL/6J strains to understand if these strains exhibit metabolic difference that protects BALB/cByJ from the hyperoxic conditions and prevents their vasculature in oxygen-induced retinopathy model. Based on our findings from the metabolic comparison between two mouse strains, we hypothesized that the medium-chain fatty acid, octanoate, can feed into the TCA and serve as an alternative energy source in response to hyperoxia. Our systems levels analysis of OIR model shows that the medium chain fatty acid can serve as an alternative source to feed TCA. We here, for the first time, demonstrate that the retina can use medium-chain fatty acid octanoate to replenish TCA in normoxic and at a higher rate in hyperoxic conditions.
Collapse
|
12
|
Lail H, Mabb AM, Parent MB, Pinheiro F, Wanders D. Effects of Dietary Methionine Restriction on Cognition in Mice. Nutrients 2023; 15:4950. [PMID: 38068808 PMCID: PMC10707861 DOI: 10.3390/nu15234950] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/16/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Dietary restriction of the essential amino acid, methionine, has been shown to induce unique metabolic protection. The peripheral benefits of methionine restriction (MR) are well established and include improvements in metabolic, energy, inflammatory, and lifespan parameters in preclinical models. These benefits all occur despite MR increasing energy intake, making MR an attractive dietary intervention for the prevention or reversal of many metabolic and chronic conditions. New and emerging evidence suggests that MR also benefits the brain and promotes cognitive health. Despite widespread interest in MR over the past few decades, many findings are limited in scope, and gaps remain in our understanding of its comprehensive effects on the brain and cognition. This review details the current literature investigating the impact of MR on cognition in various mouse models, highlights some of the key mechanisms responsible for its cognitive benefits, and identifies gaps that should be addressed in MR research moving forward. Overall findings indicate that in animal models, MR is associated with protection against obesity-, age-, and Alzheimer's disease-induced impairments in learning and memory that depend on different brain regions, including the prefrontal cortex, amygdala, and hippocampus. These benefits are likely mediated by increases in fibroblast growth factor 21, alterations in methionine metabolism pathways, reductions in neuroinflammation and central oxidative stress, and potentially alterations in the gut microbiome, mitochondrial function, and synaptic plasticity.
Collapse
Affiliation(s)
- Hannah Lail
- Department of Nutrition, Georgia State University, 140 Decatur St SE, Atlanta, GA 30303, USA; (H.L.); (F.P.)
- Department of Chemistry, Georgia State University, 100 Piedmont Ave., Atlanta, GA 30303, USA
| | - Angela M. Mabb
- Neuroscience Institute, Georgia State University, 100 Piedmont Ave., Atlanta, GA 30302, USA; (A.M.M.); (M.B.P.)
- Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30302, USA
| | - Marise B. Parent
- Neuroscience Institute, Georgia State University, 100 Piedmont Ave., Atlanta, GA 30302, USA; (A.M.M.); (M.B.P.)
- Department of Psychology, Georgia State University, 140 Decatur St SE, Atlanta, GA 30303, USA
| | - Filipe Pinheiro
- Department of Nutrition, Georgia State University, 140 Decatur St SE, Atlanta, GA 30303, USA; (H.L.); (F.P.)
- Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Desiree Wanders
- Department of Nutrition, Georgia State University, 140 Decatur St SE, Atlanta, GA 30303, USA; (H.L.); (F.P.)
