1
|
Zhang A, Sun B, Nan C, Cong L, Zhao Z, Liu L. Effects of 3D-printed exosome-functionalized brain acellular matrix hydrogel on neuroinflammation in rats following cerebral hemorrhage. Stem Cell Res Ther 2025; 16:196. [PMID: 40254565 PMCID: PMC12010578 DOI: 10.1186/s13287-025-04332-3] [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: 01/28/2025] [Accepted: 04/10/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND Exosome-based therapeutics have garnered significant attention for intracerebral hemorrhage (ICH) treatment due to their capacity to regulate metabolic dysregulation, restore cellular homeostasis, and modulate the injury microenvironment via bioactive cargoes such as microRNAs and proteins. However, rapid systemic clearance and enzymatic degradation critically limit their therapeutic efficacy. To address this challenge, we engineered a three-dimensional (3D) bioprinted scaffold capable of encapsulating and sustaining the release of human umbilical cord mesenchymal stem cell-derived exosomes (hUCMSC-exos). METHODS Based on previous research [1-3], the scaffold was composed of a decellularized brain matrix (dECM), gelatin-methacryloyl (GelMA), and silk fibroin (SF) crosslinked with a photoinitiator. hUCMSC-exos were precisely incorporated via extrusion-based 3D bioprinting. Release kinetics were assessed via in vitro elution and in vivo imaging. An ICH rat model received stereotaxic implantation of the exosome-laden scaffold (dECM@exo). Neuroinflammatory markers (IL-6, TNF-α, IL-10) and apoptotic activity (JC-1, Annexin V/PI, TUNEL) were quantified. Neurological outcomes were longitudinally evaluated using the modified Longa scale, Bederson scoring, and sensorimotor tests (rotarod, forelimb placement) at 1, 4, 7 and 14 days post-ICH. RESULTS dECM@exo demonstrated sustained exosome release over 14 days, significantly promoting neural tissue regeneration while attenuating perihematomal edema. Mechanistically, the scaffold modulated pathological MMP activity and inflammatory cytokine expression, thereby restoring extracellular matrix homeostasis and reducing neuronal apoptosis. CONCLUSIONS The findings demonstrate that the 3D biological scaffold dECM@exo effectively maintains microenvironmental homeostasis in the early stages of ICH and improves outcomes associated with the condition. dECM@exo is poised to serve as a robust platform for drug delivery and biotherapy in ICH treatment, offering a viable alternative for managing this condition.
Collapse
Affiliation(s)
- Aobo Zhang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China.
| | - Boyu Sun
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Chengrui Nan
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Lulu Cong
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Zongmao Zhao
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China
| | - Liqiang Liu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, 050000, Hebei, China.
| |
Collapse
|
2
|
Jiang Y, Wang LY, Liu Y, Li JJ, Zhang SQ, Feng XJ, Yang CJ, Zhou Y. Cold atmospheric plasma-activated saline alleviates secondary injury post-SCI by inhibiting extracellular matrix remodeling and infiltration of proinflammatory macrophages. Exp Neurol 2024; 383:115004. [PMID: 39428043 DOI: 10.1016/j.expneurol.2024.115004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/30/2024] [Accepted: 10/14/2024] [Indexed: 10/22/2024]
Abstract
BACKGROUND Cold atmospheric plasma (CAP) has been shown to improve the recovery of transected peripheral nerves. We determined the protective role of CAP-activated saline (CAP-AS) treatment in the acute and subacute stages of spinal cord injury (SCI) in mice. METHODS C57BL/6 SCI mice were treated with CAP-AS for 14 days. Injury recovery was assessed weekly for four weeks by conducting motor function tests, including the Basso Mouse Scale (BMS) and footprint test. Transcriptome analysis was conducted on day 14 to elucidate potential mechanisms, which were further validated through immunofluorescence examinations of the injured spinal cord tissues on day 28 and the levels of proinflammatory cytokines produced by macrophages in vitro. RESULTS Compared to the SCI group, the CAP-AS-treated groups presented significantly better hindlimb motor function after four weeks. The downregulated (SCI vs. SCI + CAP-AS, with CAP-AS activated for 20 min) differentially expressed genes (DEGs) were enriched in the extracellular region, extracellular matrix (ECM), and ECM-receptor interaction. In