101
|
Abstract
Blood–Brain Barrier Dysfunction in Aging Induces Hyperactivation of TGFβ Signaling and Chronic Yet Reversible Neural Dysfunction Senatorov VV Jr, Friedman AR, Milikovsky DZ, et al. Sci Transl Med. 2019;11(521):eaaw8283. doi:10.1126/scitranslmed.aaw8283 . A growing body of evidence shows that epileptic activity is frequent but often undiagnosed in patients with Alzheimer disease (AD) and has major therapeutic implications. Here, we analyzed electroencephalogram (EEG) data from patients with AD and found an EEG signature of transient slowing of the cortical network that we termed paroxysmal slow wave events (PSWEs). The occurrence per minute of the PSWEs was correlated with level of cognitive impairment. Interictal (between seizures) PSWEs were also found in patients with epilepsy, localized to cortical regions displaying blood–brain barrier (BBB) dysfunction, and in 3 rodent models with BBB pathology: aged mice, young 5× familial AD model, and status epilepticus–induced epilepsy in young rats. To investigate the potential causative role of BBB dysfunction in network modifications underlying PSWEs, we infused the serum protein albumin directly into the cerebral ventricles of naive young rats. Infusion of albumin, but not artificial cerebrospinal fluid control, resulted in high incidence of PSWEs. Our results identify PSWEs as an EEG manifestation of nonconvulsive seizures in patients with AD and suggest BBB pathology as an underlying mechanism and as a promising therapeutic target.
Collapse
|
102
|
Sutherland TC, Geoffroy CG. The Influence of Neuron-Extrinsic Factors and Aging on Injury Progression and Axonal Repair in the Central Nervous System. Front Cell Dev Biol 2020; 8:190. [PMID: 32269994 PMCID: PMC7109259 DOI: 10.3389/fcell.2020.00190] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/06/2020] [Indexed: 12/21/2022] Open
Abstract
In the aging western population, the average age of incidence for spinal cord injury (SCI) has increased, as has the length of survival of SCI patients. This places great importance on understanding SCI in middle-aged and aging patients. Axon regeneration after injury is an area of study that has received substantial attention and made important experimental progress, however, our understanding of how aging affects this process, and any therapeutic effort to modulate repair, is incomplete. The growth and regeneration of axons is mediated by both neuron intrinsic and extrinsic factors. In this review we explore some of the key extrinsic influences on axon regeneration in the literature, focusing on inflammation and astrogliosis, other cellular responses, components of the extracellular matrix, and myelin proteins. We will describe how each element supports the contention that axonal growth after injury in the central nervous system shows an age-dependent decline, and how this may affect outcomes after a SCI.
Collapse
Affiliation(s)
- Theresa C Sutherland
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, TX, United States
| | - Cédric G Geoffroy
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, TX, United States
| |
Collapse
|
103
|
Chen C, Zhong X, Smith DK, Tai W, Yang J, Zou Y, Wang LL, Sun J, Qin S, Zhang CL. Astrocyte-Specific Deletion of Sox2 Promotes Functional Recovery After Traumatic Brain Injury. Cereb Cortex 2020; 29:54-69. [PMID: 29161339 DOI: 10.1093/cercor/bhx303] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Accepted: 10/19/2017] [Indexed: 12/19/2022] Open
Abstract
Injury to the adult brain induces activation of local astrocytes, which serves as a compensatory response that modulates tissue damage and recovery. However, the mechanism governing astrocyte activation during brain injury remains largely unknown. Here we provide in vivo evidence that SOX2, a transcription factor critical for stem cells and brain development, is also required for injury-induced activation of adult cortical astrocytes. Genome-wide chromatin immunoprecipitation-seq analysis of mouse cortical tissues reveals that SOX2 binds to regulatory regions of genes associated with signaling pathways that control glial cell activation, such as Nr2e1, Mmd2, Wnt7a, and Akt2. Astrocyte-specific deletion of Sox2 in adult mice greatly diminishes glial response to controlled cortical impact injury and, most unexpectedly, dampens injury-induced cortical loss and benefits behavioral recovery of mice after injury. Together, these results uncover an essential role of SOX2 in somatic cells under pathological conditions and indicate that SOX2-dependent astrocyte activation could be targeted for functional recovery after traumatic brain injury.
Collapse
Affiliation(s)
- Chunhai Chen
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA.,Department of Occupational Health, Third Military Medical University, Chongqing, China
| | - Xiaoling Zhong
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA
| | - Derek K Smith
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA
| | - Wenjiao Tai
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA
| | - Jianjing Yang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA
| | - Yuhua Zou
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA
| | - Lei-Lei Wang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA
| | - Jiahong Sun
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA
| | - Song Qin
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA.,Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Center of Neural Injury and Repair, Shanghai Tenth People's Hospital Affiliated with Tongji University, Shanghai, China
| | - Chun-Li Zhang
- Department of Molecular Biology, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, 6000 Harry Hines Blvd, Dallas, TX, USA
| |
Collapse
|
104
|
Pous L, Deshpande SS, Nath S, Mezey S, Malik SC, Schildge S, Bohrer C, Topp K, Pfeifer D, Fernández-Klett F, Doostkam S, Galanakis DK, Taylor V, Akassoglou K, Schachtrup C. Fibrinogen induces neural stem cell differentiation into astrocytes in the subventricular zone via BMP signaling. Nat Commun 2020; 11:630. [PMID: 32005867 PMCID: PMC6994610 DOI: 10.1038/s41467-020-14466-y] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 01/09/2020] [Indexed: 12/30/2022] Open
Abstract
Neural stem/progenitor cells (NSPCs) originating from the subventricular zone (SVZ) contribute to brain repair during CNS disease. The microenvironment within the SVZ stem cell niche controls NSPC fate. However, extracellular factors within the niche that trigger astrogliogenesis over neurogenesis during CNS disease are unclear. Here, we show that blood-derived fibrinogen is enriched in the SVZ niche following distant cortical brain injury in mice. Fibrinogen inhibited neuronal differentiation in SVZ and hippocampal NSPCs while promoting astrogenesis via activation of the BMP receptor signaling pathway. Genetic and pharmacologic depletion of fibrinogen reduced astrocyte formation within the SVZ after cortical injury, reducing the contribution of SVZ-derived reactive astrocytes to lesion scar formation. We propose that fibrinogen is a regulator of NSPC-derived astrogenesis from the SVZ niche via BMP receptor signaling pathway following injury.
