1
|
Campos J, Palha AT, Fernandes LS, Cibrão JR, Pinho TS, Serra SC, Silva NA, Michael-Titus AT, Salgado AJ. Modeling Spinal Cord Injury in a Dish with Hyperosmotic Stress: Population-Specific Effects and the Modulatory Role of Mesenchymal Stromal Cell Secretome. Int J Mol Sci 2025; 26:3298. [PMID: 40244122 PMCID: PMC11989751 DOI: 10.3390/ijms26073298] [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: 01/17/2025] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
Innovations in spinal cord injury (SCI) models are crucial for developing effective therapies. This study introduces a novel in vitro SCI model using cultures of primary mixed spinal cord cells from rat pups, featuring key spinal cord cell types. This model offers distinct advantages in terms of feasibility, reproducibility, and cost-effectiveness, requiring only basic cell culture equipment. Following hyperosmotic stress via sorbitol treatment, the model recapitulated SCI pathophysiological hallmarks, with a 65% reduction in cell viability and gradual cell death over 48 h, making it ideal for evaluating neuroprotective agents. Notably, the human adipose tissue stem cell (hASC) secretome provided significant protection: it preserved metabolic viability, reduced β amyloid precursor protein (β-APP) expression in surviving neurons, and modulated the shift in the astrocytic morphotype. A transcriptomic profile of the effect of the hASC secretome treatment showed significant functional enrichments related to cell proliferation and cycle progression pathways. In addition to supporting the use of the hASC secretome as a therapy for SCI, this study is the first to use sorbitol as a hyperosmolar stressor to recapitulate key aspects of SCI pathophysiology. Thereby, this model can be used as a promising platform for evaluating therapeutic agents targeting neuroprotection and neuroregeneration, offering outputs related to cell death, neuronal stress, and protection, as well as induction of glial reactivity.
Collapse
Affiliation(s)
- Jonas Campos
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (J.C.); (A.T.P.); (L.S.F.); (J.R.C.); (T.S.P.); (S.C.S.); (N.A.S.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimaraes, Portugal
| | - Ana T. Palha
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (J.C.); (A.T.P.); (L.S.F.); (J.R.C.); (T.S.P.); (S.C.S.); (N.A.S.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimaraes, Portugal
| | - Luís S. Fernandes
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (J.C.); (A.T.P.); (L.S.F.); (J.R.C.); (T.S.P.); (S.C.S.); (N.A.S.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimaraes, Portugal
| | - Jorge R. Cibrão
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (J.C.); (A.T.P.); (L.S.F.); (J.R.C.); (T.S.P.); (S.C.S.); (N.A.S.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimaraes, Portugal
| | - Tiffany S. Pinho
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (J.C.); (A.T.P.); (L.S.F.); (J.R.C.); (T.S.P.); (S.C.S.); (N.A.S.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimaraes, Portugal
| | - Sofia C. Serra
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (J.C.); (A.T.P.); (L.S.F.); (J.R.C.); (T.S.P.); (S.C.S.); (N.A.S.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimaraes, Portugal
| | - Nuno A. Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (J.C.); (A.T.P.); (L.S.F.); (J.R.C.); (T.S.P.); (S.C.S.); (N.A.S.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimaraes, Portugal
| | - Adina T. Michael-Titus
- Centre for Neuroscience, Surgery and Trauma, The Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London E1 2AT, UK;
| | - António J. Salgado
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus de Gualtar, University of Minho, 4710-057 Braga, Portugal; (J.C.); (A.T.P.); (L.S.F.); (J.R.C.); (T.S.P.); (S.C.S.); (N.A.S.)
