1
|
Rangel-Sosa MM, Mann F, Chauvet S. Pancreatic Schwann cell reprogramming supports cancer-associated neuronal remodeling. Glia 2024; 72:1840-1861. [PMID: 38961612 DOI: 10.1002/glia.24586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 07/05/2024]
Abstract
The peripheral nervous system is a key regulator of cancer progression. In pancreatic ductal adenocarcinoma (PDAC), the sympathetic branch of the autonomic nervous system inhibits cancer development. This inhibition is associated with extensive sympathetic nerve sprouting in early pancreatic cancer precursor lesions. However, the underlying mechanisms behind this process remain unclear. This study aimed to investigate the roles of pancreatic Schwann cells in the structural plasticity of sympathetic neurons. We examined the changes in the number and distribution of Schwann cells in a transgenic mouse model of PDAC and in a model of metaplastic pancreatic lesions induced by chronic inflammation. Schwann cells proliferated and expanded simultaneously with new sympathetic nerve sprouts in metaplastic/neoplastic pancreatic lesions. Sparse genetic labeling showed that individual Schwann cells in these lesions had a more elongated and branched structure than those under physiological conditions. Schwann cells overexpressed neurotrophic factors, including glial cell-derived neurotrophic factor (GDNF). Sympathetic neurons upregulated the GDNF receptors and exhibited enhanced neurite growth in response to GDNF in vitro. Selective genetic deletion of Gdnf in Schwann cells completely blocked sympathetic nerve sprouting in metaplastic pancreatic lesions in vivo. This study demonstrated that pancreatic Schwann cells underwent adaptive reprogramming during early cancer development, supporting a protective antitumor neuronal response. These finding could help to develop new strategies to modulate cancer associated neural plasticity.
Collapse
Affiliation(s)
| | - Fanny Mann
- Aix Marseille Univ, CNRS, IBDM, Marseille, France
| | | |
Collapse
|
2
|
Yoo K, Jo YW, Yoo T, Hann SH, Park I, Kim YE, Kim YL, Rhee J, Song IW, Kim JH, Baek D, Kong YY. Muscle-resident mesenchymal progenitors sense and repair peripheral nerve injury via the GDNF-BDNF axis. eLife 2024; 13:RP97662. [PMID: 39324575 PMCID: PMC11426970 DOI: 10.7554/elife.97662] [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] [Indexed: 09/27/2024] Open
Abstract
Fibro-adipogenic progenitors (FAPs) are muscle-resident mesenchymal progenitors that can contribute to muscle tissue homeostasis and regeneration, as well as postnatal maturation and lifelong maintenance of the neuromuscular system. Recently, traumatic injury to the peripheral nerve was shown to activate FAPs, suggesting that FAPs can respond to nerve injury. However, questions of how FAPs can sense the anatomically distant peripheral nerve injury and whether FAPs can directly contribute to nerve regeneration remained unanswered. Here, utilizing single-cell transcriptomics and mouse models, we discovered that a subset of FAPs expressing GDNF receptors Ret and Gfra1 can respond to peripheral nerve injury by sensing GDNF secreted by Schwann cells. Upon GDNF sensing, this subset becomes activated and expresses Bdnf. FAP-specific inactivation of Bdnf (Prrx1Cre; Bdnffl/fl) resulted in delayed nerve regeneration owing to defective remyelination, indicating that GDNF-sensing FAPs play an important role in the remyelination process during peripheral nerve regeneration. In aged mice, significantly reduced Bdnf expression in FAPs was observed upon nerve injury, suggesting the clinical relevance of FAP-derived BDNF in the age-related delays in nerve regeneration. Collectively, our study revealed the previously unidentified role of FAPs in peripheral nerve regeneration, and the molecular mechanism behind FAPs' response to peripheral nerve injury.
Collapse
Affiliation(s)
- Kyusang Yoo
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Young-Woo Jo
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Takwon Yoo
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Sang-Hyeon Hann
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Inkuk Park
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yea-Eun Kim
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Ye Lynne Kim
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Joonwoo Rhee
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - In-Wook Song
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Ji-Hoon Kim
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
- Molecular Recognition Research Center, Korea Institute of Science and Technology, Seoul, Republic of Korea
| | - Daehyun Baek
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| | - Young-Yun Kong
- School of Biological Sciences, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
3
|
Pinzon-Herrera L, Magness J, Apodaca-Reyes H, Sanchez J, Almodovar J. Surface Modification of Nerve Guide Conduits with ECM Coatings and Investigating Their Impact on Schwann Cell Response. Adv Healthc Mater 2024; 13:e2304103. [PMID: 38400540 DOI: 10.1002/adhm.202304103] [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: 11/21/2023] [Revised: 02/06/2024] [Indexed: 02/25/2024]
Abstract
In this study, layer-by-layer coatings composed of heparin and collagen are proposed as an extracellular mimetic environment on nerve guide conduits (NGC) to modulate the behavior of Schwann cells (hSCs). The authors evaluated the stability, degradation over time, and bioactivity of six bilayers of heparin/collagen layer-by-layer coatings, denoted as (HEP/COL)6. The stability study reveals that (HEP/COL)6 is stable after incubating the coatings in cell media for up to 21 days. The impact of (HEP/COL)6 on hSCs viability, protein expression, and migration is evaluated. These assays show that hSCs cultured in (HEP/COL)6 have enhanced protein expression and migration. This condition increases the expression of neurotrophic and immunomodulatory factors up to 1.5-fold compared to controls, and hSCs migrated 1.34 times faster than in the uncoated surfaces. Finally, (HEP/COL)6 is also applied to a commercial collagen-based NGC, NeuraGen, and hSC viability and adhesion are studied after 6 days of culture. The morphology of NeuraGen is not altered by the presence of (HEP/COL)6 and a nearly 170% increase of the cell viability is observed in the condition where NeuraGen is used with (HEP/COL)6. Additionally, cell adhesion on the coated samples is successfully demonstrated. This work demonstrates the reparative enhancing potential of extracellular mimetic coatings.
Collapse
Affiliation(s)
- Luis Pinzon-Herrera
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, 3202 Bell Engineering Center, Fayetteville, AR, 72701, USA
- Department of Chemical, Biochemical, and Environmental Engineering, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD, 21250, USA
| | - John Magness
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, 3202 Bell Engineering Center, Fayetteville, AR, 72701, USA
| | - Hector Apodaca-Reyes
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, 3202 Bell Engineering Center, Fayetteville, AR, 72701, USA
| | - Jesus Sanchez
- Science & Mathematics Division, Northwest Arkansas Community College, 1418 Burns Hall, Bentonville, AR, 72712, USA
| | - Jorge Almodovar
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, 3202 Bell Engineering Center, Fayetteville, AR, 72701, USA
- Department of Chemical, Biochemical, and Environmental Engineering, University of Maryland Baltimore County, 1000 Hilltop Circle, Baltimore, MD, 21250, USA
| |
Collapse
|
4
|
Bortolozzi A, Fico G, Berk M, Solmi M, Fornaro M, Quevedo J, Zarate CA, Kessing LV, Vieta E, Carvalho AF. New Advances in the Pharmacology and Toxicology of Lithium: A Neurobiologically Oriented Overview. Pharmacol Rev 2024; 76:323-357. [PMID: 38697859 PMCID: PMC11068842 DOI: 10.1124/pharmrev.120.000007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 05/05/2024] Open
Abstract
Over the last six decades, lithium has been considered the gold standard treatment for the long-term management of bipolar disorder due to its efficacy in preventing both manic and depressive episodes as well as suicidal behaviors. Nevertheless, despite numerous observed effects on various cellular pathways and biologic systems, the precise mechanism through which lithium stabilizes mood remains elusive. Furthermore, there is recent support for the therapeutic potential of lithium in other brain diseases. This review offers a comprehensive examination of contemporary understanding and predominant theories concerning the diverse mechanisms underlying lithium's effects. These findings are based on investigations utilizing cellular and animal models of neurodegenerative and psychiatric disorders. Recent studies have provided additional support for the significance of glycogen synthase kinase-3 (GSK3) inhibition as a crucial mechanism. Furthermore, research has shed more light on the interconnections between GSK3-mediated neuroprotective, antioxidant, and neuroplasticity processes. Moreover, recent advancements in animal and human models have provided valuable insights into how lithium-induced modifications at the homeostatic synaptic plasticity level may play a pivotal role in its clinical effectiveness. We focused on findings from translational studies suggesting that lithium may interface with microRNA expression. Finally, we are exploring the repurposing potential of lithium beyond bipolar disorder. These recent findings on the therapeutic mechanisms of lithium have provided important clues toward developing predictive models of response to lithium treatment and identifying new biologic targets. SIGNIFICANCE STATEMENT: Lithium is the drug of choice for the treatment of bipolar disorder, but its mechanism of action in stabilizing mood remains elusive. This review presents the latest evidence on lithium's various mechanisms of action. Recent evidence has strengthened glycogen synthase kinase-3 (GSK3) inhibition, changes at the level of homeostatic synaptic plasticity, and regulation of microRNA expression as key mechanisms, providing an intriguing perspective that may help bridge the mechanistic gap between molecular functions and its clinical efficacy as a mood stabilizer.
Collapse
Affiliation(s)
- Analia Bortolozzi
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Giovanna Fico
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Michael Berk
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Marco Solmi
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Michele Fornaro
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Joao Quevedo
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Carlos A Zarate
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Lars V Kessing
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Eduard Vieta
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| | - Andre F Carvalho
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Spanish National Research Council (CSIC), Barcelona, Spain (A.B.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.B., G.F., E.V.); Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), ISCIII, Madrid, Spain (A.B., G.F., E.V.); Hospital Clinic, Institute of Neuroscience, University of Barcelona, Barcelona, Spain (G.F., E.V.); IMPACT - The Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Geelong, Victoria, Australia (M.B., A.F.C.); Department of Psychiatry, University of Ottawa, Ontario, Canada (M.S.); The Champlain First Episode Psychosis Program, Department of Mental Health, The Ottawa Hospital, Ontario, Canada (M.S.); Department of Child and Adolescent Psychiatry, Charité Universitätsmedizin, Berlin, Germany (M.S.); Section of Psychiatry, Department of Neuroscience, Reproductive Science and Odontostomatology, Federico II University of Naples, Naples, Italy (M.F.); Center of Excellence on Mood Disorders, Faillace Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UT Health), Houston, Texas (J.Q.); Experimental Therapeutics and Pathophysiology Branch, National Institute of Mental Health, National Institutes of Health, Bethesda, Maryland (C.A.Z.); Copenhagen Affective Disorders Research Centre (CADIC), Psychiatric Center Copenhagen, Rigshospitalet, Denmark (L.V.K.); and Department of Clinical Medicine, University of Copenhagen, Denmark (L.V.K.)
| |
Collapse
|
5
|
Gregory HN, Guillemot-Legris O, Crouch D, Williams G, Phillips JB. Electrospun aligned tacrolimus-loaded polycaprolactone biomaterials for peripheral nerve repair. Regen Med 2024; 19:171-187. [PMID: 37818696 DOI: 10.2217/rme-2023-0151] [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] [Indexed: 10/12/2023] Open
Abstract
Background: Efficacious repair of peripheral nerve injury is an unmet clinical need. The implantation of biomaterials containing neurotrophic drugs at the injury site could promote nerve regeneration and improve outcomes for patients. Materials & methods: Random and aligned electrospun poly-ε-caprolactone scaffolds containing encapsulated tacrolimus were fabricated, and the gene expression profile of Schwann cells (SCs) cultured on the surface was elucidated. On aligned fibers, the morphology of SCs and primary rat neurons was investigated. Results: Both scaffold types exhibited sustained release of drug, and the gene expression of SCs was modulated by both nanofibrous topography and the presence of tacrolimus. Aligned fibers promoted the alignment of SCs and orientated outgrowth from neurons. Conclusion: Electrospun PCL scaffolds with tacrolimus hold promise for the repair of peripheral nerve injury.