Collapse
Affiliation(s)
- Lauriane Pous
- Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Sachin S Deshpande
- Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Suvra Nath
- Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Szilvia Mezey
- Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Subash C Malik
- Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Sebastian Schildge
- Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Christian Bohrer
- Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Könül Topp
- Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Dietmar Pfeifer
- Department of Hematology, Oncology and Stem Cell Transplantation, University Medical Center Freiburg, University of Freiburg, 79106, Freiburg, Germany
| | - Francisco Fernández-Klett
- Department of Neuropsychiatry & Laboratory of Molecular Psychiatry, Charité - Universitätsmedizin Berlin, 10117, Berlin, Germany
| | - Soroush Doostkam
- Institute of Neuropathology, University Medical Center Freiburg, University of Freiburg, 79104, Freiburg, Germany
| | - Dennis K Galanakis
- Department of Pathology, State University of New York, Stony Brook, NY, 11794, USA
| | - Verdon Taylor
- Department of Biomedicine, Embryology and Stem Cell Biology, University of Basel, Mattenstrasse 28, CH-4058, Basel, Switzerland
| | - Katerina Akassoglou
- Gladstone Institutes, San Francisco, CA, 94158, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Christian Schachtrup
- Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany.
- Center for Basics in NeuroModulation (NeuroModulBasics), Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
105
|
Vilar R, Fish RJ, Casini A, Neerman-Arbez M. Fibrin(ogen) in human disease: both friend and foe. Haematologica 2020; 105:284-296. [PMID: 31949010 PMCID: PMC7012490 DOI: 10.3324/haematol.2019.236901] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 11/21/2019] [Indexed: 12/19/2022] Open
Abstract
Fibrinogen is an abundant protein synthesized in the liver, present in human blood plasma at concentrations ranging from 1.5-4 g/L in healthy individuals with a normal half-life of 3-5 days. With fibrin, produced by thrombin-mediated cleavage, fibrinogen plays important roles in many physiological processes. Indeed, the formation of a stable blood clot, containing polymerized and cross-linked fibrin, is crucial to prevent blood loss and drive wound healing upon vascular injury. A balance between clotting, notably the conversion of fibrinogen to fibrin, and fibrinolysis, the proteolytic degradation of the fibrin mesh, is essential. Disruption of this equilibrium can cause disease in distinct manners. While some pathological conditions are the consequence of altered levels of fibrinogen, others are related to structural properties of the molecule. The source of fibrinogen expression and the localization of fibrin(ogen) protein also have clinical implications. Low levels of fibrinogen expression have been detected in extra-hepatic tissues, including carcinomas, potentially contributing to disease. Fibrin(ogen) deposits at aberrant sites including the central nervous system or kidney, can also be pathological. In this review, we discuss disorders in which fibrinogen and fibrin are implicated, highlighting mechanisms that may contribute to disease.
Collapse
Affiliation(s)
- Rui Vilar
- Department of Genetic Medicine and Development, University of Geneva Faculty of Medicine
| | - Richard J Fish
- Department of Genetic Medicine and Development, University of Geneva Faculty of Medicine
| | - Alessandro Casini
- Division of Angiology and Hemostasis, University Hospitals and University of Geneva Faculty of Medicine
| | - Marguerite Neerman-Arbez
- Department of Genetic Medicine and Development, University of Geneva Faculty of Medicine .,iGE3, Institute of Genetics and Genomics in Geneva, Geneva, Switzerland
| |
Collapse
|
106
|
Feliu A, Mestre L, Carrillo-Salinas FJ, Yong VW, Mecha M, Guaza C. 2-arachidonoylglycerol reduces chondroitin sulphate proteoglycan production by astrocytes and enhances oligodendrocyte differentiation under inhibitory conditions. Glia 2020; 68:1255-1273. [PMID: 31894889 DOI: 10.1002/glia.23775] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 11/22/2019] [Accepted: 12/17/2019] [Indexed: 01/21/2023]
Abstract
The failure to remyelinate and regenerate is a critical impediment to recovery in multiple sclerosis (MS), resulting in severe dysfunction and disability. The chondroitin sulfate proteoglycans (CSPGs) that accumulate in MS lesions are thought to be linked to the failure to regenerate, impeding oligodendrocyte precursor cell (OPC) differentiation and neuronal growth. The potential of endocannabinoids to influence MS progression may reflect their capacity to enhance repair processes. Here, we investigated how 2-arachidonoylglycerol (2-AG) may affect the production of the CSPGs neurocan and brevican by astrocytes in culture. In addition, we studied whether 2-AG promotes oligodendrocyte differentiation under inhibitory conditions in vitro. Following treatment with 2-AG or by enhancing its endogenous tone through the use of inhibitors of its hydrolytic enzymes, CSPG production by rat and human TGF-β1 stimulated astrocytes was reduced. These effects of 2-AG might reflect its influence on TGF-β1/SMAD pathway, signaling that is involved in CSPG upregulation. The matrix generated from 2-AG-treated astrocytes is less inhibitory to oligodendrocyte differentiation and significantly, 2-AG administration directly promotes the differentiation of rat and human oligodendrocytes cultured under inhibitory conditions. Overall, the data obtained favor targeting the endocannabinoid system to neutralize CSPG accumulation and to enhance oligodendrocyte differentiation.
