1
|
Zhang J, Sun J, Li J, Xia H. Targeting the GDF15 Signalling for Obesity Treatment: Recent Advances and Emerging Challenges. J Cell Mol Med 2024; 28:e70251. [PMID: 39700016 DOI: 10.1111/jcmm.70251] [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: 03/09/2024] [Revised: 11/06/2024] [Accepted: 11/15/2024] [Indexed: 12/21/2024] Open
Abstract
The growth differentiation factor 15 (GDF15)-glial cell-derived neurotrophic factor family receptor alpha-like (GFRAL) pathway plays a crucial role in the regulation of metabolism, appetite and body weight control. Obesity is an increasingly prevalent chronic disease worldwide, necessitating effective treatment strategies. Recent preclinical and clinical studies have highlighted that targeting the GDF15-GFRAL signalling pathway is a promising approach for treating obesity, particularly because it has minimal impact on skeletal muscle mass, which is essential to preserve during weight loss. Given its distinctive mechanisms, the GDF15-GFRAL axis represents an attractive target for addressing various metabolic disorders, especially obesity. In this review, we will explore how the GDF15-GFRAL axis is regulated, its distribution in the body and its role in the regulation of metabolism, appetite and obesity. Additionally, we will discuss recent advances and potential challenges in targeting the GDF15-GFRAL axis for obesity treatment.
Collapse
Affiliation(s)
- Jincheng Zhang
- Division of Abdominal Tumor Multimodality Treatment, Department of Medical Oncology, Cancer Center and National Clinical Research Center for Geriatrics and Laboratory of Molecular Targeted Therapy in Oncology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- School of Physical Education and Sports, Sichuan University, Chengdu, China
- Research Institute of Molecular Exercise Science, Hungarian University of Sports Science, Budapest, Hungary
| | - Jingquan Sun
- School of Physical Education and Sports, Sichuan University, Chengdu, China
| | - Jielang Li
- Division of Abdominal Tumor Multimodality Treatment, Department of Medical Oncology, Cancer Center and National Clinical Research Center for Geriatrics and Laboratory of Molecular Targeted Therapy in Oncology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Hongwei Xia
- Division of Abdominal Tumor Multimodality Treatment, Department of Medical Oncology, Cancer Center and National Clinical Research Center for Geriatrics and Laboratory of Molecular Targeted Therapy in Oncology, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
2
|
Zhang H, Mulya A, Nieuwoudt S, Vandanmagsar B, McDowell R, Heintz EC, Zunica ER, Collier JJ, Bozadjieva-Kramer N, Seeley RJ, Axelrod CL, Kirwan JP. GDF15 Mediates the Effect of Skeletal Muscle Contraction on Glucose-Stimulated Insulin Secretion. Diabetes 2023; 72:1070-1082. [PMID: 37224335 PMCID: PMC10382648 DOI: 10.2337/db22-0019] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 05/21/2023] [Indexed: 05/26/2023]
Abstract
Exercise is a first-line treatment for type 2 diabetes and preserves β-cell function by hitherto unknown mechanisms. We postulated that proteins from contracting skeletal muscle may act as cellular signals to regulate pancreatic β-cell function. We used electric pulse stimulation (EPS) to induce contraction in C2C12 myotubes and found that treatment of β-cells with EPS-conditioned medium enhanced glucose-stimulated insulin secretion (GSIS). Transcriptomics and subsequent targeted validation revealed growth differentiation factor 15 (GDF15) as a central component of the skeletal muscle secretome. Exposure to recombinant GDF15 enhanced GSIS in cells, islets, and mice. GDF15 enhanced GSIS by upregulating the insulin secretion pathway in β-cells, which was abrogated in the presence of a GDF15 neutralizing antibody. The effect of GDF15 on GSIS was also observed in islets from GFRAL-deficient mice. Circulating GDF15 was incrementally elevated in patients with pre- and type 2 diabetes and positively associated with C-peptide in humans with overweight or obesity. Six weeks of high-intensity exercise training increased circulating GDF15 concentrations, which positively correlated with improvements in β-cell function in patients with type 2 diabetes. Taken together, GDF15 can function as a contraction-induced protein that enhances GSIS through activating the canonical signaling pathway in a GFRAL-independent manner. ARTICLE HIGHLIGHTS Exercise improves glucose-stimulated insulin secretion through direct interorgan communication. Contracting skeletal muscle releases growth differentiation factor 15 (GDF15), which is required to synergistically enhance glucose-stimulated insulin secretion. GDF15 enhances glucose-stimulated insulin secretion by activating the canonical insulin release pathway. Increased levels of circulating GDF15 after exercise training are related to improvements in β-cell function in patients with type 2 diabetes.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA
| | - Anny Mulya
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Stephan Nieuwoudt
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Bolormaa Vandanmagsar
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA
| | - Ruth McDowell
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
| | - Elizabeth C. Heintz
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA
| | - Elizabeth R.M. Zunica
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA
| | - J. Jason Collier
- Islet Biology and Inflammation Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA
| | - Nadejda Bozadjieva-Kramer
- Department of Surgery, University of Michigan, Ann Arbor, MI
- Veterans Affairs Ann Arbor Healthcare System, Research Service, Ann Arbor, MI
| | - Randy J. Seeley
- Department of Surgery, University of Michigan, Ann Arbor, MI
| | - Christopher L. Axelrod
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA
| | - John P. Kirwan
- Department of Physiology and Biophysics, School of Medicine, Case Western Reserve University, Cleveland, OH
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH
- Integrated Physiology and Molecular Medicine Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA
| |
Collapse
|
3
|
Ma Y, Li W, Fan C, Wang Y, Jiang H, Yang W. Comprehensive Analysis of Long Non-Coding RNAs N4-Acetylcytidine in Alzheimer's Disease Mice Model Using High-Throughput Sequencing. J Alzheimers Dis 2022; 90:1659-1675. [PMID: 36314201 DOI: 10.3233/jad-220564] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND N4-acetylcytidine (ac4C), an important posttranscriptional modification, is involved in various disease processes. Long noncoding RNAs (lncRNAs) regulate gene expression mainly through epigenetic modification, transcription, and posttranscriptional modification. Alzheimer's disease (AD) is a neurodegenerative disease characterized by amyloidosis of the brain. However, the role of lncRNA ac4C modification in AD remains unclear. OBJECTIVE In this study, we investigated the association between ac4C modification and AD, and the underlying mechanisms of ac4C modification in AD. METHODS The male 9-month-old APP/PS1 double transgenic mice, age- and sex-matched wild type (WT) mice were used in this study. Then, ac4C-RIP-seq and RNA-seq were used to comprehensively analyze lncRNA ac4C modification in AD mice. The lncRNA-miRNA-mRNA regulatory networks using Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses showed the regulatory relationships among these three lncRNAs and AD. RESULTS The results showed that there were 120 significantly different ac4C peaks located on 102 lncRNAs in AD, of which 55 were hyperacetylated and 47 were hypoacetylated. Simultaneously, 231 differentially expressed lncRNAs were identified, including 138 upregulated lncRNAs and 93 downregulated lncRNAs. Moreover, 3 lncRNAs, lncRNA Gm26508, lncRNA A430046D13Rik, and lncRNA 9530059O14Rik, showed significant changes in both the ac4C and RNA levels using conjoint analysis. CONCLUSION The abundance of lncRNA ac4C modification is significantly different in AD and indicates that lncRNA ac4C is associated with the occurrence and development of AD, which could provide a basis for further exploration of the related regulatory mechanisms.
Collapse
Affiliation(s)
- Yanzhen Ma
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Weizu Li
- Department of Pharmacology, Basic Medicine College, Key Laboratory of Anti-inflammatory and Immunopharmacology, Ministry of Education, Anhui Medical University, Hefei, Anhui, China
| | - Chang Fan
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Yongzhong Wang
- Key Laboratory of Xin'an Medicine of the Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, China.,Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Hui Jiang
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China.,Key Laboratory of Xin'an Medicine of the Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, China
| | - Wenming Yang
- Key Laboratory of Xin'an Medicine of the Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, China.,Encephalopathy Center, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China
| |
Collapse
|
4
|
Kim S, Larrous F, Varet H, Legendre R, Feige L, Dumas G, Matsas R, Kouroupi G, Grailhe R, Bourhy H. Early Transcriptional Changes in Rabies Virus-Infected Neurons and Their Impact on Neuronal Functions. Front Microbiol 2021; 12:730892. [PMID: 34970230 PMCID: PMC8713068 DOI: 10.3389/fmicb.2021.730892] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 10/25/2021] [Indexed: 12/24/2022] Open
Abstract
Rabies is a zoonotic disease caused by rabies virus (RABV). As rabies advances, patients develop a variety of severe neurological symptoms that inevitably lead to coma and death. Unlike other neurotropic viruses that can induce symptoms of a similar range, RABV-infected post-mortem brains do not show significant signs of inflammation nor the structural damages on neurons. This suggests that the observed neurological symptoms possibly originate from dysfunctions of neurons. However, many aspects of neuronal dysfunctions in the context of RABV infection are only partially understood, and therefore require further investigation. In this study, we used differentiated neurons to characterize the RABV-induced transcriptomic changes at the early time-points of infection. We found that the genes modulated in response to the infection are particularly involved in cell cycle, gene expression, immune response, and neuronal function-associated processes. Comparing a wild-type RABV to a mutant virus harboring altered matrix proteins, we found that the RABV matrix protein plays an important role in the early down-regulation of host genes, of which a significant number is involved in neuronal functions. The kinetics of differentially expressed genes (DEGs) are also different between the wild type and mutant virus datasets. The number of modulated genes remained constant upon wild-type RABV infection up to 24 h post-infection, but dramatically increased in the mutant condition. This result suggests that the intact viral matrix protein is important to control the size of host gene modulation. We then examined the signaling pathways previously studied in relation to the innate immune responses against RABV, and found that these pathways contribute to the changes in neuronal function-associated processes. We further examined a set of regulated genes that could impact neuronal functions collectively, and demonstrated in calcium imaging that indeed the spontaneous activity of neurons is influenced by RABV infection. Overall, our findings suggest that neuronal function-associated genes are modulated by RABV early on, potentially through the viral matrix protein-interacting signaling molecules and their downstream pathways.