Collapse
Affiliation(s)
- Holly N Gregory
- UCL School of Pharmacy, University College London, London, WC1N 1AX, UK
- UCL Centre for Nerve Engineering, London, WC1N 1AX, UK
| | - Owein Guillemot-Legris
- UCL School of Pharmacy, University College London, London, WC1N 1AX, UK
- UCL Centre for Nerve Engineering, London, WC1N 1AX, UK
| | - Daisy Crouch
- UCL School of Pharmacy, University College London, London, WC1N 1AX, UK
- UCL Centre for Nerve Engineering, London, WC1N 1AX, UK
| | - Gareth Williams
- UCL School of Pharmacy, University College London, London, WC1N 1AX, UK
- UCL Centre for Nerve Engineering, London, WC1N 1AX, UK
| | - James B Phillips
- UCL School of Pharmacy, University College London, London, WC1N 1AX, UK
- UCL Centre for Nerve Engineering, London, WC1N 1AX, UK
| |
Collapse
|
6
|
Reshamwala R, Shah M. Regenerative Approaches in the Nervous System. Regen Med 2023. [DOI: 10.1007/978-981-19-6008-6_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
7
|
Klymenko A, Lutz D. Melatonin signalling in Schwann cells during neuroregeneration. Front Cell Dev Biol 2022; 10:999322. [PMID: 36299487 PMCID: PMC9589221 DOI: 10.3389/fcell.2022.999322] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
It has widely been thought that in the process of nerve regeneration Schwann cells populate the injury site with myelinating, non–myelinating, phagocytic, repair, and mesenchyme–like phenotypes. It is now clear that the Schwann cells modify their shape and basal lamina as to accommodate re–growing axons, at the same time clear myelin debris generated upon injury, and regulate expression of extracellular matrix proteins at and around the lesion site. Such a remarkable plasticity may follow an intrinsic functional rhythm or a systemic circadian clock matching the demands of accurate timing and precision of signalling cascades in the regenerating nervous system. Schwann cells react to changes in the external circadian clock clues and to the Zeitgeber hormone melatonin by altering their plasticity. This raises the question of whether melatonin regulates Schwann cell activity during neurorepair and if circadian control and rhythmicity of Schwann cell functions are vital aspects of neuroregeneration. Here, we have focused on different schools of thought and emerging concepts of melatonin–mediated signalling in Schwann cells underlying peripheral nerve regeneration and discuss circadian rhythmicity as a possible component of neurorepair.
Collapse
|
8
|
Transplantation of human neural progenitor cells secreting GDNF into the spinal cord of patients with ALS: a phase 1/2a trial. Nat Med 2022; 28:1813-1822. [PMID: 36064599 PMCID: PMC9499868 DOI: 10.1038/s41591-022-01956-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 07/18/2022] [Indexed: 11/08/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) involves progressive motor neuron loss, leading to paralysis and death typically within 3–5 years of diagnosis. Dysfunctional astrocytes may contribute to disease and glial cell line-derived neurotrophic factor (GDNF) can be protective. Here we show that human neural progenitor cells transduced with GDNF (CNS10-NPC-GDNF) differentiated to astrocytes protected spinal motor neurons and were safe in animal models. CNS10-NPC-GDNF were transplanted unilaterally into the lumbar spinal cord of 18 ALS participants in a phase 1/2a study (NCT02943850). The primary endpoint of safety at 1 year was met, with no negative effect of the transplant on motor function in the treated leg compared with the untreated leg. Tissue analysis of 13 participants who died of disease progression showed graft survival and GDNF production. Benign neuromas near delivery sites were common incidental findings at post-mortem. This study shows that one administration of engineered neural progenitors can provide new support cells and GDNF delivery to the ALS patient spinal cord for up to 42 months post-transplantation. A phase 1/2a study shows that human neural progenitor cells modified to release the growth factor GDNF are safely transplanted into the spinal cord of patients with ALS, with cell survival and GDNF production for over 3 years.
Collapse
|
9
|
Dong C, Ubogu EE. Pro-inflammatory cytokines and leukocyte integrins associated with chronic neuropathic pain in traumatic and inflammatory neuropathies: Initial observations and hypotheses. Front Immunol 2022; 13:935306. [PMID: 35983047 PMCID: PMC9378781 DOI: 10.3389/fimmu.2022.935306] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 07/12/2022] [Indexed: 11/13/2022] Open
Abstract
Leukocyte infiltration and persistence within peripheral nerves have been implicated in chronic nociception pathogenesis in murine peripheral neuropathy models. Endoneurial cytokine and chemokine expression contribute to leukocyte infiltration and maintenance of a pro-inflammatory state that delays peripheral nerve recovery and promotes chronic pain behaviors in these mice. However, there has been a failure to translate murine model data into safe and effective treatments for chronic neuropathic pain in peripheral neuropathy patients, or develop reliable biomarkers that may help diagnose or determine treatment responses in affected patients. Initial work showed that persistent sciatic nerve CD11b+ CD45+ leukocyte infiltration was associated with disease severity in three mouse models of inflammatory and traumatic peripheral neuropathies, implying a direct contributing role in disease pathogenesis. In support of this, CD11b+ leukocytes were also seen in the sural nerve biopsies of chronic neuropathic pain patients with three different peripheral neuropathies. Systemic CD11b antagonism using a validated function-neutralizing monoclonal antibody effectively treated chronic nociception following unilateral sciatic nerve crush injury (a representative traumatic neuropathy model associated with axonal degeneration and increased blood-nerve barrier permeability) and does not cause drug addiction behaviors in adult mice. These data suggest that CD11b could be an effective molecular target for chronic neuropathic pain treatment in inflammatory and traumatic peripheral neuropathies. Despite known murine peripheral neuropathy model limitations, our initial work suggests that early expression of pro-inflammatory cytokines, such as tissue inhibitor of metalloproteinases-1 may predict subsequent chronic nociception development following unilateral sciatic nerve crush injury. Studies aligning animal model investigation with observational data from well-characterized human peripheral neuropathies, including transcriptomics and proteomics, as well as animal model studies using a human clinical trial design should foster the identification of clinically relevant biomarkers and effective targeted treatments with limited addiction potential for chronic neuropathic pain in peripheral neuropathy patients.
Collapse
|
10
|
Wang Y, Liang Y, Huang J, Gao Y, Xu Z, Ni X, Yang Y, Yang X, Zhao Y. Proteomic Analysis of Silk Fibroin Reveals Diverse Biological Function of Different Degumming Processing From Different Origin. Front Bioeng Biotechnol 2022; 9:777320. [PMID: 35198548 PMCID: PMC8859422 DOI: 10.3389/fbioe.2021.777320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/29/2021] [Indexed: 11/13/2022] Open
Abstract
Silk, as a kind of natural fibrin, has been prepared into various biomaterials due to its excellent biocompatibility and mechanicalness. However, there are some controversies on the biocompatibility of silk fibroin (SF), especially when it coexists with sericin. In this study, two kinds of silk from Jiangsu and Zhejiang were degummed with two concentrations of Na2CO3 solution, respectively, to obtain four kinds of silk fibroin. The effects of different degumming treatments on silk fibroin properties were analyzed by means of color reaction, apparent viscosity measurement, and transmission electron microscope and isobaric tags for relative and absolute quantification analyses, and the effects of different silk fibroin membranes on the growth of Schwann cells were evaluated. The results showed that the natural silk from Zhejiang treated with 0.05% Na2CO3 solution had a fuller structure, higher apparent viscosity, and better protein composition. While SF obtained by degumming with 0.5% Na2CO3 solution was more beneficial to cell adhesion and proliferation due to the thorough removal of sericin. This study may provide important theoretical and experimental bases for the selection of biomaterials for fabricating artificial nerve grafts.
Collapse
Affiliation(s)
- Yaling Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China.,School of Pharmacy, Nantong University, Nantong, China
| | - Yunyun Liang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Jiacen Huang
- School of Public Health, Nantong University, Nantong, China
| | - Yisheng Gao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Zhixin Xu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Xuejun Ni
- Affiliated Hospital of Nantong University, Nantong University, Nantong, China
| | - Yumin Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Xiaoming Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China.,School of Public Health, Nantong University, Nantong, China
| | - Yahong Zhao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China.,School of Public Health, Nantong University, Nantong, China
| |
Collapse
|
11
|
Jessen KR, Mirsky R. The Role of c-Jun and Autocrine Signaling Loops in the Control of Repair Schwann Cells and Regeneration. Front Cell Neurosci 2022; 15:820216. [PMID: 35221918 PMCID: PMC8863656 DOI: 10.3389/fncel.2021.820216] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 12/30/2021] [Indexed: 12/12/2022] Open
Abstract
After nerve injury, both Schwann cells and neurons switch to pro-regenerative states. For Schwann cells, this involves reprogramming of myelin and Remak cells to repair Schwann cells that provide the signals and mechanisms needed for the survival of injured neurons, myelin clearance, axonal regeneration and target reinnervation. Because functional repair cells are essential for regeneration, it is unfortunate that their phenotype is not robust. Repair cell activation falters as animals get older and the repair phenotype fades during chronic denervation. These malfunctions are important reasons for the poor outcomes after nerve damage in humans. This review will discuss injury-induced Schwann cell reprogramming and the concept of the repair Schwann cell, and consider the molecular control of these cells with emphasis on c-Jun. This transcription factor is required for the generation of functional repair cells, and failure of c-Jun expression is implicated in repair cell failures in older animals and during chronic denervation. Elevating c-Jun expression in repair cells promotes regeneration, showing in principle that targeting repair cells is an effective way of improving nerve repair. In this context, we will outline the emerging evidence that repair cells are sustained by autocrine signaling loops, attractive targets for interventions aimed at promoting regeneration.
Collapse
Affiliation(s)
- Kristjan R. Jessen
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | | |
Collapse
|
12
|
A ‘Relay’-Type Drug-Eluting Nerve Guide Conduit: Computational Fluid Dynamics Modeling of the Drug Eluting Efficiency of Various Drug Release Systems. Pharmaceutics 2022; 14:pharmaceutics14020230. [PMID: 35213963 PMCID: PMC8874367 DOI: 10.3390/pharmaceutics14020230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/16/2022] [Accepted: 01/17/2022] [Indexed: 01/27/2023] Open
Abstract
Nerve guidance conduits (NGCs) are tubular scaffolds that act as a bridge between the proximal and distal ends of the native nerve to facilitate the nerve regeneration. The application of NGCs is mostly limited to nerve defects less than 3 mm due to the lack of sufficient cells in the lumen. The development of drug-release-system-embedded NGCs has the potential to improve the nerve regeneration performance by providing long-term release of growth factors. However, most of the past works only focused on one type of drug release system, limiting the variation in drug release system types and features. Therefore, in this study, computer-aided design (CAD) models were constructed and Computational Fluid Dynamics (CFD) simulations were carried out to investigate the effect of growth factor transporting efficiency on different drug release systems. To overcome the challenges posed by the current NGCs in treating long nerve gap injuries (>4 cm), a novel ‘relay’ NGC design is first proposed in this paper and has the potential to improve the nerve regeneration performance to next level. The intermediate cavities introduced along the length of the multi-channel NGCs act as a relay to further enhance the cell concentrations or growth factor delivery as well as the regeneration performance. Four different drug release systems, namely, a single-layer microsphere system, a double-layer microsphere system, bulk hydrogel, and hydrogel film, were chosen for the simulation. The results show that the double-layer microsphere system achieves the highest growth factor volume fraction among all the drug release systems. For the single-layer microsphere system, growth factor concentration can be significantly improved by increasing the microsphere quantities and decreasing the diameter and adjacent distance of microspheres. Bulk hydrogel systems hold the lowest growth factor release performance, and the growth factor concentration monotonically increased with the increase of film thickness in the hydrogel film system. Owing to the easy fabrication of hydrogel film and the even distribution of growth factors, the hydrogel film system can be regarded as a strong candidate in drug-eluting NGCs. The use of computational simulations can be regarded as a guideline for the design and application of drug release systems, as well as a promising tool for further nerve tissue engineering study.
Collapse
|
13
|
Khaitin A. Calcium in Neuronal and Glial Response to Axotomy. Int J Mol Sci 2021; 22:ijms222413344. [PMID: 34948141 PMCID: PMC8706492 DOI: 10.3390/ijms222413344] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 12/09/2021] [Accepted: 12/10/2021] [Indexed: 11/16/2022] Open
Abstract
Neurotrauma assumes an instant or delayed disconnection of axons (axotomy), which affects not only neurons, but surrounding glia as well. Not only mechanically injured glia near the site of disconnection, especially transection, is subjected to the damage, but also glia that is remote from the lesion site. Glial cells, which surround the neuronal body, in turn, support neuron survival, so there is a mutual protection between neuron and glia. Calcium signaling is a central mediator of all post-axotomy events, both in neuron and glia, playing a critical role in their survival/regeneration or death/degeneration. The involvement of calcium in post-axotomy survival of the remote, mechanically intact glia is poorly studied. The purpose of this review is to sum up the calcium-involving mechanisms in responses of neurons and glial cells to axotomy to show their importance and to give some suggestions for future research of remote glia in this context.
