351
|
Murray TE, Richards CM, Robert-Gostlin VN, Bernath AK, Lindhout IA, Klegeris A. Potential neurotoxic activity of diverse molecules released by astrocytes. Brain Res Bull 2022; 189:80-101. [PMID: 35988785 DOI: 10.1016/j.brainresbull.2022.08.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 07/04/2022] [Accepted: 08/14/2022] [Indexed: 11/02/2022]
Abstract
Astrocytes are the main support cells of the central nervous system. They also participate in neuroimmune reactions. In response to pathological and immune stimuli, astrocytes transform to reactive states characterized by increased release of inflammatory mediators. Some of these molecules are neuroprotective and inflammation resolving while others, including reactive oxygen species (ROS), nitric oxide (NO), matrix metalloproteinase (MMP)- 9, L-glutamate, and tumor necrosis factor α (TNF), are well-established toxins known to cause damage to surrounding cells and tissues. We hypothesized that similar to microglia, the brain immune cells, reactive astrocytes can release a broader set of diverse molecules that are potentially neurotoxic. A literature search was conducted to identify such molecules using the following two criteria: 1) evidence of their expression and secretion by astrocytes and 2) direct neurotoxic action. This review describes 14 structurally diverse molecules as less-established astrocyte neurotoxins, including C-X-C motif chemokine ligand (CXCL)10, CXCL12/CXCL12(5-67), FS-7-associated surface antigen ligand (FasL), macrophage inflammatory protein (MIP)- 2α, TNF-related apoptosis inducing ligand (TRAIL), pro-nerve growth factor (proNGF), pro-brain-derived neurotrophic factor (proBDNF), chondroitin sulfate proteoglycans (CSPGs), cathepsin (Cat)B, group IIA secretory phospholipase A2 (sPLA2-IIA), amyloid beta peptides (Aβ), high mobility group box (HMGB)1, ceramides, and lipocalin (LCN)2. For some of these molecules, further studies are required to establish either their direct neurotoxic effects or the full spectrum of stimuli that induce their release by astrocytes. Only limited studies with human-derived astrocytes and neurons are available for most of these potential neurotoxins, which is a knowledge gap that should be addressed in the future. We also summarize available evidence of the role these molecules play in select neuropathologies where reactive astrocytes are a key feature. A comprehensive understanding of the full spectrum of neurotoxins released by reactive astrocytes is key to understanding neuroinflammatory diseases characterized by the adverse activation of these cells and may guide the development of novel treatment strategies.
Collapse
Affiliation(s)
- Taryn E Murray
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada
| | - Christy M Richards
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada
| | - Victoria N Robert-Gostlin
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada
| | - Anna K Bernath
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada
| | - Ivan A Lindhout
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada
| | - Andis Klegeris
- Department of Biology, University of British Columbia Okanagan Campus, Kelowna, British Columbia V1V 1V7, Canada.
| |
Collapse
|
352
|
Wide bandgap semiconductor nanomembranes as a long-term biointerface for flexible, implanted neuromodulator. Proc Natl Acad Sci U S A 2022; 119:e2203287119. [PMID: 35939711 PMCID: PMC9388084 DOI: 10.1073/pnas.2203287119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Electrical neuron stimulation holds promise for treating chronic neurological disorders, including spinal cord injury, epilepsy, and Parkinson's disease. The implementation of ultrathin, flexible electrodes that can offer noninvasive attachment to soft neural tissues is a breakthrough for timely, continuous, programable, and spatial stimulations. With strict flexibility requirements in neural implanted stimulations, the use of conventional thick and bulky packages is no longer applicable, posing major technical issues such as short device lifetime and long-term stability. We introduce herein a concept of long-lived flexible neural electrodes using silicon carbide (SiC) nanomembranes as a faradic interface and thermal oxide thin films as an electrical barrier layer. The SiC nanomembranes were developed using a chemical vapor deposition (CVD) process at the wafer level, and thermal oxide was grown using a high-quality wet oxidation technique. The proposed material developments are highly scalable and compatible with MEMS technologies, facilitating the mass production of long-lived implanted bioelectrodes. Our experimental results showed excellent stability of the SiC/silicon dioxide (SiO2) bioelectronic system that can potentially last for several decades with well-maintained electronic properties in biofluid environments. We demonstrated the capability of the proposed material system for peripheral nerve stimulation in an animal model, showing muscle contraction responses comparable to those of a standard non-implanted nerve stimulation device. The design concept, scalable fabrication approach, and multimodal functionalities of SiC/SiO2 flexible electronics offer an exciting possibility for fundamental neuroscience studies, as well as for neural stimulation-based therapies.
Collapse
|
353
|
Li H, He B, Zhang X, Hao H, Yang T, Sun C, Song H, Wang Y, Zhou Y, Zhu Z, Hu Y, Wang Y. D-dopachrome tautomerase drives astroglial inflammation via NF-κB signaling following spinal cord injury. Cell Biosci 2022; 12:128. [PMID: 35965310 PMCID: PMC9375920 DOI: 10.1186/s13578-022-00867-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 07/30/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Reactive astrocytes are increasingly recognized as crucial regulators of innate immunity in degenerative or damaged central nervous system (CNS). Many proinflammatory mediators have been shown to drive inflammatory cascades of astrocytes through activation of NF-κB, thereby affecting the functional outcome of the insulted CNS. D-dopachrome tautomerase (D-DT), a newly described cytokine and a close homolog of proinflammatory macrophage migration inhibitory factor (MIF), has been revealed to share receptor and overlapping functional spectrum with MIF, but little is known about its roles in the neuropathological progression of the CNS and relevant regulatory mechanisms.
Results
D-DT protein levels were significantly elevated within neurons and astrocytes following SCI. Analysis of transcriptome profile revealed that D-DT was able to activate multiple signal pathways of astrocytes, which converged to NF-κB, a hub regulator governing proinflammatory response. Rat D-DT recombinant protein was efficient in inducing the production of inflammatory cytokines from astrocytes through interaction with CD74 receptor. Activation of mitogen-activated protein kinases (MAPKs) and NF-κB was observed to be essential for the transduction of D-DT signaling. Administration of D-DT specific inhibitor at lesion sites of the cord resulted in significant attenuation of NF-κB activation and reduction of the inflammatory cytokines following SCI, and accordingly improved the recovery of locomotor functions.
Conclusion
Collectively, D-DT is a novel proinflammatory mediator of astrocytes following SCI. Insights of its cell-specific expression and relevant proinflammatory mechanisms will provide clues for the control of CNS inflammation.
Collapse
|
354
|
Lichterfeld Y, Kalinski L, Schunk S, Schmakeit T, Feles S, Frett T, Herrmann H, Hemmersbach R, Liemersdorf C. Hypergravity Attenuates Reactivity in Primary Murine Astrocytes. Biomedicines 2022; 10:biomedicines10081966. [PMID: 36009513 PMCID: PMC9405820 DOI: 10.3390/biomedicines10081966] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/31/2022] [Accepted: 08/11/2022] [Indexed: 12/11/2022] Open
Abstract
Neuronal activity is the key modulator of nearly every aspect of behavior, affecting cognition, learning, and memory as well as motion. Hence, disturbances of the transmission of synaptic signals are the main cause of many neurological disorders. Lesions to nervous tissues are associated with phenotypic changes mediated by astrocytes becoming reactive. Reactive astrocytes form the basis of astrogliosis and glial scar formation. Astrocyte reactivity is often targeted to inhibit axon dystrophy and thus promote neuronal regeneration. Here, we aim to understand the impact of gravitational loading induced by hypergravity to potentially modify key features of astrocyte reactivity. We exposed primary murine astrocytes as a model system closely resembling the in vivo reactivity phenotype on custom-built centrifuges for cultivation as well as for live-cell imaging under hypergravity conditions in a physiological range (2g and 10g). We revealed spreading rates, migration velocities, and stellation to be diminished under 2g hypergravity. In contrast, proliferation and apoptosis rates were not affected. In particular, hypergravity attenuated reactivity induction. We observed cytoskeletal remodeling of actin filaments and microtubules under hypergravity. Hence, the reorganization of these key elements of cell structure demonstrates that fundamental mechanisms on shape and mobility of astrocytes are affected due to altered gravity conditions. In future experiments, potential target molecules for pharmacological interventions that attenuate astrocytic reactivity will be investigated. The ultimate goal is to enhance neuronal regeneration for novel therapeutic approaches.
Collapse
Affiliation(s)
- Yannick Lichterfeld
- Department of Gravitational Biology, Institute of Aerospace Medicine, German Aerospace Center, 51147 Cologne, Germany
| | - Laura Kalinski
- Department of Gravitational Biology, Institute of Aerospace Medicine, German Aerospace Center, 51147 Cologne, Germany
| | - Sarah Schunk
- Department of Gravitational Biology, Institute of Aerospace Medicine, German Aerospace Center, 51147 Cologne, Germany
| | - Theresa Schmakeit
- Department of Gravitational Biology, Institute of Aerospace Medicine, German Aerospace Center, 51147 Cologne, Germany
| | - Sebastian Feles
- Department of Gravitational Biology, Institute of Aerospace Medicine, German Aerospace Center, 51147 Cologne, Germany
| | - Timo Frett
- Department of Muscle and Bone Metabolism, Institute of Aerospace Medicine, German Aerospace Center, 51147 Cologne, Germany
| | - Harald Herrmann
- Institute of Neuropathology, University of Erlangen, 91054 Erlangen, Germany
| | - Ruth Hemmersbach
- Department of Gravitational Biology, Institute of Aerospace Medicine, German Aerospace Center, 51147 Cologne, Germany
| | - Christian Liemersdorf
- Department of Gravitational Biology, Institute of Aerospace Medicine, German Aerospace Center, 51147 Cologne, Germany
- Correspondence: ; Tel.: +49-176-811-09-333
| |
Collapse
|
355
|
Liu T, Zhu W, Zhang X, He C, Liu X, Xin Q, Chen K, Wang H. Recent Advances in Cell and Functional Biomaterial Treatment for Spinal Cord Injury. BIOMED RESEARCH INTERNATIONAL 2022; 2022:5079153. [PMID: 35978649 PMCID: PMC9377911 DOI: 10.1155/2022/5079153] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/17/2022] [Accepted: 07/25/2022] [Indexed: 12/17/2022]
Abstract
Spinal cord injury (SCI) is a devastating central nervous system disease caused by accidental events, resulting in loss of sensory and motor function. Considering the multiple effects of primary and secondary injuries after spinal cord injury, including oxidative stress, tissue apoptosis, inflammatory response, and neuronal autophagy, it is crucial to understand the underlying pathophysiological mechanisms, local microenvironment changes, and neural tissue functional recovery for preparing novel treatment strategies. Treatment based on cell transplantation has become the forefront of spinal cord injury therapy. The transplanted cells provide physical and nutritional support for the damaged tissue. At the same time, the implantation of biomaterials with specific biological functions at the site of the SCI has also been proved to improve the local inhibitory microenvironment and promote axonal regeneration, etc. The combined transplantation of cells and functional biomaterials for SCI treatment can result in greater neuroprotective and regenerative effects by regulating cell differentiation, enhancing cell survival, and providing physical and directional support for axon regeneration and neural circuit remodeling. This article reviews the pathophysiology of the spinal cord, changes in the microenvironment after injury, and the mechanisms and strategies for spinal cord regeneration and repair. The article will focus on summarizing and discussing the latest intervention models based on cell and functional biomaterial transplantation and the latest progress in combinational therapies in SCI repair. Finally, we propose the future prospects and challenges of current treatment regimens for SCI repair, to provide references for scientists and clinicians to seek better SCI repair strategies in the future.
Collapse
Affiliation(s)
- Tianyi Liu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Wenhao Zhu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Xiaoyu Zhang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Chuan He
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Xiaolong Liu
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Qiang Xin
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| | - Kexin Chen
- Institute of Translational Medicine, First Hospital of Jilin University, Changchun 130021, China
| | - Haifeng Wang
- Department of Neurosurgery, First Hospital of Jilin University, Changchun 130021, China
| |
Collapse
|
356
|
Feng Y, Peng Y, Jie J, Yang Y, Yang P. The immune microenvironment and tissue engineering strategies for spinal cord regeneration. Front Cell Neurosci 2022; 16:969002. [PMID: 35990891 PMCID: PMC9385973 DOI: 10.3389/fncel.2022.969002] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Regeneration of neural tissue is limited following spinal cord injury (SCI). Successful regeneration of injured nerves requires the intrinsic regenerative capability of the neurons and a suitable microenvironment. However, the local microenvironment is damaged, including insufficient intraneural vascularization, prolonged immune responses, overactive immune responses, dysregulated bioenergetic metabolism and terminated bioelectrical conduction. Among them, the immune microenvironment formed by immune cells and cytokines plays a dual role in inflammation and regeneration. Few studies have focused on the role of the immune microenvironment in spinal cord regeneration. Here, we summarize those findings involving various immune cells (neutrophils, monocytes, microglia and T lymphocytes) after SCI. The pathological changes that occur in the local microenvironment and the function of immune cells are described. We also summarize and discuss the current strategies for treating SCI with tissue-engineered biomaterials from the perspective of the immune microenvironment.
Collapse
Affiliation(s)
- Yuan Feng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yong Peng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Jing Jie
- Department of Clinical Laboratory, The First People’s Hospital of Nantong, The Second Affiliated Hospital of Nantong University, Nantong, China
- Jing Jie,
| | - Yumin Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Yumin Yang,
| | - Pengxiang Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
- Institute of Cancer Prevention and Treatment, Heilongjiang Academy of Medical Science, Harbin Medical University, Harbin, China
- *Correspondence: Pengxiang Yang,
| |
Collapse
|
357
|
Lee K, Choi JO, Hwang A, Bae HW, Kim CY. Ciliary Neurotrophic Factor Derived From Astrocytes Protects Retinal Ganglion Cells Through PI3K/AKT, JAK/STAT, and MAPK/ERK Pathways. Invest Ophthalmol Vis Sci 2022; 63:4. [PMID: 35925584 PMCID: PMC9363680 DOI: 10.1167/iovs.63.9.4] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 07/11/2022] [Indexed: 11/24/2022] Open
Abstract
Purpose The purpose of this study was to investigate the roles of ciliary neurotrophic factor (CNTF) on the protective effects of astrocytes on retinal ganglion cells (RGCs). Methods Primary RGCs were isolated from neonatal rats. Oxidative stress was induced, and the effects of co-culture with astrocytes and CNTF treatment on RGCs were evaluated. The pathways commonly altered by astrocytes and CNTF were investigated. Effects of each pathway were investigated using pathway inhibitors against PI3K/AKT, JAK/STAT, and MAPK/ERK. RNA sequencing was performed to identify the genes upregulated and downregulated by CNTF treatment. Results Astrocytes improved the viability and increased β3-tubulin expression in RGCs. The concentration of CNTF increased in the RGC-astrocyte co-culture medium. The protective effects of astrocytes were abolished by neutralization with the anti-CNTF antibody; thus, CNTF may play an important role in the effects mediated by astrocytes. Furthermore, CNTF treatment alone enhanced the viability and β3-tubulin expression of RGCs and increased the population of viable RGCs under oxidative stress. The PI3K/AKT pathway was associated with both RGC viability and β3-tubulin expression. However, the JAK/STAT pathway increased the viability of RGCs, whereas the MAPK/ERK pathway was associated with β3-tubulin expression. RNA sequencing revealed the CNTF-upregulated genes associated with response to DNA damage and downregulated genes associated with photoreceptor cell differentiation. Conclusions Our data revealed protective effects of astrocyte-derived CNTF on RGCs. In addition, we showed that multiple pathways exert these protective effects and identified the novel genes involved. These results may be helpful in developing treatments for RGC injury.
