1
|
Testa A, Quaglia F, Naranjo NM, Verrillo CE, Shields CD, Lin S, Pickles MW, Hamza DF, Von Schalscha T, Cheresh DA, Leiby B, Liu Q, Ding J, Kelly WK, Hooper DC, Corey E, Plow EF, Altieri DC, Languino LR. Targeting the αVβ3/NgR2 pathway in neuroendocrine prostate cancer. Matrix Biol 2023; 124:49-62. [PMID: 37956856 PMCID: PMC10823877 DOI: 10.1016/j.matbio.2023.11.003] [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: 07/18/2023] [Revised: 10/25/2023] [Accepted: 11/08/2023] [Indexed: 11/15/2023]
Abstract
Highly aggressive, metastatic, neuroendocrine prostate cancer, which typically develops from prostate cancer cells acquiring resistance to androgen deprivation therapy, is associated with limited treatment options and hence poor prognosis. We have previously demonstrated that the αVβ3 integrin is over-expressed in neuroendocrine prostate cancer. We now show that LM609, a monoclonal antibody that specifically targets the human αVβ3 integrin, hinders the growth of neuroendocrine prostate cancer patient-derived xenografts in vivo. Our group has recently identified a novel αVβ3 integrin binding partner, NgR2, responsible for regulating the expression of neuroendocrine markers and for inducing neuroendocrine differentiation in prostate cancer cells. Through in vitro functional assays, we here demonstrate that NgR2 is crucial in promoting cell adhesion to αVβ3 ligands. Moreover, we describe for the first time co-fractionation of αVβ3 integrin and NgR2 in small extracellular vesicles derived from metastatic prostate cancer patients' plasma. These prostate cancer patient-derived small extracellular vesicles have a functional impact on human monocytes, increasing their adhesion to fibronectin. The monocytes incubated with small extracellular vesicles do not show an associated change in conventional polarization marker expression and appear to be in an early stage that may be defined as "adhesion competent". Overall, these findings allow us to better understand integrin-directed signaling and cell-cell communication during cancer progression. Furthermore, our results pave the way for new diagnostic and therapeutic perspectives for patients affected by neuroendocrine prostate cancer.
Collapse
Affiliation(s)
- Anna Testa
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, United States; Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Fabio Quaglia
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, United States; Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Nicole M Naranjo
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, United States; Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Cecilia E Verrillo
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, United States; Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Christopher D Shields
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, United States; Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Stephen Lin
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, United States; Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Maxwell W Pickles
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, United States; Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Drini F Hamza
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, United States; Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Tami Von Schalscha
- Department of Pathology, Moores Cancer Center, and Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, United States
| | - David A Cheresh
- Department of Pathology, Moores Cancer Center, and Sanford Consortium for Regenerative Medicine, University of California San Diego, La Jolla, CA, United States
| | - Benjamin Leiby
- Division of Biostatistics, Department of Pharmacology, Physiology, and Cancer Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| | - Qin Liu
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA, United States
| | - Jianyi Ding
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA, United States
| | - William K Kelly
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, United States
| | - D Craig Hooper
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, United States; Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA, United States
| | - Edward F Plow
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Dario C Altieri
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, United States
| | - Lucia R Languino
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, United States; Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
2
|
Ecrement A, Barnig C. Unlocking the potential of NK cells: NgR1-mediated modulation of immunological synapse stability. Allergy 2023; 78:3285-3286. [PMID: 37661661 DOI: 10.1111/all.15873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/10/2023] [Accepted: 08/22/2023] [Indexed: 09/05/2023]
Affiliation(s)
| | - Cindy Barnig
- Université de Franche-Comté, EFS, INSERM, UMR RIGHT, Besançon, France
- CHU Besançon, Department of Chest Disease, Besançon, France
| |
Collapse
|
3
|
Dave BP, Shah KC, Shah MB, Chorawala MR, Patel VN, Shah PA, Shah GB, Dhameliya TM. Unveiling the modulation of Nogo receptor in neuroregeneration and plasticity: Novel aspects and future horizon in a new frontier. Biochem Pharmacol 2023; 210:115461. [PMID: 36828272 DOI: 10.1016/j.bcp.2023.115461] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 02/25/2023]
Abstract
Neurodegenerative diseases (NDs) such as Alzheimer's, Parkinson's, Multiple Sclerosis, Hereditary Spastic Paraplegia, and Amyotrophic Lateral Sclerosis have emerged as the most dreaded diseases due to a lack of precise diagnostic tools and efficient therapies. Despite the fact that the contributing factors of NDs are still unidentified, mounting evidence indicates the possibility that genetic and cellular changes may lead to the significant production of abnormally misfolded proteins. These misfolded proteins lead to damaging effects thereby causing neurodegeneration. The association between Neurite outgrowth factor (Nogo) with neurological diseases and other peripheral diseases is coming into play. Three isoforms of Nogo have been identified Nogo-A, Nogo-B and Nogo-C. Among these, Nogo-A is mainly responsible for neurological diseases as it is localized in the CNS (Central Nervous System), whereas Nogo-B and Nogo-C are responsible for other diseases such as colitis, lung, intestinal injury, etc. Nogo-A, a membrane protein, had first been described as a CNS-specific inhibitor of axonal regeneration. Several recent studies have revealed the role of Nogo-A proteins and their receptors in modulating neurite outgrowth, branching, and precursor migration during nervous system development. It may also modulate or affect the inhibition of growth during the developmental processes of the CNS. Information about the effects of other ligands of Nogo protein on the CNS are yet to be discovered however several pieces of evidence have suggested that it may also influence the neuronal maturation of CNS and targeting Nogo-A could prove to be beneficial in several neurodegenerative diseases.
Collapse
Affiliation(s)
- Bhavarth P Dave
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad 380009, Gujarat, India
| | - Kashvi C Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad 380009, Gujarat, India
| | - Maitri B Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad 380009, Gujarat, India
| | - Mehul R Chorawala
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad 380009, Gujarat, India.
| | - Vishvas N Patel
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad 380009, Gujarat, India
| | - Palak A Shah
- Department of Pharmacology, K. B. Institute of Pharmaceutical Education and Research, Gandhinagar 380023, Gujarat, India
| | - Gaurang B Shah
- Department of Pharmacology and Pharmacy Practice, L. M. College of Pharmacy, Opp. Gujarat University, Ahmedabad 380009, Gujarat, India
| | - Tejas M Dhameliya
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, Nirma University, Ahmedabad-382481, Gujarat, India
| |
Collapse
|
4
|
van Gelderen TA, Ladisa C, Salazar-Moscoso M, Folgado C, Habibi HR, Ribas L. Metabolomic and transcriptomic profiles after immune stimulation in the zebrafish testes. Genomics 2023; 115:110581. [PMID: 36796654 DOI: 10.1016/j.ygeno.2023.110581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 02/09/2023] [Accepted: 02/11/2023] [Indexed: 02/16/2023]
Abstract
Fish farms are prone to disease outbreaks and stress due to high-density rearing conditions in tanks and sea cages, adversely affecting growth, reproduction, and metabolism. To understand the molecular mechanisms affected in the gonads of breeder fish after an immune challenge, we investigated the metabolome and the transcriptome profiles in zebrafish testes after inducing an immune response. After 48 h of the immune challenge, ultra-high-performance liquid chromatography (LC-MS) and transcriptomic analysis by RNA-seq (Illumina) resulted in 20 different released metabolites and 80 differentially expressed genes. Among these, glutamine and succinic acid were the most abundant metabolites released and 27,5% of the genes belong to either the immune or reproduction systems. Pathway analysis based on metabolomic and transcriptomic crosstalk identified cad and iars genes that act simultaneously with succinate metabolite. This study deciphers interactions between reproduction and immune systems and provides a basis to improve protocols in generating more resistant broodstock.
Collapse
Affiliation(s)
- T A van Gelderen
- Institut de Ciències del Mar - Consejo Superior de Investigaciones Científicas (ICM-CSIC), Department of Renewable Marine Resources, 08003 Barcelona, Spain
| | - C Ladisa
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - M Salazar-Moscoso
- Institut de Ciències del Mar - Consejo Superior de Investigaciones Científicas (ICM-CSIC), Department of Renewable Marine Resources, 08003 Barcelona, Spain
| | - C Folgado
- Institut de Ciències del Mar - Consejo Superior de Investigaciones Científicas (ICM-CSIC), Department of Renewable Marine Resources, 08003 Barcelona, Spain
| | - H R Habibi
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
| | - L Ribas
- Institut de Ciències del Mar - Consejo Superior de Investigaciones Científicas (ICM-CSIC), Department of Renewable Marine Resources, 08003 Barcelona, Spain.
| |
Collapse
|
5
|
Kong A, Liu T, Deng S, Xu S, Luo Y, Li J, Du Z, Wang L, Xu X, Fan X. Novel antidepressant-like properties of the fullerenol in an LPS-induced depressive mouse model. Int Immunopharmacol 2023; 116:109792. [PMID: 36738679 DOI: 10.1016/j.intimp.2023.109792] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 01/12/2023] [Accepted: 01/24/2023] [Indexed: 02/05/2023]
Abstract
Depression is a common mental disease and is highly prevalent in populations. Dysregulated neuroinflammation and concomitant-activated microglia are involved in the pathogenesis of depression. Experimental evidence has indicated that fullerenol exerts anti-neuroinflammation and protective effects against neurological diseases. Here, we evaluated fullerenol's effects against lipopolysaccharide (LPS)-induced mouse depressive-like behaviors. Fullerenol treatment produced an antidepressant-like effect, as indicated by preventing the LPS-induced reduction in the sucrose preference and shortening the immobility durations in both the tail suspension test and the forced swim test. We found that fullerenol treatment mitigated LPS-induced hippocampal microglia activation and released proinflammatory cytokines. Meanwhile, fullerenol promoted hippocampus neurogenesis, evidenced by increased DCX-positive cells in LPS-treated mice. Hippocampal RNA-Seq analysis revealed proinflammatory cytokine and neurogenesis involved in fullerenol's antidepressant-like effects. Our data indicate that fullerenol exerts antidepressant effects, which might be due to beneficial functions in reducing neuroinflammatory processes and promoting neurogenesis in the hippocampus.
