1
|
Villano G, Pontisso P. Protease activated receptor 2 as a novel druggable target for the treatment of metabolic dysfunction-associated fatty liver disease and cancer. Front Immunol 2024; 15:1397441. [PMID: 39464875 PMCID: PMC11502361 DOI: 10.3389/fimmu.2024.1397441] [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: 03/07/2024] [Accepted: 09/23/2024] [Indexed: 10/29/2024] Open
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is spreading worldwide, largely due to unhealthy lifestyles that contribute to the rise in diabetes, metabolic syndrome, and obesity. In this situation, the progression of injury to metabolic steatohepatitis can evolve to cirrhosis and, eventually, to hepatocellular carcinoma (HCC). It is well known that serine protease enzymes with different functions in cellular homeostasis act as signaling molecules that regulate liver inflammation by activating the protease-activated receptors (PARs) family members, expressed on the cellular plasma membrane. Among them, PAR2 plays a central role in the activation of signaling pathways in response to changes in the extracellular microenvironment. Experimental data have provided evidence that PAR2 is involved not only in inflammatory response but also in insulin resistance, lipid metabolism, and cancer. The major aims of this narrative review are addressed to assess PAR2 involvement in inflammation, metabolism, and liver disease progression and to explore possible therapeutic strategies, based on PAR2 inhibition, in order to prevent its biological effects in the context of MAFLD and cancer.
Collapse
Affiliation(s)
- Gianmarco Villano
- Department of Surgical, Oncological and Gastroenterological Sciences, University of Padova, Padova, Italy
| | | |
Collapse
|
2
|
Xu K, Wang L, Lin M, He G. Update on protease-activated receptor 2 in inflammatory and autoimmune dermatological diseases. Front Immunol 2024; 15:1449126. [PMID: 39364397 PMCID: PMC11446762 DOI: 10.3389/fimmu.2024.1449126] [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: 06/14/2024] [Accepted: 09/03/2024] [Indexed: 10/05/2024] Open
Abstract
Protease-activated receptor 2 (PAR2) is a cell-surface receptor expressed in various cell types, including keratinocytes, neurons, immune and inflammatory cells. Activation of PAR2, whether via its canonical or biased pathways, triggers a series of signaling cascades that mediate numerous functions. This review aims to highlight the emerging roles and interactions of PAR2 in different skin cells. It specifically summarizes the latest insights into the roles of PAR2 in skin conditions such as atopic dermatitis (AD), psoriasis, vitiligo and melasma. It also considers these roles from the perspective of the cutaneous microenvironment in relation to other inflammatory and autoimmune dermatological disorders. Additionally, the review explores PAR2's involvement in associated comorbidities from both cutaneous and extracutaneous diseases. Therefore, PAR2 may serve as a key target for interactions among various cells within the local skin environment.
Collapse
Affiliation(s)
- Kejia Xu
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease Related Molecular Network and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Lin Wang
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease Related Molecular Network and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Mao Lin
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
| | - Gu He
- Department of Dermatology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease Related Molecular Network and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Eftekhari R, Ewanchuk BW, Rawji KS, Yates RM, Noorbakhsh F, Kuipers HF, Hollenberg MD. Blockade of Proteinase-Activated Receptor 2 (PAR2) Attenuates Neuroinflammation in Experimental Autoimmune Encephalomyelitis. J Pharmacol Exp Ther 2024; 388:12-22. [PMID: 37699708 DOI: 10.1124/jpet.123.001685] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/17/2023] [Accepted: 08/08/2023] [Indexed: 09/14/2023] Open
Abstract
Proteinase-activated receptor-2 (PAR2), which modulates inflammatory responses, is elevated in the central nervous system in multiple sclerosis (MS) and in its murine model, experimental autoimmune encephalomyelitis (EAE). In PAR2-null mice, disease severity of EAE is markedly diminished. We therefore tested whether inhibiting PAR2 activation in vivo might be a viable strategy for the treatment of MS. Using the EAE model, we show that a PAR2 antagonist, the pepducin palmitoyl-RSSAMDENSEKKRKSAIK-amide (P2pal-18S), attenuates EAE progression by affecting immune cell function. P2pal-18S treatment markedly diminishes disease severity and reduces demyelination, as well as the infiltration of T-cells and macrophages into the central nervous system. Moreover, P2pal-18S decreases granulocyte-macrophage colony-stimulating factor (GM-CSF) production and T-cell activation in cultured splenocytes and prevents macrophage polarization in vitro. We conclude that PAR2 plays a key role in regulating neuroinflammation in EAE and that PAR2 antagonists represent promising therapeutic agents for treating MS and other neuroinflammatory diseases. SIGNIFICANCE STATEMENT: Proteinase-activated receptor-2 modulates inflammatory responses and is increased in multiple sclerosis lesions. We show that the proteinase-activated receptor-2 antagonist palmitoyl-RSSAMDENSEKKRKSAIK-amide reduces disease in the murine experimental autoimmune encephalomyelitis model of multiple sclerosis by inhibiting T-cell and macrophage activation and infiltration into the central nervous system, making it a potential treatment for multiple sclerosis.