Collapse
Affiliation(s)
- Kwanghyun Lee
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Ophthalmology, National Health Insurance Service Ilsan Hospital, Goyang, Gyeonggi-do, Republic of Korea
| | - Jin-Ok Choi
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Ahreum Hwang
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hyoung Won Bae
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Chan Yun Kim
- Institute of Vision Research, Department of Ophthalmology, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
358
|
Zhang Q, Li D, Zhao H, Zhang X. Decitabine attenuates ischemic stroke by reducing astrocytes proliferation in rats. PLoS One 2022; 17:e0272482. [PMID: 35917376 PMCID: PMC9345475 DOI: 10.1371/journal.pone.0272482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 07/14/2022] [Indexed: 11/18/2022] Open
Abstract
DNA methylation regulates epigenetic gene expression in ischemic stroke. Decitabine attenuates ischemic stroke by inhibiting DNA methylation. However, the underlying mechanism of this effect is not known. A model of ischemic stroke in Sprague-Dawley rats was induced through middle cerebral artery occlusion followed by reperfusion step. The rats were randomly treated with decitabine or vehicle by a one-time intraperitoneal injection. Sham rats received similar treatments. Four days after treatment, the rats were perfused with saline or 4% paraformaldehyde after which the brain was excised. DNA methylation level and brain infarct volume were determined by dot blot and histochemistry, respectively. The cellular co-localization and quantitative analysis of DNA methylation were assessed by immunohistochemistry and expression levels of cdkn1b (p27) mRNA and protein were measured by qRT-PCR and immunohistochemistry, respectively. The proliferation of astrocytes and number of neurons were determined by immunohistochemistry. Rats treated with decitabine showed hypomethylation and reduced infarct volume in the cortex. DNA methylation was decreased in astrocytes. Decitabine upregulated p27 mRNA and protein expression levels and attenuated the proliferation of astrocytes in vivo and vitro. Decitabine promotes p27 gene expression possibly by inhibiting its DNA methylation, thereby decreases the proliferation of astrocytes, neuronal death and infarct volume after ischemic stroke.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Dan Li
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Haihua Zhao
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Xu Zhang
- Department of Human Anatomy, College of Basic Medical Sciences, China Medical University, Shenyang, China
- * E-mail:
| |
Collapse
|
359
|
Corrigendum: Purinergic signaling systems across comparative models of spinal cord injury. Neural Regen Res 2022; 18:689-696. [PMID: 36018196 PMCID: PMC9727416 DOI: 10.4103/1673-5374.350234] [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] [Indexed: 12/15/2022] Open
Abstract
[This corrects the article DOI: 10.4103/1673-5374.338993].
Collapse
|
360
|
Xie J, Li J, Ma J, Li M, Wang X, Fu X, Ma Y, Yang H, Li B, Saijilafu. Magnesium Oxide/Poly(l-lactide-co-ε-caprolactone) Scaffolds Loaded with Neural Morphogens Promote Spinal Cord Repair through Targeting the Calcium Influx and Neuronal Differentiation of Neural Stem Cells. Adv Healthc Mater 2022; 11:e2200386. [PMID: 35587044 PMCID: PMC11469078 DOI: 10.1002/adhm.202200386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/22/2022] [Indexed: 11/08/2022]
Abstract
Because of the limited regenerative ability of the central nervous system (CNS), effective treatments for spinal cord injury (SCI) are still lacking. After SCI, neuron loss and axon regeneration failure often result in irreversible functional impairment. The calcium overload induced by the N-methyl-D-aspartate receptor (NMDAR) overactivation is critical for cell death in SCI. It has been reported that the magnesium ion (Mg2+ ) can competitively block the NMDAR and reduce the calcium influx, and that sonic hedgehog (Shh) and retinoic acid (RA) are the critical regulators of neuronal differentiation of endogenous neural stem cells (NSCs). Here, magnesium oxide (MgO)/poly (l-lactide-co-ε-caprolactone) (PLCL) scaffold loaded with purmorphamine (PUR, a Shh signaling agonist) and RA is developed and its feasibility in SCI repair is tested. The results showed that the Mg2+ released from MgO attenuated cell apoptosis by blocking the calcium influx, and the PUR/RA promoted the recruitment and neuronal differentiation of endogenous NSCs, thereby reducing the glial scar formation at the SCI lesion site. Furthermore, implantation of PUR/RA-loaded MgO/PLCL scaffold facilitates the partial recovery of a locomotor function of SCI mouse in vivo. Together, findings from this study imply that PUR/RA-loaded MgO/PLCL scaffold may be a promising biomaterial for the clinical treatment of SCI.
Collapse
Affiliation(s)
- Jile Xie
- Department of OrthopaedicsThe First Affiliated Hospital of Soochow University899 Pinghai RoadSuzhouJiangsu215006China
| | - Jiaying Li
- Orthopaedic InstituteMedical CollegeSoochow University1 Shizi RoadSuzhouJiangsu215006China
| | - Jinjin Ma
- Orthopaedic InstituteMedical CollegeSoochow University1 Shizi RoadSuzhouJiangsu215006China
| | - Meimei Li
- Orthopaedic InstituteMedical CollegeSoochow University1 Shizi RoadSuzhouJiangsu215006China
| | - Xingran Wang
- Orthopaedic InstituteMedical CollegeSoochow University1 Shizi RoadSuzhouJiangsu215006China
| | - Xinya Fu
- Orthopaedic InstituteMedical CollegeSoochow University1 Shizi RoadSuzhouJiangsu215006China
| | - Yanxia Ma
- Orthopaedic InstituteMedical CollegeSoochow University1 Shizi RoadSuzhouJiangsu215006China
| | - Huilin Yang
- Department of OrthopaedicsThe First Affiliated Hospital of Soochow University899 Pinghai RoadSuzhouJiangsu215006China
- Orthopaedic InstituteMedical CollegeSoochow University1 Shizi RoadSuzhouJiangsu215006China
| | - Bin Li
- Department of OrthopaedicsThe First Affiliated Hospital of Soochow University899 Pinghai RoadSuzhouJiangsu215006China
- Orthopaedic InstituteMedical CollegeSoochow University1 Shizi RoadSuzhouJiangsu215006China
| | - Saijilafu
- Department of OrthopaedicsThe First Affiliated Hospital of Soochow University899 Pinghai RoadSuzhouJiangsu215006China
- Orthopaedic InstituteMedical CollegeSoochow University1 Shizi RoadSuzhouJiangsu215006China
| |
Collapse
|
361
|
Hou J, Bi H, Ge Q, Teng H, Wan G, Yu B, Jiang Q, Gu X. Heterogeneity analysis of astrocytes following spinal cord injury at single-cell resolution. FASEB J 2022; 36:e22442. [PMID: 35816276 DOI: 10.1096/fj.202200463r] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 06/21/2022] [Accepted: 06/24/2022] [Indexed: 12/11/2022]
Abstract
Astrocytes play many important functions in response to spinal cord injury (SCI) in an activated manner, including clearance of necrotic tissue, formation of protective barrier, maintenance of microenvironment balance, interaction with immune cells, and formation of the glial scar. More and more studies have shown that the astrocytes are heterogeneous, such as inflammatory astrocyte 1 (A1) and neuroprotective astrocyte 2 (A2) types. However, the subtypes of astrocyte resulting from SCI have not been clearly defined. In this study, using single-cell RNA sequencing, we constructed the transcriptomic profile of astrocytes from uninjured spinal cord tissue and injured tissue nearby the lesion epicenter at 0.5, 1, 3, 7, 14, 60, and 90 days after mouse hemisection spinal cord surgery. Our analysis uncovered six transcriptionally distinct astrocyte states, including Atp1b2+ , S100a4+ , Gpr84+ , C3+ /G0s2+ , GFAP+ /Tm4sf1+ , and Gss+ /Cryab+ astrocytes. We used these new signatures combined with canonical astrocyte markers to determine the distribution of morphologically and physiologically distinct astrocyte population at injured sites by immunofluorescence staining. Then we identified the dynamic evolution process of each astrocyte subtype following SCI. Finally, we also revealed the evolution of highly expressed genes in these astrocyte subtypes at different phases of SCI. Together, we provided six astrocyte subtypes at single-cell resolution following SCI. These data not only contribute to understand the heterogeneity of astrocytes during SCI but also help to find new astrocyte subtypes as a target for SCI repair.
Collapse
Affiliation(s)
- Jinxing Hou
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People's Republic of China.,Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, People's Republic of China
| | - Huiru Bi
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People's Republic of China.,Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, People's Republic of China
| | - Qiting Ge
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People's Republic of China.,Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, People's Republic of China
| | - Huajian Teng
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People's Republic of China.,Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, People's Republic of China
| | - Guoqiang Wan
- State Key Laboratory of Pharmaceutical Biotechnology, MOE Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Nanjing University, Nanjing, People's Republic of China
| | - Bin Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, People's Republic of China
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, People's Republic of China.,Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, Nanjing, People's Republic of China
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, People's Republic of China
| |
Collapse
|
362
|
Cornelison C, Fadel S. Clickable Biomaterials for Modulating Neuroinflammation. Int J Mol Sci 2022; 23:8496. [PMID: 35955631 PMCID: PMC9369181 DOI: 10.3390/ijms23158496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/27/2022] [Accepted: 07/27/2022] [Indexed: 02/04/2023] Open
Abstract
Crosstalk between the nervous and immune systems in the context of trauma or disease can lead to a state of neuroinflammation or excessive recruitment and activation of peripheral and central immune cells. Neuroinflammation is an underlying and contributing factor to myriad neuropathologies including neurodegenerative diseases like Alzheimer's disease and Parkinson's disease; autoimmune diseases like multiple sclerosis; peripheral and central nervous system infections; and ischemic and traumatic neural injuries. Therapeutic modulation of immune cell function is an emerging strategy to quell neuroinflammation and promote tissue homeostasis and/or repair. One such branch of 'immunomodulation' leverages the versatility of biomaterials to regulate immune cell phenotypes through direct cell-material interactions or targeted release of therapeutic payloads. In this regard, a growing trend in biomaterial science is the functionalization of materials using chemistries that do not interfere with biological processes, so-called 'click' or bioorthogonal reactions. Bioorthogonal chemistries such as Michael-type additions, thiol-ene reactions, and Diels-Alder reactions are highly specific and can be used in the presence of live cells for material crosslinking, decoration, protein or cell targeting, and spatiotemporal modification. Hence, click-based biomaterials can be highly bioactive and instruct a variety of cellular functions, even within the context of neuroinflammation. This manuscript will review recent advances in the application of click-based biomaterials for treating neuroinflammation and promoting neural tissue repair.
Collapse
Affiliation(s)
- Chase Cornelison
- Department of Biomedical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA;
| | | |
Collapse
|
363
|
Jin Z, Tian L, Zhang Y, Zhang X, Kang J, Dong H, Huang N, Pan L, Ning B. Apigenin inhibits fibrous scar formation after acute spinal cord injury through TGFβ/SMADs signaling pathway. CNS Neurosci Ther 2022; 28:1883-1894. [PMID: 35906830 PMCID: PMC9532920 DOI: 10.1111/cns.13929] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 07/07/2022] [Accepted: 07/15/2022] [Indexed: 11/30/2022] Open
Abstract
AIM To investigate the effect of apigenin on fibrous scar formation after mouse spinal cord injury (SCI). METHODS The pneumatic impactor strike method was used to establish an SCI model. Mice were intraperitoneally injected with 5 mg/kg or 20 mg/kg apigenin daily for 28 days after SCI. The Basso Mouse Scale (BMS) score, hematoxylin-eosin staining, and immunohistochemical staining were used to assess the effect of apigenin on scar formation and motor function recovery. Western blotting and qRT-PCR were used to detect the expression of fibrosis-related parameters in spinal cord tissue homogenates. NIH-3 T3 cells and mouse primary spinal cord fibroblasts, α-Smooth muscle actin (α-SMA), collagen 1, and fibronectin were used to evaluate apigenin's effect in vitro. Western blotting and immunofluorescence techniques were used to study the effect of apigenin on TGFβ/SMADs signaling. RESULTS Apigenin inhibited fibrous scar formation in the mouse spinal cord and promoted the recovery of motor function. It reduced the expression of fibroblast-related parameters and increased the content of nerve growth factor in vivo, decreasing myofibroblast activation and collagen fiber formation by inhibiting TGFβ-induced SMAD2/3 phosphorylation and nuclear translocation in vitro. CONCLUSION Apigenin inhibits fibrous scar formation after SCI by decreasing fibrosis-related factor expression through TGFβ/SMADs signaling.
Collapse
Affiliation(s)
- Zhengxin Jin
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Lige Tian
- Tianjin Medical University, Tianjin, China
| | - Ying Zhang
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Xiaodi Zhang
- School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Jianning Kang
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Hui Dong
- Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Nana Huang
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Liuzhu Pan
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China
| | - Bin Ning
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, China.,Jinan Central Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
364
|
Cheng Y, Yin Y, Zhang A, Bernstein AM, Kawaguchi R, Gao K, Potter K, Gilbert HY, Ao Y, Ou J, Fricano-Kugler CJ, Goldberg JL, He Z, Woolf CJ, Sofroniew MV, Benowitz LI, Geschwind DH. Transcription factor network analysis identifies REST/NRSF as an intrinsic regulator of CNS regeneration in mice. Nat Commun 2022; 13:4418. [PMID: 35906210 PMCID: PMC9338053 DOI: 10.1038/s41467-022-31960-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 07/08/2022] [Indexed: 01/30/2023] Open
Abstract
The inability of neurons to regenerate long axons within the CNS is a major impediment to improving outcome after spinal cord injury, stroke, and other CNS insults. Recent advances have uncovered an intrinsic program that involves coordinate regulation by multiple transcription factors that can be manipulated to enhance growth in the peripheral nervous system. Here, we use a systems genomics approach to characterize regulatory relationships of regeneration-associated transcription factors, identifying RE1-Silencing Transcription Factor (REST; Neuron-Restrictive Silencer Factor, NRSF) as a predicted upstream suppressor of a pro-regenerative gene program associated with axon regeneration in the CNS. We validate our predictions using multiple paradigms, showing that mature mice bearing cell type-specific deletions of REST or expressing dominant-negative mutant REST show improved regeneration of the corticospinal tract and optic nerve after spinal cord injury and optic nerve crush, which is accompanied by upregulation of regeneration-associated genes in cortical motor neurons and retinal ganglion cells, respectively. These analyses identify a role for REST as an upstream suppressor of the intrinsic regenerative program in the CNS and demonstrate the utility of a systems biology approach involving integrative genomics and bio-informatics to prioritize hypotheses relevant to CNS repair.