- ICVS/3B’s—PT Government Associate Laboratory, 4805-017 Guimaraes, Portugal
| |
Collapse
|
2
|
Yang Q, Zhang H, Jin Z, Zhang B, Wang Y. Effects of Valproic Acid Therapy on Rats with Spinal Cord Injury: A Systematic Review and Meta-Analysis. World Neurosurg 2024; 182:12-28. [PMID: 37923014 DOI: 10.1016/j.wneu.2023.10.135] [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: 10/04/2023] [Revised: 10/27/2023] [Accepted: 10/28/2023] [Indexed: 11/07/2023]
Abstract
OBJECTIVE To systematically evaluate the efficacy of valproic acid (VPA) in rats with spinal cord injury (SCI) to reduce the risk of clinical conversion and provide a valuable reference for future animal and clinical studies. METHODS We searched scientific databases, including PubMed, Ovid-Embase, Web of Science, and Scopus databases. The relevant literature was searched from the establishment date of the database to June 28, 2023. The search results were screened, data were extracted, and the quality of the literature was evaluated independently by 2 reviewers. RESULTS Among 656 nonduplicated references, 14 articles were included for meta-analysis. The summary results showed that the overall Basso, Beattie and Bresnahan scores of the VPA intervention group were significantly higher than those in the control group at 1-6 weeks after VPA intervention. Subgroup analysis showed that the injury model, administration dose, rat strain, country of study, or follow-up duration had no significant effect on the efficacy of VPA on rats with SCI. In addition, mesh analysis showed that high doses of the VPA group had a better effect on SCI rats, compared with the low dose group and the medium dose group. CONCLUSIONS To date, this is the first systematic evaluation of the potential effects of VPA on motor recovery in rats with SCI. We concluded that VPA can promote motor recovery in rats with SCI, and higher doses of VPA seem to be more effective in rats with SCI. However, the limited quality and sample of included studies reduced the application of this meta-analysis. In the future, more high-quality, direct comparative studies are needed to explore this issue in depth.
Collapse
Affiliation(s)
- Qinglin Yang
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Huaibin Zhang
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Zhuanmei Jin
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Baolin Zhang
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| | - Yongping Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou, China; Department of Orthopedics, The First Hospital of Lanzhou University, Lanzhou, China.
| |
Collapse
|
3
|
Patil V, Bohara R, Krishna Kanala V, McMahon S, Pandit A. Models and approaches to comprehend and address glial inflammation following spinal cord injury. Drug Discov Today 2023; 28:103722. [PMID: 37482236 DOI: 10.1016/j.drudis.2023.103722] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/10/2023] [Accepted: 07/18/2023] [Indexed: 07/25/2023]
Abstract
Spinal cord injury (SCI) culminates in chronic inflammation and glial scar formation driven by the activation of microglia and astrocytes. Current anti-inflammatory strategies to treat glial activation associated with SCI have several limitations. Existing in vitro and ex vivo models studying molecular mechanisms associated with inflammation focus only on the acute phase. However, the progression of glial cell-derived inflammation over the acute-to-chronic phases has not been assessed. Understanding this progression will help establish a framework for evaluating therapeutic strategies. Additionally, new models could be useful as high-throughput screening (HTS) platforms. This review aims to highlight currently available models and future methods that could facilitate screening of novel therapeutics for SCI.
Collapse
Affiliation(s)
- Vaibhav Patil
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Raghvendra Bohara
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Vijaya Krishna Kanala
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Siobhan McMahon
- Anatomy, School of Medicine, University of Galway, Galway, Ireland
| | - Abhay Pandit
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland.
| |
Collapse
|
4
|
Guinard I, Nguyen T, Brassard-Jollive N, Weber J, Ruch L, Reininger L, Brouard N, Eckly A, Collin D, Lanza F, Léon C. Matrix stiffness controls megakaryocyte adhesion, fibronectin fibrillogenesis, and proplatelet formation through Itgβ3. Blood Adv 2023; 7:4003-4018. [PMID: 37171626 PMCID: PMC10410137 DOI: 10.1182/bloodadvances.2022008680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 04/13/2023] [Accepted: 04/17/2023] [Indexed: 05/13/2023] Open
Abstract
Megakaryocytes (MKs) are the precursor cells of platelets, located in the bone marrow (BM). Once mature, they extend elongated projections named proplatelets through sinusoid vessels, emerging from the marrow stroma into the circulating blood. Not all signals from the microenvironment that regulate proplatelet formation are understood, particularly those from the BM biomechanics. We sought to investigate how MKs perceive and adapt to modifications of the stiffness of their environment. Although the BM is one of the softest tissue of the body, its rigidification results from excess fibronectin (FN), and other matrix protein deposition occur upon myelofibrosis. Here, we have shown that mouse MKs are able to detect the stiffness of a FN-coated substrate and adapt their morphology accordingly. Using a polydimethylsiloxane substrate with stiffness varying from physiological to pathological marrow, we found that a stiff matrix favors spreading, intracellular contractility, and FN fibrils assembly at the expense of proplatelet formation. Itgb3, but not Itgb1, is required for stiffness sensing, whereas both integrins are involved in fibrils assembly. In contrast, soft substrates promote proplatelet formation in an Itgb3-dependent manner, consistent with the ex vivo decrease in proplatelet formation and the in vivo decrease in platelet number in Itgb3-deficient mice. Our findings demonstrate the importance of environmental stiffness for MK functions with potential pathophysiological implications during pathologies that deregulate FN deposition and modulate stiffness in the marrow.
