1
|
Aberle T, Wegner M. Like a G6-nal: transcriptional control of G-protein coupled receptors during oligodendroglial development. Neural Regen Res 2025; 20:2001-2002. [PMID: 39254558 PMCID: PMC11691466 DOI: 10.4103/nrr.nrr-d-24-00520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/06/2024] [Accepted: 06/20/2024] [Indexed: 09/11/2024] Open
Affiliation(s)
- Tim Aberle
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Wegner
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
2
|
He T, Qin L, Chen S, Huo S, Li J, Zhang F, Yi W, Mei Y, Xiao G. Bone-derived factors mediate crosstalk between skeletal and extra-skeletal organs. Bone Res 2025; 13:49. [PMID: 40307216 PMCID: PMC12044029 DOI: 10.1038/s41413-025-00424-1] [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: 10/28/2024] [Revised: 03/15/2025] [Accepted: 03/20/2025] [Indexed: 05/02/2025] Open
Abstract
Bone has long been acknowledged as a fundamental structural entity that provides support and protection to the body's organs. However, emerging research indicates that bone plays a crucial role in the regulation of systemic metabolism. This is achieved through the secretion of a variety of hormones, cytokines, metal ions, extracellular vesicles, and other proteins/peptides, collectively referred to as bone-derived factors (BDFs). BDFs act as a medium through which bones can exert targeted regulatory functions upon various organs, thereby underscoring the profound and concrete implications of bone in human physiology. Nevertheless, there remains a pressing need for further investigations to elucidate the underlying mechanisms that inform the effects of bone on other body systems. This review aims to summarize the current findings related to the roles of these significant modulators across different organs and metabolic contexts by regulating critical genes and signaling pathways in vivo. It also addresses their involvement in the pathogenesis of various diseases affecting the musculoskeletal system, circulatory system, glucose and lipid metabolism, central nervous system, urinary system, and reproductive system. The insights gained from this review may contribute to the development of innovative therapeutic strategies through a focused approach to bone secretomes. Continued research into BDFs is expected to enhance our understanding of bone as a multifunctional organ with diverse regulatory roles in human health.
Collapse
Affiliation(s)
- Tailin He
- Department of Rheumatology and Immunology, Shenzhen Third People's Hospital, Shenzhen, 518112, China
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), 100101, Beijing, China
- Department of Biochemistry, Homeostatic Medicine Institute, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Lei Qin
- Department of Orthopedics, Shenzhen Nanshan People's Hospital, and the 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, 518052, China
| | - Sheng Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Shaochuan Huo
- Shenzhen Hospital of Guangzhou University of Chinese Medicine (Futian), Shenzhen 518000, China, Shenzhen Research Institute of Guangzhou University of Traditional Medicine (Futian), Shenzhen, 518000, China
| | - Jie Li
- Department of Biochemistry, Homeostatic Medicine Institute, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China
| | - Fuping Zhang
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), 100101, Beijing, China
| | - Weihong Yi
- Department of Orthopedics, Shenzhen Nanshan People's Hospital, and the 6th Affiliated Hospital of Shenzhen University Medical School, Shenzhen, 518052, China
| | - Yifang Mei
- Department of Rheumatology and Immunology, Shenzhen Third People's Hospital, Shenzhen, 518112, China.
| | - Guozhi Xiao
- Department of Biochemistry, Homeostatic Medicine Institute, School of Medicine, Shenzhen Key Laboratory of Cell Microenvironment, Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Southern University of Science and Technology, Shenzhen, 518055, China.
| |
Collapse
|
3
|
Li J, Lou S, Bian X. Osteocalcin and GPR158: linking bone and brain function. Front Cell Dev Biol 2025; 13:1564751. [PMID: 40337551 PMCID: PMC12055796 DOI: 10.3389/fcell.2025.1564751] [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: 01/22/2025] [Accepted: 04/10/2025] [Indexed: 05/09/2025] Open
Abstract
Osteocalcin (OCN), a small protein secreted by osteoblasts, has attracted significant attention for its role as an endocrine factor in regulating the central nervous system (CNS) via the bone-brain axis. As a critical receptor for OCN, G protein-coupled receptor 158 (GPR158) facilitates the proliferation, differentiation, and survival of neural cells while directly influencing neurons' structural and functional plasticity, thereby modulating cognitive function. Additionally, GPR158 is involved in cellular energy metabolism and interacts with proteins such as regulators of G protein signaling 7 (RGS7), broadening the understanding of OCN's impact on neural activity. Notably, GPR158 displays region- and cell type-specific bidirectional effects under certain pathological conditions, such as tumor development and mood regulation, adding complexity to its mechanisms of action. Although the precise biological mechanisms underlying the OCN/GPR158 signaling pathway remain incompletely understood, its association with neurodegenerative diseases (NDs), including Alzheimer's disease (AD) and Parkinson's disease (PD), is becoming increasingly evident. Thus, a systematic summary of OCN/GPR158 in CNS regulation and NDs will deepen understanding of its role in brain function and support the development of new therapeutic targets and strategies.
Collapse
Affiliation(s)
- Jingjing Li
- Physical Education College, Shanghai University, Shanghai, China
| | - Shujie Lou
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Xuepeng Bian
- Department of Rehabilitation, School of International Medical Technology, Shanghai Sanda University, Shanghai, China
| |
Collapse
|
4
|
Freus NK, Wank I, Häfele M, Kalinichenko LS, Müller CP, Strobelt S, Ludwig A, Hess A, Kreitz S. Pharmacological and resting state fMRI reveal Osteocalcin's effects on mouse brain regions with high Gpr37 and Gpr158 expression. Sci Rep 2025; 15:10116. [PMID: 40128223 PMCID: PMC11933355 DOI: 10.1038/s41598-025-95000-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/18/2025] [Indexed: 03/26/2025] Open
Abstract
Osteocalcin (OCN) is an endocrine hormone that signals in the periphery, regulating male fertility, energy expenditure and glucose homeostasis. It can also cross the blood-brain-barrier and act on the brain via receptors GPR37 and GPR158. In the brain, OCN influences neurotransmitter synthesis of serotonin, norepinephrine, and dopamine. OCN's function is related to cognitive and memory performance and lack of OCN is associated with anxiety and depression-like behavior in mice. We used multiparametric magnetic resonance imaging (MRI) including pharmacological MRI and resting state functional MRI, along with gene expression data for Gpr37 and Gpr158 to investigate the physiological effects of intravenously administered OCN on the wild type mouse brain. We found four core brain regions (brainstem, limbic output, association cortex, and basal ganglia) that are highly relevant in all three analytical modalities (i.e. pharmacological, resting state MRI and gene expression) and play therefore a major role in mediating OCN's effect in the brain. This study provides the first imaging data of the physiological impact of OCN on the mouse brain, suggesting its potential role in modulating brain function and its relevance as a candidate for further investigation in anxiety, depression, and cognitive impairments.
Collapse
Affiliation(s)
- Natalia K Freus
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-University Erlangen-Nuremberg, Fahrstraße 17, 91054, Erlangen, Germany
| | - Isabel Wank
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-University Erlangen-Nuremberg, Fahrstraße 17, 91054, Erlangen, Germany
| | - Maximilian Häfele
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-University Erlangen-Nuremberg, Fahrstraße 17, 91054, Erlangen, Germany
| | - Liubov S Kalinichenko
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Schwabachanlage 6, 91054, Erlangen, Germany
| | - Christian P Müller
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, Schwabachanlage 6, 91054, Erlangen, Germany
- Central Institute for Mental Health, J5, 68159, Mannheim, Germany
| | - Sandra Strobelt
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-University Erlangen-Nuremberg, Fahrstraße 17, 91054, Erlangen, Germany
| | - Andreas Ludwig
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-University Erlangen-Nuremberg, Fahrstraße 17, 91054, Erlangen, Germany
- FAU NeW - Research Center for New Bioactive Compounds, Friedrich-Alexander- University Erlangen-Nuremberg, Erlangen, Germany
| | - Andreas Hess
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-University Erlangen-Nuremberg, Fahrstraße 17, 91054, Erlangen, Germany
- Institute of Neuroradiology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Schwabachanlage 6, 91054, Erlangen, Germany
- FAU NeW - Research Center for New Bioactive Compounds, Friedrich-Alexander- University Erlangen-Nuremberg, Erlangen, Germany
| | - Silke Kreitz
- Institute of Experimental and Clinical Pharmacology and Toxicology, Friedrich-Alexander-University Erlangen-Nuremberg, Fahrstraße 17, 91054, Erlangen, Germany.
- Institute of Neuroradiology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nuremberg, Schwabachanlage 6, 91054, Erlangen, Germany.
| |
Collapse
|
5
|
Tang Y, Wang Z, Cao J, Tu Y. Bone-brain crosstalk in osteoarthritis: pathophysiology and interventions. Trends Mol Med 2025; 31:281-295. [PMID: 39438197 DOI: 10.1016/j.molmed.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 09/23/2024] [Accepted: 09/26/2024] [Indexed: 10/25/2024]
Abstract
Osteoarthritis (OA) is a prevalent articular disorder characterized by joint degeneration and persistent pain; it imposes a significant burden on both individuals and society. While OA has traditionally been viewed as a localized peripheral disorder, recent preclinical and clinical studies have revealed the crucial interconnections between the bone and the brain, highlighting the systemic nature of OA. The neuronal pathway, molecular signaling, circadian rhythms, and genetic underpinnings within the bone-brain axis play vital roles in the complex interplay that contributes to OA initiation and progression. This review explores emerging evidence of the crosstalk between the bone and brain in OA progression, and discusses the potential contributions of the bone-brain axis to the development of effective interventions for managing OA.
Collapse
Affiliation(s)
- Yilan Tang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhiyan Wang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jin Cao
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100105, China
| | - Yiheng Tu
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing 100101, China; Department of Psychology, University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
6
|
Rivagorda M, Romeo-Guitart D, Blanchet V, Mailliet F, Boitez V, Barry N, Milunov D, Siopi E, Goudin N, Moriceau S, Guerrera C, Leibovici M, Saha S, Codogno P, Morselli E, Morel E, Armand AS, Oury F. A primary cilia-autophagy axis in hippocampal neurons is essential to maintain cognitive resilience. NATURE AGING 2025; 5:450-467. [PMID: 39984747 PMCID: PMC11922775 DOI: 10.1038/s43587-024-00791-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 12/06/2024] [Indexed: 02/23/2025]
Abstract
Blood-borne factors are essential to maintain neuronal synaptic plasticity and cognitive resilience throughout life. One such factor is osteocalcin (OCN), a hormone produced by osteoblasts that influences multiple physiological processes, including hippocampal neuronal homeostasis. However, the mechanism through which this blood-borne factor communicates with neurons remains unclear. Here we show the importance of a core primary cilium (PC) protein-autophagy axis in mediating the effects of OCN. We found that the OCN receptor GPR158 is present at the PC of hippocampal neurons and mediates the regulation of autophagy machinery by OCN. During aging, autophagy and PC core proteins are reduced in neurons, and restoring their levels is sufficient to improve cognitive impairments in aged mice. Mechanistically, the induction of this axis by OCN is dependent on the PC-dependent cAMP response element-binding protein signaling pathway. Altogether, this study demonstrates that the PC-autophagy axis is a gateway to mediate communication between blood-borne factors and neurons, and it advances understanding of the mechanisms involved in age-related cognitive decline.
Collapse
Affiliation(s)
- Manon Rivagorda
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
- Institute for Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
| | - David Romeo-Guitart
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
| | - Victoria Blanchet
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
| | - François Mailliet
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
| | - Valérie Boitez
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
| | - Natalie Barry
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
| | | | - Eleni Siopi
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
| | - Nicolas Goudin
- Platform for Image Analyses, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UAR 3633, Paris, France
| | - Stéphanie Moriceau
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
- Platform for Neurobehavioral and Metaboblism, Institut Imagine, Structure Fédérative de Recherche Necker, 26 INSERM US24/CNRS UAR, Paris, France
| | - Chiara Guerrera
- Platform for Proteomic Analyses, Structure Fédérative de Recherche Necker, INSERM US24/CNRS UAR 3633, Paris, France
| | - Michel Leibovici
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
| | | | - Patrice Codogno
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 6, Paris, France
| | - Eugenia Morselli
- Department of Basic Sciences, Faculty of Medicine and Sciences, Universidad San Sebastián, Santiago, Chile
| | - Etienne Morel
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 6, Paris, France
| | - Anne-Sophie Armand
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France
| | - Franck Oury
- Université Paris Cité, INSERM UMR-U1151, CNRS UMR-8253, Institut Necker Enfants Malades, Team 8, Paris, France.
| |
Collapse
|
7
|
Ma Q, Tian JL, Lou Y, Guo R, Ma XR, Wu JB, Yang J, Tang BJ, Li S, Qiu M, Duan S, Zhao JW, Zhang J, Xu ZZ. Oligodendrocytes drive neuroinflammation and neurodegeneration in Parkinson's disease via the prosaposin-GPR37-IL-6 axis. Cell Rep 2025; 44:115266. [PMID: 39913287 DOI: 10.1016/j.celrep.2025.115266] [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: 11/15/2023] [Revised: 10/28/2024] [Accepted: 01/15/2025] [Indexed: 02/28/2025] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease and is difficult to treat due to its elusive mechanisms. Recent studies have identified a striking association between oligodendrocytes and PD progression, yet how oligodendrocytes regulate the pathogenesis of PD is still unknown. Here, we show that G-protein-coupled receptor 37 (GPR37) is upregulated in oligodendrocytes of the substantia nigra and that prosaposin (PSAP) secretion is increased in parkinsonian mice. The released PSAP can induce interleukin (IL)-6 upregulation and secretion from oligodendrocytes via a GPR37-dependent pathway, resulting in enhanced neuroinflammation, dopamine neuron degeneration, and behavioral deficits. GPR37 deficiency in oligodendrocytes prevents neurodegeneration in multiple PD models. Finally, the hallmarks of the PSAP-GPR37-IL-6 axis are observed in patients with PD. Thus, our results reveal that dopaminergic neurons interact with oligodendrocytes via secreted PSAP, and our findings identify the PSAP-GPR37-IL-6 axis as a driver of PD pathogenesis and a potential therapeutic target that might alleviate PD progression in patients.