Collapse
Affiliation(s)
- Rahil Eftekhari
- Department of Physiology & Pharmacology (R.E., M.D.H.), Department of Medicine (R.E., M.D.H.), Department of Clinical Neurosciences (R.E., K.S.R., H.F.K.), Department of Biochemistry and Molecular Biology (B.W.E., R.M.Y.), Department of Comparative Biology and Experimental Medicine (B.W.E., R.M.Y.), and Department of Cell Biology and Anatomy (H.F.K.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran (R.E., F.N.)
| | - Benjamin W Ewanchuk
- Department of Physiology & Pharmacology (R.E., M.D.H.), Department of Medicine (R.E., M.D.H.), Department of Clinical Neurosciences (R.E., K.S.R., H.F.K.), Department of Biochemistry and Molecular Biology (B.W.E., R.M.Y.), Department of Comparative Biology and Experimental Medicine (B.W.E., R.M.Y.), and Department of Cell Biology and Anatomy (H.F.K.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran (R.E., F.N.)
| | - Khalil S Rawji
- Department of Physiology & Pharmacology (R.E., M.D.H.), Department of Medicine (R.E., M.D.H.), Department of Clinical Neurosciences (R.E., K.S.R., H.F.K.), Department of Biochemistry and Molecular Biology (B.W.E., R.M.Y.), Department of Comparative Biology and Experimental Medicine (B.W.E., R.M.Y.), and Department of Cell Biology and Anatomy (H.F.K.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran (R.E., F.N.)
| | - Robin M Yates
- Department of Physiology & Pharmacology (R.E., M.D.H.), Department of Medicine (R.E., M.D.H.), Department of Clinical Neurosciences (R.E., K.S.R., H.F.K.), Department of Biochemistry and Molecular Biology (B.W.E., R.M.Y.), Department of Comparative Biology and Experimental Medicine (B.W.E., R.M.Y.), and Department of Cell Biology and Anatomy (H.F.K.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran (R.E., F.N.)
| | - Farshid Noorbakhsh
- Department of Physiology & Pharmacology (R.E., M.D.H.), Department of Medicine (R.E., M.D.H.), Department of Clinical Neurosciences (R.E., K.S.R., H.F.K.), Department of Biochemistry and Molecular Biology (B.W.E., R.M.Y.), Department of Comparative Biology and Experimental Medicine (B.W.E., R.M.Y.), and Department of Cell Biology and Anatomy (H.F.K.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran (R.E., F.N.)
| | - Hedwich F Kuipers
- Department of Physiology & Pharmacology (R.E., M.D.H.), Department of Medicine (R.E., M.D.H.), Department of Clinical Neurosciences (R.E., K.S.R., H.F.K.), Department of Biochemistry and Molecular Biology (B.W.E., R.M.Y.), Department of Comparative Biology and Experimental Medicine (B.W.E., R.M.Y.), and Department of Cell Biology and Anatomy (H.F.K.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran (R.E., F.N.)
| | - Morley D Hollenberg
- Department of Physiology & Pharmacology (R.E., M.D.H.), Department of Medicine (R.E., M.D.H.), Department of Clinical Neurosciences (R.E., K.S.R., H.F.K.), Department of Biochemistry and Molecular Biology (B.W.E., R.M.Y.), Department of Comparative Biology and Experimental Medicine (B.W.E., R.M.Y.), and Department of Cell Biology and Anatomy (H.F.K.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran (R.E., F.N.)
| |
Collapse
|
4
|
Soeda M, Ohka S, Nishizawa D, Iseki M, Yamaguchi K, Arita H, Hanaoka K, Kato J, Ogawa S, Hiranuma A, Hasegawa J, Nakayama K, Ebata Y, Hayashida M, Ichinohe T, Fukuda KI, Ikeda K. Single-Nucleotide Polymorphisms of the PAR2 and IL-17A Genes Are Significantly Associated with Chronic Pain. Int J Mol Sci 2023; 24:17627. [PMID: 38139455 PMCID: PMC10744199 DOI: 10.3390/ijms242417627] [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: 10/24/2023] [Revised: 12/10/2023] [Accepted: 12/14/2023] [Indexed: 12/24/2023] Open
Abstract
Patients with chronic pain are affected psychologically and socially. There are also individual differences in treatment efficacy. Insufficient research has been conducted on genetic polymorphisms that are related to individual differences in the susceptibility to chronic pain. Autoimmune disorders can lead to inflammation and chronic pain; therefore, we focused on the autoimmune-related protease-activated receptor 2 (PAR2/F2RL1) and interleukin 17A (IL-17A/IL17A) genes. PAR2 and IL-17A are associated with autoimmune diseases that lead to chronic pain, and PAR2 regulates T-helper (Th) cell activation and differentiation. We hypothesized that the PAR2 and IL-17A genes are associated with chronic pain. The present study used a case-control design to statistically examine associations between genetic polymorphisms and the vulnerability to chronic pain. The rs2243057 polymorphism of the PAR2 gene and rs3819025 polymorphism of the IL-17A gene were previously reported to be associated with pain- or autoimmune-related phenotypes. Thus, these polymorphisms were investigated in the present study. We found that both rs2243057 and rs3819025 were significantly associated with a susceptibility to chronic pain. The present findings revealed autoimmune-related genetic factors that are involved in individual differences in chronic pain, further aiding understanding of the pathomechanism that underlies chronic pain and possibly contributing to future personalized medicine.
