1
|
Stacchiotti C, Mazzella di Regnella S, Cinotti M, Spalloni A, Volpe E. Neuroinflammation and Amyotrophic Lateral Sclerosis: Recent Advances in Anti-Inflammatory Cytokines as Therapeutic Strategies. Int J Mol Sci 2025; 26:3854. [PMID: 40332510 PMCID: PMC12028049 DOI: 10.3390/ijms26083854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
Neuroinflammation is an inflammatory response occurring within the central nervous system (CNS). The process is marked by the production of pro-inflammatory cytokines, chemokines, small-molecule messengers, and reactive oxygen species. Microglia and astrocytes are primarily involved in this process, while endothelial cells and infiltrating blood cells contribute to neuroinflammation when the blood-brain barrier (BBB) is damaged. Neuroinflammation is increasingly recognized as a pathological hallmark of several neurological diseases, including amyotrophic lateral sclerosis (ALS), and is closely linked to neurodegeneration, another key feature of ALS. In fact, neurodegeneration is a pathological trigger for inflammation, and neuroinflammation, in turn, contributes to motor neuron (MN) degeneration through the induction of synaptic dysfunction, neuronal death, and inhibition of neurogenesis. Importantly, resolution of acute inflammation is crucial for avoiding chronic inflammation and tissue destruction. Inflammatory processes are mediated by soluble factors known as cytokines, which are involved in both promoting and inhibiting inflammation. Cytokines with anti-inflammatory properties may exert protective roles in neuroinflammatory diseases, including ALS. In particular, interleukin (IL)-10, transforming growth factor (TGF)-β, IL-4, IL-13, and IL-9 have been shown to exert an anti-inflammatory role in the CNS. Other recently emerging immune regulatory cytokines in the CNS include IL-35, IL-25, IL-37, and IL-27. This review describes the current understanding of neuroinflammation in ALS and highlights recent advances in the role of anti-inflammatory cytokines within CNS with a particular focus on their potential therapeutic applications in ALS. Furthermore, we discuss current therapeutic strategies aimed at enhancing the anti-inflammatory response to modulate neuroinflammation in this disease.
Collapse
Affiliation(s)
- Costanza Stacchiotti
- Molecular Neuroimmunology Unit, Santa Lucia Foundation, 00143 Rome, Italy; (C.S.); (S.M.d.R.); (M.C.); (E.V.)
- Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Simona Mazzella di Regnella
- Molecular Neuroimmunology Unit, Santa Lucia Foundation, 00143 Rome, Italy; (C.S.); (S.M.d.R.); (M.C.); (E.V.)
| | - Miriam Cinotti
- Molecular Neuroimmunology Unit, Santa Lucia Foundation, 00143 Rome, Italy; (C.S.); (S.M.d.R.); (M.C.); (E.V.)
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, 00185 Rome, Italy
| | - Alida Spalloni
- Molecular Neurobiology Unit, Santa Lucia Foundation, 00143 Rome, Italy
| | - Elisabetta Volpe
- Molecular Neuroimmunology Unit, Santa Lucia Foundation, 00143 Rome, Italy; (C.S.); (S.M.d.R.); (M.C.); (E.V.)
| |
Collapse
|
2
|
Hellenbrand DJ, Lee JS, Mickelson EJ, Baer MC, Ott EL, Martinson NR, Ceelen MR, Hilger KH, Nielsen BE, Jacobs AN, Mishra RR, Hurley SA, Murphy WL, Hanna AS. Mineral coated microparticles delivering Interleukin-4, Interleukin-10, and Interleukin-13 reduce inflammation and improve function after spinal cord injury in a rat. Exp Neurol 2025; 386:115179. [PMID: 39914642 DOI: 10.1016/j.expneurol.2025.115179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 01/29/2025] [Accepted: 02/02/2025] [Indexed: 02/09/2025]
Abstract
After spinal cord injury (SCI) there is excessive inflammation and extensive infiltration of immune cells that leads to additional neural damage. Interleukin (IL)-4, IL-10, and IL-13 are anti-inflammatories that have been shown to reduce several pro-inflammatory species, alter macrophage state, and provide neuroprotection. However, these anti-inflammatories have a short half-life, do not cross the blood-spinal cord barrier, and large systemic doses of ant-inflammatory cytokines can cause increased susceptibility to infections. In this study, we used mineral coated microparticles (MCMs) to bind, stabilize and deliver biologically active IL-4, IL-10, and IL-13 in a sustained manner directly to the injury site. Rats with a T10 SCI were given an intraspinal injection of cytokine-loaded MCMs 6 h post-injury. Testing of 27 cytokine/chemokine levels 24 h post-injury demonstrated that MCMs delivering IL-4, IL-10, and IL-13 significantly reduced inflammation (P < 0.0001). Rats treated with MCMs+(IL-4, IL-10, IL-13) had significantly higher Basso-Beattie-Bresnahan locomotor rating scores (P = 0.0021), Ladder Rung Test scores (P = 0.0021), and significantly longer latency threshold with the Hargreaves Test (P = 0.0123), compared to Injured Controls. Analyses of post-fixed spinal cords revealed significantly less spinal cord atrophy (P = 0.0344) in rats treated with MCMs+(IL-4, IL-10, IL-13), and diffusion tensor imaging tractography revealed significantly more tracts spanning the injury site (P = 0.0025) in rats treated with MCMs+(IL-4, IL-10, IL-13) compared to Injured Controls. In conclusion, MCMs delivering IL-4, IL-10, and IL-13 significantly reduced inflammation post-SCI, resulting in significantly less spinal cord damage and a significant improvement in hind limb function.
Collapse
Affiliation(s)
- Daniel J Hellenbrand
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jae Sung Lee
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Ethan J Mickelson
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
| | - Matthew C Baer
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
| | - Emily L Ott
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
| | - Natalie R Martinson
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
| | - Matthew R Ceelen
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
| | - Keegan H Hilger
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
| | - Brooke E Nielsen
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
| | - Alison N Jacobs
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
| | - Raveena R Mishra
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA
| | - Samuel A Hurley
- Department of Radiology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - William L Murphy
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI 53705, USA; Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA; Forward BIO Institute, University of Wisconsin-Madison, Madison, WI 53705, USA.
| | - Amgad S Hanna
- Department of Neurosurgery, University of Wisconsin-Madison School of Medicine and Public Health, Madison, WI 53705, USA; Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705, USA.
| |
Collapse
|
3
|
Liu Y, Zhao C, Zhang R, Pang Y, Li L, Feng S. Progression of mesenchymal stem cell regulation on imbalanced microenvironment after spinal cord injury. Stem Cell Res Ther 2024; 15:343. [PMID: 39354635 PMCID: PMC11446099 DOI: 10.1186/s13287-024-03914-x] [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: 04/27/2024] [Accepted: 09/01/2024] [Indexed: 10/03/2024] Open
Abstract
Spinal cord injury (SCI) results in significant neural damage and inhibition of axonal regeneration due to an imbalanced microenvironment. Extensive evidence supports the efficacy of mesenchymal stem cell (MSC) transplantation as a therapeutic approach for SCI. This review aims to present an overview of MSC regulation on the imbalanced microenvironment following SCI, specifically focusing on inflammation, neurotrophy and axonal regeneration. The application, limitations and future prospects of MSC transplantation are discussed as well. Generally, a comprehensive perspective is provided for the clinical translation of MSC transplantation for SCI.
Collapse
Affiliation(s)
- Yifan Liu
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
- Institute of Medical Sciences, The Second Hospital of Shandong University, Shandong University Center for Orthopaedics, Cheeloo College of Medicine, Shandong University, Jinan, 250033, Shandong, China
| | - Chenxi Zhao
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
| | - Rong Zhang
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
| | - Yilin Pang
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China
| | - Linquan Li
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China
| | - Shiqing Feng
- Department of Orthopaedics, Qilu Hospital of Shandong University, Shandong University Centre for Orthopaedics, Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, 107 West Wenhua Road, Lixia District, Jinan, 250012, Shandong, China.
- Institute of Medical Sciences, The Second Hospital of Shandong University, Shandong University Center for Orthopaedics, Cheeloo College of Medicine, Shandong University, Jinan, 250033, Shandong, China.
- Department of Orthopedics, International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Tianjin Medical University General Hospital, 154 Anshan Road, Heping District, Tianjin, 300052, China.
| |
Collapse
|
4
|
Schepers M, Hendrix S, Mussen F, van Breedam E, Ponsaerts P, Lemmens S, Hellings N, Ricciarelli R, Fedele E, Bruno O, Brullo C, Prickaerts J, Van Broeckhoven J, Vanmierlo T. Amelioration of functional and histopathological consequences after spinal cord injury through phosphodiesterase 4D (PDE4D) inhibition. Neurotherapeutics 2024; 21:e00372. [PMID: 38760316 PMCID: PMC11284540 DOI: 10.1016/j.neurot.2024.e00372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 04/15/2024] [Accepted: 05/01/2024] [Indexed: 05/19/2024] Open
Abstract
Spinal cord injury (SCI) is a life-changing event that severely impacts the patient's quality of life. Modulating neuroinflammation, which exacerbates the primary injury, and stimulating neuro-regenerative repair mechanisms are key strategies to improve functional recovery. Cyclic adenosine monophosphate (cAMP) is a second messenger crucially involved in both processes. Following SCI, intracellular levels of cAMP are known to decrease over time. Therefore, preventing cAMP degradation represents a promising strategy to suppress inflammation while stimulating regeneration. Intracellular cAMP levels are controlled by its hydrolyzing enzymes phosphodiesterases (PDEs). The PDE4 family is most abundantly expressed in the central nervous system (CNS) and its inhibition has been shown to be therapeutically relevant for managing SCI pathology. Unfortunately, the use of full PDE4 inhibitors at therapeutic doses is associated with severe emetic side effects, hampering their translation toward clinical applications. Therefore, in this study, we evaluated the effect of inhibiting specific PDE4 subtypes (PDE4B and PDE4D) on inflammatory and regenerative processes following SCI, as inhibitors selective for these subtypes have been demonstrated to be well-tolerated. We reveal that administration of the PDE4D inhibitor Gebr32a, even when starting 2 dpi, but not the PDE4B inhibitor A33, improved functional as well as histopathological outcomes after SCI, comparable to results obtained with the full PDE4 inhibitor roflumilast. Furthermore, using a luminescent human iPSC-derived neurospheroid model, we show that PDE4D inhibition stabilizes neural viability by preventing apoptosis and stimulating neuronal differentiation. These findings strongly suggest that specific PDE4D inhibition offers a novel therapeutic approach for SCI.
Collapse
Affiliation(s)
- Melissa Schepers
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, 6229ER Maastricht, the Netherlands; University MS Centre (UMSC) Hasselt - Pelt, Belgium
| | - Sven Hendrix
- Institute for Translational Medicine, Medical School Hamburg, 20457 Hamburg, Germany
| | - Femke Mussen
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, 6229ER Maastricht, the Netherlands; Department of Immunology and Infection, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium
| | - Elise van Breedam
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, 2610 Wilrijk, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, 2610 Wilrijk, Belgium
| | - Stefanie Lemmens
- Department of Immunology and Infection, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium
| | - Niels Hellings
- University MS Centre (UMSC) Hasselt - Pelt, Belgium; Department of Immunology and Infection, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium
| | - Roberta Ricciarelli
- IRCCS Ospedale Policlinico San Martino, 16100 Genoa, Italy; Department of Experimental Medicine, Section of General Pathology, University of Genova, 16100 Genoa, Italy
| | - Ernesto Fedele
- IRCCS Ospedale Policlinico San Martino, 16100 Genoa, Italy; Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genoa, 16100 Genoa, Italy
| | - Olga Bruno
- Department of Pharmacy, Section of Medicinal Chemistry, University of Genoa, 16100 Genoa, Italy
| | - Chiara Brullo
- Department of Pharmacy, Section of Medicinal Chemistry, University of Genoa, 16100 Genoa, Italy
| | - Jos Prickaerts
- Peitho Translational, 6229ER Maastricht, the Netherlands
| | - Jana Van Broeckhoven
- University MS Centre (UMSC) Hasselt - Pelt, Belgium; Department of Immunology and Infection, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium
| | - Tim Vanmierlo
- Department of Neuroscience, Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, 3500 Hasselt, Belgium; Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, 6229ER Maastricht, the Netherlands; University MS Centre (UMSC) Hasselt - Pelt, Belgium.
| |
Collapse
|
5
|
Guo J, Yang T, Zhang W, Yu K, Xu X, Li W, Song L, Gu X, Cao R, Cui S. Inhibition of CD44 suppresses the formation of fibrotic scar after spinal cord injury via the JAK2/STAT3 signaling pathway. iScience 2024; 27:108935. [PMID: 38323002 PMCID: PMC10846335 DOI: 10.1016/j.isci.2024.108935] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 11/17/2023] [Accepted: 01/12/2024] [Indexed: 02/08/2024] Open
Abstract
Fibrotic scar is one of the main impediments to axon regeneration following spinal cord injury (SCI). In this study, we found that CD44 was upregulated during the formation of fibrotic scar, and blocking CD44 by IM7 caused downregulation of fibrosis-related extracellular matrix proteins at both 2 and 12 weeks post-spinal cord injury. More Biotinylated dextran amine (BDA)-traced corticospinal tract axons crossed the scar area and extended into the distal region after IM7 administration. A recovery of motor and sensory function was observed based on Basso Mouse Scale (BMS) scores and tail-flick test. In vitro experiments revealed that inhibiting CD44 and JAK2/STAT3 signaling pathway decreased the proliferation, differentiation, and migration of fibroblasts induced by the inflammatory supernatant. Collectively, these findings highlight the critical role of CD44 and its downstream JAK2/STAT3 signaling pathway in fibrotic scar formation, suggesting a potential therapeutic target for SCI.
Collapse
Affiliation(s)
- Jin Guo
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, Jilin Province 130033, China
| | - Tuo Yang
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, Jilin Province 130033, China
| | - Weizhong Zhang
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, Jilin Province 130033, China
| | - Kaiming Yu
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, Jilin Province 130033, China
| | - Xiong Xu
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, Jilin Province 130033, China
| | - Weizhen Li
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, Jilin Province 130033, China
| | - Lili Song
- Department of Hand & Microsurgery, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Rangjuan Cao
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, Jilin Province 130033, China
| | - Shusen Cui
- Department of Hand and Foot Surgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province 130033, China
- Key Laboratory of Peripheral Nerve Injury and Regeneration of Jilin Province, Changchun, Jilin Province 130033, China
| |
Collapse
|
6
|
Wang J, Tian F, Cao L, Du R, Tong J, Ding X, Yuan Y, Wang C. Macrophage polarization in spinal cord injury repair and the possible role of microRNAs: A review. Heliyon 2023; 9:e22914. [PMID: 38125535 PMCID: PMC10731087 DOI: 10.1016/j.heliyon.2023.e22914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 11/17/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
The prevention, treatment, and rehabilitation of spinal cord injury (SCI) have always posed significant medical challenges. After mechanical injury, disturbances in microcirculation, edema formation, and the generation of free radicals lead to additional damage, impeding effective repair processes and potentially exacerbating further dysfunction. In this context, inflammatory responses, especially the activation of macrophages, play a pivotal role. Different phenotypes of macrophages have distinct effects on inflammation. Activation of classical macrophage cells (M1) promotes inflammation, while activation of alternative macrophage cells (M2) inhibits inflammation. The polarization of macrophages is crucial for disease healing. A non-coding RNA, known as microRNA (miRNA), governs the polarization of macrophages, thereby reducing inflammation following SCI and facilitating functional recovery. This study elucidates the inflammatory response to SCI, focusing on the infiltration of immune cells, specifically macrophages. It examines their phenotype and provides an explanation of their polarization mechanisms. Finally, this paper introduces several well-known miRNAs that contribute to macrophage polarization following SCI, including miR-155, miR-130a, and miR-27 for M1 polarization, as well as miR-22, miR-146a, miR-21, miR-124, miR-223, miR-93, miR-132, and miR-34a for M2 polarization. The emphasis is placed on their potential therapeutic role in SCI by modulating macrophage polarization, as well as the present developments and obstacles of miRNA clinical therapy.