Collapse
Affiliation(s)
- Qiang Ma
- Department of Anesthesiology of First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Center for Rehabilitation Medicine, Department of Anesthesiology and Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China.
| | - Jin-Lan Tian
- Department of Anesthesiology of First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China; Nanhu Brain-Computer Interface Institute, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 311100, China
| | - Yao Lou
- Center for Rehabilitation Medicine, Department of Anesthesiology and Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China
| | - Ran Guo
- Center for Rehabilitation Medicine, Department of Anesthesiology and Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China.
| | - Xiao-Ru Ma
- Department of Pathology of Sir Run Run Shaw Hospital and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jian-Bin Wu
- Department of Pathology of Sir Run Run Shaw Hospital and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jing Yang
- Department of Anesthesiology of First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Center for Rehabilitation Medicine, Department of Anesthesiology and Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China
| | - Bing-Jie Tang
- Department of Anesthesiology of First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China; Nanhu Brain-Computer Interface Institute, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 311100, China
| | - Shun Li
- Center for Rehabilitation Medicine, Department of Anesthesiology and Department of Pain Management, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou 310014, China
| | - Mengsheng Qiu
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou 311121, China
| | - Shumin Duan
- Department of Anesthesiology of First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China; Nanhu Brain-Computer Interface Institute, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 311100, China
| | - Jing-Wei Zhao
- Department of Pathology of Sir Run Run Shaw Hospital and Department of Human Anatomy, Histology and Embryology, System Medicine Research Center, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Jing Zhang
- Department of Anesthesiology of First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China; Department of Pathology of First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310002, China; National Health and Disease Human Brain Tissue Resource Center, Zhejiang University, Hangzhou 310002, China
| | - Zhen-Zhong Xu
- Department of Anesthesiology of First Affiliated Hospital and School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, China; Liangzhu Laboratory, MOE Frontier Science Center for Brain Science and Brain-machine Integration, State Key Laboratory of Brain-machine Intelligence, Zhejiang University, Hangzhou 311121, China; Nanhu Brain-Computer Interface Institute, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University, Hangzhou 311100, China.
| |
Collapse
|
8
|
Zhao X, Yao M, Wang Y, Feng C, Yang Y, Tian L, Bao C, Li X, Zhu X, Zhang X. Neuroregulation during Bone Formation and Regeneration: Mechanisms and Strategies. ACS APPLIED MATERIALS & INTERFACES 2025; 17:7223-7250. [PMID: 39869030 DOI: 10.1021/acsami.4c16786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
The skeleton is highly innervated by numerous nerve fibers. These nerve fibers, in addition to transmitting information within the bone and mediating bone sensations, play a crucial role in regulating bone tissue formation and regeneration. Traditional bone tissue engineering (BTE) often fails to achieve satisfactory outcomes when dealing with large-scale bone defects, which is frequently related to the lack of effective reconstruction of the neurovascular network. In recent years, increasing research has revealed the critical role of nerves in bone metabolism. Nerve fibers regulate bone cells through neurotransmitters, neuropeptides, and peripheral glial cells. Furthermore, nerves also coordinate with the vascular and immune systems to jointly construct a microenvironment favorable for bone regeneration. As a signaling driver of bone formation, neuroregulation spans the entire process of bone physiological activities from the embryonic formation to postmaturity remodeling and repair. However, there is currently a lack of comprehensive summaries of these regulatory mechanisms. Therefore, this review sketches out the function of nerves during bone formation and regeneration. Then, we elaborate on the mechanisms of neurovascular coupling and neuromodulation of bone immunity. Finally, we discuss several novel strategies for neuro-bone tissue engineering (NBTE) based on neuroregulation of bone, focusing on the coordinated regeneration of nerve and bone tissue.
Collapse
Affiliation(s)
- Xiangrong Zhao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Meilin Yao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Yuyi Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Cong Feng
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Yuhan Yang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Luoqiang Tian
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Chongyun Bao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Med-X Center for Materials, Sichuan University, Chengdu 610041, Sichuan, China
| | - Xiangfeng Li
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xiangdong Zhu
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu 610064, China
| |
Collapse
|
9
|
Chen H, Wang W, Cui W, Tu C, Han Y, Zhang C, Yang L, Huang X, Zhang Q, Lu L. Seipin Deficiency Impairs Motor Coordination in Mice by Compromising Spinal Cord Myelination. Neuromolecular Med 2025; 27:12. [PMID: 39869141 DOI: 10.1007/s12017-025-08834-4] [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: 12/17/2024] [Accepted: 01/17/2025] [Indexed: 01/28/2025]
Abstract
The integrity of the myelin sheath of the spinal cord (SC) is essential for motor coordination. Seipin is an endoplasmic reticulum transmembrane protein highly expressed in adipose tissue and motor neurons in the SC. It was reported Seipin deficiency induced lipid dysregulation and neurobehavioral deficits, but the underlying mechanism, especially in SC, remains to be elucidated. In present study, we found that Seipin and myelin basic protein (MBP) increased synchronously in SC of developmental stage of mice. Demyelination impaired motor coordination as well as MBP and Seipin expression, which were alleviated by remyelination. Moreover, Seipin deficiency impaired motor coordination of mice, accompanied by hypomyelination in spinal cord. Mechanistically, we further demonstrated that myelin content as labeled by Fluormyelin, myelin basic protein (MBP) was down-regulated by Seipin deficiency. Seipin deficiency led to reduction of myelin-forming oligodendrocytes (OLs) density in spinal cord. Notably, administration of rosiglitazone (RG), a classic PPARγ activator, successfully restored the phenotypes manifested by Seipin deficiency including reduced OLs density, hypomyelination, as well as motor dyscoordination. In summary, present study revealed that Seipin deficiency disrupted motor coordination by compromising myelination in SC, and RG treatment could rescue the phenotypes. This study throws light on the mechanism underlying Seipin deficiency associated disorders and paves ways for developing therapeutics toward those diseases.
Collapse
Affiliation(s)
- Hong Chen
- Department of Anatomy, School of Basic Medical Sciences, Shanxi Medical University, No 56, Xinjian Nan Road, Taiyuan, 030001, Shanxi, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
- Department of Spine Surgery, Yuncheng Central Hospital of Shanxi Province, Shanxi Medical University, No. 3690, Hedong East Road, Yanhu District, Yuncheng, 044020, Shanxi, China
| | - Wenru Wang
- Department of Anatomy, School of Basic Medical Sciences, Shanxi Medical University, No 56, Xinjian Nan Road, Taiyuan, 030001, Shanxi, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Wenli Cui
- Department of Anatomy, School of Basic Medical Sciences, Shanxi Medical University, No 56, Xinjian Nan Road, Taiyuan, 030001, Shanxi, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Chuanyun Tu
- Department of Anatomy, School of Basic Medical Sciences, Shanxi Medical University, No 56, Xinjian Nan Road, Taiyuan, 030001, Shanxi, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Yuanyuan Han
- Department of Anatomy, School of Basic Medical Sciences, Shanxi Medical University, No 56, Xinjian Nan Road, Taiyuan, 030001, Shanxi, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Chengwu Zhang
- Department of Anatomy, School of Basic Medical Sciences, Shanxi Medical University, No 56, Xinjian Nan Road, Taiyuan, 030001, Shanxi, China
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Liu Yang
- First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Xintao Huang
- First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China
| | - Qin Zhang
- Department of Spine Surgery, Yuncheng Central Hospital of Shanxi Province, Shanxi Medical University, No. 3690, Hedong East Road, Yanhu District, Yuncheng, 044020, Shanxi, China.
| | - Li Lu
- Department of Anatomy, School of Basic Medical Sciences, Shanxi Medical University, No 56, Xinjian Nan Road, Taiyuan, 030001, Shanxi, China.
- Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
10
|
Bian X, Wang Y, Zhang W, Ye C, Li J. GPR37 and its neuroprotective mechanisms: bridging osteocalcin signaling and brain function. Front Cell Dev Biol 2024; 12:1510666. [PMID: 39633709 PMCID: PMC11614806 DOI: 10.3389/fcell.2024.1510666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Accepted: 11/11/2024] [Indexed: 12/07/2024] Open
Abstract
Osteocalcin (OCN) is a hormone secreted by osteoblasts and has attracted widespread attention for its role in regulating brain function. Clinical studies indicate a positive correlation between levels of circulating OCN and cognitive performance. Indeed, lower circulating OCN has been detected in various neurodegenerative diseases (NDs), while OCN supplementation under certain conditions may improve cognitive function. GPR37, a G protein-coupled receptor, has recently been identified as a receptor for OCN. It exhibits distinct expression patterns across various brain regions and cell types, potentially influencing its functional roles within the brain. Research indicates that GPR37 regulates neuronal migration, cell proliferation, differentiation, and myelination. Furthermore, GPR37 has been shown to mitigate inflammation and apoptosis through various mechanisms, exerting neuroprotective effects. However, its regulatory influence on brain function exhibits inconsistency, highlighting a duality in its actions. Therefore, this review thoroughly summarizes the roles and mechanisms of GPR37 in modulating cellular physiological activities and its involvement in immune responses, stress reactions, and neuroprotection. It aims to enhance the understanding of how GPR37 modulates brain function and facilitate the identification of novel therapeutic targets or strategies for related diseases.
Collapse
Affiliation(s)
- Xuepeng Bian
- Department of Rehabilitation, School of International Medical Technology, Shanghai Sanda University, Shanghai, China
| | - Yangping Wang
- Physical Education College, Shanghai University, Shanghai, China
| | - Weijie Zhang
- Physical Education College, Shanghai University, Shanghai, China
| | - Changlin Ye
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Jingjing Li
- Physical Education College, Shanghai University, Shanghai, China
| |
Collapse
|
11
|
Cherchi F, Swire M, Lecca D. Editorial: Role of ion channels and metabotropic receptors in oligodendrogliogenesis: novel targets for demyelinating pathologies. Front Cell Neurosci 2024; 18:1517363. [PMID: 39600308 PMCID: PMC11588474 DOI: 10.3389/fncel.2024.1517363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 10/31/2024] [Indexed: 11/29/2024] Open
Affiliation(s)
- Federica Cherchi
- Department of Neuroscience, Psychology, Drugs and Child Health Area, Università degli Studi di Firenze, Florence, Italy
| | - Matthew Swire
- Wolfson Institute for Biomedical Research, University College London, London, United Kingdom
| | - Davide Lecca
- Department of Pharmaceutical Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
12
|
Wang X, Ju J, Xie Y, Hang L. Emerging roles of the G-protein-coupled receptor 37 in neurological diseases and pain. Neuroscience 2024; 559:199-208. [PMID: 39244010 DOI: 10.1016/j.neuroscience.2024.08.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 08/11/2024] [Accepted: 08/21/2024] [Indexed: 09/09/2024]
Abstract
Neurological disorders and pain are prevalent clinical issues that severely impact patients' quality of life and daily functioning. With the advancing exploration of these disease mechanisms, G protein-coupled receptor 37 (GPR37) has emerged as a critical protein, garnering widespread attention in the scientific community. As a member of the G protein-coupled receptor family, GPR37 features a seven-transmembrane helix structure and is widely expressed in various brain regions, including the substantia nigra and striatum. In addition to neurons, GPR37 is also detected in immune cells within the nervous system, indicating its potential role in neuron-immune cell interactions. Research has shown that the expression level of GPR37 in neurological disorders can affect neuron survival, cellular signaling, and overall neurological health. Abnormal expression of GPR37 is often associated with disease progression and symptom exacerbation in neurological disorders such as Parkinson's disease and stroke. In the context of pain, GPR37 alleviates pain and inflammatory responses by regulating the phagocytic activity and polarization state of macrophages. This article aims to delve into the mechanistic roles of GPR37 in neurological disorders and pain. Through a comprehensive literature review, we summarize the latest research on GPR37's involvement in neurological diseases and pain, highlighting its critical roles in neural signaling, inflammatory responses, and neuroprotection. This understanding expands the comprehension of GPR37's biological functions and provides new perspectives for improving the clinical outcomes of patients with neurological disorders and pain.
