1
|
Khavandegar A, Ahmadi NS, Mousavi MA, Ramezani Z, Khodadoust E, Hasan Zadeh Tabatabaei MS, Hasanpour Segherlou Z, Zeinaddini-Meymand A, Nasehi F, Moafi M, RayatSanati K, Masoomi R, Hamidi S, Pourkhodadad S, Rahimi-Movaghar V. The potential role of RhoA/ROCK-inhibition on locomotor recovery after spinal cord injury: a systematic review of in-vivo studies. Spinal Cord 2025; 63:95-126. [PMID: 39956860 DOI: 10.1038/s41393-025-01064-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 01/27/2025] [Accepted: 02/06/2025] [Indexed: 02/18/2025]
Abstract
STUDY DESIGN Systematic Review. OBJECTIVES To thoroughly assess the existing literature regarding the impact of anti-RhoA/ROCK agents or procedures on functional recovery in animal models of SCI. SETTING Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences. METHODS A comprehensive search was conducted in Ovid MEDLINE, Embase, Scopus, and Web of Science Core Collection utilizing a combination of keywords. All in-vivo animal studies of acute or chronic SCI that evaluated the pharmacological effects of Rho/ROCK inhibitors in English literature were included in this study. RESULTS Totally, 2320 articles were identified, of which, 60 papers were included for further analysis. A total of 47 (78%) studies were conducted merely on rats, 9 (15%) on mice, 3 (5%) used both, and the remaining used other animals. Y-27632, Fasudil, C3 Transferase and its derivatives (C3-05/PEP-C3/CT04/C3bot154-182/C3bot26mer(156-181)), Ibuprofen, Electroacupuncture (EA), SiRhoA, miR-133b, miR-135-5p, miR-381, miR-30b, Statins, 17β-estradiol, β-elemene, Lentivirus-mediated PGC-1a, Repulsive guidance molecule (RGMa), Local profound hypothermia, Jisuikang (JSK), Hyperbaric oxygen (HBO), Lv-shRhoA (Notch-1 inhibitor), Anti-Ryk antibody, LINGO-antagonist, BA-210, p21Cip1/WAF1, ORL-1 antagonist, Epigallocatechin-3-gallate (EGCG), Tamsulosin, AAV.ULK1.DN, and Indomethacin were the 28 reported agents/procedures with anti-RhoA/ROCK effects. The pooled SMD for BBB scores was 0.41 (p = 0.048) in the first week, 0.85 (p < 0.001) in the second week, 1.22 (p = 0.010) in the third week, and 1.53 (p = 0.001) in the fourth week. CONCLUSION Of the 28 identified anti-RhoA/ROCK agents, all but two (C3bot and its derivatives and EGCG) demonstrated promising results. The results of the meta-analysis cautiously indicate a significant increase in BBB scores over time after SCI.
Collapse
Affiliation(s)
- Armin Khavandegar
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Negar Sadat Ahmadi
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Alsadat Mousavi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Ramezani
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Elaheh Khodadoust
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | - Fatemeh Nasehi
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Maral Moafi
- Cell Biology and Anatomical Sciences, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kimia RayatSanati
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Rasool Masoomi
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Sorour Hamidi
- Department of Neurosurgery, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Soheila Pourkhodadad
- Department of Pharmacy and Chemical Biology, Emory University, School of Medicine, Atlanta, GA, USA
| | - Vafa Rahimi-Movaghar
- Sina Trauma and Surgery Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Couto M, Vasconcelos DP, Pereira CL, Neto E, Sarmento B, Lamghari M. Neuro-Immunomodulatory Potential of Nanoenabled 4D Bioprinted Microtissue for Cartilage Tissue Engineering. Adv Healthc Mater 2025; 14:e2400496. [PMID: 38850170 PMCID: PMC11834377 DOI: 10.1002/adhm.202400496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 06/03/2024] [Indexed: 06/10/2024]
Abstract
Cartilage defects trigger post-traumatic inflammation, leading to a catabolic metabolism in chondrocytes and exacerbating cartilage degradation. Current treatments aim to relieve pain but fail to target the inflammatory process underlying osteoarthritis (OA) progression. Here, a human cartilage microtissue (HCM) nanoenabled with ibuprofen-loaded poly(lactic-co-glycolic acid) nanoparticles (ibu-PLGA NPs) is 4D-bioprinted to locally mitigate inflammation and impair nerve sprouting. Under an in vitro inflamed environment, the nanoenabled HCM exhibits chondroprotective potential by decreasing the interleukin (IL)1β and IL6 release, while sustaining extracellular matrix (ECM) production. In vivo, assessments utilizing the air pouch mouse model affirm the nanoenabled HCM non-immunogenicity. Nanoenabled HCM-derived secretomes do not elicit a systemic immune response and decrease locally the recruitment of mature dendritic cells and the secretion of multiple inflammatory mediators and matrix metalloproteinases when compared to inflamed HCM condition. Notably, the nanoenabled HCM secretome has no impact on the innervation profile of the skin above the pouch cavity, suggesting a potential to impede nerve growth. Overall, HCM nanoenabled with ibu-PLGA NPs emerges as a potent strategy to mitigate inflammation and protect ECM without triggering nerve growth, introducing an innovative and promising approach in the cartilage tissue engineering field.
Collapse
Affiliation(s)
- Marina Couto
- i3S ‐ Instituto de Investigação e Inovação em Saúde, Universidade do Porto, PortugalRua Alfredo Allen, 208Porto4200‐125Portugal
- INEB ‐ Instituto Nacional de Engenharia Biomédica, Universidade do PortoRua Alfredo Allen, 208Porto4200‐125Portugal
- Instituto Ciências Biomédicas Abel SalazarUniversidade do Porto – ICBASRua Jorge de Viterbo Ferreira 228Porto4050–313Portugal
| | - Daniela Pereira Vasconcelos
- i3S ‐ Instituto de Investigação e Inovação em Saúde, Universidade do Porto, PortugalRua Alfredo Allen, 208Porto4200‐125Portugal
- INEB ‐ Instituto Nacional de Engenharia Biomédica, Universidade do PortoRua Alfredo Allen, 208Porto4200‐125Portugal
| | - Catarina Leite Pereira
- i3S ‐ Instituto de Investigação e Inovação em Saúde, Universidade do Porto, PortugalRua Alfredo Allen, 208Porto4200‐125Portugal
- INEB ‐ Instituto Nacional de Engenharia Biomédica, Universidade do PortoRua Alfredo Allen, 208Porto4200‐125Portugal
| | - Estrela Neto
- i3S ‐ Instituto de Investigação e Inovação em Saúde, Universidade do Porto, PortugalRua Alfredo Allen, 208Porto4200‐125Portugal
- INEB ‐ Instituto Nacional de Engenharia Biomédica, Universidade do PortoRua Alfredo Allen, 208Porto4200‐125Portugal
- Escola Superior de SaúdeInstituto Politécnico do PortoRua Dr. António Bernardino de Almeida 400Porto4200‐072Portugal
| | - Bruno Sarmento
- i3S ‐ Instituto de Investigação e Inovação em Saúde, Universidade do Porto, PortugalRua Alfredo Allen, 208Porto4200‐125Portugal
- INEB ‐ Instituto Nacional de Engenharia Biomédica, Universidade do PortoRua Alfredo Allen, 208Porto4200‐125Portugal
- Instituto Universitário de Ciências da Saúde – IUCS‐CESPURua Central de Gandra, 1317Gandra4585‐116Portugal
| | - Meriem Lamghari
- i3S ‐ Instituto de Investigação e Inovação em Saúde, Universidade do Porto, PortugalRua Alfredo Allen, 208Porto4200‐125Portugal
- INEB ‐ Instituto Nacional de Engenharia Biomédica, Universidade do PortoRua Alfredo Allen, 208Porto4200‐125Portugal
| |
Collapse
|
3
|
Bourguignon L, Lukas LP, Kondiles BR, Tong B, Lee JJ, Gomes T, Tetzlaff W, Kramer JLK, Walter M, Jutzeler CR. Impact of commonly administered drugs on the progression of spinal cord injury: a systematic review. COMMUNICATIONS MEDICINE 2024; 4:213. [PMID: 39448737 PMCID: PMC11502874 DOI: 10.1038/s43856-024-00638-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 10/09/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Complications arising from acute traumatic spinal cord injury (SCI) are routinely managed by various pharmacological interventions. Despite decades of clinical application, the potential impact on neurological recovery has been largely overlooked. This study aims to highlight commonly administered drugs with potential disease-modifying effects. METHODS This systematic literature review included studies referenced in PubMed, Scopus and Web of Science from inception to March 31st, 2021, which assess disease-modifying properties on neurological and/or functional recovery of drugs routinely administered following spinal cord injury. Drug effects were classified as positive, negative, mixed, no effect, or not (statistically) reported. Risk of bias was assessed separately for animal, randomized clinical trials, and observational human studies. RESULTS We analyzed 394 studies conducting 486 experiments that evaluated 144 unique or combinations of drugs. 195 of the 464 experiments conducted on animals (42%) and one study in humans demonstrate positive disease-modifying properties on neurological and/or functional outcomes. Methylprednisolone, melatonin, estradiol, and atorvastatin are the most common drugs associated with positive effects. Two studies on morphine and ethanol report negative effects on recovery. CONCLUSION Despite a large heterogeneity observed in study protocols, research from bed to bench and back to bedside provides an alternative approach to identify new candidate drugs in the context of SCI. Future research in human populations is warranted to determine if introducing drugs like melatonin, estradiol, or atorvastatin would contribute to enhancing neurological outcomes after acute SCI.
Collapse
Affiliation(s)
- Lucie Bourguignon
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland.
- Schulthess Klinik, Zurich, Switzerland.
| | - Louis P Lukas
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland.
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland.
- Schulthess Klinik, Zurich, Switzerland.
| | - Bethany R Kondiles
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
| | - Bobo Tong
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada
| | - Jaimie J Lee
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada
| | - Tomás Gomes
- Department of Biosystems Science and Engineering, ETH Zurich, Basel, Switzerland
| | - Wolfram Tetzlaff
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
| | - John L K Kramer
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, Canada
- Department of Anesthesiology, Pharmacology, and Therapeutics, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Matthias Walter
- Department of Urology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Catherine R Jutzeler
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Schulthess Klinik, Zurich, Switzerland
| |
Collapse
|
4
|
Forston MJ, Jordan SL, Cesarz GM, Burke DA, Shum-Siu A, Petruska JC, Magnuson DSK. Combining clinically common drugs with hindlimb stretching in spinal cord injured rodents. Spinal Cord 2024; 62:574-583. [PMID: 39187628 DOI: 10.1038/s41393-024-01023-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 08/02/2024] [Accepted: 08/13/2024] [Indexed: 08/28/2024]
Abstract
STUDY DESIGN Preclinical pilot study. OBJECTIVES To explore peripheral and central nociceptive mechanisms that contribute to muscle stretch-induced locomotor deficits following spinal cord injury. SETTING Kentucky Spinal Cord Injury Research Center, Louisville, KY, USA. METHODS Ten female Sprague-Dawley rats received moderate, 25 g/cm T10 contusion injuries and recovered for 4 weeks. Rats were divided into three groups: Morphine/Ibuprofen-treated, Acetaminophen/Baclofen-treated, and saline control. Each group received daily hindlimb muscle stretching during weeks 4, 5, 9, and 10 post-injury and drugs were administered with stretching during weeks 4 and 9 only. Locomotor function was assessed throughout the experiment using the BBB Open Field Locomotor Scale. Hindlimb responses including spasticity, writhing, and clonic-like vibrations during muscle stretching were classified and scored. RESULTS Consistent with our previous studies, hindlimb muscle stretching caused significant deficits in locomotor recovery following spinal cord injury. Baclofen and Ibuprofen partially mitigated the stretching effect, but none of the drugs significantly prevented the drop in locomotor function during stretching. Interestingly, treatment with Baclofen or Ibuprofen significantly reduced hindlimb responses such as spasticity and writhing during stretching, while Morphine exacerbated clonic-like vibrations in response to stretching maneuvers. CONCLUSIONS These findings suggest that stretching may inhibit locomotor recovery through combined mechanisms of peripheral inflammation and sensitization of nociceptive afferents. When combined with central sprouting and loss of descending controls after SCI, this results in exaggerated nociceptive input during stretching. The inability of the applied clinical drugs to mitigate the detrimental effects of stretching highlights the complexity of the stretching phenomenon and emphasizes the need for further investigation.
Collapse
Affiliation(s)
- Morgan J Forston
- Interdisciplinary Program in Translational Neuroscience, University of Louisville, Louisville, KY, USA
- Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY, USA
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY, USA
| | - Savannah L Jordan
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY, USA
- Department of Bioengineering, J.B. Speed School of Engineering, University of Louisville, Louisville, KY, USA
| | - Greta M Cesarz
- Department of Health and Sports Sciences, University of Louisville, Louisville, KY, USA
| | - Darlene A Burke
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY, USA
| | - Alice Shum-Siu
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY, USA
| | - Jeffrey C Petruska
- Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY, USA.
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY, USA.
- Department of Neurological Surgery, University of Louisville, Louisville, KY, USA.
| | - David S K Magnuson
- Interdisciplinary Program in Translational Neuroscience, University of Louisville, Louisville, KY, USA.
- Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY, USA.
- Kentucky Spinal Cord Injury Research Center, University of Louisville, Louisville, KY, USA.
- Department of Bioengineering, J.B. Speed School of Engineering, University of Louisville, Louisville, KY, USA.