Collapse
Affiliation(s)
- Andrey Khaitin
- Academy of Biology and Biotechnology, Southern Federal University, 344090 Rostov-on-Don, Russia
| |
Collapse
|
14
|
Cintron-Colon AF, Almeida-Alves G, VanGyseghem JM, Spitsbergen JM. GDNF to the rescue: GDNF delivery effects on motor neurons and nerves, and muscle re-innervation after peripheral nerve injuries. Neural Regen Res 2021; 17:748-753. [PMID: 34472460 PMCID: PMC8530131 DOI: 10.4103/1673-5374.322446] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Peripheral nerve injuries commonly occur due to trauma, like a traffic accident. Peripheral nerves get severed, causing motor neuron death and potential muscle atrophy. The current golden standard to treat peripheral nerve lesions, especially lesions with large (≥ 3 cm) nerve gaps, is the use of a nerve autograft or reimplantation in cases where nerve root avulsions occur. If not tended early, degeneration of motor neurons and loss of axon regeneration can occur, leading to loss of function. Although surgical procedures exist, patients often do not fully recover, and quality of life deteriorates. Peripheral nerves have limited regeneration, and it is usually mediated by Schwann cells and neurotrophic factors, like glial cell line-derived neurotrophic factor, as seen in Wallerian degeneration. Glial cell line-derived neurotrophic factor is a neurotrophic factor known to promote motor neuron survival and neurite outgrowth. Glial cell line-derived neurotrophic factor is upregulated in different forms of nerve injuries like axotomy, sciatic nerve crush, and compression, thus creating great interest to explore this protein as a potential treatment for peripheral nerve injuries. Exogenous glial cell line-derived neurotrophic factor has shown positive effects in regeneration and functional recovery when applied in experimental models of peripheral nerve injuries. In this review, we discuss the mechanism of repair provided by Schwann cells and upregulation of glial cell line-derived neurotrophic factor, the latest findings on the effects of glial cell line-derived neurotrophic factor in different types of peripheral nerve injuries, delivery systems, and complementary treatments (electrical muscle stimulation and exercise). Understanding and overcoming the challenges of proper timing and glial cell line-derived neurotrophic factor delivery is paramount to creating novel treatments to tend to peripheral nerve injuries to improve patients’ quality of life.
Collapse
Affiliation(s)
| | | | | | - John M Spitsbergen
- Biological Sciences Department, Western Michigan University, Kalamazoo, MI, USA
| |
Collapse
|
15
|
Yokoi T, Uemura T, Takamatsu K, Shintani K, Onode E, Hama S, Miyashima Y, Okada M, Nakamura H. Fate and contribution of induced pluripotent stem cell-derived neurospheres transplanted with nerve conduits to promote peripheral nerve regeneration in mice. Biomed Mater Eng 2021; 32:171-181. [PMID: 33780359 DOI: 10.3233/bme-201182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND We previously demonstrated that a bioabsorbable nerve conduit coated with mouse induced pluripotent stem cell (iPSC)-derived neurospheres accelerated peripheral nerve regeneration in mice. OBJECTIVE We examined the fate and utility of iPSC-derived neurospheres transplanted with nerve conduits for the treatment of sciatic nerve gaps in mice. METHODS Complete 5-mm defects were created in sciatic nerves and reconstructed using nerve conduits that were either uncoated or coated with mouse iPSC-derived neurospheres. The survival of the neurospheres on the nerve conduits was tracked using an in vivo imaging. The localization of the transplanted cells and regenerating axons was examined histologically. The gene expression levels in the nerve conduits were evaluated. RESULTS The neurospheres survived for at least 14 days, peaking at 4--7 days after implantation. The grafted neurospheres remained as Schwann-like cells within the nerve conduits and migrated into the regenerated axons. The expression levels of ATF3, BDNF, and GDNF in the nerve conduit coated with neurospheres were upregulated. CONCLUSIONS Mouse iPSC-derived neurospheres transplanted with nerve conduits for the treatment of sciatic nerve defects in mice migrated into regenerating axons, survived as Schwann-like cells, and promoted axonal growth with an elevation in the expression of nerve regeneration-associated trophic factors.
Collapse
Affiliation(s)
- Takuya Yokoi
- Department of Orthopaedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Takuya Uemura
- Department of Orthopaedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan.,Department of Orthopaedic Surgery, Osaka General Hospital of West Japan Railway Company, Osaka, Japan
| | - Kiyohito Takamatsu
- Department of Orthopaedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan.,Department of Orthopaedic Surgery, Yodogawa Christian Hospital, Osaka, Japan
| | - Kosuke Shintani
- Department of Pediatric Orthopaedic Surgery, Osaka City General Hospital, Osaka, Japan
| | - Ema Onode
- Department of Orthopaedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Shunpei Hama
- Department of Orthopaedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yusuke Miyashima
- Department of Orthopaedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Mitsuhiro Okada
- Department of Orthopaedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Hiroaki Nakamura
- Department of Orthopaedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
16
|
Jones GH, Rong C, Shariq AS, Mishra A, Machado-Vieira R. Intracellular Signaling Cascades in Bipolar Disorder. Curr Top Behav Neurosci 2021; 48:101-132. [PMID: 32860212 DOI: 10.1007/7854_2020_157] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Bipolar spectrum disorders carry a significant public health burden. Disproportionately high rates of suicide, incarceration, and comorbid medical conditions necessitate an extraordinary focus on understanding the intricacies of this disease. Elucidating granular, intracellular details seems to be a necessary preamble to advancing promising therapeutic opportunities. In this chapter, we review a wide range of intracellular mechanisms including mitochondrial energetics, calcium signaling, neuroinflammation, the microbiome, neurotransmitter metabolism, glycogen synthase kinase 3-beta (GSK3β), protein kinase C (PKC) and diacylglycerol (DAG), and neurotrophins (especially BDNF), as well as the glutamatergic, dopaminergic, purinergic, and neurohormonal systems. Owing to the relative lack of understanding and effective therapeutic options compared to the rest of the spectrum, special attention is paid in the chapter to the latest developments in bipolar depression. Likewise, from a therapeutic standpoint, special attention should be paid to the pervasive mechanistic actions of lithium as a means of amalgamating numerous, disparate cascades into a digestible cognitive topology.
Collapse
Affiliation(s)
- Gregory H Jones
- Department of Psychiatry, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Carola Rong
- Department of Psychiatry, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Aisha S Shariq
- Department of Psychiatry, Texas Tech University Health Science Center, El Paso, TX, USA
- Texas Tech University Health Science Center, Paul L. Foster School of Medicine, El Paso, TX, USA
| | - Abhinav Mishra
- Texas Tech University Health Science Center, Paul L. Foster School of Medicine, El Paso, TX, USA
| | - Rodrigo Machado-Vieira
- Department of Psychiatry, University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
17
|
Patritti-Cram J, Coover RA, Jankowski MP, Ratner N. Purinergic signaling in peripheral nervous system glial cells. Glia 2021; 69:1837-1851. [PMID: 33507559 PMCID: PMC8192487 DOI: 10.1002/glia.23969] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 01/02/2023]
Abstract
To facilitate analyses of purinergic signaling in peripheral nerve glia, we review recent literature and catalog purinergic receptor mRNA expression in cultured mouse Schwann cells (SCs). Purinergic signaling can decrease developmental SC proliferation, and promote SC differentiation. The purinergic receptors P2RY2 and P2RX7 are implicated in nerve development and in the ratio of Remak SCs to myelinating SCs in differentiated peripheral nerve. P2RY2, P2RX7, and other receptors are also implicated in peripheral neuropathies and SC tumors. In SC tumors lacking the tumor suppressor NF1, the SC pathway that suppresses SC growth through P2RY2‐driven β‐arrestin‐mediated AKT signaling is aberrant. SC‐released purinergic agonists acting through SC and/or neuronal purinergic receptors activate pain responses. In all these settings, purinergic receptor activation can result in calcium‐independent and calcium‐dependent release of SC ATP and UDP, growth factors, and cytokines that may contribute to disease and nerve repair. Thus, current research suggests that purinergic agonists and/or antagonists might have the potential to modulate peripheral glia function in development and in disease.
Collapse
Affiliation(s)
- Jennifer Patritti-Cram
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Robert A Coover
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Department of Basic Pharmaceutical Sciences, High Point University, High Point, North Carolina, USA
| | - Michael P Jankowski
- Department of Anesthesia, Division of Pain Management, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA.,Center for Understanding Pediatric Pain, Cincinnati Children's Hospital Medical Center and University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| | - Nancy Ratner
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
18
|
Tiong YL, Ng KY, Koh RY, Ponnudurai G, Chye SM. Melatonin promotes Schwann cell dedifferentiation and proliferation through the Ras/Raf/ERK and MAPK pathways, and glial cell-derived neurotrophic factor expression. Exp Ther Med 2020; 20:16. [PMID: 32934681 PMCID: PMC7471953 DOI: 10.3892/etm.2020.9143] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 11/22/2019] [Indexed: 12/24/2022] Open
Abstract
Upon peripheral nerve injury (PNI), continuous proliferation of Schwann cells is critical for axon regeneration and tubular reconstruction for nerve regeneration. Melatonin is a hormone that is able to induce proliferation in various cell types. In the present study, the effects of melatonin on promoting Schwann cell proliferation and the molecular mechanism involved were investigated. The present results showed that melatonin enhanced the melatonin receptors (MT1 and MT2) expression in Schwann cells. Melatonin induced Schwann cell dedifferentiation into progenitor-like Schwann cells, as observed by immunofluorescence staining, which showed Sox2 marker expression. In addition, melatonin enhanced Schwann cell proliferation, mediated by the upregulation of glial cell-derived neurotropic factor (GNDF) and protein kinase C (PKC). Furthermore, the Ras/Raf/ERK and MAPK signaling pathways were also involved in Schwann cell dedifferentiation and proliferation. In conclusion, melatonin induced Schwann cell dedifferentiation and proliferation via the Ras/Raf/ERK, MAPK and GDNF/PKC pathways. The present results suggested that melatonin could be used to enhance the recovery of PNI.
Collapse
Affiliation(s)
- Yee Lian Tiong
- School of Postgraduate, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Khuen Yen Ng
- School of Pharmacy, Monash University Malaysia, Subang Jaya, Selangor 47500, Malaysia
| | - Rhun Yian Koh
- School of Health Science, International Medical University, Kuala Lumpur 57000, Malaysia
| | | | - Soi Moi Chye
- School of Health Science, International Medical University, Kuala Lumpur 57000, Malaysia
| |
Collapse
|
19
|
Min Q, Parkinson DB, Dun XP. Migrating Schwann cells direct axon regeneration within the peripheral nerve bridge. Glia 2020; 69:235-254. [PMID: 32697392 DOI: 10.1002/glia.23892] [Citation(s) in RCA: 142] [Impact Index Per Article: 28.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/03/2020] [Accepted: 07/08/2020] [Indexed: 12/12/2022]
Abstract
Schwann cells within the peripheral nervous system possess a remarkable regenerative potential. Current research shows that peripheral nerve-associated Schwann cells possess the capacity to promote repair of multiple tissues including peripheral nerve gap bridging, skin wound healing, digit tip repair as well as tooth regeneration. One of the key features of the specialized repair Schwann cells is that they become highly motile. They not only migrate into the area of damaged tissue and become a key component of regenerating tissue but also secrete signaling molecules to attract macrophages, support neuronal survival, promote axonal regrowth, activate local mesenchymal stem cells, and interact with other cell types. Currently, the importance of migratory Schwann cells in tissue regeneration is most evident in the case of a peripheral nerve transection injury. Following nerve transection, Schwann cells from both proximal and distal nerve stumps migrate into the nerve bridge and form Schwann cell cords to guide axon regeneration. The formation of Schwann cell cords in the nerve bridge is key to successful peripheral nerve repair following transection injury. In this review, we first examine nerve bridge formation and the behavior of Schwann cell migration in the nerve bridge, and then discuss how migrating Schwann cells direct regenerating axons into the distal nerve. We also review the current understanding of signals that could activate Schwann cell migration and signals that Schwann cells utilize to direct axon regeneration. Understanding the molecular mechanism of Schwann cell migration could potentially offer new therapeutic strategies for peripheral nerve repair.