Collapse
Affiliation(s)
- Yuyan Cheng
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Yuqin Yin
- Department of Neurosurgery, Boston Children's Hospital, Boston, MA, 02115, USA
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Neurosurgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Alice Zhang
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Alexander M Bernstein
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Riki Kawaguchi
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Psychiatry, Semel Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Kun Gao
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Kyra Potter
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Hui-Ya Gilbert
- Department of Neurosurgery, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Yan Ao
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jing Ou
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Catherine J Fricano-Kugler
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jeffrey L Goldberg
- Byers Eye Institute and Wu Tsai Neuroscience Institute, Stanford University, Palo Alto, CA, 94305, USA
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Neurology, Harvard Medical School, Boston, MA, 02115, USA
| | - Clifford J Woolf
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA
- Department of Neurology, Harvard Medical School, Boston, MA, 02115, USA
| | - Michael V Sofroniew
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Larry I Benowitz
- Department of Neurosurgery, Boston Children's Hospital, Boston, MA, 02115, USA.
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA.
- Department of Neurosurgery, Harvard Medical School, Boston, MA, 02115, USA.
- Department of Ophthalmology, Harvard Medical School, Boston, MA, 02115, USA.
| | - Daniel H Geschwind
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Psychiatry, Semel Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
365
|
Perelroizen R, Philosof B, Budick-Harmelin N, Chernobylsky T, Ron A, Katzir R, Shimon D, Tessler A, Adir O, Gaoni-Yogev A, Meyer T, Krivitsky A, Shidlovsky N, Madi A, Ruppin E, Mayo L. Astrocyte immunometabolic regulation of the tumour microenvironment drives glioblastoma pathogenicity. Brain 2022; 145:3288-3307. [PMID: 35899587 DOI: 10.1093/brain/awac222] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 05/18/2022] [Accepted: 06/09/2022] [Indexed: 12/12/2022] Open
Abstract
Malignant brain tumours are the cause of a disproportionate level of morbidity and mortality among cancer patients, an unfortunate statistic that has remained constant for decades. Despite considerable advances in the molecular characterization of these tumours, targeting the cancer cells has yet to produce significant advances in treatment. An alternative strategy is to target cells in the glioblastoma microenvironment, such as tumour-associated astrocytes. Astrocytes control multiple processes in health and disease, ranging from maintaining the brain's metabolic homeostasis, to modulating neuroinflammation. However, their role in glioblastoma pathogenicity is not well understood. Here we report that depletion of reactive astrocytes regresses glioblastoma and prolongs mouse survival. Analysis of the tumour-associated astrocyte translatome revealed astrocytes initiate transcriptional programmes that shape the immune and metabolic compartments in the glioma microenvironment. Specifically, their expression of CCL2 and CSF1 governs the recruitment of tumour-associated macrophages and promotes a pro-tumourigenic macrophage phenotype. Concomitantly, we demonstrate that astrocyte-derived cholesterol is key to glioma cell survival, and that targeting astrocytic cholesterol efflux, via ABCA1, halts tumour progression. In summary, astrocytes control glioblastoma pathogenicity by reprogramming the immunological properties of the tumour microenvironment and supporting the non-oncogenic metabolic dependency of glioblastoma on cholesterol. These findings suggest that targeting astrocyte immunometabolic signalling may be useful in treating this uniformly lethal brain tumour.
Collapse
Affiliation(s)
- Rita Perelroizen
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Bar Philosof
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Noga Budick-Harmelin
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Tom Chernobylsky
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ariel Ron
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Rotem Katzir
- Cancer Data Science Laboratory (CDSL), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Dor Shimon
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Adi Tessler
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Orit Adir
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Anat Gaoni-Yogev
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Tom Meyer
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Avivit Krivitsky
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Nuphar Shidlovsky
- Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Asaf Madi
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Eytan Ruppin
- Cancer Data Science Laboratory (CDSL), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, USA
| | - Lior Mayo
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.,Shmunis School of Biomedicine and Cancer Research, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
366
|
Qin Q, Wang T, Xu Z, Liu S, Zhang H, Du Z, Wang J, Wang Y, Wang Z, Yuan S, Wu J, He W, Wang C, Yan X, Wang Y, Jiang X. Ectoderm-derived frontal bone mesenchymal stem cells promote traumatic brain injury recovery by alleviating neuroinflammation and glutamate excitotoxicity partially via FGF1. Stem Cell Res Ther 2022; 13:341. [PMID: 35883153 PMCID: PMC9327213 DOI: 10.1186/s13287-022-03032-6] [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: 12/22/2021] [Accepted: 07/04/2022] [Indexed: 11/16/2022] Open
Abstract
Background Traumatic brain injury (TBI) leads to cell and tissue impairment, as well as functional deficits. Stem cells promote structural and functional recovery and thus are considered as a promising therapy for various nerve injuries. Here, we aimed to investigate the role of ectoderm-derived frontal bone mesenchymal stem cells (FbMSCs) in promoting cerebral repair and functional recovery in a murine TBI model. Methods A murine TBI model was established by injuring C57BL/6 N mice with moderate-controlled cortical impact to evaluate the extent of brain damage and behavioral deficits. Ectoderm-derived FbMSCs were isolated from the frontal bone and their characteristics were assessed using multiple differentiation assays, flow cytometry and microarray analysis. Brain repairment and functional recovery were analyzed at different days post-injury with or without FbMSC application. Behavioral tests were performed to assess learning and memory improvements. RNA sequencing analysis, immunofluorescence staining, and quantitative reverse-transcription polymerase chain reaction (qRT-PCR) were used to examine inflammation reaction and neural regeneration. In vitro co-culture analysis and quantification of glutamate transportation were carried out to explore the possible mechanism of neurogenesis and functional recovery promoted by FbMSCs. Results Ectoderm-derived FbMSCs showed fibroblast like morphology and osteogenic differentiation capacity. FbMSCs were CD105, CD29 positive and CD45, CD31 negative. Different from mesoderm-derived MSCs, FbMSCs expressed the ectoderm-specific transcription factor Tfap2β. TBI mice showed impaired learning and memory deficits. Microglia and astrocyte activation, as well as neural damage, were significantly increased post-injury. FbMSC application ameliorated the behavioral deficits of TBI mice and promoted neural regeneration. RNA sequencing analysis showed that signal pathways related to inflammation decreased, whereas those related to neural activation increased. Immunofluorescence staining and qRT-PCR data revealed that microglial activation and astrocyte polarization to the A1 phenotype were suppressed by FbMSC application. In addition, FGF1 secreted from FbMSCs enhanced glutamate transportation by astrocytes and alleviated the cytotoxic effect of excessive glutamate on neurons. Conclusions Ectoderm-derived FbMSC application significantly alleviated neuroinflammation, brain injury, and excitatory toxicity to neurons, improved cognition and behavioral deficits in TBI mice. Therefore, ectoderm-derived FbMSCs could be ideal therapeutic candidates for TBI which mostly affect cells from the same embryonic origins as FbMSCs. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-022-03032-6.
Collapse
Affiliation(s)
- Qiaozhen Qin
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China.,Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing, 100124, People's Republic of China
| | - Ting Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Zhenhua Xu
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Shuirong Liu
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Heyang Zhang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Zhangzhen Du
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Jianing Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Yadi Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Zhenning Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Shanshan Yuan
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Jiamei Wu
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Wenyan He
- China National Clinical Research Center for Neurological Diseases, Jing-Jin Center for Neuroinflammation, Beijing Tiantan Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Changzhen Wang
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China
| | - Xinlong Yan
- Faculty of Environmental and Life Sciences, Beijing University of Technology, Beijing, 100124, People's Republic of China.
| | - Yan Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China. .,Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China.
| | - Xiaoxia Jiang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Road, Haidian District, Beijing, 100850, People's Republic of China. .,Anhui Medical University, Hefei, 230032, Anhui, People's Republic of China.
| |
Collapse
|
367
|
Activation of NLRP3 Is Required for a Functional and Beneficial Microglia Response after Brain Trauma. Pharmaceutics 2022; 14:pharmaceutics14081550. [PMID: 35893807 PMCID: PMC9332196 DOI: 10.3390/pharmaceutics14081550] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 07/20/2022] [Accepted: 07/22/2022] [Indexed: 12/04/2022] Open
Abstract
Despite the numerous research studies on traumatic brain injury (TBI), many physiopathologic mechanisms remain unknown. TBI is a complex process, in which neuroinflammation and glial cells play an important role in exerting a functional immune and damage-repair response. The activation of the NLRP3 inflammasome is one of the first steps to initiate neuroinflammation and so its regulation is essential. Using a closed-head injury model and a pharmacological (MCC950; 3 mg/kg, pre- and post-injury) and genetical approach (NLRP3 knockout (KO) mice), we defined the transcriptional and behavioral profiles 24 h after TBI. Wild-type (WT) mice showed a strong pro-inflammatory response, with increased expression of inflammasome components, microglia and astrocytes markers, and cytokines. There was no difference in the IL1β production between WT and KO, nor compensatory mechanisms of other inflammasomes. However, some microglia and astrocyte markers were overexpressed in KO mice, resulting in an exacerbated cytokine expression. Pretreatment with MCC950 replicated the behavioral and blood-brain barrier results observed in KO mice and its administration 1 h after the lesion improved the damage. These findings highlight the importance of NLRP3 time-dependent activation and its role in the fine regulation of glial response.
Collapse
|
368
|
Nguyen TK, Yadav S, Truong TA, Han M, Barton M, Leitch M, Guzman P, Dinh T, Ashok A, Vu H, Dau V, Haasmann D, Chen L, Park Y, Do TN, Yamauchi Y, Rogers JA, Nguyen NT, Phan HP. Integrated, Transparent Silicon Carbide Electronics and Sensors for Radio Frequency Biomedical Therapy. ACS NANO 2022; 16:10890-10903. [PMID: 35816450 PMCID: PMC9332346 DOI: 10.1021/acsnano.2c03188] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
The integration of micro- and nanoelectronics into or onto biomedical devices can facilitate advanced diagnostics and treatments of digestive disorders, cardiovascular diseases, and cancers. Recent developments in gastrointestinal endoscopy and balloon catheter technologies introduce promising paths for minimally invasive surgeries to treat these diseases. However, current therapeutic endoscopy systems fail to meet requirements in multifunctionality, biocompatibility, and safety, particularly when integrated with bioelectronic devices. Here, we report materials, device designs, and assembly schemes for transparent and stable cubic silicon carbide (3C-SiC)-based bioelectronic systems that facilitate tissue ablation, with the capability for integration onto the tips of endoscopes. The excellent optical transparency of SiC-on-glass (SoG) allows for direct observation of areas of interest, with superior electronic functionalities that enable multiple biological sensing and stimulation capabilities to assist in electrical-based ablation procedures. Experimental studies on phantom, vegetable, and animal tissues demonstrated relatively short treatment times and low electric field required for effective lesion removal using our SoG bioelectronic system. In vivo experiments on an animal model were conducted to explore the versatility of SoG electrodes for peripheral nerve stimulation, showing an exciting possibility for the therapy of neural disorders through electrical excitation. The multifunctional features of SoG integrated devices indicate their high potential for minimally invasive, cost-effective, and outcome-enhanced surgical tools, across a wide range of biomedical applications.
Collapse
Affiliation(s)
- Tuan-Khoa Nguyen
- Queensland
Micro and Nanotechnology Centre, Griffith
University, Brisbane, Queensland 4111, Australia
| | - Sharda Yadav
- Queensland
Micro and Nanotechnology Centre, Griffith
University, Brisbane, Queensland 4111, Australia
| | - Thanh-An Truong
- Queensland
Micro and Nanotechnology Centre, Griffith
University, Brisbane, Queensland 4111, Australia
- School
of Mechanical and Manufacturing Engineering, The University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Mengdi Han
- Department
of Biomedical Engineering, College of Future Technology, Peking University, Beijing 100871, China
| | - Matthew Barton
- School
of Nursing and Midwifery, Griffith University, Brisbane, Queensland 4111, Australia
- Menzies
Health Institute Queensland, Brisbane, Queensland 4222, Australia
| | - Michael Leitch
- School
of Nursing and Midwifery, Griffith University, Brisbane, Queensland 4111, Australia
| | - Pablo Guzman
- Queensland
Micro and Nanotechnology Centre, Griffith
University, Brisbane, Queensland 4111, Australia
| | - Toan Dinh
- Centre
for Future Materials, University of Southern
Queensland, Toowoomba, Queensland 4305, Australia
| | - Aditya Ashok
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Hieu Vu
- School
of Engineering and Built Environment, Griffith
University, Brisbane, Queensland 4215, Australia
| | - Van Dau
- School
of Engineering and Built Environment, Griffith
University, Brisbane, Queensland 4215, Australia
| | - Daniel Haasmann
- Queensland
Micro and Nanotechnology Centre, Griffith
University, Brisbane, Queensland 4111, Australia
| | - Lin Chen
- State
Key Laboratory for Mechanical Behavior of Materials, School of Materials
Science and Engineering, Xi’an Jiaotong
University, Xi’an 710049, Shaanxi, People’s Republic of China
| | - Yoonseok Park
- Querrey
Simpson Institute for Bioelectronics, Northwestern
University, Evanston, Illinois 60208, United States
- Department
of Advanced Materials Engineering for Information and Electronics, Kyung Hee University, Yongin 17104, Republic
of Korea
| | - Thanh Nho Do
- Graduate
School of Biomedical Engineering, The University
of New South Wales, Sydney, New South Wales 2032, Australia
| | - Yusuke Yamauchi
- Australian
Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Queensland 4072, Australia
- JST-ERATO
Yamauchi Materials Space-Tectonics Project, Kagami Memorial Research
Institute for Science and Technology, Waseda
University, Tokyo 169-0051, Japan
| | - John A. Rogers
- Querrey
Simpson Institute for Bioelectronics, Northwestern
University, Evanston, Illinois 60208, United States
- Department
of Materials Science and Engineering, Department of Mechanical Engineering,
Department of Biomedical Engineering, Departments of Electrical and
Computer Engineering and Chemistry, and Department of Neurological
Surgery, Northwestern University, Evanston, Illinois 60208, United States
| | - Nam-Trung Nguyen
- Queensland
Micro and Nanotechnology Centre, Griffith
University, Brisbane, Queensland 4111, Australia
| | - Hoang-Phuong Phan
- Queensland
Micro and Nanotechnology Centre, Griffith
University, Brisbane, Queensland 4111, Australia
- School
of Mechanical and Manufacturing Engineering, The University of New South Wales, Sydney, New South Wales 2052, Australia
| |
Collapse
|
369
|
Colitti N, Desmoulin F, Le Friec A, Labriji W, Robert L, Michaux A, Conchou F, Cirillo C, Loubinoux I. Long-Term Intranasal Nerve Growth Factor Treatment Favors Neuron Formation in de novo Brain Tissue. Front Cell Neurosci 2022; 16:871532. [PMID: 35928573 PMCID: PMC9345199 DOI: 10.3389/fncel.2022.871532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 06/08/2022] [Indexed: 11/13/2022] Open
Abstract
Objective To date, no safe and effective pharmacological treatment has been clinically validated for improving post-stroke neurogenesis. Growth factors are good candidates but low safety has limited their application in the clinic. An additional restraint is the delivery route. Intranasal delivery presents many advantages. Materials and Methods A brain lesion was induced in twenty-four rats. Nerve growth factor (NGF) 5 μg/kg/day or vehicle was given intranasally from day 10 post-lesion for two periods of five weeks, separated by a two-week wash out period with no treatment. Lesion volume and atrophy were identified by magnetic resonance imaging (MRI). Anxiety and sensorimotor recovery were measured by behavior tests. Neurogenesis, angiogenesis and inflammation were evaluated by histology at 12 weeks. Results Remarkable neurogenesis occurred and was visible at the second and third months after the insult. Tissue reconstruction was clearly detected by T2 weighted MRI at 8 and 12 weeks post-lesion and confirmed by histology. In the new tissue (8.1% of the lesion in the NGF group vs. 2.4%, in the control group at 12 weeks), NGF significantly increased the percentage of mature neurons (19% vs. 7%). Angiogenesis and inflammation were not different in the two groups. Sensorimotor recovery was neither improved nor hampered by NGF during the first period of treatment, but NGF treatment limited motor recovery in the second period. Interpretation The first five-week period of treatment was very well tolerated. This study is the first presenting the effects of a long treatment with NGF and has shown an important tissue regeneration rate at 8 and 12 weeks post-injury. NGF may have increased neuronal differentiation and survival and favored neurogenesis and neuron survival through subventricular zone (SVZ) neurogenesis or reprogramming of reactive astrocytes. For the first time, we evidenced a MRI biomarker of neurogenesis and tissue reconstruction with T2 and diffusion weighted imaging.
