1
|
Ikeda T, Takahashi K, Higashi M, Komiya H, Asano T, Ogasawara A, Kubota S, Hashiguchi S, Kunii M, Tanaka K, Tada M, Doi H, Takeuchi H, Takei K, Tanaka F. Lateral olfactory tract usher substance (LOTUS), an endogenous Nogo receptor antagonist, ameliorates disease progression in amyotrophic lateral sclerosis model mice. Cell Death Discov 2023; 9:454. [PMID: 38097540 PMCID: PMC10721829 DOI: 10.1038/s41420-023-01758-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 11/22/2023] [Accepted: 11/29/2023] [Indexed: 12/17/2023] Open
Abstract
Nogo-Nogo receptor 1 (NgR1) signaling is significantly implicated in neurodegeneration in amyotrophic lateral sclerosis (ALS). We previously showed that lateral olfactory tract usher substance (LOTUS) is an endogenous antagonist of NgR1 that prevents all myelin-associated inhibitors (MAIs), including Nogo, from binding to NgR1. Here we investigated the role of LOTUS in ALS pathogenesis by analyzing G93A-mutated human superoxide dismutase 1 (SOD1) transgenic (Tg) mice, as an ALS model, as well as newly generated LOTUS-overexpressing SOD1 Tg mice. We examined expression profiles of LOTUS and MAIs and compared motor functions and survival periods in these mice. We also investigated motor neuron survival, glial proliferation in the lumbar spinal cord, and neuromuscular junction (NMJ) morphology. We analyzed downstream molecules of NgR1 signaling such as ROCK2, LIMK1, cofilin, and ataxin-2, and also neurotrophins. In addition, we investigated LOTUS protein levels in the ventral horn of ALS patients. We found significantly decreased LOTUS expression in both SOD1 Tg mice and ALS patients. LOTUS overexpression in SOD1 Tg mice increased lifespan and improved motor function, in association with prevention of motor neuron loss, reduced gliosis, increased NMJ innervation, maintenance of cofilin phosphorylation dynamics, decreased levels of ataxin-2, and increased levels of brain-derived neurotrophic factor (BDNF). Reduced LOTUS expression may enhance neurodegeneration in SOD1 Tg mice and ALS patients by activating NgR1 signaling, and in this study LOTUS overexpression significantly ameliorated ALS pathogenesis. LOTUS might serve as a promising therapeutic target for ALS.
Collapse
Affiliation(s)
- Takuya Ikeda
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Keita Takahashi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan.
| | - Minatsu Higashi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Hiroyasu Komiya
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Tetsuya Asano
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Akihiro Ogasawara
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Shun Kubota
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Shunta Hashiguchi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Misako Kunii
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Kenichi Tanaka
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Mikiko Tada
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Hiroshi Doi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Hideyuki Takeuchi
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan
| | - Kohtaro Takei
- Molecular Medical Bioscience Laboratory, Yokohama City University Graduate School of Medical Life Science, Yokohama, 236-0004, Japan
| | - Fumiaki Tanaka
- Department of Neurology and Stroke Medicine, Yokohama City University Graduate School of Medicine, Yokohama, 236-0004, Japan.
| |
Collapse
|
2
|
Rashidbenam Z, Ozturk E, Pagnin M, Theotokis P, Grigoriadis N, Petratos S. How does Nogo receptor influence demyelination and remyelination in the context of multiple sclerosis? Front Cell Neurosci 2023; 17:1197492. [PMID: 37361998 PMCID: PMC10285164 DOI: 10.3389/fncel.2023.1197492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/15/2023] [Indexed: 06/28/2023] Open
Abstract
Multiple sclerosis (MS) can progress with neurodegeneration as a consequence of chronic inflammatory mechanisms that drive neural cell loss and/or neuroaxonal dystrophy in the central nervous system. Immune-mediated mechanisms can accumulate myelin debris in the disease extracellular milieu during chronic-active demyelination that can limit neurorepair/plasticity and experimental evidence suggests that potentiated removal of myelin debris can promote neurorepair in models of MS. The myelin-associated inhibitory factors (MAIFs) are integral contributors to neurodegenerative processes in models of trauma and experimental MS-like disease that can be targeted to promote neurorepair. This review highlights the molecular and cellular mechanisms that drive neurodegeneration as a consequence of chronic-active inflammation and outlines plausible therapeutic approaches to antagonize the MAIFs during the evolution of neuroinflammatory lesions. Moreover, investigative lines for translation of targeted therapies against these myelin inhibitors are defined with an emphasis on the chief MAIF, Nogo-A, that may demonstrate clinical efficacy of neurorepair during progressive MS.
Collapse
Affiliation(s)
- Zahra Rashidbenam
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Ezgi Ozturk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Maurice Pagnin
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Paschalis Theotokis
- Laboratory of Experimental Neurology and Neuroimmunology, Department of Neurology, AHEPA University Hospital, Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, Department of Neurology, AHEPA University Hospital, Thessaloniki, Greece
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
3
|
Modulation of the Microglial Nogo-A/NgR Signaling Pathway as a Therapeutic Target for Multiple Sclerosis. Cells 2022; 11:cells11233768. [PMID: 36497029 PMCID: PMC9737582 DOI: 10.3390/cells11233768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/23/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Current therapeutics targeting chronic phases of multiple sclerosis (MS) are considerably limited in reversing the neural damage resulting from repeated inflammation and demyelination insults in the multi-focal lesions. This inflammation is propagated by the activation of microglia, the endogenous immune cell aiding in the central nervous system homeostasis. Activated microglia may transition into polarized phenotypes; namely, the classically activated proinflammatory phenotype (previously categorized as M1) and the alternatively activated anti-inflammatory phenotype (previously, M2). These transitional microglial phenotypes are dynamic states, existing as a continuum. Shifting microglial polarization to an anti-inflammatory status may be a potential therapeutic strategy that can be harnessed to limit neuroinflammation and further neurodegeneration in MS. Our research has observed that the obstruction of signaling by inhibitory myelin proteins such as myelin-associated inhibitory factor, Nogo-A, with its receptor (NgR), can regulate microglial cell function and activity in pre-clinical animal studies. Our review explores the microglial role and polarization in MS pathology. Additionally, the potential therapeutics of targeting Nogo-A/NgR cellular mechanisms on microglia migration, polarization and phagocytosis for neurorepair in MS and other demyelination diseases will be discussed.
Collapse
|
4
|
Comparative Analysis of Neurodegeneration and Axonal Dysfunction Biomarkers in the Cerebrospinal Fluid of Patients with Multiple Sclerosis. J Clin Med 2022; 11:jcm11144122. [PMID: 35887886 PMCID: PMC9324050 DOI: 10.3390/jcm11144122] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 07/08/2022] [Accepted: 07/13/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Given the significant role of neurodegeneration in the progression of multiple sclerosis (MS) and insufficient therapies, there is an urgent need to better understand this pathology and to find new biomarkers that could provide important insight into the biological mechanisms of the disease. Thus, the present study aimed to compare different neurodegeneration and axonal dysfunction biomarkers in MS and verify their potential clinical usefulness. METHODS A total of 59 patients, who underwent CSF analysis during their diagnostics, were enrolled in the study. Quantitative analysis of neurodegeneration biomarkers was performed through immunological tests. Oligoclonal bands were detected by isoelectric focusing on agarose gel, whereas the concentrations of immunoglobulins and albumin were measured using nephelometry. RESULTS Our studies showed that NfL, RTN4, and tau protein enabled the differentiation of MS patients from the control group. Additionally, the baseline CSF NfL levels positively correlated with the tau and MRI results, whereas the RTN4 concentrations were associated with the immunoglobulin quotients. The AUC for NfL was the highest among the tested proteins, although the DeLong test of the ROC curves showed no significant difference between the AUCs for NfL and RTN4. CONCLUSION The CSF NfL, RTN-4, and tau levels at the time of diagnosis could be potential diagnostic markers of multiple sclerosis, although NfL seems to have the best clinical value.