Collapse
Affiliation(s)
- Jiawei Wang
- School and Hospital of Stomatology, Shanxi Medical University, Shanxi Taiyuan, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Taiyuan, China
| | - Feng Tian
- School and Hospital of Stomatology, Shanxi Medical University, Shanxi Taiyuan, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Taiyuan, China
| | - Lili Cao
- School and Hospital of Stomatology, Shanxi Medical University, Shanxi Taiyuan, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Taiyuan, China
| | - Ruochen Du
- Experimental Animal Center, Shanxi Medical University, Shanxi Taiyuan, China
| | - Jiahui Tong
- School and Hospital of Stomatology, Shanxi Medical University, Shanxi Taiyuan, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Taiyuan, China
| | - Xueting Ding
- Experimental Animal Center, Shanxi Medical University, Shanxi Taiyuan, China
| | - Yitong Yuan
- Experimental Animal Center, Shanxi Medical University, Shanxi Taiyuan, China
| | - Chunfang Wang
- School and Hospital of Stomatology, Shanxi Medical University, Shanxi Taiyuan, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Taiyuan, China
| |
Collapse
|
7
|
O'Carroll R, Reynolds JP, Al-Roqi M, Aiyegbusi ED, Dooley D. ThermoCyte: an inexpensive open-source temperature control system for in vitro live-cell imaging. ROYAL SOCIETY OPEN SCIENCE 2023; 10:231037. [PMID: 38034122 PMCID: PMC10685113 DOI: 10.1098/rsos.231037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 11/06/2023] [Indexed: 12/02/2023]
Abstract
Live-cell imaging is a common technique in microscopy to investigate dynamic cellular behaviour and permits the accurate and relevant analysis of a wide range of cellular and tissue parameters, such as motility, cell division, wound healing responses and calcium (Ca2+) signalling in cell lines, primary cell cultures and ex vivo preparations. Furthermore, this can occur under many experimental conditions, making live-cell imaging indispensable for biological research. Systems which maintain cells at physiological conditions outside of a CO2 incubator are often bulky, expensive and use proprietary components. Here we present an inexpensive, open-source temperature control system for in vitro live-cell imaging. Our system 'ThermoCyte', which is constructed from standard electronic components, enables precise tuning, control and logging of a temperature 'set point' for imaging cells at physiological temperature. We achieved stable thermal dynamics, with reliable temperature cycling and a standard deviation of 0.42°C over 1 h. Furthermore, the device is modular in nature and is adaptable to the researcher's specific needs. This represents simple, inexpensive and reliable tool for laboratories to carry out custom live-cell imaging protocols, on a standard laboratory bench, at physiological temperature.
Collapse
Affiliation(s)
- Ross O'Carroll
- School of Medicine, Health Sciences Centre, University College Dublin, Belfield, Dublin D04 V1W8, Ireland
| | - James P. Reynolds
- School of Medicine, Health Sciences Centre, University College Dublin, Belfield, Dublin D04 V1W8, Ireland
- Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin D04 V1W8, Ireland
| | - Mazen Al-Roqi
- School of Medicine, Health Sciences Centre, University College Dublin, Belfield, Dublin D04 V1W8, Ireland
| | - Emmanuelle Damilola Aiyegbusi
- School of Medicine, Health Sciences Centre, University College Dublin, Belfield, Dublin D04 V1W8, Ireland
- Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin D04 V1W8, Ireland
| | - Dearbhaile Dooley
- School of Medicine, Health Sciences Centre, University College Dublin, Belfield, Dublin D04 V1W8, Ireland
- Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin D04 V1W8, Ireland
| |
Collapse
|
8
|
Walsh CM, Wychowaniec JK, Costello L, Brougham DF, Dooley D. An In Vitro and Ex Vivo Analysis of the Potential of GelMA Hydrogels as a Therapeutic Platform for Preclinical Spinal Cord Injury. Adv Healthc Mater 2023; 12:e2300951. [PMID: 37114899 PMCID: PMC11468190 DOI: 10.1002/adhm.202300951] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Indexed: 04/29/2023]
Abstract
Spinal cord injury (SCI) is a devastating condition with no curative therapy currently available. Immunomodulation can be applied as a therapeutic strategy to drive alternative immune cell activation and promote a proregenerative injury microenvironment. Locally injected hydrogels carrying immunotherapeutic cargo directly to injured tissue offer an encouraging treatment approach from an immunopharmacological perspective. Gelatin methacrylate (GelMA) hydrogels are promising in this regard, however, detailed analysis on the immunogenicity of GelMA in the specific context of the SCI microenvironment is lacking. Here, the immunogenicity of GelMA hydrogels formulated with a translationally relevant photoinitiator is analyzed in vitro and ex vivo. 3% (w/v) GelMA, synthesized from gelatin type-A, is first identified as the optimal hydrogel formulation based on mechanical properties and cytocompatibility. Additionally, 3% GelMA-A does not alter the expression profile of key polarization markers in BV2 microglia or RAW264.7 macrophages after 48 h. Finally, it is shown for the first time that 3% GelMA-A can support the ex vivo culture of primary murine organotypic spinal cord slices for 14 days with no direct effect on glial fibrillary acidic protein (GFAP+ ) astrocyte or ionized calcium-binding adaptor molecule 1 (Iba-1+ ) microglia reactivity. This provides evidence that GelMA hydrogels can act as an immunotherapeutic hydrogel-based platform for preclinical SCI.
Collapse
Affiliation(s)
- Ciara M. Walsh
- School of MedicineHealth Sciences CentreUniversity College DublinBelfieldDublinD04 V1W8Ireland
- UCD Conway Institute of Biomolecular & Biomedical ResearchUniversity College DublinBelfieldDublinD04 V1W8Ireland
| | - Jacek K. Wychowaniec
- School of ChemistryUniversity College DublinBelfieldDublinD04 V1W8Ireland
- AO Research Institute DavosClavadelerstrasse 8Davos7270Switzerland
| | - Louise Costello
- School of MedicineHealth Sciences CentreUniversity College DublinBelfieldDublinD04 V1W8Ireland
| | - Dermot F. Brougham
- School of ChemistryUniversity College DublinBelfieldDublinD04 V1W8Ireland
| | - Dearbhaile Dooley
- School of MedicineHealth Sciences CentreUniversity College DublinBelfieldDublinD04 V1W8Ireland
- UCD Conway Institute of Biomolecular & Biomedical ResearchUniversity College DublinBelfieldDublinD04 V1W8Ireland
| |
Collapse
|
9
|
Hamad I, Van Broeckhoven J, Cardilli A, Hellings N, Strowig T, Lemmens S, Hendrix S, Kleinewietfeld M. Effects of Recombinant IL-13 Treatment on Gut Microbiota Composition and Functional Recovery after Hemisection Spinal Cord Injury in Mice. Nutrients 2023; 15:4184. [PMID: 37836468 PMCID: PMC10574124 DOI: 10.3390/nu15194184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/06/2023] [Accepted: 09/11/2023] [Indexed: 10/15/2023] Open
Abstract
In recent years, the gut-central nervous system axis has emerged as a key factor in the pathophysiology of spinal cord injury (SCI). Interleukin-13 (IL-13) has been shown to have anti-inflammatory and neuroprotective effects in SCI. The aim of this study was to investigate the changes in microbiota composition after hemisection injury and to determine whether systemic recombinant (r)IL-13 treatment could alter the gut microbiome, indirectly promoting functional recovery. The gut microbiota composition was determined by 16S rRNA gene sequencing, and correlations between gut microbiota alterations and functional recovery were assessed. Our results showed that there were no changes in alpha diversity between the groups before and after SCI, while PERMANOVA analysis for beta diversity showed significant differences in fecal microbial communities. Phylogenetic classification of bacterial families revealed a lower abundance of the Bacteroidales S24-7 group and a higher abundance of Lachnospiraceae and Lactobacillaceae in the post-SCI group. Systemic rIL-13 treatment improved functional recovery 28 days post-injury and microbiota analysis revealed increased relative abundance of Clostridiales vadin BB60 and Acetitomaculum and decreased Anaeroplasma, Ruminiclostridium_6, and Ruminococcus compared to controls. Functional assessment with PICRUSt showed that genes related to glyoxylate cycle and palmitoleate biosynthesis-I were the predominant signatures in the rIL-13-treated group, whereas sulfolactate degradation super pathway and formaldehyde assimilation-I were enriched in controls. In conclusion, our results indicate that rIL-13 treatment promotes changes in gut microbial communities and may thereby contribute indirectly to the improvement of functional recovery in mice, possibly having important implications for the development of novel treatment options for SCI.
Collapse
Affiliation(s)
- Ibrahim Hamad
- VIB Laboratory of Translational Immunomodulation, Center for Inflammation Research (IRC), Hasselt University, 3590 Diepenbeek, Belgium (A.C.)
- Department of Immunology and Infection, Biomedical Research Institute (BIOMED), Hasselt University, 3590 Diepenbeek, Belgium; (J.V.B.); (N.H.)
| | - Jana Van Broeckhoven
- Department of Immunology and Infection, Biomedical Research Institute (BIOMED), Hasselt University, 3590 Diepenbeek, Belgium; (J.V.B.); (N.H.)
| | - Alessio Cardilli
- VIB Laboratory of Translational Immunomodulation, Center for Inflammation Research (IRC), Hasselt University, 3590 Diepenbeek, Belgium (A.C.)
- Department of Immunology and Infection, Biomedical Research Institute (BIOMED), Hasselt University, 3590 Diepenbeek, Belgium; (J.V.B.); (N.H.)
| | - Niels Hellings
- Department of Immunology and Infection, Biomedical Research Institute (BIOMED), Hasselt University, 3590 Diepenbeek, Belgium; (J.V.B.); (N.H.)
| | - Till Strowig
- Department of Microbial Immune Regulation, Helmholtz Center for Infection Research, 38124 Braunschweig, Germany
| | - Stefanie Lemmens
- Department of Immunology and Infection, Biomedical Research Institute (BIOMED), Hasselt University, 3590 Diepenbeek, Belgium; (J.V.B.); (N.H.)
| | - Sven Hendrix
- Institute for Translational Medicine, Medical School Hamburg, 20457 Hamburg, Germany
| | - Markus Kleinewietfeld
- VIB Laboratory of Translational Immunomodulation, Center for Inflammation Research (IRC), Hasselt University, 3590 Diepenbeek, Belgium (A.C.)
- Department of Immunology and Infection, Biomedical Research Institute (BIOMED), Hasselt University, 3590 Diepenbeek, Belgium; (J.V.B.); (N.H.)
| |
Collapse
|
10
|
Deng J, Meng F, Gao J, Zhang K, Liu Z, Li M, Liu X, Li J, Wang Y, Zhang L, Tang P. Early-phase rotator training impairs tissue repair and functional recovery after spinal cord injury. Heliyon 2023; 9:e18158. [PMID: 37519672 PMCID: PMC10372239 DOI: 10.1016/j.heliyon.2023.e18158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 05/27/2023] [Accepted: 07/10/2023] [Indexed: 08/01/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating disorder that often results in severe sensorimotor function impairment with limited recovery of function. In recent years, rehabilitation training for spinal cord injury has gradually emerged, and some of them play an important role in the repair of spinal cord injury However, the optimal training regimen for SCI remains to be determined. In this study, we explore the effects of rotarod training (began at 7 days post-injury) on the recovery of motor function after SCI, as well as its possible repair mechanism from the aspects of function and histopathological changes, the behaviors of specific trophic factors and cytokines, and the expression profile of specific genes. Multiple functional assessments showed that rotarod training initiated at 7 days post-injury is unsuitable for promoting neuro-electrophysiological improvement and trunk stability, but impaired functional coordination and motor recovery. In addition, rotarod training has negative effects on spinal cord repair after SCI, which is manifested as an increase of lesion area, a decrease in neuronal viability, a deterioration in immuno-microenvironment and remyelination, a significant reduction in the expression of trophic factors and an increase in the expression of pro-inflammatory factors. RNA sequencing suggested that the genes associated with angiogenesis and synaptogenesis were significantly downregulated and the PI3K-AKT pathway was inhibited, which was detrimental to spinal cord repair and impeded nerve regeneration. These results indicate that immediate rotarod training after SCI is currently unsuitable for rehabilitation in mice.
Collapse
Affiliation(s)
- Junhao Deng
- Department of Orthopedics, The Chinese PLA General Hospital, Beijing, 100853, China
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Fanqi Meng
- Department of Anesthesiology, Xuanwu Hospital Capital Medical University, Beijing, 100050, China
| | - Jianpeng Gao
- Department of Orthopedics, The Chinese PLA General Hospital, Beijing, 100853, China
| | - Kexue Zhang
- Department of Pediatric Surgery, The Chinese PLA General Hospital, Beijing, 100853, China
| | - Zhongyang Liu
- Department of Orthopedics, The Chinese PLA General Hospital, Beijing, 100853, China
| | - Ming Li
- Department of Orthopedics, The Chinese PLA General Hospital, Beijing, 100853, China
| | - Xiao Liu
- Department of Orthopedics, The Chinese PLA General Hospital, Beijing, 100853, China
| | - Jiantao Li
- Department of Orthopedics, The Chinese PLA General Hospital, Beijing, 100853, China
| | - Yu Wang
- Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Institue of Orthopaedics, The Chinese PLA General Hospital, Beijing, 100853, China
| | - Licheng Zhang
- Department of Orthopedics, The Chinese PLA General Hospital, Beijing, 100853, China
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| | - Peifu Tang
- Department of Orthopedics, The Chinese PLA General Hospital, Beijing, 100853, China
- School of Life Sciences, Tsinghua University, Beijing, 100084, China
| |
Collapse
|
11
|
Vangansewinkel T, Lemmens S, Tiane A, Geurts N, Dooley D, Vanmierlo T, Pejler G, Hendrix S. Therapeutic administration of mouse mast cell protease 6 improves functional recovery after traumatic spinal cord injury in mice by promoting remyelination and reducing glial scar formation. FASEB J 2023; 37:e22939. [PMID: 37130013 DOI: 10.1096/fj.202201942rr] [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: 11/26/2022] [Revised: 04/06/2023] [Accepted: 04/14/2023] [Indexed: 05/03/2023]
Abstract
Traumatic spinal cord injury (SCI) most often leads to permanent paralysis due to the inability of axons to regenerate in the adult mammalian central nervous system (CNS). In the past, we have shown that mast cells (MCs) improve the functional outcome after SCI by suppressing scar tissue formation at the lesion site via mouse mast cell protease 6 (mMCP6). In this study, we investigated whether recombinant mMCP6 can be used therapeutically to improve the functional outcome after SCI. Therefore, we applied mMCP6 locally via an intrathecal catheter in the subacute phase after a spinal cord hemisection injury in mice. Our findings showed that hind limb motor function was significantly improved in mice that received recombinant mMCP6 compared with the vehicle-treated group. In contrast to our previous findings in mMCP6 knockout mice, the lesion size and expression levels of the scar components fibronectin, laminin, and axon-growth-inhibitory chondroitin sulfate proteoglycans were not affected by the treatment with recombinant mMCP6. Surprisingly, no difference in infiltration of CD4+ T cells and reactivity of Iba-1+ microglia/macrophages at the lesion site was observed between the mMCP6-treated mice and control mice. Additionally, local protein levels of the pro- and anti-inflammatory mediators IL-1β, IL-2, IL-4, IL-6, IL-10, TNF-α, IFNγ, and MCP-1 were comparable between the two treatment groups, indicating that locally applied mMCP6 did not affect inflammatory processes after injury. However, the increase in locomotor performance in mMCP6-treated mice was accompanied by reduced demyelination and astrogliosis in the perilesional area after SCI. Consistently, we found that TNF-α/IL-1β-astrocyte activation was decreased and that oligodendrocyte precursor cell (OPC) differentiation was increased after recombinant mMCP6 treatment in vitro. Mechanistically, this suggests effects of mMCP6 on reducing astrogliosis and improving (re)myelination in the spinal cord after injury. In conclusion, these data show for the first time that recombinant mMCP6 is therapeutically active in enhancing recovery after SCI.