Collapse
Affiliation(s)
- Ines Guinard
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Thao Nguyen
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Noémie Brassard-Jollive
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Josiane Weber
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Laurie Ruch
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Laura Reininger
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Nathalie Brouard
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Anita Eckly
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | | | - François Lanza
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| | - Catherine Léon
- UMR_S1255, INSERM, Etablissement Français du Sang-Grand Est, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Strasbourg, France
| |
Collapse
|
5
|
Hanna ME, Pfister BJ. Advancements in in vitro models of traumatic brain injury. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2022. [DOI: 10.1016/j.cobme.2022.100430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
6
|
Feng Y, Li Y, Shen PP, Wang B. Gene-Modified Stem Cells for Spinal Cord Injury: a Promising Better Alternative Therapy. Stem Cell Rev Rep 2022; 18:2662-2682. [PMID: 35587330 DOI: 10.1007/s12015-022-10387-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2022] [Indexed: 12/18/2022]
Abstract
Stem cell therapy holds great promise for the treatment of spinal cord injury (SCI), which can reverse neurodegeneration and promote tissue regeneration via its pluripotency and ability to secrete neurotrophic factors. Although various stem cell-based approaches have shown certain therapeutic effects when applied to the treatment of SCI, their clinical efficacies have been disappointing. Thus, it is an urgent need to further enhance the neurological benefits of stem cells through bioengineering strategies including genetic engineering. In this review, we summarize the progress of stem cell therapy for SCI and the prospect of genetically modified stem cells, focusing on the genome editing tools and functional molecules involved in SCI repair, trying to provide a deeper understanding of genetically modified stem cell therapy and more applicable clinical strategies for SCI repair.
Collapse
Affiliation(s)
- Yirui Feng
- Clinical Stem Cell Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, School of Life Science, Nanjing University, Nanjing, Jiangsu Province, China
| | - Yu Li
- Clinical Stem Cell Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, School of Life Science, Nanjing University, Nanjing, Jiangsu Province, China
| | - Ping-Ping Shen
- State Key Laboratory of Pharmaceutical Biotechnology and the Comprehensive Cancer Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, School of Life Science, Nanjing University, Nanjing, Jiangsu Province, China.
| | - Bin Wang
- Clinical Stem Cell Center, the Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu Province, China.
| |
Collapse
|
7
|
Alternative Brain Slice-on-a-Chip for Organotypic Culture and Effective Fluorescence Injection Testing. Int J Mol Sci 2022; 23:ijms23052549. [PMID: 35269696 PMCID: PMC8910551 DOI: 10.3390/ijms23052549] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/21/2022] [Accepted: 02/23/2022] [Indexed: 11/22/2022] Open
Abstract
Mouse brain slices are one of the most common models to study brain development and functioning, increasing the number of study models that integrate microfluidic systems for hippocampal slice cultures. This report presents an alternative brain slice-on-a-chip, integrating an injection system inside the chip to dispense a fluorescent dye for long-term monitoring. Hippocampal slices have been cultured inside these chips, observing fluorescence signals from living cells, maintaining the cytoarchitecture of the slices. Having fluorescence images of biological samples inside the chip demonstrates the effectiveness of the staining process using the injection method avoiding leaks or biological contamination. The technology developed in this study presents a significant improvement in the local administration of reagents within a brain slice-on-a-chip system, which could be a suitable option for organotypic cultures in a microfluidic chip acting as a highly effective bioreactor.