Collapse
Affiliation(s)
- Moe Soeda
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; (M.S.); (S.O.); (D.N.)
- Department of Oral Health and Clinical Science, Tokyo Dental College, Tokyo 101-0061, Japan
| | - Seii Ohka
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; (M.S.); (S.O.); (D.N.)
| | - Daisuke Nishizawa
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; (M.S.); (S.O.); (D.N.)
| | - Masako Iseki
- Department of Anesthesiology & Pain Medicine, Juntendo University School of Medicine, Tokyo 113-8431, Japan; (M.I.)
| | - Keisuke Yamaguchi
- Department of Anesthesiology & Pain Medicine, Juntendo University School of Medicine, Tokyo 113-8431, Japan; (M.I.)
| | - Hideko Arita
- Department of Anesthesiology, Pain Relief Center, JR Tokyo General Hospital, Tokyo 151-8528, Japan; (H.A.); (K.H.)
| | - Kazuo Hanaoka
- Department of Anesthesiology, Pain Relief Center, JR Tokyo General Hospital, Tokyo 151-8528, Japan; (H.A.); (K.H.)
| | - Jitsu Kato
- Department of Anesthesiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Setsuro Ogawa
- University Research Center, Nihon University, Tokyo 173-8610, Japan
| | - Ayako Hiranuma
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; (M.S.); (S.O.); (D.N.)
- Department of Surgery, Toho University Sakura Medical Center, Chiba 285-8741, Japan
| | - Junko Hasegawa
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; (M.S.); (S.O.); (D.N.)
| | - Kyoko Nakayama
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; (M.S.); (S.O.); (D.N.)
| | - Yuko Ebata
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; (M.S.); (S.O.); (D.N.)
| | - Masakazu Hayashida
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; (M.S.); (S.O.); (D.N.)
- Department of Anesthesiology & Pain Medicine, Juntendo University School of Medicine, Tokyo 113-8431, Japan; (M.I.)
- Department of Anesthesiology, Saitama Medical University International Medical Center, Saitama 350-1298, Japan
| | - Tatsuya Ichinohe
- Department of Dental Anesthesiology, Tokyo Dental College, Tokyo 101-0061, Japan;
| | - Ken-ichi Fukuda
- Department of Oral Health and Clinical Science, Tokyo Dental College, Tokyo 101-0061, Japan
| | - Kazutaka Ikeda
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan; (M.S.); (S.O.); (D.N.)
| |
Collapse
|
5
|
Mason BN, Hassler SN, DeFea K, Boitano S, Vagner J, Price TJ, Dussor G. PAR2 activation in the dura causes acute behavioral responses and priming to glyceryl trinitrate in a mouse migraine model. J Headache Pain 2023; 24:42. [PMID: 37072694 PMCID: PMC10114383 DOI: 10.1186/s10194-023-01574-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 04/03/2023] [Indexed: 04/20/2023] Open
Abstract
BACKGROUND Migraine is a severely debilitating disorder that affects millions of people worldwide. Studies have indicated that activation of protease-activated receptor-2 (PAR2) in the dura mater causes headache responses in preclinical models. It is also well known that vasodilators such as nitric oxide (NO) donors can trigger migraine attacks in migraine patients but not controls. In the current study we examined whether activation of PAR2 in the dura causes priming to the NO donor glyceryl trinitrate (GTN). METHODS A preclinical behavioral model of migraine was used where stimuli (PAR2 agonists: 2at-LIGRL-NH2 (2AT) or neutrophil elastase (NE); and IL-6) were applied to the mouse dura through an injection made at the intersection of the lamdoidal and sagittal sutures on the skull. Following dural injection, periorbital von Frey thresholds and facial grimace responses were measured until their return to baseline. GTN was then given by intraperitoneal injection and periorbital hypersensitivity and facial grimace responses observed until they returned to baseline. RESULTS We found that application of the selective PAR2 agonist 2at-LIGRL-NH2 (2AT) onto the dura causes headache-related behavioral responses in WT but not PAR2-/- mice with no differences between sexes. Additionally, dural PAR2 activation with 2AT caused priming to GTN (1 mg/kg) at 14 days after primary dural stimulation. PAR2-/- mice showed no priming to GTN. We also tested behavioral responses to the endogenous protease neutrophil elastase, which can cleave and activate PAR2. Dural neutrophil elastase caused both acute responses and priming to GTN in WT but not PAR2-/- mice. Finally, we show that dural IL-6 causes acute responses and priming to GTN that is identical in WT and PAR2-/- mice, indicating that IL-6 does not act through PAR2 in this model. CONCLUSIONS These results indicate that PAR2 activation in the meninges can cause acute headache behavioral responses and priming to an NO donor, and support further exploration of PAR2 as a novel therapeutic target for migraine.
