1
|
Li S, Wu L, Xie J, Zhou G, Wen X, Deng L, Lin S, Liu G, Chen S, Xiao Z. Edaravone Improves Motor Dysfunction Following Brachial Plexus Avulsion Injury in Rats. ACS Chem Neurosci 2025; 16:479-489. [PMID: 39791183 DOI: 10.1021/acschemneuro.4c00717] [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: 01/12/2025] Open
Abstract
Brachial plexus root avulsion (BPRA) is often caused by road collisions, leading to total loss of motor function in the upper limb. At present, effective treatment options remain limited. Edaravone (EDA), a substance that eliminates free radicals, exhibits numerous biological properties, including neuroprotective, antioxidant and anti-inflammatory effects. However, the specific role and molecular mechanisms of EDA in the treatment of BPRA remain to be fully elucidated. The present study used a rat model of BPRA, following avulsion of the fifth, sixth and seventh cervical (C5, C6 and C7) anterior roots. Notably, C6 was replanted following a subcutaneous injection of either saline or 30 mg/kg/day EDA for seven continuous days. Subsequently, behavioral, histochemical, Western blot and reverse transcription-quantitative PCR (RT-PCR) analyses were conducted. Results of the present study revealed that treatment with EDA improves motor dysfunction, indicated by the increased Grooming test score, usage of the affected limb, and Irvine, Beatties and Bresnahan (IBB) score, following BPRA. In addition, EDA reduced the death of motoneurons (MNs), indicated by the increased number of Nissl-positive neuron, at the site of the affected limb, inhibited neuroinflammation and cellular pyroptosis, indicated by the decreased expression levels of IL-1β, IL-6, TNF-α, IL-18, p-p65, NLRP3, GSDMD and Caspase-1, improved the morphology of the abnormal myocutaneous nerve fibers, promoted axon remyelination, indicated by increased mRNA expression levels of remyelination-associated genes, including egr2, GAP-43, hmgcr, L1CAM, mpz, pmp22 and prx and demyelination-associated genes, including ngfr, notch1, pou3f1 and sox2, and alleviated muscle atrophy, indicated by the increased weight and volume of biceps brachii muscle, and the decreased number of fibroblasts and increased diameters in the fibers. Collectively, results of the present study suggested that EDA may support axonal remyelination and inhibit pyroptosis-associated neuroinflammation, enhancing MN survival and facilitating functional motor recovery. Thus, the present study may provide a novel theoretical basis for the use of EDA in the treatment of BPRA.
Collapse
Affiliation(s)
- Sijing Li
- Department of Neurology, Multi-Omics Research Center for Brain Disorders,The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Clinical Research Center for Immune-Related Encephalopathy of Hunan Province (The First Affiliated Hospital), Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Lin Wu
- Department of Neurology, Multi-Omics Research Center for Brain Disorders,The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Clinical Research Center for Immune-Related Encephalopathy of Hunan Province (The First Affiliated Hospital), Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Department of Neurology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421000, People's Republic of China
| | - Juan Xie
- Department of Neurology, Multi-Omics Research Center for Brain Disorders,The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Clinical Research Center for Immune-Related Encephalopathy of Hunan Province (The First Affiliated Hospital), Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Department of Emergency, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421000, People's Republic of China
| | - Guijuan Zhou
- Department of Neurology, Multi-Omics Research Center for Brain Disorders,The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Clinical Research Center for Immune-Related Encephalopathy of Hunan Province (The First Affiliated Hospital), Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Xuanwei Wen
- Department of Neurology, Multi-Omics Research Center for Brain Disorders,The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Clinical Research Center for Immune-Related Encephalopathy of Hunan Province (The First Affiliated Hospital), Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Limin Deng
- Department of Neurology, Multi-Omics Research Center for Brain Disorders,The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Clinical Research Center for Immune-Related Encephalopathy of Hunan Province (The First Affiliated Hospital), Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Shudong Lin
- Department of Neurology, Multi-Omics Research Center for Brain Disorders,The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Clinical Research Center for Immune-Related Encephalopathy of Hunan Province (The First Affiliated Hospital), Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Guozhi Liu
- Department of Neurology, Multi-Omics Research Center for Brain Disorders,The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Clinical Research Center for Immune-Related Encephalopathy of Hunan Province (The First Affiliated Hospital), Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Shuangxi Chen
- Department of Neurology, Multi-Omics Research Center for Brain Disorders,The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Clinical Research Center for Immune-Related Encephalopathy of Hunan Province (The First Affiliated Hospital), Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| | - Zijian Xiao
- Department of Neurology, Multi-Omics Research Center for Brain Disorders,The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
- Clinical Research Center for Immune-Related Encephalopathy of Hunan Province (The First Affiliated Hospital), Hengyang Medical School, University of South China, Hengyang, Hunan 421001, China
| |
Collapse
|
2
|
Snijders GJLJ, Gigase FAJ. Neuroglia in mood disorders. HANDBOOK OF CLINICAL NEUROLOGY 2025; 210:287-302. [PMID: 40148049 DOI: 10.1016/b978-0-443-19102-2.00010-7] [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: 03/29/2025]
Abstract
Multiple lines of evidence indicate that mood disorders, such as major depressive and bipolar disorder, are associated with abnormalities in neuroglial cells. This chapter discusses the existing literature investigating the potential role of astrocytes, oligodendrocytes, and microglia in mood pathology. We will describe evidence from in vivo imaging, postmortem, animal models based on (stress) paradigms that mimic depressive-like behavior, and biomarker studies in blood and cerebrospinal fluid in patients with mood disorders. The effect of medication used in the treatment of mood disorders, such as antidepressants and lithium, on glial function is discussed. Lastly, we highlight the most relevant findings about potential deficiencies in glia-glia crosstalk in mood disorders. Overall, decreased astrocyte and oligodendrocyte density and expression and microglial changes in homeostatic functions have frequently been put forward in MDD pathology. Studies of BD report similar findings to some extent; however, the evidence is less well established. Together, these findings are suggestive of reduced glial cell function leading to potential white matter abnormalities, glutamate dysregulation, disrupted neuronal functioning, and neurotransmission. However, more research is required to better understand the exact mechanisms underlying glial cell contributions to mood disorder development.
Collapse
Affiliation(s)
- Gijsje J L J Snijders
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States.
| | - Frederieke A J Gigase
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
3
|
Wu L, Chen S, He B, Zhou G, Xu Y, Zhu G, Xie J, Deng L, Wen X, Li S, Xiao Z. Acetylglutamine facilitates motor recovery and alleviates neuropathic pain after brachial plexus root avulsion in rats. J Transl Med 2023; 21:563. [PMID: 37612586 PMCID: PMC10464467 DOI: 10.1186/s12967-023-04399-7] [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] [Received: 02/01/2023] [Accepted: 07/29/2023] [Indexed: 08/25/2023] Open
Abstract
BACKGROUND Brachial plexus root avulsion (BPRA), a disabling peripheral nerve injury, induces substantial motoneuron death, motor axon degeneration and denervation of biceps muscles, leading to the loss of upper limb motor function. Acetylglutamine (N-acetyl-L-glutamine, NAG) has been proven to exert neuroprotective and anti-inflammatory effects on various disorders of the nervous system. Thus, the present study mainly focused on the influence of NAG on motor and sensory recovery after BPRA in rats and the underlying mechanisms. METHODS Male adult Sprague Dawley (SD) rats were subjected to BPRA and reimplantation surgery and subsequently treated with NAG or saline. Behavioral tests were conducted to evaluate motor function recovery and the mechanical pain threshold of the affected forelimb. The morphological appearance of the spinal cord, musculocutaneous nerve, and biceps brachii was assessed by histological staining. Quantitative real-time PCR (qRT‒PCR) was used to measure the mRNA levels of remyelination and regeneration indicators in myocutaneous nerves. The protein levels of inflammatory and pyroptotic indicators in the spinal cord anterior horn were measured using Western blotting. RESULTS NAG significantly accelerated the recovery of motor function in the injured forelimbs, enhanced motoneuronal survival in the anterior horn of the spinal cord, inhibited the expression of proinflammatory cytokines and pyroptosis pathway factors, facilitated axonal remyelination in the myocutaneous nerve and alleviated atrophy of the biceps brachii. Additionally, NAG attenuated neuropathic pain following BPRA. CONCLUSION NAG promotes functional motor recovery and alleviates neuropathic pain by enhancing motoneuronal survival and axonal remyelination and inhibiting the pyroptosis pathway after BPRA in rats, laying the foundation for the use of NAG as a novel treatment for BPRA.