- Department of Neurological Surgery, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
5
|
Kellaway SC, Ullrich MM, Dziemidowicz K. Electrospun drug-loaded scaffolds for nervous system repair. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1965. [PMID: 38740385 DOI: 10.1002/wnan.1965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 05/16/2024]
Abstract
Nervous system injuries, encompassing peripheral nerve injury (PNI), spinal cord injury (SCI), and traumatic brain injury (TBI), present significant challenges to patients' wellbeing. Traditional treatment approaches have limitations in addressing the complexity of neural tissue regeneration and require innovative solutions. Among emerging strategies, implantable materials, particularly electrospun drug-loaded scaffolds, have gained attention for their potential to simultaneously provide structural support and controlled release of therapeutic agents. This review provides a thorough exploration of recent developments in the design and application of electrospun drug-loaded scaffolds for nervous system repair. The electrospinning process offers precise control over scaffold characteristics, including mechanical properties, biocompatibility, and topography, crucial for creating a conducive environment for neural tissue regeneration. The large surface area of the resulting fibrous networks enhances biomolecule attachment, influencing cellular behaviors such as adhesion, proliferation, and migration. Polymeric electrospun materials demonstrate versatility in accommodating a spectrum of therapeutics, from small molecules to proteins. This enables tailored interventions to accelerate neuroregeneration and mitigate inflammation at the injury site. A critical aspect of this review is the examination of the interplay between structural properties and pharmacological effects, emphasizing the importance of optimizing both aspects for enhanced therapeutic outcomes. Drawing upon the latest advancements in the field, we discuss the promising outcomes of preclinical studies using electrospun drug-loaded scaffolds for nervous system repair, as well as future perspectives and considerations for their design and implementation. This article is categorized under: Implantable Materials and Surgical Technologies > Nanomaterials and Implants Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Simon C Kellaway
- Department of Pharmacology, UCL School of Pharmacy, London, United Kingdom
| | - Mathilde M Ullrich
- Department of Pharmacology, UCL School of Pharmacy, London, United Kingdom
- Department of Pharmaceutics, UCL School of Pharmacy, London, United Kingdom
| | - Karolina Dziemidowicz
- Department of Pharmacology, UCL School of Pharmacy, London, United Kingdom
- Department of Pharmaceutics, UCL School of Pharmacy, London, United Kingdom
| |
Collapse
|
6
|
Duarte D, Correia C, Reis RL, Pashkuleva I, Peixoto D, Alves NM. Bioadhesive Hyaluronic Acid-Based Hydrogels for Spinal Cord Injury. Biomacromolecules 2024; 25:1592-1601. [PMID: 38377534 DOI: 10.1021/acs.biomac.3c01186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Spinal cord injuries (SCI) have devastating physical, psychological, and psychosocial consequences for patients. One challenge of nerve tissue repair is the lack of a natural extracellular matrix (ECM) that guides the regenerating axons. Hyaluronic acid (HA) is a major ECM component and plays a fundamental role in facilitating lesion healing. Herein, we developed HA-based adhesive hydrogels by modification of HA with dopamine, a mussel-inspired compound with excellent adhesive properties in an aqueous environment. The hydrogels were loaded with the anti-inflammatory drug ibuprofen and the response of neuronal cells (SH-SY5Y) was evaluated in terms of viability, morphology, and adhesion. The obtained results suggested that the developed materials can bridge lesion gaps, guide axonal growth, and simultaneously act as a vehicle for the delivery of bioactive compounds.
Collapse
Affiliation(s)
- Diogo Duarte
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco/Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Cátia Correia
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco/Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco/Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Iva Pashkuleva
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco/Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Daniela Peixoto
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco/Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| | - Natália M Alves
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco/Guimarães, Portugal
- ICVS/3B's-PT Government Associate Laboratory, 4710-057 Braga/Guimarães, Portugal
| |
Collapse
|
7
|
Guha L, Kumar H. Drug Repurposing for Spinal Cord Injury: Progress Towards Therapeutic Intervention for Primary Factors and Secondary Complications. Pharmaceut Med 2023; 37:463-490. [PMID: 37698762 DOI: 10.1007/s40290-023-00499-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2023] [Indexed: 09/13/2023]
Abstract
Spinal cord injury (SCI) encompasses a plethora of complex mechanisms like the involvement of major cell death pathways, neurodegeneration of spinal cord neurons, overexpression of glutaminergic transmission and inflammation cascade, along with different co-morbidities like neuropathic pain, urinary and sexual dysfunction, respiratory and cardiac failures, making it one of the leading causes of morbidity and mortality globally. Corticosteroids such as methylprednisolone and dexamethasone, and non-steroidal anti-inflammatory drugs such as naproxen, aspirin and ibuprofen are the first-line treatment options for SCI, inhibiting primary and secondary progression by preventing inflammation and action of reactive oxygen species. However, they are constrained by a short effective drug administration window and their pharmacological action being limited to symptomatic relief of the secondary effects related to spinal cord injury only. Although post-injury rehabilitation treatments may enable functional recovery, they take a long time to show results. Drug repurposing might be an innovative method for expanding therapy alternatives, utilising drugs that are already approved by various esteemed federal agencies throughout the world. Reutilising a drug molecule to treat SCI can eliminate the need for expensive and lengthy drug discovery processes and pave the way for new therapeutic approaches in SCI. This review summarises marketed drugs that could be repurposed based on their safety and efficacy data. We also discuss their mechanisms of action and provide a list of repurposed drugs under clinical trials for SCI therapy.
Collapse
Affiliation(s)
- Lahanya Guha
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Opposite Air Force Station, Palaj, P.O-382355, Gandhinagar, Gujarat, India
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Opposite Air Force Station, Palaj, P.O-382355, Gandhinagar, Gujarat, India.
| |
Collapse
|
8
|
Rocha DN, Carvalho ED, Pires LR, Gardin C, Zanolla I, Szewczyk PK, Machado C, Fernandes R, Stachewicz U, Zavan B, Relvas JB, Pêgo AP. It takes two to remyelinate: A bioengineered platform to study astrocyte-oligodendrocyte crosstalk and potential therapeutic targets in remyelination. BIOMATERIALS ADVANCES 2023; 151:213429. [PMID: 37148597 DOI: 10.1016/j.bioadv.2023.213429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Revised: 03/21/2023] [Accepted: 04/12/2023] [Indexed: 05/08/2023]
Abstract
The loss of the myelin sheath insulating axons is the hallmark of demyelinating diseases. These pathologies often lead to irreversible neurological impairment and patient disability. No effective therapies are currently available to promote remyelination. Several elements contribute to the inadequacy of remyelination, thus understanding the intricacies of the cellular and signaling microenvironment of the remyelination niche might help us to devise better strategies to enhance remyelination. Here, using a new in vitro rapid myelinating artificial axon system based on engineered microfibres, we investigated how reactive astrocytes influence oligodendrocyte (OL) differentiation and myelination ability. This artificial axon culture system enables the effective uncoupling of molecular cues from the biophysical properties of the axons, allowing the detailed study of the astrocyte-OL crosstalk. Oligodendrocyte precursor cells (OPCs) were cultured on poly(trimethylene carbonate-co-ε-caprolactone) copolymer electrospun microfibres that served as surrogate axons. This platform was then combined with a previously established tissue engineered glial scar model of astrocytes embedded in 1 % (w/v) alginate matrices, in which astrocyte reactive phenotype was acquired using meningeal fibroblast conditioned medium. OPCs were shown to adhere to uncoated engineered microfibres and differentiate into myelinating OL. Reactive astrocytes were found to significantly impair OL differentiation ability, after six and eight days in a co-culture system. Differentiation impairment was seen to be correlated with astrocytic miRNA release through exosomes. We found significantly reduction on the expression of pro-myelinating miRNAs (miR-219 and miR-338) and an increase in anti-myelinating miRNA (miR-125a-3p) content between reactive and quiescent astrocytes. Additionally, we show that OPC differentiation inhibition could be reverted by rescuing the activated astrocytic phenotype with ibuprofen, a chemical inhibitor of the small rhoGTPase RhoA. Overall, these findings show that modulating astrocytic function might be an interesting therapeutic avenue for demyelinating diseases. The use of these engineered microfibres as an artificial axon culture system will enable the screening for potential therapeutic agents that promote OL differentiation and myelination while providing valuable insight on the myelination/remyelination processes.
Collapse
Affiliation(s)
- Daniela N Rocha
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal; Instituto de Engenharia Biomédica (INEB), Universidade do Porto, 4200-135 Porto, Portugal; Faculdade de Engenharia da Universidade do Porto (FEUP), 4200-465 Porto, Portugal
| | - Eva D Carvalho
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal; Instituto de Engenharia Biomédica (INEB), Universidade do Porto, 4200-135 Porto, Portugal; Faculdade de Engenharia da Universidade do Porto (FEUP), 4200-465 Porto, Portugal
| | - Liliana R Pires
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto, 4200-135 Porto, Portugal; Faculdade de Engenharia da Universidade do Porto (FEUP), 4200-465 Porto, Portugal
| | - Chiara Gardin
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, 48033 Ravenna, Italy
| | - Ilaria Zanolla
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Piotr K Szewczyk
- Faculty of Metals Engineering and Industrial Computer Science, AGH University of Science and Technology, 30-059 Krakow, Poland
| | - Cláudia Machado
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
| | - Rui Fernandes
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal
| | - Urszula Stachewicz
- Faculty of Metals Engineering and Industrial Computer Science, AGH University of Science and Technology, 30-059 Krakow, Poland
| | - Barbara Zavan
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - João B Relvas
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal; Instituto de Biologia Molecular e Celular (IBMC), Universidade do Porto, 4200-135 Porto, Portugal; Department of Biomedicine, Faculty of Medicine, Universidade do Porto, 4200-319 Porto, Portugal
| | - Ana P Pêgo
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, 4200-135 Porto, Portugal; Instituto de Engenharia Biomédica (INEB), Universidade do Porto, 4200-135 Porto, Portugal; Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, 4050-343 Porto, Portugal.
| |
Collapse
|
9
|
Rayner MLD, Kellaway SC, Kingston I, Guillemot-Legris O, Gregory H, Healy J, Phillips JB. Exploring the Nerve Regenerative Capacity of Compounds with Differing Affinity for PPARγ In Vitro and In Vivo. Cells 2022; 12:cells12010042. [PMID: 36611836 PMCID: PMC9818498 DOI: 10.3390/cells12010042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/09/2022] [Accepted: 12/10/2022] [Indexed: 12/24/2022] Open
Abstract
Damage to peripheral nerves can cause debilitating consequences for patients such as lifelong pain and disability. At present, no drug treatments are routinely given in the clinic following a peripheral nerve injury (PNI) to improve regeneration and remyelination of damaged nerves. Appropriately targeted therapeutic agents have the potential to be used at different stages following nerve damage, e.g., to maintain Schwann cell viability, induce and sustain a repair phenotype to support axonal growth, or promote remyelination. The development of therapies to promote nerve regeneration is currently of high interest to researchers, however, translation to the clinic of drug therapies for PNI is still lacking. Studying the effect of PPARγ agonists for treatment of peripheral nerve injures has demonstrated significant benefits. Ibuprofen, a non-steroidal anti-inflammatory drug (NSAID), has reproducibly demonstrated benefits in vitro and in vivo, suggested to be due to its agonist action on PPARγ. Other NSAIDs have demonstrated differing levels of PPARγ activation based upon their affinity. Therefore, it was of interest to determine whether affinity for PPARγ of selected drugs corresponded to an increase in regeneration. A 3D co-culture in vitro model identified some correlation between these two properties. However, when the drug treatments were screened in vivo, in a crush injury model in a rat sciatic nerve, the same correlation was not apparent. Further differences were observed between capacity to increase axon number and improvement in functional recovery. Despite there not being a clear correlation between affinity and size of effect on regeneration, all selected PPARγ agonists improved regeneration, providing a panel of compounds that could be explored for use in the treatment of PNI.
Collapse
Affiliation(s)
- Melissa L. D. Rayner
- Department of Pharmacology, School of Pharmacy, University College London, London WC1N 1AX, UK
- Centre for Nerve Engineering, University College London, London WC1N 6BT, UK
- Correspondence:
| | - Simon C. Kellaway
- Department of Pharmacology, School of Pharmacy, University College London, London WC1N 1AX, UK
- Centre for Nerve Engineering, University College London, London WC1N 6BT, UK
| | - Isabel Kingston
- Department of Pharmacology, School of Pharmacy, University College London, London WC1N 1AX, UK
- Centre for Nerve Engineering, University College London, London WC1N 6BT, UK
| | - Owein Guillemot-Legris
- Department of Pharmacology, School of Pharmacy, University College London, London WC1N 1AX, UK
- Centre for Nerve Engineering, University College London, London WC1N 6BT, UK
| | - Holly Gregory
- Department of Pharmacology, School of Pharmacy, University College London, London WC1N 1AX, UK
- Centre for Nerve Engineering, University College London, London WC1N 6BT, UK
| | - Jess Healy
- Centre for Nerve Engineering, University College London, London WC1N 6BT, UK
| | - James B. Phillips
- Department of Pharmacology, School of Pharmacy, University College London, London WC1N 1AX, UK
- Centre for Nerve Engineering, University College London, London WC1N 6BT, UK
| |
Collapse
|
10
|
Sawaguchi S, Suzuki R, Oizumi H, Ohbuchi K, Mizoguchi K, Yamamoto M, Miyamoto Y, Yamauchi J. Hypomyelinating Leukodystrophy 8 (HLD8)-Associated Mutation of POLR3B Leads to Defective Oligodendroglial Morphological Differentiation Whose Effect Is Reversed by Ibuprofen. Neurol Int 2022; 14:212-244. [PMID: 35225888 PMCID: PMC8884015 DOI: 10.3390/neurolint14010018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 02/03/2022] [Accepted: 02/14/2022] [Indexed: 11/16/2022] Open
Abstract
POLR3B and POLR3A are the major subunits of RNA polymerase III, which synthesizes non-coding RNAs such as tRNAs and rRNAs. Nucleotide mutations of the RNA polymerase 3 subunit b (polr3b) gene are responsible for hypomyelinating leukodystrophy 8 (HLD8), which is an autosomal recessive oligodendroglial cell disease. Despite the important association between POLR3B mutation and HLD8, it remains unclear how mutated POLR3B proteins cause oligodendroglial cell abnormalities. Herein, we show that a severe HLD8-associated nonsense mutation (Arg550-to-Ter (R550X)) primarily localizes POLR3B proteins as protein aggregates into lysosomes in the FBD-102b cell line as an oligodendroglial precursor cell model. Conversely, wild type POLR3B proteins were not localized in lysosomes. Additionally, the expression of proteins with the R550X mutation in cells decreased lysosome-related signaling through the mechanistic target of rapamycin (mTOR). Cells harboring the mutant constructs did not exhibit oligodendroglial cell differentiated phenotypes, which have widespread membranes that extend from their cell body. However, cells harboring the wild type constructs exhibited differentiated phenotypes. Ibuprofen, which is a non-steroidal anti-inflammatory drug (NSAID), improved the defects in their differentiation phenotypes and signaling through mTOR. These results indicate that the HLD8-associated POLR3B proteins with the R550X mutation are localized in lysosomes, decrease mTOR signaling, and inhibit oligodendroglial cell morphological differentiation, and ibuprofen improves these cellular pathological effects. These findings may reveal some of the molecular and cellular pathological mechanisms underlying HLD8 and their amelioration.
Collapse
Affiliation(s)
- Sui Sawaguchi
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Hachioji 192-0392, Japan; (S.S.); (R.S.); (Y.M.)
| | - Rimi Suzuki
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Hachioji 192-0392, Japan; (S.S.); (R.S.); (Y.M.)
| | - Hiroaki Oizumi
- Tsumura Research Laboratories, Tsumura & Co., Inashiki 200-1192, Japan; (H.O.); (K.O.); (K.M.); (M.Y.)
| | - Katsuya Ohbuchi
- Tsumura Research Laboratories, Tsumura & Co., Inashiki 200-1192, Japan; (H.O.); (K.O.); (K.M.); (M.Y.)
| | - Kazushige Mizoguchi
- Tsumura Research Laboratories, Tsumura & Co., Inashiki 200-1192, Japan; (H.O.); (K.O.); (K.M.); (M.Y.)
| | - Masahiro Yamamoto
- Tsumura Research Laboratories, Tsumura & Co., Inashiki 200-1192, Japan; (H.O.); (K.O.); (K.M.); (M.Y.)
| | - Yuki Miyamoto
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Hachioji 192-0392, Japan; (S.S.); (R.S.); (Y.M.)