Collapse
Affiliation(s)
- Bianca N Mason
- Department of Neuroscience, University of Texas at Dallas, 800 West Campbell Rd, Richardson, TX, 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Shayne N Hassler
- Department of Neuroscience, University of Texas at Dallas, 800 West Campbell Rd, Richardson, TX, 75080, USA
| | | | - Scott Boitano
- Department of Physiology, University of Arizona, Tucson, 85724, USA
| | - Josef Vagner
- Bio5 Institute, University of Arizona, Tucson, AZ, 85724, USA
| | - Theodore J Price
- Department of Neuroscience, University of Texas at Dallas, 800 West Campbell Rd, Richardson, TX, 75080, USA
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Greg Dussor
- Department of Neuroscience, University of Texas at Dallas, 800 West Campbell Rd, Richardson, TX, 75080, USA.
- Center for Advanced Pain Studies, University of Texas at Dallas, Richardson, TX, 75080, USA.
| |
Collapse
|
6
|
Peach CJ, Edgington-Mitchell LE, Bunnett NW, Schmidt BL. Protease-activated receptors in health and disease. Physiol Rev 2023; 103:717-785. [PMID: 35901239 PMCID: PMC9662810 DOI: 10.1152/physrev.00044.2021] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 07/08/2022] [Accepted: 07/10/2022] [Indexed: 11/22/2022] Open
Abstract
Proteases are signaling molecules that specifically control cellular functions by cleaving protease-activated receptors (PARs). The four known PARs are members of the large family of G protein-coupled receptors. These transmembrane receptors control most physiological and pathological processes and are the target of a large proportion of therapeutic drugs. Signaling proteases include enzymes from the circulation; from immune, inflammatory epithelial, and cancer cells; as well as from commensal and pathogenic bacteria. Advances in our understanding of the structure and function of PARs provide insights into how diverse proteases activate these receptors to regulate physiological and pathological processes in most tissues and organ systems. The realization that proteases and PARs are key mediators of disease, coupled with advances in understanding the atomic level structure of PARs and their mechanisms of signaling in subcellular microdomains, has spurred the development of antagonists, some of which have advanced to the clinic. Herein we review the discovery, structure, and function of this receptor system, highlight the contribution of PARs to homeostatic control, and discuss the potential of PAR antagonists for the treatment of major diseases.
Collapse
Affiliation(s)
- Chloe J Peach
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York
- Department of Neuroscience and Physiology and Neuroscience Institute, Grossman School of Medicine, New York University, New York, New York
| | - Laura E Edgington-Mitchell
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
- Bluestone Center for Clinical Research, Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, New York
| | - Nigel W Bunnett
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York
- Department of Neuroscience and Physiology and Neuroscience Institute, Grossman School of Medicine, New York University, New York, New York
| | - Brian L Schmidt
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, New York
- Bluestone Center for Clinical Research, Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, New York
| |
Collapse
|
7
|
Intervention of neuroinflammation in the traumatic brain injury trajectory: In vivo and clinical approaches. Int Immunopharmacol 2022; 108:108902. [PMID: 35729835 DOI: 10.1016/j.intimp.2022.108902] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/25/2022] [Accepted: 05/24/2022] [Indexed: 12/11/2022]
|
8
|
Zhao JF, Ren T, Li XY, Guo TL, Liu CH, Wang X. Research Progress on the Role of Microglia Membrane Proteins or Receptors in Neuroinflammation and Degeneration. Front Cell Neurosci 2022; 16:831977. [PMID: 35281298 PMCID: PMC8913711 DOI: 10.3389/fncel.2022.831977] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 01/26/2022] [Indexed: 01/01/2023] Open
Abstract
Microglia are intrinsic immune cells of the central nervous system and play a dual role (pro-inflammatory and anti-inflammatory) in the homeostasis of the nervous system. Neuroinflammation mediated by microglia serves as an important stage of ischemic hypoxic brain injury, cerebral hemorrhage disease, neurodegeneration and neurotumor of the nervous system and is present through the whole course of these diseases. Microglial membrane protein or receptor is the basis of mediating microglia to play the inflammatory role and they have been found to be upregulated by recognizing associated ligands or sensing changes in the nervous system microenvironment. They can then allosterically activate the downstream signal transduction and produce a series of complex cascade reactions that can activate microglia, promote microglia chemotactic migration and stimulate the release of proinflammatory factor such as TNF-α, IL-β to effectively damage the nervous system and cause apoptosis of neurons. In this paper, several representative membrane proteins or receptors present on the surface of microglia are systematically reviewed and information about their structures, functions and specific roles in one or more neurological diseases. And on this basis, some prospects for the treatment of novel coronavirus neurological complications are presented.