Collapse
|
8
|
Zhou C, Hu S, Botchway BOA, Zhang Y, Liu X. Valproic Acid: A Potential Therapeutic for Spinal Cord Injury. Cell Mol Neurobiol 2021; 41:1441-1452. [PMID: 32725456 PMCID: PMC11448682 DOI: 10.1007/s10571-020-00929-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/20/2020] [Indexed: 02/06/2023]
Abstract
The lack of an effective pharmaceutical agent for spinal cord injury (SCI) is a current problematic situation for clinicians, as the rate of motor vehicle accidents among young adults is on the rise. SCI contributes to the high disability rate. Presently, evidences detailing the precise pathological mechanisms in SCI are limited, compounding to the unavailability of an effective treatment method. Surgery, though not a complete curative method, is useful in managing some of the associated symptoms of secondary SCI. Autophagy and inflammation are contributive factors to both exacerbation and improvement of SCI. The mammalian target of rapamycin (mTOR) signaling pathway is a key player in the regulation of inflammatory response and autophagy. Valproic acid (VPA), a clinically used antiepileptic drug, has been suggested to improve neurological conditions, including SCI. This report reviewed the correlation between mTOR and autophagy, as well as autophagy's role and the therapeutic effects of VPA in SCI. VPA regulates autophagy by potentially inhibiting mTORC1, a complex of mTOR, while also hindering inflammatory response. Conclusively, an effective treatment for SCI could lie in the timely regulation of mTOR signaling pathway, and VPA could be the potential drug that improves SCI owing to its propensity to regulate the mTOR signaling pathway.
Collapse
Affiliation(s)
- Conghui Zhou
- Department of Histology and Embryology, Medical College, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China
| | - Songfeng Hu
- Department of Orthopedics, Shaoxing Hospital of Traditional Chinese Medicine, Shaoxing, 312000, Zhejiang Province, China
| | - Benson O A Botchway
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
| | - Yong Zhang
- Department of Histology and Embryology, Medical College, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China
| | - Xuehong Liu
- Department of Histology and Embryology, Medical College, Shaoxing University, Shaoxing, 312000, Zhejiang Province, China.
| |
Collapse
|
9
|
Koroglu OF, Gunata M, Vardi N, Yildiz A, Ates B, Colak C, Tanriverdi LH, Parlakpinar H. Protective effects of naringin on valproic acid-induced hepatotoxicity in rats. Tissue Cell 2021; 72:101526. [PMID: 33756270 DOI: 10.1016/j.tice.2021.101526] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 02/22/2021] [Accepted: 03/09/2021] [Indexed: 02/06/2023]
Abstract
Valproic acid (VPA) is mainly prescribed to treat epilepsy. VPA has been reported to be associated with many adverse effects, including hepatotoxicity. Naringin (NRG) is a natural, therapeutically active flavanone glycoside with anti-inflammatory, anti-apoptotic, and antioxidant. The current study was therefore designed to investigate the protective effect of NRG against the VPA-induced experimental hepatotoxicity model. For this purpose, 24 Wistar albino rats were randomly divided into three groups as control (Vehicle), VPA (500 mg/kg), and NRG + VPA (100 mg/kg NRG + 500 mg/kg VPA) groups. The agents were administered via oral gavage for 14 days. Blood and liver tissue samples were taken on the end of the experiment. Biochemical analyzes were performed on the blood and liver samples. Also, malondialdehyde (MDA), superoxide dismutase (SOD) enzyme, glutathione (GSH) content, catalase (CAT) enzyme levels were examined in the liver tissue samples. Histopathological changes (hydropic degeneration and congestion) in the VPA group were increased significantly when compared to the control group (p < 0.05). We also found a decrease in enzymes of serum liver function in the VPA group. However, NRG has been shown not to prevent histopathological changes in the VPA group. According to our results with this experiment protocol, NRG could not exert sufficient protection against VPA-induced hepatotoxicity.
Collapse
Affiliation(s)
- Omer Faruk Koroglu
- Medical Student, Faculty of Medicine, Inonu University, Malatya, 44280, Turkey
| | - Mehmet Gunata
- Department of Medical Pharmacology, Faculty of Medicine, Inonu University, Malatya, 44280, Turkey
| | - Nigar Vardi
- Department of Histology and Embryology, Faculty of Medicine, Inonu University, Malatya, 44280, Turkey
| | - Azibe Yildiz
- Department of Histology and Embryology, Faculty of Medicine, Inonu University, Malatya, 44280, Turkey
| | - Burhan Ates
- Department of Chemistry, Faculty of Science and Arts, İnonu University, Malatya, 44280, Turkey
| | - Cemil Colak
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Inonu University, Malatya, 44280, Turkey
| | - Lokman Hekim Tanriverdi
- Department of Medical Pharmacology, Faculty of Medicine, Inonu University, Malatya, 44280, Turkey
| | - Hakan Parlakpinar
- Department of Medical Pharmacology, Faculty of Medicine, Inonu University, Malatya, 44280, Turkey.