Collapse
Affiliation(s)
- Tim Vangansewinkel
- Cardio and Organ Systems, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- VIB, Center for Brain & Disease Research, Laboratory of Neurobiology, Leuven, Belgium
| | - Stefanie Lemmens
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Assia Tiane
- Department of Neuroscience, Faculty of Medicine and Life Sciences, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- University MS Center (UMSC) Hasselt-Pelt, Hasselt, Belgium
| | - Nathalie Geurts
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Dearbhaile Dooley
- School of Medicine, Health Sciences Centre, University College Dublin, Belfield, Ireland
- UCD Conway Institute of Biomolecular & Biomedical Research University College Dublin, Belfield, Ireland
| | - Tim Vanmierlo
- Department of Neuroscience, Faculty of Medicine and Life Sciences, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
- Department Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- University MS Center (UMSC) Hasselt-Pelt, Hasselt, Belgium
| | - Gunnar Pejler
- Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Sven Hendrix
- Institute for Translational Medicine, Medical School Hamburg, Hamburg, Germany
| |
Collapse
|
12
|
Huang LY, Sun X, Pan HX, Wang L, He CQ, Wei Q. Cell transplantation therapies for spinal cord injury focusing on bone marrow mesenchymal stem cells: Advances and challenges. World J Stem Cells 2023; 15:385-399. [PMID: 37342219 PMCID: PMC10277963 DOI: 10.4252/wjsc.v15.i5.385] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/17/2023] [Accepted: 03/21/2023] [Indexed: 05/26/2023] Open
Abstract
Spinal cord injury (SCI) is a devastating condition with complex pathological mechanisms that lead to sensory, motor, and autonomic dysfunction below the site of injury. To date, no effective therapy is available for the treatment of SCI. Recently, bone marrow-derived mesenchymal stem cells (BMMSCs) have been considered to be the most promising source for cellular therapies following SCI. The objective of the present review is to summarize the most recent insights into the cellular and molecular mechanism using BMMSC therapy to treat SCI. In this work, we review the specific mechanism of BMMSCs in SCI repair mainly from the following aspects: Neuroprotection, axon sprouting and/or regeneration, myelin regeneration, inhibitory microenvironments, glial scar formation, immunomodulation, and angiogenesis. Additionally, we summarize the latest evidence on the application of BMMSCs in clinical trials and further discuss the challenges and future directions for stem cell therapy in SCI models.
Collapse
Affiliation(s)
- Li-Yi Huang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital/West China School of Medicine, Sichuan University, Chengdu 610044, Sichuan Province, China
| | - Xin Sun
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital/West China School of Medicine, Sichuan University, Chengdu 610044, Sichuan Province, China
| | - Hong-Xia Pan
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital/West China School of Medicine, Sichuan University, Chengdu 610044, Sichuan Province, China
| | - Lu Wang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital/West China School of Medicine, Sichuan University, Chengdu 610044, Sichuan Province, China
| | - Cheng-Qi He
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital/West China School of Medicine, Sichuan University, Chengdu 610044, Sichuan Province, China
| | - Quan Wei
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital/West China School of Medicine, Sichuan University, Chengdu 610044, Sichuan Province, China
| |
Collapse
|
13
|
Hu H, Chen X, Zhao K, Zheng W, Gao C. Recent Advances in Biomaterials-Based Therapies for Alleviation and Regeneration of Traumatic Brain Injury. Macromol Biosci 2023; 23:e2200577. [PMID: 36758541 DOI: 10.1002/mabi.202200577] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/27/2023] [Indexed: 02/11/2023]
Abstract
Traumatic brain injury (TBI), a major public health problem accompanied with numerous complications, usually leads to serve disability and huge financial burden. The adverse and unfavorable pathological environment triggers a series of secondary injuries, resulting in serious loss of nerve function and huge obstacle of endogenous nerve regeneration. With the advances in adaptive tissue regeneration biomaterials, regulation of detrimental microenvironment to reduce the secondary injury and to promote the neurogenesis becomes possible. The adaptive biomaterials could respond and regulate biochemical, cellular, and physiological events in the secondary injury, including excitotoxicity, oxidative stress, and neuroinflammation, to rebuild circumstances suitable for regeneration. In this review, the development of pathology after TBI is discussed, followed by the introduction of adaptive biomaterials based on various pathological characteristics. The adaptive biomaterials carried with neurotrophic factors and stem cells for TBI treatment are then summarized. Finally, the current drawbacks and future perspective of biomaterials for TBI treatment are suggested.
Collapse
Affiliation(s)
- Haijun Hu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Xiping Chen
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Kefei Zhao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Weiwei Zheng
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310027, China.,Center for Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing, 312099, China
| |
Collapse
|
14
|
Walsh CM, Gull K, Dooley D. Motor rehabilitation as a therapeutic tool for spinal cord injury: New perspectives in immunomodulation. Cytokine Growth Factor Rev 2023; 69:80-89. [PMID: 36114092 DOI: 10.1016/j.cytogfr.2022.08.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 08/27/2022] [Indexed: 02/07/2023]
Abstract
Traumatic spinal cord injury (SCI) is a devastating condition that significantly impacts motor, sensory and autonomic function in patients. Despite advances in therapeutic approaches, there is still no curative therapy currently available. Neuroinflammation is a persisting event of the secondary injury phase of SCI that affects functional recovery, and modulation of the inflammatory response towards a beneficial anti-inflammatory state can improve recovery in preclinical SCI models. In human SCI patients, rehabilitative exercise, or motor rehabilitation as we will refer to it from here on out, remains the cornerstone of treatment to increase functional capacity and prevent secondary health implications. Motor rehabilitation is known to have anti-inflammatory effects; however, current literature is lacking in the description of the effect of motor rehabilitation on inflammation in the context of SCI. Understanding the effect on different inflammatory markers after SCI should enable the optimization of motor rehabilitation as a therapeutic regime. This review extensively describes the effect of motor rehabilitation on selected inflammatory mediators in both preclinical and human SCI studies. Additionally, we summarize how the type, duration, and intensity of motor rehabilitation can affect the inflammatory response after SCI. In doing so, we introduce a new perspective on how motor rehabilitation can be optimized as an immunomodulatory therapy to improve patient outcome after SCI.
Collapse
Affiliation(s)
- Ciara M Walsh
- School of Medicine, Health Sciences Centre, University College Dublin, Belfield, Dublin 4, Ireland; UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Khadija Gull
- School of Medicine, Health Sciences Centre, University College Dublin, Belfield, Dublin 4, Ireland
| | - Dearbhaile Dooley
- School of Medicine, Health Sciences Centre, University College Dublin, Belfield, Dublin 4, Ireland; UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
15
|
Huang T, Wu J, Mu J, Gao J. Advanced Therapies for Traumatic Central Nervous System Injury: Delivery Strategy Reinforced Efficient Microglial Manipulation. Mol Pharm 2023; 20:41-56. [PMID: 36469398 DOI: 10.1021/acs.molpharmaceut.2c00605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Traumatic central nervous system (CNS) injuries, including spinal cord injury and traumatic brain injury, are challenging enemies of human health. Microglia, the main component of the innate immune system in CNS, can be activated postinjury and are key participants in the pathological procedure and development of CNS trauma. Activated microglia can be typically classified into pro-inflammatory (M1) and anti-inflammatory (M2) phenotypes. Reducing M1 polarization while promoting M2 polarization is thought to be promising for CNS injury treatment. However, obstacles such as the low permeability of the blood-brain barrier and short retention time in circulation limit the therapeutic outcomes of administrated drugs, and rational delivery strategies are necessary for efficient microglial regulation. To this end, proper administration methods and delivery systems like nano/microcarriers and scaffolds are investigated to augment the therapeutic effects of drugs, while some of these delivery systems have self-efficacies in microglial manipulation. Besides, systems based on cell and cell-derived exosomes also show impressive effects, and some underlying targeting mechanisms of these delivery systems have been discovered. In this review, we introduce the roles of microglia play in traumatic CNS injuries, discuss the potential targets for the polarization regulation of microglial phenotype, and summarize recent studies and clinical trials about delivery strategies on enhancing the effect of microglial regulation and therapeutic outcome, as well as targeting mechanisms post CNS trauma.
Collapse
Affiliation(s)
- Tianchen Huang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiahe Wu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer, Pharmacology and Toxicology Research of Zhejiang Province, Affiliated, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Jiafu Mu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Jinhua Institute of Zhejiang University, Jinhua 321002, China
| |
Collapse
|
16
|
Li M, Chen H, Zhu M. Mesenchymal stem cells for regenerative medicine in central nervous system. Front Neurosci 2022; 16:1068114. [PMID: 36583105 PMCID: PMC9793714 DOI: 10.3389/fnins.2022.1068114] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
Mesenchymal stem cells (MSCs) are multipotent stem cells, whose paracrine and immunomodulatory potential has made them a promising candidate for central nervous system (CNS) regeneration. Numerous studies have demonstrated that MSCs can promote immunomodulation, anti-apoptosis, and axon re-extension, which restore functional neural circuits. The therapeutic effects of MSCs have consequently been evaluated for application in various CNS diseases including spinal cord injury, cerebral ischemia, and neurodegenerative disease. In this review, we will focus on the research works published in the field of mechanisms and therapeutic effects of MSCs in CNS regeneration.
Collapse
Affiliation(s)
- Man Li
- Department of Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hong Chen
- Department of Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mingxin Zhu
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China,*Correspondence: Mingxin Zhu,
| |
Collapse
|
17
|
Lima R, Monteiro A, Salgado AJ, Monteiro S, Silva NA. Pathophysiology and Therapeutic Approaches for Spinal Cord Injury. Int J Mol Sci 2022; 23:ijms232213833. [PMID: 36430308 PMCID: PMC9698625 DOI: 10.3390/ijms232213833] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 11/07/2022] [Indexed: 11/12/2022] Open
Abstract
Spinal cord injury (SCI) is a disabling condition that disrupts motor, sensory, and autonomic functions. Despite extensive research in the last decades, SCI continues to be a global health priority affecting thousands of individuals every year. The lack of effective therapeutic strategies for patients with SCI reflects its complex pathophysiology that leads to the point of no return in its function repair and regeneration capacity. Recently, however, several studies started to uncover the intricate network of mechanisms involved in SCI leading to the development of new therapeutic approaches. In this work, we present a detailed description of the physiology and anatomy of the spinal cord and the pathophysiology of SCI. Additionally, we provide an overview of different molecular strategies that demonstrate promising potential in the modulation of the secondary injury events that promote neuroprotection or neuroregeneration. We also briefly discuss other emerging therapies, including cell-based therapies, biomaterials, and epidural electric stimulation. A successful therapy might target different pathologic events to control the progression of secondary damage of SCI and promote regeneration leading to functional recovery.
Collapse
Affiliation(s)
- Rui Lima
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s Associate Laboratory, PT Government Associated Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - Andreia Monteiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s Associate Laboratory, PT Government Associated Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - António J. Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s Associate Laboratory, PT Government Associated Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - Susana Monteiro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s Associate Laboratory, PT Government Associated Laboratory, 4806-909 Braga/Guimarães, Portugal
| | - Nuno A. Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
- ICVS/3B’s Associate Laboratory, PT Government Associated Laboratory, 4806-909 Braga/Guimarães, Portugal
- Correspondence:
| |
Collapse
|
18
|
Pang Y, Liu X, Wang X, Shi X, Ma L, Zhang Y, Zhou T, Zhao C, Zhang X, Fan B, Hao J, Li W, Zhao X, Zhang R, Zhou S, Kong X, Feng S, Yao X. Edaravone Modulates Neuronal GPX4/ACSL4/5-LOX to Promote Recovery After Spinal Cord Injury. Front Cell Dev Biol 2022; 10:849854. [PMID: 35903552 PMCID: PMC9318422 DOI: 10.3389/fcell.2022.849854] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 03/24/2022] [Indexed: 01/20/2023] Open
Abstract
The FDA-approved drug edaravone has a neuroprotective effect on spinal cord injury (SCI) and many other central nervous system diseases. However, its molecular mechanism remains unclear. Since edaravone is a lipid peroxidation scavenger, we hypothesize that edaravone exerts its neuroprotective effect by inhibiting ferroptosis in SCI. Edaravone treatment after SCI upregulates glutathione peroxidase 4 (GPX4) and system Xc-light chain (xCT), which are anti-ferroptosis proteins. It downregulates pro-ferroptosis proteins Acyl-CoA synthetase long-chain family member 4 (ACSL4) and 5-lipoxygenase (5-LOX). The most significant changes in edaravone treatment occur in the acute phase, two days post injury. Edaravone modulates neuronal GPX4/ACSL4/5-LOX in the spinal segment below the lesion, which is critical for maintaining locomotion. Moreover, the GPX4/ACSL4/5-LOX in motor neuron is also modulated by edaravone in the spinal cord. Therefore, secondary injury below the lesion site is reversed by edaravone via ferroptosis inhibition. The cytokine array revealed that edaravone upregulated some anti-inflammatory cytokines such as IL-10, IL-13, and adiponectin. Edaravone reduced microgliosis and astrogliosis, indicating reduced neuroinflammation. Edaravone has a long-term effect on neuronal survival, spinal cord tissue sparing, and motor function recovery. In summary, we revealed a novel mechanism of edaravone in inhibiting neuronal ferroptosis in SCI. This mechanism may be generalizable to other neurological diseases.