Collapse
|
5
|
Kulczyńska-Przybik A, Dulewicz M, Słowik A, Borawska R, Kułakowska A, Kochanowicz J, Mroczko B. The Clinical Significance of Cerebrospinal Fluid Reticulon 4 (RTN4) Levels in the Differential Diagnosis of Neurodegenerative Diseases. J Clin Med 2021; 10:jcm10225281. [PMID: 34830564 PMCID: PMC8622503 DOI: 10.3390/jcm10225281] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/09/2021] [Accepted: 11/11/2021] [Indexed: 11/16/2022] Open
Abstract
Neurodegenerative diseases (NDs) belong to the top global causes of mortality. Diagnostic approaches to improve early diagnosis and differentiation of these diseases are constantly being sought. Therefore, we aimed to assess the cerebrospinal fluid (CSF) concentrations of Reticulon 4 (RTN4) in patients with neurodegenerative diseases and evaluate the potential clinical usefulness of this protein. RTNs are transmembrane proteins mediating neuroanatomical plasticity and functional recovery after central nervous system injury or diseases. According to our best knowledge, this is the first investigation providing the data concerning the dynamic of CSF RTN4 protein levels in patients with different NDs. Methods: Overall, 77 newly diagnosed patients with neurodegenerative diseases, including Alzheimer’s disease (AD), Parkinson’s disease (PD), and multiple sclerosis (MS), as well as 21 controls, were enrolled in the study. The CSF concentrations of tested proteins were assessed using immunological assays. Results: We revealed significantly higher CSF RTN4A levels in patients with AD, PD, and MS in comparison to the controls. Moreover, the comparative analysis of RTN4 concentration between different neurodegenerative diseases revealed the highest concentration of RTN4A in AD patients and a statistically significant difference between AD vs. PD, and AD vs. MS groups. The increased CSF level of the protein correlated with Tau, and pTau181 proteins in AD as well as in PD patients. Conclusions: Our study presents a previously not identified clinical utility of RTN4 in the differential diagnosis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Agnieszka Kulczyńska-Przybik
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland; (M.D.); (R.B.); (B.M.)
- Correspondence:
| | - Maciej Dulewicz
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland; (M.D.); (R.B.); (B.M.)
| | - Agnieszka Słowik
- Department of Neurology, Jagiellonian University, 30-688 Kraków, Poland;
| | - Renata Borawska
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland; (M.D.); (R.B.); (B.M.)
| | - Alina Kułakowska
- Department of Neurology, Medical University of Bialystok, 15-269 Bialystok, Poland; (A.K.); (J.K.)
| | - Jan Kochanowicz
- Department of Neurology, Medical University of Bialystok, 15-269 Bialystok, Poland; (A.K.); (J.K.)
| | - Barbara Mroczko
- Department of Neurodegeneration Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland; (M.D.); (R.B.); (B.M.)
- Department of Biochemical Diagnostics, Medical University of Bialystok, 15-269 Bialystok, Poland
| |
Collapse
|
6
|
The Implication of Reticulons (RTNs) in Neurodegenerative Diseases: From Molecular Mechanisms to Potential Diagnostic and Therapeutic Approaches. Int J Mol Sci 2021; 22:ijms22094630. [PMID: 33924890 PMCID: PMC8125174 DOI: 10.3390/ijms22094630] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/21/2021] [Accepted: 04/23/2021] [Indexed: 02/07/2023] Open
Abstract
Reticulons (RTNs) are crucial regulatory factors in the central nervous system (CNS) as well as immune system and play pleiotropic functions. In CNS, RTNs are transmembrane proteins mediating neuroanatomical plasticity and functional recovery after central nervous system injury or diseases. Moreover, RTNs, particularly RTN4 and RTN3, are involved in neurodegeneration and neuroinflammation processes. The crucial role of RTNs in the development of several neurodegenerative diseases, including Alzheimer's disease (AD), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), or other neurological conditions such as brain injury or spinal cord injury, has attracted scientific interest. Reticulons, particularly RTN-4A (Nogo-A), could provide both an understanding of early pathogenesis of neurodegenerative disorders and be potential therapeutic targets which may offer effective treatment or inhibit disease progression. This review focuses on the molecular mechanisms and functions of RTNs and their potential usefulness in clinical practice as a diagnostic tool or therapeutic strategy.
Collapse
|
7
|
Bakhuraysah MM, Theotokis P, Lee JY, Alrehaili AA, Aui PM, Figgett WA, Azari MF, Abou-Afech JP, Mackay F, Siatskas C, Alderuccio F, Strittmatter SM, Grigoriadis N, Petratos S. B-cells expressing NgR1 and NgR3 are localized to EAE-induced inflammatory infiltrates and are stimulated by BAFF. Sci Rep 2021; 11:2890. [PMID: 33536561 PMCID: PMC7858582 DOI: 10.1038/s41598-021-82346-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 01/14/2021] [Indexed: 02/01/2023] Open
Abstract
We have previously reported evidence that Nogo-A activation of Nogo-receptor 1 (NgR1) can drive axonal dystrophy during the neurological progression of experimental autoimmune encephalomyelitis (EAE). However, the B-cell activating factor (BAFF/BlyS) may also be an important ligand of NgR during neuroinflammation. In the current study we define that NgR1 and its homologs may contribute to immune cell signaling during EAE. Meningeal B-cells expressing NgR1 and NgR3 were identified within the lumbosacral spinal cords of ngr1+/+ EAE-induced mice at clinical score 1. Furthermore, increased secretion of immunoglobulins that bound to central nervous system myelin were shown to be generated from isolated NgR1- and NgR3-expressing B-cells of ngr1+/+ EAE-induced mice. In vitro BAFF stimulation of NgR1- and NgR3-expressing B cells, directed them into the cell cycle DNA synthesis phase. However, when we antagonized BAFF signaling by co-incubation with recombinant BAFF-R, NgR1-Fc, or NgR3 peptides, the B cells remained in the G0/G1 phase. The data suggest that B cells express NgR1 and NgR3 during EAE, being localized to infiltrates of the meninges and that their regulation is governed by BAFF signaling.
Collapse
Affiliation(s)
- Maha M Bakhuraysah
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, 3004, Australia
- Faculty of Applied Medical Sciences, Taif University, Taif, 26521, Kingdom of Saudi Arabia
| | - Paschalis Theotokis
- Laboratory of Experimental Neurology and Neuroimmunology, Department of Neurology, AHEPA University Hospital, 54636, Thessaloniki, Macedonia, Greece
| | - Jae Young Lee
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, 3004, Australia
- Toolgen Inc., Gasan Digital-Ro, 08594, Geumcheon, Seoul, Korea
| | - Amani A Alrehaili
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, 3004, Australia
- Faculty of Applied Medical Sciences, Taif University, Taif, 26521, Kingdom of Saudi Arabia
| | - Pei-Mun Aui
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, 3004, Australia
| | - William A Figgett
- Department of Microbiology and Immunology, School of Biomedical Science, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, 3000, Australia
| | - Michael F Azari
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, 3004, Australia
| | - John-Paul Abou-Afech
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, 3004, Australia
| | - Fabienne Mackay
- Department of Microbiology and Immunology, School of Biomedical Science, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, 3000, Australia
- QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | | | - Frank Alderuccio
- Department of Immunology and Pathology, Central Clinical School, Monash University, Prahran, VIC, 3004, Australia
| | - Stephen M Strittmatter
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, 06536, USA
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, Department of Neurology, AHEPA University Hospital, 54636, Thessaloniki, Macedonia, Greece
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, 3004, Australia.