Collapse
Affiliation(s)
- Xinxin Wang
- Department of Anesthesiology, Kunshan Hospital Affiliated to Jiangsu University, Suzhou, 215300, China.
| | - Jiajun Ju
- Gusu College, Nanjing Medical University, Department of Anesthesiology, The First People's Hospital of Kunshan, Suzhou 215300, China.
| | - Yafei Xie
- Department of Anesthesiology, Kunshan Hospital Affiliated to Jiangsu University, Suzhou, 215300, China.
| | - Lihua Hang
- Department of Anesthesiology, Kunshan Hospital Affiliated to Jiangsu University, Suzhou, 215300, China.
| |
Collapse
|
13
|
Wang W, Wang Y, Su L, Zhang M, Zhang T, Zhao J, Ma H, Zhang D, Ji F, Jiao RD, Li H, Xu Y, Chen L, Jiao J. Endothelial Cells Mediated by STING Regulate Oligodendrogenesis and Myelination During Brain Development. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308508. [PMID: 39136074 PMCID: PMC11481185 DOI: 10.1002/advs.202308508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 07/30/2024] [Indexed: 10/17/2024]
Abstract
Oligodendrocyte precursor cells (OPCs) migrate extensively using blood vessels as physical scaffolds in the developing central nervous system. Although the association of OPCs with the vasculature is critical for migration, the regulatory mechanisms important for OPCs proliferative and oligodendrocyte development are unknown. Here, a correlation is demonstrated between the developing vasculature and OPCs response during brain development. Deletion of endothelial stimulator of interferon genes (STING) disrupts angiogenesis by inhibiting farnesyl-diphosphate farnesyltransferase 1 (FDFT1) and thereby reducing cholesterol synthesis. Furthermore, the perturbation of metabolic homeostasis in endothelial cells increases interleukin 17D production which mediates the signal transduction from endothelial cells to OPCs, which inhibits oligodendrocyte development and myelination and causes behavioral abnormalities in adult mice. Overall, these findings indicate how the endothelial STING maintains metabolic homeostasis and contributes to oligodendrocyte precursor cells response in the developing neocortex.
Collapse
Affiliation(s)
- Wenwen Wang
- Key Laboratory of Organ Regeneration and ReconstructionChinese Academy of ScienceBeijing100101China
- School of Life SciencesUniversity of Science and Technology of ChinaHefei230026China
| | - Yanyan Wang
- Key Laboratory of Organ Regeneration and ReconstructionChinese Academy of ScienceBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Libo Su
- Key Laboratory of Organ Regeneration and ReconstructionChinese Academy of ScienceBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Mengtian Zhang
- Key Laboratory of Organ Regeneration and ReconstructionChinese Academy of ScienceBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Tianyu Zhang
- Key Laboratory of Organ Regeneration and ReconstructionChinese Academy of ScienceBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Jinyue Zhao
- Key Laboratory of Organ Regeneration and ReconstructionChinese Academy of ScienceBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Hongyan Ma
- Key Laboratory of Organ Regeneration and ReconstructionChinese Academy of ScienceBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Dongming Zhang
- Key Laboratory of Organ Regeneration and ReconstructionChinese Academy of ScienceBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Fen Ji
- Key Laboratory of Organ Regeneration and ReconstructionChinese Academy of ScienceBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | | | - Hong Li
- Key Laboratory of Organ Regeneration and ReconstructionChinese Academy of ScienceBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
| | - Yuming Xu
- Department of NeurologyThe First Affiliated Hospital of Zhengzhou UniversityZhengzhou450000China
| | - Lei Chen
- Department of NeurologyWest China HospitalSichuan UniversityChengdu610041China
| | - Jianwei Jiao
- Key Laboratory of Organ Regeneration and ReconstructionChinese Academy of ScienceBeijing100101China
- University of Chinese Academy of SciencesBeijing100049China
- Co‐Innovation Center of NeuroregenerationNantong UniversityNantong226001China
- Beijing Institute for Stem Cell and Regenerative MedicineInstitute for Stem Cell and RegenerationChinese Academy of SciencesBeijing100101China
| |
Collapse
|
14
|
Qian Z, Wang Z, Li B, Meng X, Kuang Z, Li Y, Yang Y, Ye K. Thy1-ApoE4/C/EBPβ double transgenic mice act as a sporadic model with Alzheimer's disease. Mol Psychiatry 2024; 29:3040-3055. [PMID: 38658772 PMCID: PMC11449781 DOI: 10.1038/s41380-024-02565-x] [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: 11/02/2023] [Revised: 04/10/2024] [Accepted: 04/15/2024] [Indexed: 04/26/2024]
Abstract
Early onset familial Alzheimer's disease (FAD) with APP, PS1/2 (presenilins) mutation accounts for only a small portion of AD cases, and most are late-onset sporadic. However, majority of AD mouse models are developed to mimic the genetic cause of human AD by overexpressing mutated forms of human APP, PS1/2, and/or Tau protein, though there is no Tau mutation in AD, and no single mouse model recapitulates all aspects of AD pathology. Here, we report Thy1-ApoE4/C/EBPβ double transgenic mouse model that demonstrates key AD pathologies in an age-dependent manner in absence of any human APP or PS1/2 mutation. Using the clinical diagnosis criteria, we show that this mouse model exhibits tempo-spatial features in AD patient brains, including progressive cognitive decline associated with brain atrophy, which is accompanied with extensive neuronal degeneration. Remarkably, the mice display gradual Aβ aggregation and neurofibrillary tangles formation in the brain validated by Aβ PET and Tau PET. Moreover, the mice reveal widespread neuroinflammation as shown in AD brains. Hence, Thy1-ApoE4/C/EBPβ mouse model acts as a sporadic AD mouse model, reconstituting the major AD pathologies.
Collapse
Affiliation(s)
- Zhengjiang Qian
- Faculty of Life and Health Sciences, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China
| | - ZhiHao Wang
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, China
| | - Bowei Li
- Shenzhen Institute of Advanced Technology, University of Chinese Academy of Science, Shenzhen, Guangdong Province, 518055, China
| | - Xin Meng
- Faculty of Life and Health Sciences, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China
| | - Zhonghua Kuang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China
| | - Yanjiao Li
- Faculty of Life and Health Sciences, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China
| | - Yongfeng Yang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China
| | - Keqiang Ye
- Faculty of Life and Health Sciences, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China.
| |
Collapse
|
15
|
Li Q, Li C, Zhang X. Research Progress on the Effects of Different Exercise Modes on the Secretion of Exerkines After Spinal Cord Injury. Cell Mol Neurobiol 2024; 44:62. [PMID: 39352588 PMCID: PMC11445308 DOI: 10.1007/s10571-024-01497-y] [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: 06/13/2024] [Accepted: 09/16/2024] [Indexed: 10/04/2024]
Abstract
Exercise training is a conventional treatment strategy throughout the entire treatment process for patients with spinal cord injury (SCI). Currently, exercise modalities for SCI patients primarily include aerobic exercise, endurance training, strength training, high-intensity interval training, and mind-body exercises. These exercises play a positive role in enhancing skeletal muscle function, inducing neuroprotection and regeneration, thereby influencing neural plasticity, reducing limb spasticity, and improving motor function and daily living abilities in SCI patients. However, the mechanism by which exercise training promotes functional recovery after SCI is still unclear, and there is no consensus on a unified and standardized exercise treatment plan. Different exercise methods may bring different benefits. After SCI, patients' physical activity levels decrease significantly due to factors such as motor dysfunction, which may be a key factor affecting changes in exerkines. The changes in exerkines of SCI patients caused by exercise training are an important and highly relevant and visual evaluation index, which may provide a new research direction for revealing the intrinsic mechanism by which exercise promotes functional recovery after SCI. Therefore, this article summarizes the changes in the expression of common exerkines (neurotrophic factors, inflammatory factors, myokines, bioactive peptides) after SCI, and intends to analyze the impact and role of different exercise methods on functional recovery after SCI from the perspective of exerkines mechanism. We hope to provide theoretical basis and data support for scientific exercise treatment programs after SCI.
Collapse
Affiliation(s)
- Qianxi Li
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China
| | - Chenyu Li
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China
| | - Xin Zhang
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, 100084, China.
| |
Collapse
|
16
|
Zhang J, Xie L, Li H, Li S, Gao X, Zhang M. Selenomethionine Promotes Milk Protein and Fat Synthesis and Proliferation of Mammary Epithelial Cells through the GPR37-mTOR-S6K1 Signaling. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:19505-19516. [PMID: 39177123 DOI: 10.1021/acs.jafc.4c03911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2024]
Abstract
Selenomethionine (SeMet) is an important nutrient, but its role in milk synthesis and the GPCR related to SeMet sensing is still largely unknown. Here, we determined the dose-dependent role of SeMet on milk protein and fat synthesis and proliferation of mammary epithelial cells (MECs), and we also uncovered the GPCR-mediating SeMet function. At 24 h postdelivery, lactating mother mice were fed a maintenance diet supplemented with 0, 5, 10, 20, 40, and 80 mg/kg SeMet, and the feeding process lasted for 18 days. The 10 mg/kg group had the best increase in milk production, weight gain of offspring mice, and mammary gland weight and acinar size, whereas a higher concentration of SeMet gradually decreased the weight gain of the offspring mice and showed toxic effects. Transcriptome sequencing was performed to find the differentially expressed genes (DEGs) between the mammary gland tissues of mother mice in the 10 mg/kg SeMet treatment group and the control group. A total of 258 DEGs were screened out, including 82 highly expressed genes including GPR37 and 176 lowly expressed genes. SeMet increased milk protein and fat synthesis in HC11 cells and cell proliferation, mTOR and S6K1 phosphorylation, and expression of GPR37 in a dose-dependent manner. GPR37 knockdown decreased milk protein and fat synthesis in HC11 cells and cell proliferation and blocked SeMet stimulation on mTOR and S6K1 phosphorylation. Taken together, our data demonstrate that SeMet can promote milk protein and fat synthesis and proliferation of MECs and functions through the GPR37-mTOR-S6K1 signaling pathway.
Collapse
Affiliation(s)
- Jinlong Zhang
- College of Animal Science and Technology, Yangtze University, Jingmi Road 88, Jingzhou 434025, China
| | - Liping Xie
- College of Animal Science and Technology, Yangtze University, Jingmi Road 88, Jingzhou 434025, China
| | - Heqian Li
- College of Animal Science and Technology, Yangtze University, Jingmi Road 88, Jingzhou 434025, China
| | - Siqi Li
- College of Animal Science and Technology, Yangtze University, Jingmi Road 88, Jingzhou 434025, China
| | - Xuejun Gao
- College of Animal Science and Technology, Yangtze University, Jingmi Road 88, Jingzhou 434025, China
| | - Minghui Zhang
- College of Animal Science and Technology, Yangtze University, Jingmi Road 88, Jingzhou 434025, China
| |
Collapse
|
17
|
Qi X, He X, Peng Y, He X, Yang Q, Jiao K, Liu H. Roles of osteocalcin in the central nervous system. CNS Neurosci Ther 2024; 30:e70016. [PMID: 39252492 PMCID: PMC11386255 DOI: 10.1111/cns.70016] [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: 06/27/2024] [Revised: 08/04/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND Bone-derived protein osteocalcin, which has beneficial effects on brain function, may be a future research direction for neurological disorders. A growing body of evidence suggests a link between osteocalcin and neurological disorders, but the exact relationship is contradictory and unclear. SCOPE OF REVIEW The aim of this review is to summarize the current research on the interaction between osteocalcin and the central nervous system and to propose some speculative future research directions. MAJOR CONCLUSIONS In the normal central nervous system, osteocalcin is involved in neuronal structure, neuroprotection, and the regulation of cognition and anxiety. Studies on osteocalcin-related abnormalities in the central nervous system are divided into animal model studies and human studies, depending on the subject. In humans, the link between osteocalcin and brain function is inconsistent. These conflicting data may be due to methodological inconsistencies. By reviewing the related literature on osteocalcin, some comorbidities of the bone and nervous system and future research directions related to osteocalcin are proposed.