Collapse
Affiliation(s)
- Yilin Pang
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, China
| | - Xinjie Liu
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, China
| | - Xu Wang
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, China
| | - Xuelian Shi
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Center for Cardiovascular Diseases, Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Lei Ma
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan Zhang
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, China
| | - Tiangang Zhou
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, China
| | - Chenxi Zhao
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, China
| | - Xu Zhang
- Tianjin Key Laboratory of Metabolic Diseases, Department of Physiology and Pathophysiology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, Center for Cardiovascular Diseases, Research Center of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Baoyou Fan
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, China
| | - Jian Hao
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenxiang Li
- Orthopedic Research Center of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaoqing Zhao
- Orthopedic Research Center of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Rong Zhang
- Orthopedic Research Center of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Songlin Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Nantong University, Jiangsu, China
| | - Xiaohong Kong
- Orthopedic Research Center of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shiqing Feng
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, China
- Orthopedic Research Center of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xue Yao
- Tianjin Key Laboratory of Spine and Spinal Cord, International Science and Technology Cooperation Base of Spinal Cord Injury, Department of Orthopedics, International Chinese Musculoskeletal Research Society Collaborating Center for Spinal Cord Injury, Tianjin Medical University General Hospital, Tianjin, China
- Orthopedic Research Center of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, China
- *Correspondence: Xue Yao,
| |
Collapse
|
19
|
Van Broeckhoven J, Erens C, Sommer D, Scheijen E, Sanchez S, Vidal PM, Dooley D, Van Breedam E, Quarta A, Ponsaerts P, Hendrix S, Lemmens S. Macrophage-based delivery of interleukin-13 improves functional and histopathological outcomes following spinal cord injury. J Neuroinflammation 2022; 19:102. [PMID: 35488301 PMCID: PMC9052547 DOI: 10.1186/s12974-022-02458-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 04/07/2022] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND Spinal cord injury (SCI) elicits a robust neuroinflammatory reaction which, in turn, exacerbates the initial mechanical damage. Pivotal players orchestrating this response are macrophages (Mφs) and microglia. After SCI, the inflammatory environment is dominated by pro-inflammatory Mφs/microglia, which contribute to secondary cell death and prevent regeneration. Therefore, reprogramming Mφ/microglia towards a more anti-inflammatory and potentially neuroprotective phenotype has gained substantial therapeutic interest in recent years. Interleukin-13 (IL-13) is a potent inducer of such an anti-inflammatory phenotype. In this study, we used genetically modified Mφs as carriers to continuously secrete IL-13 (IL-13 Mφs) at the lesion site. METHODS Mφs were genetically modified to secrete IL-13 (IL-13 Mφs) and were phenotypically characterized using qPCR, western blot, and ELISA. To analyze the therapeutic potential, the IL-13 Mφs were intraspinally injected at the perilesional area after hemisection SCI in female mice. Functional recovery and histopathological improvements were evaluated using the Basso Mouse Scale score and immunohistochemistry. Neuroprotective effects of IL-13 were investigated using different cell viability assays in murine and human neuroblastoma cell lines, human neurospheroids, as well as murine organotypic brain slice cultures. RESULTS In contrast to Mφs prestimulated with recombinant IL-13, perilesional transplantation of IL-13 Mφs promoted functional recovery following SCI in mice. This improvement was accompanied by reduced lesion size and demyelinated area. The local anti-inflammatory shift induced by IL-13 Mφs resulted in reduced neuronal death and fewer contacts between dystrophic axons and Mφs/microglia, suggesting suppression of axonal dieback. Using IL-4Rα-deficient mice, we show that IL-13 signaling is required for these beneficial effects. Whereas direct neuroprotective effects of IL-13 on murine and human neuroblastoma cell lines or human neurospheroid cultures were absent, IL-13 rescued murine organotypic brain slices from cell death, probably by indirectly modulating the Mφ/microglia responses. CONCLUSIONS Collectively, our data suggest that the IL-13-induced anti-inflammatory Mφ/microglia phenotype can preserve neuronal tissue and ameliorate axonal dieback, thereby promoting recovery after SCI.
Collapse
Affiliation(s)
- Jana Van Broeckhoven
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, 3590, Diepenbeek, Belgium
| | - Céline Erens
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, 3590, Diepenbeek, Belgium
| | - Daniela Sommer
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, 3590, Diepenbeek, Belgium
| | - Elle Scheijen
- Department of Neurosciences, Biomedical Research Institute, Hasselt University, 3590, Diepenbeek, Belgium
| | - Selien Sanchez
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, 3590, Diepenbeek, Belgium
| | - Pia M Vidal
- Neuroimmunology and Regeneration of the Central Nervous System Unit, Biomedical Science Research Laboratory, Basic Sciences Department, Faculty of Medicine, Universidad Católica de la Santísima Concepción, 4090541, Concepción, Chile
| | - Dearbhaile Dooley
- School of Medicine, Health Sciences Centre, University College Dublin, Belfield, Dublin 4, Ireland.,UCD Conway Institute of Biomolecular & Biomedical Research University College Dublin, Belfield, Dublin 4, Ireland
| | - Elise Van Breedam
- Laboratory of Experimental Hematology, University of Antwerp, 2610, Wilrijk, Belgium.,Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, 2610, Wilrijk, Belgium
| | - Alessandra Quarta
- Laboratory of Experimental Hematology, University of Antwerp, 2610, Wilrijk, Belgium.,Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, 2610, Wilrijk, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, University of Antwerp, 2610, Wilrijk, Belgium.,Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, 2610, Wilrijk, Belgium
| | - Sven Hendrix
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, 3590, Diepenbeek, Belgium. .,Medical School Hamburg, Am Kaiserkai 1, 20457, Hamburg, Germany.
| | - Stefanie Lemmens
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, 3590, Diepenbeek, Belgium
| |
Collapse
|
20
|
Xu X, Liang Z, Lin Y, Rao J, Lin F, Yang Z, Wang R, Chen C. Comparing the Efficacy and Safety of Cell Transplantation for Spinal Cord Injury: A Systematic Review and Bayesian Network Meta-Analysis. Front Cell Neurosci 2022; 16:860131. [PMID: 35444516 PMCID: PMC9013778 DOI: 10.3389/fncel.2022.860131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Accepted: 03/11/2022] [Indexed: 11/13/2022] Open
Abstract
ObjectiveTo compare the safety and effectiveness of transplanted cells from different sources for spinal cord injury (SCI).DesignA systematic review and Bayesian network meta-analysis.Data SourcesMedline, Embase, and the Cochrane Central Register of Controlled Trials.Study SelectionWe included randomized controlled trials, case–control studies, and case series related to cell transplantation for SCI patients, that included at least 1 of the following outcome measures: American Spinal Cord Injury Association (ASIA) Impairment Scale (AIS grade), ASIA motor score, ASIA sensory score, the Functional Independence Measure score (FIM), International Association of Neurorestoratology Spinal Cord Injury Functional Rating Scale (IANR-SCIFRS), or adverse events. Follow-up data were analyzed at 6 and 12 months.ResultsForty-four eligible trials, involving 1,266 patients, investigated 6 treatments: olfactory ensheathing cells (OECs), neural stem cells/ neural progenitor cells (NSCs), mesenchymal stem cells (MSCs), Schwann cells, macrophages, and combinations of cells (MSCs plus Schwann cells). Macrophages improved the AIS grade at 12 months (mean 0.42, 95% credible interval: 0–0.91, low certainty) and FIM score at 12 months (42.83, 36.33–49.18, very low certainty). MSCs improved the AIS grade at 6 months (0.42, 0.15–0.73, moderate certainty), the motor score at 6 months (4.43, 0.91–7.78, moderate certainty), light touch at 6 (10.01, 5.81–13.88, moderate certainty) and 12 months (11.48, 6.31–16.64, moderate certainty), pinprick score at 6 (14.54, 9.76–19.46, moderate certainty) and 12 months (12.48, 7.09–18.12, moderate certainty), and the IANR-SCIFRS at 6 (3.96, 0.62–6.97, moderate certainty) and 12 months (5.54, 2.45–8.42, moderate certainty). OECs improved the FIM score at 6 months (9.35, 1.71–17.00, moderate certainty). No intervention improved the motor score significantly at 12 months. The certainty of other interventions was low or very low. Overall, the number of adverse events associated with transplanted cells was low.ConclusionsPatients with SCI who receive transplantation of macrophages, MSCs, NSCs, or OECs may have improved disease prognosis. MSCs are the primary recommendations. Further exploration of the mechanism of cell transplantation in the treatment of SCI, transplantation time window, transplantation methods, and monitoring of the number of transplanted cells and cell survival is needed.Systematic Review Registrationhttps://www.crd.york.ac.uk/PROSPERO/#recordDetails, identifier: CRD 42021282043.
Collapse
|
21
|
Vankriekelsvenne E, Chrzanowski U, Manzhula K, Greiner T, Wree A, Hawlitschka A, Llovera G, Zhan J, Joost S, Schmitz C, Ponsaerts P, Amor S, Nutma E, Kipp M, Kaddatz H. Transmembrane protein 119 is neither a specific nor a reliable marker for microglia. Glia 2022; 70:1170-1190. [PMID: 35246882 DOI: 10.1002/glia.24164] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/14/2022] [Accepted: 02/17/2022] [Indexed: 12/12/2022]
Abstract
Microglia are the resident innate immune cells of the central nervous system (CNS) parenchyma. To determine the impact of microglia on disease development and progression in neurodegenerative and neuroinflammatory diseases, it is essential to distinguish microglia from peripheral macrophages/monocytes, which are eventually equally recruited. It has been suggested that transmembrane protein 119 (TMEM119) serves as a reliable microglia marker that discriminates resident microglia from blood-derived macrophages in the human and murine brain. Here, we investigated the validity of TMEM119 as a microglia marker in four in vivo models (cuprizone intoxication, experimental autoimmune encephalomyelitis (EAE), permanent filament middle cerebral artery occlusion (fMCAo), and intracerebral 6-hydroxydopamine (6-OHDA) injections) as well as post mortem multiple sclerosis (MS) brain tissues. In all applied animal models and post mortem MS tissues, we found increased densities of ionized calcium-binding adapter molecule 1+ (IBA1+ ) cells, paralleled by a significant decrease in TMEM119 expression. In addition, other cell types in peripheral tissues (i.e., follicular dendritic cells and brown adipose tissue) were also found to express TMEM119. In summary, this study demonstrates that TMEM119 is not exclusively expressed by microglia nor does it label all microglia, especially under cellular stress conditions. Since novel transgenic lines have been developed to label microglia using the TMEM119 promotor, downregulation of TMEM119 expression might interfere with the results and should, thus, be considered when working with these transgenic mouse models.
Collapse
Affiliation(s)
| | - Uta Chrzanowski
- Rostock University Medical Center, Institute of Anatomy, Rostock, Germany.,Faculty of Medicine, LMU Munich, Institute of Anatomy II, Munich, Germany
| | - Katerina Manzhula
- Rostock University Medical Center, Institute of Anatomy, Rostock, Germany
| | - Theresa Greiner
- Rostock University Medical Center, Institute of Anatomy, Rostock, Germany
| | - Andreas Wree
- Rostock University Medical Center, Institute of Anatomy, Rostock, Germany
| | | | - Gemma Llovera
- Institute for Stroke and Dementia Research, Klinikum der Universität München, Munich, Germany
| | - Jiangshan Zhan
- Rostock University Medical Center, Institute of Anatomy, Rostock, Germany
| | - Sarah Joost
- Rostock University Medical Center, Institute of Anatomy, Rostock, Germany
| | - Christoph Schmitz
- Faculty of Medicine, LMU Munich, Institute of Anatomy II, Munich, Germany
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| | - Sandra Amor
- Department of Pathology, Amsterdam UMC, VUMC Site, Amsterdam, The Netherlands.,Barts and The London School of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, UK
| | - Erik Nutma
- Department of Pathology, Amsterdam UMC, VUMC Site, Amsterdam, The Netherlands
| | - Markus Kipp
- Rostock University Medical Center, Institute of Anatomy, Rostock, Germany
| | - Hannes Kaddatz
- Rostock University Medical Center, Institute of Anatomy, Rostock, Germany
| |
Collapse
|
22
|
Pang QM, Chen SY, Fu SP, Zhou H, Zhang Q, Ao J, Luo XP, Zhang T. Regulatory Role of Mesenchymal Stem Cells on Secondary Inflammation in Spinal Cord Injury. J Inflamm Res 2022; 15:573-593. [PMID: 35115806 PMCID: PMC8802142 DOI: 10.2147/jir.s349572] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/24/2021] [Indexed: 12/13/2022] Open
Affiliation(s)
- Qi-Ming Pang
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
| | - Si-Yu Chen
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
| | - Sheng-Ping Fu
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
| | - Hui Zhou
- The First School of Clinical Medicine, Zunyi Medical University, Zunyi, People’s Republic of China
| | - Qian Zhang
- Department of Human Anatomy, Zunyi Medical University, Zunyi, People’s Republic of China
| | - Jun Ao
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
| | - Xiao-Ping Luo
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
| | - Tao Zhang
- Key Laboratory of Cell Engineering of Guizhou Province and Regenerative Medicine Centre, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
- Department of Orthopedics, Affiliated Hospital of Zunyi Medical University, Zunyi, People’s Republic of China
- Correspondence: Tao Zhang; Qian Zhang, Email ;
| |
Collapse
|
23
|
Deng J, Li M, Meng F, Liu Z, Wang S, Zhang Y, Li M, Li Z, Zhang L, Tang P. 3D spheroids of human placenta-derived mesenchymal stem cells attenuate spinal cord injury in mice. Cell Death Dis 2021; 12:1096. [PMID: 34803160 PMCID: PMC8606575 DOI: 10.1038/s41419-021-04398-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 11/01/2021] [Accepted: 11/10/2021] [Indexed: 12/23/2022]
Abstract
Mesenchymal stem cell (MSC) is an absorbing candidate for cell therapy in treating spinal cord injury (SCI) due to its great potential for multiple cell differentiation, mighty paracrine secretion as well as vigorous immunomodulatory effect, of which are beneficial to the improvement of functional recovery post SCI. However, the therapeutic effects of MSC on SCI have been limited because of the gradual loss of MSC stemness in the process of expanding culture. Therefore, in this study, we aimed to maintain those beneficial properties of MSC via three-dimensional spheroid cell culture and then compared them with conventionally-cultured MSCs in the treatment of SCI both in vitro and in vivo with the aid of two-photon microscope. We found that 3D human placenta-derived MSCs (3D-HPMSCs) demonstrated a significant increase in secretion of anti-inflammatory factors and trophic factors like VEGF, PDGF, FGF via QPCR and Bio-Plex assays, and showed great potentials on angiogenesis and neurite morphogenesis when co-cultured with HUVECs or DRGs in vitro. After transplantation into the injured spinal cord, 3D-HPMSCs managed to survive for the entire experiment and retained their advantageous properties in secretion, and exhibited remarkable effects on neuroprotection by minimizing the lesion cavity, inhibiting the inflammation and astrogliosis, and promoting angiogenesis. Further investigation of axonal dieback via two-photon microscope indicated that 3D-HPMSCs could effectively alleviate axonal dieback post injury. Further, mice only treated with 3D-HPMSCs obtained substantial improvement of functional recovery on electrophysiology, BMS score, and Catwalk analysis. RNA sequencing suggested that the 3D-HPMSCs structure organization-related gene was significantly changed, which was likely to potentiate the angiogenesis and inflammation regulation after SCI. These results suggest that 3D-HPMSCs may hold great potential for the treatment of SCI.
Collapse
Affiliation(s)
- Junhao Deng
- Medical School of Chinese PLA, Beijing, 100853, China
- Department of Orthopedics, The Chinese PLA General Hospital, Beijing, 100853, China
| | - Miao Li
- Key Laboratory of Chemical Genomics, Shenzhen Graduate School, Peking University, Shenzhen, 518055, China
- Department of Anesthesiology, Columbia University, New York, NY, 10032, USA
| | - Fanqi Meng
- Department of Spine Surgery, Peking University People's hospital, Beijing, 100044, China
| | - Zhongyang Liu
- Department of Orthopedics, The Chinese PLA General Hospital, Beijing, 100853, China
| | - Song Wang
- Department of Orthopedics, The Chinese PLA General Hospital, Beijing, 100853, China
- Medical college, Nankai University, Tianjin, 300071, China
| | - Yuan Zhang
- IBM Research-China, Beijing, 100193, China
| | - Ming Li
- Department of Orthopedics, The Chinese PLA General Hospital, Beijing, 100853, China
| | - Zhirui Li
- Department of Orthopedics, The Chinese PLA General Hospital, Beijing, 100853, China.
| | - Licheng Zhang
- Department of Orthopedics, The Chinese PLA General Hospital, Beijing, 100853, China.
| | - Peifu Tang
- Medical School of Chinese PLA, Beijing, 100853, China.