Collapse
Affiliation(s)
- Lin Wu
- Department of Neurology, Multi-Omics Research Center for Brain Disorders, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hunan, 421001, Hengyang, China
- Clinical Research Center for Immune-Related Encephalopathy of Hunan Province (The First Affiliated Hospital), Hengyang Medical School, University of South China, Hunan, 421001, Hengyang, China
- Department of Neurology, The Second Affiliated Hospital, Hengyang Medical School, University of South China, Hunan, 421000, Hengyang, People's Republic of China
| | - Shuangxi Chen
- Department of Neurology, Multi-Omics Research Center for Brain Disorders, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hunan, 421001, Hengyang, China
- Clinical Research Center for Immune-Related Encephalopathy of Hunan Province (The First Affiliated Hospital), Hengyang Medical School, University of South China, Hunan, 421001, Hengyang, China
| | - Bing He
- Department of Neurology, Multi-Omics Research Center for Brain Disorders, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hunan, 421001, Hengyang, China
- Clinical Research Center for Immune-Related Encephalopathy of Hunan Province (The First Affiliated Hospital), Hengyang Medical School, University of South China, Hunan, 421001, Hengyang, China
| | - Guijuan Zhou
- Department of Neurology, Multi-Omics Research Center for Brain Disorders, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hunan, 421001, Hengyang, China
- Clinical Research Center for Immune-Related Encephalopathy of Hunan Province (The First Affiliated Hospital), Hengyang Medical School, University of South China, Hunan, 421001, Hengyang, China
| | - Yan Xu
- Department of Neurology, Multi-Omics Research Center for Brain Disorders, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hunan, 421001, Hengyang, China
- Clinical Research Center for Immune-Related Encephalopathy of Hunan Province (The First Affiliated Hospital), Hengyang Medical School, University of South China, Hunan, 421001, Hengyang, China
| | - Guanghua Zhu
- Department of Neurology, Multi-Omics Research Center for Brain Disorders, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hunan, 421001, Hengyang, China
- Clinical Research Center for Immune-Related Encephalopathy of Hunan Province (The First Affiliated Hospital), Hengyang Medical School, University of South China, Hunan, 421001, Hengyang, China
| | - Juan Xie
- Department of Neurology, Multi-Omics Research Center for Brain Disorders, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hunan, 421001, Hengyang, China
- Clinical Research Center for Immune-Related Encephalopathy of Hunan Province (The First Affiliated Hospital), Hengyang Medical School, University of South China, Hunan, 421001, Hengyang, China
| | - Limin Deng
- Department of Neurology, Multi-Omics Research Center for Brain Disorders, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hunan, 421001, Hengyang, China
- Clinical Research Center for Immune-Related Encephalopathy of Hunan Province (The First Affiliated Hospital), Hengyang Medical School, University of South China, Hunan, 421001, Hengyang, China
| | - Xuanwei Wen
- Department of Neurology, Multi-Omics Research Center for Brain Disorders, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hunan, 421001, Hengyang, China
- Clinical Research Center for Immune-Related Encephalopathy of Hunan Province (The First Affiliated Hospital), Hengyang Medical School, University of South China, Hunan, 421001, Hengyang, China
| | - Sijing Li
- Department of Neurology, Multi-Omics Research Center for Brain Disorders, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hunan, 421001, Hengyang, China
- Clinical Research Center for Immune-Related Encephalopathy of Hunan Province (The First Affiliated Hospital), Hengyang Medical School, University of South China, Hunan, 421001, Hengyang, China
| | - Zijian Xiao
- Department of Neurology, Multi-Omics Research Center for Brain Disorders, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hunan, 421001, Hengyang, China.
- Clinical Research Center for Immune-Related Encephalopathy of Hunan Province (The First Affiliated Hospital), Hengyang Medical School, University of South China, Hunan, 421001, Hengyang, China.
| |
Collapse
|
4
|
Delfino G, Bénardais K, Graff J, Samama B, Antal MC, Ghandour MS, Boehm N. Oligodendroglial primary cilium heterogeneity during development and demyelination/remyelination. Front Cell Neurosci 2022; 16:1049468. [PMID: 36505511 PMCID: PMC9729284 DOI: 10.3389/fncel.2022.1049468] [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: 09/20/2022] [Accepted: 11/03/2022] [Indexed: 11/25/2022] Open
Abstract
The primary cilium (PC) has emerged as an indispensable cellular antenna essential for signal transduction of important cell signaling pathways. The rapid acquisition of knowledge about PC biology has raised attention to PC as a therapeutic target in some neurological and psychiatric diseases. However, the role of PC in oligodendrocytes and its participation in myelination/remyelination remain poorly understood. Oligodendrocyte precursor cells (OPCs) give rise to oligodendrocytes during central nervous system (CNS) development. In adult, a small percentage of OPCs remains as undifferentiated cells located sparsely in the different regions of the CNS. These cells can regenerate oligodendrocytes and participate to certain extent in remyelination. This study aims characterize PC in oligodendrocyte lineage cells during post-natal development and in a mouse model of demyelination/remyelination. We show heterogeneity in the frequency of cilium presence on OPCs, depending on culture conditions in vitro and cerebral regions in vivo during development and demyelination/remyelination. In vitro, Lithium chloride (LiCl), Forskolin and Chloral Hydrate differentially affect cilium, depending on culture environment and PC length correlates with the cell differentiation state. Beside the role of PC as a keeper of cell proliferation, our results suggest its involvement in myelination/remyelination.
Collapse
Affiliation(s)
- Giada Delfino
- ICube Laboratory UMR 7357, Team IMIS, Strasbourg, France,Institut d’Histologie, Service Central de Microscopie Electronique, Faculté de Médecine, Université de Strasbourg, Strasbourg, France,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France,*Correspondence: Giada Delfino,
| | - Karelle Bénardais
- ICube Laboratory UMR 7357, Team IMIS, Strasbourg, France,Institut d’Histologie, Service Central de Microscopie Electronique, Faculté de Médecine, Université de Strasbourg, Strasbourg, France,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France,Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Julien Graff
- Institut d’Histologie, Service Central de Microscopie Electronique, Faculté de Médecine, Université de Strasbourg, Strasbourg, France,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Brigitte Samama
- ICube Laboratory UMR 7357, Team IMIS, Strasbourg, France,Institut d’Histologie, Service Central de Microscopie Electronique, Faculté de Médecine, Université de Strasbourg, Strasbourg, France,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France,Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Maria Cristina Antal
- ICube Laboratory UMR 7357, Team IMIS, Strasbourg, France,Institut d’Histologie, Service Central de Microscopie Electronique, Faculté de Médecine, Université de Strasbourg, Strasbourg, France,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France,Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - M. Said Ghandour
- ICube Laboratory UMR 7357, Team IMIS, Strasbourg, France,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France
| | - Nelly Boehm
- ICube Laboratory UMR 7357, Team IMIS, Strasbourg, France,Institut d’Histologie, Service Central de Microscopie Electronique, Faculté de Médecine, Université de Strasbourg, Strasbourg, France,Fédération de Médecine Translationnelle de Strasbourg (FMTS), Strasbourg, France,Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| |
Collapse
|
5
|
Balçıkanlı Z, Culha I, Dilsiz P, Aydin MS, Ates N, Beker MC, Baltaci SB, Koc HI, Yigitbasi A, Gündogar M, Doeppner TR, Hermann DM, Kilic E. Lithium promotes long-term neurological recovery after spinal cord injury in mice by enhancing neuronal survival, gray and white matter remodeling, and long-distance axonal regeneration. Front Cell Neurosci 2022; 16:1012523. [PMID: 36439202 PMCID: PMC9693752 DOI: 10.3389/fncel.2022.1012523] [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: 08/05/2022] [Accepted: 10/25/2022] [Indexed: 09/20/2023] Open
Abstract
Spinal cord injury (SCI) induces neurological deficits associated with long-term functional impairments. Since the current treatments remain ineffective, novel therapeutic options are needed. Besides its effect on bipolar mood disorder, lithium was reported to have neuroprotective activity in different neurodegenerative conditions, including SCI. In SCI, the effects of lithium on long-term neurological recovery and neuroplasticity have not been assessed. We herein investigated the effects of intraperitoneally administered lithium chloride (LiCl) on motor coordination recovery, electromyography (EMG) responses, histopathological injury and remodeling, and axonal plasticity in mice exposed to spinal cord transection. At a dose of 0.2, but not 2.0 mmol/kg, LiCl enhanced motor coordination and locomotor activity starting at 28 days post-injury (dpi), as assessed by a set of behavioral tests. Following electrical stimulation proximal to the hemitransection, LiCl at 0.2 mmol/kg decreased the latency and increased the amplitude of EMG responses in the denervated hindlimb at 56 dpi. Functional recovery was associated with reduced gray and white matter atrophy rostral and caudal to the hemitransection, increased neuronal survival and reduced astrogliosis in the dorsal and ventral horns caudal to the hemitransection, and increased regeneration of long-distance axons proximal and distal to the lesion site in mice receiving 0.2 mmol/kg, but not 2 mmol/kg LiCl, as assessed by histochemical and immunohistochemical studies combined with anterograde tract tracing. Our results indicate that LiCl induces long-term neurological recovery and neuroplasticity following SCI.
Collapse
Affiliation(s)
- Zeynep Balçıkanlı
- Department of Physiology, Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Istanbul, Turkey
- Department of Physiology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Irem Culha
- Department of Physiology, Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Istanbul, Turkey
- Department of Physiology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Pelin Dilsiz
- Department of Physiology, Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Istanbul, Turkey
- Department of Physiology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Mehmet Serif Aydin
- Department of Physiology, Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Istanbul, Turkey
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, Turkey
| | - Nilay Ates
- Department of Physiology, Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Istanbul, Turkey
- Department of Pharmacology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Mustafa Caglar Beker
- Department of Physiology, Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Istanbul, Turkey
- Department of Physiology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Saltuk Bugra Baltaci
- Department of Physiology, Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Istanbul, Turkey
- Department of Physiology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Halil I. Koc
- Department of Physiology, Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Istanbul, Turkey
- Department of Physiology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| | - Ahmet Yigitbasi
- Department of Hematology, Medical Faculty, Trakya University, Edirne, Turkey
| | - Mustafa Gündogar
- Department of Physiology, Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Istanbul, Turkey
- Department of Endodontics, Faculty of Dentistry, Istanbul Medipol University, Istanbul, Turkey
| | - Thorsten R. Doeppner
- Department of Physiology, Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Istanbul, Turkey
- Department of Neurology, University Hospital Gießen, Göttingen, Germany
| | - Dirk M. Hermann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ertugrul Kilic
- Department of Physiology, Regenerative and Restorative Medical Research Center, Istanbul Medipol University, Istanbul, Turkey
- Department of Physiology, School of Medicine, Istanbul Medipol University, Istanbul, Turkey
| |
Collapse
|
6
|
Farmani AR, Salmeh MA, Golkar Z, Moeinzadeh A, Ghiasi FF, Amirabad SZ, Shoormeij MH, Mahdavinezhad F, Momeni S, Moradbeygi F, Ai J, Hardy JG, Mostafaei A. Li-Doped Bioactive Ceramics: Promising Biomaterials for Tissue Engineering and Regenerative Medicine. J Funct Biomater 2022; 13:162. [PMID: 36278631 PMCID: PMC9589997 DOI: 10.3390/jfb13040162] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/18/2022] [Accepted: 09/20/2022] [Indexed: 12/03/2022] Open
Abstract
Lithium (Li) is a metal with critical therapeutic properties ranging from the treatment of bipolar depression to antibacterial, anticancer, antiviral and pro-regenerative effects. This element can be incorporated into the structure of various biomaterials through the inclusion of Li chloride/carbonate into polymeric matrices or being doped in bioceramics. The biocompatibility and multifunctionality of Li-doped bioceramics present many opportunities for biomedical researchers and clinicians. Li-doped bioceramics (capable of immunomodulation) have been used extensively for bone and tooth regeneration, and they have great potential for cartilage/nerve regeneration, osteochondral repair, and wound healing. The synergistic effect of Li in combination with other anticancer drugs as well as the anticancer properties of Li underline the rationale that bioceramics doped with Li may be impactful in cancer treatments. The role of Li in autophagy may explain its impact in regenerative, antiviral, and anticancer research. The combination of Li-doped bioceramics with polymers can provide new biomaterials with suitable flexibility, especially as bio-ink used in 3D printing for clinical applications of tissue engineering. Such Li-doped biomaterials have significant clinical potential in the foreseeable future.
