1
|
Zhang W, Sun M, Liu N, Li X, Sun J, Wang M. Curcumin ameliorates astrocyte inflammation through AXL in cuprizone-induced mice. Toxicol Appl Pharmacol 2025; 494:117170. [PMID: 39586379 DOI: 10.1016/j.taap.2024.117170] [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: 08/20/2024] [Revised: 10/30/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
Curcumin has gained global attention owning to its anti-inflammatory, antioxidant, anticancer, and antimicrobial activities. Curcumin has recently been shown to have well-documented effects on neuroinflammation in multiple sclerosis (MS). Astrocytes, the most widely distributed glial cells in the brain, have a significant influence on the regulation of neuroinflammation in MS. However, it is unknown how curcumin exerts neuroprotective effects in astrocytes. To elucidate the mechanism underlying the effects of curcumin on astrocytes, we explored the effect of curcumin on cuprizone (CPZ)-induced mice in vivo and on primary astrocytes in vitro. In this study, we observed that curcumin significantly ameliorated myelin loss and reduced astrocyte activation in the corpus callosum (CC) region in mice induced with CPZ, and in primary astrocytes stimulated with lipopolysaccharide (LPS). Meanwhile, our research indicated that curcumin may exert neuroprotective effects in CPZ-induced mice by downregulating astrocyte-mediated inflammation by AXL. This study provides new insights into possible targeted therapies for MS.
Collapse
Affiliation(s)
- Wenjing Zhang
- Department of Neurology, Lanzhou University Second Hospital, 730030 Lanzhou, China
| | - Mengjiao Sun
- Department of Neurology, Lanzhou University Second Hospital, 730030 Lanzhou, China
| | - Ning Liu
- Department of Neurology, Lanzhou University Second Hospital, 730030 Lanzhou, China
| | - Xiaoling Li
- Department of Neurology, Lanzhou University Second Hospital, 730030 Lanzhou, China
| | - Jing Sun
- Department of Neurology, Lanzhou University Second Hospital, 730030 Lanzhou, China
| | - Manxia Wang
- Department of Neurology, Lanzhou University Second Hospital, 730030 Lanzhou, China.
| |
Collapse
|
2
|
Bian P, Zhang H, Ye C, Luo C, Jiang H, Wang Y, Dong Y, Yang J, Zhang F, Wang X, Zhang Y, Jia Z, Lei Y. GAS6 as a potential target to alleviate neuroinflammation during Japanese encephalitis in mouse models. J Neuroinflammation 2024; 21:231. [PMID: 39300526 PMCID: PMC11411859 DOI: 10.1186/s12974-024-03225-1] [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: 03/19/2024] [Accepted: 09/05/2024] [Indexed: 09/22/2024] Open
Abstract
Viral encephalitis is characterized by inflammation of the brain parenchyma caused by a variety of viruses, among which the Japanese encephalitis (JE) virus (JEV) is a typical representative arbovirus. Neuronal death, neuroinflammation, and breakdown of the blood brain barrier (BBB) constitute vicious circles of JE progression. Currently, there is no effective therapy to prevent this damage. Growth arrest specific gene 6 (GAS6) is a secreted growth factor that binds to the TYRO3, AXL, and MERTK (TAM) family of receptor tyrosine kinases and has been demonstrated to participate in neuroprotection and suppression of inflammation in many central nervous system (CNS) diseases which has great potential for JE intervention. In this study, we found that GAS6 expression in the brain was decreased and was reversely correlated with viral load and neuronal loss. Mice with GAS6/TAM signalling deficiency showed higher mortality and accelerated neuroinflammation during peripheral JEV infection, accompanied by BBB breakdown. GAS6 directly promoted the expression of tight junction proteins in bEnd.3 cells and strengthened BBB integrity, partly via AXL. Mice administered GAS6 were more resistant to JEV infection due to increased BBB integrity, as well as decreased viral load and neuroinflammation. Thus, targeted GAS6 delivery may represent a strategy for the prevention and treatment of JE especially in patients with impaired BBB.
Collapse
Affiliation(s)
- Peiyu Bian
- Department of Geriatrics, Xijing Hospital, Air Force Medical University, Xi'an, 710027, China
| | - Haijun Zhang
- Xijing 986 Hospital, Air Force Medical University, Xi'an, 710054, China
| | - Chuantao Ye
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Chuanyu Luo
- Norinco General Hospital, Xi'an, 710065, China
| | - Hong Jiang
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China
| | - Yuan Wang
- Department of Microbiology, School of Preclinical Medicine, Air Force Medical University, Xi'an, 710032, China
| | - Yangchao Dong
- Department of Microbiology, School of Preclinical Medicine, Air Force Medical University, Xi'an, 710032, China
| | - Jing Yang
- Department of Microbiology, School of Preclinical Medicine, Air Force Medical University, Xi'an, 710032, China
| | - Fanglin Zhang
- Department of Microbiology, School of Preclinical Medicine, Air Force Medical University, Xi'an, 710032, China
| | - Xiaoming Wang
- Department of Geriatrics, Xijing Hospital, Air Force Medical University, Xi'an, 710027, China
| | - Ying Zhang
- Department of Infectious Diseases, Tangdu Hospital, Air Force Medical University, Xi'an, 710038, China.
| | - Zhansheng Jia
- Department of Infectious Diseases, Xi'an International Medical Center Hospital, Xi'an, 710100, China.
| | - Yingfeng Lei
- Department of Microbiology, School of Preclinical Medicine, Air Force Medical University, Xi'an, 710032, China.
| |
Collapse
|
3
|
Luo J, Lin S. Dietary vitamin K intake is associated with decreased neurofilament light chain among middle-aged and older adults from the NHANES. Front Nutr 2024; 11:1396707. [PMID: 39346641 PMCID: PMC11428379 DOI: 10.3389/fnut.2024.1396707] [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: 03/09/2024] [Accepted: 09/02/2024] [Indexed: 10/01/2024] Open
Abstract
Purpose Neurofilament-light chain (NfL) is associated with neurodegenerative diseases, which are increasingly prevalent with aging. Vitamin K has been shown a neuroprotective effect. Therefore, we aimed to explore the potential relationship between dietary vitamin K intake and serum NfL. Methods This study was conducted on the 2013-2014 cycles of the National Health and Nutrition Examination Survey, a multi-site population-based study of the US general population. Serum NfL level was measured using a highly sensitive immunoassay. Dietary vitamin K intake was estimated from two-day dietary recall interviews, and its relationship with NfL was determined using linear regression models. Results The study included a total of 1,533 participants with a median age of 46 years, comprising 801 women (52.2%) and 732 men (47.8%). The median dietary intake of vitamin K was 81.6 μg/d, and the median serum NfL was 12 pg./mL. After adjusting for potential confounding factors in the full model, individuals with higher dietary vitamin K intake had lower serum NfL levels (Q4 vs. Q1, β = -4.92, 95%CI: -7.66, -2.19, p = 0.002). A non-linear negative dose-response association is found between dietary vitamin K intake and serum NfL levels (P for non-linearity = 0.008); this association reaches a plateau when the dietary vitamin K intake is higher than 200 μg/d. According to the results of stratified analysis, the relationship between dietary vitamin K intake and serum NfL levels was stronger in the population of middle-aged and older adults. Conclusion The present study suggested a negative association between dietary vitamin K intake and serum NfL levels in the general US population, especially in middle-aged and older adults. This study might offer a novel nutritional idea for the primary prevention and mechanism exploration of neurodegenerative diseases.
Collapse
Affiliation(s)
- Jing Luo
- School of Rehabilitation, Jiangsu College of Nursing, Huai'an, Jiangsu, China
| | - Song Lin
- Department of Clinical Nutrition, The Affiliated Huaian No. 1 People’s Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| |
Collapse
|
4
|
Ramos RL, De Heredia MMB, Zhang Y, Stout RF, Tindi JO, Wu L, Schwartz GJ, Botbol YM, Sidoli S, Poojari A, Rakowski-Anderson T, Shafit-Zagardo B. Patient-specific mutation of Dync1h1 in mice causes brain and behavioral deficits. Neurobiol Dis 2024; 199:106594. [PMID: 39025270 DOI: 10.1016/j.nbd.2024.106594] [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: 02/29/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/20/2024] Open
Abstract
AIMS Cytoplasmic dynein heavy chain (DYNC1H1) is a multi-subunit protein complex that provides motor force for movement of cargo on microtubules and traffics them back to the soma. In humans, mutations along the DYNC1H1 gene result in intellectual disabilities, cognitive delays, and neurologic and motor deficits. The aim of the study was to generate a mouse model to a newly identified de novo heterozygous DYNC1H1 mutation, within a functional ATPase domain (c9052C > T(P3018S)), identified in a child with motor deficits, and intellectual disabilities. RESULTS P3018S heterozygous (HET) knockin mice are viable; homozygotes are lethal. Metabolic and EchoMRI™ testing show that HET mice have a higher metabolic rate, are more active, and have less body fat compared to wildtype mice. Neurobehavioral studies show that HET mice perform worse when traversing elevated balance beams, and on the negative geotaxis test. Immunofluorescent staining shows neuronal migration abnormalities in the dorsal and lateral neocortex with heterotopia in layer I. Neuron-subtype specific transcription factors CUX1 and CTGF identified neurons from layers II/III and VI respectively in cortical layer I, and abnormal pyramidal neurons with MAP2+ dendrites projecting downward from the pial surface. CONCLUSION The HET mice are a good model for the motor deficits seen in the child, and highlights the importance of cytoplasmic dynein in the maintenance of cortical function and dendritic orientation relative to the pial surface. Our results are discussed in the context of other dynein mutant mice and in relation to clinical presentation in humans with DYNC1H1 mutations.
Collapse
Affiliation(s)
- Raddy L Ramos
- Department of Biomedical Sciences, New York Institute of Technology, College of Osteopathic Medicine, Riland Academic Health Center, Room 26, Old Westbury, NY 11568, United States of America
| | | | - Yongwei Zhang
- Cancer Center, Albert Einstein College of Medicine, 1301 Morris Park Ave, Price Building, Rm 269, Bronx, NY 10461, United States of America.
| | - Randy F Stout
- Department of Biomedical Sciences, New York Institute of Technology, College of Osteopathic Medicine, Riland Academic Health Center, Room 22, Old Westbury, NY 11568, United States of America.
| | - Jaafar O Tindi
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Kennedy Center, Rm 501, 1410 Pelham Parkway S., Bronx, NY 10461, United States of America.
| | - Liching Wu
- Dept of Medicine, Albert Einstein College of Medicine, United States of America.
| | - Gary J Schwartz
- The Fleischer Institute for Diabetes and Metabolism, Albert Einstein College of Medicine, United States of America.
| | - Yair M Botbol
- Department of Pathology, Albert Einstein College of Medicine, 1300 Morris Park Ave., Forchheimer Building, Rm 520, Bronx, NY 10461, United States of America.
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein, United States of America.
| | - Ankita Poojari
- Thomas J. Long School of Pharmacy, University of the Pacific, Stockton, CA, United States of America.
| | - Tammy Rakowski-Anderson
- Institute for Animal Studies, Albert Einstein College of Medicine, Van Etten Building, Room 463, Bronx, NY 10461, United States of America.
| | - Bridget Shafit-Zagardo
- Department of Pathology, Albert Einstein College of Medicine, 1300 Morris Park Ave., Forchheimer Building 514, Bronx, NY 10461, United States of America.
| |
Collapse
|
5
|
Asadian N, Aprico A, Chen M, Yuen D, Johnston APR, Kilpatrick TJ, Binder MD. The therapeutic effect of GAS6 in remyelination is dependent upon Tyro3. Glia 2024; 72:1392-1401. [PMID: 38572807 DOI: 10.1002/glia.24534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 04/05/2024]
Abstract
Multiple sclerosis is an autoimmune disease of the central nervous system (CNS) characterized by demyelination, axonal damage and, for the majority of people, a decline in neurological function in the long-term. Remyelination could assist in the protection of axons and their functional recovery, but such therapies are not, as yet, available. The TAM (Tyro3, Axl, and MERTK) receptor ligand GAS6 potentiates myelination in vitro and promotes recovery in pre-clinical models of MS. However, it has remained unclear which TAM receptor is responsible for transducing this effect and whether post-translational modification of GAS6 is required. In this study, we show that the promotion of myelination requires post-translational modification of the GLA domain of GAS6 via vitamin K-dependent γ-carboxylation. We also confirmed that the intracerebroventricular provision of GAS6 for 2 weeks to demyelinated wild-type (WT) mice challenged with cuprizone increased the density of myelinated axons in the corpus callosum by over 2-fold compared with vehicle control. Conversely, the provision of GAS6 to Tyro3 KO mice did not significantly improve the density of myelinated axons. The improvement in remyelination following the provision of GAS6 to WT mice was also accompanied by an increased density of CC1+ve mature oligodendrocytes compared with vehicle control, whereas this improvement was not observed in the absence of Tyro3. This effect occurs independent of any influence on microglial activation. This work therefore establishes that the remyelinative activity of GAS6 is dependent on Tyro3 and includes potentiation of oligodendrocyte numbers.
Collapse
Affiliation(s)
- Negar Asadian
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Andrea Aprico
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Moore Chen
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Daniel Yuen
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Angus P R Johnston
- Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Trevor J Kilpatrick
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| | - Michele D Binder
- The Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia
| |
Collapse
|
6
|
Olveda GE, Barasa MN, Hill RA. Microglial phagocytosis of single dying oligodendrocytes is mediated by CX3CR1 but not MERTK. Cell Rep 2024; 43:114385. [PMID: 38935500 PMCID: PMC11304498 DOI: 10.1016/j.celrep.2024.114385] [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: 12/04/2023] [Revised: 04/10/2024] [Accepted: 06/05/2024] [Indexed: 06/29/2024] Open
Abstract
Oligodendrocyte death is common in aging and neurodegenerative disease. In these conditions, dying oligodendrocytes must be efficiently removed to allow remyelination and to prevent a feedforward degenerative cascade. Removal of this cellular debris is thought to primarily be carried out by resident microglia. To investigate the cellular dynamics underlying how microglia do this, we use a single-cell cortical demyelination model combined with longitudinal intravital imaging of dual-labeled transgenic mice. Following phagocytosis, single microglia clear the targeted oligodendrocyte and its myelin sheaths in one day via a precise, rapid, and stereotyped sequence. Deletion of the fractalkine receptor, CX3CR1, delays the microglial phagocytosis of the cell soma but has no effect on clearance of myelin sheaths. Unexpectedly, deletion of the phosphatidylserine receptor, MERTK, has no effect on oligodendrocyte or myelin sheath clearance. Thus, separate molecular signals are used to detect, engage, and clear distinct sub-compartments of dying oligodendrocytes to maintain tissue homeostasis.
Collapse
Affiliation(s)
- Genaro E Olveda
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA
| | - Maryanne N Barasa
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA
| | - Robert A Hill
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA.
| |
Collapse
|
7
|
Xing J, Wang K, Xu YC, Pei ZJ, Yu QX, Liu XY, Dong YL, Li SF, Chen Y, Zhao YJ, Yao F, Ding J, Hu W, Zhou RP. Efferocytosis: Unveiling its potential in autoimmune disease and treatment strategies. Autoimmun Rev 2024; 23:103578. [PMID: 39004157 DOI: 10.1016/j.autrev.2024.103578] [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: 01/26/2024] [Revised: 07/06/2024] [Accepted: 07/07/2024] [Indexed: 07/16/2024]
Abstract
Efferocytosis is a crucial process whereby phagocytes engulf and eliminate apoptotic cells (ACs). This intricate process can be categorized into four steps: (1) ACs release "find me" signals to attract phagocytes, (2) phagocytosis is directed by "eat me" signals emitted by ACs, (3) phagocytes engulf and internalize ACs, and (4) degradation of ACs occurs. Maintaining immune homeostasis heavily relies on the efficient clearance of ACs, which eliminates self-antigens and facilitates the generation of anti-inflammatory and immunosuppressive signals that maintain immune tolerance. However, any disruptions occurring at any of the efferocytosis steps during apoptosis can lead to a diminished efficacy in removing apoptotic cells. Factors contributing to this inefficiency encompass dysregulation in the release and recognition of "find me" or "eat me" signals, defects in phagocyte surface receptors, bridging molecules, and other signaling pathways. The inadequate clearance of ACs can result in their rupture and subsequent release of self-antigens, thereby promoting immune responses and precipitating the onset of autoimmune diseases such as systemic lupus erythematosus, rheumatoid arthritis, type 1 diabetes, and multiple sclerosis. A comprehensive understanding of the efferocytosis process and its implications can provide valuable insights for developing novel therapeutic strategies that target this process to prevent or treat autoimmune diseases.