Collapse
Affiliation(s)
- Jun-Feng Zhao
- Department of Neurosurgery, Affiliated Dalian No. 3 People’s Hospital, Dalian Medical University, Dalian, China
| | - Tong Ren
- Department of Neurosurgery, Affiliated Dalian No. 3 People’s Hospital, Dalian Medical University, Dalian, China
| | - Xiang-Yu Li
- Department of Neurosurgery, Affiliated Dalian No. 3 People’s Hospital, Dalian Medical University, Dalian, China
| | - Tian-Lin Guo
- Department of Neurosurgery, Affiliated Dalian No. 3 People’s Hospital, Dalian Medical University, Dalian, China
| | - Chun-Hui Liu
- Department of Neurosurgery, Beijing Tiantan Hospital, Beijing, China
- Chun-Hui Liu,
| | - Xun Wang
- Department of Neurosurgery, Affiliated Dalian No. 3 People’s Hospital, Dalian Medical University, Dalian, China
- *Correspondence: Xun Wang,
| |
Collapse
|
9
|
Protease-activated receptor 2 activation induces behavioural changes associated with depression-like behaviour through microglial-independent modulation of inflammatory cytokines. Psychopharmacology (Berl) 2022; 239:229-242. [PMID: 34888704 PMCID: PMC8770450 DOI: 10.1007/s00213-021-06040-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 11/30/2021] [Indexed: 11/20/2022]
Abstract
RATIONALE Major depressive disorder (MDD) is a leading cause of disability worldwide but currently prescribed treatments do not adequately ameliorate the disorder in a significant portion of patients. Hence, a better appreciation of its aetiology may lead to the development of novel therapies. OBJECTIVES In the present study, we have built on our previous findings indicating a role for protease-activated receptor-2 (PAR2) in sickness behaviour to determine whether the PAR2 activator, AC264613, induces behavioural changes similar to those observed in depression-like behaviour. METHODS AC264613-induced behavioural changes were examined using the open field test (OFT), sucrose preference test (SPT), elevated plus maze (EPM), and novel object recognition test (NOR). Whole-cell patch clamping was used to investigate the effects of PAR2 activation in the lateral habenula with peripheral and central cytokine levels determined using ELISA and quantitative PCR. RESULTS Using a blood-brain barrier (BBB) permeable PAR2 activator, we reveal that AC-264613 (AC) injection leads to reduced locomotor activity and sucrose preference in mice but is without effect in anxiety and memory-related tasks. In addition, we show that AC injection leads to elevated blood sera IL-6 levels and altered cytokine mRNA expression within the brain. However, neither microglia nor peripheral lymphocytes are the source of these altered cytokine profiles. CONCLUSIONS These data reveal that PAR2 activation results in behavioural changes often associated with depression-like behaviour and an inflammatory profile that resembles that seen in patients with MDD and therefore PAR2 may be a target for novel antidepressant therapies.
Collapse
|
10
|
Liu Y, Li H, Hu J, Wu Z, Meng J, Hayashi Y, Nakanishi H, Qing H, Ni J. Differential Expression and Distinct Roles of Proteinase-Activated Receptor 2 in Microglia and Neurons in Neonatal Mouse Brain After Hypoxia-Ischemic Injury. Mol Neurobiol 2021; 59:717-730. [PMID: 34762231 DOI: 10.1007/s12035-021-02594-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 10/04/2021] [Indexed: 02/05/2023]
Abstract
Regulation of microglial activation and neuroinflammation are critical factors in the pathogenesis of ischemic brain injury. Interest in protease-activated receptor 2 (PAR2) as a pharmaceutical target for various diseases is creasing. However, it is unclear the expression and functions of PAR2 in hypoxia-ischemic (HI) brain injury. Mice with HI and cells with oxygen-glucose deprivation and reoxygenation (OGD/R) were studied. Immunoblot and qRT-PCR were used to study the differential gene expression in cultured microglia and neurons. Immunofluorescent staining was used to study the expression pattern of PAR2 in the HI brain and phagocytotic activity of microglia after OGD/R. In neonatal mice brain after HI, we found PAR2 expression was abundant in neurons, but barely in microglia from the contralateral side of cortex and hippocampus. Conversely, PAR2 expression was barely in neurons while significantly increased in activated microglia from the ipsilateral side of cortex and hippocampus. The activations of PAR2 were increased in both microglia and neuron in a cell model of OGD/R. PAR2 activation mediated the cross-talk between microglia and neurons including the following: microglial PAR2 mediated inflammatory responses that induced neuronal damage; neuronal PAR2 regulated chemokines that recruited activated microglia to damage area; microglia PAR2 controlled the phagocytosis of degenerating neurons. These data suggested differential expression and distinct roles of PAR2 in microglia and neurons after HI injury; thereby, interventions targeting PAR2 may provide insights into the inflammatory-related diseases.