Collapse
Affiliation(s)
- Qing Min
- School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei Province, People's Republic of China
| | - David B Parkinson
- Peninsula Medical School, Faculty of Health, Plymouth University, Plymouth, Devon, UK
| | - Xin-Peng Dun
- School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei Province, People's Republic of China
- Peninsula Medical School, Faculty of Health, Plymouth University, Plymouth, Devon, UK
- The Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, People's Republic of China
| |
Collapse
|
20
|
Fadia NB, Bliley JM, DiBernardo GA, Crammond DJ, Schilling BK, Sivak WN, Spiess AM, Washington KM, Waldner M, Liao HT, James IB, Minteer DM, Tompkins-Rhoades C, Cottrill AR, Kim DY, Schweizer R, Bourne DA, Panagis GE, Asher Schusterman M, Egro FM, Campwala IK, Simpson T, Weber DJ, Gause T, Brooker JE, Josyula T, Guevara AA, Repko AJ, Mahoney CM, Marra KG. Long-gap peripheral nerve repair through sustained release of a neurotrophic factor in nonhuman primates. Sci Transl Med 2020; 12:12/527/eaav7753. [DOI: 10.1126/scitranslmed.aav7753] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 08/26/2019] [Accepted: 11/25/2019] [Indexed: 01/09/2023]
Abstract
Severe injuries to peripheral nerves are challenging to repair. Standard-of-care treatment for nerve gaps >2 to 3 centimeters is autografting; however, autografting can result in neuroma formation, loss of sensory function at the donor site, and increased operative time. To address the need for a synthetic nerve conduit to treat large nerve gaps, we investigated a biodegradable poly(caprolactone) (PCL) conduit with embedded double-walled polymeric microspheres encapsulating glial cell line–derived neurotrophic factor (GDNF) capable of providing a sustained release of GDNF for >50 days in a 5-centimeter nerve defect in a rhesus macaque model. The GDNF-eluting conduit (PCL/GDNF) was compared to a median nerve autograft and a PCL conduit containing empty microspheres (PCL/Empty). Functional testing demonstrated similar functional recovery between the PCL/GDNF-treated group (75.64 ± 10.28%) and the autograft-treated group (77.49 ± 19.28%); both groups were statistically improved compared to PCL/Empty-treated group (44.95 ± 26.94%). Nerve conduction velocity 1 year after surgery was increased in the PCL/GDNF-treated macaques (31.41 ± 15.34 meters/second) compared to autograft (25.45 ± 3.96 meters/second) and PCL/Empty (12.60 ± 3.89 meters/second) treatment. Histological analyses included assessment of Schwann cell presence, myelination of axons, nerve fiber density, and g-ratio. PCL/GDNF group exhibited a statistically greater average area occupied by individual Schwann cells at the distal nerve (11.60 ± 33.01 μm2) compared to autograft (4.62 ± 3.99 μm2) and PCL/Empty (4.52 ± 5.16 μm2) treatment groups. This study demonstrates the efficacious bridging of a long peripheral nerve gap in a nonhuman primate model using an acellular, biodegradable nerve conduit.
Collapse
Affiliation(s)
- Neil B. Fadia
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jacqueline M. Bliley
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | - Donald J. Crammond
- Department of Neurosurgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | - Wesley N. Sivak
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Alexander M. Spiess
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Kia M. Washington
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Matthias Waldner
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Han-Tsung Liao
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Isaac B. James
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Danielle M. Minteer
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | - Adam R. Cottrill
- Department of Neuroscience, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Deok-Yeol Kim
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Riccardo Schweizer
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Debra A. Bourne
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - George E. Panagis
- Department of Biology, University of Pittsburgh, Greensburg, PA 15601, USA
| | - M. Asher Schusterman
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Francesco M. Egro
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | - Tyler Simpson
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Douglas J. Weber
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Physical Medicine and Rehabilitation, University of Pittsburgh, Pittsburgh, PA 15213, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Trent Gause
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jack E. Brooker
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Tvisha Josyula
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Astrid A. Guevara
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Alexander J. Repko
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | | | - Kacey G. Marra
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15213, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| |
Collapse
|
21
|
Carvalho CR, Oliveira JM, Reis RL. Modern Trends for Peripheral Nerve Repair and Regeneration: Beyond the Hollow Nerve Guidance Conduit. Front Bioeng Biotechnol 2019; 7:337. [PMID: 31824934 PMCID: PMC6882937 DOI: 10.3389/fbioe.2019.00337] [Citation(s) in RCA: 180] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 10/30/2019] [Indexed: 12/13/2022] Open
Abstract
Peripheral nerve repair and regeneration remains among the greatest challenges in tissue engineering and regenerative medicine. Even though peripheral nerve injuries (PNIs) are capable of some degree of regeneration, frail recovery is seen even when the best microsurgical technique is applied. PNIs are known to be very incapacitating for the patient, due to the deprivation of motor and sensory abilities. Since there is no optimal solution for tackling this problem up to this day, the evolution in the field is constant, with innovative designs of advanced nerve guidance conduits (NGCs) being reported every day. As a basic concept, a NGC should act as a physical barrier from the external environment, concomitantly acting as physical guidance for the regenerative axons across the gap lesion. NGCs should also be able to retain the naturally released nerve growth factors secreted by the damaged nerve stumps, as well as reducing the invasion of scar tissue-forming fibroblasts to the injury site. Based on the neurobiological knowledge related to the events that succeed after a nerve injury, neuronal subsistence is subjected to the existence of an ideal environment of growth factors, hormones, cytokines, and extracellular matrix (ECM) factors. Therefore, it is known that multifunctional NGCs fabricated through combinatorial approaches are needed to improve the functional and clinical outcomes after PNIs. The present work overviews the current reports dealing with the several features that can be used to improve peripheral nerve regeneration (PNR), ranging from the simple use of hollow NGCs to tissue engineered intraluminal fillers, or to even more advanced strategies, comprising the molecular and gene therapies as well as cell-based therapies.
Collapse
Affiliation(s)
- Cristiana R. Carvalho
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, Guimarães, Portugal
| | - Joaquim M. Oliveira
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, Guimarães, Portugal
| | - Rui L. Reis
- 3B's Research Group, I3Bs – Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
- The Discoveries Centre for Regenerative and Precision Medicine, Headquarters at University of Minho, Avepark, Guimarães, Portugal
| |
Collapse
|
22
|
Sarhane KA, Tuffaha SH, Ibrahim Z, Cashman CR, Krick K, Martin R, Broyles JM, Cooney DS, Lee WPA, Mi R, Mao HQ, Höke A, Brandacher G. Glial Cell Line-Derived Neurotrophic Factor and Chondroitinase Promote Axonal Regeneration in a Chronic Denervation Animal Model. Neurotherapeutics 2019; 16:1283-1295. [PMID: 31148054 PMCID: PMC6985423 DOI: 10.1007/s13311-019-00745-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Functional recovery following nerve injury declines when target re-innervation is delayed. Currently, no intervention exists to improve outcomes after prolonged denervation. We explored the neuroregenerative effects of glial cell line-derived neurotrophic factor (GDNF) and chondroitinase (CDN) in a chronic denervation animal model. A fibrin-based sustained delivery method for growth factors was optimized in vitro and in vivo, and then tested in our animal model. GDNF, CDN, and GDNF+CDN were injected into the denervated stump at the time of nerve repair. Histomorphometry and retrograde labeling were used to assess axonal regeneration. The mechanisms promoting such regeneration were explored with immunofluorescence. Five weeks after repair, the GDNF+CDN group had the highest number and maturity of axons. GDNF was noted to preferentially promote axonal maturity, whereas CDN predominantly increased the number of axons. GDNF favored motor neuron regeneration, and upregulated Ki67 in Schwann cells. CDN did not favor motor versus sensory regeneration and was noted to cleave inhibitory endoneurial proteoglycans. Early measures of nerve regeneration after delayed repair are improved by activating Schwann cells and breaking down the inhibitory proteoglycans in the distal nerve segment, suggesting a role for GDNF+CDN to be translated for human nerve repairs.
Collapse
Affiliation(s)
- Karim A Sarhane
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University, Ross Research Building/Suite 749D, 720 Rutland Avenue, Baltimore, Maryland, 21205, USA
- Department of Surgery, University of Toledo College of Medicine, Toledo, Ohio, USA
| | - Sami H Tuffaha
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University, Ross Research Building/Suite 749D, 720 Rutland Avenue, Baltimore, Maryland, 21205, USA
| | - Zuhaib Ibrahim
- Institute for Advanced Reconstruction, Shrewsbury, New Jersey, USA
| | - Christopher R Cashman
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland, USA
| | - Kellin Krick
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Russell Martin
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Justin M Broyles
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University, Ross Research Building/Suite 749D, 720 Rutland Avenue, Baltimore, Maryland, 21205, USA
| | - Damon S Cooney
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University, Ross Research Building/Suite 749D, 720 Rutland Avenue, Baltimore, Maryland, 21205, USA
| | - W P Andrew Lee
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University, Ross Research Building/Suite 749D, 720 Rutland Avenue, Baltimore, Maryland, 21205, USA
| | - Ruifa Mi
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland, USA
| | - Hai-Quan Mao
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, Maryland, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, Maryland, USA
| | - Ahmet Höke
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland, USA
- Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland, USA
| | - Gerald Brandacher
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University, Ross Research Building/Suite 749D, 720 Rutland Avenue, Baltimore, Maryland, 21205, USA.
| |
Collapse
|
23
|
Controlling the dose-dependent, synergistic and temporal effects of NGF and GDNF by encapsulation in PLGA microparticles for use in nerve guidance conduits for the repair of large peripheral nerve defects. J Control Release 2019; 304:51-64. [DOI: 10.1016/j.jconrel.2019.05.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 04/18/2019] [Accepted: 05/01/2019] [Indexed: 12/13/2022]
|
24
|
Hercher D, Kerbl M, Schuh CMAP, Heinzel J, Gal L, Stainer M, Schmidhammer R, Hausner T, Redl H, Nógrádi A, Hacobian A. Spatiotemporal Differences in Gene Expression Between Motor and Sensory Autografts and Their Effect on Femoral Nerve Regeneration in the Rat. Front Cell Neurosci 2019; 13:182. [PMID: 31139050 PMCID: PMC6519304 DOI: 10.3389/fncel.2019.00182] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 04/12/2019] [Indexed: 12/31/2022] Open
Abstract
To improve the outcome after autologous nerve grafting in the clinic, it is important to understand the limiting variables such as distinct phenotypes of motor and sensory Schwann cells. This study investigated the properties of phenotypically different autografts in a 6 mm femoral nerve defect model in the rat, where the respective femoral branches distally of the inguinal bifurcation served as homotopic, or heterotopic autografts. Axonal regeneration and target reinnervation was analyzed by gait analysis, electrophysiology, and wet muscle mass analysis. We evaluated regeneration-associated gene expression between 5 days and 10 weeks after repair, in the autografts as well as the proximal, and distal segments of the femoral nerve using qRT-PCR. Furthermore we investigated expression patterns of phenotypically pure ventral and dorsal roots. We identified highly significant differences in gene expression of a variety of regeneration-associated genes along the central – peripheral axis in healthy femoral nerves. Phenotypically mismatched grafting resulted in altered spatiotemporal expression of neurotrophic factor BDNF, GDNF receptor GFRα1, cell adhesion molecules Cadm3, Cadm4, L1CAM, and proliferation associated Ki67. Although significantly higher quadriceps muscle mass following homotopic nerve grafting was measured, we did not observe differences in gait analysis, and electrophysiological parameters between treatment paradigms. Our study provides evidence for phenotypic commitment of autologous nerve grafts after injury and gives a conclusive overview of temporal expression of several important regeneration-associated genes after repair with sensory or motor graft.
