1
|
Protzuk OA, Samuel MA, Seward KR, Keshishian CA, Bendale GS, Isaacs JE. Induced Pseudomembrane Enrichment in Long Nerve Allograft Reconstruction. Muscle Nerve 2025; 71:662-669. [PMID: 40052754 PMCID: PMC11887527 DOI: 10.1002/mus.28362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 03/10/2025]
Abstract
INTRODUCTION/AIMS Long nerve defects are typically reconstructed with autograft or processed acellular nerve allograft (PNA). PNA is convenient and avoids donor morbidity but lacks the neurotrophic environment of autograft. Increased levels of neurotrophic factors have been identified in pseudomembranes induced around silicone implanted between nerve ends. This study aimed to determine if pseudomembrane can be reliably induced around silicone implanted between nerve ends, and if this enhances regeneration of PNA inset within using a staged technique. METHODS Lewis rats (n = 24) underwent resection of a 15-mm sciatic nerve. The defect was filled with a silicone tube (n = 12) (MA) or the nerve ends were secured to a muscle bed (n = 12) (NMA). After 4 weeks, the silicone was replaced with PNA threaded within the pseudomembrane tunnel. In both groups, PNA was used to reconstruct the nerve defect. Weekly neuromotor assessment was performed with sciatic function index (SFI). At 16 weeks, muscle recovery was assessed, and nerve samples were obtained for histomorphometry. RESULTS The MA group's average normalized muscle weight was 46.25% versus the NMA group's 33.19% (p < 0.05). The MA group's average normalized muscle girth was 78.25% versus the NMA group's 60.73% (p < 0.05). Axon counts, g-ratio, and muscle force were not statistically different. At Week 15, the MA group had a significantly higher average SFI: -82.25 versus the NMA group -95.03 (p < 0.05). DISCUSSION PNA inset within induced pseudomembrane sheath enhanced muscle reinnervation. A staged membrane enhancement technique may be effective for improving PNA efficacy in peripheral nerve injury reconstruction.
Collapse
Affiliation(s)
- Omar A. Protzuk
- Department of Orthopaedic SurgeryHarvard Medical School, Mass General Brigham, Brigham & Women's HospitalBostonMassachusettsUSA
| | - Mariam A. Samuel
- Division of Hand Surgery, Department of Orthopaedic SurgeryVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Kriston R. Seward
- Division of Hand Surgery, Department of Orthopaedic SurgeryVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Christopher A. Keshishian
- Division of Hand Surgery, Department of Orthopaedic SurgeryVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Geetanjali S. Bendale
- Division of Hand Surgery, Department of Orthopaedic SurgeryVirginia Commonwealth UniversityRichmondVirginiaUSA
| | - Jonathan E. Isaacs
- Division of Hand Surgery, Department of Orthopaedic SurgeryVirginia Commonwealth UniversityRichmondVirginiaUSA
| |
Collapse
|
2
|
Fujimaki Y, Kondo K, Nishijima H, Kikuta S, Yamasoba T. Granulocyte colony-stimulating factor promotes regeneration of severed facial nerve in rats. Front Neurosci 2024; 18:1442614. [PMID: 39712221 PMCID: PMC11662712 DOI: 10.3389/fnins.2024.1442614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 11/04/2024] [Indexed: 12/24/2024] Open
Abstract
Background and aim The administration of growth and neurotrophic factors has been attempted experimentally as a new therapeutic strategy for severe facial paralysis. Granulocyte colony-stimulating factor (G-CSF) has an effect on the treatment of central nervous system injuries, such as cerebral infarction and spinal cord injury. This study aimed at examining the effects of G-CSF on facial nerve regeneration in rats. Methods The left facial nerve of rats was either partially resected (resection group) or severed and sutured (suture group) at the main trunk outside the temporal bone. In each surgical group, saline or G-CSF was administered via the gelatin hydrogel drug delivery system. The suture group was further divided into two subgroups for the late administration of G-CSF (2 weeks after surgical treatment) or immediate administration of G-CSF after surgical treatment. Recovery of the facial nerve was assessed by the evaluation of facial movements (after 12 weeks), complex muscle action potential amplitude measurements (after 2, 4, 8, and 12 weeks), electroneurography values (after 12 weeks), and histological evaluation (comparison of myelinated axon diameters among the groups). Results Recovery of the function and morphology of damaged nerves was faster in the suture groups than in the resection group. In the suture groups, recovery was faster for G-CSF-treated rats than for saline-treated rats. Furthermore, recovery was faster in the group that received G-CSF immediately after surgical treatment than in the group that received G-CSF 2 weeks later. However, the group that received G-CSF 2 weeks later also showed faster recovery than did the control group. Conclusion G-CSF effectively promoted nerve regeneration during facial nerve paralysis. Thus, G-CSF may be a potential treatment strategy for injured facial nerves as it has been safely administered in clinical treatments for hematological diseases.
Collapse
Affiliation(s)
- Yoko Fujimaki
- Department of Otorhinolaryngology and Head and Neck Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kenji Kondo
- Department of Otorhinolaryngology and Head and Neck Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hironobu Nishijima
- Department of Otorhinolaryngology and Head and Neck Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Shu Kikuta
- Department of Otolaryngology-Head and Neck Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Tatsuya Yamasoba
- Department of Otorhinolaryngology and Head and Neck Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
3
|
Sokolowski I, Kucharska-Lusina A, Miller E, Poplawski T, Majsterek I. Exploring the Gene Expression and Plasma Protein Levels of HSP90, HSP60, and GDNF in Multiple Sclerosis Patients and Healthy Controls. Curr Issues Mol Biol 2024; 46:11668-11680. [PMID: 39451573 PMCID: PMC11505768 DOI: 10.3390/cimb46100693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 10/12/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic neurodegenerative disease characterized by immune-mediated inflammation and neurodegeneration in the central nervous system (CNS). In this study; we aimed to investigate the gene expression and plasma protein levels of three neuroprotective genes-heat shock proteins (HSP90 and HSP60) and glial cell line-derived neurotrophic factor (GDNF)-in MS patients compared to healthy controls. Forty patients with relapsing-remitting MS and 40 healthy volunteers participated in this study. Gene expression was measured using reverse transcription quantitative real-time PCR, and protein levels were assessed via ELISA. The results showed a significant increase in HSP90 (1.7-fold) and HSP60 (2-fold) gene expression in MS patients compared to controls, along with corresponding increases in protein levels (1.5-fold for both HSP90 and HSP60). In contrast, GDNF gene expression and protein levels were significantly reduced in MS patients, with a 7-fold decrease in gene expression and a 1.6-fold reduction in protein levels. Notably, a non-linear relationship between GDNF gene expression and protein concentration was observed in MS patients, suggesting complex regulatory mechanisms influencing GDNF in the disease. The upregulation of HSP90 and HSP60 in MS highlights their roles in immune regulation and stress responses, while the reduction in GDNF indicates impaired neuroprotection. These findings suggest that HSP90, HSP60, and GDNF could serve as biomarkers for disease progression and as potential therapeutic targets in MS, offering promising avenues for future research and treatment development.
Collapse
Affiliation(s)
- Igor Sokolowski
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (I.S.); (A.K.-L.); (I.M.)
| | - Aleksandra Kucharska-Lusina
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (I.S.); (A.K.-L.); (I.M.)
| | - Elzbieta Miller
- Department of Neurological Rehabilitation, Medical University of Lodz, Milionowa 14, 93-113 Lodz, Poland;
| | - Tomasz Poplawski
- Department of Microbiology and Pharmaceutical Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland
| | - Ireneusz Majsterek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, Mazowiecka 5, 92-215 Lodz, Poland; (I.S.); (A.K.-L.); (I.M.)
| |
Collapse
|
4
|
Kakoty V, Sarathlal KC, Kaur P, Wadhwa P, Vishwas S, Khan FR, Alhazmi AYM, Almasoudi HH, Gupta G, Chellappan DK, Paudel KR, Kumar D, Dua K, Singh SK. Unraveling the role of glial cell line-derived neurotrophic factor in the treatment of Parkinson's disease. Neurol Sci 2024; 45:1409-1418. [PMID: 38082050 DOI: 10.1007/s10072-023-07253-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 12/02/2023] [Indexed: 03/16/2024]
Abstract
Parkinson's disease is the second most common neurodegenerative condition with its prevalence projected to 8.9 million individuals globally in the year 2019. Parkinson's disease affects both motor and certain non-motor functions of an individual. Numerous research has focused on the neuroprotective effect of the glial cell line-derived neurotrophic factor (GDNF) in Parkinson's disease. Discovered in 1993, GDNF is a neurotrophic factor identified from the glial cells which was found to have selective effects on promoting survival and regeneration of certain populations of neurons including the dopaminergic nigrostriatal pathway. Given this property, recent studies have focused on the exogenous administration of GDNF for relieving Parkinson's disease-related symptoms both at a pre-clinical and a clinical level. This review will focus on enumerating the molecular connection between Parkinson's disease and GDNF and shed light on all the available drug delivery approaches to facilitate the selective delivery of GDNF into the brain paving the way as a potential therapeutic candidate for Parkinson's disease in the future.
Collapse
Affiliation(s)
- Violina Kakoty
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India
| | - K C Sarathlal
- Department of Non-Communicable Disease, Translational Health Science and Technology Institute, Faridabad, India
| | - Palwinder Kaur
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India
| | - Pankaj Wadhwa
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India
| | - Sukriti Vishwas
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India
| | - Farhan R Khan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Al-Quwayiyah, Shaqra University, Riyadh, Saudi Arabia
| | | | - Hassan Hussain Almasoudi
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Najran University, Najran, 61441, Saudi Arabia
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- School of Pharmacy, Graphic Era Hill University, Dehradun, 248007, India
- School of Pharmacy, Suresh Gyan Vihar University, Jagatpura, Mahal Road, Jaipur, India
| | | | - Keshav Raj Paudel
- Centre for Inflammation, Faculty of Science, School of Life Sciences, Centenary Institute and University of Technology Sydney, Sydney, NSW, 2050, Australia
| | - Dileep Kumar
- Department of Entomology and Nematology, UC Davis Comprehensive Cancer Center, University of California Davis, One Shields Avenue, Davis, CA, 95616, USA
| | - Kamal Dua
- School of Health, University of Technology Sydney, Ultimo, NSW, 2007, Australia
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Sachin Kumar Singh
- School of Pharmaceutical Sciences, Lovely Professional University, Jalandhar-Delhi G.T Road, Phagwara, Punjab, India.
- Faculty of Health, Australian Research Centre in Complementary and Integrative Medicine, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| |
Collapse
|
5
|
Ishijima T, Nakajima K. Restoration of injured motoneurons reduces microglial proliferation in the adult rat facial nucleus. J Neuropathol Exp Neurol 2024; 83:168-180. [PMID: 38263471 DOI: 10.1093/jnen/nlad116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2024] Open
Abstract
In the axotomized facial nucleus (axotFN), the levels of choline acetyltransferase, vesicular acetylcholine transporter, and gamma amino butyric acid A receptor α1 are decreased, after which the microglia begin to proliferate around injured motoneuron cell bodies. We conjectured that an injury signal released from the injured motoneurons triggers the microglial proliferation in the axotFN. However, it is unclear whether the level of microglial proliferation is dependent on the degree of motoneuronal insult. In this study, we investigated the relationship between the extents of motoneuronal injury and microglial proliferation in a rat axotFN model. Administration of glial cell line-derived neurotrophic factor, N-acetyl L-cysteine, or salubrinal at the transection site ameliorated the increase in c-Jun and the reductions in levels of phosphorylated cAMP response element binding protein (p-CREB) and functional molecules in the injured motoneurons. Concurrently, the levels of the microglial marker ionized calcium-binding adapter molecule 1 and of macrophage colony-stimulating factor (cFms), proliferating cell nuclear antigen, and p-p38/p38 were significantly downregulated in microglia. These results demonstrate that the recovery of motoneuron function resulted in the reduction in microglial proliferation. We conclude that the degree of neuronal injury regulates the levels of microglial proliferation in the axotFN.