Collapse
Affiliation(s)
- Yicong Liu
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.,Stomatology Hospital, School of Stomatology, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Hui Li
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China
| | - Jiangqi Hu
- Department of Prosthodontics, Beijing Stomatological Hospital, School of Stomatology, Capital Medical University, Beijing, 100081, China
| | - Zhou Wu
- Department of Aging Science and Pharmacology, Faculty of Dental Science, Kyushu University, Fukuoka, 812-8582, Japan
| | - Jie Meng
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yoshinori Hayashi
- Department of Physiology, Nihon University School of Dentistry, Tokyo, 101-8310, Japan
| | - Hiroshi Nakanishi
- Department of Pharmacology, Faculty of Pharmacy, Yasuda Women's University, Hiroshima, 731-0153, Japan
| | - Hong Qing
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
| | - Junjun Ni
- Key Laboratory of Molecular Medicine and Biotherapy, Department of Biology, School of Life Science, Beijing Institute of Technology, Beijing, 100081, China.
| |
Collapse
|
11
|
The development of proteinase-activated receptor-2 modulators and the challenges involved. Biochem Soc Trans 2021; 48:2525-2537. [PMID: 33242065 PMCID: PMC7752072 DOI: 10.1042/bst20200191] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 10/13/2020] [Accepted: 11/02/2020] [Indexed: 11/30/2022]
Abstract
Protease-activated receptor-2 (PAR2) has been extensively studied since its discovery in the mid-1990. Despite the advances in understanding PAR2 pharmacology, it has taken almost 25 years for the first inhibitor to reach clinical trials, and so far, no PAR2 antagonist has been approved for human use. Research has employed classical approaches to develop a wide array of PAR2 agonists and antagonists, consisting of peptides, peptoids and antibodies to name a few, with a surge in patent applications over this period. Recent breakthroughs in PAR2 structure determination has provided a unique insight into proposed PAR2 ligand binding sites. Publication of the first crystal structures of PAR2 resolved in complex with two novel non-peptide small molecule antagonists (AZ8838 and AZ3451) revealed two distinct binding pockets, originally presumed to be allosteric sites, with a PAR2 antibody (Fab3949) used to block tethered ligand engagement with the peptide-binding domain of the receptor. Further studies have proposed orthosteric site occupancy for AZ8838 as a competitive antagonist. One company has taken the first PAR2 antibody (MEDI0618) into phase I clinical trial (NCT04198558). While this first-in-human trial is at the early stages of the assessment of safety, other research into the structural characterisation of PAR2 is still ongoing in an attempt to identify new ways to target receptor activity. This review will focus on the development of novel PAR2 modulators developed to date, with an emphasis placed upon the advances made in the pharmacological targeting of PAR2 activity as a strategy to limit chronic inflammatory disease.
Collapse
|
12
|
Soeda M, Ohka S, Nishizawa D, Hasegawa J, Nakayama K, Ebata Y, Ichinohe T, Fukuda KI, Ikeda K. Cold pain sensitivity is associated with single-nucleotide polymorphisms of PAR2/ F2RL1 and TRPM8. Mol Pain 2021; 17:17448069211002009. [PMID: 33765896 PMCID: PMC8822448 DOI: 10.1177/17448069211002009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Pain sensitivity differs individually, but the mechanisms and genetic factors that underlie these differences are not fully understood. To investigate genetic factors that are involved in sensing cold pain, we applied a cold-induced pain test and evaluated protease-activated receptor 2 (PAR2/F2RL1) and transient receptor potential melastatin 8 (TRPM8), which are related to pain. We statistically investigated the associations between genetic polymorphisms and cold pain sensitivity in 461 healthy patients who were scheduled to undergo cosmetic orthognathic surgery for mandibular prognathism. We found an association between cold pain sensitivity and the rs2243057 polymorphism of the PAR2 gene. We also found a significant association between cold pain sensitivity and the rs12992084 polymorphism of the TRPM8 gene. Carriers of the minor A allele of the rs2243057 polymorphism of PAR2 and minor C allele of the rs12992084 polymorphism of TRPM8 exhibited a longer latency to pain perception in the cold-induced pain test, reflecting a decrease in cold pain sensitivity. These results suggest that genetic polymorphisms of both PAR2 and TRPM8 are involved in individual differences in cold pain sensitivity.