Collapse
Affiliation(s)
- Xiao‐Shan Qi
- Department of RadiologyAffiliated Hospital of Zunyi Medical University, Engineering Research Center of Intelligent Medical Imaging in Guizhou Higher Education lnstitutions, Medical Imaging Center of Guizhou ProvinceZunyiChina
- The First Clinical Medical CollegeZunyi Medical UniversityZunyiChina
| | - Xin He
- Department of RadiologyAffiliated Hospital of Zunyi Medical University, Engineering Research Center of Intelligent Medical Imaging in Guizhou Higher Education lnstitutions, Medical Imaging Center of Guizhou ProvinceZunyiChina
| | - Ying Peng
- Department of RadiologyAffiliated Hospital of Zunyi Medical University, Engineering Research Center of Intelligent Medical Imaging in Guizhou Higher Education lnstitutions, Medical Imaging Center of Guizhou ProvinceZunyiChina
| | - Xing‐Hong He
- Department of RadiologyAffiliated Hospital of Zunyi Medical University, Engineering Research Center of Intelligent Medical Imaging in Guizhou Higher Education lnstitutions, Medical Imaging Center of Guizhou ProvinceZunyiChina
| | - Qian‐Yu Yang
- The First Clinical Medical CollegeZunyi Medical UniversityZunyiChina
| | - Kai Jiao
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, School of StomatologyThe Fourth Military Medical UniversityXi‘anChina
| | - Heng Liu
- Department of RadiologyAffiliated Hospital of Zunyi Medical University, Engineering Research Center of Intelligent Medical Imaging in Guizhou Higher Education lnstitutions, Medical Imaging Center of Guizhou ProvinceZunyiChina
| |
Collapse
|
18
|
Li Z, Li J, Qu G, Chen K, Liu Y, Li S, Chen C, Zhao Y, Huang J, Wang P, Wu X, Ren J. Multiscale hydrogel regulates mesenchymal stem cell fate for bone regeneration. CELL REPORTS PHYSICAL SCIENCE 2024; 5:102181. [DOI: 10.1016/j.xcrp.2024.102181] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
|
19
|
Yang M, Lin Z, Zhuang L, Pan L, Wang R, Chen H, Hu Z, Shen W, Zhuo J, Yang X, Li H, He C, Yang Z, Xie Q, Dong S, Chen J, Su R, Wei X, Yin J, Zheng S, Lu D, Xu X. An inflammatory liquid fingerprint predicting tumor recurrence after liver transplantation for hepatocellular carcinoma. MedComm (Beijing) 2024; 5:e678. [PMID: 39188937 PMCID: PMC11345533 DOI: 10.1002/mco2.678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 08/28/2024] Open
Abstract
Tumor recurrence is a life-threatening complication after liver transplantation (LT) for hepatocellular carcinoma (HCC). Precise recurrence risk stratification before transplantation is essential for the management of recipients. Here, we aimed to establish an inflammation-related prediction model for posttransplant HCC recurrence based on pretransplant peripheral cytokine profiling. Two hundred and ninety-three patients who underwent LT in two independent medical centers were enrolled, and their pretransplant plasma samples were sent for cytokine profiling. We identified four independent risk factors, including alpha-fetoprotein, systemic immune-inflammation index, interleukin 6, and osteocalcin in the training cohort (n = 190) by COX regression analysis. A prediction model named inflammatory fingerprint (IFP) was established based on the above factors. The IFP effectively predicted posttransplant recurrence (area under the receiver operating characteristic curve [AUROC]: 0.792, C-index: 0.736). The high IFP group recipients had significantly worse 3-year recurrence-free survival rates (37.9 vs. 86.9%, p < 0.001). Simultaneous T-cell profiling revealed that recipients with high IFP were characterized by impaired T cell function. The IFP also performed well in the validation cohort (n = 103, AUROC: 0.807, C-index: 0.681). In conclusion, the IFP efficiently predicted posttransplant HCC recurrence and helped to refine pretransplant risk stratification. Impaired T cell function might be the intrinsic mechanism for the high recurrence risk of recipients in the high IFP group.
Collapse
Affiliation(s)
- Modan Yang
- Department of Breast SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhouChina
- NHC Key Laboratory of Combined Multi‐Organ TransplantationZhejiang UniversityHangzhouChina
| | - Zuyuan Lin
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang ProvinceAffiliated Hangzhou First People's Hospital, School of Medicine, Westlake UniversityHangzhouChina
- Zhejiang University School of MedicineHangzhouChina
| | - Li Zhuang
- Department of Hepatobiliary and Pancreatic SurgeryShulan (Hangzhou) HospitalHangzhouChina
| | - Linhui Pan
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hangzhou First People's HospitalSchool of MedicineWestlake UniversityHangzhouChina
| | - Rui Wang
- Zhejiang University School of MedicineHangzhouChina
| | - Hao Chen
- Zhejiang University School of MedicineHangzhouChina
| | - Zhihang Hu
- Zhejiang University School of MedicineHangzhouChina
| | - Wei Shen
- Zhejiang University School of MedicineHangzhouChina
| | - Jianyong Zhuo
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hangzhou First People's HospitalSchool of MedicineWestlake UniversityHangzhouChina
| | - Xinyu Yang
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang ProvinceAffiliated Hangzhou First People's Hospital, School of Medicine, Westlake UniversityHangzhouChina
- Zhejiang University School of MedicineHangzhouChina
| | - Huigang Li
- Zhejiang University School of MedicineHangzhouChina
| | - Chiyu He
- Zhejiang University School of MedicineHangzhouChina
| | - Zhe Yang
- Department of Hepatobiliary and Pancreatic SurgeryShulan (Hangzhou) HospitalHangzhouChina
| | - Qinfen Xie
- Department of Hepatobiliary and Pancreatic SurgeryShulan (Hangzhou) HospitalHangzhouChina
| | - Siyi Dong
- National Center for Healthcare Quality Management in Liver TransplantHangzhouChina
| | - Junli Chen
- National Center for Healthcare Quality Management in Liver TransplantHangzhouChina
| | - Renyi Su
- Zhejiang University School of MedicineHangzhouChina
| | - Xuyong Wei
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang ProvinceAffiliated Hangzhou First People's Hospital, School of Medicine, Westlake UniversityHangzhouChina
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hangzhou First People's HospitalSchool of MedicineWestlake UniversityHangzhouChina
| | - Junjie Yin
- Department of Hepatobiliary and Pancreatic SurgeryAffiliated Hangzhou First People's HospitalSchool of MedicineWestlake UniversityHangzhouChina
| | - Shusen Zheng
- NHC Key Laboratory of Combined Multi‐Organ TransplantationZhejiang UniversityHangzhouChina
- Department of Hepatobiliary and Pancreatic SurgeryShulan (Hangzhou) HospitalHangzhouChina
- National Center for Healthcare Quality Management in Liver TransplantHangzhouChina
| | - Di Lu
- Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive SurgeryZhejiang Provincial People's Hospital (Affiliated People's Hospital)School of Clinical MedicineHangzhou Medical CollegeHangzhouChina
| | - Xiao Xu
- Department of Hepatobiliary & Pancreatic Surgery and Minimally Invasive SurgeryZhejiang Provincial People's Hospital (Affiliated People's Hospital)School of Clinical MedicineHangzhou Medical CollegeHangzhouChina
- Institute of Translational MedicineZhejiang University School of MedicineHangzhouChina
| |
Collapse
|
20
|
Ji PC, Xie YS, Guo WK, Fu B, Chen XM. p38 Signaling Mediates Naringin-Induced Osteogenic Differentiation of Porcine Metanephric Mesenchymal Cells. Chin J Integr Med 2024; 30:818-825. [PMID: 38850479 DOI: 10.1007/s11655-024-3761-1] [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] [Accepted: 01/23/2024] [Indexed: 06/10/2024]
Abstract
OBJECTIVE To explore the potential of metanephric mesenchymal cells (MMCs) for osteogenesis and naringin's ability to enhance this process and its molecular mechanism. METHODS Porcine MMCs at 70 days of gestation were used as tool cells, cultured in osteogenic induction medium, identified by immunocytochemistry staining. Osteogenic potential of porcine MMCs and naringin's ability to enhance this process was tested by detecting changes in cell viability, alkaline phosphatase (ALP) activity, the expression of runt-related transcription factor 2 (Runx2), osteopontin (OPN) and osteocalcin (OCN), and the formation of mineralized nodules, and the application of the p38 signaling pathway inhibitor SB203580 vitiated the osteogenesis-promoting effect of naringin. RESULTS Immunocytochemical staining showed that the cells were Vimentin and Six2(+), E-cadherin and CK-18(-). Naringin can activate the p38 signaling pathway to enhance the osteogenesis of porcine MMCs by increasing cell viability, ALP activity, the expressions of Runx2, OPN and OCN, and the formation of mineralized nodules (P<0.05). The application of p38 signaling pathway inhibitor SB203580 vitiated the osteogenesis-promoting effect of naringin, manifested by decreased ALP activity, the expressions of Runx2, OPN and OCN, and the formation of mineralized nodules (P<0.05). CONCLUSION Naringin, the active ingredient of Chinese herbal medicine Rhizoma Drynariae for nourishing Shen (Kidney) and strengthening bone, enhances the osteogenic differentiation of renal MMCs through the p38 signaling pathway.
Collapse
Affiliation(s)
- Peng-Cheng Ji
- Chinese PLA Medical School, Beijing, 100853, China
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing, 100853, China
| | - Yuan-Sheng Xie
- Chinese PLA Medical School, Beijing, 100853, China.
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing, 100853, China.
- School of Medicine, Nankai University, Tianjin, 300071, China.
| | - Wen-Kai Guo
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing, 100853, China
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Bo Fu
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing, 100853, China
| | - Xiang-Mei Chen
- Department of Nephrology, First Medical Center of Chinese PLA General Hospital, Chinese PLA Institute of Nephrology, State Key Laboratory of Kidney Diseases, National Clinical Research Center for Kidney Diseases, Beijing Key Laboratory of Kidney Diseases, Beijing, 100853, China
| |
Collapse
|
21
|
Wanionok NE, Morel GR, Fernández JM. Osteoporosis and Alzheimer´s disease (or Alzheimer´s disease and Osteoporosis). Ageing Res Rev 2024; 99:102408. [PMID: 38969142 DOI: 10.1016/j.arr.2024.102408] [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: 02/28/2024] [Revised: 07/02/2024] [Accepted: 07/02/2024] [Indexed: 07/07/2024]
Abstract
Alzheimer's disease (AD) and osteoporosis are two diseases that mainly affect elderly people, with increases in the occurrence of cases due to a longer life expectancy. Several epidemiological studies have shown a reciprocal association between both diseases, finding an increase in incidence of osteoporosis in patients with AD, and a higher burden of AD in osteoporotic patients. This epidemiological relationship has motivated the search for molecules, genes, signaling pathways and mechanisms that are related to both pathologies. The mechanisms found in these studies can serve to improve treatments and establish better patient care protocols.
Collapse
Affiliation(s)
- Nahuel E Wanionok
- Laboratorio de Osteopatías y Metabolismo Mineral (LIOMM), Facultad de Cs. Exactas. Universidad Nacional de La Plata UNLP-CIC, Argentina
| | - Gustavo R Morel
- Biochemistry Research Institute of La Plata "Professor Doctor Rodolfo R. Brenner" (INIBIOLP), Argentina
| | - Juan M Fernández
- Laboratorio de Osteopatías y Metabolismo Mineral (LIOMM), Facultad de Cs. Exactas. Universidad Nacional de La Plata UNLP-CIC, Argentina.
| |
Collapse
|
22
|
Liu Z, Liu M, Xiong Y, Wang Y, Bu X. Crosstalk between bone and brain in Alzheimer's disease: Mechanisms, applications, and perspectives. Alzheimers Dement 2024; 20:5720-5739. [PMID: 38824621 PMCID: PMC11350061 DOI: 10.1002/alz.13864] [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: 02/05/2024] [Revised: 04/01/2024] [Accepted: 04/02/2024] [Indexed: 06/04/2024]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease that involves multiple systems in the body. Numerous recent studies have revealed bidirectional crosstalk between the brain and bone, but the interaction between bone and brain in AD remains unclear. In this review, we summarize human studies of the association between bone and brain and provide an overview of their interactions and the underlying mechanisms in AD. We review the effects of AD on bone from the aspects of AD pathogenic proteins, AD risk genes, neurohormones, neuropeptides, neurotransmitters, brain-derived extracellular vesicles (EVs), and the autonomic nervous system. Correspondingly, we elucidate the underlying mechanisms of the involvement of bone in the pathogenesis of AD, including bone-derived hormones, bone marrow-derived cells, bone-derived EVs, and inflammation. On the basis of the crosstalk between bone and the brain, we propose potential strategies for the management of AD with the hope of offering novel perspectives on its prevention and treatment. HIGHLIGHTS: The pathogenesis of AD, along with its consequent changes in the brain, may involve disturbing bone homeostasis. Degenerative bone disorders may influence the progression of AD through a series of pathophysiological mechanisms. Therefore, relevant bone intervention strategies may be beneficial for the comprehensive management of AD.
Collapse
Affiliation(s)
- Zhuo‐Ting Liu
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease (Third Military Medical University)ChongqingChina
| | - Ming‐Han Liu
- Department of OrthopaedicsXinqiao Hospital, Third Military Medical UniversityChongqingChina
| | - Yan Xiong
- Department of OrthopaedicsDaping Hospital, Third Military Medical UniversityChongqingChina
| | - Yan‐Jiang Wang
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease (Third Military Medical University)ChongqingChina
- Institute of Brain and IntelligenceThird Military Medical UniversityChongqingChina
| | - Xian‐Le Bu
- Department of Neurology and Centre for Clinical NeuroscienceDaping Hospital, State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical UniversityChongqingChina
- Chongqing Key Laboratory of Ageing and Brain DiseasesChongqingChina
- Key Laboratory of Geriatric Cardiovascular and Cerebrovascular Disease (Third Military Medical University)ChongqingChina
- Institute of Brain and IntelligenceThird Military Medical UniversityChongqingChina
| |
Collapse
|
23
|
Channuwong P, Speight V, Yuan Y, Yao S, Yoshimura M, Bauermann FV, Ranjan A, Adisakwattana S, Cheng H. Hyperglycemia from Diabetes Potentiates Uncarboxylated Osteocalcin-Stimulated Insulin Secretion in Rat INS-1 Pancreatic β-Cells. Nutrients 2024; 16:2384. [PMID: 39125265 PMCID: PMC11313777 DOI: 10.3390/nu16152384] [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: 06/27/2024] [Revised: 07/16/2024] [Accepted: 07/20/2024] [Indexed: 08/12/2024] Open
Abstract
Uncarboxylated osteocalcin (ucOC) is a hormone secreted by osteoblasts that strengthens bone during mineralization and is a biomarker for ongoing bone formation. It also regulates glucose homeostasis by stimulating insulin secretion from pancreatic β-cells. However, its effect on β-cells under hyperglycemic diabetic conditions is unclear. The objective of this study was to investigate ucOC's effect on insulin secretion in β-cells maintained under high glucose conditions. We hypothesized that hyperglycemia potentiates insulin secretion in response to ucOC stimulation. Using INS-1 cells, we performed insulin secretion experiments, intracellular calcium recordings, and RT-qPCR to determine ucOC's effect on glucose-stimulated insulin secretion (GSIS)-related genes. The results reveal that ucOC significantly increased insulin secretion under hyperglycemic conditions compared to lower glucose levels. High glucose conditions also potentiated the effect of ucOC on calcium signals, which enhanced insulin secretion. The increase in intracellular calcium was due to an influx from the extracellular space via voltage-dependent calcium channels (VDCCs). Interestingly, the treatment of cells with NPS-2143, a GPRC6A blocker, failed to abolish the calcium signals. Uncarboxylated osteocalcin upregulated the expression of GSIS-related genes under high glucose conditions (450 mg/dL) compared to cells under standard culture conditions (200 mg/dL). In conclusion, hyperglycemia potentiates ucOC-induced insulin secretion in β-cells by opening VDCCs and upregulating GSIS genes. These findings provide a better understanding of ucOC's mechanism in the diabetic state and could lead to alternative treatments to stimulate insulin secretion.