- Department of Pharmacology, National Research Institute for Child Health and Development, Setagaya 157-8535, Japan
| | - Junji Yamauchi
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Hachioji 192-0392, Japan; (S.S.); (R.S.); (Y.M.)
- Department of Pharmacology, National Research Institute for Child Health and Development, Setagaya 157-8535, Japan
- Correspondence: ; Tel.: +81-42-676-7164; Fax: +81-42-676-8841
| |
Collapse
|
11
|
Zawadzka M, Kwaśniewska A, Miazga K, Sławińska U. Perspectives in the Cell-Based Therapies of Various Aspects of the Spinal Cord Injury-Associated Pathologies: Lessons from the Animal Models. Cells 2021; 10:cells10112995. [PMID: 34831217 PMCID: PMC8616284 DOI: 10.3390/cells10112995] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/25/2021] [Accepted: 10/31/2021] [Indexed: 02/07/2023] Open
Abstract
Traumatic injury of the spinal cord (SCI) is a devastating neurological condition often leading to severe dysfunctions, therefore an improvement in clinical treatment for SCI patients is urgently needed. The potential benefits of transplantation of various cell types into the injured spinal cord have been intensively investigated in preclinical SCI models and clinical trials. Despite the many challenges that are still ahead, cell transplantation alone or in combination with other factors, such as artificial matrices, seems to be the most promising perspective. Here, we reviewed recent advances in cell-based experimental strategies supporting or restoring the function of the injured spinal cord with a particular focus on the regenerative mechanisms that could define their clinical translation.
Collapse
|
12
|
Repurposing Small Molecules to Target PPAR-γ as New Therapies for Peripheral Nerve Injuries. Biomolecules 2021; 11:biom11091301. [PMID: 34572514 PMCID: PMC8465622 DOI: 10.3390/biom11091301] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 08/08/2021] [Accepted: 08/14/2021] [Indexed: 12/21/2022] Open
Abstract
The slow rate of neuronal regeneration that follows peripheral nerve repair results in poor recovery, particularly where reinnervation of muscles is delayed, leading to atrophy and permanent loss of function. There is a clear clinical need to develop drug treatments that can accelerate nerve regeneration safely, restoring connections before the target tissues deteriorate irreversibly. The identification that the Rho/Rho-associated kinase (ROCK) pathway acts to limit neuronal growth rate is a promising advancement towards the development of drugs. Targeting Rho or ROCK directly can act to suppress the activity of this pathway; however, the pathway can also be modulated through the activation of upstream receptors; one of particular interest being peroxisome proliferator-activated receptor gamma (PPAR-γ). The connection between the PPAR-γ receptor and the Rho/ROCK pathway is the suppression of the conversion of inactive guanosine diphosphate (GDP)-Rho to active guanosine triphosphate GTP-Rho, resulting in the suppression of Rho/ROCK activity. PPAR-γ is known for its role in cellular metabolism that leads to cell growth and differentiation. However, more recently there has been a growing interest in targeting PPAR-γ in peripheral nerve injury (PNI). The localisation and expression of PPAR-γ in neural cells following a PNI has been reported and further in vitro and in vivo studies have shown that delivering PPAR-γ agonists following injury promotes nerve regeneration, leading to improvements in functional recovery. This review explores the potential of repurposing PPAR-γ agonists to treat PNI and their prospective translation to the clinic.
Collapse
|
13
|
Yang Y, Zhang L, Huang M, Sui R, Khan S. Reconstruction of the cervical spinal cord based on motor function restoration and mitigation of oxidative stress and inflammation through eNOS/Nrf2 signaling pathway using ibuprofen-loaded nanomicelles. ARAB J CHEM 2021. [DOI: 10.1016/j.arabjc.2021.103289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
14
|
Vinton J, Aninweze A, Birgbauer E. Ibuprofen does not inhibit RhoA-mediated growth cone collapse of embryonic chicken retinal axons by LPA. Exp Brain Res 2021; 239:2969-2977. [PMID: 34322723 DOI: 10.1007/s00221-021-06172-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 05/15/2021] [Indexed: 11/28/2022]
Abstract
Lysophosphatidic acid (LPA) is a bioactive lysophospholipid that causes neuronal growth cones to collapse and neurites to retract through a RhoA-ROCK mediated pathway. It has been reported that the NSAID ibuprofen improves regeneration after spinal cord injury through a mechanism of inhibiting RhoA. This leads to the hypothesis that ibuprofen should block LPA-mediated growth cone collapse. We tested this hypothesis by treating embryonic chick retinal neurons with ibuprofen followed by LPA. Retinal growth cones collapsed with LPA in the presence of ibuprofen similar to control; however, growth cone collapse was effectively blocked by a ROCK inhibitor. Thus, our results do not support the designation of ibuprofen as a direct RhoA inhibitor.
Collapse
Affiliation(s)
- James Vinton
- Department of Biology, Winthrop University, Rock Hill, SC, 29733, USA
| | - Adaeze Aninweze
- Department of Biology, Winthrop University, Rock Hill, SC, 29733, USA
| | - Eric Birgbauer
- Department of Biology, Winthrop University, Rock Hill, SC, 29733, USA.
| |
Collapse
|
15
|
Upadhyay A, Amanullah A, Joshi V, Dhiman R, Prajapati VK, Poluri KM, Mishra A. Ibuprofen-based advanced therapeutics: breaking the inflammatory link in cancer, neurodegeneration, and diseases. Drug Metab Rev 2021; 53:100-121. [PMID: 33820460 DOI: 10.1080/03602532.2021.1903488] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Ibuprofen is a classical nonsteroidal anti-inflammatory drug (NSAID) highly prescribed to reduce acute pain and inflammation under an array of conditions, including rheumatoid arthritis, osteoarthritis, dysmenorrhea, and gout. Ibuprofen acts as a potential inhibitor for cyclooxygenase enzymes (COX-1 and COX-2). In the past few decades, research on this small molecule has led to identifying other possible therapeutic benefits. Anti-tumorigenic and neuroprotective functions of Ibuprofen are majorly recognized in recent literature and need further consideration. Additionally, several other roles of this anti-inflammatory molecule have been discovered and subjected to experimental assessment in various diseases. However, the major challenge faced by Ibuprofen and other drugs of similar classes is their side effects, and tendency to cause gastrointestinal injury, generate cardiovascular risks, modulate hepatic and acute kidney diseases. Future research should also be conducted to deduce new methods and approaches of suppressing the unwanted toxic changes mediated by these drugs and develop new therapeutic avenues so that these small molecules continue to serve the purposes. This article primarily aims to develop a comprehensive and better understanding of Ibuprofen, its pharmacological features, therapeutic benefits, and possible but less understood medicinal properties apart from major challenges in its future application.KEY POINTSIbuprofen, an NSAID, is a classical anti-inflammatory therapeutic agent.Pro-apoptotic roles of NSAIDs have been explored in detail in the past, holding the key in anti-cancer therapies.Excessive and continuous use of NSAIDs may have several side effects and multiple organ damage.Hyperactivated Inflammation initiates multifold detrimental changes in multiple pathological conditions.Targeting inflammatory pathways hold the key to several therapeutic strategies against many diseases, including cancer, microbial infections, multiple sclerosis, and many other brain diseases.
Collapse
Affiliation(s)
- Arun Upadhyay
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| | - Ayeman Amanullah
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| | - Vibhuti Joshi
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| | - Rohan Dhiman
- Laboratory of Mycobacterial Immunology, Department of Life Science, National Institute of Technology, Rourkela, Odisha, India
| | - Vijay Kumar Prajapati
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, Ajmer, Rajasthan, India
| | - Krishna Mohan Poluri
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Amit Mishra
- Cellular and Molecular Neurobiology Unit, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| |
Collapse
|
16
|
Lambrechts MJ, Cook JL. Nonsteroidal Anti-Inflammatory Drugs and Their Neuroprotective Role After an Acute Spinal Cord Injury: A Systematic Review of Animal Models. Global Spine J 2021; 11:365-377. [PMID: 32875860 PMCID: PMC8013945 DOI: 10.1177/2192568220901689] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
STUDY DESIGN Systematic review. OBJECTIVE Spinal cord injuries (SCIs) resulting in motor deficits can be devastating injuries resulting in millions of health care dollars spent per incident. Nonsteroidal anti-inflammatory drugs (NSAIDs) are a potential class of drugs that could improve motor function after an SCI. This systematic review utilizes PRISMA guidelines to evaluate the effectiveness of NSAIDs for SCI. METHODS PubMed/MEDLINE, CINAHL, PsycINFO, Embase, and Scopus were reviewed linking the keywords of "ibuprofen," "meloxicam," "naproxen," "ketorolac," "indomethacin," "celecoxib," "ATB-346," "NSAID," and "nonsteroidal anti-inflammatory drug" with "spinal." Results were reviewed for relevance and included if they met inclusion criteria. The SYRCLE checklist was used to assess sources of bias. RESULTS A total of 2960 studies were identified in the PubMed/MEDLINE database using the above-mentioned search criteria. A total of 461 abstracts were reviewed in Scopus, 340 in CINAHL, 179 in PsycINFO, and 7632 in Embase. A total of 15 articles met the inclusion criteria. CONCLUSIONS NSAIDs' effectiveness after SCI is largely determined by its ability to inhibit Rho-A. NSAIDs are a promising therapeutic option in acute SCI patients because they appear to decrease cord edema and inflammation, increase axonal sprouting, and improve motor function with minimal side effects. Studies are limited by heterogeneity, small sample size, and the use of animal models, which might not replicate the therapeutic effects in humans. There are no published human studies evaluating the safety and efficacy of these drugs after a traumatic cord injury. There is a need for well-designed prospective studies evaluating ibuprofen or indomethacin after adult spinal cord injuries.
Collapse
Affiliation(s)
| | - James L. Cook
- University of Missouri, Columbia, MO, USA,James L. Cook, University of Missouri, Missouri Orthopaedic Institute (4028A), 1100 Virginia Ave, Columbia, MO 65212, USA.
| |
Collapse
|
17
|
Luo M, Li YQ, Lu YF, Wu Y, Liu R, Zheng YR, Yin M. Exploring the potential of RhoA inhibitors to improve exercise-recoverable spinal cord injury: A systematic review and meta-analysis. J Chem Neuroanat 2020; 111:101879. [PMID: 33197553 DOI: 10.1016/j.jchemneu.2020.101879] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/23/2020] [Accepted: 10/24/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND The spinal cord is one of the central nervous system. Spinal cord injury (SCI) will cause loss of physical function and dysfunction below the injury site, causing them to lose sensation and mobility, thereby reducing the quality of life of patients. Although regular rehabilitation management can reduce its severity, the current effective treatment methods are limited to the treatment of secondary injuries to SCI. The purpose of treatment should not only include the restoration of the histology of the lesion, but also should focus on the restoration of sensory and mobility and. The key to effective treatment is to reduce secondary injuries. RhoA inhibitor can improve the pathophysiological changes related to secondary injury and promote the recovery of activity ability, so it may become a clinical drug for the treatment of SCI. This article systematically analyzed the effects of RhoA inhibitors on the promotion of axon regeneration and the recovery of mobility and compared the therapeutic effects of different inhibitors on SCI and their effects on physical function recovery. METHODS We used a meta-analysis to systematically evaluate the effects of Rho inhibitors on SCI treatment and the recovery of body function. RESULTS 21 articles (738 animals) were identified in the literatures search. Studies were selected if they reported the therapeutic effects of RhoA/ROCK inhibitors (BA-210, EGCG, β-elemene, C3-exoenzmye, LINGO-1-Fc, Ibuprofen, SiRhoA, iRhoA + FK506, Fasudil, p21Cip1/WAF1, HA-1007, Y-27,632 and C3bot154-182). We measure the functional recovery by BBB and BMS scores. The random effect model of weighted mean difference (WMD, 95 % confidence interval) was used to analyze the effects. The WMD of the forest graph was 2.277; 95 % CI: 1.705∼2.849, P < 0.001, suggesting that RhoA inhibitors can effectively treat SCI. In addition to EGCG, all the other agents also showed the effects on the activity recovery post-SCI (P < 0.05). CONCLUSION β-elemene, LINGO-1-Fc, Ibuprofen, SiRhoA, RhoA + FK506, Fasudil, p21Cip1/WAF1 and Y-27,632 have similar effects to BA-210, they can promote axon germination and nerve fiber regeneration after thoracic spinal cord injury and reduce the formation of syringomyelia and protect white matter, thereby improving locomotor recovery. RhoA inhibitors have great potential to restore motor function and provide a new trend for the treatment of SCI.
Collapse
Affiliation(s)
- Min Luo
- The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China.
| | - Yu Qing Li
- Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan Province, China.
| | - Ya Feng Lu
- The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China.
| | - Yue Wu
- The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China.
| | - RenShuai Liu
- The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China.
| | - Yu Rong Zheng
- The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China.
| | - Mei Yin
- The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan Province, China.
| |
Collapse
|
18
|
Kamath SM, Sridhar K, Jaison D, Gopinath V, Ibrahim BKM, Gupta N, Sundaram A, Sivaperumal P, Padmapriya S, Patil SS. Fabrication of tri-layered electrospun polycaprolactone mats with improved sustained drug release profile. Sci Rep 2020; 10:18179. [PMID: 33097770 PMCID: PMC7584580 DOI: 10.1038/s41598-020-74885-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Accepted: 09/28/2020] [Indexed: 12/16/2022] Open
Abstract
Modulation of initial burst and long term release from electrospun fibrous mats can be achieved by sandwiching the drug loaded mats between hydrophobic layers of fibrous polycaprolactone (PCL). Ibuprofen (IBU) loaded PCL fibrous mats (12% PCL-IBU) were sandwiched between fibrous polycaprolactone layers during the process of electrospinning, by varying the polymer concentrations (10% (w/v), 12% (w/v)) and volume of coat (1 ml, 2 ml) in flanking layers. Consequently, 12% PCL-IBU (without sandwich layer) showed burst release of 66.43% on day 1 and cumulative release (%) of 86.08% at the end of 62 days. Whereas, sandwich groups, especially 12% PCLSW-1 & 2 (sandwich layers-1 ml and 2 ml of 12% PCL) showed controlled initial burst and cumulative (%) release compared to 12% PCL-IBU. Moreover, crystallinity (%) and hydrophobicity of the sandwich models imparted control on ibuprofen release from fibrous mats. Further, assay for cytotoxicity and scanning electron microscopic images of cell seeded mats after 5 days showed the mats were not cytotoxic. Nuclear Magnetic Resonance spectroscopic analysis revealed weak interaction between ibuprofen and PCL in nanofibers which favors the release of ibuprofen. These data imply that concentration and volume of coat in flanking layer imparts tighter control on initial burst and long term release of ibuprofen.