Collapse
Affiliation(s)
- Jing Xing
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; School of pharmacy, Anhui Medical University, Hefei 230032, China
| | - Ke Wang
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Yu-Cai Xu
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; School of pharmacy, Anhui Medical University, Hefei 230032, China
| | - Ze-Jun Pei
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; School of pharmacy, Anhui Medical University, Hefei 230032, China
| | - Qiu-Xia Yu
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; School of pharmacy, Anhui Medical University, Hefei 230032, China
| | - Xing-Yu Liu
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; School of pharmacy, Anhui Medical University, Hefei 230032, China
| | - Ya-Lu Dong
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; School of pharmacy, Anhui Medical University, Hefei 230032, China
| | - Shu-Fang Li
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Yong Chen
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Ying-Jie Zhao
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Feng Yao
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Jie Ding
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Wei Hu
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China.
| | - Ren-Peng Zhou
- Department of Clinical Pharmacology, the Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; The Key Laboratory of Anti-inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei 230032, China.
| |
Collapse
|
8
|
D’Onghia D, Colangelo D, Bellan M, Tonello S, Puricelli C, Virgilio E, Apostolo D, Minisini R, Ferreira LL, Sozzi L, Vincenzi F, Cantello R, Comi C, Pirisi M, Vecchio D, Sainaghi PP. Gas6/TAM system as potential biomarker for multiple sclerosis prognosis. Front Immunol 2024; 15:1362960. [PMID: 38745659 PMCID: PMC11091300 DOI: 10.3389/fimmu.2024.1362960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 04/03/2024] [Indexed: 05/16/2024] Open
Abstract
Introduction The protein growth arrest-specific 6 (Gas6) and its tyrosine kinase receptors Tyro-3, Axl, and Mer (TAM) are ubiquitous proteins involved in regulating inflammation and apoptotic body clearance. Multiple sclerosis (MS) is the most common inflammatory demyelinating disease of the central nervous system leading to progressive and irreversible disability if not diagnosed and treated promptly. Gas6 and TAM receptors have been associated with neuronal remyelination and stimulation of oligodendrocyte survival. However, few data are available regarding clinical correlation in MS patients. We aimed to evaluate soluble levels of these molecules in the cerebrospinal fluid (CSF) and serum at MS diagnosis and correlate them with short-term disease severity. Methods In a prospective cohort study, we enrolled 64 patients with a diagnosis of clinical isolated syndrome (CIS), radiological isolated syndrome (RIS) and relapsing-remitting (RR) MS according to the McDonald 2017 Criteria. Before any treatment initiation, we sampled the serum and CSF, and collected clinical data: disease course, presence of gadolinium-enhancing lesions, and expanded disability status score (EDSS). At the last clinical follow-up, we assessed EDSS and calculated MS severity score (MSSS) and age-related MS severity (ARMSS). Gas6 and TAM receptors were determined using an ELISA kit (R&D Systems) and compared to neurofilament (NFLs) levels evaluated with SimplePlex™ fluorescence-based immunoassay. Results At diagnosis, serum sAxl was higher in patients receiving none or low-efficacy disease-modifying treatments (DMTs) versus patients with high-efficacy DMTs (p = 0.04). Higher CSF Gas6 and serum sAXL were associated with an EDSS <3 at diagnosis (p = 0.04; p = 0.037). Serum Gas6 correlates to a lower MSSS (r2 = -0.32, p = 0.01). Serum and CSF NFLs were confirmed as disability biomarkers in our cohort according to EDSS (p = 0.005; p = 0.002) and MSSS (r2 = 0.27, p = 0.03; r2 = 0.39, p = 0.001). Results were corroborated using multivariate analysis. Conclusions Our data suggest a protective role of Gas6 and its receptors in patients with MS and suitable severity disease biomarkers.
Collapse
Affiliation(s)
- Davide D’Onghia
- Department of Translational Medicine, University of Piemonte Orientale (UPO), Novara, Italy
- Center for Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale (UPO), Novara, Italy
| | - Donato Colangelo
- Department of Health Sciences, Pharmacology, University of Piemonte Orientale (UPO), Novara, Italy
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale (UPO), Novara, Italy
| | - Mattia Bellan
- Department of Translational Medicine, University of Piemonte Orientale (UPO), Novara, Italy
- Center for Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale (UPO), Novara, Italy
- Internal Medicine and Rheumatology Unit, Azienda Ospedaliera Universitaria (AOU) “Maggiore della Carita”, Novara, Italy
| | - Stelvio Tonello
- Department of Translational Medicine, University of Piemonte Orientale (UPO), Novara, Italy
- Center for Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale (UPO), Novara, Italy
| | - Chiara Puricelli
- Department of Health Sciences, Clinical Biochemistry, University of Piemonte Orientale (UPO), Novara, Italy
| | - Eleonora Virgilio
- Department of Translational Medicine, Neurology Unit, Maggiore Della Carità Hospital, University of Piemonte Orientale, Novara, Italy
| | - Daria Apostolo
- Department of Translational Medicine, University of Piemonte Orientale (UPO), Novara, Italy
| | - Rosalba Minisini
- Department of Translational Medicine, University of Piemonte Orientale (UPO), Novara, Italy
| | - Luciana L. Ferreira
- Department of Translational Medicine, University of Piemonte Orientale (UPO), Novara, Italy
| | - Leonardo Sozzi
- Department of Translational Medicine, University of Piemonte Orientale (UPO), Novara, Italy
| | - Federica Vincenzi
- Department of Translational Medicine, University of Piemonte Orientale (UPO), Novara, Italy
| | - Roberto Cantello
- Department of Translational Medicine, Neurology Unit, Maggiore Della Carità Hospital, University of Piemonte Orientale, Novara, Italy
| | - Cristoforo Comi
- Department of Translational Medicine, Neurology Unit, S. Andrea Hospital, University of Piemonte Orientale (UPO), Vercelli, Italy
| | - Mario Pirisi
- Department of Translational Medicine, University of Piemonte Orientale (UPO), Novara, Italy
- Center for Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale (UPO), Novara, Italy
- Internal Medicine and Rheumatology Unit, Azienda Ospedaliera Universitaria (AOU) “Maggiore della Carita”, Novara, Italy
| | - Domizia Vecchio
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale (UPO), Novara, Italy
- Department of Translational Medicine, Neurology Unit, Maggiore Della Carità Hospital, University of Piemonte Orientale, Novara, Italy
| | - Pier Paolo Sainaghi
- Department of Translational Medicine, University of Piemonte Orientale (UPO), Novara, Italy
- Center for Autoimmune and Allergic Diseases (CAAD), University of Piemonte Orientale (UPO), Novara, Italy
- Department of Health Sciences, Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale (UPO), Novara, Italy
- Internal Medicine and Rheumatology Unit, Azienda Ospedaliera Universitaria (AOU) “Maggiore della Carita”, Novara, Italy
| |
Collapse
|
9
|
Lee YJ, Kim M, Kim HS, Kang JL. Administration of Gas6 attenuates lung fibrosis via inhibition of the epithelial-mesenchymal transition and fibroblast activation. Cell Biol Toxicol 2024; 40:20. [PMID: 38578518 PMCID: PMC10997547 DOI: 10.1007/s10565-024-09858-5] [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: 12/01/2023] [Accepted: 03/25/2024] [Indexed: 04/06/2024]
Abstract
The epithelial-mesenchymal transition (EMT) and fibroblast activation are major events in idiopathic pulmonary fibrosis pathogenesis. Here, we investigated whether growth arrest-specific protein 6 (Gas6) plays a protective role in lung fibrosis via suppression of the EMT and fibroblast activation. rGas6 administration inhibited the EMT in isolated mouse ATII cells 14 days post-BLM treatment based on morphologic cellular alterations, changes in mRNA and protein expression profiles of EMT markers, and induction of EMT-activating transcription factors. BLM-induced increases in gene expression of fibroblast activation-related markers and the invasive capacity of primary lung fibroblasts in primary lung fibroblasts were reversed by rGas6 administration. Furthermore, the hydroxyproline content and collagen accumulation in interstitial areas with damaged alveolar structures in lung tissue were reduced by rGas6 administration. Targeting Gas6/Axl signaling events with specific inhibitors of Axl (BGB324), COX-2 (NS-398), EP1/EP2 receptor (AH-6809), or PGD2 DP2 receptor (BAY-u3405) reversed the inhibitory effects of rGas6 on EMT and fibroblast activation. Finally, we confirmed the antifibrotic effects of Gas6 using Gas6-/- mice. Therefore, Gas6/Axl signaling events play a potential role in inhibition of EMT process and fibroblast activation via COX-2-derived PGE2 and PGD2 production, ultimately preventing the development of pulmonary fibrosis.
Collapse
Affiliation(s)
- Ye-Ji Lee
- Department of Physiology, College of Medicine, Ewha Womans University, 25 Magokdong-Ro 2-Gil, Gangseo-Gu, Seoul, 07804, Korea
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, 25 Magokdong-Ro 2-Gil, Gangseo-Gu, Seoul, 07804, Korea
| | - Minsuk Kim
- Department of Pharmacology, College of Medicine, Ewha Womans University, 25 Magokdong-Ro 2-Gil, Gangseo-Gu, Seoul, 07804, Korea
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, 25 Magokdong-Ro 2-Gil, Gangseo-Gu, Seoul, 07804, Korea
| | - Hee-Sun Kim
- Department of Molecular Medicine, College of Medicine, Ewha Womans University, 25 Magokdong-Ro 2-Gil, Gangseo-Gu, Seoul, 07804, Korea
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, 25 Magokdong-Ro 2-Gil, Gangseo-Gu, Seoul, 07804, Korea
| | - Jihee Lee Kang
- Department of Physiology, College of Medicine, Ewha Womans University, 25 Magokdong-Ro 2-Gil, Gangseo-Gu, Seoul, 07804, Korea.
- Inflammation-Cancer Microenvironment Research Center, College of Medicine, Ewha Womans University, 25 Magokdong-Ro 2-Gil, Gangseo-Gu, Seoul, 07804, Korea.
| |
Collapse
|
10
|
Binder MD, Nwoke EC, Morwitch E, Dwyer C, Li V, Xavier A, Lea RA, Lechner-Scott J, Taylor BV, Ponsonby AL, Kilpatrick TJ. HLA-DRB1*15:01 and the MERTK Gene Interact to Selectively Influence the Profile of MERTK-Expressing Monocytes in Both Health and MS. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200190. [PMID: 38150649 PMCID: PMC10752576 DOI: 10.1212/nxi.0000000000200190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 10/31/2023] [Indexed: 12/29/2023]
Abstract
BACKGROUND AND OBJECTIVES HLA-DRB1*15:01 (DR15) and MERTK are 2 risk genes for multiple sclerosis (MS). The variant rs7422195 is an expression quantitative trait locus for MERTK in CD14+ monocytes; cells with phagocytic and immunomodulatory potential. We aimed to understand how drivers of disease risk and pathogenesis vary with HLA and MERTK genotype and disease activity. METHODS We investigated how proportions of monocytes vary with HLA and MERTK genotype and disease activity in MS. CD14+ monocytes were isolated from patients with MS at relapse (n = 40) and 3 months later (n = 23). Healthy controls (HCs) underwent 2 blood collections 3 months apart. Immunophenotypic profiling of monocytes was performed by flow cytometry. Methylation of 35 CpG sites within and near the MERTK gene was assessed in whole blood samples of individuals experiencing their first episode of clinical CNS demyelination (n = 204) and matched HCs (n = 345) using an Illumina EPIC array. RESULTS DR15-positive patients had lower proportions of CD14+ MERTK+ monocytes than DR15-negative patients, independent of genotype at the MERTK SNP rs7422195. Proportions of CD14+ MERTK+ monocytes were further reduced during relapse in DR15-positive but not DR15-negative patients. Patients homozygous for the major G allele at rs7422195 exhibited higher proportions of CD14+ MERTK+ monocytes at both relapse and remission compared with controls. We observed that increased methylation of the MERTK gene was significantly associated with the presence of DR15. DISCUSSION DR15 and MERTK genotype independently influence proportions of CD14+ MERTK+ monocytes in MS. We confirmed previous observations that the MERTK risk SNP rs7422195 is associated with altered MERTK expression in monocytes. We identified that expression of MERTK is stratified by disease in people homozygous for the major G allele of rs7422195. The finding that the proportion of CD14+ MERTK+ monocytes is reduced in DR15-positive individuals supports prior data identifying genetic links between these 2 loci in influencing MS risk. DR15 genotype-dependent alterations in methylation of the MERTK gene provides a molecular link between these loci and identifies a potential mechanism by which MERTK expression is influenced by DR15. This links DR15 haplotype to MS susceptibility beyond direct influence on antigen presentation and suggests the need for HLA-based stratification of approaches to MERTK as a therapeutic target.
