1
|
Mazuecos L, Artigas-Jerónimo S, Pintado C, Gómez O, Rubio B, Arribas C, Andrés A, Villar M, Gallardo N. Central leptin signaling deficiency induced by leptin receptor antagonist leads to hypothalamic proteomic remodeling. Life Sci 2024; 346:122649. [PMID: 38626868 DOI: 10.1016/j.lfs.2024.122649] [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: 11/29/2023] [Revised: 04/07/2024] [Accepted: 04/11/2024] [Indexed: 04/20/2024]
Abstract
AIMS Leptin irresponsiveness, which is often associated with obesity, can have significant impacts on the hypothalamic proteome of individuals, including those who are lean. While mounting evidence on leptin irresponsiveness has focused on obese individuals, understanding the early molecular and proteomic changes associated with deficient hypothalamic leptin signaling in lean individuals is essential for early intervention and prevention of metabolic disorders. Leptin receptor antagonists block the binding of leptin to its receptors, potentially reducing its effects and used in cases where excessive leptin activity might be harmful. MATERIALS AND METHODS In this work, we blocked the central actions of leptin in lean male adult Wistar rat by chronically administering intracerebroventricularly the superactive leptin receptor antagonist (SLA) (D23L/L39A/D40A/F41A) and investigated its impact on the hypothalamic proteome using label-free sequential window acquisition of all theoretical fragment ion spectra mass spectrometry (SWATH-MS) for quantitative proteomics. KEY FINDINGS Our results show an accumulation of proteins involved in mRNA processing, mRNA stability, and translation in the hypothalamus of SLA-treated rats. Conversely, hypothalamic leptin signaling deficiency reduces the representation of proteins implicated in energy metabolism, neural circuitry, and neurotransmitter release. SIGNIFICANCE The alterations in the adult rat hypothalamic proteome contribute to dysregulate appetite, metabolism, and energy balance, which are key factors in the development and progression of obesity and related metabolic disorders. Additionally, using bioinformatic analysis, we identified a series of transcription factors that are potentially involved in the upstream regulatory mechanisms responsible for the observed signature.
Collapse
Affiliation(s)
- Lorena Mazuecos
- Biochemistry Section, Faculty of Sciences and Chemical Technologies, University of Castilla-La Mancha, Avda. Camilo Jose Cela 10, 13071 Ciudad Real, Spain; DOE, Regional Center for Biomedical Research (CRIB), Castilla-La Mancha, Spain
| | - Sara Artigas-Jerónimo
- Biochemistry Section, Faculty of Sciences and Chemical Technologies, University of Castilla-La Mancha, Avda. Camilo Jose Cela 10, 13071 Ciudad Real, Spain; DOE, Regional Center for Biomedical Research (CRIB), Castilla-La Mancha, Spain
| | - Cristina Pintado
- Biochemistry Section, Faculty of Environmental Sciences and Biochemistry, University of Castilla-La Mancha, Avda. Carlos III s/n, 45071 Toledo, Spain; DOE, Regional Center for Biomedical Research (CRIB), Castilla-La Mancha, Spain
| | - Oscar Gómez
- Biochemistry Section, Faculty of Environmental Sciences and Biochemistry, University of Castilla-La Mancha, Avda. Carlos III s/n, 45071 Toledo, Spain; DOE, Regional Center for Biomedical Research (CRIB), Castilla-La Mancha, Spain
| | - Blanca Rubio
- Biochemistry Section, Faculty of Sciences and Chemical Technologies, University of Castilla-La Mancha, Avda. Camilo Jose Cela 10, 13071 Ciudad Real, Spain; DOE, Regional Center for Biomedical Research (CRIB), Castilla-La Mancha, Spain
| | - Carmen Arribas
- Biochemistry Section, Faculty of Environmental Sciences and Biochemistry, University of Castilla-La Mancha, Avda. Carlos III s/n, 45071 Toledo, Spain; DOE, Regional Center for Biomedical Research (CRIB), Castilla-La Mancha, Spain
| | - Antonio Andrés
- Biochemistry Section, Faculty of Sciences and Chemical Technologies, University of Castilla-La Mancha, Avda. Camilo Jose Cela 10, 13071 Ciudad Real, Spain; DOE, Regional Center for Biomedical Research (CRIB), Castilla-La Mancha, Spain
| | - Margarita Villar
- Biochemistry Section, Faculty of Sciences and Chemical Technologies, University of Castilla-La Mancha, Avda. Camilo Jose Cela 10, 13071 Ciudad Real, Spain; SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005 Ciudad Real, Spain.
| | - Nilda Gallardo
- Biochemistry Section, Faculty of Sciences and Chemical Technologies, University of Castilla-La Mancha, Avda. Camilo Jose Cela 10, 13071 Ciudad Real, Spain; DOE, Regional Center for Biomedical Research (CRIB), Castilla-La Mancha, Spain.