Collapse
Affiliation(s)
- S Manjunath Kamath
- Department of Translational Medicine and Research, SRM Medical College, SRMIST, Kattankulathur, Tamil Nadu, 603203, India.
| | - K Sridhar
- Institute of Craniofacial, Aesthetic & Plastic Surgery (ICAPS), SRM Institute for Medical Sciences (SIMS), Chennai, Tamil Nadu, 600026, India
| | - D Jaison
- Nanotechnology Research Center (NRC), SRMIST, Kattankulathur, Tamil Nadu, 603203, India
| | - V Gopinath
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - B K Mohamed Ibrahim
- Institute of Craniofacial, Aesthetic & Plastic Surgery (ICAPS), SRM Institute for Medical Sciences (SIMS), Chennai, Tamil Nadu, 600026, India
| | - Nilkantha Gupta
- Department of Translational Medicine and Research, SRM Medical College, SRMIST, Kattankulathur, Tamil Nadu, 603203, India
| | - A Sundaram
- Department of Pathology, SRM Medical College, SRMIST, Kattankulathur, Tamil Nadu, 603203, India
| | - P Sivaperumal
- Department of Pharmacology, Saveetha Dental College (SDC), Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, India
| | - S Padmapriya
- Electrochemical Systems Laboratory, SRM Research Institute, SRMIST, Kattankulathur, Tamil Nadu, 603203, India
| | - S Shantanu Patil
- Department of Translational Medicine and Research, SRM Medical College, SRMIST, Kattankulathur, Tamil Nadu, 603203, India
| |
Collapse
|
19
|
Sami A, Selzer ME, Li S. Advances in the Signaling Pathways Downstream of Glial-Scar Axon Growth Inhibitors. Front Cell Neurosci 2020; 14:174. [PMID: 32714150 PMCID: PMC7346763 DOI: 10.3389/fncel.2020.00174] [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] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 05/22/2020] [Indexed: 12/15/2022] Open
Abstract
Axon growth inhibitors generated by reactive glial scars play an important role in failure of axon regeneration after CNS injury in mature mammals. Among the inhibitory factors, chondroitin sulfate proteoglycans (CSPGs) are potent suppressors of axon regeneration and are important molecular targets for designing effective therapies for traumatic brain injury or spinal cord injury (SCI). CSPGs bind with high affinity to several transmembrane receptors, including two members of the leukocyte common antigen related (LAR) subfamily of receptor protein tyrosine phosphatases (RPTPs). Recent studies demonstrate that multiple intracellular signaling pathways downstream of these two RPTPs mediate the growth-inhibitory actions of CSPGs. A better understanding of these signaling pathways may facilitate development of new and effective therapies for CNS disorders characterized by axonal disconnections. This review will focus on recent advances in the downstream signaling pathways of scar-mediated inhibition and their potential as the molecular targets for CNS repair.
Collapse
Affiliation(s)
- Armin Sami
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Department of Anatomy and Cell Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Michael E Selzer
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Department of Neurology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| | - Shuxin Li
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States.,Department of Anatomy and Cell Biology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
| |
Collapse
|
20
|
Kargozar S, Mozafari M, Ghenaatgar-Kasbi M, Baino F. Bioactive Glasses and Glass/Polymer Composites for Neuroregeneration: Should We Be Hopeful? APPLIED SCIENCES 2020; 10:3421. [DOI: 10.3390/app10103421] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Bioactive glasses (BGs) have been identified as highly versatile materials in tissue engineering applications; apart from being used for bone repair for many years, they have recently shown promise for the regeneration of peripheral nerves as well. They can be formulated in different shapes and forms (micro-/nanoparticles, micro-/nanofibers, and tubes), thus potentially meeting the diverse requirements for neuroregeneration. Mechanical and biological improvements in three-dimensional (3D) polymeric scaffolds could be easily provided by adding BGs to their composition. Various types of silicate, borate, and phosphate BGs have been examined for use in neuroregeneration. In general, BGs show good compatibility with the nervous system compartments both in vitro and in vivo. Functionalization and surface modification plus doping with therapeutic ions make BGs even more effective in peripheral nerve regeneration. Moreover, the combination of BGs with conductive polymers is suggested to improve neural cell functions at injured sites. Taking advantage of BGs combined with novel technologies in tissue engineering, like 3D printing, can open new horizons in reconstructive approaches for the nervous system. Although there are great potential opportunities in BG-based therapies for peripheral nerve regeneration, more research should still be performed to carefully assess the pros and cons of BGs in neuroregeneration strategies.
Collapse
Affiliation(s)
- Saeid Kargozar
- Tissue Engineering Research Group (TERG), Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad 917794-8564, Iran
| | - Masoud Mozafari
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran 1449614535, Iran
| | - Maryam Ghenaatgar-Kasbi
- Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad 917794-8564, Iran
| | - Francesco Baino
- Institute of Materials Physics and Engineering, Applied Science and Technology Department, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| |
Collapse
|
21
|
Bighinati A, Focarete ML, Gualandi C, Pannella M, Giuliani A, Beggiato S, Ferraro L, Lorenzini L, Giardino L, Calzà L. Improved Functional Recovery in Rat Spinal Cord Injury Induced by a Drug Combination Administered with an Implantable Polymeric Delivery System. J Neurotrauma 2020; 37:1708-1719. [PMID: 32212901 DOI: 10.1089/neu.2019.6949] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Spinal cord injury (SCI) is an incurable condition, in which a cascade of cellular and molecular events triggered by inflammation and excitotoxicity impairs endogenous regeneration, namely remyelination and axonal outgrowth. We designed a treatment solution based on an implantable biomaterial (electrospun poly (l-lactic acid) [PLLA]) loaded with ibuprofen and triiodothyronine (T3) to counteract inflammation, thus improving endogenous regeneration. In vivo efficacy was tested by implanting the drug-loaded PLLA in the rat model of T8 contusion SCI. We observed the expected recovery of locomotion beginning on day 7. In PLLA-implanted rats (i.e., controls), the recovery stabilized at 21 days post-lesion (DPL), after which no further improvement was observed. On the contrary, in PLLA + ibuprofen (Ibu) + T3 (PLLA-Ibu-T3) rats a further recovery and a significant treatment effect were observed, also confirmed by the gait analysis on 49 DPL. Glutamate release at 24 h and 8 DPL was reduced in PLLA-Ibu-T3- compared to PLLA-implanted rats, such as the estimated lesion volume at 60 DPL. The myelin- and 200-neurofilament-positive area fraction was higher in PLLA-Ibu-T3-implanted rats, where the percentage of astrocytes was significantly reduced. The implant of a PLLA electrospun scaffold loaded with Ibu and T3 significantly improves the endogenous regeneration, leading to an improvement of functional locomotion outcome in the SCI.
Collapse
Affiliation(s)
- Andrea Bighinati
- Department of Veterinary Medical Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Maria Letizia Focarete
- Health Sciences and Technologies (HST) CIRI-SDV, Alma Mater Studiorum-University of Bologna, Bologna, Italy.,Department of Chemistry "Giacomo Ciamician" and National Consortium of Materials Science and Technology (INSTM, Bologna RU), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Chiara Gualandi
- Department of Chemistry "Giacomo Ciamician" and National Consortium of Materials Science and Technology (INSTM, Bologna RU), Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | | | - Alessandro Giuliani
- Department of Veterinary Medical Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Sarah Beggiato
- Department of Life Sciences and Biotechnology, Section of Medicinal and Health Products, University of Ferrara, Ferrara, Italy
| | - Luca Ferraro
- Department of Life Sciences and Biotechnology, Section of Medicinal and Health Products, University of Ferrara, Ferrara, Italy.,Iret Foundation, Ozzano Emilia, Emilia, Italy
| | - Luca Lorenzini
- Department of Veterinary Medical Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| | - Luciana Giardino
- Department of Veterinary Medical Sciences, Alma Mater Studiorum-University of Bologna, Bologna, Italy.,Health Sciences and Technologies (HST) CIRI-SDV, Alma Mater Studiorum-University of Bologna, Bologna, Italy.,Iret Foundation, Ozzano Emilia, Emilia, Italy
| | - Laura Calzà
- Health Sciences and Technologies (HST) CIRI-SDV, Alma Mater Studiorum-University of Bologna, Bologna, Italy.,Iret Foundation, Ozzano Emilia, Emilia, Italy.,Pharmacy and Biotechnology, Alma Mater Studiorum-University of Bologna, Bologna, Italy
| |
Collapse
|
22
|
Ballon Romero SS, Lee YC, Fuh LJ, Chung HY, Hung SY, Chen YH. Analgesic and Neuroprotective Effects of Electroacupuncture in a Dental Pulp Injury Model-A Basic Research. Int J Mol Sci 2020; 21:E2628. [PMID: 32283868 PMCID: PMC7178196 DOI: 10.3390/ijms21072628] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/02/2020] [Accepted: 04/05/2020] [Indexed: 12/18/2022] Open
Abstract
Irreversible pulpitis is an extremely painful condition and its consequence in the central nervous system (CNS) remains unclear. A mouse model of dental pulp injury (DPI) resembles the irreversible pulpitis profile in humans. This study sought to determine whether pain induced by DPI activates microglia and astrocytes in the trigeminal subnucleus caudalis (Vc), as well as increases levels of proinflammatory cytokines, and whether electroacupuncture (EA) can be a potential analgesic and neuroprotective therapy following DPI. Pain behavior was measured via head-withdrawal threshold (HWT) and burrowing behavior at days 1, 3, 7, 14 and 21 after DPI. A marked decrease in HWT and burrowing activity was observed from day 1 to 14 after DPI and no changes were seen on day 21. Microglial and astrocytes activation; along with high cytokine (TNFα, IL-1β, and IL-6) levels, were observed in the Vc at 21 days after DPI. These effects were attenuated by verum (local and distal) EA, as well as oral ibuprofen administration. The results suggest that DPI-induced pain and glial activations in the Vc and EA exert analgesic efficacy at both local and distal acupoints. Furthermore, verum (local and distal) EA might be associated with the modulations of microglial and astrocytes activation.
Collapse
Affiliation(s)
- Sharmely Sharon Ballon Romero
- Graduate Institute of Acupuncture Science, China Medical University, Taichung 40402, Taiwan; (S.S.B.R.); (Y.-C.L.); (H.-Y.C.); (S.-Y.H.)
| | - Yu-Chen Lee
- Graduate Institute of Acupuncture Science, China Medical University, Taichung 40402, Taiwan; (S.S.B.R.); (Y.-C.L.); (H.-Y.C.); (S.-Y.H.)
- Department of Acupuncture, China Medical University Hospital, Taichung 40447, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung 40402, Taiwan
| | - Lih-Jyh Fuh
- School of Dentistry, College of Dentistry, China Medical University; Taichung 40402, Taiwan;
| | - Hsin-Yi Chung
- Graduate Institute of Acupuncture Science, China Medical University, Taichung 40402, Taiwan; (S.S.B.R.); (Y.-C.L.); (H.-Y.C.); (S.-Y.H.)
| | - Shih-Ya Hung
- Graduate Institute of Acupuncture Science, China Medical University, Taichung 40402, Taiwan; (S.S.B.R.); (Y.-C.L.); (H.-Y.C.); (S.-Y.H.)
- Department of Medical Research, China Medical University Hospital, Taichung 40447, Taiwan
| | - Yi-Hung Chen
- Graduate Institute of Acupuncture Science, China Medical University, Taichung 40402, Taiwan; (S.S.B.R.); (Y.-C.L.); (H.-Y.C.); (S.-Y.H.)
- Chinese Medicine Research Center, China Medical University, Taichung 40402, Taiwan
- Department of Photonics and Communication Engineering, Asia University, Taichung 41354, Taiwan
- Brain Disease Research Center, China Medical University Hospital, Taichung 40447, Taiwan
| |
Collapse
|
23
|
Mari L, Behr S, Shea A, Dominguez E, Ricco C, Alcoverro E, Ekiri A, Sanchez-Masian D, De Risio L. Predictors of urinary or fecal incontinence in dogs with thoracolumbar acute non-compressive nucleus pulposus extrusion. J Vet Intern Med 2019; 33:2693-2700. [PMID: 31674064 PMCID: PMC6872617 DOI: 10.1111/jvim.15626] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 09/05/2019] [Indexed: 12/16/2022] Open
Abstract
Background Urinary (UI) and fecal (FI) incontinence occur in up to 7.5% and 32% of dogs, respectively, after thoracolumbar acute noncompressive nucleus pulposus extrusion (ANNPE). Hypotheses/Objectives To investigate clinical, diagnostic, and therapeutic predictors of UI and FI in dogs with ANNPE affecting the T3‐L3 spinal cord segments. Animals Hundred and eighty‐seven dogs with T3‐L3 ANNPE diagnosed based on clinical and MRI findings. Methods Multicenter retrospective study. Data were obtained from medical records and telephone questionnaires and analyzed by logistic regression. Results UI and FI were reported in 17 (9.1%) and 44 (23.5%) dogs, respectively. Paraplegic dogs were 3 times (95% CI = 1.25, 10.87) more likely to develop UI (P = .018) and 4 times (95% CI = 1.94, 12.56) more likely to develop FI (P = .001) compared to nonparaplegic dogs. Dogs with an intramedullary hyperintensity greater than 40% of the cross‐sectional area of the spinal cord at the same level on transverse T2‐weighted MRI images were 4 times more likely to develop UI (95% CI = 1.04, 21.72; P = .045) and FI (95% CI = 1.56, 10.39; P = .004) compared to dogs with smaller lesions. FI was 3 times (95% CI = 1.41, 7.93) more likely in dogs that were not treated with nonsteroidal anti‐inflammatory drugs (NSAIDs) after diagnosis compared to dogs administered NSAIDs (P = .006) and 2 times (95% CI = 1.12, 5.98) more likely in dogs presented with clinical signs compatible with spinal shock compared to dogs without (P = .026). Conclusion and Clinical Importance The identification of clinical, diagnostic, and therapeutic predictors of UI and FI in dogs with T3‐L3 ANNPE can help to approach these autonomic dysfunctions occurring after spinal cord injury.