contrast, the upregulated DEGs were enriched in immune response-associated pathways. Histological changes in the CAP-AS-treated groups were observed to further validate the predicted mechanisms 28 days post-injury. The alleviation of secondary injury was confirmed by an increase in GFAP-positive and NFH-positive areas, and enhanced outgrowth of 5-HT-positive fibers. Inhibited ECM remodeling was confirmed by a decrease in the areas positive for PDGFRβ, fibronectin, and laminin. A decrease in the infiltration of macrophages and activation of microglia was determined by a decrease in CD68-positive and F4/80-positive areas. The inhibitory effect of CAP-AS on inflammation was further supported by a decrease in the levels of the proinflammatory cytokines IL-1β, IL-6, and TNF-α in CAP-AS-treated M1 macrophages. CONCLUSION CAP-AS can alleviate secondary injury in SCI model mice by inhibiting ECM remodeling in injured tissues and reducing the infiltration or activation of proinflammatory macrophages/microglia.
Collapse
Affiliation(s)
- Yan Jiang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Li-Yun Wang
- Department of Dermatology and Venereology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; Joint Laboratory for Plasma Clinical Applications, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Yi Liu
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Jian-Jian Li
- Institute of Orthopaedics, Research Center for Translational Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China
| | - Sheng-Quan Zhang
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China
| | - Xiao-Jun Feng
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China.
| | - Chun-Jun Yang
- Department of Dermatology and Venereology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; Joint Laboratory for Plasma Clinical Applications, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China.
| | - Yun Zhou
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China.
| |
Collapse
|
3
|
Zhang A, Cong L, Nan C, Zhao Z, Liu L. 3D biological scaffold delivers Bergenin to reduce neuroinflammation in rats with cerebral hemorrhage. J Transl Med 2024; 22:946. [PMID: 39420402 PMCID: PMC11484212 DOI: 10.1186/s12967-024-05735-1] [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: 07/01/2024] [Accepted: 10/03/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) is a severe form of stroke characterized by high incidence and mortality rates. Currently, there is a significant lack of effective treatments aimed at improving clinical outcomes. Our research team has developed a three-dimensional (3D) biological scaffold that incorporates Bergenin, allowing for the sustained release of the compound. METHODS This 3D biological scaffold was fabricated using a combination of photoinitiator, GEMA, silk fibroin, and decellularized brain matrix (dECM) to encapsulate Bergenin through advanced 3D bioprinting techniques. The kinetics of drug release were evaluated through both in vivo and in vitro studies. A cerebral hemorrhage model was established, and a 3D biological scaffold containing Bergenin was transplanted in situ. Levels of inflammatory response, oxidative stress, and apoptosis were quantified. The neurological function of rats with cerebral hemorrhage was assessed on days 1, 3, and 5 using the turning test, forelimb placement test, Longa score, and Bederson score. RESULTS The 3D biological scaffold incorporating Bergenin significantly enhances the maintenance of drug concentration in the bloodstream, leading to a marked reduction in inflammatory markers such as IL-6, iNOS, and COX-2 levels in a cerebral hemorrhage model, primarily through the inhibition of the NF-κB pathway. Additionally, the scaffold effectively reduces the expression of hypoxia-inducible factor 1-alpha (HIF-1α) in primary cultured astrocytes, which in turn decreases the production of reactive oxygen species (ROS) and inhibits IL-6 production induced by hemin. Subsequent experiments reveal that the 3D biological scaffold containing Bergenin promotes the activation of the Nrf-2/HO-1 signaling pathway, both in vivo and in vitro, thereby preventing cell death. Moreover, the application of this 3D biological scaffold has been demonstrated to improve drug retention in the bloodstream. CONCLUSION This strategy effectively mitigates inflammation, oxidative stress, and cell death in rats with cerebral hemorrhage by inhibiting the NF-κB pathway while concurrently activating the Nrf-2/HO-1 pathway.