Collapse
Affiliation(s)
- Takashi Ishijima
- Graduate School of Science and Engineering, Soka University, Tokyo, Japan
| | - Kazuyuki Nakajima
- Graduate School of Science and Engineering, Soka University, Tokyo, Japan
- Glycan & Life Systems Integration Center, Soka University, Tokyo, Japan
| |
Collapse
|
6
|
Harry GJ. Microglia Colonization Associated with Angiogenesis and Neural Cell Development. ADVANCES IN NEUROBIOLOGY 2024; 37:163-178. [PMID: 39207692 DOI: 10.1007/978-3-031-55529-9_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
The temporal and spatial pattern of microglia colonization of the nervous system implies a role in early stages of organ development including cell proliferation, differentiation, and neurovascularization. As microglia colonize and establish within the developing nervous system, they assume a neural-specific identity and contribute to key developmental events. Their association around blood vessels implicates them in development of the vascular system or vice versa. A similar association has been reported for neural cell proliferation and associated phenotypic shifts and for cell fate differentiation to neuronal or glial phenotypes. These processes are accomplished by phagocytic activities, cell-cell contact relationships, and secretion of various factors. This chapter will present data currently available from studies evaluating the dynamic and interactive nature of these processes throughout the progression of nervous system development.
Collapse
Affiliation(s)
- G Jean Harry
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute Environmental Health Sciences, Research Triangle Park, NC, USA.
| |
Collapse
|
7
|
Bengur FB, Komatsu C, Fedor CN, Loder S, Baker JS, Totwani A, Irgebay Z, Nerone WV, Solari MG, Marra KG. Biodegradable Nerve Guide with Glial Cell Line-Derived Neurotrophic Factor Improves Recovery After Facial Nerve Injury in Rats. Facial Plast Surg Aesthet Med 2023; 25:478-486. [PMID: 36877591 PMCID: PMC10664574 DOI: 10.1089/fpsam.2022.0346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/07/2023] Open
Abstract
Background: Bioengineered nerve guides with glial cell line-derived neurotrophic factor (GDNF) support recovery after facial nerve injury by acting as regenerative scaffolds. Objective: To compare functional, electrophysiological, and histological outcomes after repair of rat facial nerve transection in control, empty nerve guide, and nerve guide with GDNF conditions. Methods: Rats underwent transection and primary repair of the buccal branch of the facial nerve and were divided into (1) transection and repair only, (2) transection and repair augmented with empty guide, (3) transection and repair augmented with GDNF-guide groups. Weekly measurements of the whisking movements were recorded. At 12 weeks, compound muscle action potentials (CMAPs) at the whisker pad were assessed, and samples were collected for histomorphometric analysis. Results: Rats in GDNF-guide group displayed the earliest peak in normalized whisking amplitude. CMAPs were significantly higher after GDNF-guide placement. Mean fiber surface area of the target muscle, axonal count of the injured branch, and the number of Schwann cells were highest with GDNF guides. Conclusion: The biodegradable nerve guide containing double-walled GDNF microspheres enhanced recovery after facial nerve transection and primary repair.
Collapse
Affiliation(s)
- Fuat Baris Bengur
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Chiaki Komatsu
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Caroline Nadia Fedor
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Shawn Loder
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jocelyn S. Baker
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Aanchal Totwani
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Zhazira Irgebay
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - W. Vincent Nerone
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Mario G. Solari
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Kacey G. Marra
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
8
|
Zochodne DW. Growth factors and molecular-driven plasticity in neurological systems. HANDBOOK OF CLINICAL NEUROLOGY 2023; 196:569-598. [PMID: 37620091 DOI: 10.1016/b978-0-323-98817-9.00017-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/26/2023]
Abstract
It has been almost 70 years since the discovery of nerve growth factor (NGF), a period of a dramatic evolution in our understanding of dynamic growth, regeneration, and rewiring of the nervous system. In 1953, the extraordinary finding that a protein found in mouse submandibular glands generated a halo of outgrowing axons has now redefined our concept of the nervous system connectome. Central and peripheral neurons and their axons or dendrites are no longer considered fixed or static "wiring." Exploiting this molecular-driven plasticity as a therapeutic approach has arrived in the clinic with a slate of new trials and ideas. Neural growth factors (GFs), soluble proteins that alter the behavior of neurons, have expanded in numbers and our understanding of the complexity of their signaling and interactions with other proteins has intensified. However, beyond these "extrinsic" determinants of neuron growth and function are the downstream pathways that impact neurons, ripe for translational development and potentially more important than individual growth factors that may trigger them. Persistent and ongoing nuances in clinical trial design in some of the most intractable and irreversible neurological conditions give hope for connecting new biological ideas with clinical benefits. This review is a targeted update on neural GFs, their signals, and new therapeutic ideas, selected from an expansive literature.
Collapse
Affiliation(s)
- Douglas W Zochodne
- Division of Neurology, Department of Medicine and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
9
|
Neuropathology of the Basal Ganglia in SNCA Transgenic Rat Model of Parkinson's Disease: Involvement of Parvalbuminergic Interneurons and Glial-Derived Neurotropic Factor. Int J Mol Sci 2022; 23:ijms231710126. [PMID: 36077524 PMCID: PMC9456397 DOI: 10.3390/ijms231710126] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 08/29/2022] [Accepted: 08/30/2022] [Indexed: 11/19/2022] Open
Abstract
Parkinson’s disease (PD) is a neurodegenerative disease characterized by the accumulation of alpha-synuclein, encoded by the SNCA gene. The main neuropathological hallmark of PD is the degeneration of dopaminergic neurons leading to striatal dopamine depletion. Trophic support by a neurotrophin called glial-derived neurotrophic factor (GDNF) is also lacking in PD. We performed immunohistochemical studies to investigate neuropathological changes in the basal ganglia of a rat transgenic model of PD overexpressing alfa-synuclein. We observed that neuronal loss also occurs in the dorsolateral part of the striatum in the advanced stages of the disease. Moreover, along with the degeneration of the medium spiny projection neurons, we found a dramatic loss of parvalbumin interneurons. A marked decrease in GDNF, which is produced by parvalbumin interneurons, was observed in the striatum and in the substantia nigra of these animals. This confirmed the involvement of the striatum in the pathophysiology of PD and the importance of GDNF in maintaining the health of the substantia nigra.
Collapse
|
10
|
Pottorf TS, Rotterman TM, McCallum WM, Haley-Johnson ZA, Alvarez FJ. The Role of Microglia in Neuroinflammation of the Spinal Cord after Peripheral Nerve Injury. Cells 2022; 11:cells11132083. [PMID: 35805167 PMCID: PMC9265514 DOI: 10.3390/cells11132083] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 12/12/2022] Open
Abstract
Peripheral nerve injuries induce a pronounced immune reaction within the spinal cord, largely governed by microglia activation in both the dorsal and ventral horns. The mechanisms of activation and response of microglia are diverse depending on the location within the spinal cord, type, severity, and proximity of injury, as well as the age and species of the organism. Thanks to recent advancements in neuro-immune research techniques, such as single-cell transcriptomics, novel genetic mouse models, and live imaging, a vast amount of literature has come to light regarding the mechanisms of microglial activation and alluding to the function of microgliosis around injured motoneurons and sensory afferents. Herein, we provide a comparative analysis of the dorsal and ventral horns in relation to mechanisms of microglia activation (CSF1, DAP12, CCR2, Fractalkine signaling, Toll-like receptors, and purinergic signaling), and functionality in neuroprotection, degeneration, regeneration, synaptic plasticity, and spinal circuit reorganization following peripheral nerve injury. This review aims to shed new light on unsettled controversies regarding the diversity of spinal microglial-neuronal interactions following injury.
Collapse
Affiliation(s)
- Tana S. Pottorf
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA; (T.S.P.); (W.M.M.); (Z.A.H.-J.)
| | - Travis M. Rotterman
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA 30318, USA;
| | - William M. McCallum
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA; (T.S.P.); (W.M.M.); (Z.A.H.-J.)
| | - Zoë A. Haley-Johnson
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA; (T.S.P.); (W.M.M.); (Z.A.H.-J.)
| | - Francisco J. Alvarez
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA; (T.S.P.); (W.M.M.); (Z.A.H.-J.)
- Correspondence:
| |
Collapse
|
11
|
Events Occurring in the Axotomized Facial Nucleus. Cells 2022; 11:cells11132068. [PMID: 35805151 PMCID: PMC9266054 DOI: 10.3390/cells11132068] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/26/2022] [Accepted: 06/27/2022] [Indexed: 01/27/2023] Open
Abstract
Transection of the rat facial nerve leads to a variety of alterations not only in motoneurons, but also in glial cells and inhibitory neurons in the ipsilateral facial nucleus. In injured motoneurons, the levels of energy metabolism-related molecules are elevated, while those of neurofunction-related molecules are decreased. In tandem with these motoneuron changes, microglia are activated and start to proliferate around injured motoneurons, and astrocytes become activated for a long period without mitosis. Inhibitory GABAergic neurons reduce the levels of neurofunction-related molecules. These facts indicate that injured motoneurons somehow closely interact with glial cells and inhibitory neurons. At the same time, these events allow us to predict the occurrence of tissue remodeling in the axotomized facial nucleus. This review summarizes the events occurring in the axotomized facial nucleus and the cellular and molecular mechanisms associated with each event.
Collapse
|
12
|
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
|
13
|
Starks AO, Owen J, Isaacs J. Evaluation of the Induced Membrane for Neurotrophic Factors. J Hand Surg Am 2022; 47:130-136. [PMID: 34865951 DOI: 10.1016/j.jhsa.2021.08.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 05/09/2021] [Accepted: 08/24/2021] [Indexed: 02/02/2023]
Abstract
PURPOSE Despite gaining popularity as a bridge for small and moderate nerve gaps, an acellular nerve allograft (ANA) lacks many of the neurotrophic characteristics of a nerve autograft. Pseudomembranes induced to form around temporary skeletal spacers are rich in growth factors. Induced membranes may have beneficial neurotrophic factors which could support ANA. METHODS Twenty-two male Sprague-Dawley rats underwent resection of 2 cm of the sciatic nerve. A silicone rod was inset in the defect of 11 experimental rats, and marking sutures only were placed in the nerve stumps of the remaining 11 control rats. After allowing 4 weeks for tissue maturation, tissue samples harvested from the induced membrane (experimental group) and the tissue bed (control group) were analyzed using Luminex multiplex assay to quantify differences in detectable levels of the following neurotrophic factors: nerve growth factor, glial-derived nerve factor, vascular endothelial growth factor, and transforming growth factor ß (TGF-ß) 1, 2, and 3, interleukin-1ß, and monocyte chemoattractant protein 1. RESULTS No difference was detected between the control and experimental groups in levels of vascular endothelial growth factor. Higher levels of TGF-ß1, TGF-ß2, TGF-ß3, glial-derived nerve factor, nerve growth factor, monocyte chemoattractant protein 1, and interleukin-1ß were detected in the experimental group. CONCLUSIONS In the setting of peripheral nerve injury, an induced membrane has higher levels of several neurotrophic factors that may support nerve regeneration compared to wound bed cicatrix. CLINICAL RELEVANCE This investigation provides impetus for further study examining the utility of using a staged induced membrane technique in conjunction with delayed nerve grafting in reconstruction of some peripheral nerve defects.