Collapse
Affiliation(s)
- Pilailak Channuwong
- Center of Excellence in Phytochemical and Functional Food for Clinical Nutrition, Department of Nutrition and Dietetics, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Victoria Speight
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Yuanying Yuan
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Shaomian Yao
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Masami Yoshimura
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Fernando V. Bauermann
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| | - Ashish Ranjan
- Department of Radiation Oncology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sirichai Adisakwattana
- Center of Excellence in Phytochemical and Functional Food for Clinical Nutrition, Department of Nutrition and Dietetics, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Henrique Cheng
- Department of Physiological Sciences, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA
| |
Collapse
|
24
|
Schmidt AL, Kremp M, Aratake T, Cui S, Lin Y, Zhong X, Lu QR, Zhang C, Qiu M, Aberle T, Wegner M. The myelination-associated G protein-coupled receptor 37 is regulated by Zfp488, Nkx2.2, and Sox10 during oligodendrocyte differentiation. Glia 2024; 72:1304-1318. [PMID: 38546197 DOI: 10.1002/glia.24530] [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: 10/12/2023] [Revised: 03/13/2024] [Accepted: 03/19/2024] [Indexed: 05/12/2024]
Abstract
Oligodendrocyte differentiation and myelination in the central nervous system are controlled and coordinated by a complex gene regulatory network that contains several transcription factors, including Zfp488 and Nkx2.2. Despite the proven role in oligodendrocyte differentiation little is known about the exact mode of Zfp488 and Nkx2.2 action, including their target genes. Here, we used overexpression of Zfp488 and Nkx2.2 in differentiating CG4 cells to identify aspects of the oligodendroglial expression profile that depend on these transcription factors. Although both transcription factors are primarily described as repressors, the detected changes argue for an additional function as activators. Among the genes activated by both Zfp488 and Nkx2.2 was the G protein-coupled receptor Gpr37 that is important during myelination. In agreement with a positive effect on Gpr37 expression, downregulation of the G protein-coupled receptor was observed in Zfp488- and in Nkx2.2-deficient oligodendrocytes in the mouse. We also identified several potential regulatory regions of the Gpr37 gene. Although Zfp488 and Nkx2.2 both bind to one of the regulatory regions downstream of the Gpr37 gene in vivo, none of the regulatory regions was activated by either transcription factor alone. Increased activation by Zfp488 or Nkx2.2 was only observed in the presence of Sox10, a transcription factor continuously present in oligodendroglial cells. Our results argue that both Zfp488 and Nkx2.2 also act as transcriptional activators during oligodendrocyte differentiation and cooperate with Sox10 to allow the expression of Gpr37 as a modulator of the myelination process.
Collapse
Affiliation(s)
- Antonia L Schmidt
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Marco Kremp
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Takaaki Aratake
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Siying Cui
- Key Laboratory of Birth Defects, Children's Hospital of Fudan University, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yifeng Lin
- Key Laboratory of Birth Defects, Children's Hospital of Fudan University, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Xiaowen Zhong
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, Ohio, USA
| | - Q Richard Lu
- Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, Ohio, USA
| | - Chengfu Zhang
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Mengsheng Qiu
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, Hangzhou Normal University, Hangzhou, China
| | - Tim Aberle
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Wegner
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
25
|
Marangon D, Castro e Silva JH, Cerrato V, Boda E, Lecca D. Oligodendrocyte Progenitors in Glial Scar: A Bet on Remyelination. Cells 2024; 13:1024. [PMID: 38920654 PMCID: PMC11202012 DOI: 10.3390/cells13121024] [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: 05/07/2024] [Revised: 06/07/2024] [Accepted: 06/10/2024] [Indexed: 06/27/2024] Open
Abstract
Oligodendrocyte progenitor cells (OPCs) represent a subtype of glia, giving rise to oligodendrocytes, the myelin-forming cells in the central nervous system (CNS). While OPCs are highly proliferative during development, they become relatively quiescent during adulthood, when their fate is strictly influenced by the extracellular context. In traumatic injuries and chronic neurodegenerative conditions, including those of autoimmune origin, oligodendrocytes undergo apoptosis, and demyelination starts. Adult OPCs become immediately activated; they migrate at the lesion site and proliferate to replenish the damaged area, but their efficiency is hampered by the presence of a glial scar-a barrier mainly formed by reactive astrocytes, microglia and the deposition of inhibitory extracellular matrix components. If, on the one hand, a glial scar limits the lesion spreading, it also blocks tissue regeneration. Therapeutic strategies aimed at reducing astrocyte or microglia activation and shifting them toward a neuroprotective phenotype have been proposed, whereas the role of OPCs has been largely overlooked. In this review, we have considered the glial scar from the perspective of OPCs, analysing their behaviour when lesions originate and exploring the potential therapies aimed at sustaining OPCs to efficiently differentiate and promote remyelination.
Collapse
Affiliation(s)
- Davide Marangon
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (D.M.); (J.H.C.e.S.)
| | - Juliana Helena Castro e Silva
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (D.M.); (J.H.C.e.S.)
| | - Valentina Cerrato
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, 10126 Turin, Italy; (V.C.); (E.B.)
- Neuroscience Institute Cavalieri Ottolenghi, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Enrica Boda
- Department of Neuroscience Rita Levi-Montalcini, University of Turin, 10126 Turin, Italy; (V.C.); (E.B.)
- Neuroscience Institute Cavalieri Ottolenghi, Regione Gonzole 10, 10043 Orbassano, Turin, Italy
| | - Davide Lecca
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Department of Pharmaceutical Sciences, Università degli Studi di Milano, 20133 Milan, Italy; (D.M.); (J.H.C.e.S.)
| |
Collapse
|
26
|
Guo X, Yang YY, Zhou R, Tian G, Shan C, Liu JM, Li R. Causal effect of blood osteocalcin on the risk of Alzheimer's disease and the mediating role of energy metabolism. Transl Psychiatry 2024; 14:205. [PMID: 38769320 PMCID: PMC11106250 DOI: 10.1038/s41398-024-02924-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/22/2024] Open
Abstract
Growing evidence suggests an association between osteocalcin (OCN), a peptide derived from bone and involved in regulating glucose and lipid metabolism, and the risk of Alzheimer's disease (AD). However, the causality of these associations and the underlying mechanisms remain uncertain. We utilized a Mendelian randomization (MR) approach to investigate the causal effects of blood OCN levels on AD and to assess the potential involvement of glucose and lipid metabolism. Independent instrumental variables strongly associated (P < 5E-08) with blood OCN levels were obtained from three independent genome-wide association studies (GWAS) on the human blood proteome (N = 3301 to 35,892). Two distinct summary statistics datasets on AD from the International Genomics of Alzheimer's Project (IGAP, N = 63,926) and a recent study including familial-proxy AD patients (FPAD, N = 472,868) were used. Summary-level data for fasting glucose (FG), 2h-glucose post-challenge, fasting insulin, HbA1c, low-density lipoprotein cholesterol, high-density lipoprotein cholesterol, total cholesterol (TC), and triglycerides were incorporated to evaluate the potential role of glucose and lipid metabolism in mediating the impact of OCN on AD risk. Our findings consistently demonstrate a significantly negative correlation between genetically determined blood OCN levels and the risk of AD (IGAP: odds ratio [OR, 95%CI] = 0.83[0.72-0.96], P = 0.013; FPAD: OR = 0.81 [0.70-0.93], P = 0.002). Similar estimates with the same trend direction were obtained using other statistical approaches. Furthermore, employing multivariable MR analysis, we found that the causal relationship between OCN levels and AD was disappeared after adjustment of FG and TC (IGAP: OR = 0.97[0.80-1.17], P = 0.753; FPAD: OR = 0.98 [0.84-1.15], P = 0.831). There were no apparent instances of horizontal pleiotropy, and leave-one-out analysis showed good stability of the estimates. Our study provides evidence supporting a protective effect of blood OCN levels on AD, which is primarily mediated through regulating FG and TC levels. Further studies are warranted to elucidate the underlying physio-pathological mechanisms.
Collapse
Affiliation(s)
- Xingzhi Guo
- Department of Geriatric Neurology, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China
- Department of Geriatric Neurology, the Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710068, Shaanxi, China
| | - Yu-Ying Yang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Rong Zhou
- Department of Geriatric Neurology, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China
- Department of Geriatric Neurology, the Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710068, Shaanxi, China
| | - Ge Tian
- Department of Geriatric Neurology, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China
| | - Chang Shan
- Department of Endocrinology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 200127, Shanghai, China
| | - Jian-Min Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
- Shanghai National Clinical Research Center for Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China.
| | - Rui Li
- Department of Geriatric Neurology, Shaanxi Provincial People's Hospital, Xi'an, 710068, Shaanxi, China.
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, Xi'an, 710072, Shaanxi, China.
- Department of Geriatric Neurology, the Third Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710068, Shaanxi, China.
| |
Collapse
|
27
|
Robertson K, Hahn O, Robinson BG, Faruk AT, Janakiraman M, Namkoong H, Kim K, Ye J, Bishop ES, Hall RA, Wyss-Coray T, Becker LS, Kaltschmidt JA. Gpr37 modulates the severity of inflammation-induced GI dysmotility by regulating enteric reactive gliosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.09.588619. [PMID: 38645163 PMCID: PMC11030428 DOI: 10.1101/2024.04.09.588619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
The enteric nervous system (ENS) is contained within two layers of the gut wall and is made up of neurons, immune cells, and enteric glia cells (EGCs) that regulate gastrointestinal (GI) function. EGCs in both inflammatory bowel disease (IBD) and irritable bowel syndrome (IBS) change in response to inflammation, referred to as reactive gliosis. Whether EGCs restricted to a specific layer or region within the GI tract alone can influence intestinal immune response is unknown. Using bulk RNA-sequencing and in situ hybridization, we identify G-protein coupled receptor Gpr37 , as a gene expressed only in EGCs of the myenteric plexus, one of the two layers of the ENS. We show that Gpr37 contributes to key components of LPS-induced reactive gliosis including activation of NF-kB and IFN-y signaling and response genes, lymphocyte recruitment, and inflammation-induced GI dysmotility. Targeting Gpr37 in EGCs presents a potential avenue for modifying inflammatory processes in the ENS.
Collapse
|
28
|
Scharf MM, Humphrys LJ, Berndt S, Di Pizio A, Lehmann J, Liebscher I, Nicoli A, Niv MY, Peri L, Schihada H, Schulte G. The dark sides of the GPCR tree - research progress on understudied GPCRs. Br J Pharmacol 2024. [PMID: 38339984 DOI: 10.1111/bph.16325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/24/2023] [Accepted: 01/08/2024] [Indexed: 02/12/2024] Open
Abstract
A large portion of the human GPCRome is still in the dark and understudied, consisting even of entire subfamilies of GPCRs such as odorant receptors, class A and C orphans, adhesion GPCRs, Frizzleds and taste receptors. However, it is undeniable that these GPCRs bring an untapped therapeutic potential that should be explored further. Open questions on these GPCRs span diverse topics such as deorphanisation, the development of tool compounds and tools for studying these GPCRs, as well as understanding basic signalling mechanisms. This review gives an overview of the current state of knowledge for each of the diverse subfamilies of understudied receptors regarding their physiological relevance, molecular mechanisms, endogenous ligands and pharmacological tools. Furthermore, it identifies some of the largest knowledge gaps that should be addressed in the foreseeable future and lists some general strategies that might be helpful in this process.