| |
Collapse
|
2
|
Neyens DM, Brenner L, Calkins R, Winzenried ET, Ritter RC, Appleyard SM. CCK-sensitive C fibers activate NTS leptin receptor-expressing neurons via NMDA receptors. Am J Physiol Regul Integr Comp Physiol 2024; 326:R383-R400. [PMID: 38105761 PMCID: PMC11381032 DOI: 10.1152/ajpregu.00238.2022] [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: 09/27/2022] [Revised: 12/07/2023] [Accepted: 12/07/2023] [Indexed: 12/19/2023]
Abstract
The hormone leptin reduces food intake through actions in the peripheral and central nervous systems, including in the hindbrain nucleus of the solitary tract (NTS). The NTS receives viscerosensory information via vagal afferents, including information from the gastrointestinal tract, which is then relayed to other central nervous system (CNS) sites critical for control of food intake. Leptin receptors (lepRs) are expressed by a subpopulation of NTS neurons, and knockdown of these receptors increases both food intake and body weight. Recently, we demonstrated that leptin increases vagal activation of lepR-expressing neurons via increased NMDA receptor (NMDAR) currents, thereby potentiating vagally evoked firing. Furthermore, chemogenetic activation of these neurons was recently shown to inhibit food intake. However, the vagal inputs these neurons receive had not been characterized. Here we performed whole cell recordings in brain slices taken from lepRCre × floxedTdTomato mice and found that lepR neurons of the NTS are directly activated by monosynaptic inputs from C-type afferents sensitive to the transient receptor potential vanilloid type 1 (TRPV1) agonist capsaicin. CCK administered onto NTS slices stimulated spontaneous glutamate release onto lepR neurons and induced action potential firing, an effect mediated by CCKR1. Interestingly, NMDAR activation contributed to the current carried by spontaneous excitatory postsynaptic currents (EPSCs) and enhanced CCK-induced firing. Peripheral CCK also increased c-fos expression in these neurons, suggesting they are activated by CCK-sensitive vagal afferents in vivo. Our results indicate that the majority of NTS lepR neurons receive direct inputs from CCK-sensitive C vagal-type afferents, with both peripheral and central CCK capable of activating these neurons and NMDARs able to potentiate these effects.
Collapse
Affiliation(s)
- Drew M Neyens
- Department of Integrated Physiology and Neuroscience, Washington State University, Pullman, Washington, United States
| | - Lynne Brenner
- Department of Integrated Physiology and Neuroscience, Washington State University, Pullman, Washington, United States
| | - Rowan Calkins
- Department of Integrated Physiology and Neuroscience, Washington State University, Pullman, Washington, United States
| | - Eric T Winzenried
- Department of Integrated Physiology and Neuroscience, Washington State University, Pullman, Washington, United States
| | - Robert C Ritter
- Department of Integrated Physiology and Neuroscience, Washington State University, Pullman, Washington, United States
| | - Suzanne M Appleyard
- Department of Integrated Physiology and Neuroscience, Washington State University, Pullman, Washington, United States
| |
Collapse
|
3
|
Higgins MBA, Glendining KA, Jasoni CL. The temporal and spatial pattern of leptin receptor-expressing cells in the developing mouse hypothalamus. J Neuroendocrinol 2024; 36:e13366. [PMID: 38279680 DOI: 10.1111/jne.13366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 12/10/2023] [Accepted: 12/30/2023] [Indexed: 01/28/2024]
Abstract
The arcuate nucleus is a crucial hypothalamic brain region involved in regulating body weight homeostasis. Neurons within the arcuate nucleus respond to peripheral metabolic signals, such as leptin, and relay these signals via neuronal projections to brain regions both within and outside the hypothalamus, ultimately causing changes in an animal's behaviour and physiology. There is a substantial amount of evidence to indicate that leptin is intimately involved with the postnatal development of arcuate nucleus melanocortin circuitry. Further, it is clear that leptin signalling directly in the arcuate nucleus is required for circuitry development. However, as leptin receptor long isoform (Leprb) mRNA is expressed in multiple nuclei within the developing hypothalamus, including the postsynaptic target regions of arcuate melanocortin projections, this raises the possibility that leptin also signals in these nuclei to promote circuitry development. Here, we used RT-qPCR and RNAscope® to reveal the spatio-temporal pattern of Leprb mRNA in the early postnatal mouse hypothalamus. We found that Leprb mRNA expression increased significantly in the arcuate nucleus, ventromedial nucleus and paraventricular nucleus of the hypothalamus from P8, in concert with the leptin surge. In the dorsomedial nucleus of the hypothalamus, increases in Leprb mRNA were slightly later, increasing significantly from P12. Using duplex RNAscope®, we found Leprb co-expressed with Sim1, Pou3f2, Mc4r and Bdnf in the paraventricular nucleus at P8. Together, these data suggest that leptin may signal in a subset of neurons postsynaptic to arcuate melanocortin neurons, as well as within the arcuate nucleus itself, to promote the formation of arcuate melanocortin circuitry during the early postnatal period.