Collapse
Affiliation(s)
- Michele D Binder
- From the Florey Institute of Neuroscience and Mental Health (M.D.B., E.C.N., E.M., C.D., V.L., A.-L.P., T.J.K.); Department of Anatomy and Physiology (M.D.B.), University of Melbourne, Parkville; Crux Biolabs (E.C.N.), Bayswater; Department of Neurology (C.D.), Royal Melbourne Hospital, Parkville; Department of Neurology (A.X., J.L.-S.), John Hunter Hospital, Newcastle; Hunter Medical Research Institute (A.X., J.L.-S.), University of Newcastle, New South Wales Genomics Research Centre (R.A.L.), Centre of Genomics and Personalised Health, Queensland University of Technology; and Menzies Institute for Medical Research (B.V.T.), University of Tasmania, Hobart, Australia
| | - Eze C Nwoke
- From the Florey Institute of Neuroscience and Mental Health (M.D.B., E.C.N., E.M., C.D., V.L., A.-L.P., T.J.K.); Department of Anatomy and Physiology (M.D.B.), University of Melbourne, Parkville; Crux Biolabs (E.C.N.), Bayswater; Department of Neurology (C.D.), Royal Melbourne Hospital, Parkville; Department of Neurology (A.X., J.L.-S.), John Hunter Hospital, Newcastle; Hunter Medical Research Institute (A.X., J.L.-S.), University of Newcastle, New South Wales Genomics Research Centre (R.A.L.), Centre of Genomics and Personalised Health, Queensland University of Technology; and Menzies Institute for Medical Research (B.V.T.), University of Tasmania, Hobart, Australia
| | - Ellen Morwitch
- From the Florey Institute of Neuroscience and Mental Health (M.D.B., E.C.N., E.M., C.D., V.L., A.-L.P., T.J.K.); Department of Anatomy and Physiology (M.D.B.), University of Melbourne, Parkville; Crux Biolabs (E.C.N.), Bayswater; Department of Neurology (C.D.), Royal Melbourne Hospital, Parkville; Department of Neurology (A.X., J.L.-S.), John Hunter Hospital, Newcastle; Hunter Medical Research Institute (A.X., J.L.-S.), University of Newcastle, New South Wales Genomics Research Centre (R.A.L.), Centre of Genomics and Personalised Health, Queensland University of Technology; and Menzies Institute for Medical Research (B.V.T.), University of Tasmania, Hobart, Australia
| | - Chris Dwyer
- From the Florey Institute of Neuroscience and Mental Health (M.D.B., E.C.N., E.M., C.D., V.L., A.-L.P., T.J.K.); Department of Anatomy and Physiology (M.D.B.), University of Melbourne, Parkville; Crux Biolabs (E.C.N.), Bayswater; Department of Neurology (C.D.), Royal Melbourne Hospital, Parkville; Department of Neurology (A.X., J.L.-S.), John Hunter Hospital, Newcastle; Hunter Medical Research Institute (A.X., J.L.-S.), University of Newcastle, New South Wales Genomics Research Centre (R.A.L.), Centre of Genomics and Personalised Health, Queensland University of Technology; and Menzies Institute for Medical Research (B.V.T.), University of Tasmania, Hobart, Australia
| | - Vivien Li
- From the Florey Institute of Neuroscience and Mental Health (M.D.B., E.C.N., E.M., C.D., V.L., A.-L.P., T.J.K.); Department of Anatomy and Physiology (M.D.B.), University of Melbourne, Parkville; Crux Biolabs (E.C.N.), Bayswater; Department of Neurology (C.D.), Royal Melbourne Hospital, Parkville; Department of Neurology (A.X., J.L.-S.), John Hunter Hospital, Newcastle; Hunter Medical Research Institute (A.X., J.L.-S.), University of Newcastle, New South Wales Genomics Research Centre (R.A.L.), Centre of Genomics and Personalised Health, Queensland University of Technology; and Menzies Institute for Medical Research (B.V.T.), University of Tasmania, Hobart, Australia
| | - Alexandre Xavier
- From the Florey Institute of Neuroscience and Mental Health (M.D.B., E.C.N., E.M., C.D., V.L., A.-L.P., T.J.K.); Department of Anatomy and Physiology (M.D.B.), University of Melbourne, Parkville; Crux Biolabs (E.C.N.), Bayswater; Department of Neurology (C.D.), Royal Melbourne Hospital, Parkville; Department of Neurology (A.X., J.L.-S.), John Hunter Hospital, Newcastle; Hunter Medical Research Institute (A.X., J.L.-S.), University of Newcastle, New South Wales Genomics Research Centre (R.A.L.), Centre of Genomics and Personalised Health, Queensland University of Technology; and Menzies Institute for Medical Research (B.V.T.), University of Tasmania, Hobart, Australia
| | - Rodney A Lea
- From the Florey Institute of Neuroscience and Mental Health (M.D.B., E.C.N., E.M., C.D., V.L., A.-L.P., T.J.K.); Department of Anatomy and Physiology (M.D.B.), University of Melbourne, Parkville; Crux Biolabs (E.C.N.), Bayswater; Department of Neurology (C.D.), Royal Melbourne Hospital, Parkville; Department of Neurology (A.X., J.L.-S.), John Hunter Hospital, Newcastle; Hunter Medical Research Institute (A.X., J.L.-S.), University of Newcastle, New South Wales Genomics Research Centre (R.A.L.), Centre of Genomics and Personalised Health, Queensland University of Technology; and Menzies Institute for Medical Research (B.V.T.), University of Tasmania, Hobart, Australia
| | - Jeannette Lechner-Scott
- From the Florey Institute of Neuroscience and Mental Health (M.D.B., E.C.N., E.M., C.D., V.L., A.-L.P., T.J.K.); Department of Anatomy and Physiology (M.D.B.), University of Melbourne, Parkville; Crux Biolabs (E.C.N.), Bayswater; Department of Neurology (C.D.), Royal Melbourne Hospital, Parkville; Department of Neurology (A.X., J.L.-S.), John Hunter Hospital, Newcastle; Hunter Medical Research Institute (A.X., J.L.-S.), University of Newcastle, New South Wales Genomics Research Centre (R.A.L.), Centre of Genomics and Personalised Health, Queensland University of Technology; and Menzies Institute for Medical Research (B.V.T.), University of Tasmania, Hobart, Australia
| | - Bruce V Taylor
- From the Florey Institute of Neuroscience and Mental Health (M.D.B., E.C.N., E.M., C.D., V.L., A.-L.P., T.J.K.); Department of Anatomy and Physiology (M.D.B.), University of Melbourne, Parkville; Crux Biolabs (E.C.N.), Bayswater; Department of Neurology (C.D.), Royal Melbourne Hospital, Parkville; Department of Neurology (A.X., J.L.-S.), John Hunter Hospital, Newcastle; Hunter Medical Research Institute (A.X., J.L.-S.), University of Newcastle, New South Wales Genomics Research Centre (R.A.L.), Centre of Genomics and Personalised Health, Queensland University of Technology; and Menzies Institute for Medical Research (B.V.T.), University of Tasmania, Hobart, Australia
| | - Anne-Louise Ponsonby
- From the Florey Institute of Neuroscience and Mental Health (M.D.B., E.C.N., E.M., C.D., V.L., A.-L.P., T.J.K.); Department of Anatomy and Physiology (M.D.B.), University of Melbourne, Parkville; Crux Biolabs (E.C.N.), Bayswater; Department of Neurology (C.D.), Royal Melbourne Hospital, Parkville; Department of Neurology (A.X., J.L.-S.), John Hunter Hospital, Newcastle; Hunter Medical Research Institute (A.X., J.L.-S.), University of Newcastle, New South Wales Genomics Research Centre (R.A.L.), Centre of Genomics and Personalised Health, Queensland University of Technology; and Menzies Institute for Medical Research (B.V.T.), University of Tasmania, Hobart, Australia
| | - Trevor J Kilpatrick
- From the Florey Institute of Neuroscience and Mental Health (M.D.B., E.C.N., E.M., C.D., V.L., A.-L.P., T.J.K.); Department of Anatomy and Physiology (M.D.B.), University of Melbourne, Parkville; Crux Biolabs (E.C.N.), Bayswater; Department of Neurology (C.D.), Royal Melbourne Hospital, Parkville; Department of Neurology (A.X., J.L.-S.), John Hunter Hospital, Newcastle; Hunter Medical Research Institute (A.X., J.L.-S.), University of Newcastle, New South Wales Genomics Research Centre (R.A.L.), Centre of Genomics and Personalised Health, Queensland University of Technology; and Menzies Institute for Medical Research (B.V.T.), University of Tasmania, Hobart, Australia
| |
Collapse
|
11
|
Jia J, Xu S, Hu J, Gan Y, Sun M, Xia S, Bao X, Zhang M, Xu Y. Growth arrest specific protein 6 alleviated white matter injury after experimental ischemic stroke. J Cereb Blood Flow Metab 2024; 44:77-93. [PMID: 37794790 PMCID: PMC10905636 DOI: 10.1177/0271678x231205078] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 09/09/2023] [Accepted: 09/11/2023] [Indexed: 10/06/2023]
Abstract
Ischemic white matter injury leads to long-term neurological deficits and lacks effective medication. Growth arrest specific protein 6 (Gas6) clears myelin debris, which is hypothesized to promote white matter integrity in experimental stroke models. By the middle cerebral artery occlusion (MCAO) stroke model, we observed that Gas6 reduced infarcted volume and behavior deficits 4 weeks after MCAO. Compared with control mice, Gas6-treatment mice represented higher FA values in the ipsilateral external capsules by MRI DTI scan. The SMI32/MBP ratio of the ipsilateral cortex and striatum was profoundly alleviated by Gas6 administration. Gas6-treatment group manifested thicker myelin sheaths than the control group by electron microscopy. We observed that Gas6 mainly promoted OPC maturation, which was closely related to microglia. Mechanically, Gas6 accelerated microglia-mediated myelin debris clearance and cholesterol transport protein expression (abca1, abcg1, apoc1, apoe) in vivo and in vitro, accordingly less myelin debris and lipid deposited in Gas6 treated stroke mice. HX531 (RXR inhibitor) administration mitigated the functions of Gas6 in speeding up debris clearance and cholesterol transport protein expression. Generally, we concluded that Gas6 cleared myelin debris and promoted cholesterol transportation protein expression through activating RXR, which could be one critical mechanism contributing to white matter repair after stroke.
Collapse
Affiliation(s)
- Junqiu Jia
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Siyi Xu
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Zhenjiang, China
| | - Jinglong Hu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yonghui Gan
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Min Sun
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Shengnan Xia
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xinyu Bao
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Meijuan Zhang
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China
| | - Yun Xu
- Department of Neurology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Department of Neurology, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Zhenjiang, China
- Department of Neurology, Nanjing Drum Tower Hospital, State Key Laboratory of Pharmaceutical Biotechnology and Institute of Translational Medicine for Brain Critical Diseases, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory for Molecular Medicine, Medical School of Nanjing University, Nanjing, China
- Jiangsu Province Stroke Center for Diagnosis and Therapy, Nanjing, China
- Nanjing Neuropsychiatry Clinic Medical Center, Nanjing, China
- Jiangsu Provincial medical key discipline, Nanjing, China
| |
Collapse
|
12
|
Olveda GE, Barasa MN, Hill RA. Microglial phagocytosis of single dying oligodendrocytes is mediated by CX3CR1 but not MERTK. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.12.11.570620. [PMID: 38168326 PMCID: PMC10760041 DOI: 10.1101/2023.12.11.570620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Oligodendrocyte death is common in aging and neurodegenerative diseases. In these conditions, single dying oligodendrocytes must be efficiently removed to allow remyelination and prevent a feed-forward degenerative cascade. Here we used a single-cell cortical demyelination model combined with longitudinal intravital imaging of dual-labeled transgenic mice to investigate the cellular dynamics underlying how brain resident microglia remove these cellular debris. Following phagocytic engagement, single microglia cleared the targeted oligodendrocyte and its myelin sheaths in one day via a precise, rapid, and stereotyped sequence. Deletion of the fractalkine receptor, CX3CR1, delayed microglia engagement with the cell soma but unexpectedly did not affect the clearance of myelin sheaths. Furthermore, and in contrast to previous reports in other demyelination models, deletion of the phosphatidylserine receptor, MERTK, did not affect oligodendrocyte or myelin sheath clearance. Thus, distinct molecular signals are used to detect, engage, and clear sub-compartments of dying oligodendrocytes to maintain tissue homeostasis.
Collapse
Affiliation(s)
- Genaro E. Olveda
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA
| | - Maryanne N. Barasa
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA
| | - Robert A. Hill
- Department of Biological Sciences, Dartmouth College, Hanover, NH 03755, USA
| |
Collapse
|
13
|
Tang J, Jin Y, Jia F, Lv T, Manaenko A, Zhang LF, Zhang Z, Qi X, Xue Y, Zhao B, Zhang X, Zhang JH, Lu J, Hu Q. Gas6 Promotes Microglia Efferocytosis and Suppresses Inflammation Through Activating Axl/Rac1 Signaling in Subarachnoid Hemorrhage Mice. Transl Stroke Res 2023; 14:955-969. [PMID: 36324028 DOI: 10.1007/s12975-022-01099-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/17/2022] [Accepted: 10/19/2022] [Indexed: 11/05/2022]
Abstract
Early brain injury (EBI) following subarachnoid hemorrhage (SAH) is characterized by rapid development of neuron apoptosis and dysregulated inflammatory response. Microglia efferocytosis plays a critical role in the clearance of apoptotic cells, attenuation of inflammation, and minimizing brain injury in various pathological conditions. Here, using a mouse SAH model, we aim to investigate whether microglia efferocytosis is involved in post-SAH inflammation and to determine the underlying signaling pathway. We hypothesized that TAM receptors and their ligands regulate this process. To prove our hypothesis, the expression and cellular location of TAM (Tyro3, Axl, and Mertk) receptors and their ligands growth arrest-specific 6 (Gas6) and Protein S (ProS1) were examined by PCR, western blots, and fluorescence immunostaining. Thirty minutes after SAH, mice received an intraventricular injection of recombinant Gas6 (rGas6) or recombinant ProS1 (rPros1) and underwent evaluations of inflammatory mediator expression, neurological deficits, and blood-brain barrier integrity at 24 h. Microglia efferocytosis of apoptotic neurons was analyzed in vivo and in vitro. The potential mechanism was determined by inhibiting or knocking down TAM receptors and Rac1 by specific inhibitors or siRNA. SAH induced upregulation of Axl and its ligand Gas6. The administration of rGas6 but not rPros1 promoted microglia efferocytosis, alleviated inflammation, and ameliorated SAH-induced BBB breakdown and neurological deficits. The beneficial effects of rGas6 were arrogated by inhibiting or knocking down Axl and Rac1. We concluded that rGas6 attenuated the development of early brain injury in mice after SAH by facilitating microglia efferocytosis and preventing inflammatory response, which is partly dependent on activation of Axl and Rac1.
Collapse
Affiliation(s)
- Junjia Tang
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
- Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Yichao Jin
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Feng Jia
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Tao Lv
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Anatol Manaenko
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lin-Feng Zhang
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Zeyu Zhang
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Xin Qi
- Discipline of Neuroscience, Department of Physiology and Anatomy, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yajun Xue
- Department of Neurosurgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Bin Zhao
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Xiaohua Zhang
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Jianfei Lu
- Discipline of Neuroscience, Department of Physiology and Anatomy, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Songjiang Institute and Songjiang Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China.
| | - Qin Hu
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| |
Collapse
|
14
|
Nguyen LT, Aprico A, Nwoke E, Walsh AD, Blades F, Avneri R, Martin E, Zalc B, Kilpatrick TJ, Binder MD. Mertk-expressing microglia influence oligodendrogenesis and myelin modelling in the CNS. J Neuroinflammation 2023; 20:253. [PMID: 37926818 PMCID: PMC10626688 DOI: 10.1186/s12974-023-02921-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/04/2023] [Indexed: 11/07/2023] Open
Abstract
BACKGROUND Microglia, an immune cell found exclusively within the CNS, initially develop from haematopoietic stem cell precursors in the yolk sac and colonise all regions of the CNS early in development. Microglia have been demonstrated to play an important role in the development of oligodendrocytes, the myelin producing cells in the CNS, as well as in myelination. Mertk is a receptor expressed on microglia that mediates immunoregulatory functions, including myelin efferocytosis. FINDINGS Here we demonstrate an unexpected role for Mertk-expressing microglia in both oligodendrogenesis and myelination. The selective depletion of Mertk from microglia resulted in reduced oligodendrocyte production in early development and the generation of pathological myelin. During demyelination, mice deficient in microglial Mertk had thinner myelin and showed signs of impaired OPC differentiation. We established that Mertk signalling inhibition impairs oligodendrocyte repopulation in Xenopus tadpoles following demyelination. CONCLUSION These data highlight the importance of microglia in myelination and are the first to identify Mertk as a regulator of oligodendrogenesis and myelin ultrastructure.
Collapse
Affiliation(s)
- Linda T Nguyen
- The Florey Institute of Neuroscience and Mental Health, Parkville, Melbourne, Australia
- Department of Anatomy and Physiology, University of Melbourne, Parkville, Melbourne, Australia
| | - Andrea Aprico
- The Florey Institute of Neuroscience and Mental Health, Parkville, Melbourne, Australia
| | - Eze Nwoke
- The Florey Institute of Neuroscience and Mental Health, Parkville, Melbourne, Australia
- Crux Biolabs, Bayswater, VIC, 3153, Australia
| | - Alexander D Walsh
- The Florey Institute of Neuroscience and Mental Health, Parkville, Melbourne, Australia
- Cognitive Neuroepigenetics Laboratory, Queensland Brain Institute, The University of Queensland, Brisbane, QLD, Australia
| | - Farrah Blades
- The Florey Institute of Neuroscience and Mental Health, Parkville, Melbourne, Australia
- Centre for Solar Biotechnology, Institute for Molecular Biosciences, University of Queensland, St Lucia, Brisbane, Australia
| | - Raphael Avneri
- Inserm, CNRS, Institut du Cerveau, AP-HP Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France
- Department of Molecular Biology, Ariel University, 40700, Ariel, Israel
| | - Elodie Martin
- Inserm, CNRS, Institut du Cerveau, AP-HP Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France
| | - Bernard Zalc
- Inserm, CNRS, Institut du Cerveau, AP-HP Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France
| | - Trevor J Kilpatrick
- The Florey Institute of Neuroscience and Mental Health, Parkville, Melbourne, Australia
| | - Michele D Binder
- The Florey Institute of Neuroscience and Mental Health, Parkville, Melbourne, Australia.