Collapse
Affiliation(s)
- Anqi Kong
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, People's Republic of China; Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China; Institute of Neuroscience, Soochow University, Suzhou 215123, People's Republic of China
| | - Tianyao Liu
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Shilong Deng
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Shiyao Xu
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, People's Republic of China; Institute of Neuroscience, Soochow University, Suzhou 215123, People's Republic of China
| | - Yi Luo
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Jianghui Li
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Zhulin Du
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Liuyongwei Wang
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China
| | - Xingshun Xu
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, People's Republic of China; Institute of Neuroscience, Soochow University, Suzhou 215123, People's Republic of China.
| | - Xiaotang Fan
- Department of Military Cognitive Psychology, School of Psychology, Third Military Medical University (Army Medical University), Chongqing, People's Republic of China.
| |
Collapse
|
6
|
Nogo-A and LINGO-1: Two Important Targets for Remyelination and Regeneration. Int J Mol Sci 2023; 24:ijms24054479. [PMID: 36901909 PMCID: PMC10003089 DOI: 10.3390/ijms24054479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 02/13/2023] [Accepted: 02/22/2023] [Indexed: 02/26/2023] Open
Abstract
Multiple sclerosis (MS) is an inflammatory disease of the central nervous system (CNS) that causes progressive neurological disability in most patients due to neurodegeneration. Activated immune cells infiltrate the CNS, triggering an inflammatory cascade that leads to demyelination and axonal injury. Non-inflammatory mechanisms are also involved in axonal degeneration, although they are not fully elucidated yet. Current therapies focus on immunosuppression; however, no therapies to promote regeneration, myelin repair, or maintenance are currently available. Two different negative regulators of myelination have been proposed as promising targets to induce remyelination and regeneration, namely the Nogo-A and LINGO-1 proteins. Although Nogo-A was first discovered as a potent neurite outgrowth inhibitor in the CNS, it has emerged as a multifunctional protein. It is involved in numerous developmental processes and is necessary for shaping and later maintaining CNS structure and functionality. However, the growth-restricting properties of Nogo-A have negative effects on CNS injury or disease. LINGO-1 is also an inhibitor of neurite outgrowth, axonal regeneration, oligodendrocyte differentiation, and myelin production. Inhibiting the actions of Nogo-A or LINGO-1 promotes remyelination both in vitro and in vivo, while Nogo-A or LINGO-1 antagonists have been suggested as promising therapeutic approaches for demyelinating diseases. In this review, we focus on these two negative regulators of myelination while also providing an overview of the available data on the effects of Nogo-A and LINGO-1 inhibition on oligodendrocyte differentiation and remyelination.
Collapse
|
7
|
Quaglia F, Krishn SR, Sossey-Alaoui K, Rana PS, Pluskota E, Park PH, Shields CD, Lin S, McCue P, Kossenkov AV, Wang Y, Goodrich DW, Ku SY, Beltran H, Kelly WK, Corey E, Klose M, Bandtlow C, Liu Q, Altieri DC, Plow EF, Languino LR. The NOGO receptor NgR2, a novel αVβ3 integrin effector, induces neuroendocrine differentiation in prostate cancer. Sci Rep 2022; 12:18879. [PMID: 36344556 PMCID: PMC9640716 DOI: 10.1038/s41598-022-21711-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 09/30/2022] [Indexed: 11/09/2022] Open
Abstract
Androgen deprivation therapies aimed to target prostate cancer (PrCa) are only partially successful given the occurrence of neuroendocrine PrCa (NEPrCa), a highly aggressive and highly metastatic form of PrCa, for which there is no effective therapeutic approach. Our group has demonstrated that while absent in prostate adenocarcinoma, the αVβ3 integrin expression is increased during PrCa progression toward NEPrCa. Here, we show a novel pathway activated by αVβ3 that promotes NE differentiation (NED). This novel pathway requires the expression of a GPI-linked surface molecule, NgR2, also known as Nogo-66 receptor homolog 1. We show here that NgR2 is upregulated by αVβ3, to which it associates; we also show that it promotes NED and anchorage-independent growth, as well as a motile phenotype of PrCa cells. Given our observations that high levels of αVβ3 and, as shown here, of NgR2 are detected in human and mouse NEPrCa, our findings appear to be highly relevant to this aggressive and metastatic subtype of PrCa. This study is novel because NgR2 role has only minimally been investigated in cancer and has instead predominantly been analyzed in neurons. These data thus pave new avenues toward a comprehensive mechanistic understanding of integrin-directed signaling during PrCa progression toward a NE phenotype.
Collapse
Affiliation(s)
- Fabio Quaglia
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Shiv Ram Krishn
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Khalid Sossey-Alaoui
- Department of Medicine, School of Medicine, MetroHealth Medical Center, Rammelkamp Center for Research, Case Western Reserve University, Cleveland, OH, USA
| | - Priyanka Shailendra Rana
- Department of Medicine, School of Medicine, MetroHealth Medical Center, Rammelkamp Center for Research, Case Western Reserve University, Cleveland, OH, USA
| | - Elzbieta Pluskota
- Cardiovascular and Metabolic Sciences Department, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Pyung Hun Park
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Christopher D Shields
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Stephen Lin
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, USA
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Peter McCue
- Department of Pathology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Andrew V Kossenkov
- Center for Systems and Computational Biology, Wistar Institute, Philadelphia, PA, USA
| | - Yanqing Wang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - David W Goodrich
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Sheng-Yu Ku
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Himisha Beltran
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - William K Kelly
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Eva Corey
- Department of Urology, University of Washington, Seattle, WA, USA
| | - Maja Klose
- Institute of Neurochemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Christine Bandtlow
- Institute of Neurochemistry, Biocenter, Medical University of Innsbruck, Innsbruck, Austria
| | - Qin Liu
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA, USA
| | - Dario C Altieri
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA, USA
| | - Edward F Plow
- Cardiovascular and Metabolic Sciences Department, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Lucia R Languino
- Prostate Cancer Discovery and Development Program, Thomas Jefferson University, Philadelphia, PA, USA.
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
8
|
Ma C, Zhang W, Cao M. Role of the Peripheral Nervous System in PD Pathology, Diagnosis, and Treatment. Front Neurosci 2021; 15:598457. [PMID: 33994915 PMCID: PMC8119739 DOI: 10.3389/fnins.2021.598457] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 03/30/2021] [Indexed: 12/13/2022] Open
Abstract
Studies on Parkinson disease (PD) have mostly focused on the central nervous system—specifically, on the loss of mesencephalic dopaminergic neurons and associated motor dysfunction. However, the peripheral nervous system (PNS) is gaining prominence in PD research, with increasing clinical attention being paid to non-motor symptoms. Researchers found abnormal deposition of α-synuclein and neuroinflammation in the PNS. Attempts have been made to use these pathological changes during the clinical diagnosis of PD. Animal studies demonstrated that combined transplantation of autologous peripheral nerves and cells with tyrosine hydroxylase activity can reduce dopaminergic neuronal damage, and similar effects were observed in some clinical trials. In this review, we will systematically explain PNS performance in PD pathology and its clinical diagnostic research, describe PNS experimental results [especially Schwann cell (SC) transplantation in the treatment of PD animal models] and the results of clinical trials, and discuss future directions. The mechanism by which SCs produce such a therapeutic effect and the safety of transplantation therapy are briefly described.
Collapse
Affiliation(s)
- Chengxiao Ma
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China.,Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Wen Zhang
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China.,Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Maohong Cao
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
9
|
Liscic RM, Alberici A, Cairns NJ, Romano M, Buratti E. From basic research to the clinic: innovative therapies for ALS and FTD in the pipeline. Mol Neurodegener 2020; 15:31. [PMID: 32487123 PMCID: PMC7268618 DOI: 10.1186/s13024-020-00373-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 03/27/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and Frontotemporal Degeneration (FTD) are neurodegenerative disorders, related by deterioration of motor and cognitive functions and short survival. Aside from cases with an inherited pathogenic mutation, the causes of the disorders are still largely unknown and no effective treatment currently exists. It has been shown that FTD may coexist with ALS and this overlap occurs at clinical, genetic, and molecular levels. In this work, we review the main pathological aspects of these complex diseases and discuss how the integration of the novel pathogenic molecular insights and the analysis of molecular interaction networks among all the genetic players represents a critical step to shed light on discovering novel therapeutic strategies and possibly tailoring personalized medicine approaches to specific ALS and FTD patients.
Collapse
Affiliation(s)
- Rajka Maria Liscic
- Department of Neurology, Johannes Kepler University, Linz, Austria
- School of Medicine, University of Osijek, Osijek, Croatia
| | - Antonella Alberici
- Neurology Unit, Department of Neurological Sciences and Vision, ASST-Spedali Civili-University of Brescia, Brescia, Italy
| | - Nigel John Cairns
- College of Medicine and Health and Living Systems Institute, University of Exeter, Exeter, UK
| | - Maurizio Romano
- Department of Life Sciences, Via Valerio 28, University of Trieste, 34127, Trieste, Italy
| | - Emanuele Buratti
- International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano 99, 34149, Trieste, Italy.
| |
Collapse
|
10
|
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
|
11
|
Spotlight on Neurotrauma Research in Canada's Leading Academic Centers. J Neurotrauma 2018; 35:1986-2004. [PMID: 30074875 DOI: 10.1089/neu.2018.29017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
12
|
Ghosh S, Hui SP. Axonal regeneration in zebrafish spinal cord. REGENERATION (OXFORD, ENGLAND) 2018; 5:43-60. [PMID: 29721326 PMCID: PMC5911453 DOI: 10.1002/reg2.99] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 03/09/2018] [Accepted: 03/13/2018] [Indexed: 12/12/2022]
Abstract
In the present review we discuss two interrelated events-axonal damage and repair-known to occur after spinal cord injury (SCI) in the zebrafish. Adult zebrafish are capable of regenerating axonal tracts and can restore full functionality after SCI. Unlike fish, axon regeneration in the adult mammalian central nervous system is extremely limited. As a consequence of an injury there is very little repair of disengaged axons and therefore functional deficit persists after SCI in adult mammals. In contrast, peripheral nervous system axons readily regenerate following injury and hence allow functional recovery both in mammals and fish. A better mechanistic understanding of these three scenarios could provide a more comprehensive insight into the success or failure of axonal regeneration after SCI. This review summarizes the present understanding of the cellular and molecular basis of axonal regeneration, in both the peripheral nervous system and the central nervous system, and large scale gene expression analysis is used to focus on different events during regeneration. The discovery and identification of genes involved in zebrafish spinal cord regeneration and subsequent functional experimentation will provide more insight into the endogenous mechanism of myelination and remyelination. Furthermore, precise knowledge of the mechanism underlying the extraordinary axonal regeneration process in zebrafish will also allow us to unravel the potential therapeutic strategies to be implemented for enhancing regrowth and remyelination of axons in mammals.
Collapse
Affiliation(s)
- Sukla Ghosh
- Department of BiophysicsMolecular Biology and BioinformaticsUniversity of Calcutta92 A. P. C. RoadKolkata 700009India
| | - Subhra Prakash Hui
- Department of BiophysicsMolecular Biology and BioinformaticsUniversity of Calcutta92 A. P. C. RoadKolkata 700009India
- Victor Chang Cardiac Research InstituteLowy Packer Building, 405 Liverpool StDarlinghurstNSW 2010Australia.
| |
Collapse
|
13
|
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
|
14
|
Khalid SI, Ampie L, Kelly R, Ladha SS, Dardis C. Immune Modulation in the Treatment of Amyotrophic Lateral Sclerosis: A Review of Clinical Trials. Front Neurol 2017; 8:486. [PMID: 28993751 PMCID: PMC5622209 DOI: 10.3389/fneur.2017.00486] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 08/31/2017] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease characterized by the degeneration of motor neurons. Though many molecular and genetic causes are thought to serve as predisposing or disease propagating factors, the underlying pathogenesis of the disease is not known. Recent discoveries have demonstrated the presence of inflammation propagating substrates in the central nervous system of patients afflicted with ALS. Over the past decade, this hypothesis has incited an effort to better understand the role of the immune system in ALS and has led to the trial of several potential immune-modulating therapies. Here, we briefly review advances in the role of such therapies. The clinical trials discussed here are currently ongoing or have been concluded at the time of writing.