Collapse
Affiliation(s)
- Seonhee Kim
- Technology Development Platform, Institut Pasteur Korea, Seongnam, South Korea
- Institut Pasteur, Université de Paris, Lyssavirus Epidemiology and Neuropathology Unit, Paris, France
- Université de Paris, Doctoral School Bio Sorbonne Paris Cité, Paris, France
| | - Florence Larrous
- Institut Pasteur, Université de Paris, Lyssavirus Epidemiology and Neuropathology Unit, Paris, France
| | - Hugo Varet
- Institut Pasteur, Université de Paris, Hub de Bioinformatique et Biostatistique, Département Biologie Computationnelle, Paris, France
- Institut Pasteur, Université de Paris, Plate-Forme Technologique Biomics, Centre de Ressources et Recherches Technologiques (C2RT), Paris, France
| | - Rachel Legendre
- Institut Pasteur, Université de Paris, Hub de Bioinformatique et Biostatistique, Département Biologie Computationnelle, Paris, France
- Institut Pasteur, Université de Paris, Plate-Forme Technologique Biomics, Centre de Ressources et Recherches Technologiques (C2RT), Paris, France
| | - Lena Feige
- Technology Development Platform, Institut Pasteur Korea, Seongnam, South Korea
- Institut Pasteur, Université de Paris, Lyssavirus Epidemiology and Neuropathology Unit, Paris, France
- Université de Paris, Doctoral School Bio Sorbonne Paris Cité, Paris, France
| | - Guillaume Dumas
- Department of Psychiatry, CHU Sainte-Justine Research Center, University of Montreal, Montreal, QC, Canada
- Mila, Quebec Artificial Intelligence Institute, University of Montreal, Montreal, QC, Canada
| | - Rebecca Matsas
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Georgia Kouroupi
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Regis Grailhe
- Technology Development Platform, Institut Pasteur Korea, Seongnam, South Korea
| | - Hervé Bourhy
- Institut Pasteur, Université de Paris, Lyssavirus Epidemiology and Neuropathology Unit, Paris, France
| |
Collapse
|
5
|
Ma Z, Viswanathan G, Sellig M, Jassal C, Choi I, Garikipati A, Xiong X, Nazo N, Rajagopal S. β-Arrestin–Mediated Angiotensin II Type 1 Receptor Activation Promotes Pulmonary Vascular Remodeling in Pulmonary Hypertension. JACC Basic Transl Sci 2021; 6:854-869. [PMID: 34869949 PMCID: PMC8617598 DOI: 10.1016/j.jacbts.2021.09.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 09/24/2021] [Accepted: 09/24/2021] [Indexed: 11/05/2022]
Abstract
We tested the effects of a β-arrestin–biased agonist (TRV023) of the angiotensin II (AngII) type 1 receptor (AT1R), which acts as a vasodilator while not blocking cellular proliferation, compared to a balanced agonist, AngII, and an antagonist, losartan, in PAH. In acute infusion, AngII increased right ventricular pressures while TRV023 and losartan did not. However, in chronic infusion in monocrotaline PAH rats, both TRV023 and AngII had significantly worse survival than losartan. Both TRV023 and AngII enhanced proliferation and migration of pulmonary artery smooth muscle cells from patients with PAH. β-arrestin-mediated AT1R signaling promotes vascular remodeling and worsens PAH, and suggests that the benefit of current PAH therapies is primarily through pulmonary vascular reverse remodeling.
Pulmonary arterial hypertension (PAH) is a disease of abnormal pulmonary vascular remodeling whose medical therapies are thought to primarily act as vasodilators but also may have effects on pulmonary vascular remodeling. The angiotensin II type 1 receptor (AT1R) is a G protein–coupled receptor that promotes vasoconstriction through heterotrimeric G proteins but also signals via β-arrestins, which promote cardioprotective effects and vasodilation through promoting cell survival. We found that an AT1R β-arrestin-biased agonist promoted vascular remodeling and worsened PAH, suggesting that the primary benefit of current PAH therapies is through pulmonary vascular reverse remodeling in addition to their vasodilation.
Collapse
|
6
|
Jiang WW, Zhang ZZ, He PP, Jiang LP, Chen JZ, Zhang XT, Hu M, Zhang YK, Ouyang XP. Emerging roles of growth differentiation factor-15 in brain disorders (Review). Exp Ther Med 2021; 22:1270. [PMID: 34594407 PMCID: PMC8456456 DOI: 10.3892/etm.2021.10705] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 08/06/2021] [Indexed: 12/14/2022] Open
Abstract
Brain disorders, such as Alzheimer's and Parkinson's disease and cerebral stroke, are an important contributor to mortality and disability worldwide, where their pathogenesis is currently a topic of intense research. The mechanisms underlying the development of brain disorders are complex and vary widely, including aberrant protein aggregation, ischemic cell necrosis and neuronal dysfunction. Previous studies have found that the expression and function of growth differentiation factor-15 (GDF15) is closely associated with the incidence of brain disorders. GDF15 is a member of the TGFβ superfamily, which is a dimer-structured stress-response protein. The expression of GDF15 is regulated by a number of proteins upstream, including p53, early growth response-1, non-coding RNAs and hormones. In particular, GDF15 has been reported to serve an important role in regulating angiogenesis, apoptosis, lipid metabolism and inflammation. For example, GDF15 can promote angiogenesis by promoting the proliferation of human umbilical vein endothelial cells, apoptosis of prostate cancer cells and fat metabolism in fasted mice, and GDF15 can decrease the inflammatory response of lipopolysaccharide-treated mice. The present article reviews the structure and biosynthesis of GDF15, in addition to the possible roles of GDF15 in Alzheimer's disease, cerebral stroke and Parkinson's disease. The purpose of the present review is to summarize the mechanism underlying the role of GDF15 in various brain disorders, which hopes to provide evidence and guide the prevention and treatment of these debilitating conditions.