Collapse
Affiliation(s)
- Moe Soeda
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.,Department of Oral Health and Clinical Science, Tokyo Dental College, Tokyo, Japan
| | - Seii Ohka
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Daisuke Nishizawa
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Junko Hasegawa
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Kyoko Nakayama
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Yuko Ebata
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Tatsuya Ichinohe
- Department of Dental Anesthesiology, Tokyo Dental College, Tokyo, Japan
| | - Ken-Ichi Fukuda
- Department of Oral Health and Clinical Science, Tokyo Dental College, Tokyo, Japan
| | - Kazutaka Ikeda
- Addictive Substance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
13
|
Ye P, Ge K, Li M, Yang L, Jin S, Zhang C, Chen X, Geng Z. Egg-laying and brooding stage-specific hormonal response and transcriptional regulation in pituitary of Muscovy duck (Cairina moschata). Poult Sci 2020; 98:5287-5296. [PMID: 31376351 DOI: 10.3382/ps/pez433] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 07/16/2019] [Indexed: 01/24/2023] Open
Abstract
Broodiness is an interesting topic in reproductive biology for its reduced egg production. The strong brooding trait of Muscovy duck has become a major factor restricting the development of its industry. Broody phenotype and environmental factors influencing broodiness in poultry have been extensively studied, but the molecular regulation mechanism of broodiness remains unclear. In this research, the Muscovy duck reproductive endocrine hormones and pituitary transcriptome profiles during egg-laying phases (LP) and brooding phases (BP) were studied. During BP (n = 19), prolactin (PRL) levels was higher, while progesterone (P4) and estradiol (E2) were lower as compared to ducks during their LP (n = 20) (P < 0.01). We then examined the pituitary transcriptome of Muscovy duck at the 2 reproductive stages. A total of 398 differentially expressed genes included 20 transcription factors were identified (fold change ≥ 1.5, P < 0.01). There were 109 upregulated and 289 downregulated genes at brooding phases (n = 6) compared with egg-laying phases (n = 6). Real-time quantitative PCR analysis was carried out to verify the transcriptome results. The present study suggested that neuroactive ligand-receptor interaction pathway, calcium signaling pathway, and response to steroid hormones biological process are critical for controlling broodiness in the ducks. Further analysis revealed that SHH, PTGS2, RLN3, and transcription factor AP-1 may act as central signal modulators of hormonal and behavioral regulation mechanism associated with broodiness.
Collapse
Affiliation(s)
- Pengfei Ye
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China.,Anhui province key laboratory of local livestock and poultry genetic resource conservation and bio-breeding, Anhui Agricultural University, Hefei 230036, P.R. China
| | - Kai Ge
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China.,Anhui province key laboratory of local livestock and poultry genetic resource conservation and bio-breeding, Anhui Agricultural University, Hefei 230036, P.R. China.,College of biological and pharmaceutical engineering, West Anhui University, Liuan 237012, China
| | - Min Li
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China.,Anhui province key laboratory of local livestock and poultry genetic resource conservation and bio-breeding, Anhui Agricultural University, Hefei 230036, P.R. China
| | - Lei Yang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China.,Anhui province key laboratory of local livestock and poultry genetic resource conservation and bio-breeding, Anhui Agricultural University, Hefei 230036, P.R. China
| | - Sihua Jin
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China.,Anhui province key laboratory of local livestock and poultry genetic resource conservation and bio-breeding, Anhui Agricultural University, Hefei 230036, P.R. China
| | - Cheng Zhang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China.,Anhui province key laboratory of local livestock and poultry genetic resource conservation and bio-breeding, Anhui Agricultural University, Hefei 230036, P.R. China
| | - Xingyong Chen
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China.,Anhui province key laboratory of local livestock and poultry genetic resource conservation and bio-breeding, Anhui Agricultural University, Hefei 230036, P.R. China
| | - Zhaoyu Geng
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China.,Anhui province key laboratory of local livestock and poultry genetic resource conservation and bio-breeding, Anhui Agricultural University, Hefei 230036, P.R. China
| |
Collapse
|
14
|
Transcriptome Analysis and Emerging Driver Identification of CD8+ T Cells in Patients with Vitiligo. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2503924. [PMID: 31885781 PMCID: PMC6899274 DOI: 10.1155/2019/2503924] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/09/2019] [Indexed: 12/12/2022]
Abstract
Activated CD8+ T cells play important roles in the pathogenesis of vitiligo. However, driving factors about the activation and migration of CD8+ T cells remain obscure. In this study, we aim to identify differentially expressed genes (DEGs) and uncover potential factors that drive the disease in melanocyte-specific CD8+ T cells in vitiligo. A total of 1147 DEGs were found through transcriptome sequencing in CD8+ T cells from lesional skin of vitiligo patients and normal controls. Based on KEGG pathway enrichment analysis and PPI, 16 upregulated and 23 downregulated genes were identified. Ultimately, 3 genes were figured out after RT-qPCR verification. The mRNA and protein expression levels of PIK3CB, HIF-1α, and F2RL1 were all elevated in CD8+ T cells from peripheral blood in vitiligo. HIF-1α and PIK3CB were significantly increased in lesional skin of vitiligo. Two CpG sites of the HIF-1α promoter were hypomethylated in vitiligo CD8+ T cells. In conclusion, HIF-1α, F2RL1, and PIK3CB may act as novel drivers for vitiligo, which are all closely associated with reactive oxygen species and possibly contribute to the activation and/or migration of melanocyte-specific CD8+ T cells in vitiligo. In addition, we uncovered a potential role for DNA hypomethylation of HIF-1α in CD8+ T cells of vitiligo.