| |
Collapse
|
8
|
Abstract
Nogo-A is considered one of the most important inhibitors of myelin-associated axonal regeneration in the central nervous system. It is mainly expressed by oligodendrocytes. Although previous studies have found regulatory roles for Nogo-A in neurite outgrowth inhibition, neuronal homeostasis, precursor migration, plasticity, and neurodegeneration, its functions in the process of oxidative injury are largely uncharacterized. In this study, oligodendrocytes were extracted from the cerebral cortex of newborn Sprague-Dawley rats. We used hydrogen peroxide (H2O2) to induce an in vitro oligodendrocyte oxidative damage model and found that endogenously expressed Nogo-A is significantly upregulated in oligodendrocytes. After recombinant virus Ad-ZsGreen-rat Nogo-A infection of oligodendrocytes, Nogo-A expression was increased, and the infected oligodendrocytes were more susceptible to acute oxidative insults and exhibited a markedly elevated rate of cell death. Furthermore, knockdown of Nogo-A expression in oligodendrocytes by Ad-ZsGreen-shRNA-Nogo-A almost completely protected against oxidative stress induced by exogenous H2O2. Intervention with a Nogo-66 antibody, a LINGO1 blocker, or Y27632, an inhibitor in the Nogo-66-NgR/p75/LINGO-1-RhoA-ROCK pathway, did not affect the death of oligodendrocytes. Ad-ZsGreen-shRNA-Nogo-A also increased the levels of phosphorylated extracellular signal-regulated kinase 1/2 and inhibited BCL2 expression in oligodendrocytes. In conclusion, Nogo-A aggravated reactive oxygen species damage in oligodendrocytes, and phosphorylated extracellular signal-regulated kinase 1/2 and BCL2 might be involved in this process. This study was approved by the Ethics Committee of Peking University People’s Hospital, China (approval No. 2018PHC081) on December 18, 2018.
Collapse
Affiliation(s)
- Yang-Yang Wang
- Department of Neurology, Peking University People's Hospital, Beijing, China
| | - Na Han
- Department of Neurology, Peking University People's Hospital, Beijing, China
| | - Dao-Jun Hong
- Department of Neurology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Jun Zhang
- Department of Neurology, Peking University People's Hospital, Beijing, China
| |
Collapse
|
9
|
Petratos S, Theotokis P, Kim MJ, Azari MF, Lee JY. That's a Wrap! Molecular Drivers Governing Neuronal Nogo Receptor-Dependent Myelin Plasticity and Integrity. Front Cell Neurosci 2020; 14:227. [PMID: 32848619 PMCID: PMC7417613 DOI: 10.3389/fncel.2020.00227] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Accepted: 06/29/2020] [Indexed: 12/19/2022] Open
Abstract
Myelin is a dynamic membrane that is important for coordinating the fast propagation of action potentials along small or large caliber axons (0.1-10 μm) some of which extend the entire length of the spinal cord. Due to the heterogeneity of electrical and energy demands of the variable neuronal populations, the axo-myelinic and axo-glial interactions that regulate the biophysical properties of myelinated axons also vary in terms of molecular interactions at the membrane interfaces. An important topic of debate in neuroscience is how myelin is maintained and modified under neuronal control and how disruption of this control (due to disease or injury) can initiate and/or propagate neurodegeneration. One of the key molecular signaling cascades that have been investigated in the context of neural injury over the past two decades involves the myelin-associated inhibitory factors (MAIFs) that interact with Nogo receptor 1 (NgR1). Chief among the MAIF superfamily of molecules is a reticulon family protein, Nogo-A, that is established as a potent inhibitor of neurite sprouting and axon regeneration. However, an understated role for NgR1 is its ability to control axo-myelin interactions and Nogo-A specific ligand binding. These interactions may occur at axo-dendritic and axo-glial synapses regulating their functional and dynamic membrane domains. The current review provides a comprehensive analysis of how neuronal NgR1 can regulate myelin thickness and plasticity under normal and disease conditions. Specifically, we discuss how NgR1 plays an important role in regulating paranodal and juxtaparanodal domains through specific signal transduction cascades that are important for microdomain molecular architecture and action potential propagation. Potential therapeutics designed to target NgR1-dependent signaling during disease are being developed in animal models since interference with the involvement of the receptor may facilitate neurological recovery. Hence, the regulatory role played by NgR1 in the axo-myelinic interface is an important research field of clinical significance that requires comprehensive investigation.
Collapse
Affiliation(s)
- Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Paschalis Theotokis
- Laboratory of Experimental Neurology and Neuroimmunology, Department of Neurology, AHEPA University Hospital, Thessaloniki, Greece
| | - Min Jung Kim
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Michael F Azari
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | | |
Collapse
|
10
|
Liu H, Su D, Liu L, Chen L, Zhao Y, Chan SO, Zhang W, Wang Y, Wang J. Identification of a new functional domain of Nogo-A that promotes inflammatory pain and inhibits neurite growth through binding to NgR1. FASEB J 2020; 34:10948-10965. [PMID: 32598099 DOI: 10.1096/fj.202000377r] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 05/28/2020] [Accepted: 06/08/2020] [Indexed: 01/10/2023]
Abstract
Nogo-A is a key inhibitory molecule to axon regeneration, and plays diverse roles in other pathological conditions, such as stroke, schizophrenia, and neurodegenerative diseases. Nogo-66 and Nogo-Δ20 fragments are two known functional domains of Nogo-A, which act through the Nogo-66 receptor (NgR1) and sphingosine-1-phosphate receptor 2 (S1PR2), respectively. Here, we reported a new functional domain of Nogo-A, Nogo-A aa 846-861, was identified in the Nogo-A-specific segment that promotes complete Freund's adjuvant (CFA)-induced inflammatory pain. Intrathecal injection of its antagonist peptide 846-861PE or the specific antibody attenuated the CFA-induced inflammatory heat hyperalgesia. The 846-861 PE reduced the content of transient receptor potential vanilloid subfamily member 1 (TRPV1) in dorsal root ganglia (DRG) and decreased the response of DRG neurons to capsaicin. These effects were accompanied by a reduction in LIMK/cofilin phosphorylation and actin polymerization. GST pull-down and fluorescence resonance energy transfer (FRET) assays both showed that Nogo-A aa 846-861 bound to NgR1. Moreover, we demonstrated that Nogo-A aa 846-861 inhibited neurite outgrowth from cortical neurons and DRG explants. We concluded that Nogo-A aa 846-861 is a novel ligand of NgR1, which activates the downstream signaling pathways that inhibit axon growth and promote inflammatory pain.