Collapse
Affiliation(s)
- Wei-Wei Jiang
- Department of Physiology, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Zi-Zhen Zhang
- Department of Medical Humanities, School of Medicine, Hunan Polytechnic of Environment and Biology, Hengyang, Hunan 421001, P.R. China
| | - Ping-Ping He
- Hunan Province Cooperative Innovation Centre for Molecular Target New Drug Study, Nursing School, University of South China, Hengyang, Hunan 421001, P.R. China.,Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Li-Ping Jiang
- Department of Physiology, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, P.R. China.,Department of Critical Care Medicine, Hunan Taihe Hospital, Changsha, Hunan 410004, P.R. China
| | - Jin-Zhi Chen
- Department of Physiology, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xing-Ting Zhang
- Department of Physiology, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Mi Hu
- Department of Physiology, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Yang-Kai Zhang
- Department of Physiology, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, P.R. China
| | - Xin-Ping Ouyang
- Department of Physiology, Institute of Neuroscience Research, Hengyang Key Laboratory of Neurodegeneration and Cognitive Impairment, Hengyang Medical College, University of South China, Hengyang, Hunan 421001, P.R. China.,Institute of Cardiovascular Research, Key Laboratory for Atherosclerology of Hunan Province, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, P.R. China
| |
Collapse
|
7
|
Lin W, Zhang WW, Lyu N, Cao H, Xu WD, Zhang YQ. Growth Differentiation Factor-15 Produces Analgesia by Inhibiting Tetrodotoxin-Resistant Nav1.8 Sodium Channel Activity in Rat Primary Sensory Neurons. Neurosci Bull 2021; 37:1289-1302. [PMID: 34076854 PMCID: PMC8423960 DOI: 10.1007/s12264-021-00709-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/10/2021] [Indexed: 01/01/2023] Open
Abstract
Growth differentiation factor 15 (GDF-15) is a member of the transforming growth factor-β superfamily. It is widely distributed in the central and peripheral nervous systems. Whether and how GDF-15 modulates nociceptive signaling remains unclear. Behaviorally, we found that peripheral GDF-15 significantly elevated nociceptive response thresholds to mechanical and thermal stimuli in naïve and arthritic rats. Electrophysiologically, we demonstrated that GDF-15 decreased the excitability of small-diameter dorsal root ganglia (DRG) neurons. Furthermore, GDF-15 concentration-dependently suppressed tetrodotoxin-resistant sodium channel Nav1.8 currents, and shifted the steady-state inactivation curves of Nav1.8 in a hyperpolarizing direction. GDF-15 also reduced window currents and slowed down the recovery rate of Nav1.8 channels, suggesting that GDF-15 accelerated inactivation and slowed recovery of the channel. Immunohistochemistry results showed that activin receptor-like kinase-2 (ALK2) was widely expressed in DRG medium- and small-diameter neurons, and some of them were Nav1.8-positive. Blockade of ALK2 prevented the GDF-15-induced inhibition of Nav1.8 currents and nociceptive behaviors. Inhibition of PKA and ERK, but not PKC, blocked the inhibitory effect of GDF-15 on Nav1.8 currents. These results suggest a functional link between GDF-15 and Nav1.8 in DRG neurons via ALK2 receptors and PKA associated with MEK/ERK, which mediate the peripheral analgesia of GDF-15.
Collapse
Affiliation(s)
- Wei Lin
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Wen-Wen Zhang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Ning Lyu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
| | - Hong Cao
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China
| | - Wen-Dong Xu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
- Department of Hand Surgery, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Yu-Qiu Zhang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai, 200032, China.
- Department of Neurobiology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
8
|
Yeh KH, Chang YT, Juang JMJ, Chiang FT, Teng MS, Wu S, Lin JF, Ko YL. Combined corrected QT interval and growth differentiation factor-15 level has synergistic predictive value for long-term outcome of angiographically confirmed coronary artery disease. Int J Clin Pract 2021; 75:e14180. [PMID: 33759309 DOI: 10.1111/ijcp.14180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND The corrected QT interval (QTc) predicts prognosis for the general population and patients with coronary artery disease (CAD). Growth differentiation factor-15 (GDF-15) is a biomarker of myocardial fibrosis and left ventricular (LV) remodelling. The interaction between these two parameters is unknown. SUBJECTS AND METHODS This study included 487 patients with angiographically confirmed CAD. QTc was calculated using the Bazett formula. Multiple biochemistries and GDF-15 levels were measured. The primary endpoint was total mortality, and the secondary endpoints comprised the combination of total mortality, myocardial infarction and hospitalisation for heart failure and stroke. RESULTS The mean follow-up period was 1029 ± 343 days (5-1692 days), during which 21 patients died and 47 had secondary endpoints. ROC curve analysis for the optimal cut-off value of primary endpoint is 1.12 ng/mL for GDF-15 (AUC = 0.787, P = 9.0 × 10-6 ) and 438.5 msec for QTc (AUC = 0.698, P = .002). Utilising linear regression, QTc has a positive correlation with Log-GDF-15 (r = .216, P = 1.0 × 10-6 ). Utilising Kaplan-Meier analysis, both QTc interval and GDF-15 level are significant predictors for primary end point (P = .000194, P = 2.0 × 10-6 , respectively) and secondary endpoint (P = .00028, P = 6.15 × 10-8 , respectively). When combined these two parameters together, a significant synergistic predictive power was noted for primary and secondary endpoint (P = 2.31 × 10-7 , P = 1.26 × 10-8 , respectively). This combined strategy also showed significant correlation with the severity of CAD (P < .001). CONCLUSION In Chinese patient with angiographically confirmed CAD, a combined strategy utilising an ECG parameter (QTc) and a circulating biomarker (GDF-15) has good correlation with the severity of CAD, and improves the predictive power for total mortality.