Collapse
Affiliation(s)
- David Hercher
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Markus Kerbl
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Christina M A P Schuh
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Centro de Medicina Regenerativa, Facultad de Medicina Clínica Alemana-Universidad del Desarrollo, Santiago, Chile
| | - Johannes Heinzel
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - László Gal
- Department of Anatomy, Histology and Embryology, University of Szeged, Szeged, Hungary
| | - Michaela Stainer
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Robert Schmidhammer
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Thomas Hausner
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Heinz Redl
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Antal Nógrádi
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria.,Department of Anatomy, Histology and Embryology, University of Szeged, Szeged, Hungary
| | - Ara Hacobian
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Vienna, Austria.,Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
25
|
Jessen KR, Arthur-Farraj P. Repair Schwann cell update: Adaptive reprogramming, EMT, and stemness in regenerating nerves. Glia 2019; 67:421-437. [PMID: 30632639 DOI: 10.1002/glia.23532] [Citation(s) in RCA: 234] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/20/2018] [Accepted: 09/05/2018] [Indexed: 12/16/2022]
Abstract
Schwann cells respond to nerve injury by cellular reprogramming that generates cells specialized for promoting regeneration and repair. These repair cells clear redundant myelin, attract macrophages, support survival of damaged neurons, encourage axonal growth, and guide axons back to their targets. There are interesting parallels between this response and that found in other tissues. At the cellular level, many other tissues also react to injury by cellular reprogramming, generating cells specialized to promote tissue homeostasis and repair. And at the molecular level, a common feature possessed by Schwann cells and many other cells is the injury-induced activation of genes associated with epithelial-mesenchymal transitions and stemness, differentiation states that are linked to cellular plasticity and that help injury-induced tissue remodeling. The number of signaling systems regulating Schwann cell plasticity is rapidly increasing. Importantly, this includes mechanisms that are crucial for the generation of functional repair Schwann cells and nerve regeneration, although they have no or a minor role elsewhere in the Schwann cell lineage. This encourages the view that selective tools can be developed to control these particular cells, amplify their repair supportive functions and prevent their deterioration. In this review, we discuss the emerging similarities between the injury response seen in nerves and in other tissues and survey the transcription factors, epigenetic mechanisms, and signaling cascades that control repair Schwann cells, with emphasis on systems that selectively regulate the Schwann cell injury response.
Collapse
Affiliation(s)
- Kristjan R Jessen
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Peter Arthur-Farraj
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
26
|
Dong C, Ubogu EE. GDNF enhances human blood-nerve barrier function in vitro via MAPK signaling pathways. Tissue Barriers 2018; 6:1-22. [PMID: 30523753 PMCID: PMC6389127 DOI: 10.1080/21688370.2018.1546537] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 10/29/2018] [Accepted: 11/07/2018] [Indexed: 01/24/2023] Open
Abstract
The human blood-nerve barrier (BNB) formed by endoneurial microvascular endothelial cells, serves to maintain the internal microenvironment in peripheral nerves required for normal axonal signal transduction to and from the central nervous system. The mechanisms of human BNB formation in health and disease are not fully elucidated. Prior work established a sufficient role for glial-derived neurotrophic factor (GDNF) in enhancing human BNB biophysical properties following serum withdrawal in vitro via RET-tyrosine kinase-dependent cytoskeletal remodeling. The objective of the study was to ascertain the downstream signaling pathway involved in this process and more comprehensively determine the molecular changes that may occur at human BNB intercellular junctions under the influence of GDNF. Proteomic studies suggested expression of several mitogen-activated protein kinases (MAPKs) in confluent GDNF-treated endoneurial endothelial cells following serum withdrawal. Using electric cell-substrate impedance sensing to continuously measure transendothelial electrical resistance and static transwell solute permeability assays with fluoresceinated small and large molecules to evaluate BNB biophysical function, we determined MAPK signaling was essential for GDNF-mediated BNB TEER increase following serum withdrawal downstream of RET-tyrosine kinase signaling that persisted for up to 48 hours in vitro. This increase was associated with reduced solute permeability to fluoresceinated sodium and high molecular weight dextran. Specific GDNF-mediated alterations were detected in cytoskeletal and intercellular junctional complex molecular transcripts and proteins relative to basal conditions without exogenous GDNF. This work provides novel insights into the molecular determinants and mechanisms responsible for specialized restrictive human BNB formation in health and disease.
Collapse
Affiliation(s)
- Chaoling Dong
- Neuromuscular Immunopathology Research Laboratory, Division of Neuromuscular Disease, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Eroboghene E. Ubogu
- Neuromuscular Immunopathology Research Laboratory, Division of Neuromuscular Disease, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
27
|
Cairns DM, Giordano JE, Conte S, Levin M, Kaplan DL. Ivermectin Promotes Peripheral Nerve Regeneration during Wound Healing. ACS OMEGA 2018; 3:12392-12402. [PMID: 30411007 PMCID: PMC6210064 DOI: 10.1021/acsomega.8b01451] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 09/13/2018] [Indexed: 06/08/2023]
Abstract
Peripheral nerves have the capacity to regenerate due to the presence of neuroprotective glia of the peripheral nervous system, Schwann cells. Upon peripheral nerve injury, Schwann cells create a permissive microenvironment for neuronal regrowth by taking up cytotoxic glutamate and secreting neurotrophic signaling molecules. Impaired peripheral nerve repair is often caused by a defective Schwann cell response after injury, and there is a critical need to develop new strategies to enhance nerve regeneration, especially in organisms with restricted regenerative potential, such as humans. One approach is to explore mechanisms in lower organisms, in which nerve repair is much more efficient. A recent study demonstrated that the antiparasitic drug, ivermectin, caused hyperinnervation of primordial eye tissue in Xenopus laevis tadpoles. Our study aimed to examine the role of ivermectin in mammalian nerve repair. We performed in vitro assays utilizing human induced neural stem cells (hiNSCs) in co-culture with human dermal fibroblasts (hDFs) and found that ivermectin-treated hDFs promote hiNSC proliferation and migration. We also characterized the effects of ivermectin on hDFs and found that ivermectin causes hDFs to uptake extracellular glutamate, secrete glial cell-derived neurotrophic factor, develop an elongated bipolar morphology, and express glial fibrillary acidic protein. Finally, in a corresponding in vivo model, we found that localized ivermectin treatment in a dermal wound site induced the upregulation of both glial and neuronal markers upon healing. Taken together, we demonstrate that ivermectin promotes peripheral nerve regeneration by inducing fibroblasts to adopt a glia-like phenotype.
Collapse
Affiliation(s)
- Dana M. Cairns
- Department
of Biomedical Engineering, Department of Biology, Allen Discovery Center, and Tufts University
Biomedical Engineering Research Scholars Program (TUBERS), Tufts University, Medford, Massachusetts 02155, United States
| | - Jodie E. Giordano
- Department
of Biomedical Engineering, Department of Biology, Allen Discovery Center, and Tufts University
Biomedical Engineering Research Scholars Program (TUBERS), Tufts University, Medford, Massachusetts 02155, United States
| | - Sylvia Conte
- Department
of Biomedical Engineering, Department of Biology, Allen Discovery Center, and Tufts University
Biomedical Engineering Research Scholars Program (TUBERS), Tufts University, Medford, Massachusetts 02155, United States
| | - Michael Levin
- Department
of Biomedical Engineering, Department of Biology, Allen Discovery Center, and Tufts University
Biomedical Engineering Research Scholars Program (TUBERS), Tufts University, Medford, Massachusetts 02155, United States
| | - David L. Kaplan
- Department
of Biomedical Engineering, Department of Biology, Allen Discovery Center, and Tufts University
Biomedical Engineering Research Scholars Program (TUBERS), Tufts University, Medford, Massachusetts 02155, United States
| |
Collapse
|
28
|
Dong C, Helton ES, Zhou P, Ouyang X, d'Anglemont de Tassigny X, Pascual A, López-Barneo J, Ubogu EE. Glial-derived neurotrophic factor is essential for blood-nerve barrier functional recovery in an experimental murine model of traumatic peripheral neuropathy. Tissue Barriers 2018; 6:1-22. [PMID: 29913111 DOI: 10.1080/21688370.2018.1479570] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
There is emerging evidence that glial-derived neurotrophic factor (GDNF) is a potent inducer of restrictive barrier function in tight junction-forming microvascular endothelium and epithelium, including the human blood-nerve barrier (BNB) in vitro. We sought to determine the role of GDNF in restoring BNB function in vivo by evaluating sciatic nerve horseradish peroxidase (HRP) permeability in tamoxifen-inducible GDNF conditional knockout (CKO) adult mice following non-transecting crush injury via electron microscopy, with appropriate wildtype (WT) and heterozygous (HET) littermate controls. A total of 24 age-, genotype- and sex-matched mice >12 weeks of age were injected with 30 mg/kg HRP via tail vein injection 7 or 14 days following unilateral sciatic nerve crush, and both sciatic nerves were harvested 30 minutes later for morphometric assessment by light and electron microscopy. The number and percentage of HRP-permeable endoneurial microvessels were ascertained to determine the effect of GDNF in restoring barrier function in vivo. Following sciatic nerve crush, there was significant upregulation in GDNF protein expression in WT and HET mice that was abrogated in CKO mice. GDNF significantly restored sciatic nerve BNB HRP impermeability to near normal levels by day 7, with complete restoration seen by day 14 in WT and HET mice. A significant recovery lag was observed in CKO mice. This effect was independent on VE-Cadherin or claudin-5 expression on endoneurial microvessels. These results imply an important role of GDNF in restoring restrictive BNB function in vivo, suggesting a potential strategy to re-establish the restrictive endoneurial microenvironment following traumatic peripheral neuropathies.
Collapse
Affiliation(s)
- Chaoling Dong
- a Neuromuscular Immunopathology Research Laboratory, Division of Neuromuscular Disease, Department of Neurology , University of Alabama at Birmingham , Birmingham , Alabama , United States of America
| | - E Scott Helton
- a Neuromuscular Immunopathology Research Laboratory, Division of Neuromuscular Disease, Department of Neurology , University of Alabama at Birmingham , Birmingham , Alabama , United States of America
| | - Ping Zhou
- a Neuromuscular Immunopathology Research Laboratory, Division of Neuromuscular Disease, Department of Neurology , University of Alabama at Birmingham , Birmingham , Alabama , United States of America
| | - Xuan Ouyang
- a Neuromuscular Immunopathology Research Laboratory, Division of Neuromuscular Disease, Department of Neurology , University of Alabama at Birmingham , Birmingham , Alabama , United States of America
| | - Xavier d'Anglemont de Tassigny
- b Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla , Seville , Spain
| | - Alberto Pascual
- b Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla , Seville , Spain
| | - José López-Barneo
- b Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla , Seville , Spain
| | - Eroboghene E Ubogu
- a Neuromuscular Immunopathology Research Laboratory, Division of Neuromuscular Disease, Department of Neurology , University of Alabama at Birmingham , Birmingham , Alabama , United States of America
| |
Collapse
|
29
|
Oliveira-Giacomelli Á, Naaldijk Y, Sardá-Arroyo L, Gonçalves MCB, Corrêa-Velloso J, Pillat MM, de Souza HDN, Ulrich H. Purinergic Receptors in Neurological Diseases With Motor Symptoms: Targets for Therapy. Front Pharmacol 2018; 9:325. [PMID: 29692728 PMCID: PMC5902708 DOI: 10.3389/fphar.2018.00325] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Accepted: 03/21/2018] [Indexed: 12/13/2022] Open
Abstract
Since proving adenosine triphosphate (ATP) functions as a neurotransmitter in neuron/glia interactions, the purinergic system has been more intensely studied within the scope of the central nervous system. In neurological disorders with associated motor symptoms, including Parkinson's disease (PD), motor neuron diseases (MND), multiple sclerosis (MS), amyotrophic lateral sclerosis (ALS), Huntington's Disease (HD), restless leg syndrome (RLS), and ataxias, alterations in purinergic receptor expression and activity have been noted, indicating a potential role for this system in disease etiology and progression. In neurodegenerative conditions, neural cell death provokes extensive ATP release and alters calcium signaling through purinergic receptor modulation. Consequently, neuroinflammatory responses, excitotoxicity and apoptosis are directly or indirectly induced. This review analyzes currently available data, which suggests involvement of the purinergic system in neuro-associated motor dysfunctions and underlying mechanisms. Possible targets for pharmacological interventions are also discussed.