| |
Collapse
|
10
|
Wood CR, Juárez EH, Ferrini F, Myint P, Innes J, Lossi L, Merighi A, Johnson WEB. Mesenchymal stem cell conditioned medium increases glial reactivity and decreases neuronal survival in spinal cord slice cultures. Biochem Biophys Rep 2021; 26:100976. [PMID: 33718633 PMCID: PMC7933697 DOI: 10.1016/j.bbrep.2021.100976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/22/2021] [Accepted: 02/22/2021] [Indexed: 12/13/2022] Open
Abstract
Ex vivo spinal cord slice cultures (SCSC) allow study of spinal cord circuitry, maintaining stimuli responses comparable to live animals. Previously, we have shown that mesenchymal stem/stromal cell (MSC) transplantation in vivo reduced inflammation and increased nerve regeneration but MSC survival was short-lived, highlighting that beneficial action may derive from the secretome. Previous in vitro studies of MSC conditioned medium (CM) have also shown increased neuronal growth. In this study, murine SCSC were cultured in canine MSC CM (harvested from the adipose tissue of excised inguinal fat) and cell phenotypes analysed via immunohistochemistry and confocal microscopy. SCSC in MSC CM displayed enhanced viability after propidium iodide staining. GFAP immunoreactivity was significantly increased in SCSC in MSC CM compared to controls, but with no change in proteoglycan (NG2) immunoreactivity. In contrast, culture in MSC CM significantly decreased the prevalence of βIII-tubulin immunoreactive neurites, whilst Ca2+ transients per cell were significantly increased. These ex vivo results contradict previous in vitro and in vivo reports of how MSC and their secretome may affect the microenvironment of the spinal cord after injury and highlight the importance of a careful comparison of the different experimental conditions used to assess the potential of cell therapies for the treatment of spinal cord injury. Treatment of spinal slices with conditioned medium caused cell phenotypic changes. Resident astrocytes become hypertrophic, yet neuronal axonal outgrowth reduced. Signalling cells reduced in number but increased their signalling activity. Highlights importance of simulation systems and systemic factors in CNS models.
Collapse
Affiliation(s)
- Chelsea R Wood
- Department of Biological Sciences, University of Chester, Parkgate Road, Chester, CH1 4BJ, UK
| | - Esri H Juárez
- Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, I-10095, Grugliasco, TO, Italy
| | - Francesco Ferrini
- Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, I-10095, Grugliasco, TO, Italy.,Université Laval, Department of Psychiatry and Neuroscience, G1K 7P4, Québec, Canada
| | - Peter Myint
- Veterinary Tissue Bank Ltd., No.1 The Long Barn, Brynkinalt Business Centre, Chirk, Wrexham, LL14 5NS, UK
| | - John Innes
- Veterinary Tissue Bank Ltd., No.1 The Long Barn, Brynkinalt Business Centre, Chirk, Wrexham, LL14 5NS, UK
| | - Laura Lossi
- Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, I-10095, Grugliasco, TO, Italy
| | - Adalberto Merighi
- Department of Veterinary Sciences, University of Turin, Largo Paolo Braccini 2, I-10095, Grugliasco, TO, Italy
| | - William E B Johnson
- Department of Biological Sciences, University of Chester, Parkgate Road, Chester, CH1 4BJ, UK
| |
Collapse
|
11
|
Santi S, Corridori I, Pugno NM, Motta A, Migliaresi C. Injectable Scaffold-Systems for the Regeneration of Spinal Cord: Advances of the Past Decade. ACS Biomater Sci Eng 2021; 7:983-999. [PMID: 33523634 DOI: 10.1021/acsbiomaterials.0c01779] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Nowadays, whenever is possible and as an alternative to open spine surgery, minimally invasive procedures are preferred to treat spinal cord injuries (SCI), with percutaneous injections or small incisions, that are faster, less traumatic, and require less recovery time. Injectable repair systems are based on materials that can be injected in the lesion site, can eventually be loaded with drugs or even cells, and act as scaffolds for the lesion repair. The review analyzes papers written from 2010 onward on injectable materials/systems used/proposed for the regenerative and combinatorial therapies of SCI and discusses the in vivo models that have been used to validate them.