Collapse
Affiliation(s)
- Magdalena M Scharf
- Karolinska Institutet, Dept. Physiology & Pharmacology, Sec. Receptor Biology & Signaling, Stockholm, Sweden
| | - Laura J Humphrys
- Institute of Pharmacy, University of Regensburg, Regensburg, Germany
| | - Sandra Berndt
- Rudolf Schönheimer Institute for Biochemistry, Molecular Biochemistry, University of Leipzig, Leipzig, Germany
| | - Antonella Di Pizio
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
- Chemoinformatics and Protein Modelling, Department of Molecular Life Science, School of Life Science, Technical University of Munich, Freising, Germany
| | - Juliane Lehmann
- Rudolf Schönheimer Institute for Biochemistry, Molecular Biochemistry, University of Leipzig, Leipzig, Germany
| | - Ines Liebscher
- Rudolf Schönheimer Institute for Biochemistry, Molecular Biochemistry, University of Leipzig, Leipzig, Germany
| | - Alessandro Nicoli
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
- Chemoinformatics and Protein Modelling, Department of Molecular Life Science, School of Life Science, Technical University of Munich, Freising, Germany
| | - Masha Y Niv
- The Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Lior Peri
- The Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Hannes Schihada
- Institute of Pharmaceutical Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Gunnar Schulte
- Karolinska Institutet, Dept. Physiology & Pharmacology, Sec. Receptor Biology & Signaling, Stockholm, Sweden
| |
Collapse
|
29
|
Correa Pinto Junior D, Canal Delgado I, Yang H, Clemenceau A, Corvelo A, Narzisi G, Musunuri R, Meyer Berger J, Hendricks LE, Tokumura K, Luo N, Li H, Oury F, Ducy P, Yadav VK, Li X, Karsenty G. Osteocalcin of maternal and embryonic origins synergize to establish homeostasis in offspring. EMBO Rep 2024; 25:593-615. [PMID: 38228788 PMCID: PMC10897216 DOI: 10.1038/s44319-023-00031-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/24/2023] [Accepted: 12/01/2023] [Indexed: 01/18/2024] Open
Abstract
Many physiological osteocalcin-regulated functions are affected in adult offspring of mothers experiencing unhealthy pregnancy. Furthermore, osteocalcin signaling during gestation influences cognition and adrenal steroidogenesis in adult mice. Together these observations suggest that osteocalcin may broadly function during pregnancy to determine organismal homeostasis in adult mammals. To test this hypothesis, we analyzed in unchallenged wildtype and Osteocalcin-deficient, newborn and adult mice of various genotypes and origin maintained on different genetic backgrounds, the functions of osteocalcin in the pancreas, liver and testes and their molecular underpinnings. This analysis revealed that providing mothers are Osteocalcin-deficient, Osteocalcin haploinsufficiency in embryos hampers insulin secretion, liver gluconeogenesis, glucose homeostasis, testes steroidogenesis in adult offspring; inhibits cell proliferation in developing pancreatic islets and testes; and disrupts distinct programs of gene expression in these organs and in the brain. This study indicates that osteocalcin exerts dominant functions in most organs it influences. Furthermore, through their synergistic regulation of multiple physiological functions, osteocalcin of maternal and embryonic origins contributes to the establishment and maintenance of organismal homeostasis in newborn and adult offspring.
Collapse
Affiliation(s)
- Danilo Correa Pinto Junior
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Isabella Canal Delgado
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Haiyang Yang
- Guangdong Provincial Key Laboratory of Brain Connectome, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
| | - Alisson Clemenceau
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | | | | | | | - Julian Meyer Berger
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Lauren E Hendricks
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Kazuya Tokumura
- Department of Bioactive Molecules, Pharmacology, Gifu Pharmaceutical University, Gifu, Japan
| | - Na Luo
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Hongchao Li
- Guangdong Provincial Key Laboratory of Brain Connectome, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
| | - Franck Oury
- INSERM U1151, Institut Necker Enfants-Malades (INEM), Université Paris Descartes-Sorbonne, Paris Cité, Paris, France.
| | - Patricia Ducy
- Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA.
| | - Vijay K Yadav
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA.
| | - Xiang Li
- Guangdong Provincial Key Laboratory of Brain Connectome, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China.
| | - Gerard Karsenty
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA.
| |
Collapse
|
30
|
Qian Z, Li B, Meng X, Liao J, Wang G, Li Y, Luo Q, Ye K. Inhibition of asparagine endopeptidase (AEP) effectively treats sporadic Alzheimer's disease in mice. Neuropsychopharmacology 2024; 49:620-630. [PMID: 38030711 PMCID: PMC10789813 DOI: 10.1038/s41386-023-01774-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 11/12/2023] [Accepted: 11/15/2023] [Indexed: 12/01/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease with cognitive dysfunction as its major clinical symptom. However, there is no disease-modifying small molecular medicine to effectively slow down progression of the disease. Here, we show an optimized asparagine endopeptidase (AEP, also known as δ-secretase) inhibitor, #11 A, that displays an orderly in vivo pharmacokinetics/pharmacodynamics (PK/PD) relationship and robustly attenuates AD pathologies in a sporadic AD mouse model. #11 A is brain permeable with great oral bioavailability. It blocks AEP cleavage of APP and Tau dose-dependently, and significantly decreases Aβ40 and Aβ42 and p-Tau levels in APP/PS1 and Tau P301S mice after oral administration. Notably, #11 A strongly inhibits AEP and prevents mouse APP and Tau fragmentation by AEP, leading to reduction of mouse Aβ42 (mAβ42), mAβ40 and mouse p-Tau181 levels in Thy1-ApoE4/C/EBPβ transgenic mice in a dose-dependent manner. Repeated oral administration of #11 A substantially decreases mAβ aggregation as validated by Aβ PET assay, Tau pathology, neurodegeneration and brain volume reduction, resulting in alleviation of cognitive impairment. Therefore, our results support that #11 A is a disease-modifying preclinical candidate for pharmacologically treating AD.
Collapse
Affiliation(s)
- Zhengjiang Qian
- Faculty of Life and Health Sciences, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China
| | - Bowei Li
- Shenzhen Institute of Advanced Technology, University of Chinese Academy of Science, Shenzhen, Guangdong, 518055, China
| | - Xin Meng
- Faculty of Life and Health Sciences, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China
| | - Jianming Liao
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Guangxing Wang
- School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yanjiao Li
- Faculty of Life and Health Sciences, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China
| | - Qian Luo
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Keqiang Ye
- Faculty of Life and Health Sciences, Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, Guangdong, China.
| |
Collapse
|
31
|
Ruggiero C, Baroni M, Xenos D, Parretti L, Macchione IG, Bubba V, Laudisio A, Pedone C, Ferracci M, Magierski R, Boccardi V, Antonelli-Incalzi R, Mecocci P. Dementia, osteoporosis and fragility fractures: Intricate epidemiological relationships, plausible biological connections, and twisted clinical practices. Ageing Res Rev 2024; 93:102130. [PMID: 38030092 DOI: 10.1016/j.arr.2023.102130] [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: 09/20/2023] [Revised: 11/06/2023] [Accepted: 11/14/2023] [Indexed: 12/01/2023]
Abstract
Dementia, osteoporosis, and fragility fractures are chronic diseases, often co-existing in older adults. These conditions pose severe morbidity, long-term disability, and mortality, with relevant socioeconomic implications. While in the research arena, the discussion remains on whether dementia is the cause or the consequence of fragility fractures, healthcare professionals need a better understanding of the interplay between such conditions from epidemiological and physiological standpoints. With this review, we summarized the available literature surrounding the relationship between cognitive impairment, dementia, and both low bone mineral density (BMD) and fragility fractures. Given the strength of the bi-directional associations and their impact on the quality of life, we shed light on the biological connections between brain and bone systems, presenting the main mediators, including gut microbioma, and pathological pathways leading to the dysregulation of bone and brain metabolism. Ultimately, we synthesized the evidence about the impact of available pharmacological treatments for the prevention of fragility fractures on cognitive functions and individuals' outcomes when dementia coexists. Vice versa, the effects of symptomatic treatments for dementia on the risk of falls and fragility fractures are explored. Combining evidence alongside clinical practice, we discuss challenges and opportunities related to the management of older adults affected by cognitive impairment or dementia and at high risk for fragility fracture prevention, which leads to not only an improvement in patient health-related outcomes and survival but also a reduction in healthcare cost and socio-economic burden.
Collapse
Affiliation(s)
- C Ruggiero
- Department of Medicine, Section of Gerontology and Geriatrics, University of Perugia, Italy.
| | - M Baroni
- Department of Medicine, Section of Gerontology and Geriatrics, University of Perugia, Italy
| | - D Xenos
- Department of Medicine, Section of Gerontology and Geriatrics, University of Perugia, Italy
| | - L Parretti
- Department of Medicine, Section of Gerontology and Geriatrics, University of Perugia, Italy
| | - I G Macchione
- Department of Medicine, Section of Gerontology and Geriatrics, University of Perugia, Italy
| | - V Bubba
- Department of Medicine, Section of Gerontology and Geriatrics, University of Perugia, Italy
| | - A Laudisio
- Department of Medicine, Unit of Geriatrics, Campus Bio-Medico di Roma University, Rome, Italy
| | - C Pedone
- Department of Medicine, Unit of Geriatrics, Campus Bio-Medico di Roma University, Rome, Italy
| | - M Ferracci
- Department of Medicine, Section of Gerontology and Geriatrics, University of Perugia, Italy
| | - R Magierski
- Department of Old Age Psychiatry and Psychotic Disorders, Medical University of Lodz, Lodz, Poland
| | - V Boccardi
- Department of Medicine, Section of Gerontology and Geriatrics, University of Perugia, Italy
| | - R Antonelli-Incalzi
- Department of Medicine, Unit of Geriatrics, Campus Bio-Medico di Roma University, Rome, Italy
| | - P Mecocci
- Department of Medicine, Section of Gerontology and Geriatrics, University of Perugia, Italy
| |
Collapse
|
32
|
Liu W, Hu Q, Zhang F, Shi K, Wu J. Investigation of the causal relationship between osteocalcin and dementia: A Mendelian randomization study. Heliyon 2023; 9:e21073. [PMID: 37916108 PMCID: PMC10616355 DOI: 10.1016/j.heliyon.2023.e21073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 08/03/2023] [Accepted: 10/13/2023] [Indexed: 11/03/2023] Open
Abstract
Objective Basic medical studies have reported an improved effect of osteocalcin on cognition. We explored the causal link between osteocalcin and dementia via the implementation of Mendelian randomization methodology. Methods Genome-wide association studies were employed to identify single nucleotide polymorphisms (SNPs) showing significant correlations with osteocalcin. Subsequently, A two-sample Mendelian randomization analysis was conducted utilizing the inverse-variance-weighted (IVW) technique to assess the causal relationship between osteocalcin and various types of dementia, including Alzheimer's disease (AD), Parkinson's disease (PD), Lewy body dementia (LBD), and vascular dementia (VD). This approach aimed to minimize potential sources of confounding bias and provide more robust results. Multivariable MR (MVMR) analysis was conducted to adjust for potential genetic pleiotropy. Results The study employed three SNPs, namely rs71631868, rs9271374, and rs116843408, as genetic tools to evaluate the causal association of osteocalcin with dementia. The IVW analysis indicated that osteocalcin may have a potential protective effect against AD with an odds ratio (OR) of 0.790 (95 % CI: 0.688-0.906; P < 0.001). However, no significant relationship was observed between osteocalcin and other types of dementia. Furthermore, the MVMR analysis indicated that the impact of osteocalcin on AD remained consistent even after adjusting for age-related macular degeneration and Type 2 diabetes with an OR of 0.856 (95 % CI: 0.744-0.985; P = 0.030). Conclusions Our findings provide important insights into the role of osteocalcin in the pathogenesis of AD. Future research is required to clarify the underlying mechanisms and their clinical applications.
Collapse
Affiliation(s)
- Wangmi Liu
- The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Qiang Hu
- Pujiang Tianxian Orthopedic Hospital, Jinhua, 322200, China
| | - Feng Zhang
- The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Kesi Shi
- The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Jiayan Wu
- Department of Neurology, Chongming Branch, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 66 East Xiangyang Road, Chongming, Shanghai, China
| |
Collapse
|
33
|
Bolinger AA, Frazier A, La JH, Allen JA, Zhou J. Orphan G Protein-Coupled Receptor GPR37 as an Emerging Therapeutic Target. ACS Chem Neurosci 2023; 14:3318-3334. [PMID: 37676000 PMCID: PMC11144446 DOI: 10.1021/acschemneuro.3c00479] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/08/2023] Open
Abstract
G protein-coupled receptors (GPCRs) are successful druggable targets, making up around 35% of all FDA-approved medications. However, a large number of receptors remain orphaned, with no known endogenous ligand, representing a challenging but untapped area to discover new therapeutic targets. Among orphan GPCRs (oGPCRs) of interest, G protein-coupled receptor 37 (GPR37) is highly expressed in the central nervous system (CNS), particularly in the spinal cord and oligodendrocytes. While its cellular signaling mechanisms and endogenous receptor ligands remain elusive, GPR37 has been implicated in several important neurological conditions, including Parkinson's disease (PD), inflammation, pain, autism, and brain tumors. GPR37 structure, signaling, emerging physiology, and pharmacology are reviewed while integrating a discussion on potential therapeutic indications and opportunities.