Collapse
Affiliation(s)
- Nina Colitti
- Toulouse NeuroImaging Center (ToNIC), Inserm, University of Toulouse (UPS), Toulouse, France
| | - Franck Desmoulin
- Toulouse NeuroImaging Center (ToNIC), Inserm, University of Toulouse (UPS), Toulouse, France
| | - Alice Le Friec
- Toulouse NeuroImaging Center (ToNIC), Inserm, University of Toulouse (UPS), Toulouse, France
| | - Wafae Labriji
- Toulouse NeuroImaging Center (ToNIC), Inserm, University of Toulouse (UPS), Toulouse, France
| | - Lorenne Robert
- Toulouse NeuroImaging Center (ToNIC), Inserm, University of Toulouse (UPS), Toulouse, France
| | - Amandine Michaux
- Unit of Medical Imaging, National Veterinary School of Toulouse, University of Toulouse, Toulouse, France
| | - Fabrice Conchou
- Unit of Medical Imaging, National Veterinary School of Toulouse, University of Toulouse, Toulouse, France
| | - Carla Cirillo
- Toulouse NeuroImaging Center (ToNIC), Inserm, University of Toulouse (UPS), Toulouse, France
| | - Isabelle Loubinoux
- Toulouse NeuroImaging Center (ToNIC), Inserm, University of Toulouse (UPS), Toulouse, France
- *Correspondence: Isabelle Loubinoux,
| |
Collapse
|
370
|
He W, Zhang X, Li X, Ju D, Mao T, Lu Y, Gu Y, Qi L, Wang Q, Wu Q, Dong C. A decellularized spinal cord extracellular matrix-gel/GelMA hydrogel three-dimensional composite scaffold promotes recovery from spinal cord injury via synergism with human menstrual blood-derived stem cells. J Mater Chem B 2022; 10:5753-5764. [PMID: 35838078 DOI: 10.1039/d2tb00792d] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Spinal cord injury (SCI), as a serious disabling disease, is still haunted by lacking of effective treatments. We previously found that transplantation of menstrual blood-derived mesenchymal stem cells (MenSCs) promoted axon regeneration in rats with SCI, while the abominable microenvironment after the SCI inhibited the survival of stem cells after transplantation. Biomaterials can support the activity of stem cells and accelerate the functional reconstruction of the injured spinal cord. In this study, we constructed a novel composite scaffold consisting of the decellularized spinal cord extracellular matrix-gel (DSCG) and the GelMA hydrogel, which harbored high water retention, wettability, degradability and soft mechanical property. In vitro, the DSCG/GelMA composite scaffold provided a dual bionic microenvironment with optimized bioactive components and favorable microstructures for the adhesion, proliferation and differentiation of MenSCs. After that, we prepared MenSC-encapsulated DSCG/GelMA composite scaffolds to bridge the 2 mm gap in rats with completely transected SCI. The in vivo results showed that the combined use of the DSCG/GelMA composite scaffold with MenSCs improved the motor function, reduced the inflammatory response, promoted neuronal differentiation, and inhibited the proliferation of reactive astrocytes after spinal cord injury. Altogether, our study provided a promising novel therapeutic option of using bioactive materials synergistic with stem cells for the treatment of SCI.
Collapse
Affiliation(s)
- Wenhua He
- Department of Anatomy, Comparative Medicine Institution, Medical School of Nantong University, Nantong 226001, Jiangsu Province, China.
| | - Xuanxuan Zhang
- Department of Anatomy, Comparative Medicine Institution, Medical School of Nantong University, Nantong 226001, Jiangsu Province, China.
| | - Xiangzhe Li
- Department of Rehabilitation Medicine, Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou Science & Technology Town Hospital, Suzhou, 215153, Jiangsu Province, China.
| | - Dingyue Ju
- Department of Anatomy, Comparative Medicine Institution, Medical School of Nantong University, Nantong 226001, Jiangsu Province, China.
| | - Tiantian Mao
- Department of Anatomy, Comparative Medicine Institution, Medical School of Nantong University, Nantong 226001, Jiangsu Province, China.
| | - Yan Lu
- Department of Anatomy, Comparative Medicine Institution, Medical School of Nantong University, Nantong 226001, Jiangsu Province, China.
| | - Yu Gu
- Department of Anatomy, Comparative Medicine Institution, Medical School of Nantong University, Nantong 226001, Jiangsu Province, China.
| | - Longju Qi
- Department of Hepatic Intervention, Affiliated Nantong Hospital 3 of Nantong University, Nantong 226001, Jiangsu Province, China
| | - Qinghua Wang
- Department of Anatomy, Comparative Medicine Institution, Medical School of Nantong University, Nantong 226001, Jiangsu Province, China. .,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu Province, China.
| | - Qinfeng Wu
- Department of Rehabilitation Medicine, Suzhou Hospital Affiliated to Nanjing Medical University, Suzhou Science & Technology Town Hospital, Suzhou, 215153, Jiangsu Province, China.
| | - Chuanming Dong
- Department of Anatomy, Comparative Medicine Institution, Medical School of Nantong University, Nantong 226001, Jiangsu Province, China. .,Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu Province, China.
| |
Collapse
|
371
|
Ferroptosis: A Novel Therapeutic Direction of Spinal Cord Injury. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:7906218. [PMID: 35866036 PMCID: PMC9296343 DOI: 10.1155/2022/7906218] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 06/25/2022] [Indexed: 02/07/2023]
Abstract
An injury to the spinal cord results in a crucial central nervous system event that further causes irreversible impairment or loss of motor, autonomic, and sensory functions. A progressive pathophysiological cascade following spinal cord injury (SCI) includes ischemia/reperfusion injury, oxidative stress, proapoptotic signaling, peripheral inflammatory cell infiltration, and glutamate-mediated excitotoxicity, and regulated cell death. These complex pathological and physiological changes continue to cause cell injury over the long-term and severely limit the efficacy of clinical treatment strategies in restoring the injured nervous system. Ferroptosis is a nonapoptotic, iron-regulated kind of cell death that has recently been discovered. It is distinguished by iron overload-induced toxic lipid peroxidation associated with mitochondrial morphological changes during the cell death process. For example, after SCI, iron overload activates the reactive oxygen species generation, dysregulation of glutathione/glutathione peroxidase 4 (GSH/GPX4) metabolism, and accumulation of lipid peroxides, which cause lipid membrane deterioration and ferroptosis. Conversely, knockout or differential expression of key genes and application of lipid peroxidation inhibitors and iron chelators (e.g., deferoxamine) (e.g., SRS-16-86) can block ferroptosis and promote neuronal repair for functional recovery after SCI. Although the findings of numerous investigations have been confirmed the importance of ferroptosis in several human neurologic sicknesses and its potential in SCI, the mechanism of ferroptosis and its application in SCI has not been elucidated. This review highlights current ferroptosis research and its impact on SCI, as well as the key molecular mechanism of ferroptosis in promoting the recovery from SCI. Understanding ferroptosis' process and function in SCI could provide useful insight into the treatment and avoidance of such a destructive injury.
Collapse
|
372
|
Critical Role of Neuronal Vps35 in Blood Vessel Branching and Maturation in Developing Mouse Brain. Biomedicines 2022; 10:biomedicines10071653. [PMID: 35884959 PMCID: PMC9313219 DOI: 10.3390/biomedicines10071653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 11/17/2022] Open
Abstract
Vps35 (vacuolar protein sorting 35), a key component of retromer, plays a crucial role in selective retrieval of transmembrane proteins from endosomes to trans-Golgi networks. Dysfunctional Vps35/retromer is a risk factor for the development of neurodegenerative diseases. Vps35 is highly expressed in developing pyramidal neurons, both in the mouse neocortex and hippocampus, Although embryonic neuronal Vps35’s function in promoting neuronal terminal differentiation and survival is evident, it remains unclear whether and how neuronal Vps35 communicates with other types of brain cells, such as blood vessels (BVs), which are essential for supplying nutrients to neurons. Dysfunctional BVs contribute to the pathogenesis of various neurodegenerative disorders. Here, we provide evidence for embryonic neuronal Vps35 as critical for BV branching and maturation in the developing mouse brain. Selectively knocking out (KO) Vps35 in mouse embryonic, not postnatal, neurons results in reductions in BV branching and density, arteriole diameter, and BV-associated pericytes and microglia but an increase in BV-associated reactive astrocytes. Deletion of microglia by PLX3397 enhances these BV deficits in mutant mice. These results reveal the function of neuronal Vps35 in neurovascular coupling in the developing mouse brain and implicate BV-associated microglia as underlying this event.
Collapse
|
373
|
Zhou Y, Guo S, Botchway BOA, Zhang Y, Jin T, Liu X. Muscone Can Improve Spinal Cord Injury by Activating the Angiogenin/Plexin-B2 Axis. Mol Neurobiol 2022; 59:5891-5901. [PMID: 35809154 DOI: 10.1007/s12035-022-02948-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 06/23/2022] [Indexed: 12/01/2022]
Abstract
Spinal cord injury (SCI) is a devastating neurological disorder that usually damages sensorimotor and autonomic functions. Signaling pathways can play a key role in the repair process of SCI. The plexin-B2 acts as a receptor for angiogenin and mediates ribosomal RNA transcription, influencing cell survival and proliferation. Protein kinase B serine/threonine kinase interacts with angiogenin to form a positive feedback effect. Brain-derived neurotrophic factor (BDNF) and vascular endothelial growth factor can induce angiogenin nuclear translocation. Moreover, the BDNF can promote the secretion of angiogenin. Interestingly, all of them can activate the angiogenin/plexin-B2 axis. Muscone has anti-inflammatory and proliferative features as it can inhibit nuclear transcription factor kappa-B (NF-κB) and activate the angiogenin/plexin-B2 axis, thus being significant agent in the SCI repair process. Herein, we review the potential mechanism of angiogenin/plexin-B2 axis activation and the role of muscone in SCI treatment. Muscone may attenuate inflammatory responses and promote neuronal regeneration after SCI.
Collapse
Affiliation(s)
- Yu Zhou
- Department of Histology and Embryology, School of Medicine, Medical College, Shaoxing University, Zhejiang Province, Shaoxing, 312000, China
| | - Shitian Guo
- School of Clinical Medicine, Hangzhou Normal University, Hangzhou, China
| | - Benson O A Botchway
- Institute of Neuroscience, Zhejiang University School of Medicine, Hangzhou, China
| | - Yong Zhang
- Department of Histology and Embryology, School of Medicine, Medical College, Shaoxing University, Zhejiang Province, Shaoxing, 312000, China
| | - Tian Jin
- Department of Histology and Embryology, School of Medicine, Medical College, Shaoxing University, Zhejiang Province, Shaoxing, 312000, China
| | - Xuehong Liu
- Department of Histology and Embryology, School of Medicine, Medical College, Shaoxing University, Zhejiang Province, Shaoxing, 312000, China.
| |
Collapse
|
374
|
Bacteria reduce flagellin synthesis to evade microglia-astrocyte-driven immunity in the brain. Cell Rep 2022; 40:111033. [PMID: 35793624 DOI: 10.1016/j.celrep.2022.111033] [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] [Received: 06/15/2021] [Revised: 05/09/2022] [Accepted: 06/13/2022] [Indexed: 11/23/2022] Open
Abstract
The immune response of brain cells to invading bacteria in vivo and the mechanism used by pathogenic bacteria to escape brain immune surveillance remain largely unknown. It is believed that microglia eliminate bacteria by phagocytosis based on in vitro data. Here we find that a small percentage of microglia in the brain engulf neonatal meningitis-causing Escherichia coli (NMEC), but more microglia are activated to produce tumor necrosis factor alpha (TNFα), which activates astrocytes to secrete complement component 3 (C3) involved in anti-bacterial activity. To evade anti-bacterial activity of the immune system, NMEC senses low concentration of threonine in cerebrospinal fluid (CSF) to down-modulate the expression of flagellin and reduce microglial TNFα and astrocyte C3 production. Our findings may help develop strategies for bacterial meningitis treatment.