Collapse
Affiliation(s)
- Kuan-Hung Yeh
- Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Yao-Ting Chang
- Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - Jyh-Ming Jimmy Juang
- Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Fu-Tien Chiang
- Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- College of Medicine, National Taiwan University, Taipei, Taiwan
- Cardiovascular Center and Division of Cardiology, Fu-Jen Catholic University Hospital, New Taipei City, Taiwan
| | - Ming-Sheng Teng
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| | - Semon Wu
- Department of Life Science, Chinese Culture University, Taipei, Taiwan
| | - Jeng-Feng Lin
- Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Yu-Lin Ko
- Cardiovascular Center and Division of Cardiology, Department of Internal Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Research, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
| |
Collapse
|
9
|
Yang S, Lu W, Zhao C, Zhai Y, Wei Y, Liu J, Yu Y, Li Z, Shi J. Leukemia cells remodel marrow adipocytes via TRPV4-dependent lipolysis. Haematologica 2020; 105:2572-2583. [PMID: 33131246 PMCID: PMC7604636 DOI: 10.3324/haematol.2019.225763] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 12/18/2019] [Indexed: 11/09/2022] Open
Abstract
Remodeling of adipocyte morphology and function plays a critical role in prostate cancer development. We previously reported that leukemia cells secrete growth differentiation factor 15 (GDF15),which remodels the residual bone marrow (BM) adipocytes into small adipocytes and is associated with a poor prognosis in acute myeloid leukemia (AML) patients. However, little is known about how GDF15 drives BM adipocyte remodeling. In this study, we examined the role of the transient receptor potential vanilloid (TRPV) channels in the remodeling of BM adipocytes exposed to GDF15. We found that TRPV4 negatively regulated GDF15-induced remodeling of BM adipocytes. Furthermore, transforming growth factor-β type II receptor (TGFβRII) was identified as the main receptor for GDF15 on BM adipocytes. PI3K inhibitor treatment reduced GDF15-induced pAKT, identifying PI3K/AKT as the downstream stress response pathway. Subsequently, GDF15 reduced the expression of the transcription factor Forkhead box C1 (FOXC1) in BM adipocytes subjected to RNA-seq screening and Western blot analyse. Moreover, it was also confirmed that FOXC1 combined with the TRPV4 promoter by the Chip-qPCR experiments, which suggests that FOXC1 mediates GDF15 regulation of TRPV4. In addition, an AML mouse model exhibited smaller BM adipocytes, whereas the TRPV4 activator 4α-phorbol 12,13-didecanoate (4αPDD) partly rescued this process and increased survival. In conclusion, TRPV4 plays a critical role in BM adipocyte remodeling induced by leukemia cells, suggesting that targeting TRPV4 may constitute a novel strategy for AML therapy.
Collapse
Affiliation(s)
- Shaoxin Yang
- Department of Hematology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine
| | - Wei Lu
- Department of Hematology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine
| | - Chong Zhao
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital
| | - Yuanmei Zhai
- Department of Hematology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine
| | - Yanyu Wei
- Department of Hematology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine
| | - Jiali Liu
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital
| | - Yehua Yu
- Department of Hematology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine
| | - Zhiqiang Li
- Department of Blood Transfusion, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jun Shi
- Department of Hematology, Shanghai Ninth People's Hospital, Shanghai JiaoTong University School of Medicine
| |
Collapse
|
10
|
GDF15, an update of the physiological and pathological roles it plays: a review. Pflugers Arch 2020; 472:1535-1546. [PMID: 32936319 DOI: 10.1007/s00424-020-02459-1] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Revised: 08/06/2020] [Accepted: 09/02/2020] [Indexed: 02/07/2023]
Abstract
Growth differentiation factor 15 (GDF15) is a peptide hormone, and a divergent member of the transforming growth factor beta (TGFβ) superfamily. In normal physiology, GDF15 is expressed in multiple tissues at a low concentration. GDF15 is overexpressed during and following many pathological conditions such as tissue injury and inflammation in order to play a protective role. However, GDF15 appears to promote tumour growth in the later stages of malignant cancer. The recently identified endogenous receptor for GDF15, GDNF family receptor a-like (GFRAL), has allowed elucidation of a physiological pathway in which GDF15 regulates energy homeostasis and body weight, primarily via appetite suppression. The anorectic effect of GDF15 provides some therapeutic potential in management of cancer-related anorexia/cachexia and obesity. Despite the identification of GFRAL as a GDF15 receptor, there appears to be other signalling mechanisms utilized by GDF15 that further increase the possibility of development of therapeutic treatments, should these pathways be fully characterized. In this review, GDF15 function in both physiological and pathological conditions in various tissues will be discussed.