Collapse
Affiliation(s)
- Matt B A Higgins
- Centre for Neuroendocrinology, Department of Anatomy, University of Otago, Dunedin, New Zealand
| | - Kelly A Glendining
- Centre for Neuroendocrinology, Department of Physiology, University of Otago, Dunedin, New Zealand
| | - Christine L Jasoni
- Centre for Neuroendocrinology, Department of Anatomy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
4
|
Hristov M, Nankova A, Andreeva-Gateva P. Alterations of the glutamatergic system in diabetes mellitus. Metab Brain Dis 2024; 39:321-333. [PMID: 37747631 DOI: 10.1007/s11011-023-01299-z] [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: 05/16/2023] [Accepted: 09/17/2023] [Indexed: 09/26/2023]
Abstract
Diabetes mellitus (DM) is a chronic disease characterized by elevated blood glucose levels caused by a lack of insulin production (type 1 diabetes) or insulin resistance (type 2 diabetes). It is well known that DM is associated with cognitive deficits and metabolic and neurophysiological changes in the brain. Glutamate is the main excitatory neurotransmitter in the central nervous system that plays a key role in synaptic plasticity, learning, and memory processes. An increasing number of studies have suggested that abnormal activity of the glutamatergic system is implicated in the pathophysiology of DM. Dysfunction of glutamatergic neurotransmission in the central nervous system can provide an important neurobiological substrate for many disorders. Magnetic resonance spectroscopy (MRS) is a non-invasive technique that allows a better understanding of the central nervous system factors by measuring in vivo the concentrations of brain metabolites within the area of interest. Here, we briefly review the MRS studies that have examined glutamate levels in the brain of patients with DM. The present article also summarizes the available data on abnormalities in glutamatergic neurotransmission observed in different animal models of DM. In addition, the role of gut microbiota in the development of glutamatergic alterations in DM is addressed. We speculate that therapeutic strategies targeting the glutamatergic system may be beneficial in the treatment of central nervous system-related changes in diabetic patients.
Collapse
Affiliation(s)
- Milen Hristov
- Department of Pharmacology and Toxicology, Faculty of Medicine, Medical University of Sofia, 2 "Zdrave" St, Sofia, 1431, Bulgaria.
| | - Anelia Nankova
- Department of Endocrinology, Faculty of Medicine, Medical University of Sofia, Sofia, 1431, Bulgaria
| | - Pavlina Andreeva-Gateva
- Department of Pharmacology and Toxicology, Faculty of Medicine, Medical University of Sofia, 2 "Zdrave" St, Sofia, 1431, Bulgaria
| |
Collapse
|
5
|
Tan R, Hu X, Wang X, Sun M, Cai Z, Zhang Z, Fu Y, Chen X, An J, Lu H. Leptin Promotes the Proliferation and Neuronal Differentiation of Neural Stem Cells through the Cooperative Action of MAPK/ERK1/2, JAK2/STAT3 and PI3K/AKT Signaling Pathways. Int J Mol Sci 2023; 24:15151. [PMID: 37894835 PMCID: PMC10606644 DOI: 10.3390/ijms242015151] [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: 09/05/2023] [Revised: 10/08/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
The potential of neural stem cells (NSCs) for neurological disorders the treatment has relied in large part upon identifying the NSCs fate decision. The hormone leptin has been reported to be a crucial regulator of brain development, able to influence the glial and neural development, yet, the underlying mechanism of leptin acting on NSCs' biological characteristics is still poorly understood. This study aims to investigate the role of leptin in the biological properties of NSCs. In this study, we investigate the possibility that leptin may regulate the NSCs' fate decision, which may promote the proliferation and neuronal differentiation of NSCs and thus act positively in neurological disorders. NSCs from the embryonic cerebral cortex were used in this study. We used CCK-8 assay, ki67 immunostaining, and FACS analysis to confirm that 25-100 ng/mL leptin promotes the proliferation of NSCs in a concentration-dependent pattern. This change was accompanied by the upregulation of p-AKT and p-ERK1/2, which are the classical downstream signaling pathways of leptin receptors b (LepRb). Inhibition of PI3K/AKT or MAPK/ERK signaling pathways both abolished the effect of leptin-induced proliferation. Moreover, leptin also enhanced the directed neuronal differentiation of NSCs. A blockade of the PI3K/AKT pathway reversed leptin-stimulated neurogenesis, while a blockade of JAK2/STAT3 had no effect on it. Taken together, our results support a role for leptin in regulating the fate of NSCs differentiation and promoting NSCs proliferation, which could be a promising approach for brain repair via regulating the biological characteristics of NSCs.
Collapse
Affiliation(s)
- Ruolan Tan
- Department of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (R.T.); (X.H.); (X.W.); (M.S.); (Z.C.); (Z.Z.); (Y.F.); (X.C.)
- Department of Human Anatomy and Histo-Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Xiaoxuan Hu
- Department of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (R.T.); (X.H.); (X.W.); (M.S.); (Z.C.); (Z.Z.); (Y.F.); (X.C.)
- Department of Human Anatomy and Histo-Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Xinyi Wang
- Department of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (R.T.); (X.H.); (X.W.); (M.S.); (Z.C.); (Z.Z.); (Y.F.); (X.C.)
- Department of Human Anatomy and Histo-Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Meiqi Sun
- Department of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (R.T.); (X.H.); (X.W.); (M.S.); (Z.C.); (Z.Z.); (Y.F.); (X.C.)
| | - Zhenlu Cai
- Department of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (R.T.); (X.H.); (X.W.); (M.S.); (Z.C.); (Z.Z.); (Y.F.); (X.C.)
| | - Zixuan Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (R.T.); (X.H.); (X.W.); (M.S.); (Z.C.); (Z.Z.); (Y.F.); (X.C.)