| |
Collapse
|
13
|
Herrmann JR, Kochanek PM, Vagni VA, Janesko-Feldman K, Stezoski J, Gorse K, Jackson TC. FGF21 modulates hippocampal cold-shock proteins and CA2-subregion proteins in neonatal mice with hypoxia-ischemia. Pediatr Res 2023; 94:1355-1364. [PMID: 37193753 PMCID: PMC10690493 DOI: 10.1038/s41390-023-02652-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/30/2023] [Accepted: 05/01/2023] [Indexed: 05/18/2023]
Abstract
BACKGROUND Fibroblast growth factor 21 (FGF21) is a neuroprotectant with cognitive enhancing effects but with poorly characterized mechanism(s) of action, particularly in females. Prior studies suggest that FGF21 may regulate cold-shock proteins (CSPs) and CA2-marker proteins in the hippocampus but empirical evidence is lacking. METHODS We assessed in normothermic postnatal day (PND) 10 female mice, if hypoxic-ischemic (HI) brain injury (25 min 8% O2/92% N2) altered endogenous levels of FGF21 in serum or in the hippocampus, or its receptor β-klotho. We also tested if systemic administration of FGF21 (1.5 mg/kg) modulated hippocampal CSPs or CA2 proteins. Finally, we measured if FGF21 therapy altered markers of acute hippocampal injury. RESULTS HI increased endogenous serum FGF21 (24 h), hippocampal tissue FGF21 (4d), and decreased hippocampal β-klotho levels (4d). Exogenous FGF21 therapy modulated hippocampal CSP levels, and dynamically altered hippocampal CA2 marker expression (24 h and 4d). Finally, FGF21 ameliorated neuronal damage markers at 24 h but did not affect GFAP (astrogliosis) or Iba1 (microgliosis) levels at 4d. CONCLUSIONS FGF21 therapy modulates CSP and CA2 protein levels in the injured hippocampus. These proteins serve different biological functions, but our findings suggest that FGF21 administration modulates them in a homeostatic manner after HI. IMPACT Hypoxic-ischemic (HI) injury in female post-natal day (PND) 10 mice decreases hippocampal RNA binding motif 3 (RBM3) levels in the normothermic newborn brain. HI injury in normothermic newborn female mice alters serum and hippocampal fibroblast growth factor 21 (FGF21) levels 24 h post-injury. HI injury in normothermic newborn female mice alters hippocampal levels of N-terminal EF-hand calcium binding protein 2 (NECAB2) in a time-dependent manner. Exogenous FGF21 therapy ameliorates the HI-mediated loss of hippocampal cold-induced RNA-binding protein (CIRBP). Exogenous FGF21 therapy modulates hippocampal levels of CA2-marker proteins after HI.
Collapse
Affiliation(s)
- Jeremy R Herrmann
- Department of Critical Care Medicine, Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Rangos Research Center - 6th floor, Pittsburgh, PA, 15224, USA
| | - Patrick M Kochanek
- Department of Critical Care Medicine, Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Rangos Research Center - 6th floor, Pittsburgh, PA, 15224, USA
| | - Vincent A Vagni
- Department of Critical Care Medicine, Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Rangos Research Center - 6th floor, Pittsburgh, PA, 15224, USA
| | - Keri Janesko-Feldman
- Department of Critical Care Medicine, Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Rangos Research Center - 6th floor, Pittsburgh, PA, 15224, USA
| | - Jason Stezoski
- Department of Critical Care Medicine, Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, Rangos Research Center - 6th floor, Pittsburgh, PA, 15224, USA
| | - Kiersten Gorse
- USF Health Heart Institute, University of South Florida Morsani College of Medicine, MDD 0630, 560 Channelside Drive, Tampa, FL, 33602, USA
- Department of Molecular Pharmacology & Physiology, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Boulevard, Tampa, FL, 33612-4799, USA
| | - Travis C Jackson
- USF Health Heart Institute, University of South Florida Morsani College of Medicine, MDD 0630, 560 Channelside Drive, Tampa, FL, 33602, USA.
- Department of Molecular Pharmacology & Physiology, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Boulevard, Tampa, FL, 33612-4799, USA.