Collapse
|
375
|
Gong L, Gu Y, Han X, Luan C, Liu C, Wang X, Sun Y, Zheng M, Fang M, Yang S, Xu L, Sun H, Yu B, Gu X, Zhou S. Spatiotemporal Dynamics of the Molecular Expression Pattern and Intercellular Interactions in the Glial Scar Response to Spinal Cord Injury. Neurosci Bull 2022; 39:213-244. [PMID: 35788904 PMCID: PMC9905408 DOI: 10.1007/s12264-022-00897-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 04/28/2022] [Indexed: 12/22/2022] Open
Abstract
Nerve regeneration in adult mammalian spinal cord is poor because of the lack of intrinsic regeneration of neurons and extrinsic factors - the glial scar is triggered by injury and inhibits or promotes regeneration. Recent technological advances in spatial transcriptomics (ST) provide a unique opportunity to decipher most genes systematically throughout scar formation, which remains poorly understood. Here, we first constructed the tissue-wide gene expression patterns of mouse spinal cords over the course of scar formation using ST after spinal cord injury from 32 samples. Locally, we profiled gene expression gradients from the leading edge to the core of the scar areas to further understand the scar microenvironment, such as neurotransmitter disorders, activation of the pro-inflammatory response, neurotoxic saturated lipids, angiogenesis, obstructed axon extension, and extracellular structure re-organization. In addition, we described 21 cell transcriptional states during scar formation and delineated the origins, functional diversity, and possible trajectories of subpopulations of fibroblasts, glia, and immune cells. Specifically, we found some regulators in special cell types, such as Thbs1 and Col1a2 in macrophages, CD36 and Postn in fibroblasts, Plxnb2 and Nxpe3 in microglia, Clu in astrocytes, and CD74 in oligodendrocytes. Furthermore, salvianolic acid B, a blood-brain barrier permeation and CD36 inhibitor, was administered after surgery and found to remedy fibrosis. Subsequently, we described the extent of the scar boundary and profiled the bidirectional ligand-receptor interactions at the neighboring cluster boundary, contributing to maintain scar architecture during gliosis and fibrosis, and found that GPR37L1_PSAP, and GPR37_PSAP were the most significant gene-pairs among microglia, fibroblasts, and astrocytes. Last, we quantified the fraction of scar-resident cells and proposed four possible phases of scar formation: macrophage infiltration, proliferation and differentiation of scar-resident cells, scar emergence, and scar stationary. Together, these profiles delineated the spatial heterogeneity of the scar, confirmed the previous concepts about scar architecture, provided some new clues for scar formation, and served as a valuable resource for the treatment of central nervous system injury.
Collapse
Affiliation(s)
- Leilei Gong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Yun Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Xiaoxiao Han
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Chengcheng Luan
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Chang Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Xinghui Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Yufeng Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Mengru Zheng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Mengya Fang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Shuhai Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Lai Xu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Hualin Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Bin Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| | - Songlin Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| |
Collapse
|
376
|
Chio JCT, Punjani N, Hejrati N, Zavvarian MM, Hong J, Fehlings MG. Extracellular Matrix and Oxidative Stress Following Traumatic Spinal Cord Injury: Physiological and Pathophysiological Roles and Opportunities for Therapeutic Intervention. Antioxid Redox Signal 2022; 37:184-207. [PMID: 34465134 DOI: 10.1089/ars.2021.0120] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significance: Traumatic spinal cord injury (SCI) causes significant disruption to neuronal, glial, vascular, and extracellular elements. The spinal cord extracellular matrix (ECM) comprises structural and communication proteins that are involved in reparative and regenerative processes after SCI. In the healthy spinal cord, the ECM helps maintain spinal cord homeostasis. After SCI, the damaged ECM limits plasticity and contributes to inflammation through the expression of damage-associated molecules such as proteoglycans. Recent Advances: Considerable insights have been gained by characterizing the origins of the gliotic and fibrotic scars, which not only reduce the spread of injury but also limit neuroregeneration. These properties likely limit the success of therapies used to treat patients with SCI. The ECM, which is a major contributor to the scars and normal physiological functions of the spinal cord, represents an exciting therapeutic target to enhance recovery post-SCI. Critical Issue: Various ECM-based preclinical therapies have been developed. These include disrupting scar components, inhibiting activity of ECM metalloproteinases, and maintaining iron homeostasis. Biomaterials have also been explored. However, the majority of these treatments have not experienced successful clinical translation. This could be due to the ECM and scars' polarizing roles. Future Directions: This review surveys the complexity involved in spinal ECM modifications, discusses new ECM-based combinatorial strategies, and explores the biomaterials evaluated in clinical trials, which hope to introduce new treatments that enhance recovery after SCI. These topics will incorporate oxidative species, which are both beneficial and harmful in reparative and regenerative processes after SCI, and not often assessed in pertinent literature. Antioxid. Redox Signal. 37, 184-207.
Collapse
Affiliation(s)
- Jonathon Chon Teng Chio
- Department of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Nayaab Punjani
- Department of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Nader Hejrati
- Department of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, Canada
| | - Mohammad-Masoud Zavvarian
- Department of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada
| | - James Hong
- Department of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, Canada
| | - Michael G Fehlings
- Department of Genetics and Development, Krembil Brain Institute, University Health Network, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada.,Department of Surgery and Spine Program, University of Toronto, Toronto, Canada
| |
Collapse
|
377
|
Zhang G, Jin LQ, Rodemer W, Hu J, Root ZD, Medeiros DM, Selzer ME. The Composition and Cellular Sources of CSPGs in the Glial Scar After Spinal Cord Injury in the Lamprey. Front Mol Neurosci 2022; 15:918871. [PMID: 35832392 PMCID: PMC9271930 DOI: 10.3389/fnmol.2022.918871] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/27/2022] [Indexed: 11/13/2022] Open
Abstract
Axon regrowth after spinal cord injury (SCI) is inhibited by several types of inhibitory extracellular molecules in the central nervous system (CNS), including chondroitin sulfate proteoglycans (CSPGs), which also are components of perineuronal nets (PNNs). The axons of lampreys regenerate following SCI, even though their spinal cords contain CSPGs, and their neurons are enwrapped by PNNs. Previously, we showed that by 2 weeks after spinal cord transection in the lamprey, expression of CSPGs increased in the lesion site, and thereafter, decreased to pre-injury levels by 10 weeks. Enzymatic digestion of CSPGs in the lesion site with chondroitinase ABC (ChABC) enhanced axonal regeneration after SCI and reduced retrograde neuronal death. Lecticans (aggrecan, versican, neurocan, and brevican) are the major CSPG family in the CNS. Previously, we cloned a cDNA fragment that lies in the most conserved link-domain of the lamprey lecticans and found that lectican mRNAs are expressed widely in lamprey glia and neurons. Because of the lack of strict one-to-one orthology with the jawed vertebrate lecticans, the four lamprey lecticans were named simply A, B, C, and D. Using probes that distinguish these four lecticans, we now show that they all are expressed in glia and neurons but at different levels. Expression levels are relatively high in embryonic and early larval stages, gradually decrease, and are upregulated again in adults. Reductions of lecticans B and D are greater than those of A and C. Levels of mRNAs for lecticans B and D increased dramatically after SCI. Lectican D remained upregulated for at least 10 weeks. Multiple cells, including glia, neurons, ependymal cells and microglia/macrophages, expressed lectican mRNAs in the peripheral zone and lesion center after SCI. Thus, as in mammals, lamprey lecticans may be involved in axon guidance and neuroplasticity early in development. Moreover, neurons, glia, ependymal cells, and microglia/macrophages, are responsible for the increase in CSPGs during the formation of the glial scar after SCI.
Collapse
Affiliation(s)
- Guixin Zhang
- Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), Department of Neural Sciences, Philadelphia, PA, United States
| | - Li-Qing Jin
- Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), Department of Neural Sciences, Philadelphia, PA, United States
| | - William Rodemer
- Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), Department of Neural Sciences, Philadelphia, PA, United States
| | - Jianli Hu
- Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), Department of Neural Sciences, Philadelphia, PA, United States
| | - Zachary D. Root
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, United States
| | - Daniel M. Medeiros
- Department of Ecology and Evolutionary Biology, University of Colorado, Boulder, CO, United States
| | - Michael E. Selzer
- Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), Department of Neural Sciences, Philadelphia, PA, United States
- Department of Neurology, The Lewis Katz School of Medicine at Temple University, Philadelphia, PA, United States
- *Correspondence: Michael E. Selzer
| |
Collapse
|
378
|
Xia L, Qi J, Tang M, Liu J, Zhang D, Zhu Y, Hu B. Continual Deletion of Spinal Microglia Reforms Astrocyte Scar Favoring Axonal Regeneration. Front Pharmacol 2022; 13:881195. [PMID: 35833026 PMCID: PMC9271995 DOI: 10.3389/fphar.2022.881195] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 05/19/2022] [Indexed: 11/16/2022] Open
Abstract
Astrocyte scar formation after spinal cord injury (SCI) efficiently limits the accurate damage but physically restricts the following axon regeneration. Lately, fine tuning scar formation is becoming a novel strategy to develop SCI treatment, yet how to leverage these opposite effects remains challenging. Here, utilizing an improved drug administration approach, we show that in a mouse model of spinal cord injury, continual deletion of microglia, especially upon scar formation, by pexidartinib decreases the amount of microglia-derived collagen I and reforms the astrocyte scar. The astrocytes become less compacted in the scar, which permits axon regeneration and extension. Although continual microglia deletion did not significantly improve the locomotive performance of the SCI mice, it did ameliorate their weight loss, possibly by improving their relevant health conditions. We thus identified a novel approach to regulate astrocyte scars for improved axon regeneration, which is indicative of the clinical treatment of SCI patients.
Collapse
Affiliation(s)
- Longkuo Xia
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences (CAS), Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Jianhuan Qi
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences (CAS), Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Mingming Tang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences (CAS), Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Jing Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences (CAS), Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Da Zhang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Yanbing Zhu
- Beijing Clinical Research Institute, Beijing Friendship Hospital, Capital Medical University, Beijing, China
- *Correspondence: Yanbing Zhu, ; Baoyang Hu,
| | - Baoyang Hu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences (CAS), Beijing, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- National Stem Cell Resource Center, Institute of Zoology (CAS), Beijing, China
- *Correspondence: Yanbing Zhu, ; Baoyang Hu,
| |
Collapse
|
379
|
He X, Li Y, Deng B, Lin A, Zhang G, Ma M, Wang Y, Yang Y, Kang X. The PI3K/AKT signalling pathway in inflammation, cell death and glial scar formation after traumatic spinal cord injury: Mechanisms and therapeutic opportunities. Cell Prolif 2022; 55:e13275. [PMID: 35754255 PMCID: PMC9436900 DOI: 10.1111/cpr.13275] [Citation(s) in RCA: 119] [Impact Index Per Article: 39.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 04/17/2022] [Accepted: 05/24/2022] [Indexed: 02/06/2023] Open
Abstract
Objects Traumatic spinal cord injury (TSCI) causes neurological dysfunction below the injured segment of the spinal cord, which significantly impacts the quality of life in affected patients. The phosphoinositide 3kinase/serine‐threonine kinase (PI3K/AKT) signaling pathway offers a potential therapeutic target for the inhibition of secondary TSCI. This review summarizes updates concerning the role of the PI3K/AKT pathway in TSCI. Materials and Methods By searching articles related to the TSCI field and the PI3K/AKT signaling pathway, we summarized the mechanisms of secondary TSCI and the PI3K/AKT signaling pathway; we also discuss current and potential future treatment methods for TSCI based on the PI3K/AKT signaling pathway. Results Early apoptosis and autophagy after TSCI protect the body against injury; a prolonged inflammatory response leads to the accumulation of pro‐inflammatory factors and excessive apoptosis, as well as excessive autophagy in the surrounding normal nerve cells, thus aggravating TSCI in the subacute stage of secondary injury. Initial glial scar formation in the subacute phase is a protective mechanism for TSCI, which limits the spread of damage and inflammation. However, mature scar tissue in the chronic phase hinders axon regeneration and prevents the recovery of nerve function. Activation of PI3K/AKT signaling pathway can inhibit the inflammatory response and apoptosis in the subacute phase after secondary TSCI; inhibiting this pathway in the chronic phase can reduce the formation of glial scar. Conclusion The PI3K/AKT signaling pathway has an important role in the recovery of spinal cord function after secondary injury. Inducing the activation of PI3K/AKT signaling pathway in the subacute phase of secondary injury and inhibiting this pathway in the chronic phase may be one of the potential strategies for the treatment of TSCI.
Collapse
Affiliation(s)
- Xuegang He
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China.,The International Cooperation Base of Gansu Province for the Pain Research in Spinal Disorders, Lanzhou, China
| | - Ying Li
- Medical School of Yan'an University, Yan'an University, Yan'an, China
| | - Bo Deng
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Aixin Lin
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China.,The International Cooperation Base of Gansu Province for the Pain Research in Spinal Disorders, Lanzhou, China
| | - Guangzhi Zhang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China.,The International Cooperation Base of Gansu Province for the Pain Research in Spinal Disorders, Lanzhou, China
| | - Miao Ma
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China
| | - Yonggang Wang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China.,The International Cooperation Base of Gansu Province for the Pain Research in Spinal Disorders, Lanzhou, China
| | - Yong Yang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China.,The International Cooperation Base of Gansu Province for the Pain Research in Spinal Disorders, Lanzhou, China
| | - Xuewen Kang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, China.,The Second Clinical Medical College, Lanzhou University, Lanzhou, China.,The International Cooperation Base of Gansu Province for the Pain Research in Spinal Disorders, Lanzhou, China
| |
Collapse
|
380
|
Liu Q, Zhou S, Wang X, Gu C, Guo Q, Li X, Zhang C, Zhang N, Zhang L, Huang F. Apelin alleviated neuroinflammation and promoted endogenous neural stem cell proliferation and differentiation after spinal cord injury in rats. J Neuroinflammation 2022; 19:160. [PMID: 35725619 PMCID: PMC9208139 DOI: 10.1186/s12974-022-02518-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 06/05/2022] [Indexed: 12/14/2022] Open
Abstract
Background Spinal cord injury (SCI) causes devastating neurological damage, including secondary injuries dominated by neuroinflammation. The role of Apelin, an endogenous ligand that binds the G protein-coupled receptor angiotensin-like receptor 1, in SCI remains unclear. Thus, our aim was to investigate the effects of Apelin in inflammatory responses and activation of endogenous neural stem cells (NSCs) after SCI. Methods Apelin expression was detected in normal and injured rats, and roles of Apelin in primary NSCs were examined. In addition, we used induced pluripotent stem cells (iPSCs) as a carrier to prolong the effective duration of Apelin and evaluate its effects in a rat model of SCI. Results Co-immunofluorescence staining suggested that Apelin was expressed in both astrocytes, neurons and microglia. Following SCI, Apelin expression decreased from 1 to 14 d and re-upregulated at 28 d. In vitro, Apelin promoted NSCs proliferation and differentiation into neurons. In vivo, lentiviral-transfected iPSCs were used as a carrier to prolong the effective duration of Apelin. Transplantation of transfected iPSCs in situ immediately after SCI reduced polarization of M1 microglia and A1 astrocytes, facilitated recovery of motor function, and promoted the proliferation and differentiation of endogenous NSCs in rats. Conclusion Apelin alleviated neuroinflammation and promoted the proliferation and differentiation of endogenous NSCs after SCI, suggesting that it might be a promising target for treatment of SCI.