Collapse
Affiliation(s)
| | - Yahaira Naaldijk
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Laura Sardá-Arroyo
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Maria C. B. Gonçalves
- Department of Neurology and Neuroscience, Medical School, Federal University of São Paulo, São Paulo, Brazil
| | - Juliana Corrêa-Velloso
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Micheli M. Pillat
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Héllio D. N. de Souza
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| | - Henning Ulrich
- Department of Biochemistry, Institute of Chemistry, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
30
|
Affiliation(s)
- Lei Li
- Department of Neuroscience, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA
| | - Wen-Cheng Xiong
- Department of Neuroscience, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA
- Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio 44106, USA
| | - Lin Mei
- Department of Neuroscience, School of Medicine, Case Western Reserve University, Cleveland, Ohio 44106, USA
- Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio 44106, USA
| |
Collapse
|
31
|
Saxena A, Scaini G, Bavaresco DV, Leite C, Valvassori SS, Carvalho AF, Quevedo J. Role of Protein Kinase C in Bipolar Disorder: A Review of the Current Literature. MOLECULAR NEUROPSYCHIATRY 2017; 3:108-124. [PMID: 29230399 DOI: 10.1159/000480349] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Accepted: 08/14/2017] [Indexed: 12/19/2022]
Abstract
Bipolar disorder (BD) is a major health problem. It causes significant morbidity and imposes a burden on the society. Available treatments help a substantial proportion of patients but are not beneficial for an estimated 40-50%. Thus, there is a great need to further our understanding the pathophysiology of BD to identify new therapeutic avenues. The preponderance of evidence pointed towards a role of protein kinase C (PKC) in BD. We reviewed the literature pertinent to the role of PKC in BD. We present recent advances from preclinical and clinical studies that further support the role of PKC. Moreover, we discuss the role of PKC on synaptogenesis and neuroplasticity in the context of BD. The recent development of animal models of BD, such as stimulant-treated and paradoxical sleep deprivation, and the ability to intervene pharmacologically provide further insights into the involvement of PKC in BD. In addition, the effect of PKC inhibitors, such as tamoxifen, in the resolution of manic symptoms in patients with BD further points in that direction. Furthermore, a wide variety of growth factors influence neurotransmission through several molecular pathways that involve downstream effects of PKC. Our current understanding identifies the PKC pathway as a potential therapeutic avenue for BD.
Collapse
Affiliation(s)
- Ashwini Saxena
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Giselli Scaini
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Daniela V Bavaresco
- Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, Brazil
| | - Camila Leite
- Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, Brazil
| | - Samira S Valvassori
- Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, Brazil
| | - André F Carvalho
- Translational Psychiatry Research Group, Faculty of Medicine, Federal University of Ceara, Fortaleza, Brazil
| | - João Quevedo
- Translational Psychiatry Program, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA.,Laboratory of Neurosciences, Graduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, Brazil.,Center of Excellence on Mood Disorders, Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, Texas, USA.,Neuroscience Graduate Program, The University of Texas Graduate School of Biomedical Sciences at Houston, Houston, Texas, USA
| |
Collapse
|
32
|
Valvassori SS, Borges CP, Varela RB, Bavaresco DV, Bianchini G, Mariot E, Arent CO, Resende WR, Budni J, Quevedo J. The different effects of lithium and tamoxifen on memory formation and the levels of neurotrophic factors in the brain of male and female rats. Brain Res Bull 2017; 134:228-235. [DOI: 10.1016/j.brainresbull.2017.08.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 08/03/2017] [Accepted: 08/07/2017] [Indexed: 01/22/2023]
|
33
|
Allergic Inflammation Leads to Neuropathic Pain via Glial Cell Activation. J Neurosci 2017; 36:11929-11945. [PMID: 27881779 DOI: 10.1523/jneurosci.1981-16.2016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 09/23/2016] [Accepted: 09/27/2016] [Indexed: 01/08/2023] Open
Abstract
Allergic and atopic disorders have increased over the past few decades and have been associated with neuropsychiatric conditions, such as autism spectrum disorder and asthmatic amyotrophy. Myelitis presenting with neuropathic pain can occur in patients with atopic disorder; however, the relationship between allergic inflammation and neuropathic pain, and the underlying mechanism, remains to be established. We studied whether allergic inflammation affects the spinal nociceptive system. We found that mice with asthma, atopic dermatitis, or atopic diathesis had widespread and significantly more activated microglia and astroglia in the spinal cord than those without atopy, and displayed tactile allodynia. Microarray analysis of isolated microglia revealed a dysregulated phenotype showing upregulation of M1 macrophage markers and downregulation of M2 markers in atopic mice. Among the cell surface protein genes, endothelin receptor type B (EDNRB) was most upregulated. Immunohistochemical analysis revealed that EDNRB expression was enhanced in microglia and astroglia, whereas endothelin-1, an EDNRB ligand, was increased in serum, lungs, and epidermis of atopic mice. No EDNRA expression was found in the spinal cord. Expression of FBJ murine osteosarcoma viral oncogene homolog B was significantly higher in the dorsal horn neurons of asthma mice than nonatopic mice. The EDNRB antagonist BQ788 abolished glial and neural activation and allodynia. We found increased serum endothelin-1 in atopic patients with myelitis and neuropathic pain, and activation of spinal microglia and astroglia with EDNRB upregulation in an autopsied case. These results suggest that allergic inflammation induces diffuse glial activation, influencing the nociceptive system via the EDNRB pathway. SIGNIFICANCE STATEMENT The prevalence of allergic disorders has markedly increased over the past few decades. Allergic disorders are associated with neuropsychiatric conditions; however, the relationship between allergic inflammation and CNS complications is unknown. A peculiar myelitis presenting with persistent neuropathic pain has been reported in patients with allergic disorders. We studied how atopy exerts substantial influence on the nociceptive system. We found that mice with allergic disorders had severe allodynia with activated astroglia and microglia, and showed marked upregulation of endothelin-1 (ET-1) receptor type B (EDNRB) in the spinal cord. A selective EDNRB antagonist prevented allodynia and glial activation. Our findings suggest a novel mechanism whereby atopy induces glial activation and neuropathic pain via an ET-1/EDNRB pathway.
Collapse
|
34
|
Hsieh CH, Rau CS, Kuo PJ, Liu SH, Wu CJ, Lu TH, Wu YC, Lin CW. Knockout of toll-like receptor impairs nerve regeneration after a crush injury. Oncotarget 2017; 8:80741-80756. [PMID: 29113341 PMCID: PMC5655236 DOI: 10.18632/oncotarget.20206] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 07/12/2017] [Indexed: 01/01/2023] Open
Abstract
Background Toll-like receptors (TLRs) are involved in the initiation of Schwann cell activation and subsequent recruitment of macrophages for clearance of degenerated myelin and neuronal debris after nerve injury. The present study was designed to investigate the regenerative outcome and expression of myelination-related factors in Tlr-knockout mice following a sciatic nerve crush injury. Materials and methods A standard sciatic nerve crush injury, induced by applying constant pressure to the nerve with a No. 5 jeweler's forceps for 30 s, was performed in C57BL/6, Tlr2−/−, Tlr3−/−, Tlr4−/−, Tlr5−/−, and Tlr7−/− mice. Quantitative histomorphometric analysis of toluidine blue-stained nerve specimens and walking track analysis were performed to evaluate nerve regeneration outcomes. PCR Arrays were used to detect the expression of neurogenesis-related genes of dorsal root ganglia as well as of myelination-related genes of the distal nerve segments. Results Worse nerve regeneration after nerve crush injury was found in all Tlr-knockout mice than in C57BL/6 mice. Delayed expression of myelin genes and a different expression pattern of myelination-related neurotrophin genes and transcription factors were found in Tlr-knockout mice in comparison to C57BL/6 mice. In these TLR-mediated pathways, insulin-like growth factor 2 and brain-derived neurotrophic factor, as well as early growth response 2 and N-myc downstream-regulated gene 1, were significantly decreased in the early and late stages, respectively, of nerve regeneration after a crush injury. Conclusions Knockout of Tlr genes decreases the expression of myelination-related factors and impairs nerve regeneration after a sciatic nerve crush injury.
Collapse
Affiliation(s)
- Ching-Hua Hsieh
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan.,Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Cheng-Shyuan Rau
- Department of Neurosurgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Pao-Jen Kuo
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Shu-Hsuan Liu
- Faculty of Health Sciences, McMaster University, Hamilton, Canada
| | - Chia-Jung Wu
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Tsu-Hsiang Lu
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Yi-Chan Wu
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chia-Wei Lin
- Department of Plastic and Reconstructive Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
| |
Collapse
|
35
|
Das SR, Uz M, Ding S, Lentner MT, Hondred JA, Cargill AA, Sakaguchi DS, Mallapragada S, Claussen JC. Electrical Differentiation of Mesenchymal Stem Cells into Schwann-Cell-Like Phenotypes Using Inkjet-Printed Graphene Circuits. Adv Healthc Mater 2017; 6. [PMID: 28218474 DOI: 10.1002/adhm.201601087] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 12/22/2016] [Indexed: 01/05/2023]
Abstract
Graphene-based materials (GBMs) have displayed tremendous promise for use as neurointerfacial substrates as they enable favorable adhesion, growth, proliferation, spreading, and migration of immobilized cells. This study reports the first case of the differentiation of mesenchymal stem cells (MSCs) into Schwann cell (SC)-like phenotypes through the application of electrical stimuli from a graphene-based electrode. Electrical differentiation of MSCs into SC-like phenotypes is carried out on a flexible, inkjet-printed graphene interdigitated electrode (IDE) circuit that is made highly conductive (sheet resistance < 1 kΩ/sq) via a postprint pulse-laser annealing process. MSCs immobilized on the graphene printed IDEs and electrically stimulated/treated (etMSCs) display significant enhanced cellular differentiation and paracrine activity above conventional chemical treatment strategies [≈85% of the etMSCs differentiated into SC-like phenotypes with ≈80 ng mL-1 of nerve growth factor (NGF) secretion vs. 75% and ≈55 ng mL-1 for chemically treated MSCs (ctMSCs)]. These results help pave the way for in vivo peripheral nerve regeneration where the flexible graphene electrodes could conform to the injury site and provide intimate electrical simulation for nerve cell regrowth.
Collapse
Affiliation(s)
- Suprem R. Das
- Department of Mechanical Engineering Iowa State University Ames IA 50011 USA
- Division of Materials Science and Engineering Ames Laboratory Department of Energy Ames IA 50011 USA
| | - Metin Uz
- Department of Chemical and Biological Engineering Iowa State University Ames IA 50011 USA
| | - Shaowei Ding
- Department of Mechanical Engineering Iowa State University Ames IA 50011 USA
| | - Matthew T. Lentner
- Department of Chemical and Biological Engineering Iowa State University Ames IA 50011 USA
| | - John A. Hondred
- Department of Mechanical Engineering Iowa State University Ames IA 50011 USA
| | - Allison A. Cargill
- Department of Mechanical Engineering Iowa State University Ames IA 50011 USA
| | - Donald S. Sakaguchi
- Neuroscience Program Iowa State University Ames IA 50011 USA
- Department of Genetics Development and Cell Biology Iowa State University Ames IA 50011 USA
| | - Surya Mallapragada
- Division of Materials Science and Engineering Ames Laboratory Department of Energy Ames IA 50011 USA
- Department of Chemical and Biological Engineering Iowa State University Ames IA 50011 USA
| | - Jonathan C. Claussen
- Department of Mechanical Engineering Iowa State University Ames IA 50011 USA
- Division of Materials Science and Engineering Ames Laboratory Department of Energy Ames IA 50011 USA
| |
Collapse
|
36
|
Wang Y, Ji Y, Zhao Y, Kong Y, Gao M, Feng Q, Wu Y, Yang Y. Effects of surface functional groups on proliferation and biofunction of Schwann cells. J Biomater Appl 2016; 30:1494-504. [DOI: 10.1177/0885328216628785] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Scaffolds in tissue engineering should be rationally designed to become an adhesion substrate friendly to cells. Schwann cells play an important role in nerve regeneration and repair. Previous studies have suggested that surface chemical groups have effect on many types of cells. However, there have hitherto been few reports on Schwann cells. In this study, we investigated cell adhesion, survival, proliferation, and neurotrophic actions of Schwann cells cultured on glass coverslips modified with different chemical groups, including methyl, carboxyl, amino, hydroxyl, mercapto, and sulfonic groups. Schwann cells on amino and carboxyl surfaces had higher attachment rate, presenting good morphology, high proliferation, and strong neurotrophic functions, while on methyl surfaces, few cells can survive, cells shrunk into round shape, exhibiting poor proliferation and weak neurotrophic functions. Growth of cells on other groups was between methyl and amino, carboxyl, and had little difference among them. Our data indicated that chemical groups can regulate behavior of Schwann cells, indicating a way to design new scaffolds for peripheral nerve regeneration.