- Department of Anatomy and Physiology, University of Melbourne, Parkville, Melbourne, Australia.
| |
Collapse
|
15
|
Shafit-Zagardo B, Sidoli S, Goldman JE, DuBois JC, Corboy JR, Strittmatter SM, Guzik H, Edema U, Arackal AG, Botbol YM, Merheb E, Nagra RM, Graff S. TMEM106B Puncta Is Increased in Multiple Sclerosis Plaques, and Reduced Protein in Mice Results in Delayed Lipid Clearance Following CNS Injury. Cells 2023; 12:1734. [PMID: 37443768 PMCID: PMC10340176 DOI: 10.3390/cells12131734] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 06/12/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
During inflammatory, demyelinating diseases such as multiple sclerosis (MS), inflammation and axonal damage are prevalent early in the course. Axonal damage includes swelling, defects in transport, and failure to clear damaged intracellular proteins, all of which affect recovery and compromise neuronal integrity. The clearance of damaged cell components is important to maintain normal turnover and restore homeostasis. In this study, we used mass spectrometry to identify insoluble proteins within high-speed/mercaptoethanol/sarcosyl-insoluble pellets from purified white matter plaques isolated from the brains of individuals with relapsing-remitting MS (RRMS). We determined that the transmembrane protein 106B (TMEM106B), normally lysosome-associated, is insoluble in RRMS plaques relative to normal-appearing white matter from individuals with Alzheimer's disease and non-neurologic controls. Relative to wild-type mice, hypomorphic mice with a reduction in TMEM106B have increased axonal damage and lipid droplet accumulation in the spinal cord following myelin-oligodendrocyte-glycoprotein-induced experimental autoimmune encephalomyelitis. Additionally, the corpora callosa from cuprizone-challenged hypomorphic mice fail to clear lipid droplets efficiently during remyelination, suggesting that when TMEM106B is compromised, protein and lipid clearance by the lysosome is delayed. As TMEM106B contains putative lipid- and LC3-binding sites, further exploration of these sites is warranted.
Collapse
Affiliation(s)
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - James E Goldman
- Department of Pathology and Cell Biology, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA
| | - Juwen C DuBois
- Department of Pathology, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - John R Corboy
- Rocky Mountain MS Brain Bank, Department of Neurology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Stephen M Strittmatter
- Departments of Neurology and Neuroscience, Yale School of Medicine, Boyer Center for Molecular Medicine, New Haven, CT 06510, USA
| | - Hillary Guzik
- Analytic Imaging Facility, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Ukuemi Edema
- Department of Anatomic and Clinical Pathology, Montefiore Medical Center, Bronx, NY 10467, USA
| | - Anita G Arackal
- Department of Anatomic and Clinical Pathology, Montefiore Medical Center, Bronx, NY 10467, USA
| | - Yair M Botbol
- Department of Pathology, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Emilio Merheb
- Department of Biochemistry, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Rashed M Nagra
- UCLA Brain Bank, VA Healthcare System, Los Angeles, CA 90073, USA
| | - Sarah Graff
- Department of Biochemistry, Albert Einstein College of Medicine, New York, NY 10461, USA
| |
Collapse
|
16
|
Zhou S, Li Y, Zhang Z, Yuan Y. An insight into the TAM system in Alzheimer's disease. Int Immunopharmacol 2023; 116:109791. [PMID: 36738678 DOI: 10.1016/j.intimp.2023.109791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/13/2023] [Accepted: 01/24/2023] [Indexed: 02/05/2023]
Abstract
The TAM receptors may help delay the progression of Alzheimer's disease (AD). AD is the most common neurodegenerative disease associated with human aging. The TAM receptors, derived from the first letter of its three constituents -Tyro3, Axl, and Mertk, are associated with immune responses, cellular differentiation and migration, and clearance of apoptotic cells and debris, with the two canonical ligands, Growth Arrest Specific 6 (Gas6) and ProS1. Several kinds of research have indicated the participation of the TAM system in AD pathology. Also, the TAMs regulate multiple features of microglia, the significant sensors of disorder in the central nervous system (CNS). In this review, we describe the biology of the TAM receptors and ligands in the CNS. Then, we discuss the relationship between the TAM system and AD, specially focusing on its functional expression in the microglia. Finally, we also summarize some agents that could interfere with the TAM signaling pathways and discuss potential difficulties and strategies for drug development.
Collapse
Affiliation(s)
- Shiqi Zhou
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Yanyan Li
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Zhao Zhang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Yuhe Yuan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
17
|
Gardner AM, Atkinson JR, Wilkinson NM, Jerome AD, Bellinger CE, Sas AR, Segal BM. TAM receptor signaling dictates lesion location and clinical phenotype during experimental autoimmune encephalomyelitis. J Neuroimmunol 2023; 375:578016. [PMID: 36708633 DOI: 10.1016/j.jneuroim.2023.578016] [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: 10/30/2022] [Revised: 12/22/2022] [Accepted: 01/06/2023] [Indexed: 01/21/2023]
Abstract
Experimental autoimmune encephalomyelitis (EAE), induced by the adoptive transfer of Th17 cells, typically presents with ascending paralysis and inflammatory demyelination of the spinal cord. Brain white matter is relatively spared. Here we show that treatment of Th17 transfer recipients with a highly selective inhibitor to the TAM family of tyrosine kinase receptors results in ataxia associated with a shift of the inflammatory infiltrate to the hindbrain parenchyma. During homeostasis and preclinical EAE, hindbrain microglia express high levels of the TAM receptor Mer. Our data suggest that constitutive TAM receptor signaling in hindbrain microglia confers region-specific protection against Th17 mediated EAE.
Collapse
Affiliation(s)
- Ashley Munie Gardner
- Department of Neurology, The Ohio State University, 395 W. 12th Ave., 7th Floor, Columbus, OH 43210, USA; The Neuroscience Research Institute, The Ohio State University, 395 W. 12th Ave., 7th Floor, Columbus, OH 43210, USA; Graduate Program in Immunology, University of Michigan Medical School, 2978 Taubman Health Sciences Library, 1135 Catherine, Ann Arbor, MI 48109, USA.
| | - Jeffrey R Atkinson
- Department of Neurology, The Ohio State University, 395 W. 12th Ave., 7th Floor, Columbus, OH 43210, USA; The Neuroscience Research Institute, The Ohio State University, 395 W. 12th Ave., 7th Floor, Columbus, OH 43210, USA.
| | - Nicole M Wilkinson
- Department of Neurology, The Ohio State University, 395 W. 12th Ave., 7th Floor, Columbus, OH 43210, USA.
| | - Andrew D Jerome
- Department of Neurology, The Ohio State University, 395 W. 12th Ave., 7th Floor, Columbus, OH 43210, USA; The Neuroscience Research Institute, The Ohio State University, 395 W. 12th Ave., 7th Floor, Columbus, OH 43210, USA.
| | - Calli E Bellinger
- Department of Neurology, The Ohio State University, 395 W. 12th Ave., 7th Floor, Columbus, OH 43210, USA; The Neuroscience Research Institute, The Ohio State University, 395 W. 12th Ave., 7th Floor, Columbus, OH 43210, USA; Neuroscience Graduate Program, The Ohio State University, 255 Institute for Behavioral Medicine Research Building (IBMR), 460 Medical Center Drive, Columbus, OH 43210, USA.
| | - Andrew R Sas
- Department of Neurology, The Ohio State University, 395 W. 12th Ave., 7th Floor, Columbus, OH 43210, USA; The Neuroscience Research Institute, The Ohio State University, 395 W. 12th Ave., 7th Floor, Columbus, OH 43210, USA.
| | - Benjamin M Segal
- Department of Neurology, The Ohio State University, 395 W. 12th Ave., 7th Floor, Columbus, OH 43210, USA; The Neuroscience Research Institute, The Ohio State University, 395 W. 12th Ave., 7th Floor, Columbus, OH 43210, USA.
| |
Collapse
|
18
|
Abstract
Tyro3, Axl and Mertk are members of the TAM family of tyrosine kinase receptors. TAMs are activated by two structurally homologous ligands GAS6 and PROS1. TAM receptors and ligands are widely distributed and often co-expressed in the same cells allowing diverse functions across many systems including the immune, reproductive, vascular, and the developing as well as adult nervous systems. This review will focus specifically on TAM signaling in the nervous system, highlighting the essential roles this pathway fulfills in maintaining cell survival and homeostasis, cellular functions such as phagocytosis, immunity and tissue repair. Dysfunctional TAM signaling can cause complications in development, disruptions in homeostasis which can rouse autoimmunity, neuroinflammation and neurodegeneration. The development of therapeutics modulating TAM activities in the nervous system has great prospects, however, foremost we need a complete understanding of TAM signaling pathways.
Collapse
Affiliation(s)
- Tal Burstyn-Cohen
- Institute for Dental Sciences, Faculty of Dental Medicine, The Hebrew University-Hadassah, Jerusalem, Israel
| | - Arielle Hochberg
- Institute for Dental Sciences, Faculty of Dental Medicine, The Hebrew University-Hadassah, Jerusalem, Israel
| |
Collapse
|
19
|
Tsouki F, Williams A. Multifaceted involvement of microglia in gray matter pathology in multiple sclerosis. Stem Cells 2021; 39:993-1007. [PMID: 33754376 DOI: 10.1002/stem.3374] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 03/01/2021] [Indexed: 06/12/2023]
Abstract
In the inflammatory demyelinating neurodegenerative disease multiple sclerosis (MS), there is increasing interest in gray matter pathology, as neuronal loss and cortical atrophy correlate with disability and disease progression, and MS therapeutics fail to significantly slow or stop neurodegeneration. Microglia, the central nervous system (CNS)-resident macrophages, are extensively involved in white matter MS pathology, but are also implicated in gray matter pathology, similar to other neurodegenerative diseases, for which there is synaptic, axonal, and neuronal degeneration. Microglia display regional heterogeneity within the CNS, which reflects their highly plastic nature and their ability to deliver context-dependent responses tailored to the demands of their microenvironment. Therefore, microglial roles in the MS gray matter in part reflect and in part diverge from those in the white matter. The present review summarizes current knowledge of microglial involvement in gray matter changes in MS, in demyelination, synaptic damage, and neurodegeneration, with evidence implicating microglia in pathology, neuroprotection, and repair. As our understanding of microglial physiology and pathophysiology increases, we describe how we are moving toward potential therapeutic applications in MS, harnessing microglia to protect and regenerate the CNS.
Collapse
Affiliation(s)
- Foteini Tsouki
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh, UK
| | - Anna Williams
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, The University of Edinburgh, Edinburgh BioQuarter, Edinburgh, UK
| |
Collapse
|
20
|
Cruz-Herranz A, Oertel FC, Kim K, Cantó E, Timmons G, Sin JH, Devereux M, Baker N, Michel B, Schubert RD, Rani L, Cordano C, Baranzini SE, Green AJ. Distinctive waves of innate immune response in the retina in experimental autoimmune encephalomyelitis. JCI Insight 2021; 6:e149228. [PMID: 34100385 PMCID: PMC8262300 DOI: 10.1172/jci.insight.149228] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 04/28/2021] [Indexed: 12/27/2022] Open
Abstract
Neurodegeneration mediates neurological disability in inflammatory demyelinating diseases of the CNS. The role of innate immune cells in mediating this damage has remained controversial with evidence for destructive and protective effects. This has complicated efforts to develop treatment. The time sequence and dynamic evolution of the opposing functions are especially unclear. Given limits of in vivo monitoring in human diseases such as multiple sclerosis (MS), animal models are warranted to investigate the association and timing of innate immune activation with neurodegeneration. Using noninvasive in vivo retinal imaging of experimental autoimmune encephalitis (EAE) in CX3CR1GFP/+–knock-in mice followed by transcriptional profiling, we are able to show 2 distinct waves separated by a marked reduction in the number of innate immune cells and change in cell morphology. The first wave is characterized by an inflammatory phagocytic phenotype preceding the onset of EAE, whereas the second wave is characterized by a regulatory, antiinflammatory phenotype during the chronic stage. Additionally, the magnitude of the first wave is associated with neuronal loss. Two transcripts identified — growth arrest–specific protein 6 (GAS6) and suppressor of cytokine signaling 3 (SOCS3) — might be promising targets for enhancing protective effects of microglia in the chronic phase after initial injury.
Collapse
Affiliation(s)
- Andrés Cruz-Herranz
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Frederike C Oertel
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, California, USA.,Experimental and Clinical Research Center (ECRC), Max-Delbrück-Centrum for Molecular Medicine, and.,NeuroCure Clinical Research Center (NCRC), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Kicheol Kim
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Ester Cantó
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Garrett Timmons
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Jung H Sin
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Michael Devereux
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Nicholas Baker
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Brady Michel
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Ryan D Schubert
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Lakshmisahithi Rani
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Christian Cordano
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Sergio E Baranzini
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, California, USA
| | - Ari J Green
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, California, USA.,Department of Ophthalmology, University of California San Francisco, San Francisco, California, USA
| |
Collapse
|
21
|
Wanke F, Gutbier S, Rümmelin A, Steinberg M, Hughes LD, Koenen M, Komuczki J, Regan-Komito D, Wagage S, Hesselmann J, Thoma R, Brugger D, Christopeit T, Wang H, Point F, Hallet R, Ghosh S, Rothlin CV, Patsch C, Geering B. Ligand-dependent kinase activity of MERTK drives efferocytosis in human iPSC-derived macrophages. Cell Death Dis 2021; 12:538. [PMID: 34035216 PMCID: PMC8149813 DOI: 10.1038/s41419-021-03770-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 04/13/2021] [Accepted: 04/27/2021] [Indexed: 12/15/2022]
Abstract
Removal of apoptotic cells by phagocytes (also called efferocytosis) is a crucial process for tissue homeostasis. Professional phagocytes express a plethora of surface receptors enabling them to sense and engulf apoptotic cells, thus avoiding persistence of dead cells and cellular debris and their consequent effects. Dysregulation of efferocytosis is thought to lead to secondary necrosis and associated inflammation and immune activation. Efferocytosis in primarily murine macrophages and dendritic cells has been shown to require TAM RTKs, with MERTK and AXL being critical for clearance of apoptotic cells. The functional role of human orthologs, especially the exact contribution of each individual receptor is less well studied. Here we show that human macrophages differentiated in vitro from iPSC-derived precursor cells express both AXL and MERTK and engulf apoptotic cells. TAM RTK agonism by the natural ligand growth-arrest specific 6 (GAS6) significantly enhanced such efferocytosis. Using a newly-developed mouse model of kinase-dead MERTK, we demonstrate that MERTK kinase activity is essential for efferocytosis in peritoneal macrophages in vivo. Moreover, human iPSC-derived macrophages treated in vitro with blocking antibodies or small molecule inhibitors recapitulated this observation. Hence, our results highlight a conserved MERTK function between mice and humans, and the critical role of its kinase activity in homeostatic efferocytosis.