Collapse
Affiliation(s)
- Ahmad Reza Farmani
- Tissue Engineering and Applied Cell Sciences Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran 14166-34793, Iran
- Tissue Engineering Department, School of Advanced Technologies in Medicine, Fasa University of Medical Sciences, Fasa 74615-168, Iran
- Students’ Scientific Research Center, Tehran University of Medical Sciences, Tehran 14166-34793, Iran
| | - Mohammad Ali Salmeh
- Department of Biotechnology, School of Chemical Engineering, College of Engineering, University of Tehran, Tehran 14155-6619, Iran
| | - Zahra Golkar
- Department of Midwifery, Firoozabad Branch, Islamic Azad University, Firoozabad 74715-117, Iran
| | - Alaa Moeinzadeh
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14496-14535, Iran
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran 14496-14535, Iran
| | - Farzaneh Farid Ghiasi
- Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran 14496-14535, Iran
| | - Sara Zamani Amirabad
- Department of Chemical Engineering, Faculty of Engineering, Yasouj University, Yasouj 75918-74934, Iran
| | - Mohammad Hasan Shoormeij
- Emergency Medicine Department, Shariati Hospital, Tehran University of Medical Sciences, Tehran 14166-34793, Iran
| | - Forough Mahdavinezhad
- Anatomy Department, School of Medicine, Tehran University of Medical Sciences, Tehran 14166-34793, Iran
- Department of Infertility, Velayat Hospital, Qazvin University of Medical Sciences, Qazvin 34199-15315, Iran
| | - Simin Momeni
- Chemistry Department, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz 83151-61355, Iran
| | - Fatemeh Moradbeygi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz 71348-14336, Iran
| | - Jafar Ai
- Tissue Engineering and Applied Cell Sciences Department, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran 14166-34793, Iran
| | - John G. Hardy
- Department of Chemistry, Faraday Building, Lancaster University, Lancaster LA1 4YB, UK
- Materials Science Institute, Lancaster University, Lancaster LA1 4YW, UK
| | - Amir Mostafaei
- Department of Mechanical, Materials, and Aerospace Engineering, Illinois Institute of Technology, 10 W 32nd Street, Chicago, IL 60616, USA
| |
Collapse
|
7
|
Nicoletti VG, Pajer K, Calcagno D, Pajenda G, Nógrádi A. The Role of Metals in the Neuroregenerative Action of BDNF, GDNF, NGF and Other Neurotrophic Factors. Biomolecules 2022; 12:biom12081015. [PMID: 35892326 PMCID: PMC9330237 DOI: 10.3390/biom12081015] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 12/14/2022] Open
Abstract
Mature neurotrophic factors and their propeptides play key roles ranging from the regulation of neuronal growth and differentiation to prominent participation in neuronal survival and recovery after injury. Their signaling pathways sculpture neuronal circuits during brain development and regulate adaptive neuroplasticity. In addition, neurotrophic factors provide trophic support for damaged neurons, giving them a greater capacity to survive and maintain their potential to regenerate their axons. Therefore, the modulation of these factors can be a valuable target for treating or preventing neurologic disorders and age-dependent cognitive decline. Neuroregenerative medicine can take great advantage by the deepening of our knowledge on the molecular mechanisms underlying the properties of neurotrophic factors. It is indeed an intriguing topic that a significant interplay between neurotrophic factors and various metals can modulate the outcome of neuronal recovery. This review is particularly focused on the roles of GDNF, BDNF and NGF in motoneuron survival and recovery from injuries and evaluates the therapeutic potential of various neurotrophic factors in neuronal regeneration. The key role of metal homeostasis/dyshomeostasis and metal interaction with neurotrophic factors on neuronal pathophysiology is also highlighted as a novel mechanism and potential target for neuronal recovery. The progress in mechanistic studies in the field of neurotrophic factor-mediated neuroprotection and neural regeneration, aiming at a complete understanding of integrated pathways, offers possibilities for the development of novel neuroregenerative therapeutic approaches.
Collapse
Affiliation(s)
- Vincenzo Giuseppe Nicoletti
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), Section of Medical Biochemistry, University of Catania, 95124 Catania, Italy; (V.G.N.); (D.C.)
| | - Krisztián Pajer
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary;
| | - Damiano Calcagno
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), Section of Medical Biochemistry, University of Catania, 95124 Catania, Italy; (V.G.N.); (D.C.)
| | - Gholam Pajenda
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, Research Centre for Traumatology of the Austrian Workers, 1200 Vienna, Austria;
- Department for Trauma Surgery, Medical University Vienna, 1090 Vienna, Austria
| | - Antal Nógrádi
- Department of Anatomy, Histology and Embryology, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary;
- Correspondence: ; Tel.: +36-6-234-2855
| |
Collapse
|
8
|
Souza NM, Gonçalves MF, Ferreira LFR, Bilal M, Iqbal HMN, Soriano RN. Revisiting the Role of Biologically Active Natural and Synthetic Compounds as an Intervention to Treat Injured Nerves. Mol Neurobiol 2021; 58:4980-4998. [PMID: 34228268 DOI: 10.1007/s12035-021-02473-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 06/24/2021] [Indexed: 02/08/2023]
Abstract
Traumatic lesions in nerves present high incidence and may culminate in sensorimotor and/or autonomic dysfunctions or a total loss of function, affecting the patient's quality of life. Although the microenvironment favors peripheral nerve regeneration, the regenerative process is not always successful. Some herbs, natural products, and synthetic drugs have been studied as potential pro-regenerative interventions. We reviewed and discussed the most recent articles published over the last ten years in high impact factor journals. Even though most of the articles contemplated in this review were in vitro and animal model studies, those with herbs showed promising results. Most of them presented antioxidant and anti-inflammatory effects. Drugs of several pharmacological classes also showed optimistic outcomes in nerve functional recovery, including clinical trials. The results are hopeful; however, mechanisms of action need to be elucidated, and there is a need for more high-quality clinical studies. The study presents careful compilation of findings of dozens of compounds with consistent pro-regenerative evidence published in respected scientific journals. It may be valuable for health professionals and researchers in the field.
Collapse
Affiliation(s)
- Natália Melo Souza
- Department of Medicine, Federal University of Juiz de Fora, Governador Valadares, Minas Gerais, 35032-620, Brazil
| | - Mateus Figueiredo Gonçalves
- Department of Medicine, Federal University of Juiz de Fora, Governador Valadares, Minas Gerais, 35032-620, Brazil
| | - Luiz Fernando Romanholo Ferreira
- Graduate Program in Process Engineering, Tiradentes University (UNIT), Av. Murilo Dantas, Aracaju Sergipe, Farolândia, 30049032-490, Brazil
- Institute of Technology and Research (ITP), Tiradentes University (UNIT), Av. Murilo Dantas, Aracaju-Sergipe, Farolândia, 30049032-490, Brazil
| | - Muhammad Bilal
- School of Life Science and Food Engineering, Huaiyin Institute of Technology, Huaian, 223003, China.
| | - Hafiz M N Iqbal
- Tecnologico de Monterrey, School of Engineering and Sciences, 64849, Monterrey, Mexico.
| | - Renato Nery Soriano
- Division of Physiology and Biophysics, Department of Basic Life Sciences, Federal University of Juiz de Fora, Governador Valadares, Minas Gerais, 35010-177, Brazil.
| |
Collapse
|
9
|
Kuffler DP. Can lithium enhance the extent of axon regeneration and neurological recovery following peripheral nerve trauma? Neural Regen Res 2021; 17:948-952. [PMID: 34558506 PMCID: PMC8552832 DOI: 10.4103/1673-5374.324830] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The clinical “gold standard” technique for attempting to restore function to nerves with a gap is to bridge the gap with sensory autografts. However, autografts induce good to excellent recovery only across short nerve gaps, in young patients, and when repairs are performed a short time post nerve trauma. Even under the best of conditions, < 50% of patients recover good recovery. Although many alternative techniques have been tested, none is as effective as autografts. Therefore, alternative techniques are required that increase the percentage of patients who recover function and the extent of their recovery. This paper examines the actions of lithium, and how it appears to trigger all the cellular and molecular events required to promote axon regeneration, and how both in animal models and clinically, lithium administration enhances both the extent of axon regeneration and neurological recovery. The paper proposes more extensive clinical testing of lithium for its ability and reliability to increase the extent of axon regeneration and functional recovery.