Collapse
Affiliation(s)
- Aobo Zhang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China
| | - Lulu Cong
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China
| | - Chengrui Nan
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China
| | - Zongmao Zhao
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China
| | - Liqiang Liu
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei, 050000, China.
| |
Collapse
|
4
|
Tabet A, Apra C, Stranahan AM, Anikeeva P. Changes in Brain Neuroimmunology Following Injury and Disease. Front Integr Neurosci 2022; 16:894500. [PMID: 35573444 PMCID: PMC9093707 DOI: 10.3389/fnint.2022.894500] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 04/04/2022] [Indexed: 01/21/2023] Open
Abstract
The nervous and immune systems are intimately related in the brain and in the periphery, where changes to one affect the other and vice-versa. Immune cells are responsible for sculpting and pruning neuronal synapses, and play key roles in neuro-development and neurological disease pathology. The immune composition of the brain is tightly regulated from the periphery through the blood-brain barrier (BBB), whose maintenance is driven to a significant extent by extracellular matrix (ECM) components. After a brain insult, the BBB can become disrupted and the composition of the ECM can change. These changes, and the resulting immune infiltration, can have detrimental effects on neurophysiology and are the hallmarks of several diseases. In this review, we discuss some processes that may occur after insult, and potential consequences to brain neuroimmunology and disease progression. We then highlight future research directions and opportunities for further tool development to probe the neuro-immune interface.
Collapse
Affiliation(s)
- Anthony Tabet
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, United States
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Caroline Apra
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, United States
- Sorbonne Universite, Paris, France
| | - Alexis M. Stranahan
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, GA, United States
| | - Polina Anikeeva
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, United States
- Research Laboratory of Electronics, Massachusetts Institute of Technology, Cambridge, MA, United States
- Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA, United States
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, United States
| |
Collapse
|
5
|
Decellularised extracellular matrix-based biomaterials for repair and regeneration of central nervous system. Expert Rev Mol Med 2022; 23:e25. [PMID: 34994341 PMCID: PMC9884794 DOI: 10.1017/erm.2021.22] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The central nervous system (CNS), consisting of the brain and spinal cord, regulates the mind and functions of the organs. CNS diseases, leading to changes in neurological functions in corresponding sites and causing long-term disability, represent one of the major public health issues with significant clinical and economic burdens worldwide. In particular, the abnormal changes in the extracellular matrix under various disease conditions have been demonstrated as one of the main factors that can alter normal cell function and reduce the neuroregeneration potential in damaged tissue. Decellularised extracellular matrix (dECM)-based biomaterials have been recently utilised for CNS applications, closely mimicking the native tissue. dECM retains tissue-specific components, including proteoglycan as well as structural and functional proteins. Due to their unique composition, these biomaterials can stimulate sensitive repair mechanisms associated with CNS damages. Herein, we discuss the decellularisation of the brain and spinal cord as well as recellularisation of acellular matrix and the recent progress in the utilisation of brain and spinal cord dECM.