Collapse
Affiliation(s)
- Florian Wanke
- Immunology, Infectious Diseases and Ophthalmology (I2O) Discovery and Translational Area, Roche Innovation Center, Basel, Switzerland.
| | - Simon Gutbier
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center, Basel, Switzerland
| | - Anna Rümmelin
- Immunology, Infectious Diseases and Ophthalmology (I2O) Discovery and Translational Area, Roche Innovation Center, Basel, Switzerland.,Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center, Basel, Switzerland
| | - Malte Steinberg
- Immunology, Infectious Diseases and Ophthalmology (I2O) Discovery and Translational Area, Roche Innovation Center, Basel, Switzerland
| | - Lindsey D Hughes
- Yale School of Medicine, Department for Immunobiology, Yale University, New Haven, Connecticut, USA
| | - Mascha Koenen
- Yale School of Medicine, Department for Immunobiology, Yale University, New Haven, Connecticut, USA.,Laboratory of Molecular Metabolism, Rockefeller University, New York, New York, USA
| | - Juliana Komuczki
- Immunology, Infectious Diseases and Ophthalmology (I2O) Discovery and Translational Area, Roche Innovation Center, Basel, Switzerland
| | - Daniel Regan-Komito
- Immunology, Infectious Diseases and Ophthalmology (I2O) Discovery and Translational Area, Roche Innovation Center, Basel, Switzerland
| | - Sagie Wagage
- Yale School of Medicine, Department for Immunobiology, Yale University, New Haven, Connecticut, USA
| | - Julia Hesselmann
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center, Basel, Switzerland
| | - Ralf Thoma
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center, Basel, Switzerland
| | - Doris Brugger
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center, Basel, Switzerland
| | - Tony Christopeit
- Roche Pharma Research and Early Development, Roche Innovation Center München, Penzberg, Germany
| | - Hayian Wang
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center, Basel, Switzerland
| | - Floriane Point
- Immunology, Infectious Diseases and Ophthalmology (I2O) Discovery and Translational Area, Roche Innovation Center, Basel, Switzerland
| | - Remy Hallet
- Immunology, Infectious Diseases and Ophthalmology (I2O) Discovery and Translational Area, Roche Innovation Center, Basel, Switzerland
| | - Sourav Ghosh
- Yale School of Medicine, Department for Neurology, Yale University, New Haven, Connecticut, USA.,Yale School of Medicine, Department for Pharmacology, Yale University, New Haven, Connecticut, USA
| | - Carla V Rothlin
- Yale School of Medicine, Department for Immunobiology, Yale University, New Haven, Connecticut, USA.,Yale School of Medicine, Department for Pharmacology, Yale University, New Haven, Connecticut, USA
| | - Christoph Patsch
- Roche Pharma Research and Early Development, Therapeutic Modalities, Roche Innovation Center, Basel, Switzerland.,BlueRock Therapeutics, New York, New York, USA
| | - Barbara Geering
- Immunology, Infectious Diseases and Ophthalmology (I2O) Discovery and Translational Area, Roche Innovation Center, Basel, Switzerland
| |
Collapse
|
22
|
Multiple sclerosis risk gene Mertk is required for microglial activation and subsequent remyelination. Cell Rep 2021; 34:108835. [PMID: 33691116 DOI: 10.1016/j.celrep.2021.108835] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 12/23/2020] [Accepted: 02/17/2021] [Indexed: 12/26/2022] Open
Abstract
In multiple sclerosis (MS) and other neurological diseases, the failure to repair demyelinated lesions contributes to axonal damage and clinical disability. Here, we provide evidence that Mertk, a gene highly expressed by microglia that alters MS risk, is required for efficient remyelination. Compared to wild-type (WT) mice, Mertk-knockout (KO) mice show impaired clearance of myelin debris and remyelination following demyelination. Using single-cell RNA sequencing, we characterize Mertk-influenced responses to cuprizone-mediated demyelination and remyelination across different cell types. Mertk-KO brains show an attenuated microglial response to demyelination but an elevated proportion of interferon (IFN)-responsive microglia. In addition, we identify a transcriptionally distinct subtype of surviving oligodendrocytes specific to demyelinated lesions. The inhibitory effect of myelin debris on remyelination is mediated in part by IFNγ, which further impedes microglial clearance of myelin debris and inhibits oligodendrocyte differentiation. Together, our work establishes a role for Mertk in microglia activation, phagocytosis, and migration during remyelination.
Collapse
|
23
|
Wei S, Chang S, Dong Y, Xu L, Yuan X, Jia H, Zhang J, Liang L. Electro-acupuncture Suppresses AXL Expression in Dorsal Root Ganglion Neurons and Enhances Analgesic Effect of AXL Inhibitor in Spinal Nerve Ligation Induced-Neuropathic Pain Rats. Neurochem Res 2021; 46:504-512. [PMID: 33387191 DOI: 10.1007/s11064-020-03185-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Revised: 11/15/2020] [Accepted: 11/18/2020] [Indexed: 12/15/2022]
Abstract
Electro-acupuncture (EA) has been used for clinic analgesia for many years. However, its mechanisms are not fully understood. We recently reported that AXL, a tyrosine kinase receptor, contributes to the peripheral mechanism of neuropathic pain. We here aim to figure out the significance of EA on neuropathic pain mediated by AXL in dorsal root ganglion (DRG). Spinal nerve ligation (SNL) was used as a neuropathic pain model. EA was applied at ''Huantiao'' (GB-30) and ''Yanglingquan'' (GB-34) acupoints for 30 min daily from day 7 to day 10 after SNL. EA not only gradually attenuated SNL-induced mechanical allodynia, but also suppressed the expression of phosphorylated AXL (p-AXL) and AXL in injured DRGs of SNL rats examined by western blotting and immunofluorescence. Moreover, intrathecal injection of the subthreshold dose of AXL inhibitor TP0903, significantly prolonged the analgesic time of single EA treatment and enhanced the analgesic effect of repeated EA treatments, suggesting a synergic effect of EA and AXL inhibitor. These results indicate that AXL signaling underlies EA analgesia and combination of AXL inhibitor and EA might be a new strategy for clinic analgesia on neuropathic pain.
Collapse
Affiliation(s)
- Siqi Wei
- Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Research Center of Stomatology, Xi'an Jiaotong University College of Stomatology, Xi'an, Shaanxi, People's Republic of China
| | - Shuyang Chang
- Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
| | - Yue Dong
- Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
| | - Linping Xu
- Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
| | - Xiaocui Yuan
- Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
| | - Hong Jia
- Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China
| | - Jun Zhang
- Department of Pain Medicine, Tianjin Union Medical Center, Nankai University, Tianjin, People's Republic of China
| | - Lingli Liang
- Institute of Neuroscience, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China.
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, People's Republic of China.
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Beijing, People's Republic of China.
| |
Collapse
|
24
|
Ortiz Wilczyñski JM, Olexen CM, Errasti AE, Schattner M, Rothlin CV, Correale J, Carrera Silva EA. GAS6 signaling tempers Th17 development in patients with multiple sclerosis and helminth infection. PLoS Pathog 2020; 16:e1009176. [PMID: 33347509 PMCID: PMC7785232 DOI: 10.1371/journal.ppat.1009176] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 01/05/2021] [Accepted: 11/19/2020] [Indexed: 12/19/2022] Open
Abstract
Multiple sclerosis (MS) is a highly disabling neurodegenerative autoimmune condition in which an unbalanced immune response plays a critical role. Although the mechanisms remain poorly defined, helminth infections are known to modulate the severity and progression of chronic inflammatory diseases. The tyrosine kinase receptors TYRO3, AXL, and MERTK (TAM) have been described as inhibitors of the immune response in various inflammatory settings. We show here that patients with concurrent natural helminth infections and MS condition (HIMS) had an increased expression of the negative regulatory TAM receptors in antigen-presenting cells and their agonist GAS6 in circulating CD11bhigh and CD4+ T cells compared to patients with only MS. The Th17 subset was reduced in patients with HIMS with a subsequent downregulation of its pathogenic genetic program. Moreover, these CD4+ T cells promoted lower levels of the co-stimulatory molecules CD80, CD86, and CD40 on dendritic cells compared with CD4+ T cells from patients with MS, an effect that was GAS6-dependent. IL-10+ cells from patients with HIMS showed higher GAS6 expression levels than Th17 cells, and inhibition of phosphatidylserine/GAS6 binding led to an expansion of Th17 effector genes. The addition of GAS6 on activated CD4+ T cells from patients with MS restrains the Th17 gene expression signature. This cohort of patients with HIMS unravels a promising regulatory mechanism to dampen the Th17 inflammatory response in autoimmunity. Helminths have co-evolved with human civilization, and the rapid exclusion from their environment, in the last few decades, has tremendously affected the immune development and regulation. Moreover, several epidemiological studies have shown an inverse correlation between the exposure of these organisms and the development of autoimmunity in industrialized countries. In this sense, helminth therapy appears to be a promising concept to oppose chronic inflammatory and autoimmune diseases because they are master manipulators of host immunity, albeit the mechanisms remain poorly defined. For this reason, it is essential to decipher the main regulatory pathways to hijack the immune response in the absence of parasite infection. Our research described how helminth infection promotes regulatory mechanisms based on the tyrosine kinase TYRO3, AXL, MERTK (TAM) receptors, and their ligand GAS6 to dampen Th17 development and the inflammatory response in patients with multiple sclerosis (MS), a neurodegenerative autoimmune disorder. We show here that GAS6 plays a critical role in the regulation of pro-inflammatory cytokines, transcriptional programs, and plasticity of IL-17 subset. Our work substantiates the hypothesis that enhancing the TAM axis in a manner similar to helminth infection could be a promising alternative for autoimmune diseases.
Collapse
Affiliation(s)
- Juan M. Ortiz Wilczyñski
- Institute of Experimental Medicine, CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Cinthia M. Olexen
- Institute of Experimental Medicine, CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Andrea E. Errasti
- Institute of Pharmacology, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
| | - Mirta Schattner
- Institute of Experimental Medicine, CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Carla V. Rothlin
- Department of Immunobiology and Pharmacology, Yale University, Connecticut, United States of America
| | | | - Eugenio A. Carrera Silva
- Institute of Experimental Medicine, CONICET-National Academy of Medicine, Buenos Aires, Argentina
- * E-mail:
| |
Collapse
|
25
|
Li XL, Zhang B, Liu W, Sun MJ, Zhang YL, Liu H, Wang MX. Rapamycin Alleviates the Symptoms of Multiple Sclerosis in Experimental Autoimmune Encephalomyelitis (EAE) Through Mediating the TAM-TLRs-SOCS Pathway. Front Neurol 2020; 11:590884. [PMID: 33329339 PMCID: PMC7728797 DOI: 10.3389/fneur.2020.590884] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 11/09/2020] [Indexed: 12/29/2022] Open
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS). Our research aimed to find an immunomodulatory therapy for MS. An experimental autoimmune encephalomyelitis (EAE) mouse model of MS was established induced with the syntheticmyelin oligodendrocyte glycoprotein peptide 35-55 (MOG35-55). Fifty C57BL/6 mice were randomly divided into the Normal group, EAE group, and Rapamycin group (EAE mice treated with three different doses of rapamycin). Hematoxylin and eosin staining and Weil myelin staining were performed on the brain tissues of mice after 21 days post-immunization. The protein expression of Gas6, Tyro3, Axl, Mer in paraventricular tissues were analyzed by immunohistochemistry. The mRNA and protein expression of Gas6, Tyro3, Axl, Mer, SOCS1, SOCS3, Toll-like receptor (TLR) 3, and TLR4 were detected by quantitative real-time PCR (qRT-PCR) and Western blot, respectively. An enzyme-linked immunosorbent assay (ELISA) was used to detect the secretion of the inflammatory factors IFN-γ and IL-17. Rapamycin treatment could ameliorate the behavior impairment in EAE mice induced by MOG35-55. The expression of Gas6, Tyro3, Axl, Mer, SOCS1, and SOCS3 were decreased in EAE mice at 21 days post-immunization, while the expression of Gas6, Tyro3, Axl, and Mer in rapamycin group was higher than that in EAE group. It was accompanied by an increase in anti-inflammatory proteins SOCS1 and SOCS3, a decrease in the inflammatory proteins TLR-3, TLR-4 and in the amount of IFN-γ, and IL-17. Rapamycin injection relieved the nerve function of and the loss of myelin sheath in the EAE mice, mainly through mediating the TAM-TLRs-SOCS signaling pathway to regulate natural immunity.
Collapse
Affiliation(s)
- Xiao-ling Li
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Bo Zhang
- Department of Cardiology, The First Hospital of Lanzhou University, Lanzhou, China
| | - Wei Liu
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Meng-jiao Sun
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Ya-lan Zhang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Hui Liu
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| | - Man-xia Wang
- Department of Neurology, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
26
|
DuBois JC, Ray AK, Davies P, Shafit-Zagardo B. Anti-Axl antibody treatment reduces the severity of experimental autoimmune encephalomyelitis. J Neuroinflammation 2020; 17:324. [PMID: 33121494 PMCID: PMC7599105 DOI: 10.1186/s12974-020-01982-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 10/05/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Multiple sclerosis is an immune-mediated disease of the central nervous system (CNS) characterized by inflammation, oligodendrocytes loss, demyelination, and damaged axons. Tyro3, Axl, and MerTK belong to a family of receptor tyrosine kinases that regulate innate immune responses and CNS homeostasis. During experimental autoimmune encephalomyelitis (EAE), the mRNA expression of MerTK, Gas6, and Axl significantly increase, whereas Tyro3 and ProS1 remain unchanged. We have shown that Gas6 is neuroprotective during EAE, and since Gas6 activation of Axl may be necessary for conferring neuroprotection, we sought to determine whether α-Axl or α-MerTK antibodies, shown by others to activate their respective receptors in vivo, could effectively reduce inflammation and neurodegeneration. METHODS Mice received either α-Axl, α-MerTK, IgG isotype control, or PBS before the onset of EAE symptoms. EAE clinical course, axonal damage, demyelination, cytokine production, and immune cell activation in the CNS were used to determine the severity of EAE. RESULTS α-Axl antibody treatment significantly decreased the EAE clinical indices of female mice during chronic EAE and of male mice during both acute and chronic phases. The number of days mice were severely paralyzed also significantly decreased with α-Axl treatment. Inflammatory macrophages/microglia and the extent of demyelination significantly decreased in the spinal cords of α-Axl-treated mice during chronic EAE, with no differences in the production of pro-inflammatory cytokines. α-MerTK antibody did not influence EAE induction or progression. CONCLUSION Our data suggests that the beneficial effect of Gas6/Axl signaling observed in mice administered with Gas6 can be partially preserved by administering an activating α-Axl antibody, but not α-MerTK.
Collapse
Affiliation(s)
- Juwen C. DuBois
- Department of Pathology, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461 USA
| | - Alex K. Ray
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY USA
| | - Peter Davies
- North Shore-LIJ Health System, Feinstein Institute for Medical Research, Manhasset, NY USA
| | - Bridget Shafit-Zagardo
- Department of Pathology, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461 USA
| |
Collapse
|
27
|
Di Stasi R, De Rosa L, D'Andrea LD. Therapeutic aspects of the Axl/Gas6 molecular system. Drug Discov Today 2020; 25:2130-2148. [PMID: 33002607 DOI: 10.1016/j.drudis.2020.09.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/14/2020] [Accepted: 09/21/2020] [Indexed: 12/17/2022]
Abstract
Axl receptor tyrosine kinase (RTK) and its ligand, growth arrest-specific protein 6 (Gas6), are involved in several biological functions and participate in the development and progression of a range of malignancies and autoimmune disorders. In this review, we present this molecular system from a drug discovery perspective, highlighting its therapeutic implications and challenges that need to be addressed. We provide an update on Axl/Gas6 axis biology, exploring its role in fields ranging from angiogenesis, cancer development and metastasis, immune response and inflammation to viral infection. Finally, we summarize the molecules that have been developed to date to target the Axl/Gas6 molecular system for therapeutic and diagnostic applications.
Collapse
Affiliation(s)
- Rossella Di Stasi
- Istituto di Biostrutture e Bioimmagini, CNR, Via Mezzocannone 16, 80134 Napoli, Italy
| | - Lucia De Rosa
- Istituto di Biostrutture e Bioimmagini, CNR, Via Mezzocannone 16, 80134 Napoli, Italy
| | - Luca D D'Andrea
- Istituto di Biostrutture e Bioimmagini, CNR, Via Nizza 52, 10126 Torino, Italy.
| |
Collapse
|
28
|
Resveratrol protects retinal ganglion cell axons through regulation of the SIRT1-JNK pathway. Exp Eye Res 2020; 200:108249. [PMID: 32956685 DOI: 10.1016/j.exer.2020.108249] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Revised: 08/12/2020] [Accepted: 09/14/2020] [Indexed: 01/21/2023]
Abstract
It is reported that Ischemia and reperfusion damage (I/R damage) can lead to retinal ganglion cell (RGC) death and neurodegeneration, which in turn can lead to irreversible vision loss. In this study, we sought to understand the neuroprotective effect of resveratrol, the important activator of sirtuin1 (SIRT1), on RGC survival in I/R damage model and the molecular mechanism that mediate this effect. Our results show that resveratrol could reverse axonal swelling, holes, and the chaos of the nucleus in axons of RGCs caused by I/R. At the same time, resveratrol could also reverse the activation of retinal astrocytes and the loss of RGCs caused by I/R. Resveratrol increased the expression of SIRT1 while decreasing the phosphorylation of N-terminal kinase (JNK). SP600125(JNK inhibitor) decreased the phosphorylation of JNK while increasing the expression of SIRT1, indicating that SIRT1 and JNK can interact with each other. Simultaneous administration of resveratrol and sirtinol (SIRT1 inhibitor) neither increased the expression of SIRT1 nor decreased the phosphorylation of JNK, indicating that resveratrol affects the phosphorylation of JNK by SIRT1. In total, our research shows that resveratrol treatment significantly reduces apoptosis and axonal degeneration of RGCs, and this protection is partly mediated through the SIRT1-JNK pathway.