Collapse
Affiliation(s)
- Huaicun Liu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Dongqiang Su
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Lei Liu
- Department of Neurobiology, School of Basic Medical Sciences and Neuroscience Research Institute, Key Lab for Neuroscience, Ministry of Education of China and National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China.,PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Ling Chen
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yan Zhao
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Sun-On Chan
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Weiguang Zhang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Yun Wang
- Department of Neurobiology, School of Basic Medical Sciences and Neuroscience Research Institute, Key Lab for Neuroscience, Ministry of Education of China and National Health Commission and State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, China.,PKU-IDG/McGovern Institute for Brain Research, Peking University, Beijing, China
| | - Jun Wang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| |
Collapse
|
11
|
Shahidi SH, Kordi MR, Rajabi H, Malm C, Shah F, Quchan ASK. Exercise modulates the levels of growth inhibitor genes before and after multiple sclerosis. J Neuroimmunol 2020; 341:577172. [DOI: 10.1016/j.jneuroim.2020.577172] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 01/22/2020] [Accepted: 01/28/2020] [Indexed: 01/09/2023]
|
12
|
Metformin-induced AMPK activation stimulates remyelination through induction of neurotrophic factors, downregulation of NogoA and recruitment of Olig2+ precursor cells in the cuprizone murine model of multiple sclerosis. ACTA ACUST UNITED AC 2019; 27:583-592. [PMID: 31620963 DOI: 10.1007/s40199-019-00286-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 06/30/2019] [Indexed: 12/15/2022]
Abstract
PURPOSE Oligodendrocytes (OLGs) damage and myelin distraction is considered as a critical step in many neurological disorders especially multiple sclerosis (MS). Cuprizone (cup) animal model of MS targets OLGs degeneration and frequently used to the mechanistic understanding of de- and remyelination. The aim of this study was exploring the effects of metformin on the OLGs regeneration, myelin repair and profile of neurotrophic factors in the mice brain after cup-induced acute demyelination. METHODS Mice (C57BL/6 J) were fed with chow containing 0.2% cup for 5 weeks to induce specific OLGs degeneration and acute demyelination. Next, the cup was withdrawn to allow one-week recovery (spontaneous remyelination). At the end of this period, mature OLGs markers, myelin-associated neurite outgrowth inhibitor protein A (NogoA), premature specific OLGs transcription factor (Olig2), anti-apoptosis marker (survivin), neurotrophic factors, and AMPK activation were monitored in the presence or absence of metformin (50 mg/kg body weight/day) in the corpus callosum (CC). RESULTS Our finding indicated that consumption of metformin during the recovery period potentially induced an active form of AMPK (p-AMPK) and promoted repopulation of mature OLGs (MOG+ cells, MBP+ cells) in CC through up-regulation of BDNF, CNTF, and NGF as well as down-regulation of NogoA and recruitment of Olig2+ precursor cells. CONCLUSIONS This study for the first time reveals that metformin-induced AMPK, a master regulator of energy homeostasis, activation following toxic demyelination could potentially accelerate regeneration and supports spontaneous demyelination. These findings suggest the development of new therapeutic strategies based on AMPK activation for MS in the near future. Graphical abstract An overview of the possible molecular mechanisms of action of metformin-mediated remyelinationa.
Collapse
|
13
|
Marin MA, Carmichael ST. Mechanisms of demyelination and remyelination in the young and aged brain following white matter stroke. Neurobiol Dis 2019; 126:5-12. [DOI: 10.1016/j.nbd.2018.07.023] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 07/03/2018] [Accepted: 07/18/2018] [Indexed: 01/01/2023] Open
|
14
|
Limiting Neuronal Nogo Receptor 1 Signaling during Experimental Autoimmune Encephalomyelitis Preserves Axonal Transport and Abrogates Inflammatory Demyelination. J Neurosci 2019; 39:5562-5580. [PMID: 31061088 DOI: 10.1523/jneurosci.1760-18.2019] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 04/05/2019] [Accepted: 04/29/2019] [Indexed: 11/21/2022] Open
Abstract
We previously identified that ngr1 allele deletion limits the severity of experimental autoimmune encephalomyelitis (EAE) by preserving axonal integrity. However, whether this favorable outcome observed in EAE is a consequence of an abrogated neuronal-specific pathophysiological mechanism, is yet to be defined. Here we show that, Cre-loxP-mediated neuron-specific deletion of ngr1 preserved axonal integrity, whereas its re-expression in ngr1-/- female mice potentiated EAE-axonopathy. As a corollary, myelin integrity was preserved under Cre deletion in ngr1flx/flx , retinal ganglion cell axons whereas, significant demyelination occurred in the ngr1-/- optic nerves following the re-introduction of NgR1. Moreover, Cre-loxP-mediated axon-specific deletion of ngr1 in ngr1flx/flx mice also demonstrated efficient anterograde transport of fluorescently-labeled ChTxβ in the optic nerves of EAE-induced mice. However, the anterograde transport of ChTxβ displayed accumulation in optic nerve degenerative axons of EAE-induced ngr1-/- mice, when NgR1 was reintroduced but was shown to be transported efficiently in the contralateral non- recombinant adeno-associated virus serotype 2-transduced optic nerves of these mutant mice. We further identified that the interaction between the axonal motor protein, Kinesin-1 and collapsin response mediator protein 2 (CRMP2) was unchanged upon Cre deletion of ngr1 Whereas, this Kinesin-1/CRMP2 association was reduced when NgR1 was re-expressed in the ngr1-/- optic nerves. Our data suggest that NgR1 governs axonal degeneration in the context of inflammatory-mediated demyelination through the phosphorylation of CRMP2 by stalling axonal vesicular transport. Moreover, axon-specific deletion of ngr1 preserves axonal transport mechanisms, blunting the induction of inflammatory demyelination and limiting the severity of EAE.SIGNIFICANCE STATEMENT Multiple sclerosis (MS) is commonly induced by aberrant immune-mediated destruction of the protective sheath of nerve fibers (known as myelin). However, it has been shown that MS lesions do not only consist of this disease pattern, exhibiting heterogeneity with continual destruction of axons. Here we investigate how neuronal NgR1 can drive inflammatory-mediated axonal degeneration and demyelination within the optic nerve by analyzing its downstream signaling events that govern axonal vesicular transport. We identify that abrogating the NgR1/pCRMP2 signaling cascade can maintain Kinesin-1-dependent anterograde axonal transport to limit inflammatory-mediated axonopathy and demyelination. The ability to differentiate between primary and secondary mechanisms of axonal degeneration may uncover therapeutic strategies to limit axonal damage and progressive MS.
Collapse
|
15
|
Can We Design a Nogo Receptor-Dependent Cellular Therapy to Target MS? Cells 2018; 8:cells8010001. [PMID: 30577457 PMCID: PMC6357095 DOI: 10.3390/cells8010001] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 12/12/2018] [Accepted: 12/18/2018] [Indexed: 12/18/2022] Open
Abstract
The current landscape of therapeutics designed to treat multiple sclerosis (MS) and its pathological sequelae is saturated with drugs that modify disease course and limit relapse rates. While these small molecules and biologicals are producing profound benefits to patients with reductions in annualized relapse rates, the repair or reversal of demyelinated lesions with or without axonal damage, remains the principle unmet need for progressive forms of the disease. Targeting the extracellular pathological milieu and the signaling mechanisms that drive neurodegeneration are potential means to achieve neuroprotection and/or repair in the central nervous system of progressive MS patients. The Nogo-A receptor-dependent signaling mechanism has raised considerable interest in neurological disease paradigms since it can promulgate axonal transport deficits, further demyelination, and extant axonal dystrophy, thereby limiting remyelination. If specific therapeutic regimes could be devised to directly clear the Nogo-A-enriched myelin debris in an expedited manner, it may provide the necessary CNS environment for neurorepair to become a clinical reality. The current review outlines novel means to achieve neurorepair with biologicals that may be directed to sites of active demyelination.