Collapse
|
6
|
Cao Z, Man W, Xiong Y, Guo Y, Yang S, Liu D, Zhao H, Yang Y, Yao S, Li C, Zhao L, Sun X, Guo H, Wang G, Wang X. White matter regeneration induced by aligned fibrin nanofiber hydrogel contributes to motor functional recovery in canine T12 spinal cord injury. Regen Biomater 2021; 9:rbab069. [PMID: 35558095 PMCID: PMC9089163 DOI: 10.1093/rb/rbab069] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 09/24/2021] [Accepted: 11/16/2021] [Indexed: 11/25/2022] Open
Abstract
A hierarchically aligned fibrin hydrogel (AFG) that possesses soft stiffness and aligned nanofiber structure has been successfully proven to facilitate neuroregeneration in vitro and in vivo. However, its potential in promoting nerve regeneration in large animal models that is critical for clinical translation has not been sufficiently specified. Here, the effects of AFG on directing neuroregeneration in canine hemisected T12 spinal cord injuries were explored. Histologically obvious white matter regeneration consisting of a large area of consecutive, compact and aligned nerve fibers is induced by AFG, leading to a significant motor functional restoration. The canines with AFG implantation start to stand well with their defective legs from 3 to 4 weeks postoperatively and even effortlessly climb the steps from 7 to 8 weeks. Moreover, high-resolution multi-shot diffusion tensor imaging illustrates the spatiotemporal dynamics of nerve regeneration rapidly crossing the lesion within 4 weeks in the AFG group. Our findings indicate that AFG could be a potential therapeutic vehicle for spinal cord injury by inducing rapid white matter regeneration and restoring locomotion, pointing out its promising prospect in clinic practice.
Collapse
Affiliation(s)
- Zheng Cao
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Weitao Man
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Yuhui Xiong
- Center for Biomedical Imaging Research, Tsinghua University, Beijing 100084, China
| | - Yi Guo
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Shuhui Yang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Dongkang Liu
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - He Zhao
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
- Department of Orthopedics, Dongzhimen Hospital, Beijing 100007, China
| | - Yongdong Yang
- Department of Orthopedics, Dongzhimen Hospital, Beijing 100007, China
| | - Shenglian Yao
- School of Materials Science and Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Chuzhong Li
- Beijing Neurosurgical Institute, Beijing Tiantan Hospital, Beijing 100070, China
| | - Lingyun Zhao
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Xiaodan Sun
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| | - Hua Guo
- Center for Biomedical Imaging Research, Tsinghua University, Beijing 100084, China
| | - Guihuai Wang
- Department of Neurosurgery, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Xiumei Wang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, Beijing 100084, China
| |
Collapse
|
7
|
Kajtez J, Nilsson F, Fiorenzano A, Parmar M, Emnéus J. 3D biomaterial models of human brain disease. Neurochem Int 2021; 147:105043. [PMID: 33887378 DOI: 10.1016/j.neuint.2021.105043] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 03/21/2021] [Accepted: 04/06/2021] [Indexed: 01/25/2023]
Abstract
Inherent limitations of the traditional approaches to study brain function and disease, such as rodent models and 2D cell culture platforms, have led to the development of 3D in vitro cell culture systems. These systems, products of multidisciplinary efforts encompassing stem cell biology, materials engineering, and biofabrication, have quickly shown great potential to mimic biochemical composition, structural properties, and cellular morphology and diversity found in the native brain tissue. Crucial to these developments have been the advancements in stem cell technology and cell reprogramming protocols that allow reproducible generation of human subtype-specific neurons and glia in laboratory conditions. At the same time, biomaterials have been designed to provide cells in 3D with a microenvironment that mimics functional and structural aspects of the native extracellular matrix with increasing fidelity. In this article, we review the use of biomaterials in 3D in vitro models of neurological disorders with focus on hydrogel technology and with biochemical composition and physical properties of the in vivo environment as reference.
Collapse
Affiliation(s)
- Janko Kajtez
- Department of Experimental Medical Sciences, Wallenberg Neuroscience Center, Division of Neurobiology and Lund Stem Cell Center, BMC A11, Lund University, Lund, S-22184, Sweden.