Collapse
|
11
|
Okamoto M, Koma YI, Kodama T, Nishio M, Shigeoka M, Yokozaki H. Growth Differentiation Factor 15 Promotes Progression of Esophageal Squamous Cell Carcinoma via TGF-β Type II Receptor Activation. Pathobiology 2020; 87:100-113. [PMID: 31896114 DOI: 10.1159/000504394] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 10/28/2019] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVES Growth differentiation factor 15 (GDF15), which is derived from tumor-associated macrophages (TAM) and cancer cells, promotes progression of esophageal squamous cell carcinomas (ESCC). However, its role in the ESCC microenvironment remains unclear. Here, we examined the effects of GDF15 on ESCC cell lines and tissues. METHODS Western blotting, MTS, and Transwell migration/invasion assays were used to evaluate cell signaling, proliferation, and migration/invasion, respectively, in ESCC cell lines treated with recombinant human GDF15 (rhGDF15). ESCC cell lines were administered a TGF-βRI/II inhibitor (LY2109761), small interfering RNA against TGF-β type II receptor (TGF-βRII), or neutralizing antibody against TGF-βRII to study the role of TGF-βRII in mediating the effects of rhGDF15. The localization of GDF15 and TGF-βRII in ESCC cell lines was observed by immunofluorescence. TGF-βRII expression in ESCC tissues was analyzed by immunohistochemistry, and the relationship between clinicopathological factors and prognosis in ESCC patients was evaluated. RESULTS rhGDF15 increased levels of phosphorylated Akt, Erk1/2, and TGF-βRII in ESCC cell lines. Inhibition/knockdown of TGF-βRII suppressed rhGDF15-induced activation of Akt and Erk1/2 and enhancement of cellular proliferation, migration, and invasion. Immunofluorescence revealed that TGF-βRII and GDF15 were colocalized in ESCC cell lines. High TGF-βRII expression in ESCC tissues, as determined by immunohistochemistry, correlated with depth of invasion and increased number of infiltrating TAMs. ESCC patients with high TGF-βRII expression showed a tendency toward poor prognosis. CONCLUSIONS GDF15 promotes ESCC progression by increasing cellular proliferation, migration, and invasion via TGF-βRII signaling.
Collapse
Affiliation(s)
- Maiko Okamoto
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Yu-Ichiro Koma
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan,
| | - Takayuki Kodama
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Mari Nishio
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Manabu Shigeoka
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Hiroshi Yokozaki
- Division of Pathology, Department of Pathology, Kobe University Graduate School of Medicine, Kobe, Japan
| |
Collapse
|
12
|
Kaur S, Maslov LN, Singh N, Jaggi AS. Dual role of T-type calcium channels in anxiety-related behavior. J Basic Clin Physiol Pharmacol 2019; 31:/j/jbcpp.ahead-of-print/jbcpp-2019-0067/jbcpp-2019-0067.xml. [PMID: 31644427 DOI: 10.1515/jbcpp-2019-0067] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 08/23/2019] [Indexed: 06/10/2023]
Abstract
T-type calcium channels are low voltage activated calcium channels that are widely expressed in various brain regions including stress-responsive regions. These channels regulate the diverse functions of the central nervous system, and modulation of these channels is shown to modulate the anxiety. Studies have described that modulation of T-type calcium channels may either aggravate or ameliorate anxiety-related behavior, suggesting the dual role of these channels. The studies employing animals with overexpression of T-type calcium channels reported their anxiety-inducing role. Therefore, the blockade of these channels using various pharmacological agents such as ethosuximide, plant extracts of linalool or rosemary, and corticotropin-releasing factor (CRF) is reported to ameliorate anxiety. On the contrary, knockout of the gene encoding these channels predisposes the rodents to anxiety-related disorders, suggesting the anxiety-attenuating role of these channels. It may be possible that these channels in normal or basal state attenuate anxiety, whereas activation of these channels in stressful condition may produce anxiety. The present review describes the dual role of T-type calcium channels in anxiety-related behavior in both preclinical and clinical studies.
Collapse
Affiliation(s)
- Simranjot Kaur
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Patiala 147002, India
| | - Leonid N Maslov
- Laboratory of Experimental Cardiology, Institute of Cardiology, Tomsk 634012, Russia
| | - Nirmal Singh
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Patiala 147002, India
| | - Amteshwar Singh Jaggi
- Department of Pharmaceutical Sciences and Drug Research, Punjabi University Patiala, Patiala 147002, India, Mobile: +919501016036
| |
Collapse
|
13
|
Ha G, De Torres F, Arouche N, Benzoubir N, Ferratge S, Hatem E, Anginot A, Uzan G. GDF15 secreted by senescent endothelial cells improves vascular progenitor cell functions. PLoS One 2019; 14:e0216602. [PMID: 31075112 PMCID: PMC6510423 DOI: 10.1371/journal.pone.0216602] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 04/24/2019] [Indexed: 12/31/2022] Open
Abstract
Endothelial dysfunction (ED) is part of the first steps in the development of cardiovascular diseases (CVD). Growth Differentiation Factor 15 (GDF15) is a cytokine belonging to the Transforming Growth Factor β superfamily and its expression is increased both during ED and in CVD. Because high blood levels of GDF15 have been reported during ED, we hypothesized that GDF15 could be produced by endothelial cells in response to a vascular stress, possibly to attenuate endothelial function loss. Since senescence is mainly involved in both vascular stress and endothelial function loss, we used Endothelial Colony Forming Cells generated from adult blood (AB-ECFCs) as a model of endothelial cells to investigate GDF15 expression during cellular senescence. Then, we analyzed the potential role of GDF15 in AB-ECFC functions and senescence. When AB-ECFCs become senescent, they secrete increased levels of GDF15. We investigated GDF15 paracrine effects on non-senescent AB-ECFCs and showed that GDF15 enhanced proliferation, migration, NO production and activated several signaling pathways including AKT, ERK1/2 and SMAD2 without triggering any oxidative stress. Taken together, our results suggest that GDF15 production by senescent AB-ECFCs could act in a paracrine manner on non-senescent AB-ECFCs, and that this interaction could be beneficial to its model cells. Therefore, GDF15 could play a beneficial role in a dysfunctional vascular system as previously reported in patients with CVD, by limiting ED related to vascular stress occurring in these diseases.