Collapse
Affiliation(s)
- Sofia Santi
- BIOTech Research Center and European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Trento, Via delle Regole 101, 38123 Trento, Italy.,Department of Industrial Engineering, University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Ilaria Corridori
- Laboratory of Bio-inspired, Bionic, Nano, Meta Materials & Mechanics, Department of Civil, Environmental and Mechanical Engineering, University of Trento, Via Mesiano 77, 38123 Trento, Italy
| | - Nicola M Pugno
- Laboratory of Bio-inspired, Bionic, Nano, Meta Materials & Mechanics, Department of Civil, Environmental and Mechanical Engineering, University of Trento, Via Mesiano 77, 38123 Trento, Italy.,School of Engineering and Material Science, Queen Mary University of London, Mile End Road, E1 4NS London, United Kingdom
| | - Antonella Motta
- BIOTech Research Center and European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Trento, Via delle Regole 101, 38123 Trento, Italy.,Department of Industrial Engineering, University of Trento, Via Sommarive 9, 38123 Trento, Italy
| | - Claudio Migliaresi
- BIOTech Research Center and European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Trento, Via delle Regole 101, 38123 Trento, Italy.,Department of Industrial Engineering, University of Trento, Via Sommarive 9, 38123 Trento, Italy
| |
Collapse
|
12
|
Lin C, Calzarossa C, Fernandez-Zafra T, Liu J, Li X, Ekblad-Nordberg Å, Vazquez-Juarez E, Codeluppi S, Holmberg L, Lindskog M, Uhlén P, Åkesson E. Human ex vivo spinal cord slice culture as a useful model of neural development, lesion, and allogeneic neural cell therapy. Stem Cell Res Ther 2020; 11:320. [PMID: 32727554 PMCID: PMC7390865 DOI: 10.1186/s13287-020-01771-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/18/2020] [Accepted: 06/12/2020] [Indexed: 12/14/2022] Open
Abstract
Background There are multiple promising treatment strategies for central nervous system trauma and disease. However, to develop clinically potent and safe treatments, models of human-specific conditions are needed to complement in vitro and in vivo animal model-based studies. Methods We established human brain stem and spinal cord (cross- and longitudinal sections) organotypic cultures (hOCs) from first trimester tissues after informed consent by donor and ethical approval by the Regional Human Ethics Committee, Stockholm (lately referred to as Swedish Ethical Review Authority), and The National Board of Health and Welfare, Sweden. We evaluated the stability of hOCs with a semi-quantitative hOC score, immunohistochemistry, flow cytometry, Ca2+ signaling, and electrophysiological analysis. We also applied experimental allogeneic human neural cell therapy after injury in the ex vivo spinal cord slices. Results The spinal cord hOCs presented relatively stable features during 7–21 days in vitro (DIV) (except a slightly increased cell proliferation and activated glial response). After contusion injury performed at 7 DIV, a significant reduction of the hOC score, increase of the activated caspase-3+ cell population, and activated microglial populations at 14 days postinjury compared to sham controls were observed. Such elevation in the activated caspase-3+ population and activated microglial population was not observed after allogeneic human neural cell therapy. Conclusions We conclude that human spinal cord slice cultures have potential for future structural and functional studies of human spinal cord development, injury, and treatment strategies.
Collapse
Affiliation(s)
- Chenhong Lin
- Department of Neurobiology, Care Sciences and Society, Div. of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Cinzia Calzarossa
- Department of Neurobiology, Care Sciences and Society, Div. of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden.,Department of Neurology and Laboratory of Neuroscience, Università degli Studi diMilan, Milan, Italy
| | - Teresa Fernandez-Zafra
- Division of Molecular Neurobiology, Departmentof Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Jia Liu
- Department of Neurobiology, Care Sciences and Society, Div. of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden.,Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, People's Republic of China
| | - Xiaofei Li
- Department of Neurobiology, Care Sciences and Society, Div. of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Åsa Ekblad-Nordberg
- Department of Clinical Science, Intervention and Technology, Div. of Obstetrics and Gynecology, Karolinska Institutet, Stockholm, Sweden
| | - Erika Vazquez-Juarez
- Department of Neurobiology, Care Sciences and Society, Div. of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Simone Codeluppi
- Division of Molecular Neurobiology, Departmentof Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Lena Holmberg
- Department of Neurobiology, Care Sciences and Society, Div. of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Maria Lindskog
- Department of Neurobiology, Care Sciences and Society, Div. of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden
| | - Per Uhlén
- Division of Molecular Neurobiology, Departmentof Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Elisabet Åkesson
- Department of Neurobiology, Care Sciences and Society, Div. of Neurogeriatrics, Karolinska Institutet, Stockholm, Sweden. .,The R&D Unit, Stockholms Sjukhem, Stockholm, Sweden.