Collapse
|
29
|
Gas6 Induces Myelination through Anti-Inflammatory IL-10 and TGF-β Upregulation in White Matter and Glia. Cells 2020; 9:cells9081779. [PMID: 32722558 PMCID: PMC7465828 DOI: 10.3390/cells9081779] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/16/2020] [Accepted: 07/24/2020] [Indexed: 02/07/2023] Open
Abstract
The Gas6–TAM (Tyro3, Axl, Mer) ligand–receptor system is believed to promote central nervous system (CNS) (re)myelination and glial cell development. An additional important function of Gas6–TAM signalling appears to be the regulation of immunity and inflammation, which remains to be fully elucidated in the CNS. Here, we characterised the expression of TAM receptors and ligands in individual CNS glial cell types, observing high expression of Gas6 and the TAM receptors, Mer and Axl, in microglia, and high expression of Tyro3 in astrocytes. We also investigated the effect of Gas6 on the inflammatory cytokine response in the optic nerve and in mixed glial cell cultures from wildtype and single TAM receptor knockout mice. In wildtype and Mer-deficient cultures, Gas6 significantly stimulated the expression of the anti-inflammatory/pro-repair cytokines interleukin 10 (IL-10) and transforming growth factor β (TGF-β), whereas this effect was absent in either Tyro3 or Axl knockout cultures. Furthermore, Gas6 caused upregulation of myelin basic protein (MBP) expression in optic nerves, which was blocked by a neutralising antibody against IL-10. In conclusion, our data show that microglia are both a major source of Gas6 as well as an effector of Gas6 action in the CNS through the upregulation of anti-inflammatory and pro-repair mediators. Furthermore, the presence of both Axl and Tyro3 receptors appears to be necessary for these effects of Gas6. In addition, IL-10, alongside suppressing inflammation and immunity, mediates the pro-myelinating mechanism of Gas6 action in the optic nerve. Therefore, Gas6 may present an attractive target for novel therapeutic interventions for demyelinating as well as neuroinflammatory disorders of the CNS.
Collapse
|
30
|
Stratton JA, Holmes A, Rosin NL, Sinha S, Vohra M, Burma NE, Trang T, Midha R, Biernaskie J. Macrophages Regulate Schwann Cell Maturation after Nerve Injury. Cell Rep 2019; 24:2561-2572.e6. [PMID: 30184491 DOI: 10.1016/j.celrep.2018.08.004] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 04/15/2018] [Accepted: 07/29/2018] [Indexed: 12/26/2022] Open
Abstract
Pro-regenerative macrophages are well known for their role in promoting tissue repair; however, their specific roles in promoting regeneration of the injured nerve are not well defined. Specifically, how macrophages interact with Schwann cells following injury during remyelination has been largely unexplored. We demonstrate that after injury, including in humans, macrophages function to clear debris and persist within the nerve microenvironment. Macrophage ablation immediately preceding remyelination results in an increase in immature Schwann cell density, a reduction in remyelination, and long-term deficits in conduction velocity. Targeted RNA-seq of macrophages from injured nerve identified Gas6 as one of several candidate factors involved in regulating Schwann cell dynamics. Functional studies show that the absence of Gas6 within monocyte lineage cells impairs Schwann cell remyelination within the injured nerve. These results demonstrate a role for macrophages in regulating Schwann cell function during nerve regeneration and highlight a molecular mechanism by which this occurs.
Collapse
Affiliation(s)
- Jo Anne Stratton
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada; Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Alexandra Holmes
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada; Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Nicole L Rosin
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Sarthak Sinha
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Mohit Vohra
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada; Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Nicole E Burma
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada; Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Tuan Trang
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada; Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Rajiv Midha
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada; Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada
| | - Jeff Biernaskie
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada; Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4Z6, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada.
| |
Collapse
|
31
|
TAM Receptor Pathways at the Crossroads of Neuroinflammation and Neurodegeneration. DISEASE MARKERS 2019; 2019:2387614. [PMID: 31636733 PMCID: PMC6766163 DOI: 10.1155/2019/2387614] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 06/04/2019] [Accepted: 08/12/2019] [Indexed: 02/07/2023]
Abstract
Increasing evidence suggests that pathogenic mechanisms underlying neurodegeneration are strongly linked with neuroinflammatory responses. Tyro3, Axl, and Mertk (TAM receptors) constitute a subgroup of the receptor tyrosine kinase family, cell surface receptors which transmit signals from the extracellular space to the cytoplasm and nucleus. TAM receptors and the corresponding ligands, Growth Arrest Specific 6 and Protein S, are expressed in different tissues, including the nervous system, playing complex roles in tissue repair, inflammation and cell survival, proliferation, and migration. In the nervous system, TAM receptor signalling modulates neurogenesis and neuronal migration, synaptic plasticity, microglial activation, phagocytosis, myelination, and peripheral nerve repair, resulting in potential interest in neuroinflammatory and neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease, and Multiple Sclerosis. In Alzheimer and Parkinson diseases, a role of TAM receptors in neuronal survival and pathological protein aggregate clearance has been suggested, while in Multiple Sclerosis TAM receptors are involved in myelination and demyelination processes. To better clarify roles and pathways involving TAM receptors may have important therapeutic implications, given the fine modulation of multiple molecular processes which could be reached. In this review, we summarise the roles of TAM receptors in the central nervous system, focusing on the regulation of immune responses and microglial activities and analysing in vitro and in vivo studies regarding TAM signalling involvement in neurodegeneration.
Collapse
|
32
|
Growth arrest-specific protein 6 (Gas6) attenuates inflammatory injury and apoptosis in iodine-induced NOD.H-2 h4 mice. Int Immunopharmacol 2019; 73:333-342. [PMID: 31129420 DOI: 10.1016/j.intimp.2019.04.038] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 03/15/2019] [Accepted: 04/18/2019] [Indexed: 02/02/2023]
Abstract
PURPOSE Growth arrest-specific protein 6 (Gas6) is a vitamin K-dependent protein that plays an important role in the pathogenesis of autoimmune diseases. The purpose of this study was to explore the expression of Gas6 and its effects on autoimmune thyroiditis (AIT). METHOD A total of 24 male NOD.H-2h4 mice were randomly assigned to three groups: (1) a control group supplied with regular water; (2) a sodium iodide (NaI) group supplied with 0.005% sodium iodide water; and (3) a group treated with recombinant mouse Gas6 (rmGas6) after iodine supplementation (NaI + Gas6 group). The severity of lymphocytic infiltration in the thyroid was measured through histopathology. Serum levels of tumor necrosis factor α (TNF-α), interleukin (IL) 6 and IL-1β, as well as anti-thyroglobulin antibody (TgAb) titers were measured using an enzyme-linked immunosorbent assay. In addition, the expression of Gas6, Caspase 3, TAM receptors (Axl and MerTK), nuclear factor κB (NF-κB) and I-kappa-B α (IκB-α) were measured by Western blotting. Finally, the proportions of T cells were determined in the splenocytes of NOD.H-2h4 mice by flow cytometry. RESULTS The mRNA and protein expression of Gas6 was significantly lower in the NaI group compared to the control group. Serum levels of TgAb, TNF-α, IL-6 and IL-1β were also significantly higher in the NaI group but were dramatically reduced after rmGas6 injection. The prevalence of thyroiditis and the infiltration of lymphocytes were significantly lower in the NaI + Gas6 group compared to the NaI group. The protein expression of cleaved-Caspase 3, phosphorylation of MerTK, and NF-κB and IκB-α in the thyroid gland were significantly reduced after rmGas6 administration. The proportion of Th1, Th2 and Th17 cells in splenocytes were also significantly reduced after rmGas6 treatment, whereas there was a dramatic increase in the proportion of Treg cells. CONCLUSION Gas6 exerts an anti-inflammatory effect in a mouse model of AIT and may therefore be a potential therapeutic target.
Collapse
|
33
|
Lee CH, Chun T. Anti-Inflammatory Role of TAM Family of Receptor Tyrosine Kinases Via Modulating Macrophage Function. Mol Cells 2019; 42:1-7. [PMID: 30699285 PMCID: PMC6354059 DOI: 10.14348/molcells.2018.0419] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/04/2018] [Accepted: 12/07/2018] [Indexed: 01/06/2023] Open
Abstract
Macrophage is an important innate immune cell that not only initiates inflammatory responses, but also functions in tissue repair and anti-inflammatory responses. Regulating macrophage activity is thus critical to maintain immune homeostasis. Tyro3, Axl, and Mer are integral membrane proteins that constitute TAM family of receptor tyrosine kinases (RTKs). Growing evidence indicates that TAM family receptors play an important role in anti-inflammatory responses through modulating the function of macrophages. First, macrophages can recognize apoptotic bodies through interaction between TAM family receptors expressed on macrophages and their ligands attached to apoptotic bodies. Without TAM signaling, macrophages cannot clear up apoptotic cells, leading to broad inflammation due to over-activation of immune cells. Second, TAM signaling can prevent chronic activation of macrophages by attenuating inflammatory pathways through particular pattern recognition receptors and cytokine receptors. Third, TAM signaling can induce autophagy which is an important mechanism to inhibit NLRP3 inflammasome activation in macrophages. Fourth, TAM signaling can inhibit polarization of M1 macrophages. In this review, we will focus on mechanisms involved in how TAM family of RTKs can modulate function of macrophage associated with anti-inflammatory responses described above. We will also discuss several human diseases related to TAM signaling and potential therapeutic strategies of targeting TAM signaling.
Collapse
Affiliation(s)
- Chang-Hee Lee
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841,
Korea
| | - Taehoon Chun
- Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841,
Korea
| |
Collapse
|
34
|
Abstract
BACKGROUND Lower tidal volumes are increasingly used in acute respiratory distress syndrome, but mortality has changed little in the last 20 yr. Therefore, in addition to ventilator settings, it is important to target molecular mediators of injury. Sepsis and other inflammatory states increase circulating concentrations of Gas6, a ligand for the antiinflammatory receptor Axl, and of a soluble decoy form of Axl. We investigated the effects of lung stretch on Axl signaling. METHODS We used a mouse model of early injury from high tidal volume and assessed the effects of inhibiting Axl on in vivo lung injury (using an antagonist R428, n = 4/group). We further determined the effects of stretch on Axl activation using in vitro lung endothelial cells. RESULTS High tidal volume caused mild injury (compliance decreased 6%) as intended, and shedding of the Axl receptor (soluble Axl in bronchoalveolar fluid increased 77%). The Axl antagonist R428 blocked the principal downstream Axl target (suppressor of cytokine signaling 3 [SOCS3]) but did not worsen lung physiology or inflammation. Cyclic stretch in vitro caused Axl to become insensitive to activation by its agonist, Gas6. Finally, in vitro Axl responses were rescued by blocking stretch-activated calcium channels (using guanidinium chloride [GdCl3]), and the calcium ionophore ionomycin replicated the effect of stretch. CONCLUSIONS These data suggest that lung endothelial cell overdistention activates ion channels, and the resultant influx of Ca inactivates Axl. Downstream inactivation of Axl by stretch was not anticipated; preventing this would be required to exploit Axl receptors in reducing lung injury.
Collapse
|
35
|
Cengiz P, Zafer D, Chandrashekhar JH, Chanana V, Bogost J, Waldman A, Novak B, Kintner DB, Ferrazzano PA. Developmental differences in microglia morphology and gene expression during normal brain development and in response to hypoxia-ischemia. Neurochem Int 2019; 127:137-147. [PMID: 30639264 DOI: 10.1016/j.neuint.2018.12.016] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 12/27/2018] [Accepted: 12/27/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND Neuroinflammation plays an important role in ischemic brain injury and recovery, however the interplay between brain development and the neuroinflammatory response is poorly understood. We previously described age-dependent differences in the microglial response and the effect of microglial inhibition. Here we investigate whether age-dependent microglial responses may be related to pre-injury developmental differences in microglial phenotype. METHODS Measures of microglia morphology were quantified using semi-automated software analysis of immunostained sections from postnatal day 2 (P2), P9, P30 and P60 mice using IMARIS. Microglia were isolated from P2, P9, P30 and P60 mice, and expression of markers of classical and alternative microglial activation was assessed, as well as transforming growth factor beta (TGF-β) receptor, Serpine1, Mer Tyrosine Kinase (MerTK), and the suppressor of cytokine signaling (SOCS3). Hypoxia-ischemia (HI) was induced in P9 and P30 mice using unilateral carotid artery ligation and exposure to 10% oxygen for 50 min. Microglia morphology and microglial expression of genes in the TGF-β and MerTK pathways were determined in ipsilateral and contralateral hippocampus. RESULTS A progressive and significant increase in microglia branching morphology was seen in all brain regions from P2 to P30. No consistent classical or alternative activation profile was seen in isolated microglia. A clear transition to increased expression of TGF-β and its downstream effector serpine1 was seen between P9 and P30. A similar increase in expression was seen in MerTK and its downstream effector SOCS3. HI resulted in a significant decrease in branching morphology only in the P9 mice, and expression of TGF-β receptor, Serpine1, MerTK, and SOCS3 were elevated in P30 mice compared to P9 post-HI. CONCLUSION Microglia maturation is associated with changes in morphology and gene expression, and microglial responses to ischemia in the developing brain differ based on the age at which injury occurs.
Collapse
Affiliation(s)
- Pelin Cengiz
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| | - Dila Zafer
- Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Jayadevi H Chandrashekhar
- Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; University of Illinois at Urbana-Champaign, IL, USA
| | - Vishal Chanana
- Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Jacob Bogost
- Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Alex Waldman
- Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Emory University School of Medicine, Atlanta, GA, USA
| | - Becca Novak
- Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Douglas B Kintner
- Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Peter A Ferrazzano
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Waisman Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
36
|
Corpas R, Griñán-Ferré C, Palomera-Ávalos V, Porquet D, García de Frutos P, Franciscato Cozzolino SM, Rodríguez-Farré E, Pallàs M, Sanfeliu C, Cardoso BR. Melatonin induces mechanisms of brain resilience against neurodegeneration. J Pineal Res 2018; 65:e12515. [PMID: 29907977 DOI: 10.1111/jpi.12515] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 06/06/2018] [Indexed: 12/20/2022]
Abstract
Melatonin is an endogenous pleiotropic molecule which orchestrates regulatory functions and protective capacity against age-related ailments. The increase in circulating levels of melatonin through dietary supplements intensifies its health benefits. Investigations in animal models have shown that melatonin protects against Alzheimer's disease (AD)-like pathology, although clinical studies have not been conclusive. We hypothesized that melatonin induces changes in the brain that prevent or attenuate AD by increasing resilience. Therefore, we treated healthy nontransgenic (NoTg) and AD transgenic (3xTg-AD) 6-month-old mice with a daily dose of 10 mg/kg of melatonin until 12 months of age. As expected, melatonin reversed cognitive impairment and dementia-associated behaviors of anxiety and apathy and reduced amyloid and tau burden in 3xTg-AD mice. Remarkably, melatonin induced cognitive enhancement and higher wellness level-related behavior in NoTg mice. At the mechanism level, NF-κB and proinflammatory cytokine expressions were decreased in both NoTg and 3xTg-AD mice. The SIRT1 pathway of longevity and neuroprotection was also activated in both mouse strains after melatonin dosing. Furthermore, we explored new mechanisms and pathways not previously associated with melatonin treatment such as the ubiquitin-proteasome proteolytic system and the recently proposed neuroprotective Gas6/TAM pathway. The upregulation of proteasome activity and the modulation of Gas6 and its receptors by melatonin were similarly displayed by both NoTg and 3xTg-AD mice. Therefore, these results confirm the potential of melatonin treatment against AD pathology, by way of opening new pathways in its mechanisms of action, and demonstrating that melatonin induces cognitive enhancement and brain resilience against neurodegenerative processes.