| | - Fredrik Nilsson
- Department of Experimental Medical Sciences, Wallenberg Neuroscience Center, Division of Neurobiology and Lund Stem Cell Center, BMC A11, Lund University, Lund, S-22184, Sweden
| | - Alessandro Fiorenzano
- Department of Experimental Medical Sciences, Wallenberg Neuroscience Center, Division of Neurobiology and Lund Stem Cell Center, BMC A11, Lund University, Lund, S-22184, Sweden
| | - Malin Parmar
- Department of Experimental Medical Sciences, Wallenberg Neuroscience Center, Division of Neurobiology and Lund Stem Cell Center, BMC A11, Lund University, Lund, S-22184, Sweden
| | - Jenny Emnéus
- Department of Biotechnology and Biomedicine (DTU Bioengineering), Technical University of Denmark, Kongens Lyngby, Denmark
| |
Collapse
|
8
|
Guest JD, Moore SW, Aimetti AA, Kutikov AB, Santamaria AJ, Hofstetter CP, Ropper AE, Theodore N, Ulich TR, Layer RT. Internal decompression of the acutely contused spinal cord: Differential effects of irrigation only versus biodegradable scaffold implantation. Biomaterials 2018; 185:284-300. [DOI: 10.1016/j.biomaterials.2018.09.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2018] [Revised: 09/04/2018] [Accepted: 09/16/2018] [Indexed: 12/13/2022]
|
9
|
Proliferating NG2-Cell-Dependent Angiogenesis and Scar Formation Alter Axon Growth and Functional Recovery After Spinal Cord Injury in Mice. J Neurosci 2017; 38:1366-1382. [PMID: 29279310 DOI: 10.1523/jneurosci.3953-16.2017] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Revised: 11/18/2017] [Accepted: 12/17/2017] [Indexed: 01/10/2023] Open
Abstract
Spinal cord injury (SCI) induces a centralized fibrotic scar surrounded by a reactive glial scar at the lesion site. The origin of these scars is thought to be perivascular cells entering lesions on ingrowing blood vessels and reactive astrocytes, respectively. However, two NG2-expressing cell populations, pericytes and glia, may also influence scar formation. In the periphery, new blood vessel growth requires proliferating NG2+ pericytes; if this were also true in the CNS, then the fibrotic scar would depend on dividing NG2+ pericytes. NG2+ glial cells (also called oligodendrocyte progenitors or polydendrocytes) also proliferate after SCI and accumulate in large numbers among astrocytes in the glial scar. Their effect there, if any, is unknown. We show that proliferating NG2+ pericytes and glia largely segregate into the fibrotic and glial scars, respectively; therefore, we used a thymidine kinase/ganciclovir paradigm to ablate both dividing NG2+ cell populations to determine whether either scar was altered. Results reveal that loss of proliferating NG2+ pericytes in the lesion prevented intralesion angiogenesis and completely abolished the fibrotic scar. The glial scar was also altered in the absence of acutely dividing NG2+ cells, displaying discontinuous borders and significantly reduced GFAP density. Collectively, these changes enhanced edema, prolonged hemorrhage, and impaired forelimb functional recovery. Interestingly, after halting GCV at 14 d postinjury, scar elements and vessels entered the lesions over the next 7 d, as did large numbers of axons that were not present in controls. Collectively, these data reveal that acutely dividing NG2+ pericytes and glia play fundamental roles in post-SCI tissue remodeling.SIGNIFICANCE STATEMENT Spinal cord injury (SCI) is characterized by formation of astrocytic and fibrotic scars, both of which are necessary for lesion repair. NG2+ cells may influence both scar-forming processes. This study used a novel transgenic mouse paradigm to ablate proliferating NG2+ cells after SCI to better understand their role in repair. For the first time, our data show that dividing NG2+ pericytes are required for post-SCI angiogenesis, which in turn is needed for fibrotic scar formation. Moreover, loss of cycling NG2+ glia and pericytes caused significant multicellular tissue changes, including altered astrocyte responses and impaired functional recovery. This work reveals previously unknown ways in which proliferating NG2+ cells contribute to endogenous repair after SCI.