Collapse
Affiliation(s)
- Lorenzo Mari
- Department of Neurology/Neurosurgery, Centre for Small Animal Studies, Animal Health Trust, Newmarket, Suffolk, UK
| | - Sebastien Behr
- Neurology/Neurosurgery Service, Willows Veterinary Centre and Referral Services, Solihull, UK
| | - Anita Shea
- Department of Neurology/Neurosurgery, Centre for Small Animal Studies, Animal Health Trust, Newmarket, Suffolk, UK
| | - Elisabet Dominguez
- Diagnostic Imaging Unit, Centre for Small Animal Studies, Animal Health Trust, Newmarket, Suffolk, UK
| | - Cristoforo Ricco
- Neurology/Neurosurgery Service, Willows Veterinary Centre and Referral Services, Solihull, UK
| | - Emili Alcoverro
- Department of Small Animal Clinical Science, Institute of Veterinary Science, University of Liverpool, Neston, Cheshire, UK
| | - Abel Ekiri
- School of Veterinary Medicine, University of Surrey, Guildford, Surrey, UK
| | - Daniel Sanchez-Masian
- Department of Small Animal Clinical Science, Institute of Veterinary Science, University of Liverpool, Neston, Cheshire, UK
| | - Luisa De Risio
- Department of Neurology/Neurosurgery, Centre for Small Animal Studies, Animal Health Trust, Newmarket, Suffolk, UK
| |
Collapse
|
24
|
Hassanpour Golakani M, Mohammad MG, Li H, Gamble J, Breit SN, Ruitenberg MJ, Brown DA. MIC-1/GDF15 Overexpression Is Associated with Increased Functional Recovery in Traumatic Spinal Cord Injury. J Neurotrauma 2019; 36:3410-3421. [PMID: 31232176 DOI: 10.1089/neu.2019.6421] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Spinal cord injury (SCI) has devastating consequences, with limited therapeutic options; therefore, improving its functional outcome is a major goal. The outcome of SCI is contributed to by neuroinflammation, which may be a target for improved recovery and quality of life after injury. Macrophage inhibitory cytokine-1/growth differentiation factor 15 (MIC-1/GDF15) has been identified as a potential novel therapy for central nervous system (CNS) injury because it is an immune regulatory cytokine with neurotrophic properties. Here we used MIC-1/GDF15 knockout (KO) and overexpressing/transgenic (Tg) and wild type (WT) animals to explore its putative therapeutic benefits in a mouse model of contusive SCI. MIC-1/GDF15 Tg mice had superior locomotor recovery and reduced secondary tissue loss at 28 days compared with their KO and WT counterparts. Overexpression of MIC-1/GDF15 coincided with increased expression of monocyte chemoattractant protein-1 (MCP-1)/C-C Motif Chemokine Ligand 2 (CCL2) at the lesion site (28 days post-SCI) and enhanced recruitment of inflammatory cells to the injured spinal cord. This inflammatory cellular infiltrate included an increased frequency of macrophages and dendritic cells (DCs) that mostly preceded recruitment of cluster of differentiation (CD)4+ and CD8+ T cells. Collectively, our findings suggest hat MIC-1/GDF15 is associated with beneficial changes in the clinical course of SCI that are characterized by altered post-injury inflammation and improved functional outcome. Further investigation of MIC-1/GDF15 as a novel therapeutic target for traumatic SCI appears warranted.
Collapse
Affiliation(s)
- Masoud Hassanpour Golakani
- St. Vincent's Centre for Applied Medical Research (AMR), St Vincent's Hospital and University of New South Wales (UNSW), Sydney, New South Wales, Australia.,The Neuroinflammation Research Group, Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Mohammad G Mohammad
- Department of Medical Laboratory Sciences, College of Health Sciences and Sharjah Institute for Medical Research, University of Sharjah, Sharjah, United Arab Emirates. Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Hui Li
- St. Vincent's Centre for Applied Medical Research (AMR), St Vincent's Hospital and University of New South Wales (UNSW), Sydney, New South Wales, Australia.,The Neuroinflammation Research Group, Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Joanne Gamble
- The Neuroinflammation Research Group, Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, University of Sydney, Sydney, New South Wales, Australia
| | - Samuel N Breit
- St. Vincent's Centre for Applied Medical Research (AMR), St Vincent's Hospital and University of New South Wales (UNSW), Sydney, New South Wales, Australia
| | - Marc J Ruitenberg
- School of Biomedical Sciences, Faculty of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - David A Brown
- St. Vincent's Centre for Applied Medical Research (AMR), St Vincent's Hospital and University of New South Wales (UNSW), Sydney, New South Wales, Australia.,The Neuroinflammation Research Group, Centre for Immunology and Allergy Research, Westmead Institute for Medical Research, University of Sydney, Sydney, New South Wales, Australia.,Department of Immunopathology, Institute for Clinical Pathology and Medical Research-New South Wales Health Pathology, Westmead Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
25
|
Guercio JR, Kralic JE, Marrotte EJ, James ML. Spinal cord injury pharmacotherapy: Current research & development and competitive commercial landscape as of 2015. J Spinal Cord Med 2019; 42:102-122. [PMID: 29485334 PMCID: PMC6340271 DOI: 10.1080/10790268.2018.1439803] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
CONTEXT Current treatment of spinal cord injury (SCI) focuses on cord stabilization to prevent further injury, rehabilitation, management of non-motor symptoms, and prevention of complications. Currently, no approved treatments are available, and limited treatment options exist for symptoms and complications associated with chronic SCI. This review describes the pharmacotherapy landscape in SCI from both commercial and research and development (R&D) standpoints through March 2015. METHODS Information about specific compounds has been obtained through drug pipeline monographs in the Pharmaprojects® (Citeline, Inc., New York, New York, USA) drug database (current as of a search on May 30, 2014), websites of individual companies with compounds in development for SCI (current as of March 24, 2015), and a literature search of published R&D studies to validate the Pharmaprojects® source for selected compounds (current as of March 24, 2015). RESULTS Types of studies conducted and outcomes measured in earlier phases of development are described for compounds in clinical development Currently four primary mechanisms are under investigation and may yield promising therapeutic targets: 1) neuronal regeneration; 2) neuroprotection (including anti-inflammation); 3) axonal reconnection; and 4) neuromodulation and signal enhancement. Many other compounds are no longer under investigation for SCI are mentioned; however, in most cases, the reason for terminating their development is not clear. CONCLUSION There is urgent need to develop disease-modifying therapy for SCI, yet the commercial landscape remains small and highly fragmented with a paucity of novel late-stage compounds in R&D.
Collapse
Affiliation(s)
- Jason R. Guercio
- North American Partners in Anesthesiology, New Britain, Connecticuit, USA,Correspondence to: Michael L. James, MD, Associate Professor, Brain Injury Translational Research Center, Duke University DUMC 3094, Durham, NC 27710, USA.
| | - Jason E. Kralic
- Innervate BD Solutions, LLC, Hillsborough, North Carolina, USA
| | - Eric J. Marrotte
- Department of Neurology, Brain Injury Translational Research Center, Duke University, Durham, North Carolina, USA
| | - Michael L. James
- Department of Neurology, Brain Injury Translational Research Center, Duke University, Durham, North Carolina, USA,Department of Anesthesiology, Brain Injury Translational Research Center, Duke University, Durham, North Carolina, USA,Correspondence to: Michael L. James, MD, Associate Professor, Brain Injury Translational Research Center, Duke University DUMC 3094, Durham, NC 27710, USA.
| |
Collapse
|
26
|
Rayner MLD, Laranjeira S, Evans RE, Shipley RJ, Healy J, Phillips JB. Developing an In Vitro Model to Screen Drugs for Nerve Regeneration. Anat Rec (Hoboken) 2018; 301:1628-1637. [PMID: 30334365 PMCID: PMC6282521 DOI: 10.1002/ar.23918] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 02/24/2018] [Accepted: 03/25/2018] [Indexed: 12/29/2022]
Abstract
Peripheral nerve injuries (PNI) have a high prevalence and can be debilitating, resulting in life‐long loss or disturbance in end‐organ function, which compromises quality of life for patients. Current therapies use microsurgical approaches but there is the potential for enhancing recovery through other therapeutic modalities such as; cell‐based conduits, gene therapy and small molecules. A number of molecular targets and drugs which have the potential to improve nerve regeneration have been identified, however, there are challenges associated with moving therapies toward clinical translation. Due to the lack of detailed knowledge about the pro‐regenerative effect of potential drug treatments, there is a need for effective in vitro models to screen compounds to inform future pre‐clinical and clinical studies. The interaction between regenerating neurites and supporting Schwann cells is a key feature of the nerve environment, therefore, in vitro models that mimic this cellular association are useful tools. In this study, we have investigated various cell culture models, including simple monolayer systems and more complex 3D‐engineered co‐cultures, as models for use in PNI drug development. Anat Rec, 301:1628–1637, 2018. © 2018 The Authors. The Anatomical Record published by Wiley Periodicals, Inc. on behalf of American Association of Anatomists.
Collapse
Affiliation(s)
- Melissa L D Rayner
- Biomaterials & Tissue Engineering, UCL Eastman Dental Institute, WC1X 8LD, London, UK.,Department of Pharmacology, UCL School of Pharmacy, 29-39 Brunswick Square WC1N 1AX, London, UK.,UCL Centre for Nerve Engineering, University College London, WC1E 6BT, London, UK
| | - Simão Laranjeira
- UCL Centre for Nerve Engineering, University College London, WC1E 6BT, London, UK.,UCL Department of Mechanical Engineering, University College London, WC1E 7JEK, London, UK
| | - Rachael E Evans
- Biomaterials & Tissue Engineering, UCL Eastman Dental Institute, WC1X 8LD, London, UK.,Department of Pharmacology, UCL School of Pharmacy, 29-39 Brunswick Square WC1N 1AX, London, UK.,UCL Centre for Nerve Engineering, University College London, WC1E 6BT, London, UK
| | - Rebecca J Shipley
- UCL Centre for Nerve Engineering, University College London, WC1E 6BT, London, UK.,UCL Department of Mechanical Engineering, University College London, WC1E 7JEK, London, UK
| | - Jess Healy
- Department of Pharmacology, UCL School of Pharmacy, 29-39 Brunswick Square WC1N 1AX, London, UK.,UCL Centre for Nerve Engineering, University College London, WC1E 6BT, London, UK
| | - James B Phillips
- Biomaterials & Tissue Engineering, UCL Eastman Dental Institute, WC1X 8LD, London, UK.,Department of Pharmacology, UCL School of Pharmacy, 29-39 Brunswick Square WC1N 1AX, London, UK.,UCL Centre for Nerve Engineering, University College London, WC1E 6BT, London, UK
| |
Collapse
|
27
|
Pedram MS, Dehghan MM, Shojaee M, Fekrazad R, Sharifi D, Farzan A, Ghasemi S, AliMohammad Kalhori K. Therapeutic effects of simultaneous Photobiomodulation therapy (PBMT) and Meloxicam administration on experimental acute spinal cord injury: Rat animal model. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2018; 189:49-54. [PMID: 30312920 DOI: 10.1016/j.jphotobiol.2018.09.022] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 08/29/2018] [Accepted: 09/25/2018] [Indexed: 11/26/2022]
Abstract
STUDY DESIGN Application of Photobiomodulation therapy (PBMT) and meloxicam in acute spinal cord injury, functional recovery and histological evaluation. OBJECTIVE Evaluation of the effect of simultaneous PBMT and meloxicam on treatment of acute experimental spinal cord injury and comparing it with the effect of application of each of them separately. SETTING The study was conducted at the Department of Surgery & Radiology, Faculty of Veterinary Medicine and Institute of Biomedical Research, University of Tehran, Tehran, Iran. METHODS Twenty four rats were used in this study. A compression injury was induced to the T8-T9 segment of the spinal cord of rats using a Fogarty embolectomy catheter. Rats were randomly divided into 4 groups including: Control group, PBMT (810 nm-200 mw-8 s-2 weeks) group, Meloxicam (1 mg/kg) group, and PBMT and Meloxicam (mixed) group. After inducing injury, hind limb performance of the rats was evaluated, using BBB test and then treatment intervention was performed and continued for 2 weeks. RESULTS Four weeks after injury induction, BBB test results were significantly higher in all treatment groups in comparison to control group, however, there were no significant differences among the treatment groups. In addition, histological findings revealed no significant difference between all 4 study groups. CONCLUSION According to the results of this study we can conclude that simultaneous and separate application of PBMT and Meloxicam play an effective role in treatment of acute spinal cord injuries.
Collapse
Affiliation(s)
- Mir Sepehr Pedram
- Department of Surgery & Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran; Institute of Biomedical Research, University of Tehran, Tehran, Iran
| | - Mohammad Mehdi Dehghan
- Department of Surgery & Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran; Institute of Biomedical Research, University of Tehran, Tehran, Iran.
| | - Maryam Shojaee
- Department of Surgery & Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Reza Fekrazad
- Department of Periodontology, Dental Faculty - Laser research center in medical Sciences, AJA University of Medical Sciences & International Network for Photo Medicine and Photo Dynamic Therapy (INPMPDT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Davood Sharifi
- Department of Surgery & Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Arash Farzan
- Department of Orthodontics, School of Density, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Setareh Ghasemi
- Department of Surgery & Radiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Katayoun AliMohammad Kalhori
- Department of Oral & Maxillofacial Pathology, Dental Faculty, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| |
Collapse
|
28
|
Schiavone S, Trabace L. Small Molecules: Therapeutic Application in Neuropsychiatric and Neurodegenerative Disorders. Molecules 2018; 23:molecules23020411. [PMID: 29438357 PMCID: PMC6017408 DOI: 10.3390/molecules23020411] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Revised: 02/05/2018] [Accepted: 02/07/2018] [Indexed: 12/13/2022] Open
Abstract
In recent years, an increasing number of studies have been published, focusing on the potential therapeutic use of small catalytic agents with strong biological properties. So far, most of these works have only regarded specific clinical fields, such as oncology, infectivology and general pathology, in particular with respect to the treatment of significant inflammatory processes. However, interesting data on possible therapeutic applications of small molecules for the treatment of neuropsychiatric and neurodegenerative illnesses are emerging, especially with respect to the possibility to modulate the cellular redox state. Indeed, a crucial role of redox dysregulation in the pathogenesis of these disorders has been widely demonstrated by both pre-clinical and clinical studies, being the reduction of the total amount of free radicals a promising novel therapeutic approach for these diseases. In this review, we focused our interest on studies published during the last ten years reporting therapeutic potential of small molecules for the treatment of neuropsychiatric and neurodegenerative disorders, also based on the biological efficiency of these compounds in detecting intracellular disturbances induced by increased production of reactive oxygen species.
Collapse
Affiliation(s)
- Stefania Schiavone
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli, 20, 71122 Foggia, Italy.
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Via Napoli, 20, 71122 Foggia, Italy.
| |
Collapse
|
29
|
Wood RL, Karlinsey KS, Thompson AD, Rigby MN, Boatright GD, Pitt WG, Roeder BL, Steffensen SC, Cook AD. Baseline effects of lysophosphatidylcholine and nerve growth factor in a rat model of sciatic nerve regeneration after crush injury. Neural Regen Res 2018; 13:846-853. [PMID: 29863015 PMCID: PMC5998639 DOI: 10.4103/1673-5374.232479] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Schwann cells play a major role in helping heal injured nerves. They help clear debris, produce neurotrophins, upregulate neurotrophin receptors, and form bands of Büngner to guide the healing nerve. But nerves do not always produce enough neurotrophins and neurotrophin receptors to repair themselves. Nerve growth factor (NGF) is an important neurotrophin for promoting nerve healing and lysophosphatidylcholine (LPC) has been shown to stimulate NGF receptors (NGFR). This study tested the administration of a single intraneural injection of LPC (1 mg/mL for single LPC injection and 10 mg/mL for multiple LPC injections) at day 0 and one (day 7), two (days 5 and 7), or three (days 5, 7, and 9) injections of NGF (160 ng/mL for single injections and 80 ng/mL for multiple injections) to determine baseline effects on crushed sciatic nerves in rats. The rats were randomly divided into four groups: control, crush, crush-NGF, and crush-LPC-NGF. The healing of the nerves was measured weekly by monitoring gait; electrophysiological parameters: compound muscle action potential (CMAP) amplitudes; and morphological parameters: total fascicle areas, myelinated fiber counts, fiber densities, fiber packing, and mean g-ratio values at weeks 3 and 6. The crush, crush-NGF, and crush-LPC-NGF groups statistically differed from the control group for all six weeks for the electrophysiological parameters but only differed from the control group at week 3 for the morphological parameters. The crush, crush-NGF, and crush-LPC-NGF groups did not differ from each other over the course of the study. Single injections of LPC and NGF one week apart or multiple treatments of NGF at 5, 7 and 9 days post-injury did not alter the healing rate of the sciatic nerves during weeks 1-6 of the study. These findings are important to define the baseline effects of NGF and LPC injections, as part of a larger effort to determine the minimal dose regimen of NGF to regenerate peripheral nerves.