Collapse
Affiliation(s)
- Damien P Kuffler
- Institute of Neurobiology, Medical Sciences Campus, University of Puerto Rico, San Juan, PR, USA
| |
Collapse
|
10
|
Samarghandian S, Pourbagher-Shahri AM, Ashrafizadeh M, Khan H, Forouzanfar F, Aramjoo H, Farkhondeh T. A Pivotal Role of the Nrf2 Signaling Pathway in Spinal Cord Injury: A Prospective Therapeutics Study. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 19:207-219. [PMID: 32496994 DOI: 10.2174/1871527319666200604175118] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 02/27/2020] [Accepted: 04/11/2020] [Indexed: 12/15/2022]
Abstract
The nuclear erythroid 2-related factor 2 (Nrf2) signaling pathway has a main role against oxidative stress and inflammation. Spinal Cord Injury (SCI) leads to the high secretion of inflammatory cytokines and reactive oxygen species, which disturbs nervous system function and regeneration. Several studies have indicated that the activation of the Nrf2 signaling pathway may be effective against inflammation after SCI. The experimental studies have indicated that many chemical and natural agents act as Nrf2 inducer, which inhibits the SCI progression. Thus, the finding of novel Nrf2- inducer anti-inflammatory agents may be a valuable approach in drug discovery. In the present review, we discussed the Nrf2 signal pathway and crosstalk with the NF-κB pathway and also the impact of this pathway on inflammation in animal models of SCI. Furthermore, we discussed the regulation of Nrf2 by several phytochemicals and drugs, as well as their effects on the SCI inhibition. Therefore, the current study presented a new hypothesis of the development of anti-inflammatory agents that mediate the Nrf2 signaling pathway for treating the SCI outcomes.
Collapse
Affiliation(s)
- Saeed Samarghandian
- Healthy Ageing Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | | | - Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University, Mardan 23200, Pakistan
| | - Fatemeh Forouzanfar
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamed Aramjoo
- Student Research Committee, Lab Sciences Technology, Birjand University of Medical Sciences, Birjand, Iran
| | - Tahereh Farkhondeh
- Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| |
Collapse
|
11
|
Gaja-Capdevila N, Hernández N, Zamanillo D, Vela JM, Merlos M, Navarro X, Herrando-Grabulosa M. Neuroprotective Effects of Sigma 1 Receptor Ligands on Motoneuron Death after Spinal Root Injury in Mice. Int J Mol Sci 2021; 22:6956. [PMID: 34203381 PMCID: PMC8269081 DOI: 10.3390/ijms22136956] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/22/2021] [Accepted: 06/24/2021] [Indexed: 12/28/2022] Open
Abstract
Loss of motor neurons (MNs) after spinal root injury is a drawback limiting the recovery after palliative surgery by nerve or muscle transfers. Research based on preventing MN death is a hallmark to improve the perspectives of recovery following severe nerve injuries. Sigma-1 receptor (Sig-1R) is a protein highly expressed in MNs, proposed as neuroprotective target for ameliorating MN degenerative conditions. Here, we used a model of L4-L5 rhizotomy in adult mice to induce MN degeneration and to evaluate the neuroprotective role of Sig-1R ligands (PRE-084, SA4503 and BD1063). Lumbar spinal cord was collected at 7, 14, 28 and 42 days post-injury (dpi) for immunohistochemistry, immunofluorescence and Western blot analyses. This proximal axotomy at the immediate postganglionic level resulted in significant death, up to 40% of spinal MNs at 42 days after injury and showed markedly increased glial reactivity. Sig-1R ligands PRE-084, SA4503 and BD1063 reduced MN loss by about 20%, associated to modulation of endoplasmic reticulum stress markers IRE1α and XBP1. These pathways are Sig-1R specific since they were not produced in Sig-1R knockout mice. These findings suggest that Sig-1R is a promising target for the treatment of MN cell death after neural injuries.
Collapse
Affiliation(s)
- Núria Gaja-Capdevila
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona, 01893 Bellaterra, Spain; (N.G.-C.); (N.H.); (X.N.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Neus Hernández
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona, 01893 Bellaterra, Spain; (N.G.-C.); (N.H.); (X.N.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Daniel Zamanillo
- Welab, Parc Científic Barcelona, 08028 Barcelona, Spain; (D.Z.); (J.M.V.); (M.M.)
| | - Jose Miguel Vela
- Welab, Parc Científic Barcelona, 08028 Barcelona, Spain; (D.Z.); (J.M.V.); (M.M.)
| | - Manuel Merlos
- Welab, Parc Científic Barcelona, 08028 Barcelona, Spain; (D.Z.); (J.M.V.); (M.M.)
| | - Xavier Navarro
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona, 01893 Bellaterra, Spain; (N.G.-C.); (N.H.); (X.N.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
- Institut Guttmann Hospital de Neurorehabilitació, 08916 Badalona, Spain
| | - Mireia Herrando-Grabulosa
- Department of Cell Biology, Physiology and Immunology, Institute of Neurosciences, Universitat Autònoma de Barcelona, 01893 Bellaterra, Spain; (N.G.-C.); (N.H.); (X.N.)
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| |
Collapse
|
12
|
Huang C, Fu C, Qi ZP, Guo WL, You D, Li R, Zhu Z. Localised delivery of quercetin by thermo-sensitive PLGA-PEG-PLGA hydrogels for the treatment of brachial plexus avulsion. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2021; 48:1010-1021. [PMID: 32608269 DOI: 10.1080/21691401.2020.1770265] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Accumulating evidence indicates that oxidative stress and inflammation are implicated in brachial plexus avulsion (BPA). Quercetin has anti-inflammatory, anti-oxidant, anti-apoptotic, and neuroprotective properties. This study investigated the therapeutic efficacy of a temperature-sensitive poly(D,L-lactide-co-glycolide)-poly(ethylene-glycol)-poly(D,L-lactide-co-glycolide) (PLGA-PEG-PLGA) hydrogel sustained-release system of quercetin in BPA. In situ injections of the hydrogel loaded with different concentrations of quercetin were conducted in a rat model of BPA. Significantly reduced reactive oxygen species and interleukin-6 levels in the injured spinal cord 24 h post-surgery, increased number of anterior horn motor and functional neurons in the spinal cord 6 weeks post-surgery, thickened biceps muscle fibres and enlarged endplate area with clear structure, reduced demyelinated peripheral nerves, and significantly increased Terzis grooming test scores were found in the groups with 50 or 100 mg/mL quercetin-loaded hydrogels compared with the control and blank hydrogel groups. In conclusion, the temperature-sensitive quercetin loaded PLGA-PEG-PLGA hydrogel sustained-release system can alleviate oxidative damage and inflammation in the spinal cord, increase neuron survival rate, and promote nerve regeneration and motor function recovery in rats with early BPA. The findings suggest that this drug-loaded hydrogel has potential applications in the clinical treatment of BPA.
Collapse
Affiliation(s)
- Chao Huang
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Chuan Fu
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Zhi-Ping Qi
- Department of Orthopedic Surgery, The Second Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Wen-Lai Guo
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Di You
- Department of Anesthesiology, The Sino-Japanese Friendship Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Rui Li
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, Jilin, P. R. China
| | - Zhe Zhu
- Department of Hand Surgery, The Second Hospital of Jilin University, Changchun, Jilin, P. R. China
| |
Collapse
|
13
|
Lithium alleviated spinal cord injury (SCI)-induced apoptosis and inflammation in rats via BDNF-AS/miR-9-5p axis. Cell Tissue Res 2021; 384:301-312. [PMID: 33464390 DOI: 10.1007/s00441-020-03298-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 09/14/2020] [Indexed: 12/14/2022]
Abstract
Spinal cord injury (SCI) is a major cause of paralysis, disability and even death in severe cases. Lithium has neuroprotective effects on SCI, while the underlying mechanisms remain obscure. In the present study, we established a SCI rat model, which subsequently received lithium treatment. Results displayed that lithium treatment improved the locomotor function recovery and reduced apoptosis by increasing anti-apoptotic molecule expression and decreasing pro-apoptotic factor expression in SCI rats. Furthermore, lithium treatment alleviated the inflammatory response by inactivating the nuclear factor-kappa B (NF-κB) pathway and inhibited the expression of lncRNA brain-derived neurotrophic factor antisense (BDNF-AS) in SCI rats. Subsequent researches indicated that miR-9-5p was targeted and regulated by BDNF-AS. Lithium treatment rescued the upregulation of BDNF-AS expression and downregulation of miR-9-5p expression induced by H2O2 in SH-SY5Y cells. BDNF-AS overexpression or miR-9-5p interference attenuated the anti-apoptotic and anti-inflammatory effects of lithium chloride in SH-SY5Y cells that was damaged by H2O2 induction, revealing that lithium might act through the BDNF-AS/miR-9-5p axis. In vivo studies showed that the injection of BDNF-AS adenovirus vector or miR-9-5p inhibitor reversed the effects of lithium on the histologic morphology of spinal cord, motor function, inflammatory reaction and apoptosis in SCI rats, which was consistent with the results of in vitro studies. In conclusion, our data demonstrated that lithium reduced SCI-induced apoptosis and inflammation in rats via the BDNF-AS/miR-9-5p axis.