Collapse
|
16
|
Smedfors G, Olson L, Karlsson TE. A Nogo-Like Signaling Perspective from Birth to Adulthood and in Old Age: Brain Expression Patterns of Ligands, Receptors and Modulators. Front Mol Neurosci 2018. [PMID: 29520216 PMCID: PMC5827527 DOI: 10.3389/fnmol.2018.00042] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
An appropriate strength of Nogo-like signaling is important to maintain synaptic homeostasis in the CNS. Disturbances have been associated with schizophrenia, MS and other diseases. Blocking Nogo-like signaling may improve recovery after spinal cord injury, stroke and traumatic brain injury. To understand the interacting roles of an increasing number of ligands, receptors and modulators engaged in Nogo-like signaling, the transcriptional activity of these genes in the same brain areas from birth to old age in the normal brain is needed. Thus, we have quantitatively mapped the innate expression of 11 important genes engaged in Nogo-like signaling. Using in situ hybridization, we located and measured the amount of mRNA encoding Nogo-A, OMgp, NgR1, NgR2, NgR3, Lingo-1, Troy, Olfactomedin, LgI1, ADAM22, and MAG, in 18 different brain areas at six different ages (P0, 1, 2, 4, 14, and 104 weeks). We show gene- and area-specific activities and how the genes undergo dynamic regulation during postnatal development and become stable during adulthood. Hippocampal areas underwent the largest changes over time. We only found differences between individual cortical areas in Troy and MAG. Subcortical areas presented the largest inter-regional differences; lateral and basolateral amygdala had markedly higher expression than other subcortical areas. The widespread differences and unique expression patterns of the different genes involved in Nogo-like signaling suggest that the functional complexes could look vastly different in different areas.
Collapse
Affiliation(s)
| | - Lars Olson
- Department of Neuroscience, Karolinska Institute, Stockholm, Sweden
| | | |
Collapse
|
17
|
p75NTR and TROY: Uncharted Roles of Nogo Receptor Complex in Experimental Autoimmune Encephalomyelitis. Mol Neurobiol 2018; 55:6329-6336. [PMID: 29294247 DOI: 10.1007/s12035-017-0841-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 12/14/2017] [Indexed: 12/11/2022]
Abstract
Multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE), have been on the forefront of drug discovery for most of the myelin inhibitory molecules implicated in axonal regenerative process. Nogo-A along with its putative receptor NgR and co-receptor LINGO-1 has paved the way for the production of pharmaceutical agents such as monoclonal antibodies, which are already put into handful of clinical trials. On the other side, little progress has been made towards clarifying the role of neurotrophin receptor p75 (p75NTR) and TROY in disease progression, other key players of the Nogo receptor complex. Previous work of our lab has shown that their exact location and type of expression is harmonized in a phase-dependent manner. Here, in this review, we outline their façade in normal and diseased central nervous system (CNS) and suggest a role for p75NTR in chronic axonal regeneration whereas TROY in acute inflammation of EAE intercourse.
Collapse
|
18
|
Alrehaili AA, Lee JY, Bakhuraysah MM, Kim MJ, Aui PM, Magee KA, Petratos S. Nogo receptor expression in microglia/macrophages during experimental autoimmune encephalomyelitis progression. Neural Regen Res 2018; 13:896-907. [PMID: 29863021 PMCID: PMC5998626 DOI: 10.4103/1673-5374.232488] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Myelin-associated inhibitory factors within the central nervous system (CNS) are considered to be one of the main obstacles for axonal regeneration following disease or injury. The nogo receptor 1 (NgR1) has been well documented to play a key role in limiting axonal regrowth in the injured and diseased mammalian CNS. However, the role of nogo receptor in immune cell activation during CNS inflammation is yet to be mechanistically elucidated. Microglia/macrophages are immune cells that are regarded as pathogenic contributors to inflammatory demyelinating lesions in multiple sclerosis (MS). In this study, the animal model of MS, experimental autoimmune encephalomyelitis (EAE) was induced in ngr1+/+ and ngr1–/– female mice following injection with the myelin oligodendrocyte glycoprotein (MOG35–55) peptide. A fate-map analysis of microglia/macrophages was performed throughout spinal cord sections of EAE-induced mice at clinical scores of 0, 1, 2 and 3, respectively (increasing locomotor disability) from both genotypes, using the CD11b and Iba1 cell markers. Western immunoblotting using lysates from isolated spinal cord microglia/macrophages, along with immunohistochemistry and flow cytometric analysis, was performed to demonstrate the expression of nogo receptor and its two homologs during EAE progression. Myelin protein engulfment during EAE progression in ngr1+/+ and ngr1–/– mice was demonstrated by western immunblotting of lysates from isolated spinal cord microglia/macrophages, detecting levels of Nogo-A and MOG. The numbers of M1 and M2 microglia/macrophage phenotypes present in the spinal cords of EAE-induced ngr1+/+ and ngr1–/– mice, were assessed by flow cytometric analysis using CD38 and Erg-2 markers. A significant difference in microglia/macrophage numbers between ngr1+/+ and ngr1–/– mice was identified during the progression of the clinical symptoms of EAE, in the white versus gray matter regions of the spinal cord. This difference was unrelated to the expression of NgR on these macrophage/microglial cells. We have identified that as EAE progresses, the phagocytic activity of microglia/macrophages with myelin debris, in ngr1–/– mice, was enhanced. Moreover, we show a modulation from a predominant M1-pathogenic to the M2-neurotrophic cell phenotype in the ngr1–/– mice during EAE progression. These findings suggest that CNS-specific macrophages and microglia of ngr1–/– mice may exhibit an enhanced capacity to clear inhibitory molecules that are sequestered in inflammatory lesions.
Collapse
Affiliation(s)
- Amani A Alrehaili
- Department of Neuroscience, Central Clinical School, Monash University, Prahran Victoria, Australia; Department of Clinical Laboratories, College of Applied Medical Sciences, Taif University, Taif, Kingdom of Saudi Arabia
| | - Jae Young Lee
- Department of Neuroscience, Central Clinical School, Monash University, Prahran Victoria, Australia; Toolgen Inc., Gasan Digital-Ro, Geumcheon, Seoul, Korea
| | - Maha M Bakhuraysah
- Department of Neuroscience, Central Clinical School, Monash University, Prahran Victoria, Australia; Department of Clinical Laboratories, College of Applied Medical Sciences, Taif University, Taif, Kingdom of Saudi Arabia
| | - Min Joung Kim
- Department of Neuroscience, Central Clinical School, Monash University, Prahran Victoria, Australia
| | - Pei-Mun Aui
- Department of Neuroscience, Central Clinical School, Monash University, Prahran Victoria, Australia
| | - Kylie A Magee
- Department of Neuroscience, Central Clinical School, Monash University, Prahran Victoria, Australia
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran Victoria, Australia
| |
Collapse
|
19
|
Ineichen BV, Kapitza S, Bleul C, Good N, Plattner PS, Seyedsadr MS, Kaiser J, Schneider MP, Zörner B, Martin R, Linnebank M, Schwab ME. Nogo-A antibodies enhance axonal repair and remyelination in neuro-inflammatory and demyelinating pathology. Acta Neuropathol 2017. [PMID: 28646336 DOI: 10.1007/s00401-017-1745-3] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Two hallmarks of chronic multiple sclerosis lesions are the absence of significant spontaneous remyelination and primary as well as secondary neurodegeneration. Both characteristics may be influenced by the presence of inhibitory factors preventing myelin and neuronal repair. We investigated the potential of antibodies against Nogo-A, a well-known inhibitory protein for neuronal growth and plasticity, to enhance neuronal regeneration and remyelination in two animal models of multiple sclerosis. We induced a targeted experimental autoimmune encephalomyelitis (EAE) lesion in the dorsal funiculus of the cervical spinal cord of adult rats resulting in a large drop of skilled forelimb motor functions. We subsequently observed improved recovery of forelimb function after anti-Nogo-A treatment. Anterograde tracing of the corticospinal tract revealed enhanced axonal sprouting and arborisation within the spinal cord gray matter preferentially targeting pre-motor and motor spinal cord laminae on lesion level and above in the anti-Nogo-A-treated animals. An important additional effect of Nogo-A-neutralization was enhanced remyelination observed after lysolecithin-induced demyelination of spinal tracts. Whereas remyelinated fiber numbers in the lesion site were increased several fold, no effect of Nogo-A-inhibition was observed on oligodendrocyte precursor proliferation, migration, or differentiation. Enhancing remyelination and promoting axonal regeneration and plasticity represent important unmet medical needs in multiple sclerosis. Anti-Nogo-A antibodies hold promise as a potential new therapy for multiple sclerosis, in particular during the chronic phase of the disease when neurodegeneration and remyelination failure determine disability evolution.