| |
Collapse
|
14
|
Yong F, Yan M, Zhang L, Ji W, Zhao S, Gao Y. Analysis of Functional Promoter of Camel FGF21 Gene and Identification of Small Compounds Targeting FGF21 Protein. Vet Sci 2023; 10:452. [PMID: 37505857 PMCID: PMC10383868 DOI: 10.3390/vetsci10070452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/27/2023] [Accepted: 07/06/2023] [Indexed: 07/29/2023] Open
Abstract
The fibroblast growth factor 21 (FGF21) gene plays an important role in the mechanism of glucose and lipid metabolism and is a promising therapeutic target for metabolic disease. Camels display a unique regulation characteristic of glucose and lipid metabolism, endowing them with the ability to adapt to survive drought and chronic hunger. However, the knowledge about the camel FGF21 gene regulation and its differences between humans and mice is still limited. In this study, camel FGF21 gene promoter was obtained for ~2000 bp upstream of the transcriptional start site (TSS). Bioinformatics analysis showed that the proximal promoter region sequences near the TSS between humans and camels have high similarity. Two potential core active regions are located in the -445-612 bp region. In addition, camel FGF21 promoter contains three CpG islands (CGIs), located in the -435~-1168 bp regions, significantly more and longer than in humans and mice. The transcription factor binding prediction showed that most transcription factors, including major functional transcription factors, are the same in different species although the binding site positions in the promoter are different. These results indicated that the signaling pathways involved in FGF21 gene transcription regulation are conservative in mammals. Truncated fragments recombinant vectors and luciferase reporter assay determined that camel FGF21 core promoter is located within the 800 bp region upstream of the TSS and an enhancer may exist between the -1000 and -2000 bp region. Combining molecular docking and in silico ADMET druggability prediction, two compounds were screened as the most promising candidate drugs specifically targeting FGF21. This study expanded the functions of these small molecules and provided a foundation for drug development targeting FGF21.
Collapse
Affiliation(s)
- Fang Yong
- College of Life Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Meilin Yan
- College of Life Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Lili Zhang
- College of Life Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Wangye Ji
- College of Life Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
| | - Shuqin Zhao
- College of Life Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| | - Yuan Gao
- College of Life Science and Technology, Gansu Agricultural University, Lanzhou 730070, China
- Gansu Key Laboratory of Animal Generational Physiology and Reproductive Regulation, Lanzhou 730070, China
| |
Collapse
|
15
|
Zhu H, Liu D, Sui M, Zhou M, Wang B, Qi Q, Wang T, Zhang G, Wan F, Zhang B. CRISPRa-based activation of Fgf21 and Fndc5 ameliorates obesity by promoting adipocytes browning. Clin Transl Med 2023; 13:e1326. [PMID: 37462619 PMCID: PMC10353577 DOI: 10.1002/ctm2.1326] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 05/29/2023] [Accepted: 06/29/2023] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND Skeletal muscle-secreted myokines widely participate in lipids metabolism through autocrine, paracrine and endocrine actions. The myokines represented by FGF21 and Irisin can promote the browning of adipocytes and serve as promising targets for treating obesity. Although recombinant myokines replacement therapy and AAV (adeno-associated virus)-based myokines overexpression have shown a definite effect in ameliorating obesity, novel myokine activation strategies with higher efficacy and safety are still in pressing need. This study aimed to evaluate the therapeutic potential of a novel CRISPR-based myokines activation strategy in obesity treatments. METHODS In this study, we used lentivirus and a single AAV vector containing dCas9-VP64 with a single-guide RNA to selectively activate Fgf21 and Fndc5 expression in skeletal muscles both in vitro and in vivo. The activation efficacy of the CRISPRa system was determined by qRT-PCR, Western blotting and ELISA. The treatment effect of CRISPR-based myokines activation was tested in 3T3-L1-derived adipocytes and diet-induced obese (DIO) mice (male C57BL/6 mice, induced at 6-week-old for 10 weeks). RESULTS The virus upregulates myokines expression in both mRNA and protein levels of muscle cells in vitro and in vivo. Myokines secreted by muscle cells promoted browning of 3T3-L1-derived adipocytes. In vivo activation of myokines by AAVs can reduce body weight and fat mass, increase the adipocytes browning and improve glucose tolerance and insulin sensitivity in DIO mice. CONCLUSIONS Our study provides a novel CRISPR-based myokines activation strategy that can ameliorate obesity by promoting adipocytes browning.