- Department of Human Anatomy and Histo-Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Yali Fu
- Department of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (R.T.); (X.H.); (X.W.); (M.S.); (Z.C.); (Z.Z.); (Y.F.); (X.C.)
- Department of Human Anatomy and Histo-Embryology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China
| | - Xinlin Chen
- Department of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (R.T.); (X.H.); (X.W.); (M.S.); (Z.C.); (Z.Z.); (Y.F.); (X.C.)
| | - Jing An
- Department of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (R.T.); (X.H.); (X.W.); (M.S.); (Z.C.); (Z.Z.); (Y.F.); (X.C.)
| | - Haixia Lu
- Department of Neurobiology, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an 710061, China; (R.T.); (X.H.); (X.W.); (M.S.); (Z.C.); (Z.Z.); (Y.F.); (X.C.)
| |
Collapse
|
6
|
Recent Advances in the Knowledge of the Mechanisms of Leptin Physiology and Actions in Neurological and Metabolic Pathologies. Int J Mol Sci 2023; 24:ijms24021422. [PMID: 36674935 PMCID: PMC9860943 DOI: 10.3390/ijms24021422] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 01/05/2023] [Accepted: 01/07/2023] [Indexed: 01/13/2023] Open
Abstract
Excess body weight is frequently associated with low-grade inflammation. Evidence indicates a relationship between obesity and cancer, as well as with other diseases, such as diabetes and non-alcoholic fatty liver disease, in which inflammation and the actions of various adipokines play a role in the pathological mechanisms involved in these disorders. Leptin is mainly produced by adipose tissue in proportion to fat stores, but it is also synthesized in other organs, where leptin receptors are expressed. This hormone performs numerous actions in the brain, mainly related to the control of energy homeostasis. It is also involved in neurogenesis and neuroprotection, and central leptin resistance is related to some neurological disorders, e.g., Parkinson's and Alzheimer's diseases. In peripheral tissues, leptin is implicated in the regulation of metabolism, as well as of bone density and muscle mass. All these actions can be affected by changes in leptin levels and the mechanisms associated with resistance to this hormone. This review will present recent advances in the molecular mechanisms of leptin action and their underlying roles in pathological situations, which may be of interest for revealing new approaches for the treatment of diseases where the actions of this adipokine might be compromised.
Collapse
|
7
|
Fyn Signaling in Ischemia-Reperfusion Injury: Potential and Therapeutic Implications. Mediators Inflamm 2022; 2022:9112127. [PMID: 36157893 PMCID: PMC9499810 DOI: 10.1155/2022/9112127] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/20/2022] [Accepted: 08/27/2022] [Indexed: 11/18/2022] Open
Abstract
Ischemic stroke caused by arterial occlusion is the most common type of stroke and is one of the leading causes of disability and death, with the incidence increasing each year. Fyn is a nonreceptor tyrosine kinase belonging to the Src family of kinases (SFKs), which is related to many normal and pathological processes of the nervous system, including neurodevelopment and disease progression. In recent years, more and more evidence suggests that Fyn may be closely related to cerebral ischemia-reperfusion, including energy metabolism disorders, excitatory neurotoxicity, intracellular calcium homeostasis, free radical production, and the activation of apoptotic genes. This paper reviews the role of Fyn in the pathological process of cerebral ischemia-reperfusion, including neuroexcitotoxicity and neuroinflammation, to explore how Fyn affects specific signal cascades and leads to cerebral ischemia-reperfusion injury. In addition, Fyn also promotes the production of superoxide and endogenous NO, so as to quickly react to produce peroxynitrite, which may also mediate cerebral ischemia-reperfusion injury, which is discussed in this paper. Finally, we revealed the treatment methods related to Fyn inhibitors and discussed its potential as a clinical treatment for ischemic stroke.
Collapse
|
8
|
Harvey J. Food for Thought: Leptin and Hippocampal Synaptic Function. Front Pharmacol 2022; 13:882158. [PMID: 35784728 PMCID: PMC9247348 DOI: 10.3389/fphar.2022.882158] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 06/02/2022] [Indexed: 11/13/2022] Open
Abstract
It is well documented that the endocrine hormone, leptin controls energy homeostasis by providing key signals to specific hypothalamic nuclei. However, our knowledge of leptin’s central actions has advanced considerably over the last 20 years, with the hippocampus now established as an important brain target for this hormone. Leptin receptors are highly localised to hippocampal synapses, and increasing evidence reveals that activation of synaptically located leptin receptors markedly impacts cognitive processes, and specifically hippocampal-dependent learning and memory. Here, we review the recent actions of leptin at hippocampal synapses and explore the consequences for brain health and disease.