Collapse
Affiliation(s)
- Benjamin V Ineichen
- Brain Research Institute, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland.
- Department of Health Sciences and Technology, ETH Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland.
- Department of Neurology, University Hospital of Zurich, Zurich, Switzerland.
| | - Sandra Kapitza
- Brain Research Institute, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
- Department of Neurology, University Hospital of Zurich, Zurich, Switzerland
| | - Christiane Bleul
- Brain Research Institute, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
- Translational Genomics Research Institute, Phoenix, AZ, USA
| | - Nicolas Good
- Brain Research Institute, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Patricia S Plattner
- Brain Research Institute, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Maryam S Seyedsadr
- Brain Research Institute, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
- Department of Neurology, University Hospital of Zurich, Zurich, Switzerland
| | - Julia Kaiser
- Brain Research Institute, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
- Department of Neurology, University Hospital of Zurich, Zurich, Switzerland
| | - Marc P Schneider
- Brain Research Institute, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Björn Zörner
- Brain Research Institute, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Roland Martin
- Department of Neurology, University Hospital of Zurich, Zurich, Switzerland
| | - Michael Linnebank
- Department of Neurology, University Hospital of Zurich, Zurich, Switzerland
- Department of Neurorehabilitation, School of Medicine, HELIOS Klinik Hagen-Ambrock, Witten/Herdecke University Faculty of Health, Ambrocker Weg 60, 58091, Hagen, Germany
| | - Martin E Schwab
- Brain Research Institute, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
- Department of Health Sciences and Technology, ETH Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| |
Collapse
|
20
|
Lee JY, Kim MJ, Li L, Velumian AA, Aui PM, Fehlings MG, Petratos S. Nogo receptor 1 regulates Caspr distribution at axo-glial units in the central nervous system. Sci Rep 2017; 7:8958. [PMID: 28827698 PMCID: PMC5567129 DOI: 10.1038/s41598-017-09405-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 07/24/2017] [Indexed: 11/09/2022] Open
Abstract
Axo-glial units are highly organised microstructures propagating saltatory conduction and are disrupted during multiple sclerosis (MS). Nogo receptor 1 (NgR1) has been suggested to govern axonal damage during the progression of disease in the MS-like mouse model, experimental autoimmune encephalomyelitis (EAE). Here we have identified that adult ngr1 -/- mice, previously used in EAE and spinal cord injury experiments, display elongated paranodes, and nodes of Ranvier. Unstructured paranodal regions in ngr1 -/- mice are matched with more distributed expression pattern of Caspr. Compound action potentials of optic nerves and spinal cords from naïve ngr1 -/- mice are delayed and reduced. Molecular interaction studies revealed enhanced Caspr cleavage. Our data suggest that NgR1 may regulate axo-myelin ultrastructure through Caspr-mediated adhesion, regulating the electrophysiological signature of myelinated axons of central nervous system (CNS).
Collapse
Affiliation(s)
- Jae Young Lee
- Department of Medicine, Central Clinical School, Monash University, Prahran, Victoria, 3004, Australia
- ToolGen, Inc., #1204, Byucksan Digital Valley 6-cha, Seoul, South Korea
| | - Min Joung Kim
- Department of Medicine, Central Clinical School, Monash University, Prahran, Victoria, 3004, Australia
| | - Lijun Li
- Krembil Research Institute, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Alexander A Velumian
- Krembil Research Institute, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Krembil Neuroscience Centre, University Health Network, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Pei Mun Aui
- Department of Medicine, Central Clinical School, Monash University, Prahran, Victoria, 3004, Australia
| | - Michael G Fehlings
- Krembil Research Institute, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Krembil Neuroscience Centre, University Health Network, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Steven Petratos
- Department of Medicine, Central Clinical School, Monash University, Prahran, Victoria, 3004, Australia.
| |
Collapse
|
21
|
Abstract
Most of the current therapies, as well as many of the clinical trials, for multiple sclerosis (MS) target the inflammatory autoimmune processes, but less than 20% of all clinical trials investigate potential therapies for the chronic progressive disease stage of MS. The latter is responsible for the steadily increasing disability in many patients, and there is an urgent need for novel therapies that protect nervous system tissue and enhance axonal growth and/or remyelination. As outlined in this review, solid pre-clinical data suggest neutralization of the neurite outgrowth inhibitor Nogo-A as a potential new way to achieve both axonal and myelin repair. Several phase I clinical studies with anti-Nogo-A antibodies have been conducted in different disease paradigms including MS and spinal cord injury. Data from spinal cord injury and amyotrophic lateral sclerosis (ALS) trials accredit a good safety profile of high doses of anti-Nogo-A antibodies administered intravenously or intrathecally. An antibody against a Nogo receptor subunit, leucine rich repeat and immunoglobulin-like domain-containing protein 1 (LINGO-1), was recently shown to improve outcome in patients with acute optic neuritis in a phase II study. Nogo-A-suppressing antibodies could be novel drug candidates for the relapsing as well as the progressive MS disease stage. In this review, we summarize the available pre-clinical and clinical evidence on Nogo-A and elucidate the potential of Nogo-A-antibodies as a therapy for progressive MS.
Collapse
|
22
|
Trowsdale J, Jones DC, Barrow AD, Traherne JA. Surveillance of cell and tissue perturbation by receptors in the LRC. Immunol Rev 2016; 267:117-36. [PMID: 26284474 DOI: 10.1111/imr.12314] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The human leukocyte receptor complex (LRC) encompasses several sets of genes with a common evolutionary origin and which form a branch of the immunoglobulin superfamily (IgSF). Comparisons of LRC genes both within and between species calls for a high degree of plasticity. The drive for this unprecedented level of variation is not known, but it relates in part to interaction of several LRC products with polymorphic human leukocyte antigen (HLA) class I molecules. However, the range of other proposed ligands for LRC products indicates a dynamic set of receptors that have adapted to detect target molecules relating to numerous cellular pathways. Several receptors in the complex bind a molecular signature in collagenous ligands. Others detect a variety of motifs relating to pathogens in addition to cellular stress, attesting to the opportunistic versatility of LRC receptors.
Collapse
Affiliation(s)
- John Trowsdale
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Des C Jones
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Alexander D Barrow
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | | |
Collapse
|
23
|
Lee JY, Biemond M, Petratos S. Axonal degeneration in multiple sclerosis: defining therapeutic targets by identifying the causes of pathology. Neurodegener Dis Manag 2015; 5:527-48. [DOI: 10.2217/nmt.15.50] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Current therapeutics in multiple sclerosis (MS) target the putative inflammation and immune attack on CNS myelin. Despite their effectiveness in blunting the relapse rate in MS patients, such therapeutics do not prevent MS disease progression. Importantly, specific clinical dilemma arises through inability to predict MS progression and thereby therapeutically target axonal injury during MS, limiting permanent disability. The current review identifies immune and neurobiological principles that govern the sequelae of axonal degeneration during MS disease progression. Defining the specific disease arbiters, inflammatory and autoimmune, oligodendrocyte dystrophy and degenerative myelin, we discuss a basis for a molecular mechanism in axons that may be targeted therapeutically, in spatial and temporal manner to limit axonal degeneration and thereby halt progression of MS.