Collapse
Affiliation(s)
- Guillaume Ha
- INSERM U1197, Hôpital Paul Brousse, Villejuif, France
- Université Paris-Diderot, Paris, France
| | | | | | | | | | - Elie Hatem
- INSERM U1197, Hôpital Paul Brousse, Villejuif, France
| | | | - Georges Uzan
- INSERM U1197, Hôpital Paul Brousse, Villejuif, France
| |
Collapse
|
14
|
Zhao QR, Lu JM, Li ZY, Mei YA. Neuritin promotes neurite and spine growth in rat cerebellar granule cells via L-type calcium channel-mediated calcium influx. J Neurochem 2018; 147:40-57. [PMID: 29920676 PMCID: PMC6220818 DOI: 10.1111/jnc.14535] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 05/21/2018] [Accepted: 06/13/2018] [Indexed: 01/15/2023]
Abstract
Neuritin is a neurotrophic factor that is activated by neural activity and neurotrophins. Its major function is to promote neurite growth and branching; however, the underlying mechanisms are not fully understood. To address this issue, this study investigated the effects of neuritin on neurite and spine growth and intracellular Ca2+ concentration in rat cerebellar granule neurons (CGNs). Incubation of CGNs for 24 h with neuritin increased neurite length and spine density; this effect was mimicked by insulin and abolished by inhibiting insulin receptor (IR) or mitogen‐activated protein kinase kinase/extracellular signal‐regulated kinase (ERK) activity. Calcium imaging and western blot analysis revealed that neuritin enhanced the increase in intracellular Ca2+ level induced by high K+, and stimulated the cell surface expression of CaV1.2 and CaV1.3 α subunits of the L‐type calcium channel, which was suppressed by inhibition of IR or mitogen‐activated protein kinase kinase/ERK. Treatment with inhibitors of L‐type calcium channels, calmodulin, and calcineurin (CaN) abrogated the effects of neuritin on neurite length and spine density. A similar result was obtained by silencing nuclear factor of activated T cells c4, which is known to be activated by neuritin in CGNs. These results indicate that IR and ERK signaling as well as the Ca2+/CaN/nuclear factor of activated T cells c4 axis mediate the effects of neuritin on neurite and spine growth in CGNs. Open Practices
Open Science: This manuscript was awarded with the Open Materials Badge. For more information see: https://cos.io/our-services/open-science-badges/ ![]()
Cover Image for this issue: doi: 10.1111/jnc.14195.
Collapse
Affiliation(s)
- Qian-Ru Zhao
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and School of Life Sciences, Fudan University, Shanghai, China
| | - Jun-Mei Lu
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and School of Life Sciences, Fudan University, Shanghai, China
| | - Zhao-Yang Li
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and School of Life Sciences, Fudan University, Shanghai, China
| | - Yan-Ai Mei
- Institutes of Brain Science, State Key Laboratory of Medical Neurobiology and School of Life Sciences, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Mullican SE, Rangwala SM. Uniting GDF15 and GFRAL: Therapeutic Opportunities in Obesity and Beyond. Trends Endocrinol Metab 2018; 29:560-570. [PMID: 29866502 DOI: 10.1016/j.tem.2018.05.002] [Citation(s) in RCA: 91] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 05/04/2018] [Accepted: 05/12/2018] [Indexed: 01/25/2023]
Abstract
Growth differentiation factor-15 (GDF15) is a circulating protein that has been implicated in multiple biological processes, including energy homeostasis, body weight regulation, and cachexia driven by cancer and chronic disease. The potential to target GDF15 in the treatment of energy-intake disorders, including obesity and anorexia, is an area of intense investigation, but has been limited by the lack of an identified receptor, signaling mechanism, and target tissue. GDNF family receptor α-like (GFRAL) was recently identified as the neuronal brainstem receptor responsible for mediating the anorectic actions of GDF15. Herein, we provide a brief overview of GDF15 biology with a focus on energy homeostasis, and highlight the implications of the recent receptor identification to this field and beyond.