Collapse
Affiliation(s)
- Alexandria O Starks
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA.
| | - John Owen
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA
| | - Jonathan Isaacs
- Department of Orthopaedic Surgery, Virginia Commonwealth University, Richmond, VA
| |
Collapse
|
14
|
Li X, Li K, Chen Y, Fang F. The Role of Hippo Signaling Pathway in the Development of the Nervous System. Dev Neurosci 2021; 43:263-270. [PMID: 34350875 DOI: 10.1159/000515633] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 02/26/2021] [Indexed: 11/19/2022] Open
Abstract
Hippo signaling pathway is a highly conserved and crucial signaling pathway that controls the size of tissues and organs by regulating the proliferation, differentiation, and apoptosis of cells. The nervous system is a complicated system that participates in information collection, integration, and procession. The balance of various aspects of the nervous system is vital for the normal regulation of physiological conditions of the body, like the population and distribution of nerve cells, nerve connections, and so on. Defects in these aspects may lead to cognitive, behavioral, and neurological dysfunction, resulting in various nervous system diseases. Recently, accumulating evidence proposes that Hippo pathway maintains numerous biological functions in the nervous system development, including modulating the proliferation and differentiation of nerve cells and promoting the development of synapse, corpus callosum, and cortex. In this review, we will summarize recent findings of Hippo pathway in the nervous system to improve our understanding on its function and to provide potential therapeutic strategies of nervous system diseases in the future.
Collapse
Affiliation(s)
- Xifan Li
- Department of Human Anatomy, School of Basic Medicine Sciences, Guilin Medical University, Guilin, China
| | - Kaixuan Li
- Department of Human Anatomy, School of Basic Medicine Sciences, Guilin Medical University, Guilin, China
| | - Yu Chen
- Department of Human Anatomy, School of Basic Medicine Sciences, Guilin Medical University, Guilin, China
| | - Fang Fang
- Department of Human Anatomy, School of Basic Medicine Sciences, Guilin Medical University, Guilin, China
| |
Collapse
|
15
|
Hung TH, Liu YC, Wu CH, Chen CC, Chao H, Yang FY, Chen SF. Antenatal low-intensity pulsed ultrasound reduces neurobehavioral deficits and brain injury following dexamethasone-induced intrauterine growth restriction. BRAIN PATHOLOGY (ZURICH, SWITZERLAND) 2021; 31:e12968. [PMID: 33960564 PMCID: PMC8549022 DOI: 10.1111/bpa.12968] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 04/01/2021] [Accepted: 04/06/2021] [Indexed: 12/01/2022]
Abstract
Intrauterine growth restriction (IUGR) is a leading cause of perinatal mortality and morbidity, and IUGR survivors are at increased risk of neurodevelopmental deficits. No effective interventions are currently available to improve the structure and function of the IUGR brain before birth. This study investigated the protective effects of low‐intensity pulsed ultrasound (LIPUS) on postnatal neurodevelopmental outcomes and brain injury using a rat model of IUGR induced by maternal exposure to dexamethasone (DEX). Pregnant rats were treated with DEX (200 μg/kg, s.c.) and LIPUS daily from gestational day (GD) 14 to 19. Behavioral assessments were performed on the IUGR offspring to examine neurological function. Neuropathology, levels of neurotrophic factors, and CaMKII‐Akt‐related molecules were assessed in the IUGR brain, and expression of glucose and amino acid transporters and neurotrophic factors were examined in the placenta. Maternal LIPUS treatment increased fetal weight, fetal liver weight, and placental weight following IUGR. LIPUS treatment also increased neuronal number and myelin protein expression in the IUGR brain, and attenuated neurodevelopmental deficits at postnatal day (PND) 18. However, the number of oligodendrocytes or microglia was not affected. These changes were associated with the upregulation of brain‐derived neurotrophic factor (BDNF) and placental growth factor (PlGF) protein expression, and enhancement of neuronal CaMKII and Akt activation in the IUGR brain at PND 1. Additionally, LIPUS treatment promoted glucose transporter (GLUT) 1 production and BDNF expression in the placenta, but had no effects on GLUT3 or amino acid transporter expression. Our findings suggest that antenatal LIPUS treatment may reduce IUGR‐induced brain injury via enhancing cerebral BDNF/CaMKII/Akt signaling. These data provide new evidence that LIPUS stimulation could be considered for antenatal neuroprotective therapy in IUGR.
Collapse
Affiliation(s)
- Tai-Ho Hung
- Department of Obstetrics and Gynecology, Taipei Chang Gung Memorial Hospital, Taipei, Taiwan.,Department of Obstetrics and Gynecology, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yu-Cheng Liu
- Department of Obstetrics and Gynecology, Taipei Chang Gung Memorial Hospital, Taipei, Taiwan.,Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chun-Hu Wu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Chien-Cheng Chen
- Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan.,Graduate Institute of Gerontology and Health Care Management, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Hsien Chao
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Feng-Yi Yang
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Szu-Fu Chen
- Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, Taipei, Taiwan.,Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
16
|
Besser RR, Bowles AC, Alassaf A, Carbonero D, Maciel R, Saporta M, Agarwal A. A Chemically Defined Common Medium for Culture of C2C12 Skeletal Muscle and Human Induced Pluripotent Stem Cell Derived Spinal Spheroids. Cell Mol Bioeng 2020; 13:605-619. [PMID: 33281990 PMCID: PMC7704992 DOI: 10.1007/s12195-020-00624-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 06/03/2020] [Indexed: 01/14/2023] Open
Abstract
INTRODUCTION Multicellular platforms and linked multi organ on chip devices are powerful tools for drug discovery, and basic mechanistic studies. Often, a critical constraint is defining a culture medium optimal for all cells present in the system. In this study, we focused on the key cells of the neuromuscular junction i.e., skeletal muscle and motor neurons. METHODS Formulation of a chemically defined medium for the co-culture of C2C12 skeletal muscle cells and human induced pluripotent stem cell (hiPSC) derived spinal spheroids (SpS) was optimized. C2C12 cells in 10 experimental media conditions and 2 topographies were evaluated over a 14-day maturation period to determine the ideal medium formulation for skeletal muscle tissue development. RESULTS During early maturation, overexpression of genes for myogenesis and myopathy was observed for several media conditions, corresponding to muscle delamination and death. Together, we identified 3 media formulations that allowed for more controlled differentiation, healthier muscle tissue, and long-term culture duration. This evidence was then used to select media formulations to culture SpS and subsequently assessed axonal growth. As axonal growth in SpS cultures was comparable in all selected media conditions, our data suggest that the neuronal basal medium with no added supplements is the ideal medium formulation for both cell types. CONCLUSIONS Optimization using both topographical cues and culture media formulations provides a comprehensive analyses of culture conditions that are vital to future applications for in vitro NMJ models.
Collapse
Affiliation(s)
- Rachel R. Besser
- Department of Biomedical Engineering, DJTMF Biomedical Nanotechnology Institute, University of Miami, 1251 Memorial Dr, MEA 203, Coral Gables, FL 33146 USA
| | - Annie C. Bowles
- Department of Biomedical Engineering, DJTMF Biomedical Nanotechnology Institute, University of Miami, 1251 Memorial Dr, MEA 203, Coral Gables, FL 33146 USA
| | - Ahmad Alassaf
- Department of Biomedical Engineering, DJTMF Biomedical Nanotechnology Institute, University of Miami, 1251 Memorial Dr, MEA 203, Coral Gables, FL 33146 USA
- Department of Medical Equipment Technology, College of Applied Medical Sciences, Majmaah University, Al-Majmaah, 11952 Saudi Arabia
| | - Daniel Carbonero
- Department of Biomedical Engineering, DJTMF Biomedical Nanotechnology Institute, University of Miami, 1251 Memorial Dr, MEA 203, Coral Gables, FL 33146 USA
| | - Renata Maciel
- Department of Neurology, University of Miami Miller School of Medicine, 1120 NW 14th St, Suite 1310, Miami, FL 33136 USA
| | - Mario Saporta
- Department of Neurology, University of Miami Miller School of Medicine, 1120 NW 14th St, Suite 1310, Miami, FL 33136 USA
| | - Ashutosh Agarwal
- Department of Biomedical Engineering, DJTMF Biomedical Nanotechnology Institute, University of Miami, 1251 Memorial Dr, MEA 203, Coral Gables, FL 33146 USA
| |
Collapse
|
17
|
Sénécal V, Barat C, Tremblay MJ. The delicate balance between neurotoxicity and neuroprotection in the context of HIV-1 infection. Glia 2020; 69:255-280. [PMID: 32910482 DOI: 10.1002/glia.23904] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 08/15/2020] [Accepted: 08/16/2020] [Indexed: 12/17/2022]
Abstract
Human immunodeficiency virus type-1 (HIV-1) causes a spectrum of neurological impairments, termed HIV-associated neurocognitive disorder (HAND), following the infiltration of infected cells into the brain. Even though the implementation of antiretroviral therapy reduced the systemic viral load, the prevalence of HAND remains unchanged and infected patients develop persisting neurological disturbances affecting their quality of life. As a result, HAND have gained importance in basic and clinical researches, warranting the need of developing new adjunctive treatments. Nonetheless, a better understanding of the molecular and cellular mechanisms remains necessary. Several studies consolidated their efforts into elucidating the neurotoxic signaling leading to HAND including the deleterious actions of HIV-1 viral proteins and inflammatory mediators. However, the scope of these studies is not sufficient to address all the complexity related to HAND development. Fewer studies focused on an altered neuroprotective capacity of the brain to respond to HIV-1 infection. Neurotrophic factors are endogenous polyproteins involved in neuronal survival, synaptic plasticity, and neurogenesis. Any defects in the processing or production of these crucial factors might compose a risk factor rendering the brain more vulnerable to neuronal damages. Due to their essential roles, they have been investigated for their diverse interplays with HIV-1 infection. In this review, we present a complete description of the neurotrophic factors involved in HAND. We discuss emerging concepts for their therapeutic applications and summarize the complex mechanisms that down-regulate their production in favor of a neurotoxic environment. For certain factors, we finally address opposing roles that rather lead to increased inflammation.
Collapse
Affiliation(s)
- Vincent Sénécal
- Axe des Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Pavillon CHUL, Québec, Quebec, Canada
| | - Corinne Barat
- Axe des Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Pavillon CHUL, Québec, Quebec, Canada
| | - Michel J Tremblay
- Axe des Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Pavillon CHUL, Québec, Quebec, Canada.,Département de Microbiologie-infectiologie et immunologie, Faculté de Médecine, Université Laval, Québec, Quebec, Canada
| |
Collapse
|
18
|
GDNF synthesis, signaling, and retrograde transport in motor neurons. Cell Tissue Res 2020; 382:47-56. [PMID: 32897420 PMCID: PMC7529617 DOI: 10.1007/s00441-020-03287-6] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 08/18/2020] [Indexed: 02/06/2023]
Abstract
Glial cell line–derived neurotrophic factor (GDNF) is a 134 amino acid protein belonging in the GDNF family ligands (GFLs). GDNF was originally isolated from rat glial cell lines and identified as a neurotrophic factor with the ability to promote dopamine uptake within midbrain dopaminergic neurons. Since its discovery, the potential neuroprotective effects of GDNF have been researched extensively, and the effect of GDNF on motor neurons will be discussed herein. Similar to other members of the TGF-β superfamily, GDNF is first synthesized as a precursor protein (pro-GDNF). After a series of protein cleavage and processing, the 211 amino acid pro-GDNF is finally converted into the active and mature form of GDNF. GDNF has the ability to trigger receptor tyrosine kinase RET phosphorylation, whose downstream effects have been found to promote neuronal health and survival. The binding of GDNF to its receptors triggers several intracellular signaling pathways which play roles in promoting the development, survival, and maintenance of neuron-neuron and neuron-target tissue interactions. The synthesis and regulation of GDNF have been shown to be altered in many diseases, aging, exercise, and addiction. The neuroprotective effects of GDNF may be used to develop treatments and therapies to ameliorate neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS). In this review, we provide a detailed discussion of the general roles of GDNF and its production, delivery, secretion, and neuroprotective effects on motor neurons within the mammalian neuromuscular system.