Collapse
Affiliation(s)
- Hongtao Zhu
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Dan Liu
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Sui
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Meiling Zhou
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Beibei Wang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Qinqin Qi
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Wang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
| | - Guo Zhang
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Environmental Health, Ministry of Education, Department of Toxicology, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Wan
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Neurosurgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Bin Zhang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
16
|
Kannangara H, Cullen L, Miyashita S, Korkmaz F, Macdonald A, Gumerova A, Witztum R, Moldavski O, Sims S, Burgess J, Frolinger T, Latif R, Ginzburg Y, Lizneva D, Goosens K, Davies TF, Yuen T, Zaidi M, Ryu V. Emerging roles of brain tanycytes in regulating blood-hypothalamus barrier plasticity and energy homeostasis. Ann N Y Acad Sci 2023; 1525:61-69. [PMID: 37199228 PMCID: PMC10524199 DOI: 10.1111/nyas.15009] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Seasonal changes in food intake and adiposity in many animal species are triggered by changes in the photoperiod. These latter changes are faithfully transduced into a biochemical signal by melatonin secreted by the pineal gland. Seasonal variations, encoded by melatonin, are integrated by third ventricular tanycytes of the mediobasal hypothalamus through the detection of the thyroid-stimulating hormone (TSH) released from the pars tuberalis. The mediobasal hypothalamus is a critical brain region that maintains energy homeostasis by acting as an interface between the neural networks of the central nervous system and the periphery to control metabolic functions, including ingestive behavior, energy homeostasis, and reproduction. Among the cells involved in the regulation of energy balance and the blood-hypothalamus barrier (BHB) plasticity are tanycytes. Increasing evidence suggests that anterior pituitary hormones, specifically TSH, traditionally considered to have unitary functions in targeting single endocrine sites, display actions on multiple somatic tissues and central neurons. Notably, modulation of tanycytic TSH receptors seems critical for BHB plasticity in relation to energy homeostasis, but this needs to be proven.
Collapse
Affiliation(s)
- Hasni Kannangara
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Liam Cullen
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Sari Miyashita
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Funda Korkmaz
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Anne Macdonald
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Anisa Gumerova
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ronit Witztum
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ofer Moldavski
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Steven Sims
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Jocoll Burgess
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Tal Frolinger
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Rauf Latif
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Yelena Ginzburg
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Daria Lizneva
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Ki Goosens
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Terry F. Davies
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Tony Yuen
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Mone Zaidi
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Vitaly Ryu
- Center for Translational Medicine and Pharmacology, Icahn School of Medicine at Mount Sinai, New York, NY
- Department of Medicine and of Pharmacological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
17
|
Dali R, Estrada-Meza J, Langlet F. Tanycyte, the neuron whisperer. Physiol Behav 2023; 263:114108. [PMID: 36740135 DOI: 10.1016/j.physbeh.2023.114108] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/23/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023]
Abstract
Reciprocal communication between neurons and glia is essential for normal brain functioning and adequate physiological functions, including energy balance. In vertebrates, the homeostatic process that adjusts food intake and energy expenditure in line with physiological requirements is tightly controlled by numerous neural cell types located within the hypothalamus and the brainstem and organized in complex networks. Within these neural networks, peculiar ependymoglial cells called tanycytes are nowadays recognized as multifunctional players in the physiological mechanisms of appetite control, partly by modulating orexigenic and anorexigenic neurons. Here, we review recent advances in tanycytes' impact on hypothalamic neuronal activity, emphasizing on arcuate neurons.
Collapse
Affiliation(s)
- Rafik Dali
- Department of biomedical sciences, University of Lausanne, 1005 Lausanne, Switzerland
| | - Judith Estrada-Meza
- Department of biomedical sciences, University of Lausanne, 1005 Lausanne, Switzerland
| | - Fanny Langlet
- Department of biomedical sciences, University of Lausanne, 1005 Lausanne, Switzerland.
| |
Collapse
|
18
|
Baghdadi M, Nespital T, Mesaros A, Buschbaum S, Withers DJ, Grönke S, Partridge L. Reduced insulin signaling in neurons induces sex-specific health benefits. SCIENCE ADVANCES 2023; 9:eade8137. [PMID: 36812323 PMCID: PMC9946356 DOI: 10.1126/sciadv.ade8137] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 01/24/2023] [Indexed: 06/18/2023]
Abstract
Reduced activity of insulin/insulin-like growth factor signaling (IIS) extends health and life span in mammals. Loss of the insulin receptor substrate 1 (Irs1) gene increases survival in mice and causes tissue-specific changes in gene expression. However, the tissues underlying IIS-mediated longevity are currently unknown. Here, we measured survival and health span in mice lacking IRS1 specifically in liver, muscle, fat, and brain. Tissue-specific loss of IRS1 did not increase survival, suggesting that lack of IRS1 in more than one tissue is required for life-span extension. Loss of IRS1 in liver, muscle, and fat did not improve health. In contrast, loss of neuronal IRS1 increased energy expenditure, locomotion, and insulin sensitivity, specifically in old males. Neuronal loss of IRS1 also caused male-specific mitochondrial dysfunction, activation of Atf4, and metabolic adaptations consistent with an activated integrated stress response at old age. Thus, we identified a male-specific brain signature of aging in response to reduced IIS associated with improved health at old age.