Collapse
Affiliation(s)
| | - Leonel Ampie
- Surgical Neurology Branch, NINDS, National Institutes of Health, Bethesda, MD, United States.,Department of Neurological Surgery, University of Virginia School of Medicine, Charlottesville, VA, United States.,Georgetown University School of Medicine, Washington, DC, United States
| | - Ryan Kelly
- Georgetown University School of Medicine, Washington, DC, United States
| | - Shafeeq S Ladha
- Department of Neurology, Barrow Neurological Institute, Phoenix, AZ, United States
| | - Christopher Dardis
- Department of Neurology, Barrow Neurological Institute, Phoenix, AZ, United States
| |
Collapse
|
15
|
Ziebell JM, Ray-Jones H, Lifshitz J. Nogo presence is inversely associated with shifts in cortical microglial morphology following experimental diffuse brain injury. Neuroscience 2017; 359:209-223. [PMID: 28736137 DOI: 10.1016/j.neuroscience.2017.07.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 06/28/2017] [Accepted: 07/12/2017] [Indexed: 01/08/2023]
Abstract
Diffuse traumatic brain injury (TBI) initiates secondary pathology, including inflammation and reduced myelination. Considering these injury-related pathologies, the many states of activated microglia as demonstrated by differing morphologies would form, migrate, and function in and through fields of growth-inhibitory myelin byproduct, specifically Nogo. Here we evaluate the relationship between inflammation and reduced myelin antigenicity in the wake of diffuse TBI and present the hypothesis that the Nogo-66 receptor antagonist peptide NEP(1-40) would reverse the injury-induced shift in distribution of microglia morphologies by limiting myelin-based inhibition. Adult male rats were subjected to midline fluid percussion sham or brain injury. At 2h, 6h, 1d, 2d, 7d, and 21d post-injury, immunohistochemical staining was analyzed in sensory cortex (S1BF) for myelin antigens (myelin basic protein; MBP and CNPase), microglia morphology (ionized calcium-binding adapter protein; Iba1), Nogo receptor and Nogo. Pronounced reduction in myelin antigenicity was evident transiently at 1d post-injury, as evidenced by decreased MBP and CNPase staining, as well as loss of white matter organization, compared to sham and later injury time points. Concomitant with reduced myelin antigenicity, injury shifted microglia morphology from the predominantly ramified morphology observed in sham-injured cortex to hyper-ramified, activated, fully activated, or rod. Changes in microglial morphology were evident as early as 2h post-injury, and remained at least until day 21. Additional cohorts of uninjured and brain-injured animals received vehicle or drug (NEP(1-40), i.p., 15min and 19h post-injury) and brains were collected at 2h, 6h, 1d, 2d, or 7d post-injury. NEP(1-40) administration further shifted distributions of microglia away from an injury-induced activated morphology toward greater proportions of rod and macrophage-like morphologies compared to vehicle-treated. By 7d post-injury, no differences in the distributions of microglia were noted between vehicle and NEP(1-40). This study begins to link secondary pathologies of white matter damage and inflammation after diffuse TBI. In the injured brain, secondary pathologies co-occur and likely interact, with consequences for neuronal circuit disruption leading to neurological symptoms.
Collapse
Affiliation(s)
- Jenna M Ziebell
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA; Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA.
| | - Helen Ray-Jones
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA; Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA; Department of Biology and Biochemistry, University of Bath, Bath, England, UK
| | - Jonathan Lifshitz
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix, AZ, USA; Department of Child Health, University of Arizona College of Medicine - Phoenix, Phoenix, AZ, USA; VA Healthcare System, Phoenix, AZ, USA; Psychology, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
16
|
Theotokis P, Touloumi O, Lagoudaki R, Nousiopoulou E, Kesidou E, Siafis S, Tselios T, Lourbopoulos A, Karacostas D, Grigoriadis N, Simeonidou C. Nogo receptor complex expression dynamics in the inflammatory foci of central nervous system experimental autoimmune demyelination. J Neuroinflammation 2016; 13:265. [PMID: 27724971 PMCID: PMC5057208 DOI: 10.1186/s12974-016-0730-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Accepted: 09/22/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Nogo-A and its putative receptor NgR are considered to be among the inhibitors of axonal regeneration in the CNS. However, few studies so far have addressed the issue of local NgR complex multilateral localization within inflammation in an MS mouse model of autoimmune demyelination. METHODS Chronic experimental autoimmune encephalomyelitis (EAE) was induced in C57BL/6 mice. Analyses were performed on acute (days 18-22) and chronic (day 50) time points and compared to controls. The temporal and spatial expression of the Nogo receptor complex (NgR and coreceptors) was studied at the spinal cord using epifluorescent and confocal microscopy or real-time PCR. Data are expressed as cells/mm2, as mean % ± SEM, or as arbitrary units of integrated density. RESULTS Animals developed a moderate to severe EAE without mortality, followed by a progressive, chronic clinical course. NgR complex spatial expression varied during the main time points of EAE. NgR with coreceptors LINGO-1 and TROY was increased in the spinal cord in the acute phase whereas LINGO-1 and p75 signal seemed to be dominant in the chronic phase, respectively. NgR was detected on gray matter NeuN+ neurons of the spinal cord, within the white matter inflammatory foci (14.2 ± 4.3 % NgR+ inflammatory cells), and found to be colocalized with GAP-43+ axonal growth cones while no β-TubIII+, SMI-32+, or APP+ axons were found as NgR+. Among the NgR+ inflammatory cells, 75.6 ± 9.0 % were microglial/macrophages (lectin+), 49.6 ± 14.2 % expressed CD68 (phagocytic ED1+ cells), and no cells were Mac-3+. Of these macrophages/monocytes, only Arginase-1+/NgR+ but not iNOS+/NgR+ were present in lesions both in acute and chronic phases. CONCLUSIONS Our data describe in detail the expression of the Nogo receptor complex within the autoimmune inflammatory foci and suggest a possible immune action for NgR apart from the established inhibitory one on axonal growth. Its expression by inflammatory macrophages/monocytes could signify a possible role of these cells on axonal guidance and clearance of the lesioned area during inflammatory demyelination.
Collapse
MESH Headings
- Animals
- Antigens, Differentiation/metabolism
- Arginase/metabolism
- Central Nervous System/metabolism
- Central Nervous System/pathology
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/chemically induced
- Encephalomyelitis, Autoimmune, Experimental/complications
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Freund's Adjuvant/immunology
- Freund's Adjuvant/toxicity
- Gene Expression Regulation/drug effects
- Gene Expression Regulation/immunology
- Mice
- Mice, Inbred C57BL
- Myelin-Oligodendrocyte Glycoprotein/immunology
- Myelin-Oligodendrocyte Glycoprotein/toxicity
- Nerve Tissue Proteins/metabolism
- Nogo Proteins/genetics
- Nogo Proteins/metabolism
- Nogo Receptors/genetics
- Nogo Receptors/metabolism
- Peptide Fragments/immunology
- Peptide Fragments/toxicity
- Receptors, Nerve Growth Factor/genetics
- Receptors, Nerve Growth Factor/metabolism
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/metabolism
- Signal Transduction/drug effects
- Signal Transduction/immunology
- Signal Transduction/physiology
- Statistics, Nonparametric
Collapse
Affiliation(s)
- Paschalis Theotokis
- B’ Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Aristotle University of Thessaloniki, Stilponos Kiriakides str. 1, 546 36 Thessaloniki, Central Macedonia Greece
| | - Olga Touloumi
- B’ Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Aristotle University of Thessaloniki, Stilponos Kiriakides str. 1, 546 36 Thessaloniki, Central Macedonia Greece
| | - Roza Lagoudaki
- B’ Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Aristotle University of Thessaloniki, Stilponos Kiriakides str. 1, 546 36 Thessaloniki, Central Macedonia Greece
| | - Evangelia Nousiopoulou
- B’ Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Aristotle University of Thessaloniki, Stilponos Kiriakides str. 1, 546 36 Thessaloniki, Central Macedonia Greece
| | - Evangelia Kesidou
- B’ Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Aristotle University of Thessaloniki, Stilponos Kiriakides str. 1, 546 36 Thessaloniki, Central Macedonia Greece
| | - Spyridon Siafis
- B’ Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Aristotle University of Thessaloniki, Stilponos Kiriakides str. 1, 546 36 Thessaloniki, Central Macedonia Greece
| | - Theodoros Tselios
- Department of Chemistry, University of Patras, Rion, 265 04 Patras, Greece
| | - Athanasios Lourbopoulos
- B’ Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Aristotle University of Thessaloniki, Stilponos Kiriakides str. 1, 546 36 Thessaloniki, Central Macedonia Greece
- Institute for Stroke and Dementia Research (ISD), Feodor-Lynen-Strasse 17, 81377 Munich, Germany
| | - Dimitrios Karacostas
- B’ Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Aristotle University of Thessaloniki, Stilponos Kiriakides str. 1, 546 36 Thessaloniki, Central Macedonia Greece
| | - Nikolaos Grigoriadis
- B’ Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Aristotle University of Thessaloniki, Stilponos Kiriakides str. 1, 546 36 Thessaloniki, Central Macedonia Greece
| | - Constantina Simeonidou
- Department of Experimental Physiology, Faculty of Medicine, Aristotle University of Thessaloniki, 546 36 Thessaloniki, Central Macedonia Greece
| |
Collapse
|
17
|
Role of IL-10 in Resolution of Inflammation and Functional Recovery after Peripheral Nerve Injury. J Neurosci 2016; 35:16431-42. [PMID: 26674868 DOI: 10.1523/jneurosci.2119-15.2015] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
UNLABELLED A rapid proinflammatory response after peripheral nerve injury is required for clearance of tissue debris (Wallerian degeneration) and effective regeneration. Unlike the CNS, this response is rapidly terminated in peripheral nerves starting between 2 and 3 weeks after crush injury. We examined the expression and role of the anti-inflammatory cytokine IL-10 in the resolution of inflammation and regeneration after sciatic nerve crush injury in mice. IL-10 mRNA increased over the first 7 d after injury, whereas at the protein level, immunofluorescence labeling showed IL-10(+) cells increased almost 3-fold in the first 3 weeks, with macrophages being the major cell type expressing IL-10. The role of IL-10 in nerve injury was assessed using IL-10-null mice. Increased numbers of macrophages were found in the distal segment of IL-10-null mice at early (3 d) and late (14 and 21 d) time points, suggesting that IL-10 may play a role in controlling the early influx and the later efflux of macrophages out of the nerve. A chemokine/cytokine PCR array of the nerve 24 h after crush showed a 2- to 4-fold increase in the expression of 10 proinflammatory mediators in IL-10(-/-) mice. In addition, myelin phagocytosis in vitro by LPS stimulated bone-marrow-derived macrophages from IL-10-null mice failed to downregulate expression of proinflammatory chemokines/cytokines, suggesting that IL-10 is required for the myelin-phagocytosis-induced shift of macrophages from proinflammatory to anti-inflammatory/pro-repair phenotype. The failure to switch off inflammation in IL-10-null mice was accompanied by impaired axon regeneration and poor recovery of motor and sensory function. SIGNIFICANCE STATEMENT An appropriately regulated inflammatory response after peripheral nerve injury is essential for axon regeneration and recovery. The aim of this study was to investigate the expression and role of the anti-inflammatory cytokine IL-10 in terminating inflammation after sciatic nerve crush injury and promoting regeneration. IL-10 is rapidly expressed by macrophages after crush injury. Its role was assessed using IL-10-null mice, which showed that IL-10 plays a role in controlling the early influx and the later efflux of macrophages out of the injured nerve, reduces the expression of proinflammatory chemokines and cytokines, and is required for myelin-phagocytosis-induced shift of macrophages from proinflammatory to anti-inflammatory. Furthermore, lack of IL-10 leads to impaired axon regeneration and poor recovery of motor and sensory function.