Collapse
Affiliation(s)
- Rubén Corpas
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC and IDIBAPS, Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Christian Griñán-Ferré
- Faculty of Pharmacy and Food Sciences, Institut de Neurociències, Universitat de Barcelona and CIBERNED, Barcelona, Spain
| | - Verónica Palomera-Ávalos
- Faculty of Pharmacy and Food Sciences, Institut de Neurociències, Universitat de Barcelona and CIBERNED, Barcelona, Spain
| | - David Porquet
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC and IDIBAPS, Barcelona, Spain
| | - Pablo García de Frutos
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC and IDIBAPS, Barcelona, Spain
| | - Silvia M Franciscato Cozzolino
- Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, São Paulo, Brazil
| | - Eduard Rodríguez-Farré
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC and IDIBAPS, Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Mercè Pallàs
- Faculty of Pharmacy and Food Sciences, Institut de Neurociències, Universitat de Barcelona and CIBERNED, Barcelona, Spain
| | - Coral Sanfeliu
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC and IDIBAPS, Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Bárbara R Cardoso
- Institute for Physical Activity and Nutrition Research (IPAN), School of Exercise and Nutrition Sciences, Deakin University, Geelong, Vic., Australia
- The Florey Institute of Neuroscience and Mental Health, Melbourne, Vic., Australia
| |
Collapse
|
37
|
Growth arrest-specific gene 6 transfer promotes mesenchymal stem cell survival and cardiac repair under hypoxia and ischemia via enhanced autocrine signaling and paracrine action. Arch Biochem Biophys 2018; 660:108-120. [PMID: 30365934 DOI: 10.1016/j.abb.2018.10.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 10/08/2018] [Accepted: 10/22/2018] [Indexed: 01/10/2023]
Abstract
Poor cell viability after transplantation has restricted the therapeutic capacity of mesenchymal stem cells (MSCs) for cardiac dysfunction after myocardial infarction (MI). Growth arrest-specific gene 6 (Gas6) encodes a secreted γ-carboxyglutamic acid (Gla)-containing protein that functions in cell growth, adhesion, chemotaxis, mitogenesis and cell survival. In this study, we genetically modified MSCs with Gas6 and evaluated cell survival, cardiac function, and infarct size in a rat model of MI via intramyocardial delivery. Functional studies demonstrated that Gas6 transfer significantly reduced MSC apoptosis, increased survival of MSCs in vitro and in vivo, and that Gas6-engineered MSCs (MSCGas6)-treated animals had smaller infarct size and showed remarkably functional recovery as compared with control MSCs (MSCNull)-treated animals. Mechanistically, Gas6 could enhance phosphatidylinositol 3-kinase (PI3K)/Akt signaling and improve hypoxia-inducible factor-1 alpha (HIF-1α)-driven secretion of four major growth factors (VEGF, bFGF, SDF and IGF-1) in MSCs under hypoxia in an Axl-dependent autocrine manner. The paracrine action of MSCGas6 was further validated by coculture neonatal rat cardiomyocytes with conditioned medium from hypoxia-treated MSCGas6, as well as by pretreatment cardiomyocytes with the specific receptor inhibitors of VEGF, bFGF, SDF and IGF-1. Collectively, our data suggest that Gas6 may advance the efficacy of MSC therapy for post-infarcted heart failure via enhanced Gas6/Axl autocrine prosurvival signaling and paracrine cytoprotective action.
Collapse
|
38
|
Yu Y, Wang D, Li H, Liu Y, Xiang Z, Wu J, Jing X. IPSC‑MSC inhibition assessment in Raw 264.7 cells following oxygen and glucose deprivation reveals a distinct function for cardiopulmonary resuscitation. Mol Med Rep 2018; 17:8212-8220. [PMID: 29658608 PMCID: PMC5983996 DOI: 10.3892/mmr.2018.8864] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 11/02/2017] [Indexed: 01/01/2023] Open
Abstract
Hypoxia is a serious stress state. The nervous system is less tolerant to hypoxia, and cell death due to hypoxia is irreversible. With the incidence of cardiovascular disease gradually increasing, the sudden cardiac death rate is additionally increasing. Although cardiopulmonary resuscitation (CPR) is an important development, recovery is frequently poor. In a successful recovery population, ~40% of the population was in a vegetative state or subsequently succumbed to their condition, and ~20% had brain damage. Therefore, the recovery of the brain is of particular importance in CPR. Immune disorders are one of the major mechanisms of cerebral resuscitation following CPR. Studies have demonstrated that induced pluripotent stem cell-derived mesenchymal stem cells (IPSC-MSCs) have a strong immune regulatory effect during tissue repair and anti-inflammatory effects. IPSC-MSCs may inhibit the inflammatory response by means of the inflammatory reaction network to improve brain function following CPR, although the cellular and molecular mechanisms remain unclear. Macrophages are a bridge between innate immune and specific immune responses in the body; therefore, it was hypothesized that macrophages may be the important effector cell of the role of IPSC-MSCs in improving brain function following recovery of spontaneous respiration and circulation subsequent to cardiopulmonary resuscitation. In the present study, IPSC-MSCs were applied to the oxygen and glucose deprivation (OGD) model. It was observed that intervention with IPSC-MSCs was able to alter the polarization direction of macrophages. The difference in the proportions of M1 and M2 macrophages was statistically significant at 6, 12, 24 and 48 h (P=0.037, P<0.05) in the OGD + IPSC-MSCs group (M1, 33.48±5.6%; M2, 50.84±6.9%) and in the OGD group (M1, 83.55±7.3%; M2, 11.41±3.2%), and over time this trend was more obvious. The polarization direction of macrophages is associated with the neurogenic locus notch homolog protein 1 (Notch-1) signaling pathway. In conclusion, it was observed that IPSC-MSCs may be associated with altered macrophage polarization, which may be accomplished by inhibiting the Notch-1 signaling pathway.
Collapse
Affiliation(s)
- Yi Yu
- Emergency Department, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Dongping Wang
- Organ Transplant Center, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Hui Li
- Emergency Department, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Yujie Liu
- Department of Breast Surgery and Key Laboratory of Malignant Tumor Gene Regulation and Target Therapy of Guangdong Higher Education Institutes, Sun Yat‑Sen Memorial Hospital, Sun Yat‑Sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Zhao Xiang
- Emergency Department, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Junlin Wu
- Emergency Department, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| | - Xiaoli Jing
- Emergency Department, The First Affiliated Hospital of Sun Yat‑sen University, Guangzhou, Guangdong 510080, P.R. China
| |
Collapse
|
39
|
Shafit-Zagardo B, Gruber RC, DuBois JC. The role of TAM family receptors and ligands in the nervous system: From development to pathobiology. Pharmacol Ther 2018. [PMID: 29514053 DOI: 10.1016/j.pharmthera.2018.03.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Tyro3, Axl, and Mertk, referred to as the TAM family of receptor tyrosine kinases, are instrumental in maintaining cell survival and homeostasis in mammals. TAM receptors interact with multiple signaling molecules to regulate cell migration, survival, phagocytosis and clearance of metabolic products and cell debris called efferocytosis. The TAMs also function as rheostats to reduce the expression of proinflammatory molecules and prevent autoimmunity. All three TAM receptors are activated in a concentration-dependent manner by the vitamin K-dependent growth arrest-specific protein 6 (Gas6). Gas6 and the TAMs are abundantly expressed in the nervous system. Gas6, secreted by neurons and endothelial cells, is the sole ligand for Axl. ProteinS1 (ProS1), another vitamin K-dependent protein functions mainly as an anti-coagulant, and independent of this function can activate Tyro3 and Mertk, but not Axl. This review will focus on the role of the TAM receptors and their ligands in the nervous system. We highlight studies that explore the function of TAM signaling in myelination, the visual cortex, neural cancers, and multiple sclerosis (MS) using Gas6-/- and TAM mutant mice models.
Collapse
Affiliation(s)
- Bridget Shafit-Zagardo
- Albert Einstein College of Medicine, Department of Pathology, 1300 Morris Park Avenue, Bronx, NY 10461, United States.
| | - Ross C Gruber
- Sanofi, Neuroinflammation and MS Research, 49 New York Ave, Framingham, MA 01701, United States
| | - Juwen C DuBois
- Albert Einstein College of Medicine, Department of Pathology, 1300 Morris Park Avenue, Bronx, NY 10461, United States
| |
Collapse
|
40
|
Chen YL, Tsai YT, Chao TT, Wu YN, Chen MC, Lin YH, Liao CH, Chou SSP, Chiang HS. DAPK and CIP2A are involved in GAS6/AXL-mediated Schwann cell proliferation in a rat model of bilateral cavernous nerve injury. Oncotarget 2018; 9:6402-6415. [PMID: 29464081 PMCID: PMC5814221 DOI: 10.18632/oncotarget.23978] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Accepted: 10/28/2017] [Indexed: 12/13/2022] Open
Abstract
Purpose Impotence is one of the major complications occurring in prostate cancer patients after radical prostectomy (RP). Self-repair of the injured nerve has been observed in animal models and in patients after RP. However, the downstream signalling is not well documented. Here, we found that the DAPK/CIP2A complex is involved in GAS6/AXL-related Schwann cell proliferation. Materials and Methods The 3 groups were a sham group, a 14-day post-bilateral cavernous nerve injury (BCNI) group and a 28-day post-BCNI group. Erectile function was assessed and immunohistochemistry was performed. The rat Schwann cell RSC96 line was chosen for gene knockdown, cell viability, western blot, immunofluorescence and co-immunoprecipitation assays. Results The intracavernosal pressure was low on the 14th day after BCNI and partially increased by the 28th day. GAS6 and p-AXL expression gradually increased in the cavernous nerve after BCNI. RSC96 cells incubated with a GAS6 ligand showed increased levels of p-ERK1/2 and p-AKT. Moreover, DAPK and CIP2A.p-AXL and p-DAPK and CIP2A complexes were identified by both immunoblotting and co-immunoprecipitation. Conclusion The DAPK/CIP2A complex is involved in GAS6/AXL-related Schwann cell proliferation. CIP2A inhibits PP2A activity, which results in p-DAPK(S308) maintenance and promotes Schwann cell proliferation. CIP2A is a potential target for the treatment of nerve injury after RP.
Collapse
Affiliation(s)
- Yen-Lin Chen
- Department of Pathology, Cardinal Tien Hospital, New Taipei, Taiwan.,Department of Chemistry, Fu-Jen Catholic University, New Taipei, Taiwan.,Graduate Institute of Biomedical and Pharmaceutical Science, Fu-Jen Catholic University, New Taipei, Taiwan
| | - Yi-Ting Tsai
- Department of Pathology, Cardinal Tien Hospital, New Taipei, Taiwan
| | - Ting-Ting Chao
- Medical Research Center, Cardinal Tien Hospital, New Taipei, Taiwan
| | - Yi-No Wu
- School of Medicine, Fu-Jen Catholic University, New Taipei, Taiwan
| | - Meng-Chuan Chen
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Ying-Hung Lin
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu-Jen Catholic University, New Taipei, Taiwan
| | - Chun-Hou Liao
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu-Jen Catholic University, New Taipei, Taiwan.,Division of Urology, Department of Surgery, Cardinal Tien Hospital, New Taipei, Taiwan
| | | | - Han-Sun Chiang
- Graduate Institute of Biomedical and Pharmaceutical Science, Fu-Jen Catholic University, New Taipei, Taiwan.,Division of Urology, Department of Surgery, Cardinal Tien Hospital, New Taipei, Taiwan
| |
Collapse
|
41
|
Wisse LE, Penning R, Zaal EA, van Berkel CGM, Ter Braak TJ, Polder E, Kenney JW, Proud CG, Berkers CR, Altelaar MAF, Speijer D, van der Knaap MS, Abbink TEM. Proteomic and Metabolomic Analyses of Vanishing White Matter Mouse Astrocytes Reveal Deregulation of ER Functions. Front Cell Neurosci 2017; 11:411. [PMID: 29375313 PMCID: PMC5770689 DOI: 10.3389/fncel.2017.00411] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 12/07/2017] [Indexed: 12/20/2022] Open
Abstract
Vanishing white matter (VWM) is a leukodystrophy with predominantly early-childhood onset. Affected children display various neurological signs, including ataxia and spasticity, and die early. VWM patients have bi-allelic mutations in any of the five genes encoding the subunits of the eukaryotic translation factor 2B (eIF2B). eIF2B regulates protein synthesis rates under basal and cellular stress conditions. The underlying molecular mechanism of how mutations in eIF2B result in VWM is unknown. Previous studies suggest that brain white matter astrocytes are primarily affected in VWM. We hypothesized that the translation rate of certain astrocytic mRNAs is affected by the mutations, resulting in astrocytic dysfunction. Here we subjected primary astrocyte cultures of wild type (wt) and VWM (2b5ho) mice to pulsed labeling proteomics based on stable isotope labeling with amino acids in cell culture (SILAC) with an L-azidohomoalanine (AHA) pulse to select newly synthesized proteins. AHA was incorporated into newly synthesized proteins in wt and 2b5ho astrocytes with similar efficiency, without affecting cell viability. We quantified proteins synthesized in astrocytes of wt and 2b5ho mice. This proteomic profiling identified a total of 80 proteins that were regulated by the eIF2B mutation. We confirmed increased expression of PROS1 in 2b5ho astrocytes and brain. A DAVID enrichment analysis showed that approximately 50% of the eIF2B-regulated proteins used the secretory pathway. A small-scale metabolic screen further highlighted a significant change in the metabolite 6-phospho-gluconate, indicative of an altered flux through the pentose phosphate pathway (PPP). Some of the proteins migrating through the secretory pathway undergo oxidative folding reactions in the endoplasmic reticulum (ER), which produces reactive oxygen species (ROS). The PPP produces NADPH to remove ROS. The proteomic and metabolomics data together suggest a deregulation of ER function in 2b5ho mouse astrocytes.
Collapse
Affiliation(s)
- Lisanne E Wisse
- Pediatrics, VU University Medical Center, Amsterdam, Netherlands
| | - Renske Penning
- Biomolecular Mass Spectrometry and Proteomics Group, Utrecht Institute for Pharmaceutical Sciences, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Esther A Zaal
- Biomolecular Mass Spectrometry and Proteomics Group, Utrecht Institute for Pharmaceutical Sciences, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | | | - Timo J Ter Braak
- Pediatrics, VU University Medical Center, Amsterdam, Netherlands
| | - Emiel Polder
- Pediatrics, VU University Medical Center, Amsterdam, Netherlands
| | - Justin W Kenney
- Centre for Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Christopher G Proud
- Centre for Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Celia R Berkers
- Biomolecular Mass Spectrometry and Proteomics Group, Utrecht Institute for Pharmaceutical Sciences, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Maarten A F Altelaar
- Biomolecular Mass Spectrometry and Proteomics Group, Utrecht Institute for Pharmaceutical Sciences, Bijvoet Center for Biomolecular Research, Utrecht University, Utrecht, Netherlands
| | - Dave Speijer
- Medical Biochemistry, Academic Medical Center, Amsterdam, Netherlands
| | | | - Truus E M Abbink
- Pediatrics, VU University Medical Center, Amsterdam, Netherlands
| |
Collapse
|
42
|
Dumont CM, Piselli J, Temple S, Dai G, Thompson DM. Endothelial Cells Exposed to Fluid Shear Stress Support Diffusion Based Maturation of Adult Neural Progenitor Cells. Cell Mol Bioeng 2017; 11:117-130. [PMID: 31719881 DOI: 10.1007/s12195-017-0516-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 11/18/2017] [Indexed: 12/21/2022] Open
Abstract
Introduction The neural stem cell (NSC) niche is a highly complex cellular and biochemical milieu supporting proliferating NSCs and neural progenitor cells (NPCs) with close apposition to the vasculature, primarily comprised of endothelial cells (ECs). Current in vitro models of the niche incorporate EC-derived factors, but do not reflect the physiologically relevant hemodynamic state of the ECs or the spatial resolution observed between cells within the niche. Methods In this work, we developed a novel in vitro model of the niche that (1) incorporates ECs cultured with fluid shear stress and (2) fosters paracrine cytokine gradients between ECs and NSCs in a spatiotemporal configuration mimicking the cytoarchitecture of the subventricular niche. A modified cone and plate viscometer was used to generate a shear stress of 10 dynes cm-2 for ECs cultured on a membrane, while statically cultured NPCs are 10 or 1000 μm below the ECs. Results NPCs cultured within 10 μm of dynamic ECs exhibit increased PSA-NCAM+ and OLIG2+ cells compared to progenitors in all other culture regimes and the hemodynamic EC phenotype results in distinct progeny phenotypes. This co-culture regime yields greater release of pro-neurogenic factors, suggesting a potential mechanism for the observed progenitor maturation. Conclusions Based on these results, models incorporating ECs exposed to shear stress allow for paracrine signaling gradients and regulate NPC lineage progression with appropriate niche spatial resolution occurring at 10 μm. This model could be used to evaluate cellular or pharmacological interactions within the healthy, diseased, or aged brain.