Collapse
Affiliation(s)
- Ryan L Wood
- Department of Chemical Engineering, Brigham Young University, Provo, UT, USA
| | | | | | - Mark N Rigby
- Neuroscience Center, Brigham Young University, Provo, UT, USA
| | | | - William G Pitt
- Department of Chemical Engineering, Brigham Young University, Provo, UT, USA
| | | | - Scott C Steffensen
- Neuroscience Center; Department of Psychology, Brigham Young University, Provo, UT, USA
| | - Alonzo D Cook
- Department of Chemical Engineering; Neuroscience Center, Brigham Young University, Provo, UT, USA
| |
Collapse
|
30
|
Acute spinal cord injury: A review of pathophysiology and potential of non-steroidal anti-inflammatory drugs for pharmacological intervention. J Chem Neuroanat 2018; 87:25-31. [DOI: 10.1016/j.jchemneu.2017.08.001] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 08/07/2017] [Accepted: 08/07/2017] [Indexed: 12/21/2022]
|
31
|
Ferrera P, Zepeda A, Arias C. Nonsteroidal anti-inflammatory drugs attenuate amyloid-β protein-induced actin cytoskeletal reorganization through Rho signaling modulation. Cell Mol Neurobiol 2017; 37:1311-1318. [PMID: 28124209 PMCID: PMC11482099 DOI: 10.1007/s10571-017-0467-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 01/19/2017] [Indexed: 01/15/2023]
Abstract
Amyloid-β protein (Aβ) neurotoxicity occurs along with the reorganization of the actin-cytoskeleton through the activation of the Rho GTPase pathway. In addition to the classical mode of action of the non-steroidal anti-inflammatory drugs (NSAIDs), indomethacin, and ibuprofen have Rho-inhibiting effects. In order to evaluate the role of the Rho GTPase pathway on Aβ-induced neuronal death and on neuronal morphological modifications in the actin cytoskeleton, we explored the role of NSAIDS in human-differentiated neuroblastoma cells exposed to Aβ. We found that Aβ induced neurite retraction and promoted the formation of different actin-dependent structures such as stress fibers, filopodia, lamellipodia, and ruffles. In the presence of Aβ, both NSAIDs prevented neurite collapse and formation of stress fibers without affecting the formation of filopodia and lamellipodia. Similar results were obtained when the downstream effector, Rho kinase inhibitor Y27632, was applied in the presence of Aβ. These results demonstrate the potential benefits of the Rho-inhibiting NSAIDs in reducing Aβ-induced effects on neuronal structural alterations.
Collapse
Affiliation(s)
- Patricia Ferrera
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70-228, 04510, México, DF, Mexico
| | - Angélica Zepeda
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70-228, 04510, México, DF, Mexico
| | - Clorinda Arias
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, AP 70-228, 04510, México, DF, Mexico.
| |
Collapse
|
32
|
Pires LR, Lopes CDF, Salvador D, Rocha DN, Pêgo AP. Ibuprofen-loaded fibrous patches-taming inhibition at the spinal cord injury site. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2017; 28:157. [PMID: 28894995 DOI: 10.1007/s10856-017-5967-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 08/19/2017] [Indexed: 06/07/2023]
Abstract
It is now widely accepted that a therapeutic strategy for spinal cord injury (SCI) demands a multi-target approach. Here we propose the use of an easily implantable bilayer polymeric patch based on poly(trimethylene carbonate-co-ε-caprolactone) (P(TMC-CL)) that combines physical guidance cues provided by electrospun aligned fibres and the delivery of ibuprofen, as a mean to reduce the inhibitory environment at the lesion site by taming RhoA activation. Bilayer patches comprised a solvent cast film onto which electrospun aligned fibres have been deposited. Both layers were loaded with ibuprofen. In vitro release (37°C, in phosphate buffered saline) of the drug from the loaded scaffolds under sink condition was found to occur in the first 24 h. The released ibuprofen was shown to retain its bioactivity, as indicated by the reduction of RhoA activation when the neuronal-like cell line ND7/23 was challenged with lysophosphatidic acid. Ibuprofen-loaded P(TMC-CL) bilayer scaffolds were successfully implanted in vivo in a dorsal hemisection rat SCI model mediating the reduction of RhoA activation after 5 days of implantation in comparison to plain P(TMC-CL) scaffolds. Immunohistochemical analysis of the tissue shows βIII tubulin positive cells close to the ibuprofen-loaded patches further supporting the use of this strategy in the context of regeneration after a lesion in the spinal cord.
Collapse
Affiliation(s)
- Liliana R Pires
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INL- International Iberian Nanotechnology Laboratory, Braga, Portugal
| | - Cátia D F Lopes
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Daniela Salvador
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
| | - Daniela N Rocha
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
| | - Ana Paula Pêgo
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal.
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto (ICBAS), Porto, Portugal.
- Faculdade de Engenharia da Universidade do Porto (FEUP), Porto, Portugal.
| |
Collapse
|
33
|
Kopp MA, Liebscher T, Watzlawick R, Martus P, Laufer S, Blex C, Schindler R, Jungehulsing GJ, Knüppel S, Kreutzträger M, Ekkernkamp A, Dirnagl U, Strittmatter SM, Niedeggen A, Schwab JM. SCISSOR-Spinal Cord Injury Study on Small molecule-derived Rho inhibition: a clinical study protocol. BMJ Open 2016; 6:e010651. [PMID: 27466236 PMCID: PMC4964175 DOI: 10.1136/bmjopen-2015-010651] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 02/25/2016] [Accepted: 05/19/2016] [Indexed: 11/30/2022] Open
Abstract
INTRODUCTION The approved analgesic and anti-inflammatory drugs ibuprofen and indometacin block the small GTPase RhoA, a key enzyme that impedes axonal sprouting after axonal damage. Inhibition of the Rho pathway in a central nervous system-effective manner requires higher dosages compared with orthodox cyclooxygenase-blocking effects. Preclinical studies on spinal cord injury (SCI) imply improved motor recovery after ibuprofen/indometacin-mediated Rho inhibition. This has been reassessed by a meta-analysis of the underlying experimental evidence, which indicates an overall effect size of 20.2% regarding motor outcome achieved after ibuprofen/indometacin treatment compared with vehicle controls. In addition, ibuprofen/indometacin may also limit sickness behaviour, non-neurogenic systemic inflammatory response syndrome (SIRS), neuropathic pain and heterotopic ossifications after SCI. Consequently, 'small molecule'-mediated Rho inhibition after acute SCI warrants clinical investigation. METHODS AND ANALYSIS Protocol of an investigator-initiated clinical open-label pilot trial on high-dose ibuprofen treatment after acute traumatic, motor-complete SCI. A sample of n=12 patients will be enrolled in two cohorts treated with 2400 mg/day ibuprofen for 4 or 12 weeks, respectively. The primary safety end point is an occurrence of serious adverse events, primarily gastroduodenal bleedings. Secondary end points are pharmacokinetics, feasibility and preliminary effects on neurological recovery, neuropathic pain and heterotopic ossifications. The primary safety analysis is based on the incidence of severe gastrointestinal bleedings. Additional analyses will be mainly descriptive and casuistic. ETHICS AND DISSEMINATION The clinical trial protocol was approved by the responsible German state Ethics Board, and the Federal Institute for Drugs and Medical Devices. The study complies with the Declaration of Helsinki, the principles of Good Clinical Practice and all further applicable regulations. This safety and pharmacokinetics trial informs the planning of a subsequent randomised controlled trial. Regardless of the result of the primary and secondary outcome assessments, the clinical trial will be reported as a publication in a peer-reviewed journal. TRIAL REGISTRATION NUMBER NCT02096913; Pre-results.
Collapse
Affiliation(s)
- Marcel A Kopp
- Department of Neurology and Experimental Neurology, Spinal Cord Injury Research, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Thomas Liebscher
- Treatment Centre for Spinal Cord Injury, Trauma Hospital Berlin, Berlin, Germany
| | - Ralf Watzlawick
- Department of Neurology and Experimental Neurology, Spinal Cord Injury Research, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Peter Martus
- Department of Clinical Epidemiology and Applied Biostatistics, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Stefan Laufer
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, Eberhard Karls Universität Tübingen, Tübingen, Germany
| | - Christian Blex
- Department of Neurology and Experimental Neurology, Spinal Cord Injury Research, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Ralf Schindler
- Division of Nephrology and Intensive Care, Department of Internal Medicine, Campus Virchow-Klinikum, Charité-University Medicine Berlin, Berlin, Germany
| | - Gerhard J Jungehulsing
- Department of Neurology, Jüdisches Krankenhaus Berlin, Berlin, Germany Department of Neurology and Experimental Neurology, Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Sven Knüppel
- Department of Epidemiology, German Institute of Human Nutrition Potsdam-Rehbrücke, Germany
| | - Martin Kreutzträger
- Treatment Centre for Spinal Cord Injury, Trauma Hospital Berlin, Berlin, Germany
| | - Axel Ekkernkamp
- Trauma Surgery and Orthopedics Clinic, Trauma Hospital Berlin, Berlin, Germany
| | - Ulrich Dirnagl
- Department of Neurology and Experimental Neurology, Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Stephen M Strittmatter
- Department of Neurology, Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, USA
| | - Andreas Niedeggen
- Treatment Centre for Spinal Cord Injury, Trauma Hospital Berlin, Berlin, Germany
| | - Jan M Schwab
- Department of Neurology and Experimental Neurology, Spinal Cord Injury Research, Charité-Universitätsmedizin Berlin, Berlin, Germany Treatment Centre for Spinal Cord Injury, Trauma Hospital Berlin, Berlin, Germany Department of Neurology, Spinal Cord Injury Division, The Ohio State University, Wexner Medical Center, Columbus, USA Department of Neuroscience and Center for Brain and Spinal Cord Repair, Department of Physical Medicine and Rehabilitation, The Neurological Institute, The Ohio State University, Wexner Medical Center, Columbus, USA
| |
Collapse
|
34
|
Cragg JJ, Haefeli J, Jutzeler CR, Röhrich F, Weidner N, Saur M, Maier DD, Kalke YB, Schuld C, Curt A, Kramer JK. Effects of Pain and Pain Management on Motor Recovery of Spinal Cord–Injured Patients. Neurorehabil Neural Repair 2016; 30:753-61. [DOI: 10.1177/1545968315624777] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background. Approximately 60% of patients suffering from acute spinal cord injury (SCI) develop pain within days to weeks after injury, which ultimately persists into chronic stages. To date, the consequences of pain after SCI have been largely examined in terms of interfering with quality of life. Objective. The objective of this study was to examine the effects of pain and pain management on neurological recovery after SCI. Methods. We analyzed clinical data in a prospective multicenter observational cohort study in patients with SCI. Using mixed effects regression techniques, total motor and sensory scores were modelled at 1, 3, 6, and 12 months postinjury. Results. A total of 225 individuals were included in the study (mean age: 45.8 ± 18 years, 80% male). At 1 month postinjury, 28% of individuals with SCI reported at- or below-level neuropathic pain. While pain classification showed no effect on neurological outcomes, individuals administered anticonvulsant medications at 1 month postinjury showed significant reductions in pain intensity (2 points over 1 year; P < .05) and greater recovery in total motor scores (7.3 points over 1 year; P < .05). This drug effect on motor recovery remained significant after adjustment for injury level and injury severity, pain classification, and pain intensity. Conclusion. While initial pain classification and intensity did not reveal an effect on motor recovery following acute SCI, anticonvulsants conferred a significant beneficial effect on motor outcomes. Early intervention with anticonvulsants may have effects beyond pain management and warrant further studies to evaluate the therapeutic effectiveness in human SCI.
Collapse
Affiliation(s)
- Jacquelyn J. Cragg
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada
| | - Jenny Haefeli
- Brain and Spinal Injury Center, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | | | - Frank Röhrich
- Berufsgenossenschaftliche Klinik Bergmanstrost of Halle, Halle, Germany
| | - Norbert Weidner
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Marion Saur
- Orthopädische Klinik, Hessisch Lichtenau, Germany
| | | | - Yorck B. Kalke
- RKU Universitäts und Rehabilitationskliniken Ulm, Ulm, Germany
| | - Christian Schuld
- Spinal Cord Injury Center, Heidelberg University Hospital, Heidelberg, Germany
| | - Armin Curt
- University Hospital Balgrist, Zurich, Switzerland
| | - John K. Kramer
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada
- School of Kinesiology, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
35
|
Hu J, Yu Q, Xie L, Zhu H. Targeting the blood-spinal cord barrier: A therapeutic approach to spinal cord protection against ischemia-reperfusion injury. Life Sci 2016; 158:1-6. [PMID: 27329433 DOI: 10.1016/j.lfs.2016.06.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 06/15/2016] [Accepted: 06/17/2016] [Indexed: 12/15/2022]
Abstract
One of the principal functions of physical barriers between the blood and central nervous system protects system (i.e., blood brain barrier and blood-spinal cord barrier) is the protection from toxic and pathogenic agents in the blood. Disruption of blood-spinal cord barrier (BSCB) plays a key role in spinal cord ischemia-reperfusion injury (SCIRI). Following SCIRI, the permeability of the BSCB increases. Maintaining the integrity of the BSCB alleviates the spinal cord injury after spinal cord ischemia. This review summarizes current knowledge of the structure and function of the BSCB and its changes following SCIRI, as well as the prevention and cure of SCIRI and the role of the BSCB.