Collapse
|
19
|
Eye alignment changes caused by sustained GDNF treatment of an extraocular muscle in infant non-human primates. Sci Rep 2020; 10:11927. [PMID: 32681083 PMCID: PMC7368047 DOI: 10.1038/s41598-020-68743-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 06/04/2020] [Indexed: 12/16/2022] Open
Abstract
The ability of sustained treatment of a single extraocular muscle with glial cell line-derived neurotrophic factor (GDNF) to produce a strabismus in infant non-human primates was tested. Six infant non-human primates received a pellet containing GDNF, releasing 2 µg/day for 90 days, on one medial rectus muscle. Eye alignment was assessed up to 6 months. Five of the six animals showed a slow decrease in eye misalignment from the significant exotropia present at birth, ending with approximately 10° of exotropia. Controls became orthotropic. Misalignment averaged 8° three months after treatment ended. After sustained GDNF treatment, few changes were seen in mean myofiber cross-sectional areas compared to age-matched naïve controls. Neuromuscular junction number was unaltered in the medial rectus muscles, but were significantly reduced in the untreated lateral recti. Neuromuscular junctions on slow fibers became multiply innervated after this sustained GDNF treatment. Pitx2-positive cells significantly decreased in treated and contralateral medial rectus muscles. Our study suggests that balanced GDNF signaling plays a role in normal development and maintenance of orthotropia. Sustained GDNF treatment of one medial rectus muscle resulted in a measurable misalignment largely maintained 3 months after treatment ended. Structural changes suggest mechanisms for producing an imbalance in muscle function.
Collapse
|
20
|
Zhao X, Li B, Guan X, Sun G, Zhang M, Zhang W, Xu J, Ren X. Peg-Enhanced Behavioral Recovery After Sciatic Nerve Transection and Either Suturing Or Sleeve Conduit Deployment in Rats. J INVEST SURG 2019; 34:524-533. [PMID: 31438740 DOI: 10.1080/08941939.2019.1654047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Polyethylene glycol (PEG) has previously been reported to improve outcomes of peripheral nerve microsuturing. However, recent studies have challenged this finding. Given its clinical importance, we investigated the potential of PEG as a facilitator of peripheral nerve restoration. The sciatic nerve of 144 rats was transected and submitted either to simple suturing (Group A), PEG-enhanced suturing (Group B), and insertion in an arterial sleeve conduit without PEG (Group C), or with PEG (Group D) in equal numbers. Behavioral recovery was assessed with the sciatic function index (SFI). Nerve impulse conduction was assessed with compound muscle action potentials (CMAPs). Histology comprised standard hematoxylin/eosin staining, electron microscopy and glial cell line-derived neurotrophic factor (GDNF) immunohistochemistry. Expression of GDNF was also assessed with western blotting. Results were evaluated at weeks 1, 4, and 8. PEG treatment significantly improved behavioral recovery and morphology of nerve restoration, particularly in the sleeve conduit group, relative to that of controls. In conclusion, PEG may improve outcomes of peripheral nerve reconstruction.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Bo Li
- Department of Ultrasound, Harbin Medical University Cancer Hospital, Harbin, China
| | - Xiangchen Guan
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guiyin Sun
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Mingzhe Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Weihua Zhang
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Junfeng Xu
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xiaoping Ren
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
21
|
Duraikannu A, Krishnan A, Chandrasekhar A, Zochodne DW. Beyond Trophic Factors: Exploiting the Intrinsic Regenerative Properties of Adult Neurons. Front Cell Neurosci 2019; 13:128. [PMID: 31024258 PMCID: PMC6460947 DOI: 10.3389/fncel.2019.00128] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/14/2019] [Indexed: 01/19/2023] Open
Abstract
Injuries and diseases of the peripheral nervous system (PNS) are common but frequently irreversible. It is often but mistakenly assumed that peripheral neuron regeneration is robust without a need to be improved or supported. However, axonal lesions, especially those involving proximal nerves rarely recover fully and injuries generally are complicated by slow and incomplete regeneration. Strategies to enhance the intrinsic growth properties of reluctant adult neurons offer an alternative approach to consider during regeneration. Since axons rarely regrow without an intimately partnered Schwann cell (SC), approaches to enhance SC plasticity carry along benefits to their axon partners. Direct targeting of molecules that inhibit growth cone plasticity can inform important regenerative strategies. A newer approach, a focus of our laboratory, exploits tumor suppressor molecules that normally dampen unconstrained growth. However several are also prominently expressed in stable adult neurons. During regeneration their ongoing expression “brakes” growth, whereas their inhibition and knockdown may enhance regrowth. Examples have included phosphatase and tensin homolog deleted on chromosome ten (PTEN), a tumor suppressor that inhibits PI3K/pAkt signaling, Rb1, the protein involved in retinoblastoma development, and adenomatous polyposis coli (APC), a tumor suppressor that inhibits β-Catenin transcriptional signaling and its translocation to the nucleus. The identification of several new targets to manipulate the plasticity of regenerating adult peripheral neurons is exciting. How they fit with canonical regeneration strategies and their feasibility require additional work. Newer forms of nonviral siRNA delivery may be approaches for molecular manipulation to improve regeneration.
Collapse
Affiliation(s)
- Arul Duraikannu
- Division of Neurology, Department of Medicine, and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Anand Krishnan
- Division of Neurology, Department of Medicine, and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Ambika Chandrasekhar
- Division of Neurology, Department of Medicine, and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Douglas W Zochodne
- Division of Neurology, Department of Medicine, and Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
22
|
Guy R, Grynspan F, Ben-Zur T, Panski A, Lamdan R, Danon U, Yaffe D, Offen D. Human Muscle Progenitor Cells Overexpressing Neurotrophic Factors Improve Neuronal Regeneration in a Sciatic Nerve Injury Mouse Model. Front Neurosci 2019; 13:151. [PMID: 30872995 PMCID: PMC6400854 DOI: 10.3389/fnins.2019.00151] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 02/11/2019] [Indexed: 01/21/2023] Open
Abstract
The peripheral nervous system has an intrinsic ability to regenerate after injury. However, this process is slow, incomplete, and often accompanied by disturbing motor and sensory consequences. Sciatic nerve injury (SNI), which is the most common model for studying peripheral nerve injury, is characterized by damage to both motor and sensory fibers. The main goal of this study is to examine the feasibility of administration of human muscle progenitor cells (hMPCs) overexpressing neurotrophic factor (NTF) genes, known to protect peripheral neurons and enhance axon regeneration and functional recovery, to ameliorate motoric and sensory deficits in SNI mouse model. To this end, hMPCs were isolated from a human muscle biopsy, and manipulated to ectopically express brain-derived neurotrophic factor (BDNF), glial-cell-line-derived neurotrophic factor (GDNF), vascular endothelial growth factor (VEGF), and insulin-like growth factor (IGF-1). These hMPC-NTF were transplanted into the gastrocnemius muscle of mice after SNI, and motor and sensory functions of the mice were assessed using the CatWalk XT system and the hot plate test. ELISA analysis showed that genetically manipulated hMPC-NTF express significant amounts of BDNF, GDNF, VEGF, or IGF-1. Transplantation of 3 × 106 hMPC-NTF was shown to improve motor function and gait pattern in mice following SNI surgery, as indicated by the CatWalk XT system 7 days post-surgery. Moreover, using the hot-plate test, performed 6 days after surgery, the treated mice showed less sensory deficits, indicating a palliative effect of the treatment. ELISA analysis following transplantation demonstrated increased NTF expression levels in the gastrocnemius muscle of the treated mice, reinforcing the hypothesis that the observed positive effect was due to the transplantation of the genetically manipulated hMPC-NTF. These results show that genetically modified hMPC can alleviate both motoric and sensory deficits of SNI. The use of hMPC-NTF demonstrates the feasibility of a treatment paradigm, which may lead to rapid, high-quality healing of damaged peripheral nerves due to administration of hMPC. Our approach suggests a possible clinical application for the treatment of peripheral nerve injury.
Collapse
Affiliation(s)
- Reut Guy
- Laboratory of Neuroscience, Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | - Tali Ben-Zur
- Laboratory of Neuroscience, Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Avraham Panski
- Department of Orthopedic Surgery, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ron Lamdan
- Department of Orthopedic Surgery, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Uri Danon
- Stem Cell Medicine Ltd., Jerusalem, Israel
| | - David Yaffe
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Daniel Offen
- Laboratory of Neuroscience, Felsenstein Medical Research Center, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
23
|
Downregulation of MicroRNA-33-5p Protected Bupivacaine-Induced Apoptosis in Murine Dorsal Root Ganglion Neurons Through GDNF. Neurotox Res 2019; 35:860-866. [PMID: 30617464 DOI: 10.1007/s12640-018-9994-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Revised: 12/21/2018] [Accepted: 12/26/2018] [Indexed: 12/25/2022]
Abstract
In this work, we evaluated the functional role of microRNA-33-5p (miR-33-5p) in regulating bupivacaine (Bv)-induced neural apoptosis in dorsal root ganglion (DRG) cells. DRG was extracted from adult mice and treated with BV in vitro. A TUNEL assay was applied to assess neural apoptosis among DRG cells. A qRT-PCR assay was applied to assess miR-33-5p expression among BV-treated DRG cells. MiR-33-5p was genetically knocked down in DRG cells. Its effect on BV-induced neural apoptosis was further evaluated by TUNEL assay. Correlation between miR-33-5p and its putative downstream target gene, glial cell-derived neurotrophic factor (GDNF), was assessed by dual-luciferase activity and qRT-PCR assays, respectively. GDNF was then inhibited in miR-33-5p-downregulated DRG cells to further assess its functional regulation in BV-induced neural apoptosis. BV induced significant neural apoptosis, in a dose-dependent manner, in DRG cells in vitro. MiR-33-5p was upregulated by BV treatment, also in a dose-dependent manner in DRG cells. On the other hand, downregulation of miR-33-5p protected BV-induced DRG neural apoptosis. GDNF was shown to be inversely correlated with miR-33-5p in BV-treated DRG cells. Moreover, inhibiting GDNF was able to reverse the protection of miR-33-5p-downregulation on BV-induced DRG neural apoptosis. MiR-33-5p, through its inverse regulation on DGNF gene, modulates anesthesia-induced neural apoptosis in DRG cells.
Collapse
|
24
|
Theme 3 In vivo experimental models. Amyotroph Lateral Scler Frontotemporal Degener 2018; 19:130-153. [DOI: 10.1080/21678421.2018.1510570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
25
|
Acosta L, Morcuende S, Silva-Hucha S, Pastor AM, de la Cruz RR. Vascular Endothelial Growth Factor (VEGF) Prevents the Downregulation of the Cholinergic Phenotype in Axotomized Motoneurons of the Adult Rat. Front Mol Neurosci 2018; 11:241. [PMID: 30050409 PMCID: PMC6052088 DOI: 10.3389/fnmol.2018.00241] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 06/22/2018] [Indexed: 12/11/2022] Open
Abstract
Vascular endothelial growth factor (VEGF) was initially characterized by its activity on the vascular system. However, there is growing evidence indicating that VEGF also acts as a neuroprotective factor, and that its administration to neurons suffering from trauma or disease is able to rescue them from cell death. We questioned whether VEGF could also maintain damaged neurons in a neurotransmissive mode by evaluating the synthesis of their neurotransmitter, and whether its action would be direct or through its well-known angiogenic activity. Adult rat extraocular motoneurons were chosen as the experimental model. Lesion was performed by monocular enucleation and immediately a gelatine sponge soaked in VEGF was implanted intraorbitally. After 7 days, abducens, trochlear, and oculomotor nuclei were examined by immunohistochemistry against choline acetyltransferase (ChAT), the biosynthetic enzyme of the motoneuronal neurotransmitter acetylcholine. Lesioned motoneurons exhibited a noticeable ChAT downregulation which was prevented by VEGF administration. To explore whether this action was mediated via an increase in blood vessels or in their permeability, we performed immunohistochemistry against laminin, glucose transporter-1 and the plasmatic protein albumin. The quantification of the immunolabeling intensity against these three proteins showed no significant differences between VEGF-treated, axotomized and control animals. Therefore, the present data indicate that VEGF is able to sustain the cholinergic phenotype in damaged motoneurons, which is a first step for adequate neuromuscular neurotransmission, and that this action seems to be mediated directly on neurons since no sign of angiogenic activity was evident. These data reinforces the therapeutical potential of VEGF in motoneuronal diseases.