Collapse
|
15
|
Rajput PS, Lamb J, Kothari S, Pereira B, Soetkamp D, Wang Y, Tang J, Van Eyk JE, Mullins ES, Lyden PD. Neuron-generated thrombin induces a protective astrocyte response via protease activated receptors. Glia 2019; 68:246-262. [PMID: 31453648 DOI: 10.1002/glia.23714] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 08/02/2019] [Accepted: 08/15/2019] [Indexed: 01/08/2023]
Abstract
Astrocytes protect neurons during cerebral injury through several postulated mechanisms. Recent therapeutic attention has focused on enhancing or augmenting the neuroprotective actions of astrocytes but in some instances astrocytes can assume a neurotoxic phenotype. The signaling mechanisms that drive astrocytes toward a protective versus toxic phenotype are not fully known but cell-cell signaling via proteases acting on cell-specific receptors underlies critical mechanistic steps in neurodevelopment and disease. The protease activated receptor (PAR), resides in multiple brain cell types, and most PARs are found on astrocytes. We asked whether neuron-generated thrombin constituted an important astrocyte activation signal because our previous studies have shown that neurons contain prothrombin gene and transcribed protein. We used neuron and astrocyte mono-cell cultures exposed to oxygen-glucose deprivation and a model of middle cerebral artery occlusion. We found that ischemic neurons secrete thrombin into culture media, which leads to astrocyte activation; such astrocyte activation can be reproduced with low doses of thrombin. Media from prothrombin-deficient neurons failed to activate astrocytes and adding thrombin to such media restored activation. Astrocytes lacking PAR1 did not respond to neuron-generated thrombin. Induced astrocyte activation was antagonized dose-dependently with thrombin inhibitors or PAR1 antagonists. Ischemia-induced astrocyte activation in vivo was inhibited after neuronal prothrombin knockout, resulting in larger strokes. Restoring prothrombin to neurons with a lentiviral gene vector restored astrocyte activation and reduced stroke damage. We conclude that neuron-generated thrombin, released during ischemia, acts via PAR1 and may cause astrocyte activation and paracrine neuroprotection.
Collapse
Affiliation(s)
- Padmesh S Rajput
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Jessica Lamb
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Shweta Kothari
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Benedict Pereira
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, California
| | - Daniel Soetkamp
- The Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Yizhou Wang
- Genomics Core, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Jie Tang
- Genomics Core, Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, California
| | - Jennifer E Van Eyk
- The Smidt Heart Institute, Advanced Clinical Biosystems Research Institute, Cedars-Sinai Medical Center, Los Angeles, California
| | - Eric S Mullins
- Division of Hematology and Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, Ohio
| | - Patrick D Lyden
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, California
| |
Collapse
|
16
|
Shavit-Stein E, Artan-Furman A, Feingold E, Ben Shimon M, Itzekson-Hayosh Z, Chapman J, Vlachos A, Maggio N. Protease Activated Receptor 2 (PAR2) Induces Long-Term Depression in the Hippocampus through Transient Receptor Potential Vanilloid 4 (TRPV4). Front Mol Neurosci 2017; 10:42. [PMID: 28303089 PMCID: PMC5332813 DOI: 10.3389/fnmol.2017.00042] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Accepted: 02/07/2017] [Indexed: 11/25/2022] Open
Abstract
Protease activated receptors (PARs) are involved in regulating synaptic transmission and plasticity in the brain. While it is well-accepted that PAR1 mediates long-term potentiation (LTP) of excitatory synaptic strength, the role of PAR2 in synaptic plasticity remains not well-understood. In this study, we assessed the role of PAR2-signaling in plasticity at hippocampal Schaffer collateral-CA1 synapses. Using field potential recordings, we report that PAR2-activation leads to long-term depression (LTD) of synaptic transmission through a protein kinase A -dependent, Transient Receptor Potential Vanilloid 4 -mediated mechanism, which requires the activation of N-methyl-D-aspartate receptors. These results demonstrate that the effects of PAR2 on synaptic plasticity are distinct from what is observed upon PAR1-activation. Thus, we propose that the activation of different classes of PARs, i.e., PAR1 and PAR2, may set the threshold of synaptic plasticity in the hippocampal network by balancing LTP and LTD.
Collapse
Affiliation(s)
- Efrat Shavit-Stein
- Department of Neurology, The Chaim Sheba Medical Center Tel HaShomer, Israel
| | - Avital Artan-Furman
- Department of Neurology, The Chaim Sheba Medical Center Tel HaShomer, Israel
| | - Ekaterina Feingold
- Department of Neurology, The Chaim Sheba Medical Center Tel HaShomer, Israel
| | - Marina Ben Shimon
- Department of Neurology, The Chaim Sheba Medical CenterTel HaShomer, Israel; Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv UniversityTel Aviv, Israel
| | | | - Joab Chapman
- Department of Neurology, The Chaim Sheba Medical CenterTel HaShomer, Israel; Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv UniversityTel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv UniversityTel Aviv, Israel
| | - Andreas Vlachos
- Institute of Anatomy II, Faculty of Medicine, Heinrich-Heine-University Duesseldorf, Germany
| | - Nicola Maggio
- Department of Neurology, The Chaim Sheba Medical CenterTel HaShomer, Israel; Department of Neurology and Neurosurgery, Sackler Faculty of Medicine, Tel Aviv UniversityTel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv UniversityTel Aviv, Israel; Talpiot Medical Leadership Program, The Chaim Sheba Medical CenterTel HaShomer, Israel
| |
Collapse
|