- Department of Orthopedics, The Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
24
|
Walsh CM, Wychowaniec JK, Brougham DF, Dooley D. Functional hydrogels as therapeutic tools for spinal cord injury: New perspectives on immunopharmacological interventions. Pharmacol Ther 2021; 234:108043. [PMID: 34813862 DOI: 10.1016/j.pharmthera.2021.108043] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 02/06/2023]
Abstract
Spinal cord injury (SCI) is a complex medical and psychological challenge for which there is no curative therapy currently available. Despite major progress in pharmacological and surgical approaches, clinical trials for SCI patients have been uniformly disappointing thus far as there are many practical and biological issues yet to be resolved. Neuroinflammation is a critical event of the secondary injury phase after SCI, and recent research strategies have focused on modulating the immune response after injury to provide a more favorable recovery environment. Biomaterials can serve this purpose by providing physical and trophic support to the injured spinal cord after SCI. Of all potential biomaterials, functional hydrogels are emerging as a key component in novel treatment strategies for SCI, including controlled and localized delivery of immunomodulatory therapies to drive polarization of immune cells towards a pro-regenerative phenotype. Here, we extensively review recent developments in the use of functional hydrogels as immunomodulatory therapies for SCI. We briefly describe physicochemical properties of hydrogels and demonstrate how advanced fabrication methods lead to the required heterogeneity and hierarchical arrangements that increasingly mimic complex spinal cord tissue. We then summarize potential SCI therapeutic modalities including: (i) hydrogels alone; (ii) hydrogels as cellular or (iii) bioactive molecule delivery vehicles, and; (iv) combinatorial approaches. By linking the structural properties of hydrogels to their functions in treatment with particular focus on immunopharmacological stimuli, this may accelerate further development of functional hydrogels for SCI, and indeed next-generation central nervous system regenerative therapies.
Collapse
Affiliation(s)
- Ciara M Walsh
- School of Medicine, Health Sciences Centre, University College Dublin, Belfield, Dublin 4, Ireland; UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Jacek K Wychowaniec
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland; AO Research Institute Davos, Clavadelerstrasse 8, 7270 Davos, Switzerland
| | - Dermot F Brougham
- School of Chemistry, University College Dublin, Belfield, Dublin 4, Ireland
| | - Dearbhaile Dooley
- School of Medicine, Health Sciences Centre, University College Dublin, Belfield, Dublin 4, Ireland; UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
25
|
Stem Cell Secretome for Spinal Cord Repair: Is It More than Just a Random Baseline Set of Factors? Cells 2021; 10:cells10113214. [PMID: 34831436 PMCID: PMC8625005 DOI: 10.3390/cells10113214] [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: 07/31/2021] [Revised: 11/02/2021] [Accepted: 11/16/2021] [Indexed: 11/29/2022] Open
Abstract
Hundreds of thousands of people suffer spinal cord injuries each year. The experimental application of stem cells following spinal cord injury has opened a new era to promote neuroprotection and neuroregeneration of damaged tissue. Currently, there is great interest in the intravenous administration of the secretome produced by mesenchymal stem cells in acute or subacute spinal cord injuries. However, it is important to highlight that undifferentiated neural stem cells and induced pluripotent stem cells are able to adapt to the damaged environment and produce the so-called lesion-induced secretome. This review article focuses on current research related to the secretome and the lesion-induced secretome and their roles in modulating spinal cord injury symptoms and functional recovery, emphasizing different compositions of the lesion-induced secretome in various models of spinal cord injury.
Collapse
|
26
|
Zhang K, Li F, Yan B, Xiao DJ, Wang YS, Liu H. Comparison of the Cytokine Profile in Mesenchymal Stem Cells from Human Adipose, Umbilical Cord, and Placental Tissues. Cell Reprogram 2021; 23:336-348. [PMID: 34677101 DOI: 10.1089/cell.2021.0043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Human mesenchymal stem cells (MSCs) can be isolated from various tissues. However, the cytokine profile in different MSC types remains unclear. In this study, MSCs were extracted from adipose, umbilical cord, and placental tissues. The surface marker expression, multilineage differentiation potential, and cytokine secretion of these cells were compared. The isolated MSCs exhibited similar morphology and surface marker expression. However, they differed with regard to their differentiation potential. Adipose-MSCs (A-MSCs) exhibited a higher potential for adipogenesis and osteogenic differentiation compared with umbilical cord-MSCs (UC-MSCs) and placental-MSCs (P-MSCs). The expression levels of 80 cytokines were detected, and the data demonstrated that the three MSC types abundantly secreted insulin-like growth factor-binding protein (IGFBP)-4, IGFBP-3, tissue inhibitor of metalloproteinase (TIMP)-1, TIMP-2, IGFBP-6, monocyte chemoattractant protein-1, and granulocyte colony-stimulating factor. However, the expression levels of vascular endothelial growth factor, tumor necrosis factor alpha, interleukin (IL)-6 receptor, and IL-13 in A-MSCs were higher compared with those of UC-MSCs and P-MSCs. Moreover, the expression levels of intercellular adhesion molecule-1 and growth differentiation factor 15 were lower in A-MSCs. Kyoto Encyclopedia of Genes and Genomes analysis indicated that the "adipocytokine" and the "PI3K/Akt pathways" were enriched in A-MSCs. Taken together, the results demonstrated that MSCs from different sources exhibited differences in the secretion of specific factors. A-MSCs were associated with the expression of several proangiogenic factors and may be an improved source for angiogenesis and tissue regeneration.
Collapse
Affiliation(s)
- Kun Zhang
- Cell Therapy Center, Jinan Central Hospital, Jinan, P.R. China.,Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China.,Shandong Research Center of Transplantation and Tissue, Jinan, P.R. China
| | - Fang Li
- Cell Therapy Center, Jinan Central Hospital, Jinan, P.R. China.,Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China.,Shandong Research Center of Transplantation and Tissue, Jinan, P.R. China
| | - Bing Yan
- Department of Gastrointestinal Surgery, Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China
| | - Dong-Jie Xiao
- Cell Therapy Center, Jinan Central Hospital, Jinan, P.R. China.,Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China
| | - Yun-Shan Wang
- Cell Therapy Center, Jinan Central Hospital, Jinan, P.R. China.,Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China
| | - Hua Liu
- Cell Therapy Center, Jinan Central Hospital, Jinan, P.R. China.,Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, P.R. China.,Shandong Research Center of Transplantation and Tissue, Jinan, P.R. China
| |
Collapse
|
27
|
Van Broeckhoven J, Sommer D, Dooley D, Hendrix S, Franssen AJPM. Macrophage phagocytosis after spinal cord injury: when friends become foes. Brain 2021; 144:2933-2945. [PMID: 34244729 DOI: 10.1093/brain/awab250] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 06/12/2021] [Accepted: 06/20/2021] [Indexed: 11/14/2022] Open
Abstract
After spinal cord injury (SCI), macrophages can exert either beneficial or detrimental effects depending on their phenotype. Aside from their critical role in inflammatory responses, macrophages are also specialized in the recognition, engulfment, and degradation of pathogens, apoptotic cells, and tissue debris. They promote remyelination and axonal regeneration by removing inhibitory myelin components and cellular debris. However, excessive intracellular presence of lipids and dysregulated intracellular lipid homeostasis result in the formation of foamy macrophages. These develop a pro-inflammatory phenotype that may contribute to further neurological decline. Additionally, myelin-activated macrophages play a crucial role in axonal dieback and retraction. Here, we review the opposing functional consequences of phagocytosis by macrophages in SCI, including remyelination and regeneration versus demyelination, degeneration, and axonal dieback. Furthermore, we discuss how targeting the phagocytic ability of macrophages may have therapeutic potential for the treatment of SCI.
Collapse
Affiliation(s)
- Jana Van Broeckhoven
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Daniela Sommer
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Dearbhaile Dooley
- School of Medicine, Health Sciences Centre, University College Dublin, Belfield Dublin 4, Ireland.,UCD Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - Sven Hendrix
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium.,Medical School Hamburg, Hamburg, Germany
| | - Aimée J P M Franssen
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| |
Collapse
|
28
|
The Protein Kinase Inhibitor Midostaurin Improves Functional Neurological Recovery and Attenuates Inflammatory Changes Following Traumatic Cervical Spinal Cord Injury. Biomolecules 2021; 11:biom11070972. [PMID: 34356596 PMCID: PMC8301989 DOI: 10.3390/biom11070972] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/25/2021] [Accepted: 06/27/2021] [Indexed: 12/12/2022] Open
Abstract
Traumatic spinal cord injury (SCI) impairs neuronal function and introduces a complex cascade of secondary pathologies that limit recovery. Despite decades of preclinical and clinical research, there is a shortage of efficacious treatment options to modulate the secondary response to injury. Protein kinases are crucial signaling molecules that mediate the secondary SCI-induced cellular response and present promising therapeutic targets. The objective of this study was to examine the safety and efficacy of midostaurin—a clinically-approved multi-target protein kinase inhibitor—on cervical SCI pathogenesis. High-throughput analyses demonstrated that intraperitoneal midostaurin injection (25 mg/kg) in C6/7 injured Wistar rats altered the local inflammasome and downregulated adhesive and migratory genes at 24 h post-injury. Treated animals also exhibited enhanced recovery and restored coordination between forelimbs and hindlimbs after injury, indicating the synergistic impact of midostaurin and its dimethyl sulfoxide vehicle to improve functional recovery. Furthermore, histological analyses suggested improved tissue preservation and functionality in the treated animals during the chronic phase of injury. This study serves as a proof-of-concept experiment and demonstrates that systemic midostaurin administration is an effective strategy for mitigating cervical secondary SCI damage.
Collapse
|
29
|
An N, Yang J, Wang H, Sun S, Wu H, Li L, Li M. Mechanism of mesenchymal stem cells in spinal cord injury repair through macrophage polarization. Cell Biosci 2021; 11:41. [PMID: 33622388 PMCID: PMC7903655 DOI: 10.1186/s13578-021-00554-z] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 02/11/2021] [Indexed: 02/07/2023] Open
Abstract
Treatment and rehabilitation of spinal cord injury (SCI) is a major problem in clinical medicine. Modern medicine has achieved minimal progress in improving the functions of injured nerves in patients with SCI, mainly due to the complex pathophysiological changes that present after injury. Inflammatory reactions occurring after SCI are related to various functions of immune cells over time at different injury sites. Macrophages are important mediators of inflammatory reactions and are divided into two different subtypes (M1 and M2), which play important roles at different times after SCI. Mesenchymal stem cells (MSCs) are characterized by multi-differentiation and immunoregulatory potentials, and different treatments can have different effects on macrophage polarization. MSC transplantation has become a promising method for eliminating nerve injury caused by SCI and can help repair injured nerve tissues. Therapeutic effects are related to the induced formation of specific immune microenvironments, caused by influencing macrophage polarization, controlling the consequences of secondary injury after SCI, and assisting with function recovery. Herein, we review the mechanisms whereby MSCs affect macrophage-induced specific immune microenvironments, and discuss potential avenues of investigation for improving SCI treatment.
Collapse
Affiliation(s)
- Nan An
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, 126 Xinmin Street, Changchun, 130021, Jilin, People's Republic of China.,The Second Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Jiaxu Yang
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, 126 Xinmin Street, Changchun, 130021, Jilin, People's Republic of China.,The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Hequn Wang
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, 126 Xinmin Street, Changchun, 130021, Jilin, People's Republic of China.,The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Shengfeng Sun
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, 126 Xinmin Street, Changchun, 130021, Jilin, People's Republic of China.,The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Hao Wu
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, 126 Xinmin Street, Changchun, 130021, Jilin, People's Republic of China.,The First Hospital of Jilin University, Changchun, 130021, Jilin, China
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, 126 Xinmin Street, Changchun, 130021, Jilin, People's Republic of China.
| | - Meiying Li
- The Key Laboratory of Pathobiology, Ministry of Education, Jilin University, 126 Xinmin Street, Changchun, 130021, Jilin, People's Republic of China.
| |
Collapse
|
30
|
Abstract
Traumatic spinal cord injury (SCI) results in direct and indirect damage to neural tissues, which results in motor and sensory dysfunction, dystonia, and pathological reflex that ultimately lead to paraplegia or tetraplegia. A loss of cells, axon regeneration failure, and time-sensitive pathophysiology make tissue repair difficult. Despite various medical developments, there are currently no effective regenerative treatments. Stem cell therapy is a promising treatment for SCI due to its multiple targets and reactivity benefits. The present review focuses on SCI stem cell therapy, including bone marrow mesenchymal stem cells, umbilical mesenchymal stem cells, adipose-derived mesenchymal stem cells, neural stem cells, neural progenitor cells, embryonic stem cells, induced pluripotent stem cells, and extracellular vesicles. Each cell type targets certain features of SCI pathology and shows therapeutic effects via cell replacement, nutritional support, scaffolds, and immunomodulation mechanisms. However, many preclinical studies and a growing number of clinical trials found that single-cell treatments had only limited benefits for SCI. SCI damage is multifaceted, and there is a growing consensus that a combined treatment is needed.
Collapse
Affiliation(s)
- Liyi Huang
- Department of Rehabilitation Medicine Center, 34753West China Hospital/West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Chenying Fu
- State Key Laboratory of Biotherapy, 34753West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feng Xiong
- Department of Rehabilitation Medicine Center, 34753West China Hospital/West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Chengqi He
- Department of Rehabilitation Medicine Center, 34753West China Hospital/West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Sichuan University, Chengdu, Sichuan Province, PR China
| | - Quan Wei
- Department of Rehabilitation Medicine Center, 34753West China Hospital/West China School of Medicine, Sichuan University, Chengdu, Sichuan, PR China.,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Sichuan University, Chengdu, Sichuan Province, PR China
| |
Collapse
|
31
|
Van Dyck A, Bollaerts I, Beckers A, Vanhunsel S, Glorian N, van Houcke J, van Ham TJ, De Groef L, Andries L, Moons L. Müller glia-myeloid cell crosstalk accelerates optic nerve regeneration in the adult zebrafish. Glia 2021; 69:1444-1463. [PMID: 33502042 DOI: 10.1002/glia.23972] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 01/12/2021] [Accepted: 01/15/2021] [Indexed: 12/18/2022]
Abstract
Neurodegenerative disorders, characterized by progressive neuronal loss, eventually lead to functional impairment in the adult mammalian central nervous system (CNS). Importantly, these deteriorations are irreversible, due to the very limited regenerative potential of these CNS neurons. Stimulating and redirecting neuroinflammation was recently put forward as an important approach to induce axonal regeneration, but it remains elusive how inflammatory processes and CNS repair are intertwined. To gain more insight into these interactions, we investigated how immunomodulation affects the regenerative outcome after optic nerve crush (ONC) in the spontaneously regenerating zebrafish. First, inducing intraocular inflammation using zymosan resulted in an acute inflammatory response, characterized by an increased infiltration and proliferation of innate blood-borne immune cells, reactivation of Müller glia, and altered retinal cytokine expression. Strikingly, inflammatory stimulation also accelerated axonal regrowth after optic nerve injury. Second, we demonstrated that acute depletion of both microglia and macrophages in the retina, using pharmacological treatments with both the CSF1R inhibitor PLX3397 and clodronate liposomes, compromised optic nerve regeneration. Moreover, we observed that csf1ra/b double mutant fish, lacking microglia in both retina and brain, displayed accelerated RGC axonal regrowth after ONC, which was accompanied with unusual Müller glia proliferative gliosis. Altogether, our results highlight the importance of altered glial cell interactions in the axonal regeneration process after ONC in adult zebrafish. Unraveling the relative contribution of the different cell types, as well as the signaling pathways involved, may pinpoint new targets to stimulate repair in the vertebrate CNS.