Collapse
Affiliation(s)
- Ji Hu
- Department of Anesthesiology, Liyuan Hospital of Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430077, Hubei Province, China.
| | - Qijing Yu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China.
| | - Lijie Xie
- Department of Anesthesiology, Liyuan Hospital of Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430077, Hubei Province, China
| | - Hongfei Zhu
- Department of Anesthesiology, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, Hubei Province, China
| |
Collapse
|
36
|
Schill EM, Lake JI, Tusheva OA, Nagy N, Bery SK, Foster L, Avetisyan M, Johnson SL, Stenson WF, Goldstein AM, Heuckeroth RO. Ibuprofen slows migration and inhibits bowel colonization by enteric nervous system precursors in zebrafish, chick and mouse. Dev Biol 2016; 409:473-88. [PMID: 26586201 PMCID: PMC4862364 DOI: 10.1016/j.ydbio.2015.09.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2015] [Revised: 08/31/2015] [Accepted: 09/07/2015] [Indexed: 12/17/2022]
Abstract
Hirschsprung Disease (HSCR) is a potentially deadly birth defect characterized by the absence of the enteric nervous system (ENS) in distal bowel. Although HSCR has clear genetic causes, no HSCR-associated mutation is 100% penetrant, suggesting gene-gene and gene-environment interactions determine HSCR occurrence. To test the hypothesis that certain medicines might alter HSCR risk we treated zebrafish with medications commonly used during early human pregnancy and discovered that ibuprofen caused HSCR-like absence of enteric neurons in distal bowel. Using fetal CF-1 mouse gut slice cultures, we found that ibuprofen treated enteric neural crest-derived cells (ENCDC) had reduced migration, fewer lamellipodia and lower levels of active RAC1/CDC42. Additionally, inhibiting ROCK, a RHOA effector and known RAC1 antagonist, reversed ibuprofen effects on migrating mouse ENCDC in culture. Ibuprofen also inhibited colonization of Ret+/- mouse bowel by ENCDC in vivo and dramatically reduced bowel colonization by chick ENCDC in culture. Interestingly, ibuprofen did not affect ENCDC migration until after at least three hours of exposure. Furthermore, mice deficient in Ptgs1 (COX 1) and Ptgs2 (COX 2) had normal bowel colonization by ENCDC and normal ENCDC migration in vitro suggesting COX-independent effects. Consistent with selective and strain specific effects on ENCDC, ibuprofen did not affect migration of gut mesenchymal cells, NIH3T3, or WT C57BL/6 ENCDC, and did not affect dorsal root ganglion cell precursor migration in zebrafish. Thus, ibuprofen inhibits ENCDC migration in vitro and bowel colonization by ENCDC in vivo in zebrafish, mouse and chick, but there are cell type and strain specific responses. These data raise concern that ibuprofen may increase Hirschsprung disease risk in some genetically susceptible children.
Collapse
Affiliation(s)
- Ellen Merrick Schill
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Jonathan I Lake
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Olga A Tusheva
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Nandor Nagy
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, 55 Fruit St., Boston, MA 02114, USA; Department of Human Morphology and Developmental Biology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Saya K Bery
- Department of Pediatrics, The Children's Hospital of Philadelphia Research Institute and the Perelman School of Medicine at the University of Pennsylvania, Abramson Research Center, 3615 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Lynne Foster
- Department of Internal Medicine, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Marina Avetisyan
- Department of Pediatrics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Stephen L Johnson
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - William F Stenson
- Department of Internal Medicine, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Allan M Goldstein
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, 55 Fruit St., Boston, MA 02114, USA
| | - Robert O Heuckeroth
- Department of Pediatrics, The Children's Hospital of Philadelphia Research Institute and the Perelman School of Medicine at the University of Pennsylvania, Abramson Research Center, 3615 Civic Center Blvd, Philadelphia, PA 19104, USA.
| |
Collapse
|
37
|
Liu J, Gao HY, Wang XF. The role of the Rho/ROCK signaling pathway in inhibiting axonal regeneration in the central nervous system. Neural Regen Res 2015; 10:1892-1896. [PMID: 26807132 PMCID: PMC4705809 DOI: 10.4103/1673-5374.170325] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/17/2015] [Indexed: 02/05/2023] Open
Abstract
The Rho/Rho-associated coiled-coil containing protein kinase (Rho/ROCK) pathway is a major signaling pathway in the central nervous system, transducing inhibitory signals to block regeneration. After central nervous system damage, the main cause of impaired regeneration is the presence of factors that strongly inhibit regeneration in the surrounding microenvironment. These factors signal through the Rho/ROCK signaling pathway to inhibit regeneration. Therefore, a thorough understanding of the Rho/ROCK signaling pathway is crucial for advancing studies on regeneration and repair of the injured central nervous system.
Collapse
Affiliation(s)
- Jing Liu
- Department of Neonatology & NICU of Bayi Children's Hospital, General Hospital of Beijing Military Command of Chinese PLA, Beijing, China
| | - Hong-yan Gao
- Department of Neonatology & NICU of Bayi Children's Hospital, General Hospital of Beijing Military Command of Chinese PLA, Beijing, China
| | - Xiao-feng Wang
- Department of Neonatology & NICU of Bayi Children's Hospital, General Hospital of Beijing Military Command of Chinese PLA, Beijing, China
- Department of Neonatology, People's Hospital of Rizhao, Rizhao, Shangdong Province, China
| |
Collapse
|
38
|
Barbosa RA, Nunes TLGM, Nunes TLGM, da Paixão AO, Neto RB, Moura S, Albuquerque Junior RLC, Cândido EAF, Padilha FF, Quintans-Júnior LJ, Gomes MZ, Cardoso JC. Hydroalcoholic extract of red propolis promotes functional recovery and axon repair after sciatic nerve injury in rats. PHARMACEUTICAL BIOLOGY 2015; 54:993-1004. [PMID: 26511070 PMCID: PMC11132607 DOI: 10.3109/13880209.2015.1091844] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Revised: 07/16/2015] [Accepted: 09/04/2015] [Indexed: 06/05/2023]
Abstract
CONTEXT Peripheral axon injury and degeneration are often mediated by oxidative stress and inflammation. The hydroalcoholic extract of the red propolis (HERP) has attracted great attention because of its antioxidant and anti-inflammatory activities. OBJECTIVE The objective of this work is to study the effect of HERP on nerve repair and functional recovery after sciatic nerve injury (SNI) in rats. MATERIALS AND METHODS The chemical markers in HERP were identified using high-resolution mass spectroscopy. After axonotmesis of sciatic nerve, ibuprofen (IBP) and HERP treatments were orally administered for 28 d. Behavioural tests were performed weekly after SNI. The myelinated axon number was counted using morphometric analysis. RESULTS The compounds found in HERP were pinocembrin, formononetin, vestitol, and biochanin A. The animals that underwent SNI showed a significant decrease in motor function based on the Basso, Beattie and Bresnahan scale and sciatic functional index compared with sham animals until 7 d after the surgery (p < 0.05). After 14 and 21 d, the SNI groups treated with either HERP or IBP showed significant improvement (p < 0.01), and the SNI group treated with HERP 10 mg/kg showed accelerated motor recovery compared with the other groups (p < 0.01). SNI caused also a reduction in the myelinated axon counts, and treatment with HERP 10 mg/kg induced a significant increase in the number of myelinated fibres compared with all other groups. CONCLUSION HERP promoted regenerative responses and accelerated functional recovery after sciatic nerve crush. Thus, it can be considered to be a new strategy or complementary therapy for treating nerve injuries.
Collapse
Affiliation(s)
| | | | | | | | - Reinaldo Belo Neto
- Department of Healthy and Environment, Tiradentes University, Aracaju, Brazil
| | - Sidnei Moura
- Department of Technology, University of Caxias Do Sul, Caxias Do Sul, Brazil
| | - Ricardo Luiz Cavalcanti Albuquerque Junior
- Department of Healthy and Environment, Tiradentes University, Aracaju, Brazil
- Department of Biomaterials, Morphology and Experimental Pathology, Research and Technology Institute (ITP), Aracaju, Brazil
| | - Edna Aragão Farias Cândido
- Department of Healthy and Environment, Tiradentes University, Aracaju, Brazil
- Department of Biomaterials, Morphology and Experimental Pathology, Research and Technology Institute (ITP), Aracaju, Brazil
| | - Francine Ferreira Padilha
- Department of Healthy and Environment, Tiradentes University, Aracaju, Brazil
- Department of Biomaterials, Morphology and Experimental Pathology, Research and Technology Institute (ITP), Aracaju, Brazil
| | | | - Margarete Zanardo Gomes
- Department of Healthy and Environment, Tiradentes University, Aracaju, Brazil
- Department of Biomaterials, Morphology and Experimental Pathology, Research and Technology Institute (ITP), Aracaju, Brazil
| | - Juliana Cordeiro Cardoso
- Department of Healthy and Environment, Tiradentes University, Aracaju, Brazil
- Department of Biomaterials, Morphology and Experimental Pathology, Research and Technology Institute (ITP), Aracaju, Brazil
| |
Collapse
|
39
|
Rocha DN, Ferraz-Nogueira JP, Barrias CC, Relvas JB, Pêgo AP. Extracellular environment contribution to astrogliosis-lessons learned from a tissue engineered 3D model of the glial scar. Front Cell Neurosci 2015; 9:377. [PMID: 26483632 PMCID: PMC4586948 DOI: 10.3389/fncel.2015.00377] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 09/09/2015] [Indexed: 12/19/2022] Open
Abstract
Glial scars are widely seen as a (bio)mechanical barrier to central nervous system regeneration. Due to the lack of a screening platform, which could allow in-vitro testing of several variables simultaneously, up to now no comprehensive study has addressed and clarified how different lesion microenvironment properties affect astrogliosis. Using astrocytes cultured in alginate gels and meningeal fibroblast conditioned medium, we have built a simple and reproducible 3D culture system of astrogliosis mimicking many features of the glial scar. Cells in this 3D culture model behave similarly to scar astrocytes, showing changes in gene expression (e.g., GFAP) and increased extra-cellular matrix production (chondroitin 4 sulfate and collagen), inhibiting neuronal outgrowth. This behavior being influenced by the hydrogel network properties. Astrocytic reactivity was found to be dependent on RhoA activity, and targeting RhoA using shRNA-mediated lentivirus reduced astrocytic reactivity. Further, we have shown that chemical inhibition of RhoA with ibuprofen or indirectly targeting RhoA by the induction of extracellular matrix composition modification with chondroitinase ABC, can diminish astrogliosis. Besides presenting the extracellular matrix as a key modulator of astrogliosis, this simple, controlled and reproducible 3D culture system constitutes a good scar-like system and offers great potential in future neurodegenerative mechanism studies, as well as in drug screenings envisaging the development of new therapeutic approaches to minimize the effects of the glial scar in the context of central nervous system disease.
Collapse
Affiliation(s)
- Daniela N Rocha
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto Porto, Portugal ; Instituto de Investigação e Inovação em Saúde, Universidade do Porto Porto, Portugal ; Faculdade de Engenharia, Universidade do Porto Porto, Portugal
| | - José P Ferraz-Nogueira
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto Porto, Portugal ; Glia Cell Biology Group, Instituto de Biologia Celular e Molecular, Universidade do Porto Porto, Portugal
| | - Cristina C Barrias
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto Porto, Portugal ; Instituto de Investigação e Inovação em Saúde, Universidade do Porto Porto, Portugal
| | - João B Relvas
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto Porto, Portugal ; Glia Cell Biology Group, Instituto de Biologia Celular e Molecular, Universidade do Porto Porto, Portugal
| | - Ana P Pêgo
- Instituto de Engenharia Biomédica (INEB), Universidade do Porto Porto, Portugal ; Instituto de Investigação e Inovação em Saúde, Universidade do Porto Porto, Portugal ; Faculdade de Engenharia, Universidade do Porto Porto, Portugal ; Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto Porto, Portugal
| |
Collapse
|
40
|
Pires LR, Pêgo AP. Bridging the lesion-engineering a permissive substrate for nerve regeneration. Regen Biomater 2015; 2:203-14. [PMID: 26816642 PMCID: PMC4669012 DOI: 10.1093/rb/rbv012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Revised: 07/21/2015] [Accepted: 06/30/2015] [Indexed: 01/30/2023] Open
Abstract
Biomaterial-based strategies to restore connectivity after lesion at the spinal cord are focused on bridging the lesion and providing an favourable substrate and a path for axonal re-growth. Following spinal cord injury (SCI) a hostile environment for neuronal cell growth is established by the activation of multiple inhibitory mechanisms that hamper regeneration to occur. Implantable scaffolds can provide mechanical support and physical guidance for axon re-growth and, at the same time, contribute to alleviate the hostile environment by the in situ delivery of therapeutic molecules and/or relevant cells. Basic research on SCI has been contributing with the description of inhibitory mechanisms for regeneration as well as identifying drugs/molecules that can target inhibition. This knowledge is the background for the development of combined strategies with biomaterials. Additionally, scaffold design is significantly evolving. From the early simple hollow conduits, scaffolds with complex architectures that can modulate cell fate are currently being tested. A number of promising pre-clinical studies combining scaffolds, cells, drugs and/or nucleic acids are reported in the open literature. Overall, it is considered that to address the multi-factorial inhibitory environment of a SCI, a multifaceted therapeutic approach is imperative. The progress in the identification of molecules that target inhibition after SCI and its combination with scaffolds and/or cells are described and discussed in this review.
Collapse
Affiliation(s)
- Liliana R. Pires
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua do Campo Alegre 823, 4150-180 Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
- Faculdade de Engenharia—Universidade do Porto (FEUP), Porto, Portugal and
| | - Ana P. Pêgo
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua do Campo Alegre 823, 4150-180 Porto, Portugal
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Portugal
- Faculdade de Engenharia—Universidade do Porto (FEUP), Porto, Portugal and
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| |
Collapse
|
41
|
Martin AR, Aleksanderek I, Fehlings MG. Diagnosis and Acute Management of Spinal Cord Injury: Current Best Practices and Emerging Therapies. CURRENT TRAUMA REPORTS 2015. [DOI: 10.1007/s40719-015-0020-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
42
|
Liu X, Wang Y, Yang J, Liu Y, Zhou D, Hou M, Xiang L. Anti-edema effect of melatonin on spinal cord injury in rats. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2015; 159:220-6. [PMID: 25916278 DOI: 10.5507/bp.2015.012] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Accepted: 03/10/2015] [Indexed: 01/01/2023] Open
Abstract
AIM To determine the anti-edema effects of melatonin on spinal cord injury (SCI) in rats. METHODS A total of 150 adult male Sprague-Dawley rats were randomly allocated to the following three groups (n=50): a sham group which underwent laminectomy without dural compression; an SCI group, which underwent laminectomy followed by SCI and received saline i.p. immediately after injury and then daily for 2 days; an MT group, which underwent laminectomy followed by SCI and received a 100 mg/kg dose of melatonin i.p. immediately after SCI and then daily for 2 days. The cords were removed at 12, 24, 48 and 72 h after surgery in every group. Spinal cord edema was evaluated by determining the spinal cord water content. Expressions of AQP4 and GFAP positive cells in injured spinal cord were detected by immunohistochemical staining, and protein expressions of AQP4 and GFAP were detected by Western blotting. RESULTS Spinal cord water content was obviously increased after SCI, which was maintained almost unchanged by melatonin treatment (100 mg/kg) at 12 h after injury but was significantly reduced from 24 h to 72 h. The expressions of AQP4 and GFAP increased in the injured spinal cord segments, which were decreased by melatonin treatment (100 mg/kg) between 24 h and 72 h after SCI. CONCLUSIONS Melatonin (100 mg/kg) had anti-edema effects after acute SCI probably by down-regulating the expression level of AQP4 protein, and it may eliminate astrocytic swelling after SCI through down-regulating the expression level of GFAP protein.