Collapse
Affiliation(s)
- Ana Feliu
- Functional and Systems Neurobiology Department, Neuroimmunology Group, Instituto Cajal, CSIC, Madrid, Spain
| | - Leyre Mestre
- Functional and Systems Neurobiology Department, Neuroimmunology Group, Instituto Cajal, CSIC, Madrid, Spain
| | | | - V Wee Yong
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences and Oncology, University of Calgary, Calgary, Alberta, Canada
| | - Miriam Mecha
- Functional and Systems Neurobiology Department, Neuroimmunology Group, Instituto Cajal, CSIC, Madrid, Spain
| | - Carmen Guaza
- Functional and Systems Neurobiology Department, Neuroimmunology Group, Instituto Cajal, CSIC, Madrid, Spain
| |
Collapse
|
107
|
Li C, Ma W, Li S, Zhao Y, Zhao X, Wang H. The FDP/FIB Ratio and Blood FDP Level May Be Related to Seizures After Fever in Young Children. Front Pediatr 2020; 8:439. [PMID: 32850552 PMCID: PMC7412868 DOI: 10.3389/fped.2020.00439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 06/23/2020] [Indexed: 11/18/2022] Open
Abstract
Objective: To evaluate the relationship of the blood fibrinogen (FIB) degradation product (FDP) level and FDP/FIB ratio with seizure in young children with fever. Methods: A total of 35 children with simple febrile seizures and 80 children with fever but no seizure were selected. First, the differences in white blood cell (WBC), platelets (PLT), prothrombin time (PT), activated partial thromboplastin time (APTT), thrombin time (TT), FIB, FDP, FDP/FIB ratio, and C-reactive protein (CRP) between 35 children with simple febrile seizures and 40 randomly selected children with fever but no seizure were retrospectively analyzed. Then, an ROC curve was used to determine the diagnostic utility of the FDP level, FDP/FIB ratio, and FDP+FDP/FIB ratio, and the best diagnostic cutoff points were selected. Finally, the diagnostic specificities of the three diagnostic indicators were verified by comparison with the results of all 80 children with fever but no seizure. Results: The FDP level and FDP/FIB ratio were significantly different between the two groups (P < 0.0001) and there was a positive correlation between the FDP and FIB levels. Both the FDP level and FDP/FIB ratio had good diagnostic value. An FDP ≥ 2.0 mg/L and FDP/FIB ratio ≥ 0.5 had good diagnostic specificities. Combined application of an FDP ≥ 2.0 mg/L and FDP/FIB ratio ≥ 0.5 improved the diagnostic power. Conclusions: The blood FDP level and FDP/FIB ratio may be related to seizures after fever, and an FDP ≥ 2.0 mg/L + FDP/FIB ratio ≥ 0.5 has good diagnostic specificity.
Collapse
Affiliation(s)
- Chun Li
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Weining Ma
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shaoyi Li
- Department of Neurosurgery, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yajuan Zhao
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xuyang Zhao
- Beijing Key Laboratory of Tumor Systems Biology, Department of Pathology, Peking-Tsinghua Center for Life Sciences, School of Basic Medical Sciences, Institute of Systems Biomedicine, Peking University Health Science Center, Beijing, China
| | - Hua Wang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
108
|
Eide PK, Hansson HA. Blood-brain barrier leakage of blood proteins in idiopathic normal pressure hydrocephalus. Brain Res 2020; 1727:146547. [DOI: 10.1016/j.brainres.2019.146547] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2019] [Revised: 10/14/2019] [Accepted: 11/07/2019] [Indexed: 01/05/2023]
|
109
|
Hart CG, Dyck SM, Kataria H, Alizadeh A, Nagakannan P, Thliveris JA, Eftekharpour E, Karimi-Abdolrezaee S. Acute upregulation of bone morphogenetic protein-4 regulates endogenous cell response and promotes cell death in spinal cord injury. Exp Neurol 2019; 325:113163. [PMID: 31881217 DOI: 10.1016/j.expneurol.2019.113163] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 12/10/2019] [Accepted: 12/24/2019] [Indexed: 01/11/2023]
Abstract
Traumatic spinal cord injury (SCI) elicits a cascade of secondary injury mechanisms that induce profound changes in glia and neurons resulting in their activation, injury or cell death. The resultant imbalanced microenvironment of acute SCI also negatively impacts regenerative processes in the injured spinal cord. Thus, it is imperative to uncover endogenous mechanisms that drive these acute injury events. Here, we demonstrate that the active form of bone morphogenetic protein-4 (BMP4) is robustly and transiently upregulated in acute SCI in rats. BMP4 is a key morphogen in neurodevelopment; however, its role in SCI is not fully defined. Thus, we elucidated the ramification of BMP4 upregulation in a preclinical model of compressive/contusive SCI in the rat by employing noggin, an endogenous antagonist of BMP ligands, and LDN193189, an intracellular inhibitor of BMP signaling. In parallel, we studied cell-specific effects of BMP4 on neural precursor cells (NPCs), oligodendrocyte precursor cells (OPCs), neurons and astrocytes in vitro. We demonstrate that activation of BMP4 inhibits differentiation of spinal cord NPCs and OPCs into mature myelin-expressing oligodendrocytes, and acute blockade of BMPs promotes oligodendrogenesis, oligodendrocyte preservation and remyelination after SCI. Importantly, we report for the first time that BMP4 directly induces caspase-3 mediated apoptosis in neurons and oligodendrocytes in vitro, and noggin and LDN193189 remarkably attenuate caspase-3 activation and lipid peroxidation in acute SCI. BMP4 also enhances the production of inhibitory chondroitin sulfate proteoglycans (CSPGs) in activated astrocytes in vitro and after SCI. Interestingly, our work reveals that despite the beneficial effects of BMP inhibition in acute SCI, neither noggin nor LDN193189 treatment resulted in long-term functional recovery. Collectively, our findings suggest a role for BMP4 in regulating acute secondary injury mechanisms following SCI, and a potential target for combinatorial approaches to improve endogenous cell response and remyelination.