Collapse
Affiliation(s)
| | - Tobias Nespital
- Max-Planck Institute for Biology of Ageing, Cologne, Germany
| | - Andrea Mesaros
- Max-Planck Institute for Biology of Ageing, Cologne, Germany
| | | | - Dominic J. Withers
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- Medical Research Council London Institute of Medical Sciences, London, UK
| | | | - Linda Partridge
- Max-Planck Institute for Biology of Ageing, Cologne, Germany
- Institute of Healthy Ageing and Genetics, Evolution and Environment, University College London, London, UK
| |
Collapse
|
19
|
Geller S, Pellerin L. Tanycytes and hypothalamic FGF21: New players in the metabolic game. Cell Rep 2023; 42:111954. [PMID: 36640320 DOI: 10.1016/j.celrep.2022.111954] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/29/2022] [Accepted: 12/15/2022] [Indexed: 01/13/2023] Open
Affiliation(s)
- Sarah Geller
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland.
| | - Luc Pellerin
- Inserm U1313 IRMETIST, Université de Poitiers et CHU de Poitiers, Poitiers, France.
| |
Collapse
|
20
|
Prida E, Álvarez-Delgado S, Pérez-Lois R, Soto-Tielas M, Estany-Gestal A, Fernø J, Seoane LM, Quiñones M, Al-Massadi O. Liver Brain Interactions: Focus on FGF21 a Systematic Review. Int J Mol Sci 2022; 23:ijms232113318. [PMID: 36362103 PMCID: PMC9658462 DOI: 10.3390/ijms232113318] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/21/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
Abstract
Fibroblast growth factor 21 is a pleiotropic hormone secreted mainly by the liver in response to metabolic and nutritional challenges. Physiologically, fibroblast growth factor 21 plays a key role in mediating the metabolic responses to fasting or starvation and acts as an important regulator of energy homeostasis, glucose and lipid metabolism, and insulin sensitivity, in part by its direct action on the central nervous system. Accordingly, pharmacological recombinant fibroblast growth factor 21 therapies have been shown to counteract obesity and its related metabolic disorders in both rodents and nonhuman primates. In this systematic review, we discuss how fibroblast growth factor 21 regulates metabolism and its interactions with the central nervous system. In addition, we also state our vision for possible therapeutic uses of this hepatic-brain axis.
Collapse
Affiliation(s)
- Eva Prida
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - Sara Álvarez-Delgado
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - Raquel Pérez-Lois
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, 15706 Santiago de Compostela, Spain
| | - Mateo Soto-Tielas
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
| | - Ana Estany-Gestal
- Unidad de Metodología de la Investigación, Fundación Instituto de Investigación de Santiago (FIDIS), 15706 Santiago de Compostela, Spain
| | - Johan Fernø
- Hormone Laboratory, Department of Biochemistry and Pharmacology, Haukeland University Hospital, 5201 Bergen, Norway
| | - Luisa María Seoane
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, 15706 Santiago de Compostela, Spain
| | - Mar Quiñones
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, 15706 Santiago de Compostela, Spain
- Correspondence: (M.Q.); (O.A.-M.); Tel.: +34-981955708 (M.Q.); +34-981955522 (O.A.-M.)
| | - Omar Al-Massadi
- Instituto de Investigación Sanitaria de Santiago de Compostela, Complexo Hospitalario Universitario de Santiago (CHUS/SERGAS), Travesía da Choupana s/n, 15706 Santiago de Compostela, Spain
- CIBER de Fisiopatología de la Obesidad y la Nutrición, Instituto de Salud Carlos III, 15706 Santiago de Compostela, Spain
- Correspondence: (M.Q.); (O.A.-M.); Tel.: +34-981955708 (M.Q.); +34-981955522 (O.A.-M.)
| |
Collapse
|