Collapse
Affiliation(s)
- Lourdes Acosta
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, Seville, Spain
| | - Sara Morcuende
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, Seville, Spain
| | - Silvia Silva-Hucha
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, Seville, Spain
| | - Angel M Pastor
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, Seville, Spain
| | - Rosa R de la Cruz
- Departamento de Fisiología, Facultad de Biología, Universidad de Sevilla, Seville, Spain
| |
Collapse
|
26
|
Abstract
Age-dependent declines in muscle function are observed across species. The loss of mobility resulting from the decline in muscle function represents an important health issue and a key determinant of quality of life for the elderly. It is believed that changes in the structure and function of the neuromuscular junction are important contributors to the observed declines in motor function with increased age. Numerous studies indicate that the aging muscle is an important contributor to the deterioration of the neuromuscular junction but the cellular and molecular mechanisms driving the degeneration of the synapse remain incompletely described. Importantly, growing data from both animal models and humans indicate that exercise can rejuvenate the neuromuscular junction and improve motor function. In this review we will focus on the role of muscle-derived neurotrophin signaling in the rejuvenation of the aged neuromuscular junction in response to exercise.
Collapse
Affiliation(s)
- Tabita Kreko-Pierce
- Department of Cellular and Integrative Physiology, University of Texas Health Sciences Center at San Antonio, San Antonio, Texas, USA.,Barshoph Institute of Longevity and Aging Studies, University of Texas Health Sciences Center at San Antonio, San Antonio, Texas, USA
| | - Benjamin A Eaton
- Department of Cellular and Integrative Physiology, University of Texas Health Sciences Center at San Antonio, San Antonio, Texas, USA.,Barshoph Institute of Longevity and Aging Studies, University of Texas Health Sciences Center at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
27
|
Rosich K, Hanna BF, Ibrahim RK, Hellenbrand DJ, Hanna A. The Effects of Glial Cell Line-Derived Neurotrophic Factor after Spinal Cord Injury. J Neurotrauma 2017; 34:3311-3325. [DOI: 10.1089/neu.2017.5175] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Affiliation(s)
- Konstantin Rosich
- Department of Neurological Surgery, University of Wisconsin, Madison, Wisconsin
| | - Bishoy F. Hanna
- Department of Neurological Surgery, Ross University School of Medicine, Dominica, West Indies
| | - Rami K. Ibrahim
- Department of Neurological Surgery, University of Wisconsin, Madison, Wisconsin
| | - Daniel J. Hellenbrand
- Department of Neurological Surgery, University of Wisconsin, Madison, Wisconsin
- Department of Biomedical Engineering, University of Wisconsin, Madison, Wisconsin
| | - Amgad Hanna
- Department of Neurological Surgery, University of Wisconsin, Madison, Wisconsin
| |
Collapse
|
28
|
Response of the GABAergic System to Axotomy of the Rat Facial Nerve. Neurochem Res 2017; 43:324-339. [PMID: 29164431 DOI: 10.1007/s11064-017-2427-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 09/24/2017] [Accepted: 10/21/2017] [Indexed: 12/31/2022]
Abstract
The responses of inhibitory neurons/synapses to motoneuron injury in the cranial nervous system remain to be elucidated. In this study, we analyzed GABAA receptor (GABAAR) and GABAergic neurons at the protein level in the transected rat facial nucleus. Immunoblotting revealed that the GABAARα1 protein levels in the axotomized facial nucleus decreased significantly 5-14 days post-insult, and these levels remained low for 5 weeks. Immunohistochemical analysis indicated that the GABAARα1-expressing cells were motoneurons. We next examined the specific components of GABAergic neurons, including glutamate decarboxylase (GAD), vesicular GABA transporter (VGAT) and GABA transporter-1 (GAT-1). Immunoblotting indicated that the protein levels of GAD, VGAT and GAT-1 decreased transiently in the transected facial nucleus from 5 to 14 days post-insult, but returned to the control levels at 5 weeks post-insult. Although GABAARα1 protein levels in the transected nucleus did not return to their control levels for 5 weeks post-insult, the administration of glial cell line-derived neurotrophic factor at the cut site significantly ameliorated the reductions. Through these findings, we verified that the injured facial motoneurons suppressed the levels of GABAARα1 protein over the 5 weeks post-insult, presumably due to the deprivation of neurotrophic factor. On the other hand, the levels of the GAD, VGAT and GAT-1 proteins in GABAergic neurons were transiently reduced in the axotomized facial nucleus at 5-14 days post-insult, but recovered at 4-5 weeks post-insult.
Collapse
|
29
|
Bendella H, Rink S, Grosheva M, Sarikcioglu L, Gordon T, Angelov DN. Putative roles of soluble trophic factors in facial nerve regeneration, target reinnervation, and recovery of vibrissal whisking. Exp Neurol 2017; 300:100-110. [PMID: 29104116 DOI: 10.1016/j.expneurol.2017.10.029] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 10/25/2017] [Accepted: 10/30/2017] [Indexed: 12/15/2022]
Abstract
It is well-known that, after nerve transection and surgical repair, misdirected regrowth of regenerating motor axons may occur in three ways. The first way is that the axons enter into endoneurial tubes that they did not previously occupy, regenerate through incorrect fascicles and reinnervate muscles that they did not formerly supply. Consequently the activation of these muscles results in inappropriate movements. The second way is that, in contrast with the precise target-directed pathfinding by elongating motor nerves during embryonic development, several axons rather than a single axon grow out from each transected nerve fiber. The third way of misdirection occurs by the intramuscular terminal branching (sprouting) of each regenerating axon to culminate in some polyinnervation of neuromuscular junctions, i.e. reinnervation of junctions by more than a single axon. Presently, "fascicular" or "topographic specificity" cannot be achieved and hence target-directed nerve regeneration is, as yet, unattainable. Nonetheless, motor and sensory reinnervation of appropriate endoneurial tubes does occur and can be promoted by brief nerve electrical stimulation. This review considers the expression of neurotrophic factors in the neuromuscular system and how this expression can promote functional recovery, with emphasis on the whisking of vibrissae on the rat face in relationship to the expression of the factors. Evidence is reviewed for a role of neurotrophic factors as short-range diffusible sprouting stimuli in promoting complete functional recovery of vibrissal whisking in blind Sprague Dawley (SD)/RCS rats but not in SD rats with normal vision, after facial nerve transection and surgical repair. Briefly, a complicated time course of growth factor expression in the nerves and denervated muscles include (1) an early increase in FGF2 and IGF2, (2) reduced NGF between 2 and 14days after nerve transection and surgical repair, (3) a late rise in BDNF and (4) reduced IGF1 protein in the denervated muscles at 28days. These findings suggest that recovery of motor function after peripheral nerve injury is due, at least in part, to a complex regulation of nerve injury-associated neurotrophic factors and cytokines at the neuromuscular junctions of denervated muscles. In particular, the increase of FGF2 and concomittant decrease of NGF during the first week after facial nerve-nerve anastomosis in SD/RCS blind rats may prevent intramuscular axon sprouting and, in turn, reduce poly-innervation of the neuromuscular junction.
Collapse
Affiliation(s)
- Habib Bendella
- Department of Neurosurgery, University of Witten/Herdecke, Cologne Merheim Medical Center (CMMC), Cologne, Germany
| | - Svenja Rink
- Department of Prosthetic Dentistry, School of Dental and Oral Medicine, University of Cologne, Germany
| | - Maria Grosheva
- Department of Oto-Rhino-Laryngology, University of Cologne, Germany
| | | | - Tessa Gordon
- Department of Surgery, The Hospital for Sick Children, Toronto, Ontario M5G 1X8, Canada
| | | |
Collapse
|
30
|
Hou L, Chen W, Liu X, Qiao D, Zhou FM. Exercise-Induced Neuroprotection of the Nigrostriatal Dopamine System in Parkinson's Disease. Front Aging Neurosci 2017; 9:358. [PMID: 29163139 PMCID: PMC5675869 DOI: 10.3389/fnagi.2017.00358] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 10/19/2017] [Indexed: 12/11/2022] Open
Abstract
Epidemiological studies indicate that physical activity and exercise may reduce the risk of developing Parkinson's disease (PD), and clinical observations suggest that physical exercise can reduce the motor symptoms in PD patients. In experimental animals, a profound observation is that exercise of appropriate timing, duration, and intensity can reduce toxin-induced lesion of the nigrostriatal dopamine (DA) system in animal PD models, although negative results have also been reported, potentially due to inappropriate timing and intensity of the exercise regimen. Exercise may also minimize DA denervation-induced medium spiny neuron (MSN) dendritic atrophy and other abnormalities such as enlarged corticostriatal synapse and abnormal MSN excitability and spiking activity. Taken together, epidemiological studies, clinical observations, and animal research indicate that appropriately dosed physical activity and exercise may not only reduce the risk of developing PD in vulnerable populations but also benefit PD patients by potentially protecting the residual DA neurons or directly restoring the dysfunctional cortico-basal ganglia motor control circuit, and these benefits may be mediated by exercise-triggered production of endogenous neuroprotective molecules such as neurotrophic factors. Thus, exercise is a universally available, side effect-free medicine that should be prescribed to vulnerable populations as a preventive measure and to PD patients as a component of treatment. Future research needs to establish standardized exercise protocols that can reliably induce DA neuron protection, enabling the delineation of the underlying cellular and molecular mechanisms that in turn can maximize exercise-induced neuroprotection and neurorestoration in animal PD models and eventually in PD patients.