Collapse
Affiliation(s)
- Yaling Wang
- Key Laboratory of Eco-Textiles, Ministry of Education, Jiangnan University, Wuxi, P. R. China
- School of Chemistry and Chemical Engineering, Nantong University, Nantong, P. R. China
| | - Yawei Ji
- Department of cardiology, Affiliated Hospital of Nantong University, Nantong, P.R. China
| | - Yahong Zhao
- Key Laboratory of Eco-Textiles, Ministry of Education, Jiangnan University, Wuxi, P. R. China
- Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, P. R. China
| | - Yan Kong
- Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, P. R. China
| | - Ming Gao
- Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, P. R. China
| | - Qilin Feng
- School of Medical, Nantong University, Nantong, P. R. China
| | - Yue Wu
- Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, P. R. China
| | - Yumin Yang
- Key Laboratory of Eco-Textiles, Ministry of Education, Jiangnan University, Wuxi, P. R. China
- Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, P. R. China
| |
Collapse
|
37
|
Yang JH, Lv JG, Wang H, Nie HY. Electroacupuncture promotes the recovery of motor neuron function in the anterior horn of the injured spinal cord. Neural Regen Res 2016; 10:2033-9. [PMID: 26889195 PMCID: PMC4730831 DOI: 10.4103/1673-5374.172323] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Acupuncture has been shown to lessen the inflammatory reaction after acute spinal cord injury and reduce secondary injury. However, the mechanism of action remains unclear. In this study, a rat model of spinal cord injury was established by compressing the T8–9 segments using a modified Nystrom method. Twenty-four hours after injury, Zusanli (ST36), Xuanzhong (GB39), Futu (ST32) and Sanyinjiao (SP6) were stimulated with electroacupuncture. Rats with spinal cord injury alone were used as controls. At 2, 4 and 6 weeks after injury, acetylcholinesterase (AChE) activity at the site of injury, the number of medium and large neurons in the spinal cord anterior horn, glial cell line-derived neurotrophic factor (GDNF) mRNA expression, and Basso, Beattie and Bresnahan locomotor rating scale scores were greater in the electroacupuncture group compared with the control group. These results demonstrate that electroacupuncture increases AChE activity, up-regulates GDNF mRNA expression, and promotes the recovery of motor neuron function in the anterior horn after spinal cord injury.
Collapse
Affiliation(s)
- Jian-Hui Yang
- Rehabilitation Center, First Affiliated Hospital of Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Jian-Guo Lv
- Rehabilitation Center, First Affiliated Hospital of Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Hui Wang
- Rehabilitation Center, First Affiliated Hospital of Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| | - Hui-Yong Nie
- Rehabilitation Center, First Affiliated Hospital of Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi Province, China
| |
Collapse
|
38
|
Assunção-Silva RC, Oliveira CC, Ziv-Polat O, Gomes ED, Sahar A, Sousa N, Silva NA, Salgado AJ. Induction of neurite outgrowth in 3D hydrogel-based environments. Biomed Mater 2015; 10:051001. [DOI: 10.1088/1748-6041/10/5/051001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
39
|
Pinkernelle J, Raffa V, Calatayud MP, Goya GF, Riggio C, Keilhoff G. Growth factor choice is critical for successful functionalization of nanoparticles. Front Neurosci 2015; 9:305. [PMID: 26388717 PMCID: PMC4557102 DOI: 10.3389/fnins.2015.00305] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Accepted: 08/12/2015] [Indexed: 12/16/2022] Open
Abstract
Nanoparticles (NPs) show new characteristics compared to the corresponding bulk material. These nanoscale properties make them interesting for various applications in biomedicine and life sciences. One field of application is the use of magnetic NPs to support regeneration in the nervous system. Drug delivery requires a functionalization of NPs with bio-functional molecules. In our study, we functionalized self-made PEI-coated iron oxide NPs with nerve growth factor (NGF) and glial cell-line derived neurotrophic factor (GDNF). Next, we tested the bio-functionality of NGF in a rat pheochromocytoma cell line (PC12) and the bio-functionality of GDNF in an organotypic spinal cord culture. Covalent binding of NGF to PEI-NPs impaired bio-functionality of NGF, but non-covalent approach differentiated PC12 cells reliably. Non-covalent binding of GDNF showed a satisfying bio-functionality of GDNF:PEI-NPs, but turned out to be unstable in conjugation to the PEI-NPs. Taken together, our study showed the importance of assessing bio-functionality and binding stability of functionalized growth factors using proper biological models. It also shows that successful functionalization of magnetic NPs with growth factors is dependent on the used binding chemistry and that it is hardly predictable. For use as therapeutics, functionalization strategies have to be reproducible and future studies are needed.
Collapse
Affiliation(s)
- Josephine Pinkernelle
- Department of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University of MagdeburgMagdeburg, Germany
- Institute for Biochemistry and Cell Biology, Otto-von-Guericke University of MagdeburgMagdeburg, Germany
| | - Vittoria Raffa
- Department of Biology, University of PisaPisa, Italy
- Institute of Life Science, Scuola Superiore Sant' AnnaPisa, Italy
| | | | - Gerado F. Goya
- Aragon Institute of Nanosciences, University of ZaragozaZaragoza, Spain
- Department of Condensed Matter Physics, University of ZaragozaSpain
| | - Cristina Riggio
- Institute of Life Science, Scuola Superiore Sant' AnnaPisa, Italy
| | - Gerburg Keilhoff
- Department of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke University of MagdeburgMagdeburg, Germany
| |
Collapse
|
40
|
Dennys CN, Armstrong J, Levy M, Byun YJ, Ramdial KR, Bott M, Rossi FH, Fernández-Valle C, Franco MC, Estevez AG. Chronic inhibitory effect of riluzole on trophic factor production. Exp Neurol 2015; 271:301-7. [PMID: 26071088 PMCID: PMC4864959 DOI: 10.1016/j.expneurol.2015.05.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 05/18/2015] [Accepted: 05/23/2015] [Indexed: 12/14/2022]
Abstract
Riluzole is the only FDA approved drug for the treatment of amyotrophic lateral sclerosis (ALS). However, the drug affords moderate protection to ALS patients, extending life for a few months by a mechanism that remains controversial. In the presence of riluzole, astrocytes increase the production of factors protective to motor neurons. The stimulation of trophic factor production by motor neuron associated cells may contribute to riluzole's protective effect in ALS. Here, we investigated the effects of media conditioned by astrocytes and Schwann cells acutely or chronically incubated with riluzole on trophic factor-deprived motor neuron survival. While acute riluzole incubation induced CT-1 secretion by astrocytes and Schwann cells, chronic treatment stimulated a significant decrease in trophic factor production compared to untreated cultures. Accordingly, conditioned media from astrocytes and Schwann cells acutely treated with riluzole protected motor neurons from trophic factor deprivation-induced cell death. Motor neuron protection was prevented by incubation with CT-1 neutralizing antibodies. In contrast, conditioned media from astrocytes and Schwann cells chronically treated with riluzole was not protective. Acute and chronic treatment of mice with riluzole showed opposite effects on trophic factor production in spinal cord, sciatic nerve and brain. There was an increase in the production of CT-1 and GDNF in the spinal cord and CT-1 in the sciatic nerve during the first days of treatment with riluzole, but the levels dropped significantly after chronic treatment with the drug. Similar results were observed in brain for CT-1 and BDNF while there was no change in GDNF levels after riluzole treatment. Our results reveal that riluzole regulates long-lasting processes involving protein synthesis, which may be relevant for riluzole therapeutic effects. Changing the regimen of riluzole administration to favor the acute effect of the drug on trophic factor production by discontinuous long-term treatment may improve the outcome of ALS patient therapy.
Collapse
Affiliation(s)
- Cassandra N Dennys
- Burnett School of Biomedical Science, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd., Orlando, FL 32827, United States
| | - JeNay Armstrong
- Burnett School of Biomedical Science, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd., Orlando, FL 32827, United States
| | - Mark Levy
- Burnett School of Biomedical Science, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd., Orlando, FL 32827, United States
| | - Youn Jung Byun
- Burnett School of Biomedical Science, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd., Orlando, FL 32827, United States
| | - Kristina R Ramdial
- Burnett School of Biomedical Science, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd., Orlando, FL 32827, United States
| | - Marga Bott
- Burnett School of Biomedical Science, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd., Orlando, FL 32827, United States
| | - Fabian H Rossi
- Orlando Veteran Administration Healthcare System, Orlando, FL 32803, United States
| | - Cristina Fernández-Valle
- Burnett School of Biomedical Science, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd., Orlando, FL 32827, United States
| | - Maria Clara Franco
- Burnett School of Biomedical Science, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd., Orlando, FL 32827, United States
| | - Alvaro G Estevez
- Burnett School of Biomedical Science, College of Medicine, University of Central Florida, 6900 Lake Nona Blvd., Orlando, FL 32827, United States.
| |
Collapse
|
41
|
Bencsik N, Szíber Z, Liliom H, Tárnok K, Borbély S, Gulyás M, Rátkai A, Szűcs A, Hazai-Novák D, Ellwanger K, Rácz B, Pfizenmaier K, Hausser A, Schlett K. Protein kinase D promotes plasticity-induced F-actin stabilization in dendritic spines and regulates memory formation. J Cell Biol 2015; 210:771-83. [PMID: 26304723 PMCID: PMC4555815 DOI: 10.1083/jcb.201501114] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Accepted: 07/23/2015] [Indexed: 02/07/2023] Open
Abstract
PKD regulates the stabilization of the F-actin network within dendritic spines upon chemically induced plasticity changes and is needed for proper hippocampal LTP and spatial memory formation. Actin turnover in dendritic spines influences spine development, morphology, and plasticity, with functional consequences on learning and memory formation. In nonneuronal cells, protein kinase D (PKD) has an important role in stabilizing F-actin via multiple molecular pathways. Using in vitro models of neuronal plasticity, such as glycine-induced chemical long-term potentiation (LTP), known to evoke synaptic plasticity, or long-term depolarization block by KCl, leading to homeostatic morphological changes, we show that actin stabilization needed for the enlargement of dendritic spines is dependent on PKD activity. Consequently, impaired PKD functions attenuate activity-dependent changes in hippocampal dendritic spines, including LTP formation, cause morphological alterations in vivo, and have deleterious consequences on spatial memory formation. We thus provide compelling evidence that PKD controls synaptic plasticity and learning by regulating actin stability in dendritic spines.
Collapse
Affiliation(s)
- Norbert Bencsik
- Department of Physiology and Neurobiology, Eötvös Loránd University, H-1117 Budapest, Hungary
| | - Zsófia Szíber
- Department of Physiology and Neurobiology, Eötvös Loránd University, H-1117 Budapest, Hungary
| | - Hanna Liliom
- Department of Physiology and Neurobiology, Eötvös Loránd University, H-1117 Budapest, Hungary
| | - Krisztián Tárnok
- Department of Physiology and Neurobiology, Eötvös Loránd University, H-1117 Budapest, Hungary
| | - Sándor Borbély
- Department of Physiology and Neurobiology, Eötvös Loránd University, H-1117 Budapest, Hungary
| | - Márton Gulyás
- MTA-ELTE-NAP B Neuronal Cell Biology Research Group, H-1117 Budapest, Hungary
| | - Anikó Rátkai
- Department of Physiology and Neurobiology, Eötvös Loránd University, H-1117 Budapest, Hungary
| | - Attila Szűcs
- MTA-ELTE-NAP B Neuronal Cell Biology Research Group, H-1117 Budapest, Hungary
| | - Diána Hazai-Novák
- Department of Anatomy and Histology, Faculty of Veterinary Science, Szent István University, H-1400 Budapest, Hungary
| | - Kornelia Ellwanger
- Institute of Cell Biology and Immunology, University of Stuttgart, D-70569 Stuttgart, Germany
| | - Bence Rácz
- Department of Anatomy and Histology, Faculty of Veterinary Science, Szent István University, H-1400 Budapest, Hungary
| | - Klaus Pfizenmaier
- Institute of Cell Biology and Immunology, University of Stuttgart, D-70569 Stuttgart, Germany
| | - Angelika Hausser
- Institute of Cell Biology and Immunology, University of Stuttgart, D-70569 Stuttgart, Germany
| | - Katalin Schlett
- Department of Physiology and Neurobiology, Eötvös Loránd University, H-1117 Budapest, Hungary MTA-ELTE-NAP B Neuronal Cell Biology Research Group, H-1117 Budapest, Hungary
| |
Collapse
|
42
|
Burnstock G. An introduction to the roles of purinergic signalling in neurodegeneration, neuroprotection and neuroregeneration. Neuropharmacology 2015; 104:4-17. [PMID: 26056033 DOI: 10.1016/j.neuropharm.2015.05.031] [Citation(s) in RCA: 155] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2015] [Revised: 05/19/2015] [Accepted: 05/20/2015] [Indexed: 12/21/2022]
Abstract
Purinergic signalling appears to play important roles in neurodegeneration, neuroprotection and neuroregeneration. Initially there is a brief summary of the background of purinergic signalling, including release of purines and pyrimidines from neural and non-neural cells and their ectoenzymatic degradation, and the current characterisation of P1 (adenosine), and P2X (ion channel) and P2Y (G protein-coupled) nucleotide receptor subtypes. There is also coverage of the localization and roles of purinoceptors in the healthy central nervous system. The focus is then on the roles of purinergic signalling in trauma, ischaemia, stroke and in neurodegenerative diseases, including Alzheimer's, Parkinson's and Huntington's diseases, as well as multiple sclerosis and amyotrophic lateral sclerosis. Neuroprotective mechanisms involving purinergic signalling are considered and its involvement in neuroregeneration, including the role of adult neural stem/progenitor cells. This article is part of the Special Issue entitled 'Purines in Neurodegeneration and Neuroregeneration'.