Collapse
|
14
|
Wang J, Kuang X, Peng Z, Li C, Guo C, Fu X, Wu J, Luo Y, Rao X, Zhou X, Huang B, Tang W, Tang Y. EGCG treats ICH via up-regulating miR-137-3p and inhibiting Parthanatos. Transl Neurosci 2020; 11:371-379. [PMID: 33335777 PMCID: PMC7718614 DOI: 10.1515/tnsci-2020-0143] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/24/2020] [Accepted: 09/09/2020] [Indexed: 12/21/2022] Open
Abstract
Intracranial hemorrhage (ICH) causes high mortality and disability without effective treatment in the clinical setting. (-)-Epigallocatechin-3-gallate (EGCG) exerts an essential role in the central nervous system and offers a promising therapeutic agent for the treatment of oxidative damage-related diseases. MiR-137 can inhibit the oxidative stress and apoptosis to attenuate neuronal injury. However, the role of EGCG in regulating miR-137-3p and neuronal Parthanatos remains to be unclear. In the present study, we build the ICH mice model to investigate the antioxidant effects of EGCG via upregulating miR-137-3p and inhibiting neuronal Parthanatos. We revealed that EGCG upregulated miR-137-3p and inhibited neuronal Parthanatos, and promoted the functional recovery, alleviated ICH-induced brain injury, and reduced oxidative stress in mice following ICH. However, following the inhibition of miR-137-3p and activation of Parthanatos, EGCG was unable to exert neuroprotective roles. These combined results suggest that EGCG may upregulate miR-137-3p and inhibit neuronal Parthanatos to accelerate functional recovery in mice after ICH, laying the foundation for EGCG to be a novel strategy for the treatment of neuronal injuries related to Parthanatos.
Collapse
Affiliation(s)
- Jianjun Wang
- Affiliated hospital, Xiangnan University, Chenzhou, 423000, Hunan Province, China
- Department of Clinical, Xiangnan University, Chenzhou, 423000, Hunan Province, China
| | - Xuejun Kuang
- Affiliated hospital, Xiangnan University, Chenzhou, 423000, Hunan Province, China
| | - Zhao Peng
- Affiliated hospital, Xiangnan University, Chenzhou, 423000, Hunan Province, China
| | - Conghui Li
- Affiliated hospital, Xiangnan University, Chenzhou, 423000, Hunan Province, China
| | - Chengwu Guo
- Department of Basic Medical Sciences, Xiangnan University, Chenzhou, 423000, Hunan Province, China
| | - Xi Fu
- Department of Basic Medical Sciences, Xiangnan University, Chenzhou, 423000, Hunan Province, China
| | - Junhong Wu
- Department of Basic Medical Sciences, Xiangnan University, Chenzhou, 423000, Hunan Province, China
| | - Yang Luo
- Department of Basic Medical Sciences, Xiangnan University, Chenzhou, 423000, Hunan Province, China
| | - Xiaolin Rao
- Department of Basic Medical Sciences, Xiangnan University, Chenzhou, 423000, Hunan Province, China
| | - Xiangjuan Zhou
- Department of Basic Medical Sciences, Xiangnan University, Chenzhou, 423000, Hunan Province, China
| | - Bin Huang
- Department of Basic Medical Sciences, Xiangnan University, Chenzhou, 423000, Hunan Province, China
| | - Weijun Tang
- Department of Pharmacy, Xiangnan University, Chenzhou, 423000, Hunan Province, China
| | - Yinjuan Tang
- Department of Basic Medical Sciences, Xiangnan University, Chenzhou, 423000, Hunan Province, China
| |
Collapse
|
15
|
Chen S, He B, Zhou G, Xu Y, Wu L, Xie Y, Li Y, Chen S, Huang J, Wu H, Xiao Z. Berberine enhances L1 expression and axonal remyelination in rats after brachial plexus root avulsion. Brain Behav 2020; 10:e01792. [PMID: 32770668 PMCID: PMC7559605 DOI: 10.1002/brb3.1792] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 06/17/2020] [Accepted: 07/19/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE Enhanced remyelination of the regenerated axons results in functional re-innervation and improved functional motor recovery after brachial plexus root avulsion (BPRA). The neural cell adhesion molecule L1 (L1CAM, L1) regulates myelination and promotes regeneration after acute injury in the nervous system. Berberine (BBR) can exert neuroprotective roles against the lesion. Herein, we investigated whether berberine (BBR) can affect the expression of L1 and enhance the axonal remyelination in rats following BPRA. METHODS The surgical procedures were performed to build the rat brachial plexus avulsion and re-implantation model, and then, the rats were treated with BBR. After the rehabilitation for 12 weeks, the musculocutaneous nerves were collected for quantitative real-time PCR, Western blot analysis, and histochemical and immunofluorescence staining. RESULTS We observed that, BBR treatment ameliorated the abnormal musculocutaneous nerve fibers morphology, up-regulated the L1 expression, increased the myelination-related genes, decreased the differentiated-associated genes, and up-regulated the phosphorylation of ERK. CONCLUSION These results suggest that BBR may enhance L1 expression and promote axonal remyelination after spinal root avulsion.
Collapse
Affiliation(s)
- Shuangxi Chen
- The First Affiliated Hospital, University of South China, Hengyang, China
| | - Bing He
- The First Affiliated Hospital, University of South China, Hengyang, China
| | - Guijuan Zhou
- The First Affiliated Hospital, University of South China, Hengyang, China
| | - Yan Xu
- The First Affiliated Hospital, University of South China, Hengyang, China
| | - Lin Wu
- The First Affiliated Hospital, University of South China, Hengyang, China
| | - Yangzhi Xie
- The First Affiliated Hospital, University of South China, Hengyang, China.,Leiyang People's Hospital, Leiyang, China
| | - Yihui Li
- The First Affiliated Hospital, University of South China, Hengyang, China
| | - Shuangqin Chen
- The First Affiliated Hospital, University of South China, Hengyang, China
| | - Jianghua Huang
- The First Affiliated Hospital, University of South China, Hengyang, China
| | - Heng Wu
- The First Affiliated Hospital, University of South China, Hengyang, China
| | - Zijian Xiao
- The First Affiliated Hospital, University of South China, Hengyang, China
| |
Collapse
|
16
|
Gu XK, Li XR, Lu ML, Xu H. Lithium promotes proliferation and suppresses migration of Schwann cells. Neural Regen Res 2020; 15:1955-1961. [PMID: 32246645 PMCID: PMC7513976 DOI: 10.4103/1673-5374.280324] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/05/2019] [Accepted: 12/17/2019] [Indexed: 12/12/2022] Open
Abstract
Schwann cell proliferation, migration and remyelination of regenerating axons contribute to regeneration after peripheral nervous system injury. Lithium promotes remyelination by Schwann cells and improves peripheral nerve regeneration. However, whether lithium modulates other phenotypes of Schwann cells, especially their proliferation and migration remains elusive. In the current study, primary Schwann cells from rat sciatic nerve stumps were cultured and exposed to 0, 5, 10, 15, or 30 mM lithium chloride (LiCl) for 24 hours. The effects of LiCl on Schwann cell proliferation and migration were examined using the Cell Counting Kit-8, 5-ethynyl-2'-deoxyuridine, Transwell and wound healing assays. Cell Counting Kit-8 and 5-ethynyl-2'-deoxyuridine assays showed that 5, 10, 15, and 30 mM LiCl significantly increased the viability and proliferation rate of Schwann cells. Transwell-based migration assays and wound healing assays showed that 10, 15, and 30 mM LiCl suppressed the migratory ability of Schwann cells. Furthermore, the effects of LiCl on the proliferation and migration phenotypes of Schwann cells were mostly dose-dependent. These data indicate that lithium treatment significantly promotes the proliferation and inhibits the migratory ability of Schwann cells. This conclusion will inform strategies to promote the repair and regeneration of peripheral nerves. All of the animal experiments in this study were ethically approved by the Administration Committee of Experimental Animal Center of Nantong University, China (approval No. 20170320-017) on March 2, 2017.
Collapse
Affiliation(s)
- Xiao-Kun Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
- Department of Hand Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Xin-Rui Li
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Mei-Ling Lu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu Province, China
| | - Hui Xu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
17
|
Gouvêa-Junqueira D, Falvella ACB, Antunes ASLM, Seabra G, Brandão-Teles C, Martins-de-Souza D, Crunfli F. Novel Treatment Strategies Targeting Myelin and Oligodendrocyte Dysfunction in Schizophrenia. Front Psychiatry 2020; 11:379. [PMID: 32425837 PMCID: PMC7203658 DOI: 10.3389/fpsyt.2020.00379] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 04/15/2020] [Indexed: 12/12/2022] Open
Abstract
Oligodendrocytes are the glial cells responsible for the formation of the myelin sheath around axons. During neurodevelopment, oligodendrocytes undergo maturation and differentiation, and later remyelination in adulthood. Abnormalities in these processes have been associated with behavioral and cognitive dysfunctions and the development of various mental illnesses like schizophrenia. Several studies have implicated oligodendrocyte dysfunction and myelin abnormalities in the disorder, together with altered expression of myelin-related genes such as Olig2, CNP, and NRG1. However, the molecular mechanisms subjacent of these alterations remain elusive. Schizophrenia is a severe, chronic psychiatric disorder affecting more than 23 million individuals worldwide and its symptoms usually appear at the beginning of adulthood. Currently, the major therapeutic strategy for schizophrenia relies on the use of antipsychotics. Despite their widespread use, the effects of antipsychotics on glial cells, especially oligodendrocytes, remain unclear. Thus, in this review we highlight the current knowledge regarding oligodendrocyte dysfunction in schizophrenia, compiling data from (epi)genetic studies and up-to-date models to investigate the role of oligodendrocytes in the disorder. In addition, we examined potential targets currently investigated for the improvement of schizophrenia symptoms. Research in this area has been investigating potential beneficial compounds, including the D-amino acids D-aspartate and D-serine, that act as NMDA receptor agonists, modulating the glutamatergic signaling; the antioxidant N-acetylcysteine, a precursor in the synthesis of glutathione, protecting against the redox imbalance; as well as lithium, an inhibitor of glycogen synthase kinase 3β (GSK3β) signaling, contributing to oligodendrocyte survival and functioning. In conclusion, there is strong evidence linking oligodendrocyte dysfunction to the development of schizophrenia. Hence, a better understanding of oligodendrocyte differentiation, as well as the effects of antipsychotic medication in these cells, could have potential implications for understanding the development of schizophrenia and finding new targets for drug development.