Collapse
Affiliation(s)
- Christopher G Hart
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Scott M Dyck
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Hardeep Kataria
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Arsalan Alizadeh
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Pandian Nagakannan
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - James A Thliveris
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Eftekhar Eftekharpour
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada; Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
110
|
Streeter KA, Sunshine MD, Brant JO, Sandoval AGW, Maden M, Fuller DD. Molecular and histologic outcomes following spinal cord injury in spiny mice, Acomys cahirinus. J Comp Neurol 2019; 528:1535-1547. [PMID: 31820438 DOI: 10.1002/cne.24836] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/27/2019] [Accepted: 11/27/2019] [Indexed: 12/15/2022]
Abstract
The spiny mouse (Acomys cahirinus) appears to be unique among mammals by showing little scarring or fibrosis after skin or muscle injury, but the Acomys response to spinal cord injury (SCI) is unknown. We tested the hypothesis that Acomys would have molecular and immunohistochemical evidence of reduced spinal inflammation and fibrosis following SCI as compared to C57BL/6 mice (Mus), which similar to all mammals studied to date exhibits spinal scarring following SCI. Initial experiments used two pathway-focused RT-PCR gene arrays ("wound healing" and "neurogenesis") to evaluate tissue samples from the C2-C6 spinal cord 3 days after a C3/C4 hemi-crush injury (C3Hc). Based on the gene array results, specific genes were selected for RT-qPCR evaluation using species-specific primers. The results supported our hypothesis by showing increased inflammation and fibrosis related gene expression (Serpine 1, Plau, and Timp1) in Mus as compared to Acomys (p < .05). RT-qPCR also showed enhanced stem cell and axonal guidance related gene expression (Bmp2, GDNF, and Shh) in Acomys compared to Mus (p < .05). Immunohistochemical evaluation of the spinal lesion at 4 weeks postinjury indicated less collagen IV immunostaining in Acomys (p < .05). Glial fibrillary acidic protein (GFAP) and ionized calcium binding adaptor molecule 1(IBA1) immunostaining indicated morphological differences in the appearance of astrocytes and macrophages/microglia in Acomys. Collectively, the molecular and histologic results support the hypothesis that Acomys has reduced spinal inflammation and fibrosis following SCI. We suggest that Acomys may be a useful comparative model to study adaptive responses to SCI.
Collapse
Affiliation(s)
- Kristi A Streeter
- Department of Physical Therapy, University of Florida, Gainesville, Florida.,McKnight Brain Institute, University of Florida, Gainesville, Florida.,Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, Florida
| | - Michael D Sunshine
- Department of Physical Therapy, University of Florida, Gainesville, Florida.,McKnight Brain Institute, University of Florida, Gainesville, Florida.,Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, Florida
| | - Jason O Brant
- Department of Biology, University of Florida, Gainesville, Florida
| | | | - Malcolm Maden
- Department of Biology, University of Florida, Gainesville, Florida
| | - David D Fuller
- Department of Physical Therapy, University of Florida, Gainesville, Florida.,McKnight Brain Institute, University of Florida, Gainesville, Florida.,Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, Florida
| |
Collapse
|
111
|
Bhattrai A, Irimia A, Van Horn JD. Neuroimaging of traumatic brain injury in military personnel: An overview. J Clin Neurosci 2019; 70:1-10. [PMID: 31331746 PMCID: PMC6861663 DOI: 10.1016/j.jocn.2019.07.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 07/04/2019] [Indexed: 02/02/2023]
Abstract
BACKGROUND The incidence of blunt-force traumatic brain injury (TBI) is especially prevalent in the military, where the emergency care admission rate has been reported to be 24.6-41.8 per 10,000 soldier-years. Given substantial advancements in modern neuroimaging techniques over the past decade in terms of structural, functional, and connectomic approaches, this mode of exploration can be viewed as best suited for understanding the underlying pathology and for providing proper intervention at effective time-points. APPROACH Here we survey neuroimaging studies of mild-to-severe TBI in military veterans with the intent to aid the field in the creation of a roadmap for clinicians and researchers whose aim is to understand TBI progression. DISCUSSION Recent advancements on the quantification of neurocognitive dysfunction, cellular dysfunction, intracranial pressure, cerebral blood flow, inflammation, post-traumatic neuropathophysiology, on blood serum biomarkers and on their correlation to neuroimaging findings are reviewed to hypothesize how they can be used in conjunction with one another. This may allow clinicians and scientists to comprehensively study TBI in military service members, leading to new treatment strategies for both currently-serving as well as veteran personnel, and to improve the study of TBI more broadly.