Collapse
|
9
|
Proskura AL, Islamova MY, Vechkapova SO. Cross-Talk of the Glutamate and Leptin Receptor Pathways. Mol Biol 2021. [DOI: 10.1134/s0026893321020291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
10
|
Forte N, Boccella S, Tunisi L, Fernández-Rilo AC, Imperatore R, Iannotti FA, De Risi M, Iannotta M, Piscitelli F, Capasso R, De Girolamo P, De Leonibus E, Maione S, Di Marzo V, Cristino L. Orexin-A and endocannabinoids are involved in obesity-associated alteration of hippocampal neurogenesis, plasticity, and episodic memory in mice. Nat Commun 2021; 12:6137. [PMID: 34675233 PMCID: PMC8531398 DOI: 10.1038/s41467-021-26388-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Accepted: 09/30/2021] [Indexed: 11/20/2022] Open
Abstract
The mammalian brain stores and distinguishes among episodic memories, i.e. memories formed during the personal experience, through a mechanism of pattern separation computed in the hippocampal dentate gyrus. Decision-making for food-related behaviors, such as the choice and intake of food, might be affected in obese subjects by alterations in the retrieval of episodic memories. Adult neurogenesis in the dentate gyrus regulates the pattern separation. Several molecular factors affect adult neurogenesis and exert a critical role in the development and plasticity of newborn neurons. Orexin-A/hypocretin-1 and downstream endocannabinoid 2-arachidonoylglycerol signaling are altered in obese mice. Here, we show that excessive orexin-A/2-arachidonoylglycerol/cannabinoid receptor type-1 signaling leads to the dysfunction of adult hippocampal neurogenesis and the subsequent inhibition of plasticity and impairment of pattern separation. By inhibiting orexin-A action at orexin-1 receptors we rescued both plasticity and pattern separation impairment in obese mice, thus providing a molecular and functional mechanism to explain alterations in episodic memory in obesity. The authors show that adult hippocampal neurogenesis is altered in the dentate gyrus of obese mice with subsequent inhibition of long-term potentiation and impairment of pattern separation. Inhibition of orexin-A action at orexin-1 receptors rescued both impairments in obese mice.
Collapse
Affiliation(s)
- Nicola Forte
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche (CNR), Pozzuoli, NA, Italy
| | - Serena Boccella
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Luigi Vanvitelli, Napoli, Italy
| | - Lea Tunisi
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche (CNR), Pozzuoli, NA, Italy
| | | | - Roberta Imperatore
- Department of Science and Technology, University of Sannio, Benevento, Italy
| | - Fabio Arturo Iannotti
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche (CNR), Pozzuoli, NA, Italy
| | - Maria De Risi
- Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy.,Institute of Biochemistry and Cell Biology, Consiglio Nazionale delle Ricerche (CNR), Monterotondo Scalo, Rome, Italy
| | - Monica Iannotta
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Luigi Vanvitelli, Napoli, Italy
| | - Fabiana Piscitelli
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche (CNR), Pozzuoli, NA, Italy
| | - Raffaele Capasso
- Department of Agricultural Sciences, University of Naples Federico II, Portici, NA, Italy
| | - Paolo De Girolamo
- Department of Veterinary Medicine and Animal Productions, University Federico II, Napoli, Italy
| | - Elvira De Leonibus
- Telethon Institute of Genetics and Medicine, Pozzuoli, Naples, Italy.,Institute of Biochemistry and Cell Biology, Consiglio Nazionale delle Ricerche (CNR), Monterotondo Scalo, Rome, Italy
| | - Sabatino Maione
- Department of Experimental Medicine, Division of Pharmacology, University of Campania Luigi Vanvitelli, Napoli, Italy.,I.R.C.S.S., Neuromed, 86077, Pozzilli, Italy
| | - Vincenzo Di Marzo
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche (CNR), Pozzuoli, NA, Italy. .,Heart and Lung Research Institute of Université Laval, Québec City, QC, Canada. .,Institute for Nutrition and Functional Foods, Centre NUTRISS, Université Laval, Québec City, QC, Canada. .,Canada Excellence Research Chair on the Microbiome-Endocannabinoidome Axis in Metabolic Health, Université Laval, Québec City, QC, 61V0AG, Canada.
| | - Luigia Cristino
- Institute of Biomolecular Chemistry, Consiglio Nazionale delle Ricerche (CNR), Pozzuoli, NA, Italy.
| |
Collapse
|
11
|
Rajani V, Sengar AS, Salter MW. Src and Fyn regulation of NMDA receptors in health and disease. Neuropharmacology 2021; 193:108615. [PMID: 34051267 DOI: 10.1016/j.neuropharm.2021.108615] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/10/2021] [Accepted: 05/12/2021] [Indexed: 11/28/2022]
Abstract
The Src family kinases (SFKs) are cytoplasmic non-receptor tyrosine kinases involved in multiple signalling pathways. In the central nervous system (CNS), SFKs are key regulators of N-methyl-d-aspartate receptor (NMDAR) function and major points of convergence for neuronal transduction pathways. Physiological upregulation of NMDAR activity by members of the SFKs, namely Src and Fyn, is crucial for induction of plasticity at Schaffer collateral-CA1 synapses of the hippocampus. Aberrant SFK regulation of NMDARs is implicated in several pathological conditions in the CNS including schizophrenia and pain hypersensitivity. Here, evidence is presented to highlight the current understanding of the intermolecular interactions of SFKs within the NMDAR macromolecular complex, the upstream regulators of SFK activity on NMDAR function and the role Src and Fyn have in synaptic plasticity and metaplasticity. The targeting of SFK protein-protein interactions is discussed as a potential therapeutic strategy to restore signalling activity underlying glutamatergic dysregulation in CNS disease pathophysiology.