Collapse
Affiliation(s)
- Lijuan Hou
- Exercise Physiology Laboratory, College of Physical Education and Sports, Beijing Normal University, Beijing, China
| | - Wei Chen
- Exercise Physiology Laboratory, College of Physical Education and Sports, Beijing Normal University, Beijing, China.,Department of Exercise and Rehabilitation, Physical Education College, Hebei Normal University, Shijiazhuang, China
| | - Xiaoli Liu
- Exercise Physiology Laboratory, College of Physical Education and Sports, Beijing Normal University, Beijing, China
| | - Decai Qiao
- Exercise Physiology Laboratory, College of Physical Education and Sports, Beijing Normal University, Beijing, China
| | - Fu-Ming Zhou
- Department of Pharmacology, University of Tennessee College of Medicine, Memphis, TN, United States
| |
Collapse
|
31
|
Wang HJ, Song G, Liang J, Gao YY, Wang CJ. Involvement of integrin β1/FAK signaling in the analgesic effects induced by glial cell line-derived neurotrophic factor in neuropathic pain. Brain Res Bull 2017; 135:149-156. [DOI: 10.1016/j.brainresbull.2017.10.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 09/20/2017] [Accepted: 10/11/2017] [Indexed: 12/23/2022]
|
32
|
Tan SA, Déglon N, Zurn AD, Baetge EE, Bamber B, Kato AC, Aebischer P. Rescue of Motoneurons from Axotomy-Induced Cell Death by Polymer Encapsulated Cells Genetically Engineered to Release CNTF. Cell Transplant 2017; 5:577-87. [PMID: 8889216 DOI: 10.1177/096368979600500507] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The neurodegenerative disease amyotrophic lateral sclerosis (ALS) results from the progressive loss of motoneurons, leading to death in a few years. Ciliary neurotrophic factor (CNTF), which decreases naturally occurring and axotomy-induced cell death, may result in slowing of motoneuron loss and has been evaluated as a treatment for ALS. Effective administration of this protein to motoneurons may be hampered by the exceedingly short half-life of CNTF, and the inability to deliver effective concentration into the central nervous system after systemic administration in vivo. The constitutive release of CNTF from genetically engineered cells may represent a solution to this delivery problem. In this work, baby hamster kidney (BHK) cells stably tranfected with a chimeric plasmid construct containing the gene for human or mouse CNTF were encapsulated in polymer fibers, which prevents immune rejection and allow long-term survival of the transplanted cells. In vitro bioassays show that the encapsulated transfected cells release bioactive CNTF. In vivo, systemic delivery of human and mouse CNTF from encapsulated cells was observed to rescue 26 and 27% more facial motoneurons, respectively, as compared to capsules containing parent BHK cells 1 wk postaxotomy in neonatal rats. With local application of CNTF on the nerve stump and by systemic delivery through repeated subcutaneous injections, 15 and 13% more rescue effects were observed. These data illustrate the potential of using encapsulated genetically engineered cells to continuously release CNTF to slow down motoneuron degeneration following axotomy and suggest that encapsulated cell delivery of neurotrophic factors may provide a general method for effective administration of therapeutic proteins for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- S A Tan
- Division of Surgical Research, Lausanne University Medical School, Switzerland
| | | | | | | | | | | | | |
Collapse
|
33
|
Chiang YH, Borlongan CV, Zhou FC, Hoffer BJ, Wang Y. Transplantation of Fetal Kidney Cells: Neuroprotection and Neuroregeneration. Cell Transplant 2017; 14:1-9. [PMID: 15789657 DOI: 10.3727/000000005783983304] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Various trophic factors in the transforming growth factor-β (TGF-β) superfamily have been reported to have neuroprotective and neuroregenerative effects. Intracerebral administration of glial cell line-derived neurotrophic factor (GDNF) or bone morphogenetic proteins (BMPs), both members of the TGF-β family, reduce ischemia- or 6-hydroxydopamine (6-OHDA)-induced injury in adult rat brain. Because BMPs and GDNF are highly expressed in fetal kidney cells, transplantation of fetal kidney tissue could serve as a cellular reservoir for such molecules and protect against neuronal injury induced by ischemia, neurotoxins, or reactive oxygen species. In this review, we discuss preclinical evidence for the efficacy of fetal kidney cell transplantation in neuroprotection and regeneration models.
Collapse
Affiliation(s)
- Yung-Hsiao Chiang
- Tri-Service General Hospital, National Defense Medical Center, Taiwan
| | | | | | | | | |
Collapse
|
34
|
Ringer C, Tune S, Bertoune MA, Schwarzbach H, Tsujikawa K, Weihe E, Schütz B. Disruption of calcitonin gene-related peptide signaling accelerates muscle denervation and dampens cytotoxic neuroinflammation in SOD1 mutant mice. Cell Mol Life Sci 2017; 74:339-358. [PMID: 27554772 PMCID: PMC11107523 DOI: 10.1007/s00018-016-2337-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 08/06/2016] [Accepted: 08/08/2016] [Indexed: 12/13/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal motor neuron disease. Neuronal vacuolization and glial activation are pathologic hallmarks in the superoxide dismutase 1 (SOD1) mouse model of ALS. Previously, we found the neuropeptide calcitonin gene-related peptide (CGRP) associated with vacuolization and astrogliosis in the spinal cord of these mice. We now show that CGRP abundance positively correlated with the severity of astrogliosis, but not vacuolization, in several motor and non-motor areas throughout the brain. SOD1 mice harboring a genetic depletion of the βCGRP isoform showed reduced CGRP immunoreactivity associated with vacuolization, while motor functions, body weight, survival, and astrogliosis were not altered. When CGRP signaling was completely disrupted through genetic depletion of the CGRP receptor component, receptor activity-modifying protein 1 (RAMP1), hind limb muscle denervation, and loss of muscle performance were accelerated, while body weight and survival were not affected. Dampened neuroinflammation, i.e., reduced levels of astrogliosis in the brain stem already in the pre-symptomatic disease stage, and reduced microgliosis and lymphocyte infiltrations during the late disease phase were additional neuropathology features in these mice. On the molecular level, mRNA expression levels of brain-derived neurotrophic factor (BDNF) and those of the anti-inflammatory cytokine interleukin 6 (IL-6) were elevated, while those of several pro-inflammatory cytokines found reduced in the brain stem of RAMP1-deficient SOD1 mice at disease end stage. Our results thus identify an important, possibly dual role of CGRP in ALS pathogenesis.
Collapse
Affiliation(s)
- Cornelia Ringer
- Department of Molecular Neurosciences, Institute of Anatomy and Cell Biology, Philipps-University, Robert-Koch-Strasse 8, 35037, Marburg, Germany
- Institute of Anatomy, University of Lübeck, Lübeck, Germany
| | - Sarah Tune
- Department of Physiology, University of Lübeck, Lübeck, Germany
| | - Mirjam A Bertoune
- Department of Medical Cell Biology, Institute of Anatomy and Cell Biology, Philipps-University, Marburg, Germany
| | - Hans Schwarzbach
- Department of Medical Cell Biology, Institute of Anatomy and Cell Biology, Philipps-University, Marburg, Germany
| | - Kazutake Tsujikawa
- Laboratory of Molecular and Cellular Physiology, Graduate School of Pharmaceutical Sciences, Osaka University, Yamadaoka, Suita, Osaka, Japan
| | - Eberhard Weihe
- Department of Molecular Neurosciences, Institute of Anatomy and Cell Biology, Philipps-University, Robert-Koch-Strasse 8, 35037, Marburg, Germany.
| | - Burkhard Schütz
- Department of Molecular Neurosciences, Institute of Anatomy and Cell Biology, Philipps-University, Robert-Koch-Strasse 8, 35037, Marburg, Germany.
| |
Collapse
|
35
|
Wang B, Yuan J, Xu J, Chen X, Ying X, Dong P. Brain-derived and glial cell line-derived neurotrophic factor fusion protein immobilization to laminin. Exp Ther Med 2016; 13:178-186. [PMID: 28123487 PMCID: PMC5245157 DOI: 10.3892/etm.2016.3925] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 09/01/2016] [Indexed: 11/06/2022] Open
Abstract
Damage to the recurrent laryngeal nerve often causes hoarseness, dyspnea, dysphagia, and sometimes asphyxia due to vocal cord paralysis which result in a reduction of quality of life. Brain-derived neurotrophic factor (BDNF) and glial cell line-derived neurotrophic factor (GDNF) play critical roles in peripheral nerve regeneration. However, methods for efficiently delivering these molecules are lacking, which limits their use in clinical applications. The present study reports an effective strategy for targeting BDNF and GDNF to laminin by fusing the N-terminal domains of these molecules with agrin (NtA). More specifically, laminin-binding efficacy was assessed and sustained release assays of the delivery of BDNF or GDNF fused with NtA (LBD-BDNF or LBD-GDNF) to laminin were conducted in vitro. In addition, the bioactivity of LBD-BDNF and LBD-GDNF on laminin in vitro was investigated. LBD-BDNF and LBD-GDNF were each able to specifically bind to laminin and maintain their activity in vitro. Moreover, neurotrophic factors with NtA retained higher concentrations and bioactivity levels compared with those without NtA. The ratio of LBD-BDNF and LBD-GDNF that produced optimal effects was 4:6. BDNF and GDNF fused with NtA were effective in specifically binding to laminin. As laminin is a major component of the extracellular matrix, LBD-BDNF and LBD-GDNF may prove useful in the repair of peripheral nerve injuries.
Collapse
Affiliation(s)
- Baoxin Wang
- Department of Otolaryngology, Head and Neck Surgery, Shanghai Jiao Tong University Affiliated to Shanghai First People's Hospital, Shanghai 201620, P.R. China
| | - Junjie Yuan
- Department of Orthopedics, Shanghai Fengxian District Central Hospital, Shanghai Jiao Tong University Affiliated to Sixth People's Hospital South Campus, Shanghai 200011, P.R. China
| | - Jiafeng Xu
- School of Economics and Finance, Shanghai International Studies University, Shanghai 200083, P.R. China
| | - Xinwei Chen
- Department of Otolaryngology, Head and Neck Surgery, Shanghai Jiao Tong University Affiliated to Shanghai First People's Hospital, Shanghai 201620, P.R. China
| | - Xinjiang Ying
- Department of Otolaryngology, Head and Neck Surgery, Shanghai Jiao Tong University Affiliated to Shanghai First People's Hospital, Shanghai 201620, P.R. China
| | - Pin Dong
- Department of Otolaryngology, Head and Neck Surgery, Shanghai Jiao Tong University Affiliated to Shanghai First People's Hospital, Shanghai 201620, P.R. China
| |
Collapse
|
36
|
Ma KH, Hung HA, Svaren J. Epigenomic Regulation of Schwann Cell Reprogramming in Peripheral Nerve Injury. J Neurosci 2016; 36:9135-47. [PMID: 27581455 PMCID: PMC5005723 DOI: 10.1523/jneurosci.1370-16.2016] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 07/08/2016] [Accepted: 07/15/2016] [Indexed: 12/16/2022] Open
Abstract
UNLABELLED The rapid and dynamic transcriptional changes of Schwann cells in response to injury are critical to peripheral nerve repair, yet the epigenomic reprograming that leads to the induction of injury-activated genes has not been characterized. Polycomb Repressive Complex 2 (PRC2) catalyzes the trimethylation of lysine 27 of histone H3 (H3K27me3), which produces a transcriptionally repressive chromatin environment. We find that many promoters and/or gene bodies of injury-activated genes of mature rat nerves are occupied with H3K27me3. In contrast, the majority of distal enhancers that gain H3K27 acetylation after injury are not repressed by H3K27 methylation before injury, which is normally observed in developmentally poised enhancers. Injury induces demethylation of H3K27 in many genes, such as Sonic hedgehog (Shh), which is silenced throughout Schwann cell development before injury. In addition, experiments using a Schwann cell-specific mouse knock-out of the Eed subunit of PRC2 indicate that demethylation is a rate-limiting step in the activation of such genes. We also show that some transcription start sites of H3K27me3-repressed injury genes of uninjured nerves are bound with a mark of active promoters H3K4me3, for example, Shh and Gdnf, and the reduction of H3K27me3 results in increased trimethylation of H3K4. Our findings identify reversal of polycomb repression as a key step in gene activation after injury. SIGNIFICANCE STATEMENT Peripheral nerve regeneration after injury is dependent upon implementation of a novel genetic program in Schwann cells that supports axonal survival and regeneration. Identifying means to enhance Schwann cell reprogramming after nerve injury could be used to foster effective remyelination in the treatment of demyelinating disorders and in identifying pathways involved in regenerative process of myelination. Although recent progress has identified transcriptional determinants of successful reprogramming of the Schwann cell transcriptome after nerve injury, our results have highlighted a novel epigenomic pathway in which reversal of the Polycomb pathway of repressive histone methylation is required for activation of a significant number of injury-induced genes.