Collapse
Affiliation(s)
- Jae Young Lee
- Department of Medicine, Central Clinical School, Monash University, Prahran VIC 3004, Australia
| | - Melissa Biemond
- Department of Medicine, Central Clinical School, Monash University, Prahran VIC 3004, Australia
| | - Steven Petratos
- Department of Medicine, Central Clinical School, Monash University, Prahran VIC 3004, Australia
| |
Collapse
|
24
|
Seiler S, Di Santo S, Widmer HR. Non-canonical actions of Nogo-A and its receptors. Biochem Pharmacol 2015; 100:28-39. [PMID: 26348872 DOI: 10.1016/j.bcp.2015.08.113] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 08/31/2015] [Indexed: 12/13/2022]
Abstract
Nogo-A is a myelin associated protein and one of the most potent neurite growth inhibitors in the central nervous system. Interference with Nogo-A signaling has thus been investigated as therapeutic target to promote functional recovery in CNS injuries. Still, the finding that Nogo-A presents a fairly ubiquitous expression in many types of neurons in different brain regions, in the eye and even in the inner ear suggests for further functions besides the neurite growth repression. Indeed, a growing number of studies identified a variety of functions including regulation of neuronal stem cells, modulation of microglial activity, inhibition of angiogenesis and interference with memory formation. Aim of the present commentary is to draw attention on these less well-known and sometimes controversial roles of Nogo-A. Furthermore, we are addressing the role of Nogo-A in neuropathological conditions such as ischemic stroke, schizophrenia and neurodegenerative diseases.
Collapse
Affiliation(s)
- Stefanie Seiler
- Department of Neurosurgery, Neurocenter and Regenerative Neuroscience Cluster, University Hospital Bern and University of Bern, CH-3010 Bern, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Stefano Di Santo
- Department of Neurosurgery, Neurocenter and Regenerative Neuroscience Cluster, University Hospital Bern and University of Bern, CH-3010 Bern, Switzerland
| | - Hans Rudolf Widmer
- Department of Neurosurgery, Neurocenter and Regenerative Neuroscience Cluster, University Hospital Bern and University of Bern, CH-3010 Bern, Switzerland.
| |
Collapse
|
25
|
Li YH, Yu JZ, Xin YL, Feng L, Chai Z, Liu JC, Zhang HZ, Zhang GX, Xiao BG, Ma CG. Protective effect of a novel Rho kinase inhibitor WAR-5 in experimental autoimmune encephalomyelitis by modulating inflammatory response and neurotrophic factors. Exp Mol Pathol 2015; 99:220-8. [PMID: 26112093 DOI: 10.1016/j.yexmp.2015.06.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 06/19/2015] [Indexed: 11/19/2022]
Abstract
The Rho-kinase (ROCK) inhibitor Fasudil has proven beneficial in experimental autoimmune encephalomyelitis (EAE). Given the small safety window of Fasudil, we are looking for novel ROCK inhibitors, which have similar or stronger effect on EAE with greater safety. In this study, we report that WAR-5, a Y-27632 derivative, alleviates the clinical symptoms, attenuates myelin damage and reduces CNS inflammatory responses in EAE C57BL/6 mice at an extent similar to Fasudil, while exhibits less vasodilator and adverse reaction in vivo. WAR-5 inhibits ROCK activity, and selectively suppresses the expression of ROCK II in spleen, brain and spinal cord of EAE mice, especially in spinal cord, accompanied by decreased expression of Nogo. WAR-5 also regulates the imbalance of Th1/Th17 T cells and regulatory T cells, inhibits inflammatory microenvironment induced with NF-κB-IL-1β pathway. Importantly, WAR-5 converts M1 toward M2 microglia/macrophages that are positively correlated with BDNF and NT-3 production. Taken together, WAR-5 exhibits therapeutic potential in EAE by more selectively inhibits ROCK II, with a greater safety than Fasudil, and is worthy of further clinical study to clarify its clinical value.
Collapse
MESH Headings
- 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives
- 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology
- Amides/pharmacology
- Aminopyridines/pharmacology
- Animals
- Cytokines/metabolism
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/prevention & control
- Enzyme-Linked Immunosorbent Assay
- Female
- Immunoblotting
- Immunoenzyme Techniques
- Inflammation/drug therapy
- Inflammation/immunology
- Inflammation/metabolism
- Macrophages/drug effects
- Macrophages/immunology
- Macrophages/metabolism
- Mice
- Mice, Inbred C57BL
- Microglia/drug effects
- Microglia/immunology
- Microglia/metabolism
- Nerve Growth Factors/metabolism
- Piperidines/pharmacology
- Protein Kinase Inhibitors/pharmacology
- Pyridines/pharmacology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- rho-Associated Kinases/antagonists & inhibitors
- rho-Associated Kinases/metabolism
Collapse
Affiliation(s)
- Yan-hua Li
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong 037009, China
| | - Jie-zhong Yu
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong 037009, China
| | - Yan-le Xin
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong 037009, China
| | - Ling Feng
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong 037009, China
| | - Zhi Chai
- "2011" Collaborative Innovation Center/Research Center of Neurobiology, Shanxi University of Traditional Chinese Medicine, Taiyuan 030619, China
| | - Jian-chun Liu
- "2011" Collaborative Innovation Center/Research Center of Neurobiology, Shanxi University of Traditional Chinese Medicine, Taiyuan 030619, China
| | - Hong-zhen Zhang
- "2011" Collaborative Innovation Center/Research Center of Neurobiology, Shanxi University of Traditional Chinese Medicine, Taiyuan 030619, China
| | - Guang-Xian Zhang
- Department of Neurology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Bao-guo Xiao
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong 037009, China; Institute of Neurology, Huashan Hospital, Institutes of Brain Science and State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200025, China.
| | - Cun-gen Ma
- Institute of Brain Science, Department of Neurology, Medical School, Shanxi Datong University, Datong 037009, China; "2011" Collaborative Innovation Center/Research Center of Neurobiology, Shanxi University of Traditional Chinese Medicine, Taiyuan 030619, China.
| |
Collapse
|
26
|
Aging-Related Peculiarities of the Distribution of Myelin Basic Protein in Cerebral Structures of Gerbils. NEUROPHYSIOLOGY+ 2015. [DOI: 10.1007/s11062-015-9514-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
27
|
Lee SI, Yun J, Baek JY, Jeong YJ, Kim JA, Kang JS, Park SH, Kim SK, Park SK. NgR1 Expressed in P19 Embryonal Carcinoma Cells Differentiated by Retinoic Acid Can Activate STAT3. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2015; 19:105-9. [PMID: 25729271 PMCID: PMC4342729 DOI: 10.4196/kjpp.2015.19.2.105] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Revised: 01/02/2015] [Accepted: 01/12/2015] [Indexed: 01/09/2023]
Abstract
NgR1, a Nogo receptor, is involved in inhibition of neurite outgrowth and axonal regeneration and regulation of synaptic plasticity. P19 embryonal carcinoma cells were induced to differentiate into neuron-like cells using all trans-retinoic acid and the presence and/or function of cellular molecules, such as NgR1, NMDA receptors and STAT3, were examined. Neuronally differentiated P19 cells expressed the mRNA and protein of NgR1, which could stimulate the phosphorylation of STAT3 when activated by Nogo-P4 peptide, an active segment of Nogo-66. During the whole period of differentiation, mRNAs of all of the NMDA receptor subtypes tested (NR1, NR2A-2D) were consistently expressed, which meant that neuronally differentiated P19 cells maintained some characteristics of neurons, especially central nervous system neurons. Our results suggests that neuronally differentiated P19 cells expressing NgR1 may be an efficient and convenient in vitro model for studying the molecular mechanism of cellular events that involve NgR1 and its binding partners, and for screening compounds that activate or inhibit NgR1.