Collapse
Affiliation(s)
- Avnish Bhattrai
- USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, 2025 Zonal Avenue, SHN, Los Angeles, CA 90033, USA
| | - Andrei Irimia
- Ethel Percy Andrus Gerontology Center, USC Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Ave., Room 228C, Los Angeles, CA 90089-0191, USA.
| | - John Darrell Van Horn
- USC Mark and Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine of USC, University of Southern California, 2025 Zonal Avenue, SHN, Los Angeles, CA 90033, USA.
| |
Collapse
|
112
|
Roth M, Enström A, Aghabeick C, Carlsson R, Genové G, Paul G. Parenchymal pericytes are not the major contributor of extracellular matrix in the fibrotic scar after stroke in male mice. J Neurosci Res 2019; 98:826-842. [PMID: 31758600 PMCID: PMC7154736 DOI: 10.1002/jnr.24557] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2019] [Revised: 10/15/2019] [Accepted: 10/28/2019] [Indexed: 12/24/2022]
Abstract
Scar formation after injury of the brain or spinal cord is a common event. While glial scar formation by astrocytes has been extensively studied, much less is known about the fibrotic scar, in particular after stroke. Platelet‐derived growth factor receptor ß‐expressing (PDGFRß+) pericytes have been suggested as a source of the fibrotic scar depositing fibrous extracellular matrix (ECM) proteins after detaching from the vessel wall. However, to what extent these parenchymal PDGFRß+ cells contribute to the fibrotic scar and whether targeting these cells affects fibrotic scar formation in stroke is still unclear. Here, we utilize male transgenic mice that after a permanent middle cerebral artery occlusion stroke model have a shift from a parenchymal to a perivascular location of PDGFRß+ cells due to the loss of regulator of G‐protein signaling 5 in pericytes. We find that only a small fraction of parenchymal PDGFRß+ cells co‐label with type I collagen and fibronectin. Consequently, a reduction in parenchymal PDGFRß+ cells by ca. 50% did not affect the overall type I collagen or fibronectin deposition after stroke. The redistribution of PDGFRß+ cells to a perivascular location, however, resulted in a reduced thickening of the vascular basement membrane and changed the temporal dynamics of glial scar maturation after stroke. We demonstrate that parenchymal PDGFRß+ cells are not the main contributor to the fibrotic ECM, and therefore targeting these cells might not impact on fibrotic scar formation after stroke.
Collapse
Affiliation(s)
- Michaela Roth
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Andreas Enström
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Candice Aghabeick
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Robert Carlsson
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Guillem Genové
- Integrated Cardio Metabolic Center, Department of Medicine, Karolinska Institute, Huddinge, Sweden
| | - Gesine Paul
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden.,Department of Neurology, Scania University Hospital, Lund, Sweden.,Wallenberg Centrum for Molecular Medicine, Lund University, Lund, Sweden
| |
Collapse
|
113
|
Zhou P, Guan J, Xu P, Zhao J, Zhang C, Zhang B, Mao Y, Cui W. Cell Therapeutic Strategies for Spinal Cord Injury. Adv Wound Care (New Rochelle) 2019; 8:585-605. [PMID: 31637103 PMCID: PMC6798812 DOI: 10.1089/wound.2019.1046] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Accepted: 08/27/2019] [Indexed: 12/13/2022] Open
Abstract
Significance: Spinal cord injury (SCI) is a neurological disorder that resulted from destroyed long axis of spinal cord, affecting thousands of people every year. With the occurrence of SCI, the lesions can form cystic cavities and produce glial scar, myelin inhibitor, and inflammation that negatively impact repair of spinal cord. Therefore, SCI remains a difficult problem to overcome with present therapeutics. This review of cell therapeutics in SCI provides a systematic review of combinatory therapeutics of SCI and helps the realization of regeneration of spinal cord in the future. Recent Advances: With major breakthroughs in neurobiology in recent years, present therapeutic strategies for SCI mainly aim at nerve regeneration or neuroprotection. For nerve regeneration, the application approaches are tissue engineering and cell transplantation, while drug therapeutics is applied for neuroprotection. Cell therapeutics is a new approach that treats SCI by cell transplantation. Cell therapeutics possesses advantages of neuroprotection, immune regulation, axonal regeneration, neuron relay formation, and remyelination. Critical Issues: Neurons cannot regenerate at the site of injury. Therefore, it is essential to find a repair strategy for remyelination, axon regeneration, and functional recovery. Cell therapeutics is emerging as the most promising approach for treating SCI. Future Directions: The future application of SCI therapy in clinical practice may require a combination of multiple strategies. A comprehensive treatment of injury of spinal cord is the focus of the present research. With the combination of different cell therapy strategies, future experiments will achieve more dramatic success in spinal cord repair.