Collapse
Affiliation(s)
- Andrew A. Bolinger
- Department of Pharmacology and Toxicology, Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Andrew Frazier
- Department of Pharmacology and Toxicology, Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Jun-Ho La
- Department of Neurobiology, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - John A. Allen
- Department of Pharmacology and Toxicology, Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, Texas 77555, United States
| | - Jia Zhou
- Department of Pharmacology and Toxicology, Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, Texas 77555, United States
| |
Collapse
|
34
|
Kubi JA, Brah AS, Cheung KMC, Lee YL, Lee KF, Sze SCW, Qiao W, Yeung KWK. A new osteogenic protein isolated from Dioscorea opposita Thunb accelerates bone defect healing through the mTOR signaling axis. Bioact Mater 2023; 27:429-446. [PMID: 37152710 PMCID: PMC10160600 DOI: 10.1016/j.bioactmat.2023.04.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 04/13/2023] [Accepted: 04/17/2023] [Indexed: 05/09/2023] Open
Abstract
Delayed bone defect repairs lead to severe health and socioeconomic impacts on patients. Hence, there are increasing demands for medical interventions to promote bone defect healing. Recombinant proteins such as BMP-2 have been recognized as one of the powerful osteogenic substances that promote mesenchymal stem cells (MSCs) to osteoblast differentiation and are widely applied clinically for bone defect repairs. However, recent reports show that BMP-2 treatment has been associated with clinical adverse side effects such as ectopic bone formation, osteolysis and stimulation of inflammation. Here, we have identified one new osteogenic protein, named 'HKUOT-S2' protein, from Dioscorea opposita Thunb. Using the bone defect model, we have shown that the HKUOT-S2 protein can accelerate bone defect repair by activating the mTOR signaling axis of MSCs-derived osteoblasts and increasing osteoblastic biomineralization. The HKUOT-S2 protein can also modulate the transcriptomic changes of macrophages, stem cells, and osteoblasts, thereby enhancing the crosstalk between the polarized macrophages and MSCs-osteoblast differentiation to facilitate osteogenesis. Furthermore, this protein had no toxic effects in vivo. We have also identified HKUOT-S2 peptide sequence TKSSLPGQTK as a functional osteogenic unit that can promote osteoblast differentiation in vitro. The HKUOT-S2 protein with robust osteogenic activity could be a potential alternative osteoanabolic agent for promoting osteogenesis and bone defect repairs. We believe that the HKUOT-S2 protein may potentially be applied clinically as a new class of osteogenic agent for bone defect healing.
Collapse
Affiliation(s)
- John Akrofi Kubi
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong (HKU), Hong Kong S.A.R., PR China
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, HKU-Shenzhen Hospital, Shenzhen, 518053, PR China
| | - Augustine Suurinobah Brah
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong (HKU), Hong Kong S.A.R., PR China
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, HKU-Shenzhen Hospital, Shenzhen, 518053, PR China
| | - Kenneth Man Chee Cheung
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong (HKU), Hong Kong S.A.R., PR China
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, HKU-Shenzhen Hospital, Shenzhen, 518053, PR China
| | - Yin Lau Lee
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, HKU, 21 Sassoon Road, Hong Kong S.A.R, PR China
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, HKU- Shenzhen Hospital, Shenzhen, PR China
| | - Kai-Fai Lee
- Department of Obstetrics and Gynaecology, Li Ka Shing Faculty of Medicine, HKU, 21 Sassoon Road, Hong Kong S.A.R, PR China
- Shenzhen Key Laboratory of Fertility Regulation, Reproductive Medicine Center, HKU- Shenzhen Hospital, Shenzhen, PR China
| | - Stephen Cho Wing Sze
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Kowloon Tong, Hong Kong S.A.R, PR China
- Golden Meditech Center for NeuroRegeneration Sciences, Hong Kong Baptist University, Kowloon Tong, Hong Kong S.A.R, PR China
| | - Wei Qiao
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, Hong Kong S.A.R, PR China
| | - Kelvin Wai-Kwok Yeung
- Department of Orthopaedics and Traumatology, Li Ka Shing Faculty of Medicine, The University of Hong Kong (HKU), Hong Kong S.A.R., PR China
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, HKU-Shenzhen Hospital, Shenzhen, 518053, PR China
| |
Collapse
|
35
|
Abstract
Together, loss- and gain-of-function experiments have identified the bone-derived secreted molecule osteocalcin as a hormone with a broad reach in rodents and primates. Following its binding to one of three receptors, osteocalcin exerts a profound influence on various aspects of energy metabolism as well as steroidogenesis, neurotransmitter biosynthesis and thereby male fertility, electrolyte homeostasis, cognition, the acute stress response, and exercise capacity. Although this review focuses mostly on the regulation of energy metabolism by osteocalcin, it also touches on its other functions. Lastly, it proposes what could be a common theme between the functions of osteocalcin and between these functions and the structural functions of bone.
Collapse
Affiliation(s)
- Gerard Karsenty
- Departments of Genetics and Development, Vagelos College of Physicians & Surgeons, Columbia University, New York, New York, USA;
| |
Collapse
|
36
|
Pinto DC, Delgado IC, Yang H, Clemenceau A, Corvelo A, Narzisi G, Musunuri R, Berger JM, Hendricks LE, Tokumura K, Luo N, Li H, Oury F, Ducy P, Yadav VK, Li X, Karsenty G. Osteocalcin of maternal and embryonic origins synergize to establish homeostasis in offspring. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.11.552969. [PMID: 37645714 PMCID: PMC10462025 DOI: 10.1101/2023.08.11.552969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Many physiological functions regulated by osteocalcin are affected in adult offspring of mothers experiencing an unhealthy pregnancy. Furthermore, osteocalcin signaling during gestation influences cognition and adrenal steroidogenesis in adult mice. Together these observations suggest that osteocalcin functions during pregnancy may be a broader determinant of organismal homeostasis in adult mammals than previously thought. To test this hypothesis, we analyzed in unchallenged wildtype and Osteocalcin -deficient, newborn, and adult mice of various genotypes and origin, and that were maintained on different genetic backgrounds, the functions of osteocalcin in the pancreas, liver and testes and their molecular underpinnings. This analysis revealed that providing mothers are themselves Osteocalcin -deficient, Osteocalcin haploinsufficiency in embryos hampers insulin secretion, liver gluconeogenesis, glucose homeostasis, testes steroidogenesis in adult offspring; inhibits cell proliferation in developing pancreatic islets and testes; and disrupts distinct programs of gene expression in these organs and in the brain. This study indicates that through their synergistic regulation of multiple physiological functions, osteocalcin ofmaternal and embryonic origins contributes to the establishment and maintenance of organismal homeostasis in newborn and adult offspring.
Collapse
|
37
|
Zheng XQ, Lin JL, Huang J, Wu T, Song CL. Targeting aging with the healthy skeletal system: The endocrine role of bone. Rev Endocr Metab Disord 2023; 24:695-711. [PMID: 37402956 DOI: 10.1007/s11154-023-09812-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/29/2023] [Indexed: 07/06/2023]
Abstract
Aging is an inevitable biological process, and longevity may be related to bone health. Maintaining strong bone health can extend one's lifespan, but the exact mechanism is unclear. Bone and extraosseous organs, including the heart and brain, have complex and precise communication mechanisms. In addition to its load bearing capacity, the skeletal system secretes cytokines, which play a role in bone regulation of extraosseous organs. FGF23, OCN, and LCN2 are three representative bone-derived cytokines involved in energy metabolism, endocrine homeostasis and systemic chronic inflammation levels. Today, advanced research methods provide new understandings of bone as a crucial endocrine organ. For example, gene editing technology enables bone-specific conditional gene knockout models, which allows the study of bone-derived cytokines to be more precise. We systematically evaluated the various effects of bone-derived cytokines on extraosseous organs and their possible antiaging mechanism. Targeting aging with the current knowledge of the healthy skeletal system is a potential therapeutic strategy. Therefore, we present a comprehensive review that summarizes the current knowledge and provides insights for futures studies.
Collapse
Affiliation(s)
- Xuan-Qi Zheng
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Jia-Liang Lin
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Jie Huang
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Tong Wu
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Chun-Li Song
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China.
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China.
- Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China.
| |
Collapse
|
38
|
Cai W, Sun B, Song C, Liu F, Wu Z, Liu Z. Resveratrol induces proliferation and differentiation of mouse pre-osteoblast MC3T3-E1 by promoting autophagy. BMC Complement Med Ther 2023; 23:121. [PMID: 37060066 PMCID: PMC10103476 DOI: 10.1186/s12906-023-03943-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 03/29/2023] [Indexed: 04/16/2023] Open
Abstract
BACKGROUND In mouse, it was discovered that resveratrol (Res) enhanced osteoporosis (OP) by boosting osteogenesis. Besides, Res can also have an impact on MC3T3-E1 cells, which are crucial for the control of osteogenesis and thus increase osteogenesis. Although some articles have discovered that Res enhanced autophagy to promote the value-added differentiation of MC3T3, it is unclear exactly how this affects the process of osteogenesis in mouse. Therefore, we will show that Res encourages MC3T3-E1 proliferation and differentiation in mouse pre-osteoblasts and further investigate the autophagy-related mechanism for this impact. METHODS (1) MC3T3-E1 cells were separated into blank control group and various concentrations (0.01, 0.1, 1, 10, 100µmol/L) of group in order to determine the ideal Res concentration. In the Res group, Cell Counting Kit-8 (CCK-8) was used to measure the proliferation activity of pre-osteoblasts in mice in each group after resveratrol intervention. Alkaline Phosphatase (ALP) and alizarin red staining were used to gauge the degree of osteogenic differentiation, and RT-qPCR was used to measure the expression levels of Runx2 and OCN in the osteogenic differentiation ability of the cells. (2) In the experiment, four groups were set up: the control group, 3MA group, Res group, and Res + 3MA group. To examine cell mineralization, ALP and alizarin red staining were utilized. RT-qPCR and Western blot detection of cell autophagy activity levels and osteogenic differentiation capacity in each group following intervention. RESULTS (1) Resveratrol might increase the number of mice pre-osteoblast, with the impact being most pronounced at 10µmol/L (P < 0.05). The nodules developed substantially more often than in the blank control group, and Runx2 and OCN expressions significantly increased (P < 0.05). (2) In contrast to the Res group, after 3MA purine blocked autophagy, the Res + 3MA group's alkaline phosphatase staining and the development of mineralized nodules were reduced. Runx2, OCN, LC3II / LC3I expression decreased, p62 expression increased (P < 0.05). CONCLUSION The present study partially or indirectly demonstrated that Res may, through increased autophagy, induce osteogenic differentiation of MC3T3-E1 cells.
Collapse
Affiliation(s)
- Weiye Cai
- Department of Orthopaedics and Traumatology, The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, Sichuan, China
| | - Bin Sun
- The People's Hospital Of Jimo, Jimo, Qingdao, China
| | - Chao Song
- Department of Orthopaedics and Traumatology, The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, Sichuan, China
| | - Fei Liu
- Department of Orthopaedics and Traumatology, The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, Sichuan, China
| | - Zhengliang Wu
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zongchao Liu
- Department of Orthopaedics and Traumatology, The Affiliated Hospital of Traditional Chinese Medicine of Southwest Medical University, Luzhou, Sichuan, China.
- Luzhou Longmatan District People's Hospital, Luzhou, Sichuan Province, 646000, China.
| |
Collapse
|
39
|
Chen L, Yu Z, Xie L, He X, Mu X, Chen C, Yang W, Tong X, Liu J, Gao Z, Sun S, Xu N, Lu Z, Zheng J, Zhang Y. ANGPTL2 binds MAG to efficiently enhance oligodendrocyte differentiation. Cell Biosci 2023; 13:42. [PMID: 36855057 PMCID: PMC9976406 DOI: 10.1186/s13578-023-00970-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 01/23/2023] [Indexed: 03/02/2023] Open
Abstract
BACKGROUND Oligodendrocytes have robust regenerative ability and are key players in remyelination during physiological and pathophysiological states. However, the mechanisms of brain microenvironmental cue in regulation of the differentiation of oligodendrocytes still needs to be further investigated. RESULTS We demonstrated that myelin-associated glycoprotein (MAG) was a novel receptor for angiopoietin-like protein 2 (ANGPTL2). The binding of ANGPTL2 to MAG efficiently promoted the differentiation of oligodendrocytes in vitro, as evaluated in an HCN cell line. Angptl2-null mice had a markedly impaired myelination capacity in the early stage of oligodendrocyte development. These mice had notably decreased remyelination capacities and enhanced motor disability in a cuprizone-induced demyelinating mouse model, which was similar to the Mag-null mice. The loss of remyelination ability in Angptl2-null/Mag-null mice was similar to the Angptl2-WT/Mag-null mice, which indicated that the ANGPTL2-mediated oligodendrocyte differentiation effect depended on the MAG receptor. ANGPTL2 bound MAG to enhance its phosphorylation level and recruit Fyn kinase, which increased Fyn phosphorylation levels, followed by the transactivation of myelin regulatory factor (MYRF). CONCLUSION Our study demonstrated an unexpected cross-talk between the environmental protein (ANGPTL2) and its surface receptor (MAG) in the regulation of oligodendrocyte differentiation, which may benefit the treatment of many demyelination disorders, including multiple sclerosis.