Collapse
Affiliation(s)
- Annelies Van Dyck
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - Ilse Bollaerts
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - An Beckers
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - Sophie Vanhunsel
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - Nynke Glorian
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - Jessie van Houcke
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - Tjakko J van Ham
- Department of Clinical Genetics, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Lies De Groef
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven, Belgium.,Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| | - Lien Andries
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven, Belgium
| | - Lieve Moons
- Neural Circuit Development and Regeneration Research Group, Department of Biology, KU Leuven, Leuven, Belgium.,Leuven Brain Institute (LBI), KU Leuven, Leuven, Belgium
| |
Collapse
|
32
|
Quarta A, Berneman Z, Ponsaerts P. Functional consequences of a close encounter between microglia and brain-infiltrating monocytes during CNS pathology and repair. J Leukoc Biol 2020; 110:89-106. [PMID: 33155726 DOI: 10.1002/jlb.3ru0820-536r] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 12/12/2022] Open
Abstract
Neuroinflammation is recognized as an important factor contributing to the development and progression of several central nervous system (CNS) disorders. Upon CNS trauma or disease, parenchymal microglia highly proliferate and accumulate in and around the lesion site. In addition, blood-derived monocytes can infiltrate the inflamed CNS in response to cellular damage and/or a compromised blood-brain barrier. Both microglia and infiltrating monocytes are characterized by multiple functional states and can either display highly proinflammatory properties or promote resolution of inflammation and tissue regeneration. Despite sharing some basic immunologic functions, microglia and monocytes display many distinctive features, which ultimately define their contribution to neuropathology. Understanding how the innate immune system participates to brain disease is imperative to identify novel treatment options for CNS inflammatory disorders. In this context, existing and newly developed in vitro platforms for disease modeling are fundamental tools to investigate and modulate microglia and monocyte immune functions within a specific neuropathologic context. In this review, we first briefly summarize the current knowledge on microglia and monocyte ontogenesis, as well as their complex and interconnected contributions to the development of various CNS pathologies. Following the well-recognized concept that both microglia and monocytes can either exert neuroprotective functions or exacerbate tissue damage, we provide a comprehensive overview of cellular models currently available for in vitro study of neuroinflammatory responses. In this context, we highlight how simplified single-cell models may not always correctly recapitulate in vivo biology, hence future research should move toward novel models with higher and multicellular complexity.
Collapse
Affiliation(s)
- Alessandra Quarta
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Zwi Berneman
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
33
|
Azimzadeh M, Mahmoodi M, Kazemi M, Hakemi MG, Jafarinia M, Eslami A, Salehi H, Amirpour N. The immunoregulatory and neuroprotective effects of human adipose derived stem cells overexpressing IL-11 and IL-13 in the experimental autoimmune encephalomyelitis mice. Int Immunopharmacol 2020; 87:106808. [PMID: 32693359 DOI: 10.1016/j.intimp.2020.106808] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/29/2020] [Accepted: 07/11/2020] [Indexed: 02/05/2023]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelination disease in the central nervous system (CNS) characterized by incomplete endogenous remyelination in the chronic phase. A shift of the balance between pro and anti-inflammatory cytokines is one of the important markers in the pathogenesis of MS. This study aimed to evaluate the effects of human adipose derived stem cells (hADSCs) overexpressing interleukin 11 and interleukin 13 (IL-11, 13-hADSCs) on the experimental autoimmune encephalomyelitis (EAE), an animal model of MS.12 days after immunization of C57Bl/6 female mice with MOG35-55 and initial clinical symptoms appearance, the IL-11, 13-hADSCs were injected via the tail vein into the EAE mice. Then, the mice were sacrificed at 30 days post-immunization (DPI) and the spinal cords of experimental groups were extracted for histopathological and real-time RT-PCR studies.The results indicated that the clinical scores and mononuclear cells infiltration into the spinal cords of EAE mice were significantly reduced in mice treated with IL-11, 13-hADSCs. Likewise, the remyelination and oligodendrogenesis were significantly enhanced in the mentioned treatment group. Real-time results demonstrated that pro/anti-inflammatory cytokine genes expression was reversed in IL-11, 13-hADSCs treatment group in comparison to the untreated EAE group.Expression of IL-11 as a neurotrophic cytokine and IL-13 as an anti-inflammatory cytokine by hADSCs could increase the immunomodulatory and neuroprotective effects of hADSCs and be a powerful candidate in stem cell therapy for future treatment of MS.
Collapse
Affiliation(s)
- Maryam Azimzadeh
- Department of Anatomical Science, School Of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Merat Mahmoodi
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Kazemi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | | - Morteza Jafarinia
- Department of Immunology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Asma Eslami
- Department of Immunology, School of Medicine, Isfahan University of Medical Science, Isfahan, Iran
| | - Hossein Salehi
- Department of Anatomical Science, School Of Medicine, Isfahan University of Medical Science, Isfahan, Iran.
| | - Noushin Amirpour
- Department of Anatomical Science, School Of Medicine, Isfahan University of Medical Science, Isfahan, Iran.
| |
Collapse
|
34
|
Eslami A, Dehbashi M, Ashja-Arvan M, Salehi H, Azimzadeh M, Ganjalikhani-Hakemi M. Assessment of ability of human adipose derived stem cells for long term overexpression of IL-11 and IL-13 as therapeutic cytokines. Cytotechnology 2020; 72:773-784. [PMID: 32935166 PMCID: PMC7547926 DOI: 10.1007/s10616-020-00421-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 09/02/2020] [Indexed: 02/05/2023] Open
Abstract
Adipose-derived stem cells (ADSCs) are a type of mesenchymal stem cells with the therapeutic effects that make them one of the best sources for cell therapy. In this study, we aimed to assess the ability of human ADSCs for constant expression of IL-11 and IL-13, simultaneously. In this study, the characterized hADSCs were transduced with a lentiviral vector (PCDH-513B) containing IL-11 and IL-13 genes, and the ability of long-term expression of the transgenes was evaluated by ELISA technique on days 15, 45 and 75 after transduction. Our results indicated a high rate of transduction (more than 90%) in the isolated hADSCs. Our data showed the highest rate of expression on days 75 after transduction which was 242.67 pg/ml for IL-11 and 303.6 pg/ml for IL-13 compared with 35.2 pg/ml and 35.6 pg/ml in untreated cells, respectively (p = 0.001). Besides, MTT assay showed transduction of hADSCs with lentiviral viruses containing IL-11 and IL-13 had no adverse effect on hADSCs proliferation (p-value = 0.89). Finally, we successfully constructed a hADSC population stably overexpressing IL-11 as the neurotrophic cytokine and IL-13 as the anti-inflammatory cytokine and this transduced cells can be used for further studies in EAE mice model.
Collapse
Affiliation(s)
- Asma Eslami
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Moein Dehbashi
- Division of Genetics, Department of Cell and Molecular Biology, Faculty of Biological Sciences and Technologies, University of Isfahan, 81746-73441 Isfahan, Iran
| | - Mehnoosh Ashja-Arvan
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hossein Salehi
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Azimzadeh
- Department of Anatomical Sciences, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | | |
Collapse
|
35
|
Quarta A, Meese T, Pieters Z, Van Breedam E, Le Blon D, Van Broeckhoven J, Hendrix S, Goossens H, Hens N, Berneman Z, Van Nieuwerburgh F, Ponsaerts P. Murine induced pluripotent stem cell-derived neuroimmune cell culture models emphasize opposite immune-effector functions of interleukin 13-primed microglia and macrophages in terms of neuroimmune toxicity. Glia 2020; 69:326-345. [PMID: 32865285 DOI: 10.1002/glia.23899] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/28/2020] [Accepted: 07/30/2020] [Indexed: 12/19/2022]
Abstract
Cellular models of induced pluripotent stem cell (iPSC)-derived microglia and macrophages are an emerging toolbox to investigate neuroinflammation in vitro. We previously demonstrated that murine iPSC-microglia and iPSC-macrophages display phenotypical activation properties highly comparable to microglia and macrophages in vivo. Here we extended the characterization of iPSC-microglia and iPSC-macrophages with the analysis of their transcriptome profile. Next, these cellular models were employed to evaluate neuroimmune toxicity in vitro and to investigate the immune-modulatory properties of interleukin 13 (IL13), a cytokine known for its ability to protect against neuroinflammation-induced pathology by modulating microglia and macrophage activation. iPSC-microglia and iPSC-macrophages, in co-culture with astrocyte-committed neural stem cells (NSC), were (pre)treated with IL13 and stimulated with lipopolysaccharide (LPS) and interferon γ (IFNγ), to assess how IL13 modulates their inflammatory response. Additionally, the use of luciferase-expressing NSC (Luc-NSC) allowed real-time monitoring of immune-mediated neurotoxicity. Despite the known anti-inflammatory properties of IL13, iPSC-microglia primed with IL13 before LPS + IFNγ stimulation significantly increased NO secretion. This was associated with a marked reduction of the luminescence signal produced by Luc-NSC. Interestingly, we observed that IL13 signaling has a divergent functional outcome in microglia as compared to macrophages, as for the latter no major alterations in NO release and Luc-NSC viability were observed upon IL13 (pre)treatment. Finally, the striking IL13-induced upregulation of NO secretion by microglia under pro-inflammatory conditions was confirmed in vivo, where intracerebral delivery of IL13 increased inducible nitric oxide synthase mRNA expression. Concluding, we applied iPSC-derived neuroimmune cell culture models to identify distinct neuroimmune (toxicity) responses of microglia and macrophages to IL13-based immune modulation.
Collapse
Affiliation(s)
- Alessandra Quarta
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Tim Meese
- Laboratory of Pharmaceutical Biotechnology, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Zoë Pieters
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium.,Interuniversity Institute for Biostatistics and Statistical Bioinformatics (I-BioStat), Data Science Institute, Hasselt University, Hasselt, Belgium.,Centre for Health Economics Research and Modelling Infectious Diseases, University of Antwerp, Antwerp, Belgium
| | - Elise Van Breedam
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Debbie Le Blon
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Jana Van Broeckhoven
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Sven Hendrix
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Herman Goossens
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Niel Hens
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium.,Interuniversity Institute for Biostatistics and Statistical Bioinformatics (I-BioStat), Data Science Institute, Hasselt University, Hasselt, Belgium.,Centre for Health Economics Research and Modelling Infectious Diseases, University of Antwerp, Antwerp, Belgium
| | - Zwi Berneman
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Filip Van Nieuwerburgh
- Laboratory of Pharmaceutical Biotechnology, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium.,Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
36
|
Quarta A, Berneman Z, Ponsaerts P. Neuroprotective modulation of microglia effector functions following priming with interleukin 4 and 13: current limitations in understanding their mode-of-action. Brain Behav Immun 2020; 88:856-866. [PMID: 32224056 DOI: 10.1016/j.bbi.2020.03.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/23/2020] [Accepted: 03/24/2020] [Indexed: 12/12/2022] Open
Abstract
In recent years the long-standing theory of microglia's properties for dual polarization towards a pro- or anti-inflammatory phenotype has been deeply challenged. Furthermore, the elucidation of microglia ontogenesis exposed intrinsic differences between microglia and peripheral myeloid cells, thereby further underscoring the need to re-evaluate microglia-specific activation behavior, especially within an inflamed central nervous system (CNS) environment. This review critically summarizes recent literature on the in vitro and in vivo response of murine microglia to the immune-modulatory cytokines interleukin 4 (IL4) and interleukin 13 (IL13), i.e. those driving the so-called anti-inflammatory phenotype. Here we highlight several pivotal factors that may influence experimental outcome and/or interpretation of in vitro and in vivo studies evaluating microglia's phenotypical and functional properties upon IL4/IL13 treatment. Finally, the current therapeutic relevance of IL4/IL13-induced microglia activation in both acute and chronic CNS disorders is discussed.
Collapse
Affiliation(s)
- Alessandra Quarta
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Zwi Berneman
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
37
|
Huang L, You J, Yao Y, Xie M. Interleukin-13 Gene Modification Enhances Grafted Mesenchymal Stem Cells Survival After Subretinal Transplantation. Cell Mol Neurobiol 2020; 40:725-735. [PMID: 31792777 PMCID: PMC11448798 DOI: 10.1007/s10571-019-00768-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Accepted: 11/26/2019] [Indexed: 12/13/2022]
Abstract
Mesenchymal stem cells (MSCs) hold great potential for cell- and gene-based therapies for retinal degeneration. Limited survival is the main obstacle in achieving successful subretinal transplantation of MSCs. The present study sought to evaluate the effect of interleukin-13 (IL-13) gene modification on the phenotypic alteration of retinal microglia (RMG) and the survival of MSCs following subretinal grafting. In this study, LPS-activated RMG were cocultured with MSCs or IL-13-expressing MSCs (IL-13-MSCs) for 24 h, and activated phenotypes were detected in vitro. Western blotting was performed to quantify cytokine secretion by light-injured retinas following subretinal transplantation. The numbers of activated RMG and surviving grafted cells were analysed, and the integrity of the blood-retinal barrier (BRB) was examined in vivo. We found that, compared with normal MSCs, cocultured IL-13-MSCs suppressed the expression of pro-inflammatory factors and major histocompatibility complex II, promoted the expression of anti-inflammatory cytokines by activated RMG and simultaneously inhibited the proliferation of and phagocytosis by RMG. The subretinal transplantation of IL-13-MSCs increased the expression of neurotrophic factors, IL-13 and tight junction proteins in the host retina, decreased the number of phagocytic RMG and improved the survival of grafted cells. Furthermore, IL-13-MSCs alleviated BRB breakdown induced by subretinal injection. Our results demonstrate that IL-13-MSCs can polarize activated RMG to the neuroprotective M2 phenotype and enhance the survival of grafted MSCs against the damage stress induced by subretinal transplantation.
Collapse
Affiliation(s)
- Libin Huang
- Department of Ophthalmology, The First Affiliated Hospital of Fujian Medical University, No. 20 Chazhong Road, 350005, Fuzhou, China
| | - Junmei You
- Department of Ophthalmology, The First Affiliated Hospital of Fujian Medical University, No. 20 Chazhong Road, 350005, Fuzhou, China
| | - Yao Yao
- Department of Ophthalmology, The First Affiliated Hospital of Fujian Medical University, No. 20 Chazhong Road, 350005, Fuzhou, China
| | - Maosong Xie
- Department of Ophthalmology, The First Affiliated Hospital of Fujian Medical University, No. 20 Chazhong Road, 350005, Fuzhou, China.
| |
Collapse
|
38
|
HDAC8 Inhibition Reduces Lesional Iba-1+ Cell Infiltration after Spinal Cord Injury without Effects on Functional Recovery. Int J Mol Sci 2020; 21:ijms21124539. [PMID: 32630606 PMCID: PMC7352158 DOI: 10.3390/ijms21124539] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 06/10/2020] [Accepted: 06/21/2020] [Indexed: 12/30/2022] Open
Abstract
Pan-histone deacetylase (HDAC) inhibition with valproic acid (VPA) has beneficial effects after spinal cord injury (SCI), although with side effects. We focused on specific HDAC8 inhibition, because it is known to reduce anti-inflammatory mediators produced by macrophages (Mφ). We hypothesized that HDAC8 inhibition improves functional recovery after SCI by reducing pro-inflammatory classically activated Mφ. Specific HDAC8 inhibition with PCI-34051 reduced the numbers of perilesional Mφ as measured by histological analyses, but did not improve functional recovery (Basso Mouse Scale). We could not reproduce the published improvement of functional recovery described in contusion SCI models using VPA in our T-cut hemisection SCI model. The presence of spared fibers might be the underlying reason for the conflicting data in different SCI models.