Collapse
Affiliation(s)
- Pinghui Zhou
- Department of Orthopedics, First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China
- Anhui Province Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, P.R. China
| | - Jingjing Guan
- Department of Orthopedics, First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China
| | - Panpan Xu
- Department of Orthopedics, First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China
| | - Jingwen Zhao
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Changchun Zhang
- Department of Orthopedics, First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China
| | - Bin Zhang
- Department of Orthopedics, First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China
| | - Yingji Mao
- Department of Orthopedics, First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China
- School of Life Science, Bengbu Medical College, Bengbu, P.R. China
| | - Wenguo Cui
- Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| |
Collapse
|
114
|
Motallebnejad P, Thomas A, Swisher SL, Azarin SM. An isogenic hiPSC-derived BBB-on-a-chip. BIOMICROFLUIDICS 2019; 13:064119. [PMID: 31768205 PMCID: PMC6874510 DOI: 10.1063/1.5123476] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 11/07/2019] [Indexed: 05/03/2023]
Abstract
The blood-brain barrier (BBB) is composed of brain microvascular endothelial cells (BMECs) that regulate brain homeostasis, and astrocytes within the brain are involved in the maintenance of the BBB or modulation of its integrity in disease states via secreted factors. A major challenge in modeling the normal or diseased BBB is that conventional in vitro models lack either the physiological complexity of the BBB or key functional features such as formation of a sufficiently tight barrier. In this study, we utilized human induced pluripotent stem cell (hiPSC)-derived BMECs in a BBB-on-a-chip device that supports flow and coculture with an astrocyte-laden 3D hydrogel. The BMECs are separated from the hydrogel by a porous membrane with either 0.4 or 8.0 μm pore size, making the device suitable for studying the transport of molecules or cells, respectively, across the BBB. In addition, all cells seeded in the device are differentiated from the same hiPSC line, which could enable genetic and rare disease modeling. Formation of a confluent BMEC barrier was confirmed by immunocytochemistry of tight junction proteins and measurement of fluorescein permeability. Integrity of the barrier was further assessed by performing impedance spectroscopy in the device. Finally, the ability of this device to recapitulate a disease model of BBB disruption was demonstrated, with apical addition of TGF-β1 leading to transendothelial electrical resistance reduction and indicators of astrocyte activation. These results demonstrate the utility of the fabricated device for a broad range of applications such as drug screening and mechanistic studies of BBB disruption.
Collapse
Affiliation(s)
- Pedram Motallebnejad
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Andrew Thomas
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Sarah L. Swisher
- Department of Electrical and Computer Engineering, University of Minnesota, Minneapolis, Minnesota 55455, USA
| | - Samira M. Azarin
- Department of Chemical Engineering and Materials Science, University of Minnesota, Minneapolis, Minnesota 55455, USA
| |
Collapse
|
115
|
Divolis G, Stavropoulos A, Manioudaki M, Apostolidou A, Doulou A, Gavriil A, Dafnis I, Chroni A, Mummery C, Xilouri M, Sideras P. Activation of both transforming growth factor-β and bone morphogenetic protein signalling pathways upon traumatic brain injury restrains pro-inflammatory and boosts tissue reparatory responses of reactive astrocytes and microglia. Brain Commun 2019; 1:fcz028. [PMID: 32954268 PMCID: PMC7425383 DOI: 10.1093/braincomms/fcz028] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/24/2019] [Accepted: 10/07/2019] [Indexed: 02/06/2023] Open
Abstract
Various ligands and receptors of the transforming growth factor-β superfamily have been found upregulated following traumatic brain injury; however, the role of this signalling system in brain injury pathophysiology is not fully characterized. To address this, we utilized an acute stab wound brain injury model to demonstrate that hallmarks of transforming growth factor-β superfamily system activation, such as levels of phosphorylated Smads, ligands and target genes for both transforming growth factor-β and bone morphogenetic protein pathways, were upregulated within injured tissues. Using a bone morphogenetic protein-responsive reporter mouse model, we showed that activation of the bone morphogenetic protein signalling pathway involves primarily astrocytes that demarcate the wound area. Insights regarding the potential role of transforming growth factor-β superfamily activation in glia cells within the injured tissues were obtained indirectly by treating purified reactive astrocytes and microglia with bone morphogenetic protein-4 or transforming growth factor-β1 and characterizing changes in their transcriptional profiles. Astrocytes responded to both ligands with considerably overlapping profiles, whereas, microglia responded selectively to transforming growth factor-β1. Novel pathways, crucial for repair of tissue-injury and blood-brain barrier, such as activation of cholesterol biosynthesis and transport, production of axonal guidance and extracellular matrix components were upregulated by transforming growth factor-β1 and/or bone morphogenetic protein-4 in astrocytes. Moreover, both ligands in astrocytes and transforming growth factor-β1 in microglia shifted the phenotype of reactive glia cells towards the anti-inflammatory and tissue reparatory 'A2'-like and 'M0/M2'-like phenotypes, respectively. Increased expression of selected key components of the in vitro modulated pathways and markers of 'A2'-like astrocytes was confirmed within the wound area, suggesting that these processes could also be modulated in situ by the integrated action of transforming growth factor-β and/or bone morphogenetic protein-mediated signalling. Collectively, our study provides a comprehensive comparative analysis of transforming growth factor-β superfamily signalling in reactive astrocytes and microglia and points towards a crucial role of both transforming growth factor-β and bone morphogenetic protein pathways in modulating the inflammatory and brain injury reparatory functions of activated glia cells.