Collapse
Affiliation(s)
- Vishaal Rajani
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL, A1B 3V6, Canada; Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada
| | - Ameet S Sengar
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada
| | - Michael W Salter
- Program in Neurosciences & Mental Health, Hospital for Sick Children, Toronto, Ontario, M5G 1X8, Canada; Department of Physiology, University of Toronto, Toronto, Ontario, M5S 1A8, Canada.
| |
Collapse
|
12
|
Sahin GS, Luis Rodriguez-Llamas J, Dillon C, Medina I, Appleyard SM, Gaiarsa JL, Wayman GA. Leptin increases GABAergic synaptogenesis through the Rho guanine exchange factor β-PIX in developing hippocampal neurons. Sci Signal 2021; 14:14/683/eabe4111. [PMID: 34006608 DOI: 10.1126/scisignal.abe4111] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Developing hippocampal neurons undergo rapid synaptogenesis in response to neurotrophic signals to form and refine circuit connections. The adipokine leptin is a satiety factor with neurotrophic actions, which potentiates both glutamatergic and GABAergic synaptogenesis in the hippocampus during neonatal development. Brief exposure to leptin enhances GABAA receptor-dependent synaptic currents in hippocampal neurons. Here, using molecular and electrophysiological techniques, we found that leptin increased the surface localization of GABAA receptors and the number of functional GABAergic synapses in hippocampal cultures from male and female rat pups. Leptin increased the interaction between GABAA receptors and the Rho guanine exchange factor β-PIX (a scaffolding protein at GABAergic postsynaptic sites) in a manner dependent on the kinase CaMKK. We also found that the leptin receptor and β-PIX formed a complex, the amount of which transiently increased upon leptin receptor activation. Furthermore, Tyr985 in the leptin receptor and the SH3 domain of β-PIX are crucial for this interaction, which was required for the developmental increase in GABAergic synaptogenesis. Our results suggest a mechanism by which leptin promotes GABAergic synaptogenesis in hippocampal neurons and reveal further complexity in leptin receptor signaling and its interactome.
Collapse
Affiliation(s)
- Gulcan Semra Sahin
- Department of Integrated Physiology and Neuroscience, Washington State University, Pullman, WA 99164 USA
| | - Jose Luis Rodriguez-Llamas
- Department of Integrated Physiology and Neuroscience, Washington State University, Pullman, WA 99164 USA
| | - Crystal Dillon
- Department of Integrated Physiology and Neuroscience, Washington State University, Pullman, WA 99164 USA
| | - Igor Medina
- Aix-Marseille University UMR 1249, INSERM (Institut National de la Santé et de la Recherche Médicale) Unité 1249, INMED (Institut de Neurobiologie de la Méditerranée), Marseille, France
| | - Suzanne M Appleyard
- Department of Integrated Physiology and Neuroscience, Washington State University, Pullman, WA 99164 USA
| | - Jean-Luc Gaiarsa
- Aix-Marseille University UMR 1249, INSERM (Institut National de la Santé et de la Recherche Médicale) Unité 1249, INMED (Institut de Neurobiologie de la Méditerranée), Marseille, France
| | - Gary A Wayman
- Department of Integrated Physiology and Neuroscience, Washington State University, Pullman, WA 99164 USA.
| |
Collapse
|
13
|
Irving A, Harvey J. Regulation of hippocampal synaptic function by the metabolic hormone leptin: Implications for health and disease. Prog Lipid Res 2021; 82:101098. [PMID: 33895229 DOI: 10.1016/j.plipres.2021.101098] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 10/21/2022]
Abstract
Significant advances have been made in our understanding of the hormone, leptin and its CNS actions in recent years. It is now evident that leptin has a multitude of brain functions, that extend beyond its established role in the hypothalamic control of energy balance. Additional brain regions including the hippocampus are important targets for leptin, with a high density of leptin receptors (LepRs) expressed in specific hippocampal regions and localised to CA1 synapses. Extensive evidence indicates that leptin has pro-cognitive actions, as it rapidly modifies synaptic efficacy at excitatory Schaffer collateral (SC)-CA1 and temporoammonic (TA)-CA1 synapses and enhances performance in hippocampal-dependent memory tasks. There is a functional decline in hippocampal responsiveness to leptin with age, with significant reductions in the modulatory effects of leptin at SC-CA1 and TA-CA1 synapses in aged, compared to adult hippocampus. As leptin has pro-cognitive effects, this decline in leptin sensitivity is likely to have negative consequences for cognitive function during the aging process. Here we review how evaluation of the hippocampal actions of leptin has improved our knowledge of the regulatory brain functions of leptin in health and provided significant insight into the impact of leptin in age-related neurodegenerative disorders linked to cognitive decline.