Collapse
|
39
|
Enam SF, Kader SR, Bodkin N, Lyon JG, Calhoun M, Azrak C, Tiwari PM, Vanover D, Wang H, Santangelo PJ, Bellamkonda RV. Evaluation of M2-like macrophage enrichment after diffuse traumatic brain injury through transient interleukin-4 expression from engineered mesenchymal stromal cells. J Neuroinflammation 2020; 17:197. [PMID: 32563258 PMCID: PMC7306141 DOI: 10.1186/s12974-020-01860-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/29/2020] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Appropriately modulating inflammation after traumatic brain injury (TBI) may prevent disabilities for the millions of those inflicted annually. In TBI, cellular mediators of inflammation, including macrophages and microglia, possess a range of phenotypes relevant for an immunomodulatory therapeutic approach. It is thought that early phenotypic modulation of these cells will have a cascading healing effect. In fact, an anti-inflammatory, "M2-like" macrophage phenotype after TBI has been associated with neurogenesis, axonal regeneration, and improved white matter integrity (WMI). There already exist clinical trials seeking an M2-like bias through mesenchymal stem/stromal cells (MSCs). However, MSCs do not endogenously synthesize key signals that induce robust M2-like phenotypes such as interleukin-4 (IL-4). METHODS To enrich M2-like macrophages in a clinically relevant manner, we augmented MSCs with synthetic IL-4 mRNA to transiently express IL-4. These IL-4 expressing MSCs (IL-4 MSCs) were characterized for expression and functionality and then delivered in a modified mouse TBI model of closed head injury. Groups were assessed for functional deficits and MR imaging. Brain tissue was analyzed through flow cytometry, multi-plex ELISA, qPCR, histology, and RNA sequencing. RESULTS We observed that IL-4 MSCs indeed induce a robust M2-like macrophage phenotype and promote anti-inflammatory gene expression after TBI. However, here we demonstrate that acute enrichment of M2-like macrophages did not translate to improved functional or histological outcomes, or improvements in WMI on MR imaging. To further understand whether dysfunctional pathways underlie the lack of therapeutic effect, we report transcriptomic analysis of injured and treated brains. Through this, we discovered that inflammation persists despite acute enrichment of M2-like macrophages in the brain. CONCLUSION The results demonstrate that MSCs can be engineered to induce a stronger M2-like macrophage response in vivo. However, they also suggest that acute enrichment of only M2-like macrophages after diffuse TBI cannot orchestrate neurogenesis, axonal regeneration, or improve WMI. Here, we also discuss our modified TBI model and methods to assess severity, behavioral studies, and propose that IL-4 expressing MSCs may also have relevance in other cavitary diseases or in improving biomaterial integration into tissues.
Collapse
Affiliation(s)
- Syed Faaiz Enam
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Nicholas Bodkin
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Johnathan G Lyon
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Mark Calhoun
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Cesar Azrak
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Pooja Munnilal Tiwari
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Daryll Vanover
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Haichen Wang
- Department of Neurology, Duke University, Durham, NC, USA
| | - Philip J Santangelo
- Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | | |
Collapse
|
40
|
Ma Y, Deng M, Zhao XQ, Liu M. Alternatively Polarized Macrophages Regulate the Growth and Differentiation of Ependymal Stem Cells through the SIRT2 Pathway. Exp Neurobiol 2020; 29:150-163. [PMID: 32408405 PMCID: PMC7237271 DOI: 10.5607/en19078] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 02/06/2023] Open
Abstract
Ependymal stem cells (EpSCs) are dormant stem cells in the adult spinal cord that proliferate rapidly and migrate to the site of injury after spinal cord injury (SCI). Although they can differentiate into neurons under appropriate conditions in vitro, EpSCs mainly differentiate into astrocytes in vivo. Our previous study confirmed that alternatively polarized macrophages (M2) facilitate the differentiation of EpSCs towards neurons, but the detailed mechanism remains elusive. In the present study, we found that M2 conditioned medium could upregulate the expression of Sirtuin 2 (SIRT2) in EpSCs in vitro through the BDNF/TrkB-MEK/ERK signaling pathway. As an important deacetylase, SIRT2 deacetylated stable Ac-α-tubulin (Acetyl alpha Tubulin) in microtubules and thus promoted EpSC differentiation into neurons. The present study provides a theoretical basis and a new way to improve neural recovery, such as regulating the growth and differentiation of EpSCs by increasing the proportion of M2 cells in the local microenvironment or upregulating the expression of SIRT2 in EpSCs.
Collapse
Affiliation(s)
- Yonggang Ma
- Department of Orthopaedics, Renmin Hospital, Wuhan University, Wuhan 430060, China
| | - Ming Deng
- Department of Orthopaedics, Renmin Hospital, Wuhan University, Wuhan 430060, China
| | - Xiao-Qi Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| | - Min Liu
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan 430071, China
| |
Collapse
|
41
|
Miao W, Zhao Y, Huang Y, Chen D, Luo C, Su W, Gao Y. IL-13 Ameliorates Neuroinflammation and Promotes Functional Recovery after Traumatic Brain Injury. THE JOURNAL OF IMMUNOLOGY 2020; 204:1486-1498. [PMID: 32034062 DOI: 10.4049/jimmunol.1900909] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 01/05/2020] [Indexed: 12/16/2022]
Abstract
Microglia play essential roles in neuroinflammatory responses after traumatic brain injury (TBI). Our previous studies showed that phenotypes of microglia, as well as infiltrating macrophages, altered at different stages after CNS injury, which was correlated to functional outcomes. IL-13 is an anti-inflammatory cytokine that has been reported to protect against demyelination and spinal cord injury through immunomodulation. The effects of IL-13 in microglia/macrophage-mediated immune responses after TBI remain unknown. In this study, we showed that intranasal administration of IL-13 in male C57BL/6J mice accelerated functional recovery in the controlled cortical impact model of TBI. IL-13 treatment increased the time to fall off in the Rotarod test, reduced the number of foot faults in the foot fault test, and improved the score in the wire hang test up to 28 d after TBI. Consistent with functional improvement, IL-13 reduced neuronal tissue loss and preserved white matter integrity 6 d after TBI. Furthermore, IL-13 ameliorated the elevation of proinflammatory factors and reduced the number of proinflammatory microglia/macrophages 6 d after TBI. Additionally, IL-13 enhanced microglia/macrophage phagocytosis of damaged neurons in the peri-lesion areas. In vitro studies confirmed that IL-13 treatment inhibited the production of proinflammatory cytokines in rat primary microglia in response to LPS or dead neuron stimulation and increased the ability of microglia to engulf fluorophore-labeled latex beads or dead neurons. Collectively, we demonstrated that IL-13 treatment improved neurologic outcomes after TBI through adjusting microglia/macrophage phenotypes and inhibiting inflammatory responses. IL-13 may represent a potential immunotherapy to promote long-term recovery from TBI.
Collapse
Affiliation(s)
- Wanying Miao
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China.,Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China.,Institutes of Brain Science, Fudan University, Shanghai 200032, China; and
| | - Yongfang Zhao
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China.,Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China.,Institutes of Brain Science, Fudan University, Shanghai 200032, China; and
| | - Yichen Huang
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China.,Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China.,Institutes of Brain Science, Fudan University, Shanghai 200032, China; and
| | - Di Chen
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China.,Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China.,Institutes of Brain Science, Fudan University, Shanghai 200032, China; and
| | - Chen Luo
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China.,Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China.,Institutes of Brain Science, Fudan University, Shanghai 200032, China; and
| | - Wei Su
- Department of Neurosurgery, School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing 102218, China
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai 200032, China; .,Ministry of Education Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China.,Institutes of Brain Science, Fudan University, Shanghai 200032, China; and
| |
Collapse
|
42
|
Therajaran P, Hamilton JA, O'Brien TJ, Jones NC, Ali I. Microglial polarization in posttraumatic epilepsy: Potential mechanism and treatment opportunity. Epilepsia 2020; 61:203-215. [PMID: 31943156 DOI: 10.1111/epi.16424] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 12/16/2019] [Accepted: 12/17/2019] [Indexed: 12/17/2022]
Abstract
Owing to the complexity of the pathophysiological mechanisms driving epileptogenesis following traumatic brain injury (TBI), effective preventive treatment approaches are not yet available for posttraumatic epilepsy (PTE). Neuroinflammation appears to play a critical role in the pathogenesis of the acquired epilepsies, including PTE, but despite a large preclinical literature demonstrating the ability of anti-inflammatory treatments to suppress epileptogenesis and chronic seizures, no anti-inflammatory treatment approaches have been clinically proven to date. TBI triggers robust inflammatory cascades, suggesting that they may be relevant for the pathogenesis of PTE. A major cell type involved in such cascades is the microglial cells-brain-resident immune cells that become activated after brain injury. When activated, these cells can oscillate between different phenotypes, and such polarization states are associated with the release of various pro- and anti-inflammatory mediators that may influence brain repair processes, and also differentially contribute to the development of PTE. As the molecular mechanisms and key signaling molecules associated with microglial polarization in brain are discovered, strategies are now emerging that can modulate this polarization, promoting this as a potential therapeutic strategy for PTE. In this review, we discuss the relevant literature regarding the polarization of brain-resident immune cells following TBI and attempt to put into perspective a role in epilepsy pathogenesis. Finally, we explore potential strategies that could polarize microglia/macrophages toward a neuroprotective phenotype to mitigate PTE development.
Collapse
Affiliation(s)
- Peravina Therajaran
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia
| | - John A Hamilton
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia
| | - Terence J O'Brien
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia.,Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Neurology, Alfred Hospital, Melbourne, Victoria, Australia
| | - Nigel C Jones
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia.,Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Neurology, Alfred Hospital, Melbourne, Victoria, Australia
| | - Idrish Ali
- Department of Medicine (Royal Melbourne Hospital), Melbourne Brain Centre, University of Melbourne, Parkville, Victoria, Australia.,Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Neurology, Alfred Hospital, Melbourne, Victoria, Australia
| |
Collapse
|
43
|
Schirmer L, Hoornaert C, Le Blon D, Eigel D, Neto C, Gumbleton M, Welzel PB, Rosser AE, Werner C, Ponsaerts P, Newland B. Heparin-based, injectable microcarriers for controlled delivery of interleukin-13 to the brain. Biomater Sci 2020; 8:4997-5004. [DOI: 10.1039/d0bm01249a] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The anti-inflammatory cytokine IL-13 can be loaded and released from heparin-based cryogel biomaterials for sustained delivery to the brain.
Collapse
|
44
|
Liu AM, Chen BL, Yu LT, Liu T, Shi LL, Yu PP, Qu YB, So KF, Zhou LB. Human adipose tissue- and umbilical cord-derived stem cells: which is a better alternative to treat spinal cord injury? Neural Regen Res 2020; 15:2306-2317. [PMID: 32594054 PMCID: PMC7749492 DOI: 10.4103/1673-5374.284997] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Multiple types of stem cells have been proposed for the treatment of spinal cord injury, but their comparative information remains elusive. In this study, a rat model of T10 contusion spinal cord injury was established by the impactor method. Human umbilical cord-derived mesenchymal stem cells (UCMSCs) or human adipose tissue-derived mesenchymal stem cells (ADMSCs) (2.5 μL/injection site, 1 × 105 cells/μL) was injected on rostral and caudal of the injury segment on the ninth day after injury. Rats injected with mesenchymal stem cell culture medium were used as controls. Our results show that although transplanted UCMSCs and ADMSCs failed to differentiate into neurons or glial cells in vivo, both significantly improved motor and sensory function. After spinal cord injury, UCMSCs and ADMSCs similarly promoted spinal neuron survival and axonal regeneration, decreased glial scar and lesion cavity formation, and reduced numbers of active macrophages. Bio-Plex analysis of spinal samples showed a specific increase of interleukin-10 and decrease of tumor necrosis factor α in the ADMSC group, as well as a downregulation of macrophage inflammatory protein 3α in both UCMSC and ADMSC groups at 3 days after cell transplantation. Upregulation of interleukin-10 and interleukin-13 was observed in both UCMSC and ADMSC groups at 7 days after cell transplantation. Isobaric tagging for relative and absolute quantitation proteomics analyses showed that UCMSCs and ADMSCs induced changes of multiple genes related to axonal regeneration, neurotrophy, and cell apoptosis in common and specific manners. In conclusion, UCMSC and ADMSC transplants yielded quite similar contributions to motor and sensory recovery after spinal cord injury via anti-inflammation and improved axonal growth. However, there were some differences in cytokine and gene expression induced by these two types of transplanted cells. Animal experiments were approved by the Laboratory Animal Ethics Committee at Jinan University (approval No. 20180228026) on February 28, 2018, and the application of human stem cells was approved by the Medical Ethics Committee of Medical College of Jinan University of China (approval No. 2016041303) on April 13, 2016.
Collapse
Affiliation(s)
- Ai-Mei Liu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
| | - Bo-Li Chen
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
| | - Ling-Tai Yu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
| | - Tao Liu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
| | - Ling-Ling Shi
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
| | - Pan-Pan Yu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
| | - Yi-Bo Qu
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Kwok-Fai So
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, Guangdong Province; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Li-Bing Zhou
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University; Guangzhou Regenerative Medicine and Health Guangdong Laboratory, Guangzhou, Guangdong Province; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| |
Collapse
|
45
|
Short-term inhibition of fibrinolytic system restores locomotor function after spinal cord injury in mice. Sci Rep 2019; 9:16024. [PMID: 31690812 PMCID: PMC6831600 DOI: 10.1038/s41598-019-52621-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 10/21/2019] [Indexed: 02/06/2023] Open
Abstract
Spinal cord injury (SCI) is caused by an initial mechanical insult followed by a series of deleterious events that promote the progressive damage of affected tissues. Fibrinolysis, the process by which plasmin degrades cross-linked fibrin clots, has numerous functions in the central nervous system. However, the roles of the fibrinolytic system in SCI pathophysiology remain unknown. We investigated the roles of fibrinolysis in SCI, and explored therapeutic applications targeting fibrinolysis. Plasminogen-deficient (Plg-/-) mice exhibited significantly improved locomotor function in the early phase of SCI (the first 7 days post injury), with significant inhibition of bleeding and vascular permeability, but failed to demonstrate conclusive functional recovery. Consistent with these findings, the short-term administration of tranexamic acid (TXA) in wild-type mice over the first 3 days post injury significantly improved locomotor function after SCI, whereas prolonged TXA administration did not. Prolonged TXA administration resulted in significantly lower levels of matrix metalloproteinase activities in the spinal cord, suggesting that inhibition of the fibrinolytic system impaired tissue remodeling. Our results indicate that the fibrinolytic system has time-dependent biphasic actions following SCI. The temporally optimised modulation of fibrinolytic activity may thus be a novel therapeutic strategy to improve functional outcomes after SCI.
Collapse
|
46
|
Quarta A, Le Blon D, D'aes T, Pieters Z, Hamzei Taj S, Miró-Mur F, Luyckx E, Van Breedam E, Daans J, Goossens H, Dewilde S, Hens N, Pasque V, Planas AM, Hoehn M, Berneman Z, Ponsaerts P. Murine iPSC-derived microglia and macrophage cell culture models recapitulate distinct phenotypical and functional properties of classical and alternative neuro-immune polarisation. Brain Behav Immun 2019; 82:406-421. [PMID: 31525508 DOI: 10.1016/j.bbi.2019.09.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 09/05/2019] [Accepted: 09/12/2019] [Indexed: 12/24/2022] Open
Abstract
The establishment and validation of reliable induced pluripotent stem cell (iPSC)-derived in vitro models to study microglia and monocyte/macrophage immune function holds great potential for fundamental and translational neuro-immunology research. In this study, we first demonstrate that ramified CX3CR1+ iPSC-microglia (cultured within a neural environment) and round-shaped CX3CR1- iPSC-macrophages can easily be differentiated from newly established murine CX3CR1eGFP/+CCR2RFP/+ iPSC lines. Furthermore, we show that obtained murine iPSC-microglia and iPSC-macrophages are distinct cell populations, even though iPSC-macrophages may upregulate CX3CR1 expression when cultured within a neural environment. Next, we characterized the phenotypical and functional properties of murine iPSC-microglia and iPSC-macrophages following classical and alternative immune polarisation. While iPSC-macrophages could easily be triggered to adopt a classically-activated or alternatively-activated phenotype following, respectively, lipopolysaccharide + interferon γ or interleukin 13 (IL13) stimulation, iPSC-microglia and iPSC-macrophages cultured within a neural environment displayed a more moderate activation profile as characterised by the absence of MHCII expression upon classical immune polarisation and the absence of Ym1 expression upon alternative immune polarisation. Finally, extending our preceding in vivo studies, this striking phenotypical divergence was also observed for resident microglia and infiltrating monocytes within highly inflammatory cortical lesions in CX3CR1eGFP/+CCR2RFP/+ mice subjected to middle cerebral arterial occlusion (MCAO) stroke and following IL13-mediated therapeutic intervention thereon. In conclusion, our study demonstrates that the applied murine iPSC-microglia and iPSC-macrophage culture models are able to recapitulate in vivo microglia and monocyte/macrophage ontogeny and corresponding phenotypical/functional properties upon classical and alternative immune polarisation, and therefore represent a valuable in vitro platform to further study and modulate microglia and (infiltrating) monocyte immune responses under neuro-inflammatory conditions within a neural environment.