Collapse
Affiliation(s)
- Lu Chen
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Zhuo Yu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Li Xie
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Xiaoxiao He
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Xingmei Mu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Chiqi Chen
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Wenqian Yang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China
| | - Xiaoping Tong
- Center for Brain Science, Shanghai Children's Medical Center, Department of Anatomy and Physiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junling Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhengliang Gao
- Yangzhi Rehabilitation Hospital (Shanghai Sunshine Rehabilitation Center), Tongji Univeirsity School of Medicine, Shanghai, China
| | - Suya Sun
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - NanJie Xu
- Collaborative Innovation Center for Brain Science, Department of Anatomy and Physiology, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhigang Lu
- The Fifth People's Hospital of Shanghai, the Shanghai Key Laboratory of Medical Epigenetics, The International Co-Laboratory of Medical Epigenetics and Metabolism, Ministry of Science and Technology, Institutes of Biomedical Sciences, Shanghai Institute of Infectious Diseases and Biosecurity, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Junke Zheng
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.
| | - Yaping Zhang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital/Faculty of Basic Medicine, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai, 200025, China.
| |
Collapse
|
40
|
Inflammation and Infection in Pain and the Role of GPR37. Int J Mol Sci 2022; 23:ijms232214426. [PMID: 36430912 PMCID: PMC9692891 DOI: 10.3390/ijms232214426] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022] Open
Abstract
Inflammation is known to cause pain, and pain is of one of the cardinal signs of inflammation. Mounting evidence suggests that acute inflammation also resolves pain through specialized pro-resolving mediators (SPMs) and macrophage signaling. GPR37 is expressed by neurons and oligodendrocytes in the brain and has been implicated in multiple disorders, such as demyelination, Parkinson's disease, stroke, and cancer. Recent studies have demonstrated that GPR37 is expressed by macrophages and confers protection against infection by bacteria and parasites. Furthermore, GPR37 promotes the resolution of inflammatory pain and infection-induced pain, as the duration of pain after tissue injury and infection is prolonged in mice lacking Gpr37. Mechanistically, activation of GPR37 enhances macrophage phagocytosis, and Gpr37-deficient macrophages exhibit dysregulations of pro-inflammatory and anti-inflammatory cytokines, switching from M2- to M1-like phenotypes. We also discuss novel ligands of GPR37, including neuroprotectin D1 (NPD1), a SPM derived from docosahexaenoic acid (DHA), and bone-derived hormone osteocalcin (OCN), which can suppress oligodendrocyte differentiation and myelination. NPD1 stimulates macrophage phagocytosis via GPR37 and exhibits potent analgesic actions in various animal models of inflammatory and neuropathic pain. Targeting GPR37 may lead to novel therapeutics for treating inflammation, infection, pain, and neurological diseases.
Collapse
|
41
|
Xu J, Zhang Z, Zhao J, Meyers CA, Lee S, Qin Q, James AW. Interaction between the nervous and skeletal systems. Front Cell Dev Biol 2022; 10:976736. [PMID: 36111341 PMCID: PMC9468661 DOI: 10.3389/fcell.2022.976736] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 08/08/2022] [Indexed: 11/14/2022] Open
Abstract
The skeleton is one of the largest organ systems in the body and is richly innervated by the network of nerves. Peripheral nerves in the skeleton include sensory and sympathetic nerves. Crosstalk between bones and nerves is a hot topic of current research, yet it is not well understood. In this review, we will explore the role of nerves in bone repair and remodeling, as well as summarize the molecular mechanisms by which neurotransmitters regulate osteogenic differentiation. Furthermore, we discuss the skeleton’s role as an endocrine organ that regulates the innervation and function of nerves by secreting bone-derived factors. An understanding of the interactions between nerves and bone can help to prevent and treat bone diseases caused by abnormal innervation or nerve function, develop new strategies for clinical bone regeneration, and improve patient outcomes.
Collapse
Affiliation(s)
- Jiajia Xu
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Academy of Orthopedics, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China
| | - Zhongmin Zhang
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junjie Zhao
- Division of Spine Surgery, Department of Orthopaedics, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Carolyn A. Meyers
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Seungyong Lee
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
- Department of Physical Education, Incheon National University, Incheon, South Korea
| | - Qizhi Qin
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
| | - Aaron W. James
- Department of Pathology, Johns Hopkins University, Baltimore, MD, United States
- *Correspondence: Aaron W. James,
| |
Collapse
|
42
|
Qian Z, Liu C, Li H, Yang H, Wu J, Liu J, Li Y, Chen X, Xu J, Li X. Osteocalcin Alleviates Lipopolysaccharide-Induced Acute Inflammation via Activation of GPR37 in Macrophages. Biomedicines 2022; 10:1006. [PMID: 35625743 PMCID: PMC9138386 DOI: 10.3390/biomedicines10051006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/24/2022] [Accepted: 04/24/2022] [Indexed: 11/16/2022] Open
Abstract
The G protein-coupled receptor 37 (GPR37) has been reported to be expressed in macrophages and the activation of GPR37 by its ligand/agonist, and it can regulate macrophage-associated functions and inflammatory responses. Since our previous work identified that osteocalcin (OCN) acts as an endogenous ligand for GPR37 and can elicit various intracellular signals by interacting with GPR37, we thus hypothesized that OCN may also play a functional role in macrophage through the activation of GPR37. To verify the hypothesis, we conducted a series of in vivo and in vitro studies in lipopolysaccharide (LPS)-challenged mice and primary cultured macrophages. Our results reveal that the OCN gene deletion (OCN-/-) and wild type (WT) mice showed comparable death rates and inflammatory cytokines productions in response to a lethal dose of LPS exposure. However, the detrimental effects caused by LPS were significantly ameliorated by exogenous OCN treatments in both WT and OCN-/- mice. Notably, the protective effects of OCN were absent in GPR37-/- mice. In coordination with the in vivo results, our in vitro studies further illustrated that OCN triggered intracellular responses via GPR37 in peritoneal macrophages by regulating the release of inflammatory factors and macrophage phagocytic function. Finally, we exhibited that the adoptive transfer of OCN-treated macrophages from WT mice significantly inhibits the release of pro-inflammatory cytokines in GPR37-/- mice exposed to LPS. Taken together, these findings suggest a protective role of OCN against LPS-caused acute inflammation, by the activation of GPR37 in macrophages, and provide a potential application of the activation of the OCN/GPR37 regulatory axis as a therapeutic strategy for inflammatory diseases.
Collapse
Affiliation(s)
- Zhengjiang Qian
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China; (Z.Q.); (C.L.); (H.L.); (H.Y.)
| | - Chunhua Liu
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China; (Z.Q.); (C.L.); (H.L.); (H.Y.)
| | - Hongchao Li
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China; (Z.Q.); (C.L.); (H.L.); (H.Y.)
| | - Haiyang Yang
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China; (Z.Q.); (C.L.); (H.L.); (H.Y.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jianhao Wu
- Department of Traditional Chinese Medicine, Shenzhen University General Hospital, Shenzhen 518055, China; (J.W.); (J.L.); (J.X.)
| | - Jing Liu
- Department of Traditional Chinese Medicine, Shenzhen University General Hospital, Shenzhen 518055, China; (J.W.); (J.L.); (J.X.)
| | - Yanjiao Li
- Faculty of Life and Health Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China;
| | - Xuhui Chen
- Department of Neurology, Peking University Shenzhen Hospital, Shenzhen 518000, China;
| | - Jianyang Xu
- Department of Traditional Chinese Medicine, Shenzhen University General Hospital, Shenzhen 518055, China; (J.W.); (J.L.); (J.X.)
| | - Xiang Li
- Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, Shenzhen Key Laboratory of Viral Vectors for Biomedicine, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China; (Z.Q.); (C.L.); (H.L.); (H.Y.)
| |
Collapse
|
43
|
Massimi M, Di Pietro C, La Sala G, Matteoni R. Mouse Mutants of Gpr37 and Gpr37l1 Receptor Genes: Disease Modeling Applications. Int J Mol Sci 2022; 23:ijms23084288. [PMID: 35457105 PMCID: PMC9025225 DOI: 10.3390/ijms23084288] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/08/2022] [Accepted: 04/11/2022] [Indexed: 02/05/2023] Open
Abstract
The vertebrate G protein–coupled receptor 37 and G protein–coupled receptor 37-like 1 (GPR37 and GPR37L1) proteins have amino acid sequence homology to endothelin and bombesin-specific receptors. The prosaposin glycoprotein, its derived peptides, and analogues have been reported to interact with and activate both putative receptors. The GPR37 and GPR37L1 genes are highly expressed in human and rodent brains. GPR37 transcripts are most abundant in oligodendrocytes and in the neurons of the substantia nigra and hippocampus, while the GPR37L1 gene is markedly expressed in cerebellar Bergmann glia astrocytes. The human GPR37 protein is a substrate of parkin, and its insoluble form accumulates in brain samples from patients of inherited juvenile Parkinson’s disease. Several Gpr37 and Gpr37l1 mouse mutant strains have been produced and applied to extensive in vivo and ex vivo analyses of respective receptor functions and involvement in brain and other organ pathologies. The genotypic and phenotypic characteristics of the different mouse strains so far published are reported and discussed, and their current and proposed applications to human disease modeling are highlighted.
Collapse
|
44
|
Emerging Roles for the Orphan GPCRs, GPR37 and GPR37 L1, in Stroke Pathophysiology. Int J Mol Sci 2022; 23:ijms23074028. [PMID: 35409385 PMCID: PMC9000135 DOI: 10.3390/ijms23074028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 03/31/2022] [Accepted: 04/02/2022] [Indexed: 11/17/2022] Open
Abstract
Recent studies have shed light on the diverse and complex roles of G-protein coupled receptors (GPCRs) in the pathophysiology of stroke. These receptors constitute a large family of seven transmembrane-spanning proteins that play an intricate role in cellular communication mechanisms which drive both tissue injury and repair following ischemic stroke. Orphan GPCRs represent a unique sub-class of GPCRs for which no natural ligands have been found. Interestingly, the majority of these receptors are expressed within the central nervous system where they represent a largely untapped resource for the treatment of neurological diseases. The focus of this review will thus be on the emerging roles of two brain-expressed orphan GPCRs, GPR37 and GPR37 L1, in regulating various cellular and molecular processes underlying ischemic stroke.
Collapse
|
45
|
Dumontet T, Hammer GD. Bones and adrenal organogenesis: how embryonic osteocalcin influences lifelong adrenal function. J Clin Invest 2022; 132:157200. [PMID: 35166237 PMCID: PMC8843705 DOI: 10.1172/jci157200] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Osteocalcin is a hormone produced in bones by osteoblasts during bone formation. Numerous studies have demonstrated that adrenal gland–derived glucocorticoids inhibit osteocalcin production, which can ultimately cause deleterious bones loss. This loss establishes a unidirectional endocrine relationship between the adrenal glands and bone, however, whether osteocalcin reciprocally regulates glucocorticoid secretion remains unclear. In this issue of the JCI, Yadav and colleagues address how bone-derived osteocalcin influences adrenal organogenesis and function. Using a large variety of animal models, the authors established that embryonic osteocalcin signaling, specifically through the GPR158 receptor, regulates postnatal adrenal steroid concentrations throughout life. This work has translational potential, and we await future investigations that determine whether modulating osteocalcin levels could promote endogenous adrenocortical function in adrenocortical hypoplasia and glucocorticoid deficiency.
Collapse
|
46
|
Berger JM, Karsenty G. Osteocalcin and the Physiology of Danger. FEBS Lett 2021; 596:665-680. [PMID: 34913486 PMCID: PMC9020278 DOI: 10.1002/1873-3468.14259] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 11/29/2021] [Accepted: 12/07/2021] [Indexed: 12/02/2022]
Abstract
Bone biology has long been driven by the question as to what molecules affect cell differentiation or the functions of bone. Exploring this issue has been an extraordinarily powerful way to improve our knowledge of bone development and physiology. More recently, a second question has emerged: does bone have other functions besides making bone? Addressing this conundrum revealed that the bone-derived hormone osteocalcin affects a surprisingly large number of organs and physiological processes, including acute stress response. This review will focus on this emerging aspect of bone biology taking osteocalcin as a case study and will show how classical and endocrine functions of bone help to define a new functional identity for this tissue.
Collapse
Affiliation(s)
- Julian Meyer Berger
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, NY, 10032, USA
| | - Gerard Karsenty
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University, NY, 10032, USA
| |
Collapse
|
47
|
Hertz E, Saarinen M, Svenningsson P. GM1 Is Cytoprotective in GPR37-Expressing Cells and Downregulates Signaling. Int J Mol Sci 2021; 22:ijms222312859. [PMID: 34884663 PMCID: PMC8657933 DOI: 10.3390/ijms222312859] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/15/2021] [Accepted: 11/17/2021] [Indexed: 01/02/2023] Open
Abstract
G-protein-coupled receptors (GPCRs) are commonly pharmacologically modulated due to their ability to translate extracellular events to intracellular changes. Previously, studies have mostly focused on protein–protein interactions, but the focus has now expanded also to protein–lipid connections. GM1, a brain-expressed ganglioside known for neuroprotective effects, and GPR37, an orphan GPCR often reported as a potential drug target for diseases in the central nervous system, have been shown to form a complex. In this study, we looked into the functional effects. Endogenous GM1 was downregulated when stably overexpressing GPR37 in N2a cells (N2aGPR37-eGFP). However, exogenous GM1 specifically rescued N2aGPR37-eGFP from toxicity induced by the neurotoxin MPP+. The treatment did not alter transcription levels of GPR37 or the enzyme responsible for GM1 production, both potential mechanisms for the effect. However, GM1 treatment inhibited cAMP-dependent signaling from GPR37, here reported as potentially consecutively active, possibly contributing to the protective effects. We propose an interplay between GPR37 and GM1 as one of the many cytoprotective effects reported for GM1.
Collapse
Affiliation(s)
- Ellen Hertz
- Correspondence: (E.H.); (P.S.); Tel.: +46-8517-74-614 (E.H.)
| | | | | |
Collapse
|