Collapse
|
18
|
An H, Brettle M, Lee T, Heng B, Lim CK, Guillemin GJ, Lord MS, Klotzsch E, Geczy CL, Bryant K, Fath T, Tedla N. Soluble LILRA3 promotes neurite outgrowth and synapses formation through high affinity interaction with Nogo 66. J Cell Sci 2016; 129:1198-209. [DOI: 10.1242/jcs.182006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 01/26/2016] [Indexed: 01/24/2023] Open
Abstract
Inhibitory proteins, particularly Nogo 66, a highly conserved 66 amino acid loop of Nogo A, play key roles in limiting the intrinsic capacity of the central nervous system to regenerate after injury. Ligation of surface Nogo receptors (NgRs) and/or leukocyte immunoglobulin like receptor B2 (LILRB2) and its mouse orthologue the paired-immunoglobulin-like receptor B (PIRB) by Nogo 66 transduces inhibitory signals that potently inhibit neurite outgrowth. Here we show that soluble leukocyte immunoglobulin-like receptor A3 (LILRA3) is a high affinity receptor for Nogo 66, suggesting that LILRA3 might be a competitive antagonist to these cell surface inhibitory receptors. Consistent with this, LILRA3 significantly reversed Nogo 66-mediated inhibition of neurite outgrowth and promoted synapse formation in primary cortical neurons via regulation of the MEK/ERK pathway. LILRA3 represents a new antagonist to Nogo 66-mediated inhibition of neurite outgrowth in the CNS, a function distinct from its immune-regulatory role in leukocytes. This report is also the first to demonstrate that a member of LILR family normally not expressed in rodents exerts functions on mouse neurons through the highly homologous Nogo 66 ligand.
Collapse
Affiliation(s)
- Hongyan An
- Inflammation and Infection Research Centre, School of Medical Sciences, Department of Pathology, UNSW, Sydney, Australia
| | - Merryn Brettle
- Neurodegeneration and Repair Unit, School of Medical Sciences, Department of Anatomy, UNSW, Sydney, Australia
| | - Terry Lee
- Inflammation and Infection Research Centre, School of Medical Sciences, Department of Pathology, UNSW, Sydney, Australia
| | - Benjamin Heng
- Faculty of Medicine and Health Sciences, Department of Biomedical Sciences, Macquarie University, Australia
| | - Chai K. Lim
- Faculty of Medicine and Health Sciences, Department of Biomedical Sciences, Macquarie University, Australia
| | - Gilles J. Guillemin
- Faculty of Medicine and Health Sciences, Department of Biomedical Sciences, Macquarie University, Australia
| | - Megan S. Lord
- Graduate School of Biomedical Engineering, UNSW, Sydney, NSW 2052, Australia
| | - Enrico Klotzsch
- EMBL Australia Node in Single Molecule Science, School of Medical Sciences, ARC Centre of Excellence in Advanced Molecular Imaging, The University of New South Wales, Sydney, NSW, Australia
| | - Carolyn L. Geczy
- Inflammation and Infection Research Centre, School of Medical Sciences, Department of Pathology, UNSW, Sydney, Australia
| | - Katherine Bryant
- Inflammation and Infection Research Centre, School of Medical Sciences, Department of Pathology, UNSW, Sydney, Australia
| | - Thomas Fath
- Neurodegeneration and Repair Unit, School of Medical Sciences, Department of Anatomy, UNSW, Sydney, Australia
| | - Nicodemus Tedla
- Inflammation and Infection Research Centre, School of Medical Sciences, Department of Pathology, UNSW, Sydney, Australia
| |
Collapse
|
19
|
Bexarotene-Activated Retinoid X Receptors Regulate Neuronal Differentiation and Dendritic Complexity. J Neurosci 2015; 35:11862-76. [PMID: 26311769 DOI: 10.1523/jneurosci.1001-15.2015] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
UNLABELLED Bexarotene-activated retinoid X receptors (RXRs) ameliorate memory deficits in Alzheimer's disease mouse models, including mice expressing human apolipoprotein E (APOE) isoforms. The goal of this study was to gain further insight into molecular mechanisms whereby ligand-activated RXR can affect or restore cognitive functions. We used an unbiased approach to discover genome-wide changes in RXR cistrome (ChIP-Seq) and gene expression profile (RNA-Seq) in response to bexarotene in the cortex of APOE4 mice. Functional categories enriched in both datasets revealed that bexarotene-liganded RXR affected signaling pathways associated with neurogenesis and neuron projection development. To further validate the significance of RXR for these functions, we used mouse embryonic stem (ES) cells, primary neurons, and APOE3 and APOE4 mice treated with bexarotene. In vitro data from ES cells confirmed that bexarotene-activated RXR affected neuronal development at different levels, including proliferation of neural progenitors and neuronal differentiation, and stimulated neurite outgrowth. This effect was validated in vivo by demonstrating an increased number of neuronal progenitors after bexarotene treatment in the dentate gyrus of APOE3 and APOE4 mice. In primary neurons, bexarotene enhanced the dendritic complexity characterized by increased branching, intersections, and bifurcations. This effect was confirmed by in vivo studies demonstrating that bexarotene significantly improved the compromised dendritic structure in the hippocampus of APOE4 mice. We conclude that bexarotene-activated RXRs promote genetic programs involved in the neurogenesis and development of neuronal projections and these results have significance for the improvement of cognitive deficits. SIGNIFICANCE STATEMENT Bexarotene-activated retinoid X receptors (RXRs) ameliorate memory deficits in Alzheimer's disease mouse models, including mice expressing human apolipoprotein E (APOE) isoforms. The goal of this study was to gain further insight into molecular mechanisms whereby ligand-activated RXR can affect or restore cognitive functions. We used an unbiased approach to discover genome-wide changes in RXR cistrome (ChIP-Seq) and gene expression profile (RNA-Seq) in response to bexarotene in the cortex of APOE4 mice. Functional categories enriched in both datasets revealed that liganded RXR affected signaling pathways associated with neurogenesis and neuron projection development. The significance of RXR for these functions was validated in mouse embryonic stem cells, primary neurons, and APOE3 and APOE4 mice treated with bexarotene.
Collapse
|
20
|
Tong J, Ren Y, Wang X, Dimopoulos VG, Kesler HN, Liu W, He X, Nedergaard M, Huang JH. Assessment of Nogo-66 receptor 1 function in vivo after spinal cord injury. Neurosurgery 2015; 75:51-60. [PMID: 24594926 DOI: 10.1227/neu.0000000000000337] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
BACKGROUND Neuronal Nogo-66 receptor 1 (NgR1) has attracted attention as a converging point for mediating the effects of myelin-associate inhibitory ligands in the central nervous system, establishing the growth-restrictive environment, and limiting axon regeneration after traumatic injury. OBJECTIVE To investigate the factors that may be contributing to the discrepancy in the importance of NgR1, which has been undermined by several studies that have shown the lack of substantial axon regeneration after spinal cord injury (SCI) in NgR1-knockout or -knockdown animal models. METHODS We used mice carrying either a homozygous or heterozygous null mutation in the NgR1 gene and subjected them to either a moderate or severe SCI. RESULTS Locomotor function assessments revealed that the level of functional recovery is affected by the degree of injury suffered. NgR1 ablation enhanced local collateral sprouting in the mutant mice. Reactive astrocytes and chondroitin sulfate proteoglycans (CSPGs) are upregulated surrounding the injury site. Matrix metalloproteinase-9, which has been shown to degrade CSPGs, was significantly upregulated in the homozygous mutant mice compared with the heterozygous or wild-type mice. However, CSPG levels remained higher in the homozygous compared with the heterozygous mice, suggesting that CSPG-degrading activity of matrix metalloproteinase-9 may require the presence of NgR1. CONCLUSION Genetic ablation of NgR1 may lead to significant recovery in locomotor function after SCI. The difference in locomotor recovery we observed between the groups that suffered various degrees of injury suggests that injury severity may be a confounding factor in functional recovery after SCI.
Collapse
Affiliation(s)
- Jing Tong
- ‡Department of Neurosurgery and ‖Center for Translational Neuromedicine, University of Rochester, Rochester, New York; §Department of Neurosurgery, Fourth Affiliated Hospital of Hebei Medical University, Hebei, China; ¶Department of Neurosurgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China; and #Department of Neurosurgery, Baylor Scott & White Health, Temple, Texas
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Meeker RB, Williams KS. The p75 neurotrophin receptor: at the crossroad of neural repair and death. Neural Regen Res 2015; 10:721-5. [PMID: 26109945 PMCID: PMC4468762 DOI: 10.4103/1673-5374.156967] [Citation(s) in RCA: 127] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2015] [Indexed: 12/14/2022] Open
Abstract
The strong repair and pro-survival functions of neurotrophins at their primary receptors, TrkA, TrkB and TrkC, have made them attractive candidates for treatment of nervous system injury and disease. However, difficulties with the clinical implementation of neurotrophin therapies have prompted the search for treatments that are stable, easier to deliver and allow more precise regulation of neurotrophin actions. Recently, the p75 neurotrophin receptor (p75NTR) has emerged as a potential target for pharmacological control of neurotrophin activity, supported in part by studies demonstrating 1) regulation of neural plasticity in the mature nervous system, 2) promotion of adult neurogenesis and 3) increased expression in neurons, macrophages, microglia, astrocytes and/or Schwann cells in response to injury and neurodegenerative diseases. Although the receptor has no intrinsic catalytic activity it interacts with and modulates the function of TrkA, TrkB, and TrkC, as well as sortilin and the Nogo receptor. This provides substantial cellular and molecular diversity for regulation of neuron survival, neurogenesis, immune responses and processes that support neural function. Upregulation of the p75NTR under pathological conditions places the receptor in a key position to control numerous processes necessary for nervous system recovery. Support for this possibility has come from recent studies showing that small, non-peptide p75NTR ligands can selectively modify pro-survival and repair functions. While a great deal remains to be discovered about the wide ranging functions of the p75NTR, studies summarized in this review highlight the immense potential for development of novel neuroprotective and neurorestorative therapies.
Collapse
Affiliation(s)
- Rick B Meeker
- Department of Neurology, University of North Carolina, Chapel Hill, NC, USA
| | - Kimberly S Williams
- Curriculum in Neurobiology, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
22
|
Lang BT, Wang J, Filous AR, Au NPB, Ma CHE, Shen Y. Pleiotropic molecules in axon regeneration and neuroinflammation. Exp Neurol 2014; 258:17-23. [DOI: 10.1016/j.expneurol.2014.04.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 04/21/2014] [Accepted: 04/29/2014] [Indexed: 12/20/2022]
|
23
|
Brosius Lutz A, Barres BA. Contrasting the Glial Response to Axon Injury in the Central and Peripheral Nervous Systems. Dev Cell 2014; 28:7-17. [DOI: 10.1016/j.devcel.2013.12.002] [Citation(s) in RCA: 158] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
24
|
Litwak SA, Payne NL, Campanale N, Ozturk E, Lee JY, Petratos S, Siatskas C, Bakhuraysah M, Bernard CCA. Nogo-receptor 1 deficiency has no influence on immune cell repertoire or function during experimental autoimmune encephalomyelitis. PLoS One 2013; 8:e82101. [PMID: 24339996 PMCID: PMC3855334 DOI: 10.1371/journal.pone.0082101] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2013] [Accepted: 10/30/2013] [Indexed: 12/03/2022] Open
Abstract
The potential role of Nogo-66 Receptor 1 (NgR1) on immune cell phenotypes and their activation during neuroinflammatory diseases such as multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE), is unclear. To further understand the function of this receptor on haematopoietically-derived cells, phenotypic and functional analyses were performed using NgR1-deficient (ngr1-/-) animals. Flow cytometry-based phenotypic analyses performed on blood, spleen, thymus, lymph nodes, bone marrow and central nervous-system (CNS)-infiltrating blood cells revealed no immunological defects in naïve ngr1-/- animals versus wild-type littermate (WTLM) controls. EAE was induced by either recombinant myelin oligodendrocyte glycoprotein (rMOG), a model in which B cells are considered to contribute pathogenically, or by MOG35–55 peptide, a B cell-independent model. We have demonstrated that in ngr1-/- mice injected with MOG35–55, a significant reduction in the severity of EAE correlated with reduced axonal damage present in the spinal cord when compared to their WTLM controls. However, despite a reduction in axonal damage observed in the CNS of ngr1-/- mice at the chronic stage of disease, no clinical differences could be attributed to a specific genotype when rMOG was used as the encephalitogen. Following MOG35–55-induction of EAE, we could not derive any major changes to the immune cell populations analyzed between ngr1-/- and WTLM mice. Collectively, these data demonstrate that NgR1 has little if any effects on the repertoire of immune cells, their activation and trafficking to the CNS.