Collapse
|
10
|
The influence of fibroblast on the arachnoid leptomeningeal cells in vitro. Brain Res 2017; 1657:109-119. [PMID: 27923631 DOI: 10.1016/j.brainres.2016.12.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 11/26/2016] [Accepted: 12/02/2016] [Indexed: 11/24/2022]
Abstract
OBJECTIVE Fibroblast is pervasive in the setting of injury. Its invasion into the arachnoid tissue causes scarring, cortical adhesion of the brain, and obstruction of cerebrospinal fluid outflow. The purpose of this study is to determine the phenotypic and physiologic effects of fibroblasts on arachnoid in culture. METHODS We studied the effects of fibroblast on the arachnoid cell growth, motility, phenotypic changes, and transport properties. Immortalized rat (Rattus norvegicus, Sprague Dawley breed) arachnoid cells were grown with fibroblast on opposite sides of polyethylene membranes or co-cultured in plastic wells. Arachnoid cell growth rate and DNA content, morphology, transport physiology, and extracellular matriceal content were determined in the presence of normal and irradiated fibroblast cells. RESULTS When arachnoid cells were grown in the presence of fibroblasts, mannitol permeability increased and transepithelial electrical resistance (TEER) decreased. Arachnoid cell growth rate also significantly decreased. When arachnoid cells were grown in close proximity (i.e. on the same monolayer) with fibroblasts, the arachnoid cells were overrun by day 2, yet when physically separated, no significant change was seen in growth. Apoptosis increased markedly in arachnoid cultures in the presence of fibroblast. Fibroblast caused arachnoid cell to exhibit avoidance behavior, and irradiated fibroblast induced arachnoidal cells to move faster and exhibited greater directional changes. Subcellular glycosaminoglycan (GAG) content was significantly altered by fibroblast. INTERPRETATION Fibroblasts influence arachnoid cell's mannitol transport likely via soluble factors. While the arachnoid cells did not change morphologically, cell growth was influenced. Over time, the cells had profound changes in transport and motility. The immortalized arachnoid cell/fibroblast culture system provides a unique model mimicking the pathologic event of leptomeningeal scarring.
Collapse
|
11
|
Krencik R, Hokanson KC, Narayan AR, Dvornik J, Rooney GE, Rauen KA, Weiss LA, Rowitch DH, Ullian EM. Dysregulation of astrocyte extracellular signaling in Costello syndrome. Sci Transl Med 2016; 7:286ra66. [PMID: 25947161 DOI: 10.1126/scitranslmed.aaa5645] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Astrocytes produce an assortment of signals that promote neuronal maturation according to a precise developmental timeline. Is this orchestrated timing and signaling altered in human neurodevelopmental disorders? To address this question, the astroglial lineage was investigated in two model systems of a developmental disorder with intellectual disability caused by mutant Harvey rat sarcoma viral oncogene homolog (HRAS) termed Costello syndrome: mutant HRAS human induced pluripotent stem cells (iPSCs) and transgenic mice. Human iPSCs derived from patients with Costello syndrome differentiated to astroglia more rapidly in vitro than those derived from wild-type cell lines with normal HRAS, exhibited hyperplasia, and also generated an abundance of extracellular matrix remodeling factors and proteoglycans. Acute treatment with a farnesyl transferase inhibitor and knockdown of the transcription factor SNAI2 reduced expression of several proteoglycans in Costello syndrome iPSC-derived astrocytes. Similarly, mice in which mutant HRAS was expressed selectively in astrocytes exhibited experience-independent increased accumulation of perineuronal net proteoglycans in cortex, as well as increased parvalbumin expression in interneurons, when compared to wild-type mice. Our data indicate that astrocytes expressing mutant HRAS dysregulate cortical maturation during development as shown by abnormal extracellular matrix remodeling and implicate excessive astrocyte-to-neuron signaling as a possible drug target for treating mental impairment and enhancing neuroplasticity.