Collapse
Affiliation(s)
- Su In Lee
- College of Pharmacy, Korea University, Sejong 339-700, Korea
| | - Jieun Yun
- Bioevaluation Center, KRIBB, Ochang 363-883, Korea
| | - Ji-Young Baek
- College of Pharmacy, Korea University, Sejong 339-700, Korea
| | - Yun-Ji Jeong
- College of Pharmacy, Korea University, Sejong 339-700, Korea
| | - Jin-Ah Kim
- Bioevaluation Center, KRIBB, Ochang 363-883, Korea
| | | | - Sun Hong Park
- College of Pharmacy, Chungbuk National University, Cheongju 362-763, Korea
| | - Sang Kyum Kim
- College of Pharmacy, Chungnam National University, Daejeon 305-764, Korea
| | - Song-Kyu Park
- College of Pharmacy, Korea University, Sejong 339-700, Korea. ; Research Driven Hospital, Korea University Guro Hospital, Biomedical Research Center, Seoul 152-703, Korea
| |
Collapse
|
28
|
Pourabdolhossein F, Mozafari S, Morvan-Dubois G, Mirnajafi-Zadeh J, Lopez-Juarez A, Pierre-Simons J, Demeneix BA, Javan M. Nogo receptor inhibition enhances functional recovery following lysolecithin-induced demyelination in mouse optic chiasm. PLoS One 2014; 9:e106378. [PMID: 25184636 PMCID: PMC4153612 DOI: 10.1371/journal.pone.0106378] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2014] [Accepted: 07/29/2014] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Inhibitory factors have been implicated in the failure of remyelination in demyelinating diseases. Myelin associated inhibitors act through a common receptor called Nogo receptor (NgR) that plays critical inhibitory roles in CNS plasticity. Here we investigated the effects of abrogating NgR inhibition in a non-immune model of focal demyelination in adult mouse optic chiasm. METHODOLOGY/PRINCIPAL FINDINGS A focal area of demyelination was induced in adult mouse optic chiasm by microinjection of lysolecithin. To knock down NgR levels, siRNAs against NgR were intracerebroventricularly administered via a permanent cannula over 14 days, Functional changes were monitored by electrophysiological recording of latency of visual evoked potentials (VEPs). Histological analysis was carried out 3, 7 and 14 days post demyelination lesion. To assess the effect of NgR inhibition on precursor cell repopulation, BrdU was administered to the animals prior to the demyelination induction. Inhibition of NgR significantly restored VEPs responses following optic chiasm demyelination. These findings were confirmed histologically by myelin specific staining. siNgR application resulted in a smaller lesion size compared to control. NgR inhibition significantly increased the numbers of BrdU+/Olig2+ progenitor cells in the lesioned area and in the neurogenic zone of the third ventricle. These progenitor cells (Olig2+ or GFAP+) migrated away from this area as a function of time. CONCLUSIONS/SIGNIFICANCE Our results show that inhibition of NgR facilitate myelin repair in the demyelinated chiasm, with enhanced recruitment of proliferating cells to the lesion site. Thus, antagonizing NgR function could have therapeutic potential for demyelinating disorders such as Multiple Sclerosis.
Collapse
Affiliation(s)
- Fereshteh Pourabdolhossein
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- UMR CNRS 7221, Evolution des Régulations Endocriniennes, Département Régulations, Développement et Diversité Moléculaire, Muséum National d'Histoire Naturelle, Paris, France
| | - Sabah Mozafari
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ghislaine Morvan-Dubois
- UMR CNRS 7221, Evolution des Régulations Endocriniennes, Département Régulations, Développement et Diversité Moléculaire, Muséum National d'Histoire Naturelle, Paris, France
| | - Javad Mirnajafi-Zadeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Alejandra Lopez-Juarez
- UMR CNRS 7221, Evolution des Régulations Endocriniennes, Département Régulations, Développement et Diversité Moléculaire, Muséum National d'Histoire Naturelle, Paris, France
| | - Jacqueline Pierre-Simons
- UMR CNRS 7221, Evolution des Régulations Endocriniennes, Département Régulations, Développement et Diversité Moléculaire, Muséum National d'Histoire Naturelle, Paris, France
| | - Barbara A. Demeneix
- UMR CNRS 7221, Evolution des Régulations Endocriniennes, Département Régulations, Développement et Diversité Moléculaire, Muséum National d'Histoire Naturelle, Paris, France
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- * E-mail:
| |
Collapse
|
29
|
Winther M, Walmod PS. Neural cell adhesion molecules belonging to the family of leucine-rich repeat proteins. ADVANCES IN NEUROBIOLOGY 2014; 8:315-95. [PMID: 25300143 DOI: 10.1007/978-1-4614-8090-7_14] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Leucine-rich repeats (LRRs) are motifs that form protein-ligand interaction domains. There are approximately 140 human genes encoding proteins with extracellular LRRs. These encode cell adhesion molecules (CAMs), proteoglycans, G-protein-coupled receptors, and other types of receptors. Here we give a brief description of 36 proteins with extracellular LRRs that all can be characterized as CAMs or putative CAMs expressed in the nervous system. The proteins are involved in multiple biological processes in the nervous system including the proliferation and survival of cells, neuritogenesis, axon guidance, fasciculation, myelination, and the formation and maintenance of synapses. Moreover, the proteins are functionally implicated in multiple diseases including cancer, hearing impairment, glaucoma, Alzheimer's disease, multiple sclerosis, Parkinson's disease, autism spectrum disorders, schizophrenia, and obsessive-compulsive disorders. Thus, LRR-containing CAMs constitute a large group of proteins of pivotal importance for the development, maintenance, and regeneration of the nervous system.
Collapse
|
30
|
Chiurchiù V, Maccarrone M, Orlacchio A. The role of reticulons in neurodegenerative diseases. Neuromolecular Med 2013; 16:3-15. [PMID: 24218324 PMCID: PMC3918113 DOI: 10.1007/s12017-013-8271-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 10/23/2013] [Indexed: 01/08/2023]
Abstract
Reticulons (RTNs) are a group of membrane-associated proteins mainly responsible for shaping the tubular endoplasmic reticulum network, membrane trafficking, inhibition of axonal growth, and apoptosis. These proteins share a common sequence feature, the reticulon homology domain, which consists of paired hydrophobic stretches that are believed to induce membrane curvature by acting as a wedge in bilayer membranes. RTNs are ubiquitously expressed in all tissues, but each RTN member exhibits a unique expression pattern that prefers certain tissues or even cell types. Recently, accumulated evidence has suggested additional and unexpected roles for RTNs, including those on DNA binding, autophagy, and several inflammatory-related functions. These manifold actions of RTNs account for their ever-growing recognition of their involvement in neurodegenerative diseases like Alzheimer's disease, amyotrophic lateral sclerosis, multiple sclerosis, as well as hereditary spastic paraplegia. This review summarizes the latest discoveries on RTNs in human pathophysiology, and the engagement of these in neurodegeneration, along with the implications of these findings for a better understanding of the molecular events triggered by RTNs and their potential exploitation as next-generation therapeutics.
Collapse
Affiliation(s)
- Valerio Chiurchiù
- Laboratorio di Neurochimica dei Lipidi, Centro Europeo di Ricerca sul Cervello (CERC) - Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Santa Lucia, Rome, Italy
| | | | | |
Collapse
|