Collapse
Affiliation(s)
- Sara A. Litwak
- Multiple Sclerosis Research Group, Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Natalie L. Payne
- Multiple Sclerosis Research Group, Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Naomi Campanale
- Multiple Sclerosis Research Group, Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Ezgi Ozturk
- Multiple Sclerosis Research Group, Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Jae Young Lee
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Steven Petratos
- Central Clinical School, Monash University, Prahran, Victoria, Australia
| | - Christopher Siatskas
- Multiple Sclerosis Research Group, Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Maha Bakhuraysah
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Claude C. A. Bernard
- Multiple Sclerosis Research Group, Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
- * E-mail:
| |
Collapse
|
25
|
He BR, Xie ST, Wu MM, Hao DJ, Yang H. Phagocytic Removal of Neuronal Debris by Olfactory Ensheathing Cells Enhances Neuronal Survival and Neurite Outgrowth via p38MAPK Activity. Mol Neurobiol 2013; 49:1501-12. [DOI: 10.1007/s12035-013-8588-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 11/05/2013] [Indexed: 01/23/2023]
|
26
|
Roloff F, Ziege S, Baumgärtner W, Wewetzer K, Bicker G. Schwann cell-free adult canine olfactory ensheathing cell preparations from olfactory bulb and mucosa display differential migratory and neurite growth-promoting properties in vitro. BMC Neurosci 2013; 14:141. [PMID: 24219805 PMCID: PMC3840578 DOI: 10.1186/1471-2202-14-141] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Accepted: 11/07/2013] [Indexed: 12/04/2022] Open
Abstract
Background Transplantation of olfactory ensheathing cells (OEC) and Schwann cells (SC) is a promising therapeutic strategy to promote axonal growth and remyelination after spinal cord injury. Previous studies mainly focused on the rat model though results from primate and porcine models differed from those in the rat model. Interestingly, canine OECs show primate-like in vitro characteristics, such as absence of early senescence and abundance of stable p75NTR expression indicating that this species represents a valuable translational species for further studies. So far, few investigations have tested different glial cell types within the same study under identical conditions. This makes it very difficult to evaluate contradictory or confirmatory findings reported in various studies. Moreover, potential contamination of OEC preparations with Schwann cells was difficult to exclude. Thus, it remains rather controversial whether the different glial types display distinct cellular properties. Results Here, we established cultures of Schwann cell-free OECs from olfactory bulb (OB-OECs) and mucosa (OM-OECs) and compared them in assays to Schwann cells. These glial cultures were obtained from a canine large animal model and used for monitoring migration, phagocytosis and the effects on in vitro neurite growth. OB-OECs and Schwann cells migrated faster than OM-OECs in a scratch wound assay. Glial cell migration was not modulated by cGMP and cAMP signaling, but activating protein kinase C enhanced motility. All three glial cell types displayed phagocytic activity in a microbead assay. In co-cultures with of human model (NT2) neurons neurite growth was maximal on OB-OECs. Conclusions These data provide evidence that OB- and OM-OECs display distinct migratory behavior and interaction with neurites. OB-OECs migrate faster and enhance neurite growth of human model neurons better than Schwann cells, suggesting distinct and inherent properties of these closely-related cell types. Future studies will have to address whether, and how, these cellular properties correlate with the in vivo behavior after transplantation.
Collapse
Affiliation(s)
| | | | | | | | - Gerd Bicker
- Division of Cell Biology, University of Veterinary Medicine Hannover, Bischofsholer Damm 15/102, 30173 Hannover, Germany.
| |
Collapse
|
27
|
Su Z, Chen J, Qiu Y, Yuan Y, Zhu F, Zhu Y, Liu X, Pu Y, He C. Olfactory ensheathing cells: the primary innate immunocytes in the olfactory pathway to engulf apoptotic olfactory nerve debris. Glia 2013; 61:490-503. [PMID: 23339073 DOI: 10.1002/glia.22450] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2012] [Accepted: 11/09/2012] [Indexed: 11/11/2022]
Abstract
The olfactory system is an unusual tissue in which olfactory receptor neurons (ORNs) are continuously replaced throughout the life of mammals. Clearance of the apoptotic ORNs corpses is a fundamental process serving important functions in the regulation of olfactory nerve turnover and regeneration. However, little is known about the underlying mechanisms. Olfactory ensheathing cells (OECs) are a unique type of glial cells that wrap olfactory axons and support their continual regeneration from the olfactory epithelium to the bulb. In the present study, OECs were identified to exist in two different states, resting and reactive, in which resting OECs could be activated by LPS stimulation and functioned as phagocytes for cleaning apoptotic ORNs corpses. Confocal analysis revealed that dead ORNs debris were engulfed by OECs and co-localized with lysosome associated membrane protein 1. Moreover, phosphatidylserine (PS) receptor was identified to express on OECs, which allowed OECs to recognize apoptotic ORNs by binding to PS. Importantly, engulfment of olfactory nerve debris by OECs was found in olfactory mucosa under normal turnover and was significantly increased in the animal model of olfactory bulbectomy, while little phagocytosis by Iba-1-positive microglia/macrophages was observed. Together, these results implicate OEC as a primary innate immunocyte in the olfactory pathway, and suggest a cellular and molecular mechanism by which ORNs corpses are removed during olfactory nerve turnover and regeneration.
Collapse
Affiliation(s)
- Zhida Su
- Institute of Neuroscience and Key Laboratory of Molecular Neurobiology of Ministry of Education, Neuroscience Research Center of Changzheng Hospital, Second Military Medical University, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Elias PZ, Spector M. Treatment of penetrating brain injury in a rat model using collagen scaffolds incorporating soluble Nogo receptor. J Tissue Eng Regen Med 2012; 9:137-50. [PMID: 23038669 DOI: 10.1002/term.1621] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Revised: 05/18/2012] [Accepted: 08/25/2012] [Indexed: 02/06/2023]
Abstract
Injuries and diseases of the central nervous system (CNS) have the potential to cause permanent loss of brain parenchyma, with severe neurological consequences. Cavitary defects in the brain may afford the possibility of treatment with biomaterials that fill the lesion site while delivering therapeutic agents. This study examined the treatment of penetrating brain injury (PBI) in a rat model with collagen biomaterials and a soluble Nogo receptor (sNgR) molecule. sNgR was aimed at neutralizing myelin proteins that hinder axon regeneration by inducing growth cone collapse. Scaffolds containing sNgR were implanted in the brains of adult rats 1 week after injury and analysed 4 weeks or 8 weeks later. Histological analysis revealed that the scaffolds filled the lesion sites, remained intact with open pores and were infiltrated with cells and extracellular matrix. Immunohistochemical staining demonstrated the composition of the cellular infiltrate to include macrophages, astrocytes and vascular endothelial cells. Isolated regions of the scaffold borders showed integration with surrounding viable brain tissue that included neurons and oligodendrocytes. While axon regeneration was not detected in the scaffolds, the cellular infiltration and vascularization of the lesion site demonstrated a modification of the injury environment with implications for regenerative strategies.
Collapse
Affiliation(s)
- Paul Z Elias
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA, USA; Tissue Engineering Laboratories, VA Boston Healthcare System, Boston, MA, USA
| | | |
Collapse
|
29
|
Chao PK, Lu KT, Jhu JY, Wo YYP, Huang TC, Ro LS, Yang YL. Indomethacin protects rats from neuronal damage induced by traumatic brain injury and suppresses hippocampal IL-1β release through the inhibition of Nogo-A expression. J Neuroinflammation 2012; 9:121. [PMID: 22676811 PMCID: PMC3416695 DOI: 10.1186/1742-2094-9-121] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Accepted: 03/08/2012] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Nogo-A is a member of the reticulon family of membrane-associated proteins and plays an important role in axonal remodeling. The present study aimed to investigate alterations in Nogo-A expression following traumatic brain injury (TBI)-induced inflammation and neuronal damage. METHODS A weight-drop device was used to deliver a standard traumatic impact to rats. Western blot, RT-PCR and ELISA were used to analyze the expression of Nogo-A and IL-1β. Nogo-A antisense, and an irrelevant control oligonucleotide was intracerebroventricularly infused. We also performed H & E staining and luxol fast blue staining to evaluate the neuronal damage and demyelination resulting from TBI and various treatments. RESULTS Based on RT-PCR and western blot analyses, the expression of Nogo-A was found to be significantly upregulated in the hippocampus beginning eight hours after TBI. In addition, TBI caused an apparent elevation in IL-1β levels and severe neuronal damage and demyelination in the tested animals. All of the TBI-associated molecular and cellular consequences could be effectively reversed by treating the animals with the anti-inflammatory drug indomethacin. More importantly, the TBI-associated stimulation in the levels of both Nogo-A and IL-1β could be effectively inhibited by a specific Nogo-A antisense oligonucleotide. CONCLUSIONS Our findings suggest that the suppression of Nogo-A expression appears to be an early response conferred by indomethacin, which then leads to decreases in the levels of IL-1β and TBI-induced neuron damage.
Collapse
Affiliation(s)
- Po-Kuan Chao
- Department of Life Science, National Taiwan Normal University, 88 Section 4, Ting-Chou Road, Taipei, Taiwan
| | | | | | | | | | | | | |
Collapse
|
30
|
Drosophila GPI-mannosyltransferase 2 is required for GPI anchor attachment and surface expression of chaoptin. Vis Neurosci 2012; 29:143-56. [PMID: 22575127 DOI: 10.1017/s0952523812000181] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Glycosylphosphatidylinositol (GPI) anchors are critical for the membrane attachment of a wide variety of essential signaling and cell adhesion proteins. The GPI anchor is a complex glycolipid structure that utilizes glycosylphosphatidylinositol-mannosyltransferases (GPI-MTs) for the addition of three core mannose residues during its biosynthesis. Here, we demonstrate that Drosophila GPI-MT2 is required for the GPI-mediated membrane attachment of several GPI-anchored proteins, including the photoreceptor-specific cell adhesion molecule, chaoptin. Mutations in gpi-mt2 lead to defects in chaoptin trafficking to the plasma membrane in Drosophila photoreceptor cells. In gpi-mt2 mutants, loss of sufficient chaoptin in the membrane leads to microvillar instability, photoreceptor cell pathology, and retinal degeneration. Finally, using site-directed mutagenesis, we have identified key amino acids that are essential for GPI-MT2 function and cell viability in Drosophila. Our findings on GPI-MT2 provide a mechanistic link between GPI anchor biosynthesis and protein trafficking in Drosophila and shed light on a novel mechanism for inherited retinal degeneration.