Collapse
Affiliation(s)
- Qing Liu
- Institute of Neurobiology, Binzhou Medical University, 346 Guanhai Road, Laishan, 264003, Shandong, China
| | - Shuai Zhou
- Institute of Neurobiology, Binzhou Medical University, 346 Guanhai Road, Laishan, 264003, Shandong, China
| | - Xiao Wang
- Institute of Neurobiology, Binzhou Medical University, 346 Guanhai Road, Laishan, 264003, Shandong, China
| | - Chengxu Gu
- Institute of Neurobiology, Binzhou Medical University, 346 Guanhai Road, Laishan, 264003, Shandong, China
| | - Qixuan Guo
- Institute of Neurobiology, Binzhou Medical University, 346 Guanhai Road, Laishan, 264003, Shandong, China
| | - Xikai Li
- Institute of Neurobiology, Binzhou Medical University, 346 Guanhai Road, Laishan, 264003, Shandong, China
| | - Chunlei Zhang
- Institute of Neurobiology, Binzhou Medical University, 346 Guanhai Road, Laishan, 264003, Shandong, China
| | - Naili Zhang
- Institute of Neurobiology, Binzhou Medical University, 346 Guanhai Road, Laishan, 264003, Shandong, China
| | - Luping Zhang
- Institute of Neurobiology, Binzhou Medical University, 346 Guanhai Road, Laishan, 264003, Shandong, China.
| | - Fei Huang
- Institute of Neurobiology, Binzhou Medical University, 346 Guanhai Road, Laishan, 264003, Shandong, China. .,School of Health and Life Sciences, University of Health and Rehabilitation Sciences, 17 Shandong Road, Qingdao, 266071, China.
| |
Collapse
|
381
|
Zhao C, Rao JS, Duan H, Hao P, Shang J, Fan Y, Zhao W, Gao Y, Yang Z, Sun YE, Li X. Chronic spinal cord injury repair by NT3-chitosan only occurs after clearance of the lesion scar. Signal Transduct Target Ther 2022; 7:184. [PMID: 35710784 PMCID: PMC9203793 DOI: 10.1038/s41392-022-01010-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 03/22/2022] [Accepted: 04/06/2022] [Indexed: 11/24/2022] Open
Abstract
Spinal cord injury (SCI) is a severe damage usually leading to limb dysesthesia, motor dysfunction, and other physiological disability. We have previously shown that NT3-chitosan could trigger an acute SCI repairment in rats and non-human primates. Due to the negative effect of inhibitory molecules in glial scar on axonal regeneration, however, the role of NT3-chitosan in the treatment of chronic SCI remains unclear. Compared with the fresh wound of acute SCI, how to handle the lesion core and glial scars is a major issue related to chronic-SCI repair. Here we report, in a chronic complete SCI rat model, establishment of magnetic resonance-diffusion tensor imaging (MR-DTI) methods to monitor spatial and temporal changes of the lesion area, which matched well with anatomical analyses. Clearance of the lesion core via suction of cystic tissues and trimming of solid scar tissues before introducing NT3-chitosan using either a rigid tubular scaffold or a soft gel form led to robust neural regeneration, which interconnected the severed ascending and descending axons and accompanied with electrophysiological and motor functional recovery. In contrast, cystic tissue extraction without scar trimming followed by NT3-chitosan injection, resulted in little, if any regeneration. Taken together, after lesion core clearance, NT3-chitosan can be used to enable chronic-SCI repair and MR-DTI-based mapping of lesion area and monitoring of ongoing regeneration can potentially be implemented in clinical studies for subacute/chronic-SCI repair.
Collapse
Affiliation(s)
- Can Zhao
- Beijing Key Laboratory for Biomaterials and Neural Regeneration, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China.,Institute of Rehabilitation Engineering, China Rehabilitation Science Institute, Beijing, 100068, China
| | - Jia-Sheng Rao
- Beijing Key Laboratory for Biomaterials and Neural Regeneration, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China
| | - Hongmei Duan
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Peng Hao
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Junkui Shang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yubo Fan
- Beijing Advanced Innovation Centre for Biomedical Engineering, Key Laboratory for Biomechanics and Mechanobiology of Chinese Education Ministry, School of Biological Science and Medical Engineering, Beihang University, Beijing, 10083, China.,School of Engineering Medicine, Beihang University, Beijing, 10083, China
| | - Wen Zhao
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Yudan Gao
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China
| | - Zhaoyang Yang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
| | - Yi Eve Sun
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China. .,Department of Psychiatry and Biobehavioral Sciences, UCLA Medical School, Los Angeles, CA, 90095, USA.
| | - Xiaoguang Li
- Beijing Key Laboratory for Biomaterials and Neural Regeneration, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, 100083, China. .,Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
382
|
He T, Yang GY, Zhang Z. Crosstalk of Astrocytes and Other Cells during Ischemic Stroke. LIFE (BASEL, SWITZERLAND) 2022; 12:life12060910. [PMID: 35743941 PMCID: PMC9228674 DOI: 10.3390/life12060910] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 06/04/2022] [Accepted: 06/07/2022] [Indexed: 12/27/2022]
Abstract
Stroke is a leading cause of death and long-term disability worldwide. Astrocytes structurally compose tripartite synapses, blood–brain barrier, and the neurovascular unit and perform multiple functions through cell-to-cell signaling of neurons, glial cells, and vasculature. The crosstalk of astrocytes and other cells is complicated and incompletely understood. Here we review the role of astrocytes in response to ischemic stroke, both beneficial and detrimental, from a cell–cell interaction perspective. Reactive astrocytes provide neuroprotection through antioxidation and antiexcitatory effects and metabolic support; they also contribute to neurorestoration involving neurogenesis, synaptogenesis, angiogenesis, and oligodendrogenesis by crosstalk with stem cells and cell lineage. In the meantime, reactive astrocytes also play a vital role in neuroinflammation and brain edema. Glial scar formation in the chronic phase hinders functional recovery. We further discuss astrocyte enriched microRNAs and exosomes in the regulation of ischemic stroke. In addition, the latest notion of reactive astrocyte subsets and astrocytic activity revealed by optogenetics is mentioned. This review discusses the current understanding of the intimate molecular conversation between astrocytes and other cells and outlines its potential implications after ischemic stroke. “Neurocentric” strategies may not be sufficient for neurological protection and recovery; future therapeutic strategies could target reactive astrocytes.
Collapse
Affiliation(s)
- Tingting He
- Department of Neurology, Shanghai Tenth People’s Hospital, Tongji University, Shanghai 200072, China;
- Neuroscience and Neuroengineering Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Guo-Yuan Yang
- Neuroscience and Neuroengineering Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
- Correspondence: (G.-Y.Y.); (Z.Z.); Tel.: +86-21-62933186 (G.-Y.Y.); Fax: +86-21-62932302 (G.-Y.Y.)
| | - Zhijun Zhang
- Neuroscience and Neuroengineering Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
- Correspondence: (G.-Y.Y.); (Z.Z.); Tel.: +86-21-62933186 (G.-Y.Y.); Fax: +86-21-62932302 (G.-Y.Y.)
| |
Collapse
|
383
|
Yamada J, Maeda S, Soya M, Nishida H, Iinuma KM, Jinno S. Alleviation of cognitive deficits via upregulation of chondroitin sulfate biosynthesis by lignan sesamin in a mouse model of neuroinflammation. J Nutr Biochem 2022; 108:109093. [PMID: 35724814 DOI: 10.1016/j.jnutbio.2022.109093] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 05/09/2022] [Accepted: 05/20/2022] [Indexed: 12/29/2022]
Abstract
Lignans are plant-derived compounds that act as partial estrogen agonists. Chondroitin sulfate proteoglycans (CSPGs) represent one of the major components of the extracellular matrix (ECM). Here we aimed to understand the role of sesamin (SES), a major lignan compound, in the biosynthesis and degradation of CSPGs in the mouse hippocampus because CSPGs play a key role in the regulation of cognitive functions through the promotion of adult neurogenesis. The expression of the pro-inflammatory cytokine interleukin-1β was decreased by SES administration in the hippocampus of lipopolysaccharide (LPS)-treated mice, a model of neuroinflammation-induced cognitive deficits. The expression of genes related to biosynthesis and degradation of CSPGs in the hippocampus of LPS-treated mice was both increased and decreased by SES administration. Further, the diffuse ECM labeling of CSPGs by Wisteria floribunda agglutinin (WFA) in the hippocampus of LPS-treated mice was increased by SES administration. The densities of neural stem cells, late transit-amplifying cells, and newborn-granule cells in the hippocampus of LPS-treated mice were also increased by SES administration. Moreover, SES-induced alterations in gene expression, WFA labeling, and adult neurogenesis in LPS-treated mice were more evident in the dorsal hippocampus (center of cognition) than in the ventral hippocampus (center of emotion). Neither LPS nor SES administration affected locomotor activity, anxiety-like behavior, and depression-related behavior. However, impairments in contextual memory and sensorimotor gating in LPS-treated mice were recovered by SES administration. Our results show that SES can promote adult hippocampal neurogenesis through the upregulation of CSPGs, which may alleviate cognitive deficits induced by neuroinflammation.
Collapse
Affiliation(s)
- Jun Yamada
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shoichiro Maeda
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mariko Soya
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hidefumi Nishida
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kyoko M Iinuma
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shozo Jinno
- Department of Anatomy and Neuroscience, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
384
|
Xu X, Zhang Y. Regulation of Oxidative Stress by Long Non-coding RNAs in Central Nervous System Disorders. Front Mol Neurosci 2022; 15:931704. [PMID: 35782387 PMCID: PMC9241987 DOI: 10.3389/fnmol.2022.931704] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 05/24/2022] [Indexed: 11/13/2022] Open
Abstract
Central nervous system (CNS) disorders, such as ischemic stroke, Alzheimer’s disease, Parkinson’s disease, spinal cord injury, glioma, and epilepsy, involve oxidative stress and neuronal apoptosis, often leading to long-term disability or death. Emerging studies suggest that oxidative stress may induce epigenetic modifications that contribute to CNS disorders. Non-coding RNAs are epigenetic regulators involved in CNS disorders and have attracted extensive attention. Long non-coding RNAs (lncRNAs) are non-coding RNAs more than 200 nucleotides long and have no protein-coding function. However, these molecules exert regulatory functions at the transcriptional, post-transcriptional, and epigenetic levels. However, the major role of lncRNAs in the pathophysiology of CNS disorders, especially related to oxidative stress, remains unclear. Here, we review the molecular functions of lncRNAs in oxidative stress and highlight lncRNAs that exert positive or negative roles in oxidation/antioxidant systems. This review provides novel insights into the therapeutic potential of lncRNAs that mediate oxidative stress in CNS disorders.
Collapse
Affiliation(s)
- Xiaoman Xu
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yi Zhang
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Yi Zhang,
| |
Collapse
|
385
|
Zhang Q, Liu C, Shi R, Zhou S, Shan H, Deng L, Chen T, Guo Y, Zhang Z, Yang GY, Wang Y, Tang Y. Blocking C3d +/GFAP + A1 Astrocyte Conversion with Semaglutide Attenuates Blood-Brain Barrier Disruption in Mice after Ischemic Stroke. Aging Dis 2022; 13:943-959. [PMID: 35656116 PMCID: PMC9116904 DOI: 10.14336/ad.2021.1029] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 10/29/2021] [Indexed: 12/25/2022] Open
Abstract
Astrocytes play an essential role in the modulation of blood-brain barrier function. Neurological diseases induce the transformation of astrocytes into a neurotoxic A1 phenotype, exacerbating brain injury. However, the effect of A1 astrocytes on the BBB dysfunction after stroke is unknown. Adult male ICR mice (n=97) were subjected to 90-minute transient middle cerebral artery occlusion (tMCAO). Immunohistochemical staining of A1 (C3d) and A2 (S100A10) was performed to characterize phenotypic changes in astrocytes over time after tMCAO. The glucagon-like peptide-1 receptor agonist semaglutide was intraperitoneally injected into mice to inhibit A1 astrocytes. Infarct volume, atrophy volume, neurobehavioral outcomes, and BBB permeability were evaluated. RNA-seq was adopted to explore the potential targets and signaling pathways of A1 astrocyte-induced BBB dysfunction. Astrocytic C3d expression was increased, while expression of S100A10 was decreased in the first two weeks after tMCAO, reflecting a shift in the astrocytic phenotype. Semaglutide treatment reduced the expression of CD16/32 in microglia and C3d in astrocytes after ischemic stroke (p<0.05). Ischemia-induced brain infarct volume, atrophy volume and neuroinflammation were reduced in the semaglutide-treated mice, and neurobehavioral outcomes were improved compared to control mice (p<0.05). We further demonstrated that semaglutide treatment reduced the gap formation of tight junction proteins ZO-1, claudin-5 and occludin, as well as IgG leakage three days following tMCAO (p<0.05). In vitro experiments revealed that A1 astrocyte-conditioned medium disrupted BBB integrity. RNA-seq showed that A1 astrocytes were enriched in inflammatory factors and chemokines and significantly modulated the TNF and chemokine signaling pathways, which are closely related to barrier damage. We concluded that astrocytes undergo a phenotypic shift over time after ischemic stroke. C3d+/GFAP+ astrocytes aggravate BBB disruption, suggesting that inhibiting C3d+/GFAP+ astrocyte formation represents a novel strategy for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Qi Zhang
- 1School of Biomedical Engineering and Shanghai 6th People's Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Chang Liu
- 1School of Biomedical Engineering and Shanghai 6th People's Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Rubing Shi
- 1School of Biomedical Engineering and Shanghai 6th People's Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Shiyi Zhou
- 1School of Biomedical Engineering and Shanghai 6th People's Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Huimin Shan
- 1School of Biomedical Engineering and Shanghai 6th People's Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Lidong Deng
- 1School of Biomedical Engineering and Shanghai 6th People's Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Tingting Chen
- 1School of Biomedical Engineering and Shanghai 6th People's Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yiyan Guo
- 1School of Biomedical Engineering and Shanghai 6th People's Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Zhijun Zhang
- 1School of Biomedical Engineering and Shanghai 6th People's Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Guo-Yuan Yang
- 1School of Biomedical Engineering and Shanghai 6th People's Hospital, Shanghai Jiao Tong University, Shanghai 200030, China.,2Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Yongting Wang
- 1School of Biomedical Engineering and Shanghai 6th People's Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yaohui Tang
- 1School of Biomedical Engineering and Shanghai 6th People's Hospital, Shanghai Jiao Tong University, Shanghai 200030, China
| |
Collapse
|
386
|
Xiong XY, Tang Y, Yang QW. Metabolic changes favor the activity and heterogeneity of reactive astrocytes. Trends Endocrinol Metab 2022; 33:390-400. [PMID: 35396164 DOI: 10.1016/j.tem.2022.03.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 02/27/2022] [Accepted: 03/10/2022] [Indexed: 12/20/2022]
Abstract
Reactive astrocytes undergo morphological, molecular, metabolic, and functional remodeling in response to central nervous system (CNS) damage. However, we still know very little about how the metabolic switching of astrocytes influences, or is influenced by, reactive astrocytes in response to neurological diseases. In this review, we initially cover a brief introduction into reactive astrocyte function under pathological conditions. Subsequently, we summarize the emerging roles of glucose and lipid metabolism in reactive astrocytes in the context of CNS injury to provide a new insight into metabolic mechanisms of reactive astrocyte-mediated neuroprotection or damage. Finally, we propose that deciphering the mechanistic link between astrocyte heterogeneity metabolism and improved methods is an emerging frontier for the therapeutic investigation of CNS injury and disease.