Collapse
Affiliation(s)
- Georgios Divolis
- Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Athanasios Stavropoulos
- Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Maria Manioudaki
- Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Anastasia Apostolidou
- Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Athanasia Doulou
- Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Ariana Gavriil
- Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Ioannis Dafnis
- Institute of Biosciences and Applications, National Center for Scientific Research-Demokritos, 15341 Athens, Greece
| | - Angeliki Chroni
- Institute of Biosciences and Applications, National Center for Scientific Research-Demokritos, 15341 Athens, Greece
| | - Christine Mummery
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333ZC Leiden, The Netherlands
| | - Maria Xilouri
- Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Paschalis Sideras
- Center for Clinical Research, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| |
Collapse
|
116
|
Liu Y, Chen H, Zhao K, He W, Lin S, He J. High levels of plasma fibrinogen are related to post-stroke cognitive impairment. Brain Behav 2019; 9:e01391. [PMID: 31475471 PMCID: PMC6790326 DOI: 10.1002/brb3.1391] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 06/30/2019] [Accepted: 07/28/2019] [Indexed: 01/01/2023] Open
Abstract
INTRODUCTION Studies have shown that high levels of the fibrinogen (FIB) are related to cognitive deficits. However, the relationship between fibrinogen and cognitive deficit after stroke remains unclear. Therefore, we explored the relationship between plasma fibrinogen and post-stroke cognitive impairment (PSCI). METHODS This study is carried out in the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China. A total of 210 patients with acute ischemic stroke were enrolled in this study. Ultimately, 134 patients completed 3-month follow-up. Blood samples were collected at hospital admission. Cognitive function was evaluated 3 months after stroke. All patients underwent the Mini-Mental State Examination (MMSE) after 3 months. RESULTS Higher levels of fibrinogen were observed in patients with post-stroke cognitive impairment compared with the non-PSCI group (p < .001). Additionally, elevated plasma fibrinogen levels were independently associated with PSCI (odds ratio [OR] = 2.000, 95% CI 1.062-3.770 p = .032). The plasma fibrinogen levels were negatively correlated with the 3-month MMSE scores (r = -.171, p = .048). In a multivariate linear regression, FIB was negatively associated with the 3-month MMSE scores after adjustment for the other variables (β = -0.782, p = .035). CONCLUSION High levels of plasma fibrinogen were associated with the presence and severity of PSCI.
Collapse
Affiliation(s)
- Yuntao Liu
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Huijun Chen
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kai Zhao
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Weilei He
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Shasha Lin
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jincai He
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
117
|
Chronic Cerebral Hypoperfusion Activates the Coagulation and Complement Cascades in Alzheimer's Disease Mice. Neuroscience 2019; 416:126-136. [DOI: 10.1016/j.neuroscience.2019.07.050] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 07/29/2019] [Accepted: 07/30/2019] [Indexed: 12/22/2022]
|
118
|
Charkviani M, Muradashvili N, Lominadze D. Vascular and non-vascular contributors to memory reduction during traumatic brain injury. Eur J Neurosci 2019; 50:2860-2876. [PMID: 30793398 PMCID: PMC6703968 DOI: 10.1111/ejn.14390] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 02/06/2019] [Accepted: 02/07/2019] [Indexed: 01/09/2023]
Abstract
Traumatic brain injury (TBI) is an increasing health problem. It is a complex, progressive disease that consists of many factors affecting memory. Studies have shown that increased blood-brain barrier (BBB) permeability initiates pathological changes in neuro-vascular network but the role of cerebrovascular dysfunction and its mediated mechanisms associated with memory reduction during TBI are still not well understood. Changes in BBB, inflammation, extravasation of blood plasma components, activation of neuroglia lead to neurodegeneration. Extravasated proteins such as amyloid-beta, fibrinogen, and cellular prion protein may form degradation resistant complexes that can lead to neuronal dysfunction and degeneration. They also have the ability to activate astrocytes, and thus, can be involved in memory impairment. Understanding the triggering mechanisms and the places they originate in vasculature or in extravascular tissue may help to identify potential therapeutic targets to ameliorate memory reduction during TBI. The goal of this review is to discuss conceptual mechanisms that lead to short-term memory reduction during non-severe TBI considering distinction between vascular and non-vascular effects on neurons. Some aspects of these mechanisms need to be confirmed further. Therefore, we hope that the discussion presented bellow may lead to experiments that may clarify the triggering mechanisms of memory reduction after head trauma.
Collapse
Affiliation(s)
- Mariam Charkviani
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Nino Muradashvili
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
- Department of Basic Medicine, Caucasus International University, Tbilisi, Georgia
| | - David Lominadze
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
- Kentucky Spinal Cord Research Center, University of Louisville, School of Medicine, Louisville, KY, USA
| |
Collapse
|
119
|
Stamatovic SM, Phillips CM, Martinez-Revollar G, Keep RF, Andjelkovic AV. Involvement of Epigenetic Mechanisms and Non-coding RNAs in Blood-Brain Barrier and Neurovascular Unit Injury and Recovery After Stroke. Front Neurosci 2019; 13:864. [PMID: 31543756 PMCID: PMC6732937 DOI: 10.3389/fnins.2019.00864] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 07/31/2019] [Indexed: 12/13/2022] Open
Abstract
Cessation of blood flow leads to a complex cascade of pathophysiological events at the blood-vascular-parenchymal interface which evolves over time and space, and results in damage to neural cells and edema formation. Cerebral ischemic injury evokes a profound and deleterious upregulation in inflammation and triggers multiple cell death pathways, but it also induces a series of the events associated with regenerative responses, including vascular remodeling, angiogenesis, and neurogenesis. Emerging evidence suggests that epigenetic reprograming could play a pivotal role in ongoing post-stroke neurovascular unit (NVU) changes and recovery. This review summarizes current knowledge about post-stroke recovery processes at the NVU, as well as epigenetic mechanisms and modifiers (e.g., DNA methylation, histone modifying enzymes and microRNAs) associated with stroke injury, and NVU repair. It also discusses novel drug targets and therapeutic strategies for enhancing post-stroke recovery.