Collapse
Affiliation(s)
- C M Dumont
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180 USA.,Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180 USA
| | - J Piselli
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180 USA.,Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180 USA
| | - S Temple
- Neural Stem Cell Institute, Rensselaer, NY 12144 USA
| | - G Dai
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180 USA.,Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180 USA
| | - D M Thompson
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, Troy, NY 12180 USA.,Center for Biotechnology & Interdisciplinary Studies, Rensselaer Polytechnic Institute, Troy, NY 12180 USA
| |
Collapse
|
43
|
Ray AK, DuBois JC, Gruber RC, Guzik HM, Gulinello ME, Perumal G, Raine C, Kozakiewicz L, Williamson J, Shafit-Zagardo B. Loss of Gas6 and Axl signaling results in extensive axonal damage, motor deficits, prolonged neuroinflammation, and less remyelination following cuprizone exposure. Glia 2017; 65:2051-2069. [PMID: 28925029 DOI: 10.1002/glia.23214] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 08/10/2017] [Accepted: 08/11/2017] [Indexed: 01/08/2023]
Abstract
The TAM (Tyro3, Axl, and MerTK) family of receptor tyrosine kinases (RTKs) and their ligands, Gas6 and ProS1, are important for innate immune responses and central nervous system (CNS) homeostasis. While only Gas6 directly activates Axl, ProS1 activation of Tyro3/MerTK can indirectly activate Axl through receptor heterodimerization. Therefore, we generated Gas6-/- Axl-/- double knockout (DKO) mice to specifically examine the contribution of this signaling axis while retaining ProS1 signaling through Tyro3 and MerTK. We found that naïve young adult DKO and WT mice have comparable myelination and equal numbers of axons and oligodendrocytes in the corpus callosum. Using the cuprizone model of demyelination/remyelination, transmission electron microscopy revealed extensive axonal swellings containing autophagolysosomes and multivesicular bodies, and fewer myelinated axons in brains of DKO mice at 3-weeks recovery from a 6-week cuprizone diet. Analysis of immunofluorescent staining demonstrated more SMI32+ and APP+ axons and less myelin in the DKO mice. There were no significant differences in the number of GFAP+ astrocytes or Iba1+ microglia/macrophages between the groups of mice. However, at 6-weeks cuprizone and recovery, DKO mice had increased proinflammatory cytokine and altered suppressor of cytokine signaling (SOCS) mRNA expression supporting a role for Gas6-Axl signaling in proinflammatory cytokine suppression. Significant motor deficits in DKO mice relative to WT mice on cuprizone were also observed. These data suggest that Gas6-Axl signaling plays an important role in maintaining axonal integrity and regulating and reducing CNS inflammation that cannot be compensated for by ProS1/Tyro3/MerTK signaling.
Collapse
Affiliation(s)
- Alex K Ray
- Department of Pathology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, 10461
| | - Juwen C DuBois
- Department of Pathology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, 10461
| | - Ross C Gruber
- Neuroimmunology and MS Research, Sanofi, Framingham, Massachusetts, 01701
| | - Hillary M Guzik
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York, 10461
| | - Maria E Gulinello
- Rodent Behavioral Core, Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, 10461
| | - Geoffrey Perumal
- Department of Anatomy and Structural Biology, Albert Einstein College of Medicine, Bronx, New York, 10461
| | - Cedric Raine
- Department of Pathology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, 10461
| | - Lauren Kozakiewicz
- Department of Pathology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, 10461
| | - Julie Williamson
- Department of Pathology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, 10461
| | - Bridget Shafit-Zagardo
- Department of Pathology, Albert Einstein College of Medicine, Montefiore Medical Center, Bronx, New York, 10461
| |
Collapse
|
44
|
Tong LS, Shao AW, Ou YB, Guo ZN, Manaenko A, Dixon BJ, Tang J, Lou M, Zhang JH. Recombinant Gas6 augments Axl and facilitates immune restoration in an intracerebral hemorrhage mouse model. J Cereb Blood Flow Metab 2017; 37:1971-1981. [PMID: 27389179 PMCID: PMC5464693 DOI: 10.1177/0271678x16658490] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Axl, a tyrosine kinase receptor, was recently identified as an essential component regulating innate immune response. Suppressor of cytokine signaling 1 and suppressor of cytokine signaling 3 are potent Axl-inducible negative inflammatory regulators. This study investigated the role of Axl signaling pathway in immune restoration in an autologous blood-injection mouse model of intracerebral hemorrhage. Recombinant growth arrest-specific 6 (Gas6) and R428 were administrated as specific agonist and antagonist. In vivo knockdown of Axl or suppressor of cytokine signaling 1 and suppressor of cytokine signaling 3 by siRNA was applied. After intracerebral hemorrhage, the expression of endogenous Axl, soluble Axl, and Gas6 was increased, whereas the expression of suppressor of cytokine signaling 1 and suppressor of cytokine signaling 3 was inhibited. Recombinant growth arrest-specific 6 administration alleviated brain edema and improved neurobehavioral performances. Moreover, enhanced Axl phosphorylation with cleavage of soluble Axl (sAxl), and an upregulation of suppressor of cytokine signaling 1 and suppressor of cytokine signaling 3 were observed. In vivo knockdown of Axl and R428 administration both abolished the effect of recombinant growth arrest-specific 6 on brain edema and also decreased the expression suppressor of cytokine signaling 1 and suppressor of cytokine signaling 3. In vivo knockdown of suppressor of cytokine signaling 1 and suppressor of cytokine signaling 3 aggravated cytokine releasing despite of recombinant growth arrest-specific 6. In conclusion, Axl plays essential role in immune restoration after intracerebral hemorrhage. And recombinant growth arrest-specific 6 attenuated brain injury after intracerebral hemorrhage, probably by enhancing Axl phosphorylation and production of suppressor of cytokine signaling 1 and suppressor of cytokine signaling 3.
Collapse
Affiliation(s)
- Lu-Sha Tong
- 1 Department of Anesthesiology, School of Medicine, Loma Linda University, CA, USA.,2 Department of Neurology, School of Medicine, the 2nd Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - An-Wen Shao
- 1 Department of Anesthesiology, School of Medicine, Loma Linda University, CA, USA.,3 Department of Neurosurgery, School of Medicine, the 2nd Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Yi-Bo Ou
- 1 Department of Anesthesiology, School of Medicine, Loma Linda University, CA, USA.,4 Department of Neurosurgery, Tong-ji Hospital, Wuhan, China
| | - Zhen-Ni Guo
- 1 Department of Anesthesiology, School of Medicine, Loma Linda University, CA, USA.,5 Department of Neurology, Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Anatol Manaenko
- 1 Department of Anesthesiology, School of Medicine, Loma Linda University, CA, USA
| | - Brandon J Dixon
- 1 Department of Anesthesiology, School of Medicine, Loma Linda University, CA, USA
| | - Jiping Tang
- 2 Department of Neurology, School of Medicine, the 2nd Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Min Lou
- 2 Department of Neurology, School of Medicine, the 2nd Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - John H Zhang
- 1 Department of Anesthesiology, School of Medicine, Loma Linda University, CA, USA
| |
Collapse
|
45
|
Growth Arrest-Specific 6 Enhances the Suppressive Function of CD4 +CD25 + Regulatory T Cells Mainly through Axl Receptor. Mediators Inflamm 2017; 2017:6848430. [PMID: 28270700 PMCID: PMC5320320 DOI: 10.1155/2017/6848430] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 11/07/2016] [Accepted: 12/05/2016] [Indexed: 11/17/2022] Open
Abstract
Background. Growth arrest-specific (Gas) 6 is one of the endogenous ligands of TAM receptors (Tyro3, Axl, and Mertk), and its role as an immune modulator has been recently emphasized. Naturally occurring CD4+CD25+ regulatory T cells (Tregs) are essential for the active suppression of autoimmunity. The present study was designed to investigate whether Tregs express TAM receptors and the potential role of Gas6-TAM signal in regulating the suppressive function of Tregs. Methods. The protein and mRNA levels of TAM receptors were determined by using Western blot, immunofluorescence, flow cytometry, and RT-PCR. Then, TAM receptors were silenced using targeted siRNA or blocked with specific antibody. The suppressive function of Tregs was assessed by using a CFSE-based T cell proliferation assay. Flow cytometry was used to determine the expression of Foxp3 and CTLA4 whereas cytokines secretion levels were measured by ELISA assay. Results. Tregs express both Axl and Mertk receptors. Gas6 increases the suppressive function of Tregs in vitro and in mice. Both Foxp3 and CTLA-4 expression on Tregs are enhanced after Gas6 stimulation. Gas6 enhances the suppressive activity of Tregs mainly through Axl receptor. Conclusion. Gas6 has a direct effect on the functions of CD4+CD25+Tregs mainly through its interaction with Axl receptor.
Collapse
|
46
|
Akkermann R, Aprico A, Perera AA, Bujalka H, Cole AE, Xiao J, Field J, Kilpatrick TJ, Binder MD. The TAM receptor Tyro3 regulates myelination in the central nervous system. Glia 2017; 65:581-591. [PMID: 28145605 DOI: 10.1002/glia.23113] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 12/05/2016] [Accepted: 12/20/2016] [Indexed: 11/12/2022]
Abstract
Myelin is an essential component of the mammalian nervous system, facilitating rapid conduction of electrical impulses by axons, as well as providing trophic support to neurons. Within the central nervous system, the oligodendrocyte is the specialized neural cell responsible for producing myelin by a process that is thought to be regulated by both activity dependent and independent mechanisms but in incompletely understood ways. We have previously identified that the protein Gas6, a ligand for a family of tyrosine kinase receptors known as the TAM (Tyro3, Axl, and Mertk) receptors, directly increases oligodendrocyte induced myelination in vitro. Gas6 can bind to and activate all three TAM receptors, but the high level of expression of Tyro3 on oligodendrocytes makes this receptor the principal candidate for transducing the pro-myelinating effect of Gas6. In this study, we establish that in the absence of Tyro3, the pro-myelinating effect of Gas6 is lost, that developmental myelination is delayed and that the myelin produced is thinner than normal. We show that this effect is specific to the myelination process and not due to changes in the proliferation or differentiation of oligodendrocyte precursor cells. We have further demonstrated that the reduction in myelination is due to the loss of Tyro3 on oligodendrocytes, and this effect may be mediated by activation of Erk1. Collectively, our findings indicate the critical importance of Tyro3 in potentiating central nervous system myelination. GLIA 2017 GLIA 2017;65:581-591.
Collapse
Affiliation(s)
- Rainer Akkermann
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Andrea Aprico
- The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade (Cnr Genetics Lane), Parkville, Victoria, 3052, Australia
| | - Ashwyn A Perera
- The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade (Cnr Genetics Lane), Parkville, Victoria, 3052, Australia
| | - Helena Bujalka
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Alistair E Cole
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Junhua Xiao
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, 3010, Australia
| | - Judith Field
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, 3010, Australia.,The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade (Cnr Genetics Lane), Parkville, Victoria, 3052, Australia
| | - Trevor J Kilpatrick
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, 3010, Australia.,The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade (Cnr Genetics Lane), Parkville, Victoria, 3052, Australia
| | - Michele D Binder
- Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, 3010, Australia.,The Florey Institute of Neuroscience and Mental Health, 30 Royal Parade (Cnr Genetics Lane), Parkville, Victoria, 3052, Australia
| |
Collapse
|
47
|
Protective Effects of Pterostilbene Against Myocardial Ischemia/Reperfusion Injury in Rats. Inflammation 2017; 40:578-588. [DOI: 10.1007/s10753-016-0504-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
48
|
The Gas6/TAM System and Multiple Sclerosis. Int J Mol Sci 2016; 17:ijms17111807. [PMID: 27801848 PMCID: PMC5133808 DOI: 10.3390/ijms17111807] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Revised: 10/22/2016] [Accepted: 10/26/2016] [Indexed: 01/25/2023] Open
Abstract
Growth arrest specific 6 (Gas6) is a multimodular circulating protein, the biological actions of which are mediated by the interaction with three transmembrane tyrosine kinase receptors: Tyro3, Axl, and MerTK, collectively named TAM. Over the last few decades, many progresses have been done in the understanding of the biological activities of this highly pleiotropic system, which plays a role in the regulation of immune response, inflammation, coagulation, cell growth, and clearance of apoptotic bodies. Recent findings have further related Gas6 and TAM receptors to neuroinflammation in general and, specifically, to multiple sclerosis (MS). In this paper, we review the biology of the Gas6/TAM system and the current evidence supporting its potential role in the pathogenesis of MS.
Collapse
|
49
|
Torii T, Yamauchi J. Gas6-Tyro3 signaling is required for Schwann cell myelination and possible remyelination. Neural Regen Res 2016; 11:215-6. [PMID: 27073361 PMCID: PMC4810972 DOI: 10.4103/1673-5374.177714] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Tomohiro Torii
- Department of Pharmacology, National Center for Child Health and Development, Setagaya, Tokyo, Japan
| | - Junji Yamauchi
- Department of Pharmacology, National Center for Child Health and Development, Setagaya, Tokyo, Japan; Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Bunkyo, Tokyo, Japan
| |
Collapse
|
50
|
Healy LM, Perron G, Won SY, Michell-Robinson MA, Rezk A, Ludwin SK, Moore CS, Hall JA, Bar-Or A, Antel JP. MerTK Is a Functional Regulator of Myelin Phagocytosis by Human Myeloid Cells. THE JOURNAL OF IMMUNOLOGY 2016; 196:3375-84. [PMID: 26962228 DOI: 10.4049/jimmunol.1502562] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Accepted: 01/28/2016] [Indexed: 12/22/2022]
Abstract
Multifocal inflammatory lesions featuring destruction of lipid-rich myelin are pathologic hallmarks of multiple sclerosis. Lesion activity is assessed by the extent and composition of myelin uptake by myeloid cells present in such lesions. In the inflamed CNS, myeloid cells are comprised of brain-resident microglia, an endogenous cell population, and monocyte-derived macrophages, which infiltrate from the systemic compartment. Using microglia isolated from the adult human brain, we demonstrate that myelin phagocytosis is dependent on the polarization state of the cells. Myelin ingestion is significantly enhanced in cells exposed to TGF-β compared with resting basal conditions and markedly reduced in classically activated polarized cells. Transcriptional analysis indicated that TGF-β-treated microglia closely resembled M0 cells. The tyrosine kinase phagocytic receptor MerTK was one of the most upregulated among a select number of differentially expressed genes in TGF-β-treated microglia. In contrast, MerTK and its known ligands, growth arrest-specific 6 and Protein S, were downregulated in classically activated cells. MerTK expression and myelin phagocytosis were higher in CNS-derived microglia than observed in monocyte-derived macrophages, both basally and under all tested polarization conditions. Specific MerTK inhibitors reduced myelin phagocytosis and the resultant anti-inflammatory biased cytokine responses for both cell types. Defining and modulating the mechanisms that regulate myelin phagocytosis has the potential to impact lesion and disease evolution in multiple sclerosis. Relevant effects would include enhancing myelin clearance, increasing anti-inflammatory molecule production by myeloid cells, and thereby permitting subsequent tissue repair.
Collapse
Affiliation(s)
- Luke M Healy
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Gabrielle Perron
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - So-Yoon Won
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Montreal, Quebec H3A 2B4, Canada
| | | | - Ayman Rezk
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Samuel K Ludwin
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Montreal, Quebec H3A 2B4, Canada; Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario K7L 3N6, Canada
| | - Craig S Moore
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University, St. John's, Newfoundland A1B 3V6, Canada; and
| | - Jeffery A Hall
- Department of Neurosurgery, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Amit Bar-Or
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Jack P Antel
- Neuroimmunology Unit, Montreal Neurological Institute, McGill University, Montreal, Quebec H3A 2B4, Canada;
| |
Collapse
|