Collapse
Affiliation(s)
- Danielle Gouvêa-Junqueira
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Ana Caroline Brambilla Falvella
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - André Saraiva Leão Marcelo Antunes
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Gabriela Seabra
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Caroline Brandão-Teles
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
- Experimental Medicine Research Cluster (EMRC), University of Campinas, Campinas, Brazil
- Instituto Nacional de Biomarcadores em Neuropsiquiatria, Conselho Nacional de Desenvolvimento Científico e Tecnológico, São Paulo, Brazil
- D′Or Institute for Research and Education (IDOR), São Paulo, Brazil
| | - Fernanda Crunfli
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, Campinas, Brazil
| |
Collapse
|
18
|
Multifunctional compounds lithium chloride and methylene Blue attenuate the negative effects of diisopropylfluorophosphate on axonal transport in rat cortical neurons. Toxicology 2020; 431:152379. [PMID: 31962143 DOI: 10.1016/j.tox.2020.152379] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 01/03/2020] [Accepted: 01/18/2020] [Indexed: 12/29/2022]
Abstract
Organophosphates (OPs) are valuable as pesticides in agriculture and for controlling deadly vector-borne illnesses; however, they are highly toxic and associated with many deleterious health effects in humans including long-term neurological impairments. Antidotal treatment regimens are available to combat the symptoms of acute OP toxicity, which result from the irreversible inhibition of acetylcholinesterase (AChE). However, there are no established treatments for the long-term neurological consequences of OP exposure. In addition to AChE, OPs can negatively affect multiple protein targets as well as biological processes such as axonal transport. Given the fundamental nature of axonal transport to neuronal health, we rationalized that this process might serve as a general focus area for novel therapeutic strategies against OP toxicity. In the studies described here, we employed a multi-target, phenotypic screening, and drug repurposing strategy for the evaluations of potential novel OP-treatments using a primary neuronal culture model and time-lapse live imaging microscopy. Two multi-target compounds, lithium chloride (LiCl) and methylene blue (MB), which are FDA-approved for other indications, were evaluated for their ability to prevent the negative effects of the OP, diisopropylfluorophosphate (DFP) on axonal transport. The results indicated that both LiCl and MB prevented DFP-induced impairments in anterograde and retrograde axonal transport velocities in a concentration dependent manner. While in vivo studies will be required to confirm our in vitro findings, these experiments support the potential of LiCl and MB as repurposed drugs for the treatment of the long-term neurological deficits associated with OP exposure (currently an unmet medical need).
Collapse
|
19
|
Reactivation of Denervated Schwann Cells by Embryonic Spinal Cord Neurons to Promote Axon Regeneration and Remyelination. Stem Cells Int 2019; 2019:7378594. [PMID: 31885623 PMCID: PMC6915008 DOI: 10.1155/2019/7378594] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Accepted: 09/06/2019] [Indexed: 01/04/2023] Open
Abstract
In peripheral nerve injuries (PNIs) in which proximal axons do not regenerate quickly enough, significant chronic degeneration of Schwann cells (SCs) can occur at the distal stump of the injured nerve and obstruct regeneration. Cell transplantation can delay the degeneration of SCs, but transplanted cells fail to generate voluntary electrical impulses without downstream signal stimulation from the central nervous system. In this study, we combined cell transplantation and nerve transfer strategies to investigate whether the transplantation of embryonic spinal cord cells could benefit the microenvironment of the distal stump of the injured nerve. The experiment consisted of two stages. In the first-stage surgery, common peroneal nerves were transected, and embryonic day 14 (E14) cells or cell culture medium was transplanted into the distal stump of the CPs. Six months after the first-stage surgery, the transplanted cells were removed, and the nerve segment distal to the transplanted site was used to bridge freshly cut tibial nerves to detect whether the cell-treated graft promoted axon growth. The phenotypic changes and the neurotrophic factor expression pattern of SCs distal to the transplanted site were detected at several time points after cell transplantation and excision. The results showed that at different times after transplantation, the cells could survive and generate neurons. Thus, the neurons play the role of proximal axons to prevent chronic degeneration and fibrosis of SCs. After excision of the transplanted cells, the SCs returned to their dedifferentiated phenotype and upregulated growth-associated gene expression. The ability of SCs to be activated again allowed a favorable microenvironment to be created and enhanced the regeneration and remyelination of proximal axons. Muscle reinnervation was also elevated. This transplantation strategy could provide a treatment option for complex neurological injuries in the clinic.
Collapse
|
20
|
Enhanced regeneration and reinnervation following timed GDNF gene therapy in a cervical ventral root avulsion. Exp Neurol 2019; 321:113037. [PMID: 31425689 DOI: 10.1016/j.expneurol.2019.113037] [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: 06/14/2019] [Revised: 08/01/2019] [Accepted: 08/14/2019] [Indexed: 12/11/2022]
Abstract
Avulsion of spinal nerve roots is a severe proximal peripheral nerve lesion. Despite neurosurgical repair, recovery of function in human patients is disappointing, because spinal motor neurons degenerate progressively, axons grow slowly and the distal Schwann cells which are instrumental to supporting axon extension lose their pro-regenerative properties. We have recently shown that timed GDNF gene therapy (dox-i-GDNF) in a lumbar plexus injury model promotes axon regeneration and improves electrophysiological recovery but fails to stimulate voluntary hind paw function. Here we report that dox-i-GDNF treatment following avulsion and re-implantation of cervical ventral roots leads to sustained motoneuron survival and recovery of voluntary function. These improvements were associated with a twofold increase in motor axon regeneration and enhanced reinnervation of the hand musculature. In this cervical model the distal hand muscles are located 6,5 cm from the reimplantation site, whereas following a lumber lesion this distance is twice as long. Since the first signs of muscle reinnervation are observed 6 weeks after the lesion, this suggests that regenerating axons reached the hand musculature before a critical state of chronic denervation has developed. These results demonstrate that the beneficial effects of timed GDNF-gene therapy are more robust following spinal nerve avulsion lesions that allow reinnervation of target muscles within a relatively short time window after the lesion. This study is an important step in demonstrating the potential of timed GDNF-gene therapy to enhance axon regeneration after neurosurgical repair of a severe proximal nerve lesion.
Collapse
|
21
|
GDNF pretreatment overcomes Schwann cell phenotype mismatch to promote motor axon regeneration via sensory graft. Exp Neurol 2019; 318:258-266. [PMID: 31100319 DOI: 10.1016/j.expneurol.2019.05.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 05/10/2019] [Accepted: 05/12/2019] [Indexed: 12/11/2022]
Abstract
In the clinic, severe motor nerve injury is commonly repaired by autologous sensory nerve bridging, but the ability of Schwann cells (SCs) in sensory nerves to support motor neuron axon growth is poor due to phenotype mismatch. In vitro experiments have demonstrated that sensory-derived SCs overcome phenotypic mismatch-induced growth inhibition after pretreatment with exogenous glial cell-derived neurotrophic factor (GDNF) and induce motor neuron axonal growth. Thus, we introduced a novel staging surgery: In the first stage of surgery, the denervated sensory nerve was pretreated with sustained-release GDNF, which was encapsulated into a self-assembling peptide nanofiber scaffold (SAPNS) RADA-16I in the donor area in vivo. In the second stage of surgery, the pretreated sensory grafts were transplanted to repair motor nerve injury. Motor axon regeneration and remyelination and muscle functional recovery after the second surgery was compared to those in the control groups. The expression of genes previously shown to be differently expressed in motor and sensory SCs was also analyzed in pretreated sensory grafts by qRT-PCR to explore possible changes after exogenous GDNF application. Exogenous GDNF acted directly on the denervated sensory nerve graft in vivo, increasing the expression of endogenous GDNF and sensory SC-derived marker brain-derived neurotrophic factor (BDNF). After transplantation to repair motor nerve injury, exogenous GDNF pretreatment promoted the regeneration and remyelination of proximal motor axons and the recovery of muscle function. Further research into how phenotype, gene expression and changes in neurotrophic factors in SCs are affected by GDNF will help us design more effective methods to treat peripheral nerve injury.
Collapse
|
22
|
Chen S, Hou Y, Zhao Z, Luo Y, Lv S, Wang Q, Li J, He L, Zhou L, Wu W. Neuregulin-1 Accelerates Functional Motor Recovery by Improving Motoneuron Survival After Brachial Plexus Root Avulsion in Mice. Neuroscience 2019; 404:510-518. [PMID: 30731156 DOI: 10.1016/j.neuroscience.2019.01.054] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Revised: 01/16/2019] [Accepted: 01/28/2019] [Indexed: 12/14/2022]
Abstract
Brachial plexus root avulsion (BPRA) results in the complete loss of motor function in the upper limb, mainly due to the death of spinal motoneurons (MNs). The survival of spinal MNs is the key to the recovery of motor function. Neuregulin-1 (Nrg1) plays fundamental roles in nervous system development and nerve repair. However, its functional role in BPRA remains unclear. On the basis of our findings that Nrg1 is down-regulated in the ventral horn in a mouse model of BPRA, Nrg1 may be associated with BPRA. Here, we investigated whether recombinant Nrg1β (rNrg1β) can enhance the survival of spinal MNs and improve functional recovery in mice following BPRA. In vitro studies on primary cultured mouse MNs showed that rNrg1β increased the survival rate in a dose-dependent manner, reaching a peak at 5 nM, which increased the survival rate and enhanced the pERK levels in MNs under H2O2-induced oxidative stress. In vivo studies revealed that rNrg1β improved the functional recovery of elbow flexion, promoted the survival of MNs, enhanced the re-innervation of biceps brachii, and decreased the muscle atrophy. These results suggest that Nrg1 may provide a potential therapeutic strategy for root avulsion.