Collapse
Affiliation(s)
- Alessandra Quarta
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium; Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Debbie Le Blon
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium; Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Tine D'aes
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium; Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Zoë Pieters
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium; Interuniversity Institute for Biostatistics and Statistical Bioinformatics, Hasselt University, Belgium; Centre for Health Economics Research and Modelling Infectious Diseases, University of Antwerp, Belgium
| | - Somayyeh Hamzei Taj
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Francesc Miró-Mur
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Evi Luyckx
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium; Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium; Protein Chemistry, Proteomics and Epigenetic Signaling, University of Antwerp, Antwerp, Belgium
| | - Elise Van Breedam
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium; Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Jasmijn Daans
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium; Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Herman Goossens
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Sylvia Dewilde
- Protein Chemistry, Proteomics and Epigenetic Signaling, University of Antwerp, Antwerp, Belgium
| | - Niel Hens
- Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium; Interuniversity Institute for Biostatistics and Statistical Bioinformatics, Hasselt University, Belgium; Centre for Health Economics Research and Modelling Infectious Diseases, University of Antwerp, Belgium
| | - Vincent Pasque
- Department of Development and Regeneration, Leuven Stem Cell Institute, Leuven Cancer Institute, KU Leuven - University of Leuven, Belgium
| | - Anna M Planas
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB)-Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
| | - Mathias Hoehn
- In-vivo-NMR Laboratory, Max Planck Institute for Metabolism Research, Cologne, Germany; Department of Radiology, Leiden University Medical Center, Leiden, Netherlands
| | - Zwi Berneman
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium; Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, University of Antwerp, Antwerp, Belgium; Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
47
|
Olde Heuvel F, Holl S, Chandrasekar A, Li Z, Wang Y, Rehman R, Förstner P, Sinske D, Palmer A, Wiesner D, Ludolph A, Huber-Lang M, Relja B, Wirth T, Röszer T, Baumann B, Boeckers T, Knöll B, Roselli F. STAT6 mediates the effect of ethanol on neuroinflammatory response in TBI. Brain Behav Immun 2019; 81:228-246. [PMID: 31207335 DOI: 10.1016/j.bbi.2019.06.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2019] [Revised: 05/31/2019] [Accepted: 06/13/2019] [Indexed: 12/14/2022] Open
Abstract
Traumatic brain injury (TBI) and ethanol intoxication (EI) frequently coincide, particularly in young subjects. However, the mechanisms of their interaction remain poorly understood. Among other pathogenic pathways, TBI induces glial activation and neuroinflammation in the hippocampus, resulting in acute and chronic hippocampal dysfunction. In this regard, we investigated the role of EI affecting these responses unfolding after TBI. We used a blunt, weight-drop approach to model TBI in mice. Male mice were pre-administered with ethanol or vehicle to simulate EI. The neuroinflammatory response in the hippocampus was assessed by monitoring the expression levels of >20 cytokines, the phosphorylation status of transcription factors and the phenotype of microglia and astrocytes. We used AS1517499, a brain-permeable STAT6 inhibitor, to elucidate the role of this pathway in the EI/TBI interaction. We showed that TBI causes the elevation of IL-33, IL-1β, IL-38, TNF-α, IFN-α, IL-19 in the hippocampus at 3 h time point and concomitant EI results in the dose-dependent downregulation of IL-33, IL-1β, IL-38, TNF-α and IL-19 (but not of IFN-α) and in the selective upregulation of IL-13 and IL-12. EI is associated with the phosphorylation of STAT6 and the transcription of STAT6-controlled genes. Moreover, ethanol-induced STAT6 phosphorylation and transcriptional activation can be recapitulated in vitro by concomitant exposure of neurons to ethanol, depolarization and inflammatory stimuli (simulating the acute trauma). Acute STAT6 inhibition prevents the effects of EI on IL-33 and TNF-α, but not on IL-13 and negates acute EI beneficial effects on TBI-associated neurological impairment. Additionally, EI is associated with reduced microglial activation and astrogliosis as well as preserved synaptic density and baseline neuronal activity 7 days after TBI and all these effects are prevented by acute administration of the STAT6 inhibitor concomitant to EI. EI concomitant to TBI exerts significant immunomodulatory effects on cytokine induction and microglial activation, largely through the activation of STAT6 pathway, ultimately with beneficial outcomes.
Collapse
Affiliation(s)
- Florian Olde Heuvel
- Dept. of Neurology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Sarah Holl
- Dept. of Neurology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Akila Chandrasekar
- Dept. of Neurology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Zhenghui Li
- Dept. of Neurology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Yibin Wang
- Dept. of Neurology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Rida Rehman
- Dept. of Neurology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Philip Förstner
- Institute of Physiological Chemistry, Ulm University, N27, Albert-Einstein-Allee 11 9081 Ulm, Germany
| | - Daniela Sinske
- Institute of Physiological Chemistry, Ulm University, N27, Albert-Einstein-Allee 11 9081 Ulm, Germany
| | - Annette Palmer
- Institute of Clinical and Experimental Trauma-Immunology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Diana Wiesner
- Dept. of Neurology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany; German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| | - Albert Ludolph
- Dept. of Neurology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany; German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| | - Markus Huber-Lang
- Institute of Clinical and Experimental Trauma-Immunology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany
| | - Borna Relja
- Dept. of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe-University Frankfurt am Main, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany
| | - Thomas Wirth
- Institute of Physiological Chemistry, Ulm University, N27, Albert-Einstein-Allee 11 9081 Ulm, Germany
| | - Tamás Röszer
- Institute of Neurobiology, Ulm University, M24, ALbert-Einstein Allee 11, 89081 Ulm, Germany
| | - Bernd Baumann
- Institute of Physiological Chemistry, Ulm University, N27, Albert-Einstein-Allee 11 9081 Ulm, Germany
| | - Tobias Boeckers
- German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany; Institute of Anatomy and Cell Biology, Ulm University, M24, ALbert-Einstein Allee 11, 89081 Ulm, Germany
| | - Bernd Knöll
- Institute of Physiological Chemistry, Ulm University, N27, Albert-Einstein-Allee 11 9081 Ulm, Germany
| | - Francesco Roselli
- Dept. of Neurology, Ulm University, ZBF - Helmholtzstrasse 8/1, 89081 Ulm, Germany; German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany; Institute of Anatomy and Cell Biology, Ulm University, M24, ALbert-Einstein Allee 11, 89081 Ulm, Germany.
| |
Collapse
|
48
|
Kolosowska N, Keuters MH, Wojciechowski S, Keksa-Goldsteine V, Laine M, Malm T, Goldsteins G, Koistinaho J, Dhungana H. Peripheral Administration of IL-13 Induces Anti-inflammatory Microglial/Macrophage Responses and Provides Neuroprotection in Ischemic Stroke. Neurotherapeutics 2019; 16:1304-1319. [PMID: 31372938 PMCID: PMC6985054 DOI: 10.1007/s13311-019-00761-0] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Neuroinflammation is strongly induced by cerebral ischemia. The early phase after the onset of ischemic stroke is characterized by acute neuronal injury, microglial activation, and subsequent infiltration of blood-derived inflammatory cells, including macrophages. Therefore, modulation of the microglial/macrophage responses has increasingly gained interest as a potential therapeutic approach for the ischemic stroke. In our study, we investigated the effects of peripherally administered interleukin 13 (IL-13) in a mouse model of permanent middle cerebral artery occlusion (pMCAo). Systemic administration of IL-13 immediately after the ischemic insult significantly reduced the lesion volume, alleviated the infiltration of CD45+ leukocytes, and promoted the microglia/macrophage alternative activation within the ischemic region, as determined by arginase 1 (Arg1) immunoreactivity at 3 days post-ischemia (dpi). Moreover, IL-13 enhanced the expression of M2a alternative activation markers Arg1 and Ym1 in the peri-ischemic (PI) area, as well as increased plasma IL-6 and IL-10 levels at 3 dpi. Furthermore, IL-13 treatment ameliorated gait disturbances at day 7 and 14 and sensorimotor deficits at day 14 post-ischemia, as analyzed by the CatWalk gait analysis system and adhesive removal test, respectively. Finally, IL-13 treatment decreased neuronal cell death in a coculture model of neuroinflammation with RAW 264.7 macrophages. Taken together, delivery of IL-13 enhances microglial/macrophage anti-inflammatory responses in vivo and in vitro, decreases ischemia-induced brain cell death, and improves sensory and motor functions in the pMCAo mouse model of cerebral ischemia.
Collapse
Affiliation(s)
- Natalia Kolosowska
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Meike H. Keuters
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Sara Wojciechowski
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Velta Keksa-Goldsteine
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Mika Laine
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Tarja Malm
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Gundars Goldsteins
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jari Koistinaho
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
- Neuroscience Center, HiLIFE, University of Helsinki, Haartmaninkatu 8, Helsinki, 00290 Finland
| | - Hiramani Dhungana
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
- Neuroscience Center, Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| |
Collapse
|
49
|
Sommer D, Corstjens I, Sanchez S, Dooley D, Lemmens S, Van Broeckhoven J, Bogie J, Vanmierlo T, Vidal PM, Rose-John S, Gou-Fabregas M, Hendrix S. ADAM17-deficiency on microglia but not on macrophages promotes phagocytosis and functional recovery after spinal cord injury. Brain Behav Immun 2019; 80:129-145. [PMID: 30851378 DOI: 10.1016/j.bbi.2019.02.032] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 02/12/2019] [Accepted: 02/27/2019] [Indexed: 12/26/2022] Open
Abstract
A disintegrin and metalloproteinase 17 (ADAM17) is the major sheddase involved in the cleavage of a plethora of cytokines, cytokine receptors and growth factors, thereby playing a substantial role in inflammatory and regenerative processes after central nervous system trauma. By making use of a hypomorphic ADAM17 knockin mouse model as well as pharmacological ADAM10/ADAM17 inhibitors, we showed that ADAM17-deficiency or inhibition significantly increases clearance of apoptotic cells, promotes axon growth and improves functional recovery after spinal cord injury (SCI) in mice. Microglia-specific ADAM17-knockout (ADAM17flox+/+-Cx3Cr1 Cre+/-) mice also showed improved functional recovery similar to hypomorphic ADAM17 mice. In contrast, endothelial-specific (ADAM17flox+/+-Cdh5Pacs Cre+/-) and macrophage-specific (ADAM17flox+/+-LysM Cre+/-) ADAM17-knockout mice or bone marrow chimera with transplanted ADAM17-deficient macrophages, displayed no functional improvement compared to wild type mice. These data indicate that ADAM17 expression on microglia cells (and not on macrophages or endothelial cells) plays a detrimental role in inflammation and functional recovery after SCI.
Collapse
Affiliation(s)
- Daniela Sommer
- Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium
| | - Inge Corstjens
- Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium
| | - Selien Sanchez
- Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium
| | - Dearbhaile Dooley
- Health Science Centre, School of Medicine, University College Dublin, Dublin 4, Ireland
| | - Stefanie Lemmens
- Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium
| | | | - Jeroen Bogie
- Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium
| | - Tim Vanmierlo
- Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium; Division of Translational Neuroscience, MHeNs, Maastricht University, 6229ER Maastricht, the Netherlands
| | - Pia M Vidal
- Laboratory of Neuroimmunology, Fundación Ciencia & Vida, 7780272 Santiago, Chile
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts University Kiel, 24098 Kiel, Germany
| | | | - Sven Hendrix
- Biomedical Research Institute, Hasselt University, 3500 Hasselt, Belgium.
| |
Collapse
|
50
|
Fernández M, Baldassarro VA, Capirossi R, Montevecchi R, Bonavita J, Cescatti M, Giovannini T, Giovannini G, Uneddu M, Giovanni G, Giardino L, Calzà L. Possible Strategies to Optimize a Biomarker Discovery Approach to Correlate with Neurological Outcome in Patients with Spinal Cord Injury: A Pilot Study. J Neurotrauma 2019; 37:431-440. [PMID: 31215324 DOI: 10.1089/neu.2018.6362] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The lack of reliable diagnostic and prognostic markers for spinal cord injured (SCI) patients is a severe obstacle in development and testing of new therapies, and it also impairs appropriate rehabilitation care. The sparse available data on the biochemical composition of cerebrospinal fluid (CSF) during the acute and/or chronic phase of the lesion provide, up until now, inconsistent results. In this pilot study, we then explored the possibility of combining a multi-parametric and bioinformatic analysis of CSF for its biological properties tested on different cells types, suitable for investigating inflammation and re-myelination. The patient enrollment was based on stringent inclusion criteria; that is, cervical and thoracic SCI trauma, CSF collection within 24 h of trauma, type of surgical approach for spine stabilization, and absence of steroid therapy before CSF collection. Eleven SCI patients and four healthy controls were included, and in three patients, CSF was also collected at 3 months after lesion. We identified 19 proteins among the 60 investigated cytokines, chemokines, growth factors, and structural biomarkers, which are transiently regulated 24 h after SCI. A bioinformatic analysis indicated that interleukin (IL)-6 and IL-10 are in the core of the interconnected net of activated proteins. Cell-based experiments indicate that CSF from SCI patients stimulates astroglia derivation from neural precursor cells, and an inverse correlation between IL-8 CSF level and oligodendrocyte precursor cells generated from neural stem cells was also observed. Results from this pilot study suggest that using a combined bioanalytic and biological approach to analyze SCI CSF at different times after injury could be a useful approach for identifying reliable diagnostic and prognostic markers in SCI.
Collapse
Affiliation(s)
- Mercedes Fernández
- Department of Veterinary Medical Sciences-DIMEVET, University of Bologna, Bologna, Italy
| | - Vito Antonio Baldassarro
- Department of Health Sciences and Technologies - Interdepartmental Center for Industrial Research-CIRI-SDV, University of Bologna, Bologna, Italy.,Department of Pharmacy and Biotechnology-Fabit, University of Bologna, Bologna, Italy
| | - Rita Capirossi
- Montecatone Rehabilitation Institute SpA, Imola, Bologna, Italy
| | - Roberto Montevecchi
- Bologna Local Health Authority - Intensive Care Unit, EMS and Trauma Centre, Maggiore Hospital, Bologna, Italy
| | - Jacopo Bonavita
- Montecatone Rehabilitation Institute SpA, Imola, Bologna, Italy
| | | | | | | | - Mariella Uneddu
- Montecatone Rehabilitation Institute SpA, Imola, Bologna, Italy
| | - Gordini Giovanni
- Bologna Local Health Authority - Intensive Care Unit, EMS and Trauma Centre, Maggiore Hospital, Bologna, Italy
| | - Luciana Giardino
- Department of Veterinary Medical Sciences-DIMEVET, University of Bologna, Bologna, Italy.,Department of Health Sciences and Technologies - Interdepartmental Center for Industrial Research-CIRI-SDV, University of Bologna, Bologna, Italy.,IRET Foundation, Ozzano Emilia, Italy
| | - Laura Calzà
- Department of Health Sciences and Technologies - Interdepartmental Center for Industrial Research-CIRI-SDV, University of Bologna, Bologna, Italy.,Department of Pharmacy and Biotechnology-Fabit, University of Bologna, Bologna, Italy.,IRET Foundation, Ozzano Emilia, Italy
| |
Collapse
|