Collapse
Affiliation(s)
- Xiao-Yi Xiong
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China; International Collaborative Centre on Big Science Plan for Purinergic Signaling, Chengdu, China; Acupuncture & Chronobiology Key Laboratory of Sichuan Province, Chengdu, China.
| | - Yong Tang
- School of Acupuncture and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China; International Collaborative Centre on Big Science Plan for Purinergic Signaling, Chengdu, China; Acupuncture & Chronobiology Key Laboratory of Sichuan Province, Chengdu, China
| | - Qing-Wu Yang
- Department of Neurology, Xinqiao Hospital, The Army Medical University (Third Military Medical University), Chongqing, China; Chongqing Institute for Brain and Intelligence, Guangyang Bay Laboratory, Chongqing 400064, China.
| |
Collapse
|
387
|
Burda JE, O'Shea TM, Ao Y, Suresh KB, Wang S, Bernstein AM, Chandra A, Deverasetty S, Kawaguchi R, Kim JH, McCallum S, Rogers A, Wahane S, Sofroniew MV. Divergent transcriptional regulation of astrocyte reactivity across disorders. Nature 2022; 606:557-564. [PMID: 35614216 PMCID: PMC10027402 DOI: 10.1038/s41586-022-04739-5] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 04/07/2022] [Indexed: 01/30/2023]
Abstract
Astrocytes respond to injury and disease in the central nervous system with reactive changes that influence the outcome of the disorder1-4. These changes include differentially expressed genes (DEGs) whose contextual diversity and regulation are poorly understood. Here we combined biological and informatic analyses, including RNA sequencing, protein detection, assay for transposase-accessible chromatin with high-throughput sequencing (ATAC-seq) and conditional gene deletion, to predict transcriptional regulators that differentially control more than 12,000 DEGs that are potentially associated with astrocyte reactivity across diverse central nervous system disorders in mice and humans. DEGs associated with astrocyte reactivity exhibited pronounced heterogeneity across disorders. Transcriptional regulators also exhibited disorder-specific differences, but a core group of 61 transcriptional regulators was identified as common across multiple disorders in both species. We show experimentally that DEG diversity is determined by combinatorial, context-specific interactions between transcriptional regulators. Notably, the same reactivity transcriptional regulators can regulate markedly different DEG cohorts in different disorders; changes in the access of transcriptional regulators to DNA-binding motifs differ markedly across disorders; and DEG changes can crucially require multiple reactivity transcriptional regulators. We show that, by modulating reactivity, transcriptional regulators can substantially alter disorder outcome, implicating them as therapeutic targets. We provide searchable resources of disorder-related reactive astrocyte DEGs and their predicted transcriptional regulators. Our findings show that transcriptional changes associated with astrocyte reactivity are highly heterogeneous and are customized from vast numbers of potential DEGs through context-specific combinatorial transcriptional-regulator interactions.
Collapse
Affiliation(s)
- Joshua E Burda
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Center for Neural Science and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| | - Timothy M O'Shea
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Yan Ao
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Keshav B Suresh
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Center for Neural Science and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Shinong Wang
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Alexander M Bernstein
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Ashu Chandra
- Department of Computer Science, University of California Los Angeles, Los Angeles, CA, USA
| | - Sandeep Deverasetty
- Department of Psychiatry, University of California Los Angeles, Los Angeles, CA, USA
- Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA
| | - Riki Kawaguchi
- Department of Psychiatry, University of California Los Angeles, Los Angeles, CA, USA
- Department of Neurology, University of California Los Angeles, Los Angeles, CA, USA
| | - Jae H Kim
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Sarah McCallum
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Center for Neural Science and Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Alexandra Rogers
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Shalaka Wahane
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Michael V Sofroniew
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, CA, USA.
| |
Collapse
|
388
|
Bub A, Brenna S, Alawi M, Kügler P, Gui Y, Kretz O, Altmeppen H, Magnus T, Puig B. Multiplexed mRNA analysis of brain-derived extracellular vesicles upon experimental stroke in mice reveals increased mRNA content with potential relevance to inflammation and recovery processes. Cell Mol Life Sci 2022; 79:329. [PMID: 35639208 PMCID: PMC9156510 DOI: 10.1007/s00018-022-04357-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 04/05/2022] [Accepted: 05/09/2022] [Indexed: 12/14/2022]
Abstract
Extracellular vesicles (EVs) are lipid bilayer-enclosed structures that represent newly discovered means for cell-to-cell communication as well as promising disease biomarkers and therapeutic tools. Apart from proteins, lipids, and metabolites, EVs can deliver genetic information such as mRNA, eliciting a response in the recipient cells. In the present study, we have analyzed the mRNA content of brain-derived EVs (BDEVs) isolated 72 h after experimental stroke in mice and compared them to controls (shams) using nCounter® Nanostring panels, with or without prior RNA isolation. We found that both panels show similar results when comparing upregulated mRNAs in stroke. Notably, the highest upregulated mRNAs were related to processes of stress and immune system responses, but also to anatomical structure development, cell differentiation, and extracellular matrix organization, thus indicating that regenerative mechanisms already take place at this time-point. The five top overrepresented mRNAs in stroke mice were confirmed by RT-qPCR and, interestingly, found to be full-length. We could reveal that the majority of the mRNA cargo in BDEVs was of microglial origin and predominantly present in small BDEVs (≤ 200 nm in diameter). However, the EV population with the highest increase in the total BDEVs pool at 72 h after stroke was of oligodendrocytic origin. Our study shows that nCounter® panels are a good tool to study mRNA content in tissue-derived EVs as they can be carried out even without previous mRNA isolation, and that the mRNA cargo of BDEVs indicates a possible participation in inflammatory but also recovery processes after stroke.
Collapse
Affiliation(s)
- Annika Bub
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Santra Brenna
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Malik Alawi
- Bioinformatics Core, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Paul Kügler
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yuqi Gui
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Oliver Kretz
- III Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hermann Altmeppen
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Magnus
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Berta Puig
- Neurology Department, Experimental Research in Stroke and Inflammation (ERSI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
389
|
Zhou XH, Zhang CC, Wang L, Jin SL. Remimazolam induced cognitive dysfunction in mice via glutamate excitotoxicity. Transl Neurosci 2022; 13:104-115. [PMID: 35734308 PMCID: PMC9164290 DOI: 10.1515/tnsci-2022-0220] [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: 12/08/2021] [Revised: 04/26/2022] [Accepted: 04/26/2022] [Indexed: 11/27/2022] Open
Abstract
Objective Several lines of evidence demonstrated the role of anesthetic drugs in cognitive functions. Some anesthetic agents have been confirmed to be associated with long-term spatial memory and learning in aged animal models. Methods C57BL/6 mice were divided into four different groups based on different concentrations of remimazolam treatments. Behavioral phenotype was observed by open field, rota rod, Morris water maze, and elevated plus maze test. Western blot was performed to see the expression pattern of different proteins. Confocal microscopy images were taken for neuronal and glial cells to see the effect of remimazolam on CNS cells. Results We showed that remimazolam, a new anesthetic drug, impaired cognitive behavior. Repetitive doses of remimazolam have been found to induce neuronal loss with a significant change in morphology. Here, we showed that a higher concentration of remimazolam had a significant effect on CNS cell activation. We showed that remimazolam caused memory dysfunction by inducing neuronal apoptosis via glutamate excitotoxicity. It also exhibited amyloid β plaque in the brain via abnormal phosphorylation of tau protein. Remimazolam-mediated regulation of glial cells in mouse cortex was observed and robust activation of astrocytes and microglial cells was found. Finally, we assessed the behavioral phenotype of mice and found that treatment with remimazolam induced significant behavioral changes and memory dysfunction. Conclusions This study provides insight into the mechanism of anesthetic drug-induced memory deficits and may help improve the therapeutic effects of anesthesia agents in clinical applications.
Collapse
Affiliation(s)
- Xin-hua Zhou
- Department of Anesthesiology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhizaoju Road, Shanghai 201900, China
| | - Cheng-cheng Zhang
- Department of Anesthesiology, Changhai Hospital, The Naval Medical University, Shanghai 200433, China
| | - Ling Wang
- Department of Anesthesiology, Changhai Hospital, The Naval Medical University, Shanghai 200433, China
| | - Shan-liang Jin
- Department of Anesthesiology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, No. 639, Zhizaoju Road, Shanghai 201900, China
| |
Collapse
|
390
|
Sun C, Lin L, Yin L, Hao X, Tian J, Zhang X, Ren Y, Li C, Yang Y. Acutely Inhibiting AQP4 With TGN-020 Improves Functional Outcome by Attenuating Edema and Peri-Infarct Astrogliosis After Cerebral Ischemia. Front Immunol 2022; 13:870029. [PMID: 35592320 PMCID: PMC9110854 DOI: 10.3389/fimmu.2022.870029] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 04/11/2022] [Indexed: 01/05/2023] Open
Abstract
Background Ischemic stroke is one of the leading causes of human death and disability. Brain edema and peri-infarct astrocyte reactivity are crucial pathological changes, both involving aquaporin-4 (AQP4). Studies revealed that acute inhibition of AQP4 after stroke diminishes brain edema, however, its effect on peri-infarct astrocyte reactivity and the subacute outcome is unclear. And if diffusion-weighted imaging (DWI) could reflect the AQP4 expression patterns is uncertain. Methods Rats were subjected to middle cerebral artery occlusion (MCAO) and allocated randomly to TGN 020-treated and control groups. One day after stroke, brain swelling and lesion volumes of the rats were checked using T2-weighted imaging (T2-WI). Fourteen days after stroke, the rats successively underwent neurological examination, T2-WI and DWI with standard b-values and ultra-high b-values, apparent diffusion coefficient (ADC) was calculated correspondingly. Finally, the rats’ brains were acquired and used for glial fibrillary acidic protein (GFAP) and AQP4 immunoreactive analysis. Results At 1 day after stroke, the TGN-020-treated animals exhibited reduced brain swelling and lesion volumes compared with those in the control group. At 14 days after stroke, the TGN-020-treated animals showed fewer neurological function deficits and smaller lesion volumes. In the peri-infarct region, the control group showed evident astrogliosis and AQP4 depolarization, which were reduced significantly in the TGN-020 group. In addition, the ultra-high b-values of ADC (ADCuh) in the peri-infarct region of the TGN-020 group was higher than that of the control group. Furthermore, correlation analysis revealed that peri-infarct AQP4 polarization correlated negatively with astrogliosis extent, and ADCuh correlated positively with AQP4 polarization. Conclusion We found that acutely inhibiting AQP4 using TGN-020 promoted neurological recovery by diminishing brain edema at the early stage and attenuating peri-infarct astrogliosis and AQP4 depolarization at the subacute stage after stroke. Moreover, ADCuh could reflect the AQP4 polarization.
Collapse
Affiliation(s)
- Chengfeng Sun
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Luyi Lin
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Lekang Yin
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiaozhu Hao
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiaqi Tian
- Department of Radiology, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoxue Zhang
- Department of Radiotherapy, Shanghai Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Yan Ren
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Chanchan Li
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yanmei Yang
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
391
|
Li J, Zhu Z, Li Y, Chen Y, Hu X, Liu Y, Shi Y, Hu Y, Bi Y, Xu X, Zheng M, Cheng L, Jing J. D-4F, an apolipoprotein A-I mimetic, promotes the clearance of myelin debris and the reduction of foamy macrophages after spinal cord injury. Bioengineered 2022; 13:11794-11809. [PMID: 35546071 PMCID: PMC9276047 DOI: 10.1080/21655979.2022.2073063] [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] [Indexed: 11/25/2022] Open
Abstract
After spinal cord injury (SCI), a large number of blood-derived macrophages infiltrate the lesion site and phagocytose myelin debris to become foamy macrophages, which leads to chronic inflammation. The drug D-4F, an apolipoprotein A-I peptidomimetic made of D-amino acids, has been reported to promote the lipid metabolism of foamy macrophages in atherosclerosis. However, the role and mechanism of D-4F in SCI are still unclear. In this study, we found that D-4F can promote the removal of myelin debris, reduce the formation of foamy macrophages in the lesion core and promote neuroprotection and recovery of motor function after SCI. These beneficial functions of D-4F may be related to its ability to upregulate the expression of ATP-binding cassette transporter A1 (ABCA1), the main transporter that mediates lipid efflux in foamy macrophages because inhibiting the activity of ABCA1 can reverse the effect of D-4F in vitro. In conclusion, D-4F may be a promising candidate for treating SCI by promoting the clearance of myelin debris by foamy macrophages via the ABCA1 pathway.