Collapse
Affiliation(s)
- Robert Krencik
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Kenton C Hokanson
- Neuroscience Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Aditi R Narayan
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jill Dvornik
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Gemma E Rooney
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Katherine A Rauen
- Department of Pediatrics, University of California, Davis, Sacramento, CA 95817, USA
| | - Lauren A Weiss
- Department of Psychiatry and Institute for Human Genetics, University of California, San Francisco, San Francisco, CA 94143, USA
| | - David H Rowitch
- Department of Pediatrics, Eli and Edythe Broad Institute for Regenerative Medicine and Stem Cell Research, and Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Erik M Ullian
- Department of Ophthalmology, University of California, San Francisco, San Francisco, CA 94143, USA. Department of Physiology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
12
|
Hyaluronan Synthesis, Catabolism, and Signaling in Neurodegenerative Diseases. Int J Cell Biol 2015; 2015:368584. [PMID: 26448752 PMCID: PMC4581574 DOI: 10.1155/2015/368584] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Accepted: 04/11/2015] [Indexed: 11/18/2022] Open
Abstract
The glycosaminoglycan hyaluronan (HA), a component of the extracellular matrix, has been implicated in regulating neural differentiation, survival, proliferation, migration, and cell signaling in the mammalian central nervous system (CNS). HA is found throughout the CNS as a constituent of proteoglycans, especially within perineuronal nets that have been implicated in regulating neuronal activity. HA is also found in the white matter where it is diffusely distributed around astrocytes and oligodendrocytes. Insults to the CNS lead to long-term elevation of HA within damaged tissues, which is linked at least in part to increased transcription of HA synthases. HA accumulation is often accompanied by elevated expression of at least some transmembrane HA receptors including CD44. Hyaluronidases that digest high molecular weight HA into smaller fragments are also elevated following CNS insults and can generate HA digestion products that have unique biological activities. A number of studies, for example, suggest that both the removal of high molecular weight HA and the accumulation of hyaluronidase-generated HA digestion products can impact CNS injuries through mechanisms that include the regulation of progenitor cell differentiation and proliferation. These studies, reviewed here, suggest that targeting HA synthesis, catabolism, and signaling are all potential strategies to promote CNS repair.
Collapse
|
13
|
Benarroch EE. Brain-derived neurotrophic factor: Regulation, effects, and potential clinical relevance. Neurology 2015; 85:1417-27. [PMID: 25817841 DOI: 10.1212/wnl.0000000000002044] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
14
|
Abstract
The biochemical and biophysical properties of the extracellular matrix (ECM) dictate tissue-specific cell behaviour. The molecules that are associated with the ECM of each tissue, including collagens, proteoglycans, laminins and fibronectin, and the manner in which they are assembled determine the structure and the organization of the resultant ECM. The product is a specific ECM signature that is comprised of unique compositional and topographical features that both reflect and facilitate the functional requirements of the tissue.
Collapse
Affiliation(s)
- Janna K Mouw
- Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco
| | - Guanqing Ou
- 1] Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco. [2] University of California San Francisco and University of California Berkeley Joint Graduate Group in Bioengineering, San Francisco, California 94143, USA
| | - Valerie M Weaver
- 1] Center for Bioengineering and Tissue Regeneration, Department of Surgery, University of California, San Francisco. [2] Department of Anatomy, University of California, San Francisco. [3] Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco. [4] Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco. [5] UCSF Helen Diller Comprehensive Cancer Center, University of California, San Francisco, California 94143, USA
| |
Collapse
|
15
|
Extracellular matrix assembly: a multiscale deconstruction. Nat Rev Mol Cell Biol 2014. [PMID: 25370693 DOI: 10.1038/nrm3902 10.1038/nrm3902] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The biochemical and biophysical properties of the extracellular matrix (ECM) dictate tissue-specific cell behaviour. The molecules that are associated with the ECM of each tissue, including collagens, proteoglycans, laminins and fibronectin, and the manner in which they are assembled determine the structure and the organization of the resultant ECM. The product is a specific ECM signature that is comprised of unique compositional and topographical features that both reflect and facilitate the functional requirements of the tissue.
Collapse
|
16
|
Abstract
The biochemical and biophysical properties of the extracellular matrix (ECM) dictate tissue-specific cell behaviour. The molecules that are associated with the ECM of each tissue, including collagens, proteoglycans, laminins and fibronectin, and the manner in which they are assembled determine the structure and the organization of the resultant ECM. The product is a specific ECM signature that is comprised of unique compositional and topographical features that both reflect and facilitate the functional requirements of the tissue.
Collapse
|