Collapse
Affiliation(s)
- Ki H Ma
- Waisman Center, Cellular and Molecular Pathology Graduate Program, and
| | - Holly A Hung
- Waisman Center, Cellular and Molecular Pathology Graduate Program, and
| | - John Svaren
- Waisman Center, Department of Comparative Biosciences, University of Wisconsin, Madison, Wisconsin 53705
| |
Collapse
|
37
|
Brambilla L, Guidotti G, Martorana F, Iyer AM, Aronica E, Valori CF, Rossi D. Disruption of the astrocytic TNFR1-GDNF axis accelerates motor neuron degeneration and disease progression in amyotrophic lateral sclerosis. Hum Mol Genet 2016; 25:3080-3095. [PMID: 27288458 DOI: 10.1093/hmg/ddw161] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 05/17/2016] [Accepted: 05/19/2016] [Indexed: 11/14/2022] Open
Abstract
Considerable evidence indicates that neurodegeneration in amyotrophic lateral sclerosis (ALS) can be conditioned by a deleterious interplay between motor neurons and astrocytes. Astrocytes are the major glial component in the central nervous system (CNS) and fulfill several activities that are essential to preserve CNS homeostasis. In physiological and pathological conditions, astrocytes secrete a wide range of factors by which they exert multimodal influences on their cellular neighbours. Among others, astrocytes can secrete glial cell line-derived neurotrophic factor (GDNF), one of the most potent protective agents for motor neurons. This suggests that the modulation of the endogenous mechanisms that control the production of astrocytic GDNF may have therapeutic implications in motor neuron diseases, particularly ALS. In this study, we identified TNF receptor 1 (TNFR1) signalling as a major promoter of GDNF synthesis/release from human and mouse spinal cord astrocytes in vitro and in vivo To determine whether endogenously produced TNFα can also trigger the synthesis of GDNF in the nervous system, we then focused on SOD1G93A ALS transgenic mice, whose affected tissues spontaneously exhibit high levels of TNFα and its receptor 1 at the onset and symptomatic stage of the disease. In SOD1G93A spinal cords, we verified a strict correlation in the expression of the TNFα, TNFR1 and GDNF triad at different stages of disease progression. Yet, ablation of TNFR1 completely abolished GDNF rises in both SOD1G93A astrocytes and spinal cords, a condition that accelerated motor neuron degeneration and disease progression. Our data suggest that the astrocytic TNFR1-GDNF axis represents a novel target for therapeutic intervention in ALS.
Collapse
Affiliation(s)
- Liliana Brambilla
- Laboratory for Research on Neurodegenerative Disorders, IRCCS Fondazione Salvatore Maugeri, 27100 Pavia, Italy
| | - Giulia Guidotti
- Laboratory for Research on Neurodegenerative Disorders, IRCCS Fondazione Salvatore Maugeri, 27100 Pavia, Italy
| | - Francesca Martorana
- Laboratory for Research on Neurodegenerative Disorders, IRCCS Fondazione Salvatore Maugeri, 27100 Pavia, Italy
| | - Anand M Iyer
- Department of Pathology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Eleonora Aronica
- Department of Pathology, Academic Medical Center, University of Amsterdam, 1105 AZ Amsterdam, The Netherlands
| | - Chiara F Valori
- Department of Neuropathology, German Center for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
| | - Daniela Rossi
- Laboratory for Research on Neurodegenerative Disorders, IRCCS Fondazione Salvatore Maugeri, 27100 Pavia, Italy,
| |
Collapse
|
38
|
Wang K, Zhu X, Zhang K, Zhou F, Zhu L. Gas1 Knockdown Increases the Neuroprotective Effect of Glial Cell-Derived Neurotrophic Factor Against Glutamate-Induced Cell Injury in Human SH-SY5Y Neuroblastoma Cells. Cell Mol Neurobiol 2016; 36:603-11. [PMID: 26215053 PMCID: PMC11482505 DOI: 10.1007/s10571-015-0241-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 07/16/2015] [Indexed: 01/01/2023]
Abstract
Growth arrest-specific 1 (Gas1) protein acts as an inhibitor of cell growth and a mediator of cell death in nervous system during development and is also re-expressed in adult neurons during excitotoxic insult. Due to its structural similarity to the glial cell-derived neurotrophic factor family receptors α (GFRα), Gas1 is likely to interfere with the neuroprotective effect of GDNF. In the present study, we investigated the expression profile of Gas1 during glutamate insults in human SH-SY5Y neuroblastoma cells as well as the influence of Gas1 inhibition on the protective effect of GDNF against glutamate-induced cell injury. Our data showed that Gas1 expression was significantly increased with the treatment of glutamate in SH-SY5Y cells. The silencing of Gas1 by small interfering RNA promoted the protective effect of GDNF against glutamate-induced cytotoxicity as well as cell apoptosis, which effect was likely mediated through activating Akt/PI3 K-dependent cell survival signaling pathway and inhibiting mitochondrial-dependent cell apoptosis signaling pathway via Bad dephosphorylation blockade. In summary, this study showed the synergistic effect of Gas1 inhibition and GDNF against glutamate-induced cell injury in human SH-SY5Y neuroblastoma cells, which information might significantly contribute to better understanding the function of Gas1 in neuronal cells and form the basis of the therapeutic development of GDNF in treating human neurodegenerative diseases in the future.
Collapse
Affiliation(s)
- Ke Wang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Qianrong Road 20, Wuxi, 214063, Jiangsu, China.
| | - Xue Zhu
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Qianrong Road 20, Wuxi, 214063, Jiangsu, China
| | - Kai Zhang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Qianrong Road 20, Wuxi, 214063, Jiangsu, China
| | - Fanfan Zhou
- Faculty of Pharmacy, University of Sydney, Room 104 Medical Foundation Building K25, Sydney, NSW, 2006, Australia.
| | - Ling Zhu
- Save Sight Institute, University of Sydney, Sydney, NSW, 2000, Australia
| |
Collapse
|
39
|
Hernandez-Morato I, Tewari I, Sharma S, Pitman MJ. Blockade of glial-derived neurotrophic factor in laryngeal muscles promotes appropriate reinnervation. Laryngoscope 2016; 126:E337-42. [DOI: 10.1002/lary.25953] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 01/14/2016] [Accepted: 02/04/2016] [Indexed: 11/08/2022]
Affiliation(s)
| | - Ishan Tewari
- Department of Anatomy and Cell Biology; New York Medical College; Valhalla New York
| | - Sansar Sharma
- Department of Anatomy and Cell Biology; New York Medical College; Valhalla New York
| | - Michael J. Pitman
- Department of Otolaryngology; New York Eye and Ear Infirmary of Mount Sinai; New York New York U.S.A
| |
Collapse
|
40
|
Tajdaran K, Gordon T, Wood MD, Shoichet MS, Borschel GH. An engineered biocompatible drug delivery system enhances nerve regeneration after delayed repair. J Biomed Mater Res A 2015; 104:367-76. [DOI: 10.1002/jbm.a.35572] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Revised: 08/21/2015] [Accepted: 09/21/2015] [Indexed: 12/14/2022]
Affiliation(s)
- Kasra Tajdaran
- Division of Plastic and Reconstructive Surgery; the Hospital for Sick Children; 555 University Ave Toronto Ontario M5G1X8 Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto; Toronto Ontario Canada
| | - Tessa Gordon
- Division of Plastic and Reconstructive Surgery; the Hospital for Sick Children; 555 University Ave Toronto Ontario M5G1X8 Canada
- Division of Plastic and Reconstructive Surgery, Department of Surgery; University of Toronto; Toronto Ontario Canada
- Program in Neuroscience, the Hospital for Sick Children Research Institute; Toronto Ontario Canada
| | - Mathew D. Wood
- Division of Plastic and Reconstructive Surgery; Washington University School of Medicine; St. Louis Missouri
| | - Molly S. Shoichet
- Institute of Biomaterials and Biomedical Engineering, University of Toronto; Toronto Ontario Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto Ontario Canada
| | - Gregory H. Borschel
- Division of Plastic and Reconstructive Surgery; the Hospital for Sick Children; 555 University Ave Toronto Ontario M5G1X8 Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto; Toronto Ontario Canada
- Division of Plastic and Reconstructive Surgery, Department of Surgery; University of Toronto; Toronto Ontario Canada
- Program in Neuroscience, the Hospital for Sick Children Research Institute; Toronto Ontario Canada
| |
Collapse
|
41
|
Roam JL, Yan Y, Nguyen PK, Kinstlinger IS, Leuchter MK, Hunter DA, Wood MD, Elbert DL. A modular, plasmin-sensitive, clickable poly(ethylene glycol)-heparin-laminin microsphere system for establishing growth factor gradients in nerve guidance conduits. Biomaterials 2015; 72:112-24. [PMID: 26352518 DOI: 10.1016/j.biomaterials.2015.08.054] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Revised: 08/25/2015] [Accepted: 08/28/2015] [Indexed: 12/16/2022]
Abstract
Peripheral nerve regeneration is a complex problem that, despite many advancements and innovations, still has sub-optimal outcomes. Compared to biologically derived acellular nerve grafts and autografts, completely synthetic nerve guidance conduits (NGC), which allow for precise engineering of their properties, are promising but still far from optimal. We have developed an almost entirely synthetic NGC that allows control of soluble growth factor delivery kinetics, cell-initiated degradability and cell attachment. We have focused on the spatial patterning of glial-cell derived human neurotrophic factor (GDNF), which promotes motor axon extension. The base scaffolds consisted of heparin-containing poly(ethylene glycol) (PEG) microspheres. The modular microsphere format greatly simplifies the formation of concentration gradients of reversibly bound GDNF. To facilitate axon extension, we engineered the microspheres with tunable plasmin degradability. 'Click' cross-linking chemistries were also added to allow scaffold formation without risk of covalently coupling the growth factor to the scaffold. Cell adhesion was promoted by covalently bound laminin. GDNF that was released from these microspheres was confirmed to retain its activity. Graded scaffolds were formed inside silicone conduits using 3D-printed holders. The fully formed NGC's contained plasmin-degradable PEG/heparin scaffolds that developed linear gradients in reversibly bound GDNF. The NGC's were implanted into rats with severed sciatic nerves to confirm in vivo degradability and lack of a major foreign body response. The NGC's also promoted robust axonal regeneration into the conduit.
Collapse
Affiliation(s)
- Jacob L Roam
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Ying Yan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, Campus Box 8238, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Peter K Nguyen
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Ian S Kinstlinger
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Michael K Leuchter
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Daniel A Hunter
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, Campus Box 8238, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Matthew D Wood
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Washington University School of Medicine, Campus Box 8238, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Donald L Elbert
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA.
| |
Collapse
|
42
|
d'Anglemont de Tassigny X, Pascual A, López-Barneo J. GDNF-based therapies, GDNF-producing interneurons, and trophic support of the dopaminergic nigrostriatal pathway. Implications for Parkinson's disease. Front Neuroanat 2015; 9:10. [PMID: 25762899 PMCID: PMC4327623 DOI: 10.3389/fnana.2015.00010] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Accepted: 01/21/2015] [Indexed: 01/09/2023] Open
Abstract
The glial cell line-derived neurotrophic factor (GDNF) is a well-established trophic agent for dopaminergic (DA) neurons in vitro and in vivo. GDNF is necessary for maintenance of neuronal morphological and neurochemical phenotype and protects DA neurons from toxic damage. Numerous studies on animal models of Parkinson’s disease (PD) have reported beneficial effects of GDNF on nigrostriatal DA neuron survival. However, translation of these observations to the clinical setting has been hampered so far by side effects associated with the chronic continuous intra-striatal infusion of recombinant GDNF. In addition, double blind and placebo-controlled clinical trials have not reported any clinically relevant effect of GDNF on PD patients. In the past few years, experiments with conditional Gdnf knockout mice have suggested that GDNF is necessary for maintenance of DA neurons in adulthood. In parallel, new methodologies for exogenous GDNF delivery have been developed. Recently, it has been shown that a small population of scattered, electrically interconnected, parvalbumin positive (PV+) GABAergic interneurons is responsible for most of the GDNF produced in the rodent striatum. In addition, cholinergic striatal interneurons appear to be also involved in the modulation of striatal GDNF. In this review, we summarize current knowledge on brain GDNF delivery, homeostasis, and its effects on nigrostriatal DA neurons. Special attention is paid to the therapeutic potential of endogenous GDNF stimulation in PD.