Collapse
|
31
|
Extrinsic cellular and molecular mediators of peripheral axonal regeneration. Cell Tissue Res 2012; 349:5-14. [PMID: 22476657 DOI: 10.1007/s00441-012-1389-5] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Accepted: 02/23/2012] [Indexed: 12/11/2022]
Abstract
The ability of injured peripheral nerves to regenerate and reinnervate their original targets is a characteristic feature of the peripheral nervous system (PNS). On the other hand, neurons of the central nervous system (CNS), including retinal ganglion cell (RGC) axons, are incapable of spontaneous regeneration. In the adult PNS, axonal regeneration after injury depends on well-orchestrated cellular and molecular processes that comprise a highly reproducible series of degenerative reactions distal to the site of injury. During this fine-tuned process, named Wallerian degeneration, a remodeling of the distal nerve fragment prepares a permissive microenvironment that permits successful axonal regrowth originating from the proximal nerve fragment. Therefore, a multitude of adjusted intrinsic and extrinsic factors are important for surviving neurons, Schwann cells, macrophages and fibroblasts as well as endothelial cells in order to achieve successful regeneration. The aim of this review is to summarize relevant extrinsic cellular and molecular determinants of successful axonal regeneration in rodents that contribute to the regenerative microenvironment of the PNS.
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW The inflammatory response that accompanies neural injury involves multiple cell types and effector molecules with both positive and negative effects. Inflammation is essential for normal regeneration in the peripheral nervous system, and here we review evidence that augmenting inflammation can enhance regeneration in areas of the central nervous system in which it normally does not occur. RECENT FINDINGS Within the spinal cord, inflammation enables transplanted sensory neurons to regenerate lengthy axons and enhances the ability of a trophic factor to promote corticospinal tract sprouting. Induction of inflammation in the eye supports survival of retinal ganglion cells and enables them to regenerate injured axons through the optic nerve. These effects are linked to an atypical trophic factor, oncomodulin, along with other, better known molecules. Induction of inflammation within dorsal root ganglia, when combined with other treatments, enables peripheral sensory neurons to regenerate axons into the spinal cord. However, inflammation also has negative effects that impede recovery. SUMMARY In light of the importance of inflammation for neural repair, it is important to identify the specific cell types and molecules responsible for the positive and negative effects of inflammation and to develop treatments that tip the balance to favor repair.
Collapse
|
33
|
Chinese Medicine's Intervention Effect on Nogo-A/NgR. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2012; 2012:528482. [PMID: 22216056 PMCID: PMC3247900 DOI: 10.1155/2012/528482] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Revised: 09/28/2011] [Accepted: 11/02/2011] [Indexed: 01/31/2023]
Abstract
Cerebral vascular disease is very common in the elderly and is one of the most dangerous diseases which is hazardous to the body's health, and it is the medical specialists' study hot spot not only in the clinical field but also in the medical basic research field. Neural regeneration has been paid more and more attention in recent years. Nogo's function in the process of neural regeneration has become the focal point since it was discovered in the year 2000. Many studies elucidate that Nogo negatively affects the neural regeneration and plasticity. Chinese medicine plays an important role in the prevention and treatment of neural diseases, and recently some researches about the Chinese medicine's intervention effect on Nogo-A/NgR sprang up, so it is necessary to make a review on this aspect.
Collapse
|
34
|
Abstract
Spinal cord injury (SCI) has multiple consequences, ranging from molecular imbalances to glial scar formation to functional impairments. It is logical to think that a combination of single treatments implemented in the right order and at the right time will be required to repair the spinal cord. However, the single treatments that compose the combination therapy will need to be chosen with caution as many have multiple outcomes that may or may not be synergistic. Single treatments may also elicit unwanted side-effects and/or effects that would decrease the repair potential of other components and/or the entire combination therapy. In this chapter a number of single treatments are discussed with respect to their multiplicity of action. These include strategies to boost growth and survival (such as neurotrophins and cyclic AMP) and strategies to reduce inhibitory factors (such as antimyelin-associated growth inhibitors and digestion of glial scar-associated inhibitors). We also present an overview of combination therapies that have successfully or unsuccessfully been tested in the laboratory using animal models. To effectively design a combination therapy a number of considerations need to be made such as the nature and timing of the treatments and the method for delivery. This chapter discusses these issues as well as considerations related to chronic SCI and the logistics of bringing combination therapies to the clinic.
Collapse
Affiliation(s)
- M Oudega
- Departments of Physical Medicine and Rehabilitation, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | | | | |
Collapse
|
35
|
Steinbach K, McDonald CL, Reindl M, Schweigreiter R, Bandtlow C, Martin R. Nogo-receptors NgR1 and NgR2 do not mediate regulation of CD4 T helper responses and CNS repair in experimental autoimmune encephalomyelitis. PLoS One 2011; 6:e26341. [PMID: 22096481 PMCID: PMC3214013 DOI: 10.1371/journal.pone.0026341] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 09/25/2011] [Indexed: 11/28/2022] Open
Abstract
Myelin-associated inhibition of axonal regrowth after injury is considered one important factor that contributes to regeneration failure in the adult central nervous system (CNS). Blocking strategies targeting this pathway have been successfully applied in several nerve injury models, including experimental autoimmune encephalomyelitis (EAE), suggesting myelin-associated inhibitors (MAIs) and functionally related molecules as targets to enhance regeneration in multiple sclerosis. NgR1 and NgR2 were identified as interaction partners for the myelin proteins Nogo-A, MAG and OMgp and are probably mediating their growth-inhibitory effects on axons, although the in vivo relevance of this pathway is currently under debate. Recently, alternative functions of MAIs and NgRs in the regulation of immune cell migration and T cell differentiation have been described. Whether and to what extent NgR1 and NgR2 are contributing to Nogo and MAG-related inhibition of neuroregeneration or immunomodulation during EAE is currently unknown. Here we show that genetic deletion of both receptors does not promote functional recovery during EAE and that NgR1 and NgR2-mediated signals play a minor role in the development of CNS inflammation. Induction of EAE in Ngr1/2-double mutant mice resulted in indifferent disease course and tissue damage when compared to WT controls. Further, the development of encephalitogenic CD4+ Th1 and Th17 responses was unchanged. However, we observed a slightly increased leukocyte infiltration into the CNS in the absence of NgR1 and NgR2, indicating that NgRs might be involved in the regulation of immune cell migration in the CNS. Our study demonstrates the urgent need for a more detailed knowledge on the multifunctional roles of ligands and receptors involved in CNS regeneration failure.
Collapse
Affiliation(s)
- Karin Steinbach
- Institute for Neuroimmunology and Clinical MS-Research, Hamburg, Germany
| | - Claire L. McDonald
- Clinical Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
| | - Markus Reindl
- Clinical Department of Neurology, Innsbruck Medical University, Innsbruck, Austria
| | | | - Christine Bandtlow
- Department of Neurobiochemistry, Innsbruck Medical University, Innsbruck, Austria
| | - Roland Martin
- Institute for Neuroimmunology and Clinical MS-Research, Hamburg, Germany
- * E-mail:
| |
Collapse
|
36
|
Wang YT, Lu XM, Zhu F, Huang P, Yu Y, Zeng L, Long ZY, Wu YM. The use of a gold nanoparticle-based adjuvant to improve the therapeutic efficacy of hNgR-Fc protein immunization in spinal cord-injured rats. Biomaterials 2011; 32:7988-98. [PMID: 21784510 DOI: 10.1016/j.biomaterials.2011.07.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Accepted: 07/04/2011] [Indexed: 10/18/2022]
Abstract
As a common receptor for three myelin associated inhibitors, Nogo-66 receptor (NgR) mediates their inhibitory activities on neurite outgrowth in the adult mammalian central nervous system (CNS). Therapeutic vaccination protocol targeting NgR emulsified with Freund's adjuvant (FA) has been used in spinal cord injury (SCI) models. However, the vaccine emulsified with FA may induce some side effects, which are not suitable for further clinical application. As an adjuvant, gold nanoparticles (GNPs) could stimulate a stronger immune response without producing detectable toxicity and physiological damage than FA. There is, however, uncertainty regarding the efficacy of axon regeneration and neuroprotection in vaccines with GNPs as an adjuvant. In this investigation, a recombinant protein vaccine targeting NgR, human NgR-Fc (hNgR-Fc) fusion protein conjugated with 15 nm GNPs was prepared and its effects on axonal regeneration and functional recovery in spinal cord-injured rats were investigated. The results showed that adult rats immunized with the protein vaccine produced higher titers of anti-NgR antibody than that with FA, and the antisera promoted neurite outgrowth in presence of MAG in vitro. In a spinal cord dorsal hemisection model, vaccine immunized with GNPs promoted axonal regeneration more effectively than FA, resulted in significant protection from neuronal loss, and improved functional recovery. Thus, as an adjuvant, 15 nm GNPs can effectively boost the immunogenicity of hNgR-Fc protein vaccine, and promote the repair of spinal cord-injured rats. The utilization of GNPs, for clinical considerations, may be a more beneficial supplement than FA to the promising therapeutic vaccination strategy for promoting SCI repair.
Collapse
Affiliation(s)
- Yong-Tang Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing 400042, China.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
David S, Kroner A. Repertoire of microglial and macrophage responses after spinal cord injury. Nat Rev Neurosci 2011; 12:388-99. [PMID: 21673720 DOI: 10.1038/nrn3053] [Citation(s) in RCA: 1055] [Impact Index Per Article: 75.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Macrophages from the peripheral circulation and those derived from resident microglia are among the main effector cells of the inflammatory response that follows spinal cord trauma. There has been considerable debate in the field as to whether the inflammatory response is good or bad for tissue protection and repair. Recent studies on macrophage polarization in non-neural tissues have shed much light on their changing functional states. In the context of this literature, we discuss the activation of macrophages and microglia following spinal cord injury, and their effects on repair. Harnessing their anti-inflammatory properties could pave the way for new therapeutic strategies for spinal cord trauma.
Collapse
Affiliation(s)
- Samuel David
- The Research Institute of the McGill University Health Center, 1650 Cedar Avenue, Montreal, Quebec, Canada, H3G 1A4.
| | | |
Collapse
|
38
|
Gensel JC, Donnelly DJ, Popovich PG. Spinal cord injury therapies in humans: an overview of current clinical trials and their potential effects on intrinsic CNS macrophages. Expert Opin Ther Targets 2011; 15:505-18. [PMID: 21281256 DOI: 10.1517/14728222.2011.553605] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION Macrophage activation is a hallmark of spinal cord injury (SCI) pathology. CNS macrophages, derived from resident microglia and blood monocytes, are ubiquitous throughout the injured spinal cord, and respond to signals in the lesion environment by changing their phenotype and function. Depending on their phenotype and activation status, macrophages may initiate secondary injury mechanisms and/or promote CNS regeneration and repair. AREAS COVERED This review provides a comprehensive overview of current SCI clinical trials that are intended to promote neuroprotection, axon regeneration or cell replacement. None of these potential therapies were developed with the goal of influencing macrophage function; however, it is likely that each will have direct or indirect effects on CNS macrophages. The potential impact of each trial is discussed in the context of CNS macrophage biology. EXPERT OPINION Activation of CNS macrophages is an inevitable consequence of traumatic SCI. Given that these cells are exquisitely sensitive to changes in microenvironment, any intervention that affects tissue integrity and/or the composition of the cellular milieu will undoubtedly affect CNS macrophages. Thus, it is important to understand how current clinical trials will affect intrinsic CNS macrophages.