Collapse
Affiliation(s)
- Andrew Irving
- School of Biomolecular and Biomedical Science, The Conway Institute, University College Dublin, Dublin, Ireland
| | - Jenni Harvey
- Division of Systems Medicine, Ninewells Hospital and Medical School, University of Dundee, Dundee DD1 9SY, United Kingdom.
| |
Collapse
|
14
|
Cochrane VA, Wu Y, Yang Z, ElSheikh A, Dunford J, Kievit P, Fortin DA, Shyng SL. Leptin modulates pancreatic β-cell membrane potential through Src kinase-mediated phosphorylation of NMDA receptors. J Biol Chem 2020; 295:17281-17297. [PMID: 33037073 PMCID: PMC7863909 DOI: 10.1074/jbc.ra120.015489] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/08/2020] [Indexed: 12/23/2022] Open
Abstract
The adipocyte-derived hormone leptin increases trafficking of KATP and Kv2.1 channels to the pancreatic β-cell surface, resulting in membrane hyperpolarization and suppression of insulin secretion. We have previously shown that this effect of leptin is mediated by the NMDA subtype of glutamate receptors (NMDARs). It does so by potentiating NMDAR activity, thus enhancing Ca2+ influx and the ensuing downstream signaling events that drive channel trafficking to the cell surface. However, the molecular mechanism by which leptin potentiates NMDARs in β-cells remains unknown. Here, we report that leptin augments NMDAR function via Src kinase-mediated phosphorylation of the GluN2A subunit. Leptin-induced membrane hyperpolarization diminished upon pharmacological inhibition of GluN2A but not GluN2B, indicating involvement of GluN2A-containing NMDARs. GluN2A harbors tyrosine residues that, when phosphorylated by Src family kinases, potentiate NMDAR activity. We found that leptin increases phosphorylation of Tyr-418 in Src, an indicator of kinase activation. Pharmacological inhibition of Src or overexpression of a kinase-dead Src mutant prevented the effect of leptin, whereas a Src kinase activator peptide mimicked it. Using mutant GluN2A overexpression, we show that Tyr-1292 and Tyr-1387 but not Tyr-1325 are responsible for the effect of leptin. Importantly, β-cells from db/db mice, a type 2 diabetes mouse model lacking functional leptin receptors, or from obese diabetic human donors failed to respond to leptin but hyperpolarized in response to NMDA. Our study reveals a signaling pathway wherein leptin modulates NMDARs via Src to regulate β-cell excitability and suggests NMDARs as a potential target to overcome leptin resistance.
Collapse
Affiliation(s)
- Veronica A Cochrane
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, USA
| | - Yi Wu
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, USA
| | - Zhongying Yang
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, USA
| | - Assmaa ElSheikh
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, USA; Department of Medical Biochemistry, Tanta University, Tanta, Egypt
| | - Jeremy Dunford
- Department of Integrated Physiology and Neuroscience, College of Arts and Sciences, Washington State University, Vancouver, Washington, USA
| | - Paul Kievit
- Division of Cardiometabolic Health, Oregon National Primate Research Center, Beaverton, Oregon, USA
| | - Dale A Fortin
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, USA; Department of Integrated Physiology and Neuroscience, College of Arts and Sciences, Washington State University, Vancouver, Washington, USA.
| | - Show-Ling Shyng
- Department of Chemical Physiology and Biochemistry, Oregon Health and Science University, Portland, Oregon, USA.
| |
Collapse
|
15
|
Dumon C, Belaidouni Y, Diabira D, Appleyard SM, Wayman GA, Gaiarsa JL. Leptin down-regulates KCC2 activity and controls chloride homeostasis in the neonatal rat hippocampus. Mol Brain 2020; 13:151. [PMID: 33183317 PMCID: PMC7661183 DOI: 10.1186/s13041-020-00689-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/28/2020] [Indexed: 11/10/2022] Open
Abstract
The canonical physiological role of leptin is to regulate hunger and satiety acting on specific hypothalamic nuclei. Beyond this key metabolic function; leptin also regulates many aspects of development and functioning of neuronal hippocampal networks throughout life. Here we show that leptin controls chloride homeostasis in the developing rat hippocampus in vitro. The effect of leptin relies on the down-regulation of the potassium/chloride extruder KCC2 activity and is present during a restricted period of postnatal development. This study confirms and extends the role of leptin in the ontogenesis of functional GABAergic inhibition and helps understanding how abnormal levels of leptin may contribute to neurological disorders.