Collapse
Affiliation(s)
- Xavier d'Anglemont de Tassigny
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla Seville, Spain
| | - Alberto Pascual
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla Seville, Spain
| | - José López-Barneo
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla Seville, Spain ; Departamento de Fisiología Médica y Biofísica, Facultad de Medicina, Universidad de Sevilla Seville, Spain ; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) Madrid, Spain
| |
Collapse
|
43
|
Rosa E, Cha J, Bain JR, Fahnestock M. Calcitonin gene-related peptide regulation of glial cell-line derived neurotrophic factor in differentiated rat myotubes. J Neurosci Res 2014; 93:514-20. [PMID: 25403360 DOI: 10.1002/jnr.23512] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 09/29/2014] [Accepted: 10/09/2014] [Indexed: 01/05/2023]
Abstract
Glial cell-line derived neurotrophic factor (GDNF) is the most potent trophic factor for motoneuron survival and neuromuscular junction formation. GDNF is upregulated in injured or denervated skeletal muscle and returns to normal levels following reinnervation. However, the mechanism by which GDNF is regulated in denervated muscle is not well understood. The nerve-derived neurotransmitter calcitonin gene-related peptide (CGRP) is upregulated following neuromuscular injury and is subsequently released from motoneurons at the neuromuscular junction. CGRP also promotes nerve regeneration, but the mechanism is not well understood. The current study investigates whether this increase in CGRP regulates GDNF, thus playing a key role in promoting regeneration of injured nerves. This study demonstrates that CGRP increases GDNF secretion without affecting its transcription or translation. Rat L6 myoblasts were differentiated into myotubes and subsequently treated with CGRP. GDNF mRNA expression levels were quantified by quantitative real-time reverse transcription-polymerase chain reaction, and secreted GDNF was quantified in the conditioned medium by ELISA. CGRP treatment increased secreted GDNF protein without altering GDNF mRNA levels. The translational inhibitor cycloheximide did not affect CGRP-induced GDNF secreted protein levels, whereas the secretional inhibitor brefeldin A blocked the CGRP-induced increase in GDNF. This study highlights the importance of injury-induced upregulation of CGRP by exposing its ability to increase GDNF levels and demonstrates a secretional mechanism for regulation of this key regeneration-promoting neurotrophic factor.
Collapse
Affiliation(s)
- Elyse Rosa
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, Ontario, Canada
| | | | | | | |
Collapse
|
44
|
Neurotrophic requirements of human motor neurons defined using amplified and purified stem cell-derived cultures. PLoS One 2014; 9:e110324. [PMID: 25337699 PMCID: PMC4206291 DOI: 10.1371/journal.pone.0110324] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2014] [Accepted: 09/18/2014] [Indexed: 12/29/2022] Open
Abstract
Human motor neurons derived from embryonic and induced pluripotent stem cells (hESCs and hiPSCs) are a potentially important tool for studying motor neuron survival and pathological cell death. However, their basic survival requirements remain poorly characterized. Here, we sought to optimize a robust survival assay and characterize their response to different neurotrophic factors. First, to increase motor neuron yield, we screened a small-molecule collection and found that the Rho-associated kinase (ROCK) inhibitor Y-27632 enhances motor neuron progenitor proliferation up to 4-fold in hESC and hiPSC cultures. Next, we FACS-purified motor neurons expressing the Hb9::GFP reporter from Y-27632-amplified embryoid bodies and cultured them in the presence of mitotic inhibitors to eliminate dividing progenitors. Survival of these purified motor neurons in the absence of any other cell type was strongly dependent on neurotrophic support. GDNF, BDNF and CNTF all showed potent survival effects (EC(50) 1-2 pM). The number of surviving motor neurons was further enhanced in the presence of forskolin and IBMX, agents that increase endogenous cAMP levels. As a demonstration of the ability of the assay to detect novel neurotrophic agents, Y-27632 itself was found to support human motor neuron survival. Thus, purified human stem cell-derived motor neurons show survival requirements similar to those of primary rodent motor neurons and can be used for rigorous cell-based screening.
Collapse
|
45
|
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]
|
46
|
Hernandez-Morato I, Isseroff TF, Sharma S, Pitman MJ. Differential expression of glial-derived neurotrophic factor in rat laryngeal muscles during reinnervation. Laryngoscope 2014; 124:2750-6. [DOI: 10.1002/lary.24759] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 04/26/2014] [Indexed: 11/11/2022]
Affiliation(s)
| | - Tova F. Isseroff
- Department of Otolaryngology; The New York Eye and Ear Infirmary; New York New York U.S.A
| | - Sansar Sharma
- Department of Anatomy and Cell Biology; New York Medical College; Valhalla New York
| | - Michael J. Pitman
- Department of Otolaryngology; The New York Eye and Ear Infirmary; New York New York U.S.A
| |
Collapse
|
47
|
Berdugo-Vega G, Arias-Gil G, Rodriguez-Niedenführ M, Davies DC, Vázquez T, Pascual-Font A. GFAP immunoreactivity within the rat nucleus ambiguus after laryngeal nerve injury. J Anat 2014; 225:492-501. [PMID: 25181319 DOI: 10.1111/joa.12231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2014] [Indexed: 11/30/2022] Open
Abstract
Changes that occur in astroglial populations of the nucleus ambiguus after recurrent (RLN) or superior (SLN) laryngeal nerve injury have hitherto not been fully characterised. In the present study, rat RLN and SLN were lesioned. After 3, 7, 14, 28 or 56 days of survival, the nucleus ambiguus was investigated by means of glial fibrillary acidic protein (GFAP) immunofluorescence or a combination of GFAP immunofluorescence and the application of retrograde tracers. GFAP immunoreactivity was significantly increased 3 days after RLN resection and it remained significantly elevated until after 28 days post injury (dpi). By 56 dpi it had returned to basal levels. In contrast, following RLN transection with repair, GFAP immunoreactivity was significantly elevated at 7 dpi and remained significantly elevated until 14 dpi. It had returned to basal levels by 28 dpi. Topographical analysis of the distribution of GFAP immunoreactivity revealed that after RLN injury, GFAP immunoreactivity was increased beyond the area of the nucleus ambiguus within which RLN motor neuron somata were located. GFAP immunoreactivity was also observed in the vicinity of neuronal somata that project into the uninjured SLN. Similarly, lesion of the SLN resulted in increased GFAP immunoreactivity around the neuronal somata projecting into it and also in the vicinity of the motor neuron somata projecting into the RLN. The increase in GFAP immunoreactivity outside of the region containing the motor neurons projecting into the injured nerve, may reflect the onset of a regenerative process attempting to compensate for impairment of one of the laryngeal nerves and may occur because of the dual innervation of the posterior cricoarytenoid muscle. This dual innervation of a very specialised muscle could provide a useful model system for studying the molecular mechanisms underlying axonal regeneration process and the results of the current study could provide the basis for studies into functional regeneration following laryngeal nerve injury, with subsequent application to humans.
Collapse
Affiliation(s)
- G Berdugo-Vega
- Department of Human Anatomy and Embryology I, School of Medicine, Complutense University of Madrid, Madrid, Spain
| | | | | | | | | | | |
Collapse
|
48
|
Hashimoto M, Kanda M, Ikeno K, Hayashi Y, Nakamura T, Ogawa Y, Fukumitsu H, Nomoto H, Furukawa S. Oral Administration of Royal Jelly Facilitates mRNA Expression of Glial Cell Line-Derived Neurotrophic Factor and Neurofilament H in the Hippocampus of the Adult Mouse Brain. Biosci Biotechnol Biochem 2014; 69:800-5. [PMID: 15849420 DOI: 10.1271/bbb.69.800] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Royal jelly (RJ) is known to have a variety of biological activities toward various types of cells and tissues of animal models, but nothing is known about its effect on brain functions. Hence, we examined the effect of oral administration of RJ on the mRNA expression of various neurotrophic factors, their receptors, and neural cell markers in the mouse brain. Our results revealed that RJ selectively facilitates the mRNA expression of glial cell line-derived neurotrophic factor (GDNF), a potent neurotrophic factor acting in the brain, and neurofilament H, a specific marker predominantly found in neuronal axons, in the adult mouse hippocampus. These observations suggest that RJ shows neurotrophic effects on the mature brain via stimulation of GDNF production, and that enhanced expression of neurofilament H mRNA is involved in events subsequently caused by GDNF. RJ may play neurotrophic and/or neuroprotective roles in the adult brain through GDNF.
Collapse
Affiliation(s)
- Manabu Hashimoto
- Laboratory of Molecular Biology, Gifu Pharmaceutical University, Gifu 502-8585, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Functional effect of local administration of glial derived neurotrophic factor combined with inside-out artery graft on sciatic nerve regeneration in rat. Int J Surg 2014; 12:457-63. [DOI: 10.1016/j.ijsu.2014.03.018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Revised: 01/29/2014] [Accepted: 03/28/2014] [Indexed: 11/19/2022]
|
50
|
Gyorkos AM, Spitsbergen JM. GDNF content and NMJ morphology are altered in recruited muscles following high-speed and resistance wheel training. Physiol Rep 2014; 2:e00235. [PMID: 24744904 PMCID: PMC3966253 DOI: 10.1002/phy2.235] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Revised: 01/20/2014] [Accepted: 01/21/2014] [Indexed: 12/25/2022] Open
Abstract
Glial cell line‐derived neurotrophic factor (GDNF) may play a role in delaying the onset of aging and help compress morbidity by preventing motor unit degeneration. Exercise has been shown to alter GDNF expression differently in slow‐ and fast‐twitch myofibers. The aim was to examine the effects of different intensities (10, 20, ~30, and ~40 m·min−1) of wheel running on GDNF expression and neuromuscular junction (NMJ) plasticity in slow‐ and fast‐twitch myofibers. Male Sprague‐Dawley Rats (4 weeks old) were divided into two sedentary control groups (CON4 week, n = 5 and CON6 week, n = 5), two involuntary running groups, one at a low velocity; 10 m/min (INVOL‐low, n = 5), and one at a higher velocity; 20 m/min (INVOL‐high, n = 5), and two voluntary running groups with resistance (VOL‐R, n = 5, 120 g), and without resistance (VOL‐NR, n = 5, 4.5 g). GDNF protein content, determined by enzyme‐linked immunosorbent assay (ELISA), increased significantly in the recruited muscles. Plantaris (PLA) GDNF protein content increased 174% (P <0.05) and 161% (P <0.05) and end plate‐stained area increased 123% (P <0.05) and 72% (P <0.05) following VOL‐R, and VOL‐NR training, respectively, when compared to age‐matched controls. A relationship exists between GDNF protein content and end plate area (r = 0.880, P < 0.01, n = 15). VOL‐R training also resulted in more dispersed synapses in the PLA muscle when compared to age‐matched controls (P <0.05). Higher intensity exercise (>30 m/min) can increase GDNF protein content in fast‐twitch myofibers as well as induce changes in the NMJ morphology. These findings help to inform exercise prescription to preserve the integrity of the neuromuscular system through aging and disease. Higher intensity exercise (>30 m/min) can increase glial cell line‐derived neurotrophic factor (GDNF) protein content in fast‐twitch myofibers as well as induce changes in the neuromuscular junction (NMJ) morphology. These findings help to inform exercise prescription to preserve the integrity of the neuromuscular system through aging and disease.
Collapse
Affiliation(s)
- Amy Morrison Gyorkos
- Department of Biological Sciences, Western Michigan University, 1903 W Michigan Ave., Kalamazoo, 49008-5410, Michigan
| | - John M Spitsbergen
- Department of Biological Sciences, Western Michigan University, 1903 W Michigan Ave., Kalamazoo, 49008-5410, Michigan
| |
Collapse
|