Collapse
Affiliation(s)
- Xinwei Liu
- Department of Orthopedics, General Hospital of Shenyang Military Area Command of Chinese PLA, Rescue Center of Severe Wound and Trauma of Chinese PLA, Shenyang 110016, China
| | - Yu Wang
- Department of Orthopedics, General Hospital of Shenyang Military Area Command of Chinese PLA, Rescue Center of Severe Wound and Trauma of Chinese PLA, Shenyang 110016, China
| | - Jishun Yang
- The Medical Department, No. 100 Hospital of Chinese PLA, Suzhou 215007, China
| | - Yunen Liu
- Department of Emergency, General Hospital of Shenyang Military Area Command of Chinese PLA, Rescue Center of Severe Wound and Trauma of Chinese PLA, Shenyang 110016, China
| | - Dapeng Zhou
- Department of Orthopedics, General Hospital of Shenyang Military Area Command of Chinese PLA, Rescue Center of Severe Wound and Trauma of Chinese PLA, Shenyang 110016, China
| | - Mingxiao Hou
- Department of Emergency, General Hospital of Shenyang Military Area Command of Chinese PLA, Rescue Center of Severe Wound and Trauma of Chinese PLA, Shenyang 110016, China
| | - Liangbi Xiang
- Department of Orthopedics, General Hospital of Shenyang Military Area Command of Chinese PLA, Rescue Center of Severe Wound and Trauma of Chinese PLA, Shenyang 110016, China
| |
Collapse
|
43
|
Roloff F, Scheiblich H, Dewitz C, Dempewolf S, Stern M, Bicker G. Enhanced neurite outgrowth of human model (NT2) neurons by small-molecule inhibitors of Rho/ROCK signaling. PLoS One 2015; 10:e0118536. [PMID: 25714396 PMCID: PMC4340918 DOI: 10.1371/journal.pone.0118536] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2014] [Accepted: 01/20/2015] [Indexed: 12/02/2022] Open
Abstract
Axonal injury in the adult human central nervous system often results in loss of sensation and motor functions. Promoting regeneration of severed axons requires the inactivation of growth inhibitory influences from the tissue environment and stimulation of the neuron intrinsic growth potential. Especially glial cell derived factors, such as chondroitin sulfate proteoglycans, Nogo-A, myelin-associated glycoprotein, and myelin in general, prevent axon regeneration. Most of the glial growth inhibiting factors converge onto the Rho/ROCK signaling pathway in neurons. Although conditions in the injured nervous system are clearly different from those during neurite outgrowth in vitro, here we use a chemical approach to manipulate Rho/ROCK signalling with small-molecule agents to encourage neurite outgrowth in cell culture. The development of therapeutic treatments requires drug testing not only on neurons of experimental animals, but also on human neurons. Using human NT2 model neurons, we demonstrate that the pain reliever Ibuprofen decreases RhoA (Ras homolog gene family, member A GTPase) activation and promotes neurite growth. Inhibition of the downstream effector Rho kinase by the drug Y-27632 results in a strong increase in neurite outgrowth. Conversely, activation of the Rho pathway by lysophosphatidic acid results in growth cone collapse and eventually to neurite retraction. Finally, we show that blocking of Rho kinase, but not RhoA results in an increase in neurons bearing neurites. Due to its anti-inflammatory and neurite growth promoting action, the use of a pharmacological treatment of damaged neural tissue with Ibuprofen should be explored.
Collapse
Affiliation(s)
- Frank Roloff
- Division of Cell Biology, University of Veterinary Medicine Hannover, Bischofsholer Damm 15/102, 30173, Hannover, Germany
| | - Hannah Scheiblich
- Division of Cell Biology, University of Veterinary Medicine Hannover, Bischofsholer Damm 15/102, 30173, Hannover, Germany
| | - Carola Dewitz
- Division of Cell Biology, University of Veterinary Medicine Hannover, Bischofsholer Damm 15/102, 30173, Hannover, Germany
| | - Silke Dempewolf
- Division of Cell Biology, University of Veterinary Medicine Hannover, Bischofsholer Damm 15/102, 30173, Hannover, Germany
| | - Michael Stern
- Division of Cell Biology, University of Veterinary Medicine Hannover, Bischofsholer Damm 15/102, 30173, Hannover, Germany
| | - Gerd Bicker
- Division of Cell Biology, University of Veterinary Medicine Hannover, Bischofsholer Damm 15/102, 30173, Hannover, Germany
- Center for Systems Neuroscience, Hannover, Germany
- * E-mail:
| |
Collapse
|
44
|
Overcoming Drug Development Bottlenecks With Repurposing: Old drugs learn new tricks. Nat Med 2014; 20:590-1. [PMID: 24901567 DOI: 10.1038/nm.3595] [Citation(s) in RCA: 136] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
45
|
Chondroitin sulphate N-acetylgalactosaminyl-transferase-1 inhibits recovery from neural injury. Nat Commun 2014; 4:2740. [PMID: 24220492 PMCID: PMC3831297 DOI: 10.1038/ncomms3740] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 10/10/2013] [Indexed: 12/20/2022] Open
Abstract
Extracellular factors that inhibit axon growth and intrinsic factors that promote it affect neural regeneration. Therapies targeting any single gene have not yet simultaneously optimized both types of factors. Chondroitin sulphate (CS), a glycosaminoglycan, is the most abundant extracellular inhibitor of axon growth. Here we show that mice carrying a gene knockout for CS N-acetylgalactosaminyltransferase-1 (T1), a key enzyme in CS biosynthesis, recover more completely from spinal cord injury than wild-type mice and even chondroitinase ABC-treated mice. Notably, synthesis of heparan sulphate (HS), a glycosaminoglycan promoting axonal growth, is also upregulated in TI knockout mice because HS-synthesis enzymes are induced in the mutant neurons. Moreover, chondroitinase ABC treatment never induces HS upregulation. Taken together, our results indicate that regulation of a single gene, T1, mediates excellent recovery from spinal cord injury by optimizing counteracting effectors of axon regeneration—an extracellular inhibitor of CS and intrinsic promoters, namely, HS-synthesis enzymes. The glycosaminoglycan chondroitin sulphate inhibits axon growth. Here the authors show that mice deficient in chondroitin sulphate biosynthesis have increased levels of heparan sulphate, which is more efficient than chondroitinase in supporting recovery from spinal cord injury.
Collapse
|
46
|
Streijger F, Lee JHT, Duncan GJ, Ng MTL, Assinck P, Bhatnagar T, Plunet WT, Tetzlaff W, Kwon BK. Combinatorial treatment of acute spinal cord injury with ghrelin, ibuprofen, C16, and ketogenic diet does not result in improved histologic or functional outcome. J Neurosci Res 2014; 92:870-83. [PMID: 24658967 DOI: 10.1002/jnr.23372] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Revised: 11/28/2013] [Accepted: 01/20/2014] [Indexed: 11/09/2022]
Abstract
Because of the complex, multifaceted nature of spinal cord injury (SCI), it is widely believed that a combination of approaches will be superior to individual treatments. Therefore, we employed a rat model of cervical SCI to evaluate the combination of four noninvasive treatments that individually have been reported to be effective for acute SCI during clinically relevant therapeutic time windows. These treatments included ghrelin, ibuprofen, C16, and ketogenic diet (KD). These were selected not only because of their previously reported efficacy in SCI models but also for their potentially different mechanisms of action. The administration of ghrelin, ibuprofen, C16, and KD several hours to days postinjury was based on previous observations by others that each treatment had profound effects on the pathophysiology and functional outcome following SCI. Here we showed that, with the exception of a modest improvement in performance on the Montoya staircase test at 8-10 weeks postinjury, the combinatorial treatment with ghrelin, ibuprofen, C16, and KD did not result in any significant improvements in the rearing test, grooming test, or horizontal ladder. Histologic analysis of the spinal cords did not reveal any significant differences in tissue sparing between treatment and control groups. Although single approaches of ghrelin, ibuprofen, C16, and KD have been reported to be beneficial after SCI, our results show that the combination of the four interventions did not confer significant functional or histological improvements in a cervical model of SCI. Possible interactions among the treatments may have negated their beneficial effects, emphasizing the challenges that have to be addressed when considering combinatorial drug therapies for SCI.
Collapse
Affiliation(s)
- F Streijger
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, British Columbia, Canada; Department of Zoology, University of British Columbia, Vancouver, British Columbia, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Inosine enhances axon sprouting and motor recovery after spinal cord injury. PLoS One 2013; 8:e81948. [PMID: 24312612 PMCID: PMC3846725 DOI: 10.1371/journal.pone.0081948] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 10/18/2013] [Indexed: 11/19/2022] Open
Abstract
Although corticospinal tract axons cannot regenerate long distances after spinal cord injury, they are able to sprout collateral branches rostral to an injury site that can help form compensatory circuits in cases of incomplete lesions. We show here that inosine enhances the formation of compensatory circuits after a dorsal hemisection of the thoracic spinal cord in mature rats and improves coordinated limb use. Inosine is a naturally occurring metabolite of adenosine that crosses the cell membrane and, in neurons, activates Mst3b, a protein kinase that is part of a signal transduction pathway that regulates axon outgrowth. Compared to saline-treated controls, rats with dorsal hemisections that were treated with inosine showed three times as many synaptic contacts between corticospinal tract collaterals and long propriospinal interneurons that project from the cervical cord to the lumbar level. Inosine-treated rats also showed stronger serotonergic reinnervation of the lumbar cord than saline-treated controls, and performed well above controls in both open-field testing and a horizontal ladder rung-walking test. Inosine was equally effective whether delivered intracranially or intravenously, and has been shown to be safe for other indications in humans. Thus, inosine might be a useful therapeutic for improving outcome after spinal cord injury.
Collapse
|
48
|
Silva NA, Sousa N, Reis RL, Salgado AJ. From basics to clinical: a comprehensive review on spinal cord injury. Prog Neurobiol 2013; 114:25-57. [PMID: 24269804 DOI: 10.1016/j.pneurobio.2013.11.002] [Citation(s) in RCA: 555] [Impact Index Per Article: 46.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2013] [Revised: 11/12/2013] [Accepted: 11/12/2013] [Indexed: 12/15/2022]
Abstract
Spinal cord injury (SCI) is a devastating neurological disorder that affects thousands of individuals each year. Over the past decades an enormous progress has been made in our understanding of the molecular and cellular events generated by SCI, providing insights into crucial mechanisms that contribute to tissue damage and regenerative failure of injured neurons. Current treatment options for SCI include the use of high dose methylprednisolone, surgical interventions to stabilize and decompress the spinal cord, and rehabilitative care. Nonetheless, SCI is still a harmful condition for which there is yet no cure. Cellular, molecular, rehabilitative training and combinatorial therapies have shown promising results in animal models. Nevertheless, work remains to be done to ascertain whether any of these therapies can safely improve patient's condition after human SCI. This review provides an extensive overview of SCI research, as well as its clinical component. It starts covering areas from physiology and anatomy of the spinal cord, neuropathology of the SCI, current clinical options, neuronal plasticity after SCI, animal models and techniques to assess recovery, focusing the subsequent discussion on a variety of promising neuroprotective, cell-based and combinatorial therapeutic approaches that have recently moved, or are close, to clinical testing.
Collapse
Affiliation(s)
- Nuno A Silva
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui L Reis
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal; 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, 4806-909 Caldas das Taipas, Guimarães, Portugal
| | - António J Salgado
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
49
|
Redondo-Castro E, Navarro X. Chronic ibuprofen administration reduces neuropathic pain but does not exert neuroprotection after spinal cord injury in adult rats. Exp Neurol 2013; 252:95-103. [PMID: 24246280 DOI: 10.1016/j.expneurol.2013.11.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Accepted: 11/05/2013] [Indexed: 01/02/2023]
Abstract
Ibuprofen is commonly used as an anti-inflammatory analgesic drug, although it is not amongst the first-line treatments for neuropathic pain. Its main effects are mediated by non-specific inhibition of COX enzymes, but it also exerts some COX-independent effects, such as the inhibition of RhoA signaling and the modulation of glial activity. These effects have boosted the use of ibuprofen as a tool to promote axonal regeneration and to increase functional recovery after neural injuries, although with controversial results showing positive and negative outcomes of ibuprofen treatment in several experimental models. We have evaluated the effects of ibuprofen administered at 60 mg/kg twice a day to rats subjected to a mild spinal cord contusion. Our results indicate that ibuprofen ameliorates mechanical hyperalgesia in rats by reducing central hyperexcitability, but failed to produce improvements in the recovery of locomotion. Despite an early effect on reducing microglial reactivity, the ibuprofen treatment did not provide histological evidence of neuroprotection; indeed the volume of cord tissue spared rostral to the lesion was decreased in ibuprofen treated rats. In summary, the early modulation of neuroinflammation produced by the administration of ibuprofen seems to eventually lead to a worse resolution of detrimental events occurring in the secondary injury phase, but also to reduce the development of neuropathic pain.
Collapse
Affiliation(s)
- Elena Redondo-Castro
- Group of Neuroplasticity and Regeneration, Institute of Neurosciences and Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain
| | - Xavier Navarro
- Group of Neuroplasticity and Regeneration, Institute of Neurosciences and Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain.
| |
Collapse
|
50
|
Boato F, Rosenberger K, Nelissen S, Geboes L, Peters EM, Nitsch R, Hendrix S. Absence of IL-1β positively affects neurological outcome, lesion development and axonal plasticity after spinal cord injury. J Neuroinflammation 2013; 10:6. [PMID: 23317037 PMCID: PMC3585738 DOI: 10.1186/1742-2094-10-6] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2012] [Accepted: 12/07/2012] [Indexed: 11/10/2022] Open
Abstract
Precise crosstalk between the nervous and immune systems is important for neuroprotection and axon plasticity after injury. Recently, we demonstrated that IL-1β acts as a potent inducer of neurite outgrowth from organotypic brain slices in vitro, suggesting a potential function of IL-1β in axonal plasticity. Here, we have investigated the effects of IL-1β on axon plasticity during glial scar formation and on functional recovery in a mouse model of spinal cord compression injury (SCI). We used an IL-1β deficiency model (IL-1βKO mice) and administered recombinant IL-1β. In contrast to our hypothesis, the histological analysis revealed a significantly increased lesion width and a reduced number of corticospinal tract fibers caudal to the lesion center after local application of recombinant IL-1β. Consistently, the treatment significantly worsened the neurological outcome after SCI in mice compared with PBS controls. In contrast, the absence of IL-1β in IL-1βKO mice significantly improved recovery from SCI compared with wildtype mice. Histological analysis revealed a smaller lesion size, reduced lesion width and greatly decreased astrogliosis in the white matter, while the number of corticospinal tract fibers increased significantly 5 mm caudal to the lesion in IL-1βKO mice relative to controls. Our study for the first time characterizes the detrimental effects of IL-1β not only on lesion development (in terms of size and glia activation), but also on the plasticity of central nervous system axons after injury.
Collapse
Affiliation(s)
- Francesco Boato
- Department of Morphology & BIOMED Institute, Campus Diepenbeek, Hasselt University, Agoralaan Gebouw C, Diepenbeek, BE 3590, Belgium
| | | | | | | | | | | | | |
Collapse
|