Collapse
Affiliation(s)
- Shannon E Mullican
- Cardiovascular & Metabolism Therapeutic Area, Janssen Pharmaceuticals, Inc., Spring House, PA 19477, USA
| | - Shamina M Rangwala
- Cardiovascular & Metabolism Therapeutic Area, Janssen Pharmaceuticals, Inc., Spring House, PA 19477, USA.
| |
Collapse
|
16
|
Williams CL, Smith SM. Calcium dependence of spontaneous neurotransmitter release. J Neurosci Res 2018; 96:335-347. [PMID: 28699241 PMCID: PMC5766384 DOI: 10.1002/jnr.24116] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2017] [Revised: 06/16/2017] [Accepted: 06/19/2017] [Indexed: 01/14/2023]
Abstract
Spontaneous release of neurotransmitters is regulated by extracellular [Ca2+ ] and intracellular [Ca2+ ]. Curiously, some of the mechanisms of Ca2+ signaling at central synapses are different at excitatory and inhibitory synapses. While the stochastic activity of voltage-activated Ca2+ channels triggers a majority of spontaneous release at inhibitory synapses, this is not the case at excitatory nerve terminals. Ca2+ release from intracellular stores regulates spontaneous release at excitatory and inhibitory terminals, as do agonists of the Ca2+ -sensing receptor. Molecular machinery triggering spontaneous vesicle fusion may differ from that underlying evoked release and may be one of the sources of heterogeneity in release mechanisms.
Collapse
Affiliation(s)
- Courtney L. Williams
- Department of Medicine, Division of Pulmonary & Critical Care Medicine, Oregon Health & Science University, Portland, Oregon, 97239, USA
- Section of Pulmonary & Critical Care Medicine, VA Portland Health Care System, Portland, Oregon, USA
| | - Stephen M. Smith
- Department of Medicine, Division of Pulmonary & Critical Care Medicine, Oregon Health & Science University, Portland, Oregon, 97239, USA
- Section of Pulmonary & Critical Care Medicine, VA Portland Health Care System, Portland, Oregon, USA
| |
Collapse
|
17
|
Yang L, Chang CC, Sun Z, Madsen D, Zhu H, Padkjær SB, Wu X, Huang T, Hultman K, Paulsen SJ, Wang J, Bugge A, Frantzen JB, Nørgaard P, Jeppesen JF, Yang Z, Secher A, Chen H, Li X, John LM, Shan B, He Z, Gao X, Su J, Hansen KT, Yang W, Jørgensen SB. GFRAL is the receptor for GDF15 and is required for the anti-obesity effects of the ligand. Nat Med 2017; 23:1158-1166. [PMID: 28846099 DOI: 10.1038/nm.4394] [Citation(s) in RCA: 464] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 08/03/2017] [Indexed: 01/12/2023]
Abstract
Growth differentiation factor 15 (GDF15; also known as MIC-1) is a divergent member of the TGF-β superfamily and is associated with body-weight regulation in humans and rodents. However, the cognate receptor of GDF15 is unknown. Here we show that GDF15 binds specifically to GDNF family receptor α-like (GFRAL) with high affinity, and that GFRAL requires association with the coreceptor RET to elicit intracellular signaling in response to GDF15 stimulation. We also found that GDF15-mediated reductions in food intake and body weight of mice with obesity were abolished in GFRAL-knockout mice. We further found that GFRAL expression was limited to hindbrain neurons and not present in peripheral tissues, which suggests that GDF15-GFRAL-mediated regulation of food intake is by a central mechanism. Lastly, given that GDF15 did not increase energy expenditure in treated mice with obesity, the anti-obesity actions of the cytokine are likely driven primarily by a reduction in food intake.
Collapse
Affiliation(s)
- Linda Yang
- Novo Nordisk Research Centre China, Novo Nordisk A/S, Beijing, China
| | - Chih-Chuan Chang
- Novo Nordisk Research Centre China, Novo Nordisk A/S, Beijing, China
| | - Zhe Sun
- Novo Nordisk Research Centre China, Novo Nordisk A/S, Beijing, China
| | | | - Haisun Zhu
- Novo Nordisk Research Centre China, Novo Nordisk A/S, Beijing, China
| | | | - Xiaoai Wu
- Novo Nordisk Research Centre China, Novo Nordisk A/S, Beijing, China
| | - Tao Huang
- Novo Nordisk Research Centre China, Novo Nordisk A/S, Beijing, China
| | | | | | - Jishu Wang
- Novo Nordisk Research Centre China, Novo Nordisk A/S, Beijing, China
| | - Anne Bugge
- Global Research, Novo Nordisk A/S, Maaloev, Denmark
| | | | - Per Nørgaard
- Global Research, Novo Nordisk A/S, Maaloev, Denmark
| | | | - Zhiru Yang
- Novo Nordisk Research Centre China, Novo Nordisk A/S, Beijing, China
| | - Anna Secher
- Global Research, Novo Nordisk A/S, Maaloev, Denmark
| | - Haibin Chen
- Novo Nordisk Research Centre China, Novo Nordisk A/S, Beijing, China
| | - Xun Li
- Novo Nordisk Research Centre China, Novo Nordisk A/S, Beijing, China
| | | | - Bing Shan
- Novo Nordisk Research Centre China, Novo Nordisk A/S, Beijing, China
| | - Zhenhua He
- Novo Nordisk Research Centre China, Novo Nordisk A/S, Beijing, China
| | - Xiang Gao
- Novo Nordisk Research Centre China, Novo Nordisk A/S, Beijing, China
| | - Jing Su
- Novo Nordisk Research Centre China, Novo Nordisk A/S, Beijing, China
| | | | - Wei Yang
- Novo Nordisk Research Centre China, Novo Nordisk A/S, Beijing, China
| | | |
Collapse
|