| |
Collapse
|
13
|
Omelchenko A, Singh NK, Firestein BL. Current advances in in vitro models of central nervous system trauma. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2020; 14:34-41. [PMID: 32671312 PMCID: PMC7363028 DOI: 10.1016/j.cobme.2020.05.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
CNS trauma is a prominent cause of mortality and morbidity, and although much effort has focused on developing treatments for CNS trauma-related pathologies, little progress has been made. Pre-clinical models of TBI and SCI suffer from significant drawbacks, which result in substantial failures during clinical translation of promising pre-clinical therapies. Here, we review recent advances made in the development of in vitro models of CNS trauma, the promises and drawbacks of current in vitro CNS injury models, and the attributes necessary for future models to accurately mimic the trauma microenvironment and facilitate CNS trauma drug discovery. The goal is to provide insight for the development of future CNS injury models and to aid researchers in selecting effective models for pre-clinical research of trauma therapeutics.
Collapse
Affiliation(s)
- Anton Omelchenko
- Department of Cell Biology and Neuroscience; Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854-8082
- Neuroscience Graduate Program, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854-8082
| | - Nisha K. Singh
- Department of Cell Biology and Neuroscience; Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854-8082
- Molecular Biosciences Graduate Program, Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854-8082
| | - Bonnie L. Firestein
- Department of Cell Biology and Neuroscience; Rutgers, The State University of New Jersey, 604 Allison Road, Piscataway, NJ 08854-8082
| |
Collapse
|
14
|
Gulino M, Kim D, Pané S, Santos SD, Pêgo AP. Tissue Response to Neural Implants: The Use of Model Systems Toward New Design Solutions of Implantable Microelectrodes. Front Neurosci 2019; 13:689. [PMID: 31333407 PMCID: PMC6624471 DOI: 10.3389/fnins.2019.00689] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 06/18/2019] [Indexed: 01/28/2023] Open
Abstract
The development of implantable neuroelectrodes is advancing rapidly as these tools are becoming increasingly ubiquitous in clinical practice, especially for the treatment of traumatic and neurodegenerative disorders. Electrodes have been exploited in a wide number of neural interface devices, such as deep brain stimulation, which is one of the most successful therapies with proven efficacy in the treatment of diseases like Parkinson or epilepsy. However, one of the main caveats related to the clinical application of electrodes is the nervous tissue response at the injury site, characterized by a cascade of inflammatory events, which culminate in chronic inflammation, and, in turn, result in the failure of the implant over extended periods of time. To overcome current limitations of the most widespread macroelectrode based systems, new design strategies and the development of innovative materials with superior biocompatibility characteristics are currently being investigated. This review describes the current state of the art of in vitro, ex vivo, and in vivo models available for the study of neural tissue response to implantable microelectrodes. We particularly highlight new models with increased complexity that closely mimic in vivo scenarios and that can serve as promising alternatives to animal studies for investigation of microelectrodes in neural tissues. Additionally, we also express our view on the impact of the progress in the field of neural tissue engineering on neural implant research.
Collapse
Affiliation(s)
- Maurizio Gulino
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- FEUP – Faculdade de Engenharia, Universidade do Porto, Porto, Portugal
| | - Donghoon Kim
- Multi-Scale Robotics Lab (MSRL), Institute of Robotics and Intelligent Systems (IRIS), ETH Zurich, Zurich, Switzerland
| | - Salvador Pané
- Multi-Scale Robotics Lab (MSRL), Institute of Robotics and Intelligent Systems (IRIS), ETH Zurich, Zurich, Switzerland
| | - Sofia Duque Santos
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | - Ana Paula Pêgo
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- FEUP – Faculdade de Engenharia, Universidade do Porto, Porto, Portugal
- ICBAS – Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| |
Collapse
|