Collapse
Affiliation(s)
- Shuangxi Chen
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China
| | - Yuhui Hou
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China
| | - Zhikai Zhao
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China
| | - Yunhao Luo
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China
| | - Shiqin Lv
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China
| | - Qianghua Wang
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China
| | - Jing Li
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China
| | - Liumin He
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Libing Zhou
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China
| | - Wutian Wu
- Guangdong-Hong Kong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, China; Re-Stem Biotechnology Co., Ltd., Suzhou, China.
| |
Collapse
|
23
|
Wang T, Zeng LN, Zhu Z, Wang YH, Ding L, Luo WB, Zhang XM, He ZW, Wu HF. Effect of lentiviral vector-mediated overexpression of hypoxia-inducible factor 1 alpha delivered by pluronic F-127 hydrogel on brachial plexus avulsion in rats. Neural Regen Res 2019; 14:1069-1078. [PMID: 30762021 PMCID: PMC6404506 DOI: 10.4103/1673-5374.250629] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Brachial plexus avulsion often results in massive motor neuron death and severe functional deficits of target muscles. However, no satisfactory treatment is currently available. Hypoxia-inducible factor 1α is a critical molecule targeting several genes associated with ischemia-hypoxia damage and angiogenesis. In this study, a rat model of brachial plexus avulsion-reimplantation was established, in which C5–7 ventral nerve roots were avulsed and only the C6 root reimplanted. Different implants were immediately injected using a microsyringe into the avulsion-reimplantation site of the C6 root post-brachial plexus avulsion. Rats were randomly divided into five groups: phosphate-buffered saline, negative control of lentivirus, hypoxia-inducible factor 1α (hypoxia-inducible factor 1α overexpression lentivirus), gel (pluronic F-127 hydrogel), and gel + hypoxia-inducible factor 1α (pluronic F-127 hydrogel + hypoxia-inducible factor 1α overexpression lentivirus). The Terzis grooming test was performed to assess recovery of motor function. Scores were higher in the hypoxia-inducible factor 1α and gel + hypoxia-inducible factor 1α groups (in particular the gel + hypoxia-inducible factor 1α group) compared with the phosphate-buffered saline group. Electrophysiology, fluorogold retrograde tracing, and immunofluorescent staining were further performed to investigate neural pathway reconstruction and changes of neurons, motor endplates, and angiogenesis. Compared with the phosphate-buffered saline group, action potential latency of musculocutaneous nerves was markedly shortened in the hypoxia-inducible factor 1α and gel + hypoxia-inducible factor 1α groups. Meanwhile, the number of fluorogold-positive cells and ChAT-positive neurons, neovascular area (labeled by CD31 around avulsed sites in ipsilateral spinal cord segments), and the number of motor endplates in biceps brachii (identified by α-bungarotoxin) were all visibly increased, as well as the morphology of motor endplate in biceps brachil was clear in the hypoxia-inducible factor 1α and gel + hypoxia-inducible factor 1α groups. Taken together, delivery of hypoxia-inducible factor 1α overexpression lentiviral vectors mediated by pluronic F-127 effectively promotes spinal root regeneration and functional recovery post-brachial plexus avulsion. All animal procedures were approved by the Institutional Animal Care and Use Committee of Guangdong Medical University, China.
Collapse
Affiliation(s)
- Tao Wang
- Institute of Stem Cells and Regenerative Medicine, Department of Physiology, Guangdong Medical University, Dongguan; Department of Surgery, the Third Hospital of Guangdong Medical University (Longjiang Hospital of Shunde District), Foshan, Guangdong Province, China
| | - Li-Ni Zeng
- Institute of Stem Cells and Regenerative Medicine, Department of Physiology, Guangdong Medical University, Dongguan, Guangdong Province, China
| | - Zhe Zhu
- Hand & Foot Surgery and Reparative & Reconstruction Surgery Center, the Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Yu-Hui Wang
- Institute of Stem Cells and Regenerative Medicine, Department of Physiology, Guangdong Medical University, Dongguan; Department of Surgery, the Third Hospital of Guangdong Medical University (Longjiang Hospital of Shunde District), Foshan, Guangdong Province, China
| | - Lu Ding
- Department of Scientific Research Center, the Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, Guangdong Province, China
| | - Wei-Bin Luo
- Institute of Stem Cells and Regenerative Medicine, Department of Physiology, Guangdong Medical University, Dongguan; Department of Surgery, the Third Hospital of Guangdong Medical University (Longjiang Hospital of Shunde District), Foshan, Guangdong Province, China
| | - Xiao-Min Zhang
- Institute of Stem Cells and Regenerative Medicine, Department of Physiology, Guangdong Medical University, Dongguan, Guangdong Province, China
| | - Zhi-Wei He
- Institute of Stem Cells and Regenerative Medicine, Department of Physiology, Guangdong Medical University, Dongguan, Guangdong Province, China
| | - Hong-Fu Wu
- Institute of Stem Cells and Regenerative Medicine, Department of Physiology, Guangdong Medical University, Dongguan, Guangdong Province, China
| |
Collapse
|
24
|
Romeo-Guitart D, Casas C. Network-centric medicine for peripheral nerve injury: Treating the whole to boost endogenous mechanisms of neuroprotection and regeneration. Neural Regen Res 2019; 14:1122-1128. [PMID: 30804234 PMCID: PMC6425822 DOI: 10.4103/1673-5374.251187] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Peripheral nerve injuries caused by accidents may lead to paralysis, sensory disturbances, anaesthesia, and lack of autonomic functions. Functional recovery after disconnection of the motoneuronal soma from target tissue with proximal rupture of axons is determined by several factors: motoneuronal soma viability, proper axonal sprouting across inhibitory zones and elongation toward specific muscle, effective synapse contact rebuilding, and prevention of muscle atrophy. Therapies, such as adjuvant drugs with pleiotropic effects, that promote functional recovery after peripheral nerve injury are needed. Toward this aim, we designed a drug discovery workflow based on a network-centric molecular vision using unbiased proteomic data and neural artificial computational tools. Our focus is on boosting intrinsic capabilities of neurons for neuroprotection; this is in contrast to the common approach based on suppression of a pathobiological pathway known to be associated with disease condition. Using our workflow, we discovered neuroheal, a combination of two repurposed drugs that promotes motoneuronal soma neuroprotection, is anti-inflammatory, enhances axonal regeneration after axotomy, and reduces muscle atrophy. This drug discovery workflow has thus yielded a therapy that is close to its clinical application.
Collapse
Affiliation(s)
- David Romeo-Guitart
- Institut de Neurociències (INc) and Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona (UAB) & Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Barcelona, Spain
| | - Caty Casas
- Institut de Neurociències (INc) and Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona (UAB) & Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Barcelona, Spain
| |
Collapse
|
25
|
Husson T, Duboc JB, Quenez O, Charbonnier C, Rotharmel M, Cuenca M, Jegouzo X, Richard AC, Frebourg T, Deleuze JF, Boland A, Genin E, Debette S, Tzourio C, Campion D, Nicolas G, Guillin O, The FREX Consortium. Identification of potential genetic risk factors for bipolar disorder by whole-exome sequencing. Transl Psychiatry 2018; 8:268. [PMID: 30518751 PMCID: PMC6281607 DOI: 10.1038/s41398-018-0291-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 09/25/2018] [Accepted: 10/05/2018] [Indexed: 12/11/2022] Open
Abstract
This study aims at assessing the burden of rare (minor allele frequency < 1%) predicted damaging variants in the whole exome of 92 bipolar I disorder (BD) patients and 1051 controls of French ancestry. Patients exhibiting an extreme phenotype (earlier onset and family history of mood disorder) were preferentially included to increase the power to detect an association. A collapsing strategy was used to test the overall burden of rare variants in cases versus controls at the gene level. Only protein-truncating and predicted damaging missense variants were included in the analysis. Thirteen genes exhibited p values exceeding 10-3 and could be considered as potential risk factors for BD. Furthermore, the validity of the association was supported when the Exome Aggregation Consortium database non-Finnish European population was used as controls for eight of them. Their gene products are involved in various cerebral processes, some of which were previously implicated in BD and belong to pathways implicated in the therapeutic effect of lithium, the main mood stabilizer. However, exome-wide threshold for association study was not reached, emphasizing that larger samples are needed.