Collapse
Affiliation(s)
- Geoffrey Burnstock
- Autonomic Neuroscience Centre, University College Medical School, Rowland Hill Street, London NW3 2PF, UK; Department of Pharmacology and Therapeutics, The University of Melbourne, Australia.
| |
Collapse
|
43
|
Gan L, Qian M, Shi K, Chen G, Gu Y, Du W, Zhu G. Restorative effect and mechanism of mecobalamin on sciatic nerve crush injury in mice. Neural Regen Res 2015; 9:1979-84. [PMID: 25598780 PMCID: PMC4283280 DOI: 10.4103/1673-5374.145379] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2014] [Indexed: 12/21/2022] Open
Abstract
Mecobalamin, a form of vitamin B12 containing a central metal element (cobalt), is one of the most important mediators of nervous system function. In the clinic, it is often used to accelerate recovery of peripheral nerves, but its molecular mechanism remains unclear. In the present study, we performed sciatic nerve crush injury in mice, followed by daily intraperitoneal administration of mecobalamin (65 μg/kg or 130 μg/kg) or saline (negative control). Walking track analysis, histomorphological examination, and quantitative real-time PCR showed that mecobalamin significantly improved functional recovery of the sciatic nerve, thickened the myelin sheath in myelinated nerve fibers, and increased the cross-sectional area of target muscle cells. Furthermore, mecobalamin upregulated mRNA expression of growth associated protein 43 in nerve tissue ipsilateral to the injury, and of neurotrophic factors (nerve growth factor, brain-derived nerve growth factor and ciliary neurotrophic factor) in the L4–6 dorsal root ganglia. Our findings indicate that the molecular mechanism underlying the therapeutic effect of mecobalamin after sciatic nerve injury involves the upregulation of multiple neurotrophic factor genes.
Collapse
Affiliation(s)
- Lin Gan
- Department of Orthopedics, Wuxi No. 2 People's Hospital, Wuxi, Jiangsu Province, China
| | - Minquan Qian
- Department of Orthopedics, Wuxi No. 2 People's Hospital, Wuxi, Jiangsu Province, China
| | - Keqin Shi
- Department of Orthopedics, Wuxi No. 2 People's Hospital, Wuxi, Jiangsu Province, China
| | - Gang Chen
- Department of Orthopedics, Wuxi No. 2 People's Hospital, Wuxi, Jiangsu Province, China
| | - Yanglin Gu
- Department of Orthopedics, Wuxi No. 2 People's Hospital, Wuxi, Jiangsu Province, China
| | - Wei Du
- Department of Orthopedics, Wuxi No. 2 People's Hospital, Wuxi, Jiangsu Province, China
| | - Guoxing Zhu
- Department of Orthopedics, Wuxi No. 2 People's Hospital, Wuxi, Jiangsu Province, China
| |
Collapse
|
44
|
Darabid H, Perez-Gonzalez AP, Robitaille R. Neuromuscular synaptogenesis: coordinating partners with multiple functions. Nat Rev Neurosci 2014; 15:630-1. [DOI: 10.1038/nrn3821] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
45
|
Li G, Zhao X, Zhao W, Zhang L, Wang C, Jiang M, Gu X, Yang Y. Porous chitosan scaffolds with surface micropatterning and inner porosity and their effects on Schwann cells. Biomaterials 2014; 35:8503-13. [DOI: 10.1016/j.biomaterials.2014.05.093] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Accepted: 05/29/2014] [Indexed: 10/25/2022]
|
46
|
Zhang Y, Zhang H, Katiella K, Huang W. Chemically extracted acellular allogeneic nerve graft combined with ciliary neurotrophic factor promotes sciatic nerve repair. Neural Regen Res 2014; 9:1358-64. [PMID: 25221592 PMCID: PMC4160866 DOI: 10.4103/1673-5374.137588] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/28/2014] [Indexed: 12/01/2022] Open
Abstract
A chemically extracted acellular allogeneic nerve graft can reduce postoperative immune rejection, similar to an autologous nerve graft, and can guide neural regeneration. However, it remains poorly understood whether a chemically extracted acellular allogeneic nerve graft combined with neurotrophic factors provides a good local environment for neural regeneration. This study investigated the repair of injured rat sciatic nerve using a chemically extracted acellular allogeneic nerve graft combined with ciliary neurotrophic factor. An autologous nerve anastomosis group and a chemical acellular allogeneic nerve bridging group were prepared as controls. At 8 weeks after repair, sciatic functional index, evoked potential amplitude of the soleus muscle, triceps wet weight recovery rate, total number of myelinated nerve fibers and myelin sheath thickness were measured. For these indices, values in the three groups showed the autologous nerve anastomosis group > chemically extracted acellular nerve graft + ciliary neurotrophic factor group > chemical acellular allogeneic nerve bridging group. These results suggest that chemically extracted acellular nerve grafts combined with ciliary neurotrophic factor can repair sciatic nerve defects, and that this repair is inferior to autologous nerve anastomosis, but superior to chemically extracted acellular allogeneic nerve bridging alone.
Collapse
Affiliation(s)
- Yanru Zhang
- Institute of International Education, Zhengzhou University, Zhengzhou, Henan Province, China ; Institute of Clinical Anatomy, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Hui Zhang
- Department of Orthopedics, First Affiliated Hospital, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Kaka Katiella
- Institute of Clinical Medicine, Zhengzhou University, Zhengzhou, Henan Province, China
| | - Wenhua Huang
- Institute of Clinical Anatomy, Southern Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
47
|
Keilhoff G, Lucas B, Pinkernelle J, Steiner M, Fansa H. Effects of cerebrolysin on motor-neuron-like NSC-34 cells. Exp Cell Res 2014; 327:234-55. [PMID: 24997385 DOI: 10.1016/j.yexcr.2014.06.020] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 06/12/2014] [Accepted: 06/26/2014] [Indexed: 01/01/2023]
Abstract
Although the peripheral nervous system is capable of regeneration, this capability is limited. As a potential means of augmenting nerve regeneration, the effects of cerebrolysin (CL)--a proteolytic peptide fraction--were tested in vitro on the motor-neuron-like NSC-34 cell line and organotypic spinal cord cultures. Therefore, NSC-34 cells were subjected to mechanical stress by changing media and metabolic stress by oxygen glucose deprivation. Afterwards, cell survival/proliferation using MTT and BrdU-labeling (FACS) and neurite sprouting using ImageJ analysis were evaluated. Calpain-1, Src and α-spectrin protein expression were analyzed by Western blot. In organotypic cultures, the effect of CL on motor neuron survival and neurite sprouting was tested by immunohistochemistry. CL had a temporary anti-proliferative but initially neuroprotective effect on OGD-stressed NSC-34 cells. High-dosed or repeatedly applied CL was deleterious for cell survival. CL amplified neurite reconstruction to limited extent, affected calpain-1 protein expression and influenced calpain-mediated spectrin cleavage as a function of Src expression. In organotypic spinal cord slice cultures, CL was not able to support motor neuron survival/neurite sprouting. Moreover, it hampered astroglia and microglia activities. The data suggest that CL may have only isolated positive effects on injured spinal motor neurons. High-dosed or accumulated CL seemed to have adverse effects in treatment of spinal cord injury. Further experiments are required to optimize the conditions for a safe clinical administration of CL in spinal cord injuries.
Collapse
Affiliation(s)
- Gerburg Keilhoff
- Institute of Biochemistry and Cell Biology, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany.
| | - Benjamin Lucas
- Institute of Biochemistry and Cell Biology, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany
| | - Josephine Pinkernelle
- Institute of Biochemistry and Cell Biology, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany
| | - Michael Steiner
- Institute of Biochemistry and Cell Biology, Otto-von-Guericke University Magdeburg, Leipziger Str. 44, D-39120 Magdeburg, Germany
| | - Hisham Fansa
- Department of Plastic, Reconstructive and Aesthetic Surgery, Hand Surgery, Klinikum Bielefeld, Teutoburger Str. 50, D-33604 Bielefeld, Germany
| |
Collapse
|
48
|
Wang C, Wang H, Pang J, Li L, Zhang S, Song G, Li N, Cao J, Zhang L. Glial Cell-Derived Neurotrophic Factor Attenuates Neuropathic Pain in a Mouse Model of Chronic Constriction Injury: Possible Involvement of E-cadherin/p120ctn Signaling. J Mol Neurosci 2014; 54:156-63. [DOI: 10.1007/s12031-014-0266-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Accepted: 02/14/2014] [Indexed: 12/22/2022]
|
49
|
The use of fiber-reinforced scaffolds cocultured with Schwann cells and vascular endothelial cells to repair rabbit sciatic nerve defect with vascularization. BIOMED RESEARCH INTERNATIONAL 2013; 2013:362918. [PMID: 24490158 PMCID: PMC3893804 DOI: 10.1155/2013/362918] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Accepted: 12/09/2013] [Indexed: 11/17/2022]
Abstract
To explore the feasibility of biodegradable fiber-reinforced 3D scaffolds with satisfactory mechanical properties for the repair of long-distance sciatic nerve defect in rabbits and effects of vascularized graft in early stage on the recovery of neurological function, Schwann cells and vascular endothelial cells were cocultured in the fiber-reinforced 3D scaffolds. Experiment group which used prevascularized nerve complex for the repair of sciatic nerve defect and control group which only cultured with Schwann cells were set. The animals in both groups underwent electromyography to show the status of the neurological function recovery at 4, 8, and 16 weeks after the surgery. Sciatic nerve regeneration and myelination were observed under the light microscope and electron microscope. Myelin sheath thickness, axonal diameter, and number of myelinated nerve fiber were quantitatively analyzed using image analysis system. The recovery of foot ulcer, the velocity of nerve conduction, the number of regenerating nerve fiber, and the recovery of ultrastructure were increased in the experimental group than those in the control group. Prevascularized tissue engineered fiber-reinforced 3D scaffolds for the repair of sciatic nerve defects in rabbits can effectively promote the recovery of neurological function.
Collapse
|
50
|
Repair of the Peripheral Nerve-Remyelination that Works. Brain Sci 2013; 3:1182-97. [PMID: 24961524 PMCID: PMC4061866 DOI: 10.3390/brainsci3031182] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Revised: 07/07/2013] [Accepted: 07/19/2013] [Indexed: 12/15/2022] Open
Abstract
In this review we summarize the events known to occur after an injury in the peripheral nervous system. We have focused on the Schwann cells, as they are the most important cells for the repair process and facilitate axonal outgrowth. The environment created by this cell type is essential for the outcome of the repair process. The review starts with a description of the current state of knowledge about the initial events after injury, followed by Wallerian degeneration, and subsequent regeneration. The importance of surgical repair, carried out as soon as possible to increase the chances of a good outcome, is emphasized throughout the review. The review concludes by describing the target re-innervation, which today is one of the most serious problems for nerve regeneration. It is clear, compiling this data, that even though regeneration of the peripheral nervous system is possible, more research in this area is needed in order to perfect the outcome.
Collapse
|