Collapse
Affiliation(s)
- John C Gensel
- The Ohio State University College of Medicine, Center for Brain and Spinal Cord Repair, 795 Biomedical Research Tower, 460 West 12th Avenue, Columbus, OH 43210-1239, USA
| | | | | |
Collapse
|
39
|
The age- and amyloid-β-related increases in Nogo B contribute to microglial activation. Neurochem Int 2010; 58:161-8. [PMID: 21111015 DOI: 10.1016/j.neuint.2010.11.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2010] [Accepted: 11/10/2010] [Indexed: 12/13/2022]
Abstract
The family of reticulons include three isoforms of the Nogo protein, Nogo A, Nogo B and Nogo C. Nogo A is expressed on neuronal tissue and its primary effect is widely acknowledged to be inhibition of neurite outgrowth. Although both Nogo B and Nogo C are also expressed in neuronal tissue, their roles in the CNS remain to be identified. In this study, we set out to assess whether expression of Nogo A or Nogo B was altered in tissue prepared from aged rats in which increased microglial activation is accompanied by decreased synaptic plasticity. The data indicate that Nogo B, but not Nogo A, was markedly increased in hippocampal tissue prepared from aged rats and that, at least in vitro, Nogo B increased several markers of microglial activation. In a striking parallel with the age-related changes, we demonstrate that intracerebroventricular delivery of amyloid-β (Aβ)(1-40)+Aβ(1-42) for 8 days was associated with a depression of long-term potentiation (LTP) and an increase in markers of microglial activation and Nogo B. In both models, evidence of cell stress was identified by increased activity of caspases 8 and 3 and importantly, incubation of cultured neurons in the presence of Nogo B increased activity of both enzymes. The data identify, for the first time, an effect of Nogo B in the brain and specifically show that its expression is increased in conditions where synaptic plasticity is compromised.
Collapse
|
40
|
Llorens F, Gil V, del Río JA. Emerging functions of myelin-associated proteins during development, neuronal plasticity, and neurodegeneration. FASEB J 2010; 25:463-75. [PMID: 21059749 DOI: 10.1096/fj.10-162792] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Adult mammalian central nervous system (CNS) axons have a limited regrowth capacity following injury. Myelin-associated inhibitors (MAIs) limit axonal outgrowth, and their blockage improves the regeneration of damaged fiber tracts. Three of these proteins, Nogo-A, MAG, and OMgp, share two common neuronal receptors: NgR1, together with its coreceptors [p75(NTR), TROY, and Lingo-1]; and the recently described paired immunoglobulin-like receptor B (PirB). These proteins impair neuronal regeneration by limiting axonal sprouting. Some of the elements involved in the myelin inhibitory pathways may still be unknown, but the discovery that blocking both PirB and NgR1 activities leads to near-complete release from myelin inhibition, sheds light on one of the most competitive and intense fields of neuroregeneration study in recent decades. In parallel with the identification and characterization of the roles and functions of these inhibitory molecules in axonal regeneration, data gathered in the field strongly suggest that most of these proteins have roles other than axonal growth inhibition. The discovery of a new group of interacting partners for myelin-associated receptors and ligands, as well as functional studies within or outside the CNS environment, highlights the potential new physiological roles for these proteins in processes, such as development, neuronal homeostasis, plasticity, and neurodegeneration.
Collapse
Affiliation(s)
- Franc Llorens
- Molecular and Cellular Neurobiotechnology, Institute for Bioengineering of Catalonia, and Department of Cell Biology, University of Barcelona, Barcelona, Spain
| | | | | |
Collapse
|
41
|
Schwab ME. Functions of Nogo proteins and their receptors in the nervous system. Nat Rev Neurosci 2010; 11:799-811. [PMID: 21045861 DOI: 10.1038/nrn2936] [Citation(s) in RCA: 302] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The membrane protein Nogo-A was initially characterized as a CNS-specific inhibitor of axonal regeneration. Recent studies have uncovered regulatory roles of Nogo proteins and their receptors--in precursor migration, neurite growth and branching in the developing nervous system--as well as a growth-restricting function during CNS maturation. The function of Nogo in the adult CNS is now understood to be that of a negative regulator of neuronal growth, leading to stabilization of the CNS wiring at the expense of extensive plastic rearrangements and regeneration after injury. In addition, Nogo proteins interact with various intracellular components and may have roles in the regulation of endoplasmic reticulum (ER) structure, processing of amyloid precursor protein and cell survival.
Collapse
Affiliation(s)
- Martin E Schwab
- University of Zurich and ETH, Zurich, Winterthurerstrasse 190, CH-8057 Zurich, Switzerland.
| |
Collapse
|
42
|
Zhu Z, Ni B, Yin G, Zhou F, Liu J, Guo Q, Guo X. NgR expression in macrophages promotes nerve regeneration after spinal cord injury in rats. Arch Orthop Trauma Surg 2010; 130:945-51. [PMID: 20179954 DOI: 10.1007/s00402-010-1065-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2009] [Indexed: 10/19/2022]
Abstract
OBJECTIVE This study aimed to investigate the expression of Nogo-66 receptor (NgR) in macrophages after SCI and clarify its role in neuron regeneration. METHODS Macrophages harvested from injured spine cord of rats were stained by double immunofluorescence labeling technique to observe the expression of NgR at histological and cellular levels. Macrophages which expressed NgR were constructed in vitro, and then the effects of NgR on macrophage phagocytosis and neuraxon regeneration in three groups (NgR-macrophages group, mock group and normal macrophages group) were studied using Western blot, micro-MTT colorimetry, and LDH assay separately. RESULTS The results showed that CD68-positive macrophages in injured tissue of spine cord expressed NgR after double immunofluorescence staining on day 7 after SCI, and so did macrophages isolated and cultured from the injured spine cord. The results of Western blot showed that phagocytosis of macrophages in NgR-macrophages group was much better than that in mock group and normal macrophage group (p < 0.05). And the results of Micro-MTT colorimetry and LDH assay indicated that the capacity of neuraxon regeneration in NgR-macrophages group was significantly higher than that in the other two groups (p < 0.05). CONCLUSIONS The results suggested that there was NgR expressing in the infiltrated macrophages following SCI, which increased phagocytosis of the macrophages, and promoted post-SCI CNS regeneration in vitro.
Collapse
Affiliation(s)
- Zhuangchen Zhu
- Department of Orthopedics, Changzheng Hospital, The Second Military Medical University, Huangpu District, Shanghai, People's Republic of China.
| | | | | | | | | | | | | |
Collapse
|
43
|
Nagamoto-Combs K, Morecraft RJ, Darling WG, Combs CK. Long-term gliosis and molecular changes in the cervical spinal cord of the rhesus monkey after traumatic brain injury. J Neurotrauma 2010; 27:565-85. [PMID: 20030560 DOI: 10.1089/neu.2009.0966] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Recovery of fine motor skills after traumatic brain injury (TBI) is variable, with some patients showing progressive improvements over time while others show poor recovery. We therefore studied possible cellular mechanisms accompanying the recovery process in a non-human primate model system, in which the lateral frontal motor cortex areas controlling the preferred upper limb were unilaterally lesioned, and the animals eventually regained fine hand motor function. Immunohistochemical staining of the cervical spinal cord, the site of compensatory sprouting and degeneration of corticospinal axons, showed profound increases in immunoreactivities for major histocompatibility complex class II molecule (MHC-II) and extracellular signal-regulated kinases (ERK1/2) up to 12 months post lesion, particularly within the lateral corticospinal tract (LCST). Double immunostaining demonstrated that phosphorylated ERK1/2 colocalized within the MCH-II + microglia, suggesting a trophic role of long-term microglia activation after TBI at the site of compensatory sprouting. Active sprouting was observed in the LCST as well as in the spinal gray matter of the lesioned animals, as illustrated by increases in growth associated protein 43. Upregulation of Nogo receptor and glutamate transporter expression was also observed in this region after TBI, suggesting possible mechanisms for controlling aberrant sprouting and/or synaptic formation en route and interstitial glutamate concentration changes at the site of axon degeneration, respectively. Taken together, these changes in the non-human primate spinal cord support a long-term trophic/tropic role for reactive microglia, in particular, during functional and structural recovery after TBI.
Collapse
Affiliation(s)
- Kumi Nagamoto-Combs
- Department of Anatomy and Cell Biology, University of North Dakota School of Medicine and Health Sciences, Grand Forks, North Dakota 58202, USA
| | | | | | | |
Collapse
|
44
|
Udina E, Ladak A, Furey M, Brushart T, Tyreman N, Gordon T. Rolipram-induced elevation of cAMP or chondroitinase ABC breakdown of inhibitory proteoglycans in the extracellular matrix promotes peripheral nerve regeneration. Exp Neurol 2010; 223:143-52. [PMID: 19733561 PMCID: PMC3071985 DOI: 10.1016/j.expneurol.2009.08.026] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2009] [Revised: 08/25/2009] [Accepted: 08/26/2009] [Indexed: 11/24/2022]
Abstract
The inhibitory growth environment of myelin and extracellular matrix proteoglycans in the central nervous system may be overcome by elevating neuronal cAMP or degrading inhibitory proteoglycans with chondroitinase ABC (ChABC). In this study, we asked whether similar mechanisms operate in peripheral nerve regeneration where effective Wallerian degeneration removes myelin and extracellular proteoglycans slowly. We repaired transected common peroneal (CP) nerve in rats and either elevated cAMP in the axotomized neurons by subcutaneous rolipram, a specific inhibitor of phosphodiesterase IV, and/or promoted degradation of proteoglycans in the distal nerve stump by local ChABC administration. Rolipram treatment significantly increased the number of motoneurons that regenerated axons across the repair site at 1 and 2 weeks, and increased the number of sensory neurons that regenerated axons across the repair site at 2 weeks. Local application of ChABC had a similar effect to rolipram treatment in promoting motor axon regeneration, the effect being no greater when rolipram and ChABC were administered simultaneously. We conclude that blocking inhibitors of axon regeneration by elevating cAMP or degrading proteoglycans in the distal nerve stump promotes peripheral axon regeneration after surgical repair of a transected nerve. It is likely that elevated cAMP is sufficient to encourage axon outgrowth despite the inhibitory growth environment such that simultaneous enzymatic proteoglycan degradation does not promote more axon regeneration than either elevated cAMP or proteoglycan degradation alone.
Collapse
Affiliation(s)
- E Udina
- Division of Physical Medicine and Rehabilitation, Faculty of Medicine and Dentistry, Center for Neurosciences, University of Alberta, Edmonton, Alberta, Canada T6G 2S2
| | | | | | | | | | | |
Collapse
|
45
|
Boulenguez P, Vinay L. Strategies to restore motor functions after spinal cord injury. Curr Opin Neurobiol 2009; 19:587-600. [PMID: 19896827 DOI: 10.1016/j.conb.2009.10.005] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Revised: 09/23/2009] [Accepted: 10/12/2009] [Indexed: 12/20/2022]
Abstract
This review presents recent advances in the development of strategies to restore posture and locomotion after spinal cord injury (SCI). A set of strategies focusing on the lesion site includes prevention of secondary damages, promotion of axonal sprouting/regeneration, and replacement of lost cells. Other strategies focus on spinal central pattern generators (CPGs). Training promotes functional recovery by enhancing the plasticity of CPGs and these sublesional networks can be reactivated by means of pharmacological or electrical stimulation. It is now clear that substantial functional recovery will require a combination of strategies adapted to each phase following SCI. Finally, improvements in the understanding of the mechanisms underlying spasticity may lead to new treatments of this disabling complication affecting patients with SCI.