Collapse
Affiliation(s)
- Thomas Husson
- 0000 0004 1765 2814grid.477068.aDepartment of Research, Centre hospitalier du Rouvray, Sotteville-lès-Rouen, France ,grid.41724.34Department of Genetics, Normandy Centre for Genomic and Personalized Medicine, Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, F 76000 Rouen, France
| | - Jean-Baptiste Duboc
- 0000 0004 1765 2814grid.477068.aDepartment of Research, Centre hospitalier du Rouvray, Sotteville-lès-Rouen, France
| | - Olivier Quenez
- grid.41724.34Department of Genetics, Normandy Centre for Genomic and Personalized Medicine, Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, F 76000 Rouen, France
| | - Camille Charbonnier
- grid.41724.34Department of Genetics, Normandy Centre for Genomic and Personalized Medicine, Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, F 76000 Rouen, France
| | - Maud Rotharmel
- 0000 0004 1765 2814grid.477068.aDepartment of Research, Centre hospitalier du Rouvray, Sotteville-lès-Rouen, France ,grid.41724.34Department of Genetics, Normandy Centre for Genomic and Personalized Medicine, Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, F 76000 Rouen, France
| | - Macarena Cuenca
- 0000 0004 1765 2814grid.477068.aDepartment of Research, Centre hospitalier du Rouvray, Sotteville-lès-Rouen, France
| | - Xavier Jegouzo
- 0000 0004 1765 2814grid.477068.aDepartment of Research, Centre hospitalier du Rouvray, Sotteville-lès-Rouen, France
| | - Anne-Claire Richard
- grid.41724.34Department of Genetics, Normandy Centre for Genomic and Personalized Medicine, Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, F 76000 Rouen, France
| | - Thierry Frebourg
- grid.41724.34Department of Genetics, Normandy Centre for Genomic and Personalized Medicine, Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, F 76000 Rouen, France
| | - Jean-François Deleuze
- Centre National de Recherche en Génomique Humaine, Institut de Génomique, CEA, Evry, France
| | - Anne Boland
- Centre National de Recherche en Génomique Humaine, Institut de Génomique, CEA, Evry, France
| | - Emmanuelle Genin
- 0000 0004 0472 3249grid.411766.3Inserm UMR-1078, CHRU Brest, Univ. Brest, Brest, France
| | - Stéphanie Debette
- 0000 0001 2106 639Xgrid.412041.2Univ. Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR1219, F-33076 Bordeaux, France
| | - Christophe Tzourio
- 0000 0001 2106 639Xgrid.412041.2Univ. Bordeaux, Inserm, Bordeaux Population Health Research Center, UMR1219, F-33076 Bordeaux, France
| | - Dominique Campion
- 0000 0004 1765 2814grid.477068.aDepartment of Research, Centre hospitalier du Rouvray, Sotteville-lès-Rouen, France ,grid.41724.34Department of Genetics, Normandy Centre for Genomic and Personalized Medicine, Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, F 76000 Rouen, France
| | - Gaël Nicolas
- grid.41724.34Department of Genetics, Normandy Centre for Genomic and Personalized Medicine, Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, F 76000 Rouen, France
| | - Olivier Guillin
- Department of Research, Centre hospitalier du Rouvray, Sotteville-lès-Rouen, France. .,Department of Genetics, Normandy Centre for Genomic and Personalized Medicine, Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, F 76000, Rouen, France.
| | | |
Collapse
|
26
|
Abstract
Current strategies for promoting faster and more effective peripheral nerve healing have utilized a wide variety of techniques and approaches. Nerve grafts, conduits, and stem cell therapy all have their respective advantages. However, there are still some difficulties in attaining complete functional recovery with a single treatment modality. The utilization of adjuvant treatments, in combination with current standard-of-care methods, offers the potential to improve patient outcomes. This paper highlights the current landscape of adjuvant treatments for enhancing peripheral nerve repair and regeneration.
Collapse
|
27
|
Mohammad Jafari R, Ghahremani MH, Rahimi N, Shadboorestan A, Rashidian A, Esmaeili J, Ejtemaei Mehr S, Dehpour AR. The anticonvulsant activity and cerebral protection of chronic lithium chloride via NMDA receptor/nitric oxide and phospho-ERK. Brain Res Bull 2018; 137:1-9. [DOI: 10.1016/j.brainresbull.2017.10.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/30/2017] [Accepted: 10/31/2017] [Indexed: 02/08/2023]
|
28
|
Kim Y, Kim J, Ahn M, Shin T. Lithium ameliorates rat spinal cord injury by suppressing glycogen synthase kinase-3β and activating heme oxygenase-1. Anat Cell Biol 2017; 50:207-213. [PMID: 29043099 PMCID: PMC5639175 DOI: 10.5115/acb.2017.50.3.207] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/18/2017] [Accepted: 07/24/2017] [Indexed: 11/27/2022] Open
Abstract
Glycogen synthase kinase (GSK)-3β and related enzymes are associated with various forms of neuroinflammation, including spinal cord injury (SCI). Our aim was to evaluate whether lithium, a non-selective inhibitor of GSK-3β, ameliorated SCI progression, and also to analyze whether lithium affected the expression levels of two representative GSK-3β–associated molecules, nuclear factor erythroid 2-related factor-2 (Nrf-2) and heme oxygenase-1 (HO-1) (a target gene of Nrf-2). Intraperitoneal lithium chloride (80 mg/kg/day for 3 days) significantly improved locomotor function at 8 days post-injury (DPI); this was maintained until 14 DPI (P<0.05). Western blotting showed significantly increased phosphorylation of GSK-3β (Ser9), Nrf-2, and the Nrf-2 target HO-1 in the spinal cords of lithium-treated animals. Fewer neuropathological changes (e.g., hemorrhage, inflammatory cell infiltration, and tissue loss) were observed in the spinal cords of the lithium-treated group compared with the vehicle-treated group. Microglial activation (evaluated by measuring the immunoreactivity of ionized calcium-binding protein-1) was also significantly reduced in the lithium-treated group. These findings suggest that GSK-3β becomes activated after SCI, and that a non-specific enzyme inhibitor, lithium, ameliorates rat SCI by increasing phosphorylation of GSK-3β and the associated molecules Nrf-2 and HO-1.
Collapse
Affiliation(s)
- Yonghoon Kim
- Department of Veterinary Anatomy, College of Veterinary Medicine, Jeju National University, Jeju, Korea.,HAEMALGEUN Veterinary Medical Clinic, Jeju, Korea
| | - Jeongtae Kim
- Department of Veterinary Anatomy, College of Veterinary Medicine, Jeju National University, Jeju, Korea
| | - Meejung Ahn
- Department of Veterinary Anatomy, College of Veterinary Medicine, Jeju National University, Jeju, Korea
| | - Taekyun Shin
- Department of Veterinary Anatomy, College of Veterinary Medicine, Jeju National University, Jeju, Korea
| |
Collapse
|
29
|
Romeo-Guitart D, Forés J, Navarro X, Casas C. Boosted Regeneration and Reduced Denervated Muscle Atrophy by NeuroHeal in a Pre-clinical Model of Lumbar Root Avulsion with Delayed Reimplantation. Sci Rep 2017; 7:12028. [PMID: 28931824 PMCID: PMC5607317 DOI: 10.1038/s41598-017-11086-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 08/09/2017] [Indexed: 12/21/2022] Open
Abstract
The “gold standard” treatment of patients with spinal root injuries consists of delayed surgical reconnection of nerves. The sooner, the better, but problems such as injury-induced motor neuronal death and muscle atrophy due to long-term denervation mean that normal movement is not restored. Herein we describe a preclinical model of root avulsion with delayed reimplantation of lumbar roots that was used to establish a new adjuvant pharmacological treatment. Chronic treatment (up to 6 months) with NeuroHeal, a new combination drug therapy identified using a systems biology approach, exerted long-lasting neuroprotection, reduced gliosis and matrix proteoglycan content, accelerated nerve regeneration by activating the AKT pathway, promoted the formation of functional neuromuscular junctions, and reduced denervation-induced muscular atrophy. Thus, NeuroHeal is a promising treatment for spinal nerve root injuries and axonal regeneration after trauma.
Collapse
Affiliation(s)
- David Romeo-Guitart
- Institut de Neurociències (INc) and Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona (UAB), Bellaterra, Barcelona, Spain
| | - Joaquim Forés
- Hand and Peripheral Nerve Unit, Hospital Clínic i Provincial, Universitat de Barcelona, Barcelona, Spain
| | - Xavier Navarro
- Institut de Neurociències (INc) and Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona (UAB), Bellaterra, Barcelona, Spain
| | - Caty Casas
- Institut de Neurociències (INc) and Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona (UAB), Bellaterra, Barcelona, Spain.
| |
Collapse
|
30
|
Piñero G, Berg R, Andersen ND, Setton-Avruj P, Monje PV. Lithium Reversibly Inhibits Schwann Cell Proliferation and Differentiation Without Inducing Myelin Loss. Mol Neurobiol 2016; 54:8287-8307. [PMID: 27917448 DOI: 10.1007/s12035-016-0262-z] [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: 08/30/2016] [Accepted: 10/25/2016] [Indexed: 01/11/2023]
Abstract
This study was undertaken to examine the bioactivity, specificity, and reversibility of lithium's action on the growth, survival, proliferation, and differentiation of cultured Schwann cells (SCs). In isolated SCs, lithium promoted a state of cell cycle arrest that featured extensive cell enlargement and c-Jun downregulation in the absence of increased expression of myelin-associated markers. In addition, lithium effectively prevented mitogen-induced S-phase entry without impairing cell viability. When lithium was administered together with differentiating concentrations of cyclic adenosine monophosphate (cAMP) analogs, a dramatic inhibition of the expression of the master regulator of myelination Krox-20 was observed. Likewise, lithium antagonized the cAMP-dependent expression of various myelin markers such as protein zero, periaxin, and galactocerebroside and allowed SCs to maintain high levels of expression of immature SC markers even in the presence of high levels of cAMP and low levels of c-Jun. Most importantly, the inhibitory action of lithium on SC proliferation and differentiation was shown to be dose dependent, specific, and reversible upon removal of lithium compounds. In SC-neuron cultures, lithium suppressed myelin sheath formation while preserving axonal integrity, SC-axon contact, and basal lamina formation. Lithium was unique in its ability to prevent the onset of myelination without promoting myelin degradation or SC dedifferentiation. To conclude, our results underscored an unexpected antagonistic action of lithium on SC mitogenesis and myelin gene expression. We suggest that lithium represents an attractive pharmacological agent to safely and reversibly suppress the onset of SC proliferation, differentiation, and myelination while maintaining the integrity of pre-existing myelinated fibers.
Collapse
Affiliation(s)
- Gonzalo Piñero
- The Miami Project to Cure Paralysis and the Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
- Universidad de Buenos Aires. CONICET, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Buenos Aires, Argentina
| | - Randall Berg
- The Miami Project to Cure Paralysis and the Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Natalia Denise Andersen
- The Miami Project to Cure Paralysis and the Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Patricia Setton-Avruj
- Universidad de Buenos Aires. CONICET, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Buenos Aires, Argentina
| | - Paula Virginia Monje
- The Miami Project to Cure Paralysis and the Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|