Collapse
Affiliation(s)
- Jinxin Li
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zhenyu Zhu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yiteng Li
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yihao Chen
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xuyang Hu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yanchang Liu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yi Shi
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yao Hu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yihui Bi
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xinzhong Xu
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Meige Zheng
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Li Cheng
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Juehua Jing
- Department of Orthopedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
392
|
Lattke M, Guillemot F. Understanding astrocyte differentiation: Clinical relevance, technical challenges, and new opportunities in the omics era. WIREs Mech Dis 2022; 14:e1557. [PMID: 35546493 PMCID: PMC9539907 DOI: 10.1002/wsbm.1557] [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: 05/06/2021] [Revised: 03/23/2022] [Accepted: 03/25/2022] [Indexed: 11/06/2022]
Abstract
Astrocytes are a major type of glial cells that have essential functions in development and homeostasis of the central nervous system (CNS). Immature astrocytes in the developing CNS support neuronal maturation and possess neural-stem-cell-like properties. Mature astrocytes partially lose these functions but gain new functions essential for adult CNS homeostasis. In pathological conditions, astrocytes become "reactive", which disrupts their mature homeostatic functions and reactivates some immature astrocyte-like properties, suggesting a partial reversal of astrocyte maturation. The loss of homeostatic astrocyte functions contributes to the pathogenesis of various neurological conditions, and therefore activating maturation-promoting mechanisms may be a promising therapeutic strategy to restore homeostasis. Manipulating the mechanisms underlying astrocyte maturation might also allow to facilitate CNS regeneration by enhancing developmental functions of adult astrocytes. However, such therapeutic strategies are still some distance away because of our limited understanding of astrocyte differentiation and maturation, due to biological and technical challenges, including the high degree of similarity of astrocytes with neural stem cells and the shortcomings of astrocyte markers. Current advances in systems biology have a huge potential to overcome these challenges. Recent transcriptomic analyses have already revealed new astrocyte markers and new regulators of astrocyte differentiation. However, the epigenomic changes that presumably occur during astrocyte differentiation remain an important, largely unexplored area for future research. Emerging technologies such as CRISPR/Cas9-based functional screens will further improve our understanding of the mechanisms underlying astrocyte differentiation. This may open up new clinical approaches to restore homeostasis in neurological disorders and/or promote CNS regeneration. This article is categorized under: Neurological Diseases > Genetics/Genomics/Epigenetics Neurological Diseases > Stem Cells and Development Neurological Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Michael Lattke
- Neural Stem Cell Biology Laboratory, The Francis Crick Institute, London, UK
| | - Francois Guillemot
- Neural Stem Cell Biology Laboratory, The Francis Crick Institute, London, UK
| |
Collapse
|
393
|
Lawal O, Ulloa Severino FP, Eroglu C. The role of astrocyte structural plasticity in regulating neural circuit function and behavior. Glia 2022; 70:1467-1483. [PMID: 35535566 PMCID: PMC9233050 DOI: 10.1002/glia.24191] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 04/28/2022] [Accepted: 04/28/2022] [Indexed: 12/12/2022]
Abstract
Brain circuits undergo substantial structural changes during development, driven by the formation, stabilization, and elimination of synapses. Synaptic connections continue to undergo experience‐dependent structural rearrangements throughout life, which are postulated to underlie learning and memory. Astrocytes, a major glial cell type in the brain, are physically in contact with synaptic circuits through their structural ensheathment of synapses. Astrocytes strongly contribute to the remodeling of synaptic structures in healthy and diseased central nervous systems by regulating synaptic connectivity and behaviors. However, whether structural plasticity of astrocytes is involved in their critical functions at the synapse is unknown. This review will discuss the emerging evidence linking astrocytic structural plasticity to synaptic circuit remodeling and regulation of behaviors. Moreover, we will survey possible molecular and cellular mechanisms regulating the structural plasticity of astrocytes and their non‐cell‐autonomous effects on neuronal plasticity. Finally, we will discuss how astrocyte morphological changes in different physiological states and disease conditions contribute to neuronal circuit function and dysfunction.
Collapse
Affiliation(s)
- Oluwadamilola Lawal
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA.,Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Francesco Paolo Ulloa Severino
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA.,Department of Neuroscience and Psychology, Duke University, Durham, North Carolina, USA.,Howard Hughes Medical Institute, Duke University, Durham, North Carolina, USA
| | - Cagla Eroglu
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina, USA.,Department of Neurobiology, Duke University Medical Center, Durham, North Carolina, USA.,Howard Hughes Medical Institute, Duke University, Durham, North Carolina, USA.,Duke Institute for Brain Sciences, Durham, North Carolina, USA
| |
Collapse
|
394
|
Cooke P, Janowitz H, Dougherty SE. Neuronal Redevelopment and the Regeneration of Neuromodulatory Axons in the Adult Mammalian Central Nervous System. Front Cell Neurosci 2022; 16:872501. [PMID: 35530177 PMCID: PMC9074815 DOI: 10.3389/fncel.2022.872501] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 03/24/2022] [Indexed: 01/09/2023] Open
Abstract
One reason that many central nervous system injuries, including those arising from traumatic brain injury, spinal cord injury, and stroke, have limited recovery of function is that neurons within the adult mammalian CNS lack the ability to regenerate their axons following trauma. This stands in contrast to neurons of the adult mammalian peripheral nervous system (PNS). New evidence, provided by single-cell expression profiling, suggests that, following injury, both mammalian central and peripheral neurons can revert to an embryonic-like growth state which is permissive for axon regeneration. This “redevelopment” strategy could both facilitate a damage response necessary to isolate and repair the acute damage from injury and provide the intracellular machinery necessary for axon regrowth. Interestingly, serotonin neurons of the rostral group of raphe nuclei, which project their axons into the forebrain, display a robust ability to regenerate their axons unaided, counter to the widely held view that CNS axons cannot regenerate without experimental intervention after injury. Furthermore, initial evidence suggests that norepinephrine neurons within the locus coeruleus possess similar regenerative abilities. Several morphological characteristics of serotonin axon regeneration in adult mammals, observable using longitudinal in vivo imaging, are distinct from the known characteristics of unaided peripheral nerve regeneration, or of the regeneration seen in the spinal cord and optic nerve that occurs with experimental intervention. These results suggest that there is an alternative CNS program for axon regeneration that likely differs from that displayed by the PNS.
Collapse
Affiliation(s)
- Patrick Cooke
- Linden Lab, Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Haley Janowitz
- Linden Lab, Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Sarah E Dougherty
- Linden Lab, Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
395
|
Kahuripour M, Behroozi Z, Rahimi B, Hamblin MR, Ramezani F. The potential of curcumin for treating spinal cord injury: a meta-analysis study. Nutr Neurosci 2022; 26:560-571. [PMID: 35507337 DOI: 10.1080/1028415x.2022.2070703] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
INTRODUCTION In this paper, we conducted a meta-analysis on the curcumin effect on functional recovery provided by the Basso, Beattie, Brenham (BBB) test for rats, and the Basso mouse scale (BMS) for mice after spinal cord injury (SCI) in animal models. METHOD Data mining was performed, and the standard mean difference (SMD) between the treated and control (untreated) groups was calculated using the STATA software. Quality control and subgroup analysis were performed. RESULTS The analysis includes 24 experimental studies that showed curcumin had a strong significance in improving functional recovery after SCI (SMD = 3.38; 95% CI: 2.54-4.22; p < 0.001). When curcumin was administered daily, it had a stronger effect than single-dose treatment or weekly administration. Despite the same effect in the follow-up time before and after 4 weeks post-injury, but later 9 weeks, curcumin had only a moderate effect. Curcumin also significantly reduced the expression of GFAP (Glial fibrillary acidic protein) marker compared to untreated groups. CONCLUSION These findings suggest that daily administration of curcumin can be an effective approach to improving functional recovery after SCI.
Collapse
Affiliation(s)
- Mahnaz Kahuripour
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Behroozi
- Physiology Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran.,Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Behnaz Rahimi
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa
| | - Fatemeh Ramezani
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
396
|
Nobili P, Shen W, Milicevic K, Bogdanovic Pristov J, Audinat E, Nikolic L. Therapeutic Potential of Astrocyte Purinergic Signalling in Epilepsy and Multiple Sclerosis. Front Pharmacol 2022; 13:900337. [PMID: 35586058 PMCID: PMC9109958 DOI: 10.3389/fphar.2022.900337] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Epilepsy and multiple sclerosis (MS), two of the most common neurological diseases, are characterized by the establishment of inflammatory environment in the central nervous system that drives disease progression and impacts on neurodegeneration. Current therapeutic approaches in the treatments of epilepsy and MS are targeting neuronal activity and immune cell response, respectively. However, the lack of fully efficient responses to the available treatments obviously shows the need to search for novel therapeutic candidates that will not exclusively target neurons or immune cells. Accumulating knowledge on epilepsy and MS in humans and analysis of relevant animal models, reveals that astrocytes are promising therapeutic candidates to target as they participate in the modulation of the neuroinflammatory response in both diseases from the initial stages and may play an important role in their development. Indeed, astrocytes respond to reactive immune cells and contribute to the neuronal hyperactivity in the inflamed brain. Mechanistically, these astrocytic cell to cell interactions are fundamentally mediated by the purinergic signalling and involve metabotropic P2Y1 receptors in case of astrocyte interactions with neurons, while ionotropic P2X7 receptors are mainly involved in astrocyte interactions with autoreactive immune cells. Herein, we review the potential of targeting astrocytic purinergic signalling mediated by P2Y1 and P2X7 receptors to develop novel approaches for treatments of epilepsy and MS at very early stages.
Collapse
Affiliation(s)
- Paola Nobili
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Weida Shen
- School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Katarina Milicevic
- Center for Laser Microscopy, Institute of Physiology and Biochemistry “Ivan Djaja”, University of Belgrade, Faculty of Biology, Belgrade, Serbia
| | - Jelena Bogdanovic Pristov
- Department of Life Sciences, University of Belgrade, Institute for Multidisciplinary Research, Belgrade, Serbia
| | - Etienne Audinat
- Institute of Functional Genomics (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Ljiljana Nikolic
- Department of Neurophysiology, University of Belgrade, Institute for Biological Research Siniša Stanković, National Institute of Republic of Serbia, Belgrade, Serbia
| |
Collapse
|
397
|
Abdi IY, Ghanem SS, El-Agnaf OM. Immune-related biomarkers for Parkinson's disease. Neurobiol Dis 2022; 170:105771. [DOI: 10.1016/j.nbd.2022.105771] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/14/2022] [Accepted: 05/15/2022] [Indexed: 12/13/2022] Open
|
398
|
Chen M, Guo L, Hao J, Ni J, Lv Q, Xin X, Liao H. p75NTR Promotes Astrocyte Proliferation in Response to Cortical Stab Wound. Cell Mol Neurobiol 2022; 42:1153-1166. [PMID: 33201418 PMCID: PMC11441203 DOI: 10.1007/s10571-020-01006-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 11/11/2020] [Indexed: 01/15/2023]
Abstract
Astrogliosis after brain trauma can have a significant impact on functional recovery. However, little is known about the mechanisms underlying astrocyte proliferation and subsequent astrogliosis. In this study, we established a cortical stab wound injury mouse model and observed dramatic astrocyte activation and nerve growth factor receptor (p75NTR) upregulation near the lesion. We also found profound alterations in the cell cycle of astrocytes near the lesion, with a switch from a mitotically quiescent (G0) phase to the G2/M and S phases. However, no changes in the level of astrocyte apoptosis were observed. Cell cycle progression to the G2/M and S phases and CDK2 protein levels in response to cortical stab wound was inhibited after p75NTR knockdown in mouse astrocytes. Conversely, p75NTR overexpression in mouse astrocytes was sufficient in promoting cell cycle progression. In conclusion, our results suggested that p75NTR upregulation in astrocytes after brain injury induces cell cycle entry by promoting CDK2 expression and promoting astrocyte proliferation. Our findings provided a better understanding of astrocytic responses after cortical stab wound injury in mice.
Collapse
Affiliation(s)
- Mingming Chen
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, No. 24 Tongjiaxiang Street, Nanjing, 210009, China
| | - Linlu Guo
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, No. 24 Tongjiaxiang Street, Nanjing, 210009, China
| | - Jie Hao
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, No. 24 Tongjiaxiang Street, Nanjing, 210009, China
| | - Jie Ni
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, No. 24 Tongjiaxiang Street, Nanjing, 210009, China
| | - Qunyu Lv
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, No. 24 Tongjiaxiang Street, Nanjing, 210009, China
| | - Xiaoyan Xin
- Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Hong Liao
- New Drug Screening Center, Jiangsu Center for Pharmacodynamics Research and Evaluation, China Pharmaceutical University, No. 24 Tongjiaxiang Street, Nanjing, 210009, China.
| |
Collapse
|
399
|
Lee HG, Wheeler MA, Quintana FJ. Function and therapeutic value of astrocytes in neurological diseases. Nat Rev Drug Discov 2022; 21:339-358. [PMID: 35173313 PMCID: PMC9081171 DOI: 10.1038/s41573-022-00390-x] [Citation(s) in RCA: 278] [Impact Index Per Article: 92.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2022] [Indexed: 12/20/2022]
Abstract
Astrocytes are abundant glial cells in the central nervous system (CNS) that perform diverse functions in health and disease. Astrocyte dysfunction is found in numerous diseases, including multiple sclerosis, Alzheimer disease, Parkinson disease, Huntington disease and neuropsychiatric disorders. Astrocytes regulate glutamate and ion homeostasis, cholesterol and sphingolipid metabolism and respond to environmental factors, all of which have been implicated in neurological diseases. Astrocytes also exhibit significant heterogeneity, driven by developmental programmes and stimulus-specific cellular responses controlled by CNS location, cell-cell interactions and other mechanisms. In this Review, we highlight general mechanisms of astrocyte regulation and their potential as therapeutic targets, including drugs that alter astrocyte metabolism, and therapies that target transporters and receptors on astrocytes. Emerging ideas, such as engineered probiotics and glia-to-neuron conversion therapies, are also discussed. We further propose a concise nomenclature for astrocyte subsets that we use to highlight the roles of astrocytes and specific subsets in neurological diseases.
Collapse
Affiliation(s)
- Hong-Gyun Lee
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael A Wheeler
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Francisco J Quintana
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
400
|
Ikeda-Yorifuji I, Tsujioka H, Sakata Y, Yamashita T. Single-nucleus RNA sequencing identified cells with ependymal cell-like features enriched in neonatal mice after spinal cord injury. Neurosci Res 2022; 181:22-38. [PMID: 35452717 DOI: 10.1016/j.neures.2022.04.006] [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: 12/16/2021] [Revised: 04/09/2022] [Accepted: 04/15/2022] [Indexed: 11/30/2022]
Abstract
The adult mammalian central nervous system has limited regenerative ability, and spinal cord injury (SCI) often causes lifelong motor disability. While regeneration is limited in adults, injured spinal cord tissue can be regenerated and neural function can be almost completely restored in neonates. However, difference of cellular composition in lesion has not been well characterized. To gain insight into the age-dependent cellular reaction after SCI, we performed single-nucleus RNA sequencing, analyzing 4,076 nuclei from sham and injured spinal cords from adult and neonatal mice. Clustering analysis identified 18 cell populations. We identified previously undescribed cells with ependymal cell-like gene expression profile, the number of which was increased in neonates after SCI. Histological analysis revealed that these cells line the central canal under physiological conditions in both adults and neonates. We confirmed that they were enriched in the lesion only in neonates. We further showed that these cells were positive for the cellular markers of ependymal cells, astrocytes and radial glial cells. This study provides a deeper understanding of neonate-specific cellular responses after SCI, which may determine regenerative capacity.
Collapse
Affiliation(s)
- Iyo Ikeda-Yorifuji
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan; Department of Cardiovascular Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hiroshi Tsujioka
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan; WPI Immunology Frontier Research Center, Osaka University, Suita, Japan.
| | - Yasushi Sakata
- Department of Cardiovascular Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Toshihide Yamashita
- Department of Molecular Neuroscience, Graduate School of Medicine, Osaka University, Osaka, Japan; WPI Immunology Frontier Research Center, Osaka University, Suita, Japan; Department of Molecular Neuroscience, Graduate School of Frontier Biosciences, Osaka University, Suita, Japan; Department of Neuro-Medical Science, Graduate School of Medicine, Osaka University, Suita, Japan.
| |
Collapse
|