Collapse
Affiliation(s)
- Camille Dumon
- Aix-Marseille Univ UMR 1249, INSERM (Institut National de La Santé et de La Recherche Médicale) Unité 1249, INMED (Institut de Neurobiologie de La Méditerranée), Parc Scientifique de Luminy, Marseille, France
- Neurochlore Parc Scientifique et Technologique de Luminy, Bâtiment Beret Delaage, Zone Luminy Entreprises Biotech, Marseille, France
| | - Yasmine Belaidouni
- Aix-Marseille Univ UMR 1249, INSERM (Institut National de La Santé et de La Recherche Médicale) Unité 1249, INMED (Institut de Neurobiologie de La Méditerranée), Parc Scientifique de Luminy, Marseille, France
| | - Diabe Diabira
- Aix-Marseille Univ UMR 1249, INSERM (Institut National de La Santé et de La Recherche Médicale) Unité 1249, INMED (Institut de Neurobiologie de La Méditerranée), Parc Scientifique de Luminy, Marseille, France
| | - Suzanne M Appleyard
- Program in Neuroscience, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA
| | - Gary A Wayman
- Program in Neuroscience, Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA
| | - Jean-Luc Gaiarsa
- Aix-Marseille Univ UMR 1249, INSERM (Institut National de La Santé et de La Recherche Médicale) Unité 1249, INMED (Institut de Neurobiologie de La Méditerranée), Parc Scientifique de Luminy, Marseille, France.
| |
Collapse
|
16
|
Hypothalamic and Cell-Specific Transcriptomes Unravel a Dynamic Neuropil Remodeling in Leptin-Induced and Typical Pubertal Transition in Female Mice. iScience 2020; 23:101563. [PMID: 33083731 PMCID: PMC7522126 DOI: 10.1016/j.isci.2020.101563] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/16/2020] [Accepted: 09/10/2020] [Indexed: 01/01/2023] Open
Abstract
Epidemiological and genome-wide association studies (GWAS) have shown high correlation between childhood obesity and advance in puberty. Early age at menarche is associated with a series of morbidities, including breast cancer, cardiovascular diseases, type 2 diabetes, and obesity. The adipocyte hormone leptin signals the amount of fat stores to the neuroendocrine reproductive axis via direct actions in the brain. Using mouse genetics, we and others have identified the hypothalamic ventral premammillary nucleus (PMv) and the agouti-related protein (AgRP) neurons in the arcuate nucleus (Arc) as primary targets of leptin action in pubertal maturation. However, the molecular mechanisms underlying leptin's effects remain unknown. Here we assessed changes in the PMv and Arc transcriptional program during leptin-stimulated and typical pubertal development using overlapping analysis of bulk RNA sequecing, TRAP sequencing, and the published database. Our findings demonstrate that dynamic somatodendritic remodeling and extracellular space organization underlie leptin-induced and typical pubertal maturation in female mice. MBH DEGs between lean and Lepob mice are highly represented in development Short-term leptin to Lepob mice alters MBH DEGs associated with reproduction PMv/Arc LepRb DEGs between lean and Lepob mice are abundant in extracellular space DEGs in developing PMv/Arc are conspicuous in extracellular and neuropil remodeling
Collapse
|
17
|
Leptin Sensitizes NTS Neurons to Vagal Input by Increasing Postsynaptic NMDA Receptor Currents. J Neurosci 2020; 40:7054-7064. [PMID: 32817248 DOI: 10.1523/jneurosci.1865-19.2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/05/2019] [Accepted: 07/27/2020] [Indexed: 11/21/2022] Open
Abstract
Leptin signaling within the nucleus of the solitary tract (NTS) contributes to the control of food intake, and injections of leptin into the NTS reduce meal size and increase the efficacy of vagus-mediated satiation signals. Leptin receptors (LepRs) are expressed by vagal afferents as well as by a population of NTS neurons. However, the electrophysiological properties of LepR-expressing NTS neurons have not been well characterized, and it is unclear how leptin might act on these neurons to reduce food intake. To address this question, we recorded from LepR-expressing neurons in horizontal brain slices containing the NTS from male and female LepR-Cre X Rosa-tdTomato mice. We found that the vast majority of NTS LepR neurons received monosynaptic innervation from vagal afferent fibers and LepR neurons exhibited large synaptic NMDA receptor (NMDAR)-mediated currents compared with non-LepR neurons. During high-frequency stimulation of vagal afferents, leptin increased the size of NMDAR-mediated currents, but not AMPAR-mediated currents. Leptin also increased the size of evoked EPSPs and the ability of low-intensity solitary tract stimulation to evoke action potentials in LepR neurons. These effects of leptin were blocked by bath applying a competitive NMDAR antagonist (DCPP-ene) or by an NMDAR channel blocker applied through the recording pipette (MK-801). Last, feeding studies using male rats demonstrate that intra-NTS injections of DCPP-ene attenuate reduction of overnight food intake following intra-NTS leptin injection. Our results suggest that leptin acts in the NTS to reduce food intake by increasing NMDAR-mediated currents, thus enhancing NTS sensitivity to vagal inputs.SIGNIFICANCE STATEMENT Leptin is a hormone that critically impacts food intake and energy homeostasis. The nucleus of the solitary tract (NTS) is activated by vagal afferents from the gastrointestinal tract, which promotes termination of a meal. Injection of leptin into the NTS inhibits food intake, while knockdown of leptin receptors (LepRs) in NTS neurons increases food intake. However, little was known about how leptin acts in the NTS neurons to inhibit food intake. We found that leptin increases the sensitivity of LepR-expressing neurons to vagal inputs by increasing NMDA receptor-mediated synaptic currents and that NTS NMDAR activation contributes to leptin-induced reduction of food intake. These findings suggest a novel mechanism by which leptin, acting in the NTS, could potentiate gastrointestinal satiation signals.
Collapse
|