Collapse
Affiliation(s)
- Pascale Boulenguez
- Laboratoire Plasticité et Physio-Pathologie de Motricité (UMR6196), Centre National de Recherche Scientifique (CNRS) & Aix-Marseille Université, CNRS, 31 chemin Joseph Aiguier, F-13402 Marseille cx 20, France
| | | |
Collapse
|
46
|
Identification of BLyS (B lymphocyte stimulator), a non-myelin-associated protein, as a functional ligand for Nogo-66 receptor. J Neurosci 2009; 29:6348-52. [PMID: 19439611 DOI: 10.1523/jneurosci.5040-08.2009] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
B lymphocyte stimulator (BLyS), a tumor necrosis factor family protein essential for B cell development, was previously shown to be expressed at an elevated level in the CNS of multiple sclerosis patients. Although it may be involved in CNS diseases, its exact functions in CNS remain unknown. We hypothesize that BLyS may be a negative regulator for neuronal functions. Here Nogo-66 receptor (NgR) is identified as a high affinity receptor for BLyS, which inhibits dorsal root ganglion outgrowth in culture. The inhibition by BLyS can be reversed by a truncated NgR or by removal of glycosylphosphatidylinositol-linked proteins from neurons. More importantly, the inhibitory effect by BLyS is significantly diminished for neurons isolated from NgR(-/-) mice. Furthermore, expressions of BLyS and NgR are also found to be associated with astrocytes and macrophages/microglial cells at spinal cord injury sites. Thus, BLyS can function independently of myelin-associated inhibitors and likely serves as a redundant NgR ligand that negatively influences axonal outgrowth in CNS.
Collapse
|
47
|
Robak LA, Venkatesh K, Lee H, Raiker SJ, Duan Y, Lee-Osbourne J, Hofer T, Mage RG, Rader C, Giger RJ. Molecular basis of the interactions of the Nogo-66 receptor and its homolog NgR2 with myelin-associated glycoprotein: development of NgROMNI-Fc, a novel antagonist of CNS myelin inhibition. J Neurosci 2009; 29:5768-83. [PMID: 19420245 PMCID: PMC2779053 DOI: 10.1523/jneurosci.4935-08.2009] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2008] [Revised: 11/10/2008] [Accepted: 03/12/2009] [Indexed: 01/17/2023] Open
Abstract
Myelin-associated glycoprotein (MAG) is a sialic acid-binding Ig-family lectin that functions in neuronal growth inhibition and stabilization of axon-glia interactions. The ectodomain of MAG is comprised of five Ig-like domains and uses neuronal cell-type-specific mechanisms to signal growth inhibition. We show that the first three Ig-like domains of MAG bind with high affinity and in a sialic acid-dependent manner to the Nogo-66 receptor-1 (NgR1) and its homolog NgR2. Domains Ig3-Ig5 of MAG are sufficient to inhibit neurite outgrowth but fail to associate with NgR1 or NgR2. Nogo receptors are sialoglycoproteins comprised of 8.5 canonical leucine-rich repeats (LRR) flanked by LRR N-terminal (NT) and C-terminal (CT)-cap domains. The LRR cluster is connected through a stalk region to a membrane lipid anchor. The CT-cap domain and stalk region of NgR2, but not NgR1, are sufficient for MAG binding, and when expressed in neurons, exhibit constitutive growth inhibitory activity. The LRR cluster of NgR1 supports binding of Nogo-66, OMgp, and MAG. Deletion of disulfide loop Cys(309)-Cys(336) of NgR1 selectively increases its affinity for Nogo-66 and OMgp. A chimeric Nogo receptor variant (NgR(OMNI)) in which Cys(309)-Cys(336) is deleted and followed by a 13 aa MAG-binding motif of the NgR2 stalk, shows superior binding of OMgp, Nogo-66, and MAG compared with wild-type NgR1 or NgR2. Soluble NgR(OMNI) (NgR(OMNI)-Fc) binds strongly to membrane-bound inhibitors and promotes neurite outgrowth on both MAG and CNS myelin substrates. Thus, NgR(OMNI)-Fc may offer therapeutic opportunities following nervous system injury or disease where myelin inhibits neuronal regeneration.
Collapse
Affiliation(s)
- Laurie A. Robak
- Department of Biomedical Genetics, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14642
| | - Karthik Venkatesh
- Neurology, University of Michigan School of Medicine, The University of Michigan, Ann Arbor, Michigan 48109-2200
| | - Hakjoo Lee
- Department of Biomedical Genetics, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14642
| | - Stephen J. Raiker
- Departments of Cell and Developmental Biology and
- Neurology, University of Michigan School of Medicine, The University of Michigan, Ann Arbor, Michigan 48109-2200
- Department of Biomedical Genetics, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14642
| | - Yuntao Duan
- Departments of Cell and Developmental Biology and
- Neurology, University of Michigan School of Medicine, The University of Michigan, Ann Arbor, Michigan 48109-2200
| | - Jane Lee-Osbourne
- Department of Biomedical Genetics, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14642
| | - Thomas Hofer
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-1203
| | - Rose G. Mage
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892-1892, and
| | - Christoph Rader
- Experimental Transplantation and Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-1203
| | - Roman J. Giger
- Departments of Cell and Developmental Biology and
- Neurology, University of Michigan School of Medicine, The University of Michigan, Ann Arbor, Michigan 48109-2200
- Department of Biomedical Genetics, School of Medicine and Dentistry, University of Rochester, Rochester, New York 14642
| |
Collapse
|
48
|
Pool M, Niino M, Rambaldi I, Robson K, Bar-Or A, Fournier AE. Myelin regulates immune cell adhesion and motility. Exp Neurol 2009; 217:371-7. [PMID: 19328785 DOI: 10.1016/j.expneurol.2009.03.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2008] [Revised: 03/09/2009] [Accepted: 03/14/2009] [Indexed: 01/04/2023]
Abstract
The etiology of multiple sclerosis (MS) has not been fully elucidated, however evidence supports an autoimmune disease model notable for the infiltration of pro-inflammatory immune cells into sites of active demyelination and axonal injury. Previous findings demonstrate that neutralization of Nogo, a protein originally identified as a myelin-associated inhibitor (MAI) of axon regeneration, ameliorates experimental autoimmune encephalomyelitis (EAE), a commonly used animal model of MS. More efficient axonal regeneration was suggested as a mechanism underlying the improved EAE outcome. However, neutralization of Nogo also led to an anti-inflammatory shift of T cell cytokines during EAE suggesting that another therapeutic mechanism may involve regulation of immune cell responses. Here we report that human immune cells from healthy individuals and MS patients express Nogo receptor1 (NgR1) indicating that they may be subject to regulation by MAIs. B cells, T cells and monocytes express NgR1 in a regulated fashion upon activation. While direct stimulation of human immune cells with an inhibitory fragment of Nogo does not impact their in vitro proliferation or cytokine production, the immune cells display reduced adhesion and enhanced motility in response to myelin, effects that are in part attenuated by antagonizing NgR1 signaling. We conclude that NgR1 alters the motility of immune cells exposed to myelin and may thus impact their behaviour within the CNS, particularly under conditions when immune cell activation is heightened.
Collapse
Affiliation(s)
- Madeline Pool
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, 3801 Rue University, Montreal, Quebec, Canada H3A 2B4
| | | | | | | | | | | |
Collapse
|
49
|
Cui Q, Yin Y, Benowitz LI. The role of macrophages in optic nerve regeneration. Neuroscience 2009; 158:1039-48. [PMID: 18708126 PMCID: PMC2670061 DOI: 10.1016/j.neuroscience.2008.07.036] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2008] [Revised: 07/18/2008] [Accepted: 07/20/2008] [Indexed: 11/25/2022]
Abstract
Following injury to the nervous system, the activation of macrophages, microglia, and T-cells profoundly affects the ability of neurons to survive and to regenerate damaged axons. The primary visual pathway provides a well-defined model system for investigating the interactions between the immune system and the nervous system after neural injury. Following damage to the optic nerve in mice and rats, retinal ganglion cells, the projection neurons of the eye, normally fail to regenerate their axons and soon begin to die. Induction of an inflammatory response in the vitreous strongly enhances the survival of retinal ganglion cells and enables these cells to regenerate lengthy axons beyond the injury site. T cells modulate this response, whereas microglia are thought to contribute to the loss of retinal ganglion cells in this model and in certain ocular diseases. This review discusses the complex and sometimes paradoxical actions of blood-borne macrophages, resident microglia, and T-cells in determining the outcome of injury in the primary visual pathway.
Collapse
Affiliation(s)
- Q Cui
- Department of Ophthalmology and Visual Sciences, The Chinese University of Hong Kong, 147K Argyle Street, Kowloon, Hong Kong, PR China.
| | | | | |
Collapse
|
50
|
Richard M, Sacquet J, Jourdan F, Pellier-Monnin V. Spatio-temporal expression pattern of receptors for myelin-associated inhibitors in the developing rat olfactory system. Brain Res 2008; 1252:52-65. [PMID: 19063867 DOI: 10.1016/j.brainres.2008.11.049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2008] [Revised: 11/03/2008] [Accepted: 11/10/2008] [Indexed: 12/11/2022]
Abstract
The myelin-associated inhibitory proteins (Nogo-A, MAG and OMgp) that prevent axon regeneration in adult CNS, mediate their effects via a receptor referred as NgR1. Beside their inhibitory role in the adult CNS, Nogo-A and NgR1 might also be functionally involved in the developing nervous system. At the present time, no detailed study is available regarding either the onset of NgR1 expression during development or its spatio-temporal pattern of expression relative to the presence of Nogo-A. Two homologs of NgR1, NgR2 and NgR3, have been recently identified, but their function in the nervous system is still unknown in adult as well as during development. We have examined the spatio-temporal expression pattern of both NgR1, NgR2 and NgR3 mRNAs and corresponding proteins in the developing rat olfactory system using in situ hybridization and immunohistochemistry. From E15-E16 onwards, NgR1 mRNA was expressed by differentiating neurons in both the olfactory epithelium and the olfactory bulb. At all developmental stages, including adult animals, NgR1 protein was preferentially targeted to olfactory axons emerging from the olfactory epithelium. Using double-immunostainings in the postnatal olfactory mucosa, we confirm the neuronal localization of NgR1 and its preferential distribution along the olfactory axons. The NgR2 and NgR3 transcripts and their proteins display similar expression profiles in the olfactory system. Together, our data suggest that, in non-pathological conditions, NgR1 and its homologs may play a role in axon outgrowth in the rat olfactory system and may be relevant for the confinement of neural projections within the developing olfactory bulb.
Collapse
Affiliation(s)
- Marion Richard
- Laboratoire Neurosciences Sensorielles, Comportement, Cognition, CNRS-UMR 5020, Université de Lyon, Lyon 1, F-69366, France.
| | | | | | | |
Collapse
|