Collapse
Affiliation(s)
- Svetlana M. Stamatovic
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Chelsea M. Phillips
- Neuroscience Graduate Program, University of Michigan Medical School, Ann Arbor, MI, United States
| | | | - Richard F. Keep
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Molecular Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Anuska V. Andjelkovic
- Department of Pathology, University of Michigan Medical School, Ann Arbor, MI, United States
- Department of Neurosurgery, University of Michigan Medical School, Ann Arbor, MI, United States
| |
Collapse
|
120
|
TGF-β Secretion by M2 Macrophages Induces Glial Scar Formation by Activating Astrocytes In Vitro. J Mol Neurosci 2019; 69:324-332. [PMID: 31327154 DOI: 10.1007/s12031-019-01361-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Accepted: 06/25/2019] [Indexed: 02/07/2023]
Abstract
Transforming growth factor-β (TGF-β) is a key factor that promotes fibrosis or scar formation, which could become an obstacle in the repair of impaired axons in the central nervous system (CNS) of the human body resulting from diseases or injuries. Considering that major pathological reactions occur during this process, we focused on TGF-secreting M2 macrophages to identify the interactions between M2 macrophages and astrocytes (AS) and verify the specific mechanism of fibrosis or glial scar formation. In the present study, we used the Transwell coculturing technique and found an increase in glial fibrillary acidic protein (GFAP), neurocan, IL-13, and TGF-β expression after incubation for 48 h; the expression of these proteins decreased when additional inhibitors of the TGF-β receptor were added. We concluded that fibrosis or glial scar formation would be enhanced by the secretion of neurocan from AS, resulting from the release of TGF-β from M2 macrophages. We also used M2 macrophage-conditioned medium to further confirm this finding in a subsequent experiment. We hope that the findings in this research could provide a foundation for locating new targets for treating CNS diseases or injuries.
Collapse
|
121
|
Brown AC, Lavik E, Stabenfeldt SE. Biomimetic Strategies To Treat Traumatic Brain Injury by Leveraging Fibrinogen. Bioconjug Chem 2019; 30:1951-1956. [PMID: 31246419 DOI: 10.1021/acs.bioconjchem.9b00360] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
There were over 27 million new cases of traumatic brain injuries (TBIs) in 2016 across the globe. TBIs are often part of complicated trauma scenarios and may not be diagnosed initially because the primary clinical focus is on stabilizing the patient. Interventions used to stabilize trauma patients may inadvertently impact the outcomes of TBIs. Recently, there has been a strong interest in the trauma community toward administrating fibrinogen-containing solutions intravenously to help stabilize trauma patients. While this interventional shift may benefit general trauma scenarios, fibrinogen is associated with potentially deleterious effects for TBIs. Here, we deconstruct what components of fibrinogen may be beneficial as well as potentially harmful following TBI and extrapolate this to biomimetic approaches to treat bleeding and trauma that may also lead to better outcomes following TBI.
Collapse
Affiliation(s)
- Ashley C Brown
- Joint Department of Biomedical Engineering , North Carolina State University and The University of North Carolina at Chapel Hill , Raleigh , North Carolina 27695 , United States.,Comparative Medicine Institute , North Carolina State University , Raleigh , North Carolina 27695 , United States
| | - Erin Lavik
- Chemical, Biochemical, and Environmental Engineering , University of Maryland, Baltimore County , Baltimore , Maryland 21250 , United States
| | - Sarah E Stabenfeldt
- School of Biological and Health Systems Engineering , Arizona State University , Tempe , Arizona 85287 , United States
| |
Collapse
|
122
|
Golanov EV, Sharpe MA, Regnier-Golanov AS, Del Zoppo GJ, Baskin DS, Britz GW. Fibrinogen Chains Intrinsic to the Brain. Front Neurosci 2019; 13:541. [PMID: 31191233 PMCID: PMC6549596 DOI: 10.3389/fnins.2019.00541] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 05/09/2019] [Indexed: 11/13/2022] Open
Abstract
We observed fine fibrin deposition along the paravascular spaces in naive animals, which increased dramatically following subarachnoid hemorrhage (SAH). Following SAH, fibrin deposits in the areas remote from the hemorrhage. Traditionally it is thought that fibrinogen enters subarachnoid space through damaged blood brain barrier. However, deposition of fibrin remotely from hemorrhage suggests that fibrinogen chains Aα, Bβ, and γ can originate in the brain. Here we demonstrate in vivo and in vitro that astroglia and neurons are capable of expression of fibrinogen chains. SAH in mice was induced by the filament perforation of the circle of Willis. Four days after SAH animals were anesthetized, transcardially perfused and fixed. Whole brain was processed for immunofluorescent (IF) analysis of fibrin deposition on the brain surface or in brains slices processed for fibrinogen chains Aα, Bβ, γ immunohistochemical detection. Normal human astrocytes were grown media to confluency and stimulated with NOC-18 (100 μM), TNF-α (100 nM), ATP-γ-S (100 μM) for 24 h. Culture was fixed and washed/permeabilized with 0.1% Triton and processed for IF. Four days following SAH fibrinogen chains Aα IF associated with glia limitans and superficial brain layers increased 3.2 and 2.5 times (p < 0.05 and p < 0.01) on the ventral and dorsal brain surfaces respectively; fibrinogen chains Bβ increased by 3 times (p < 0.01) on the dorsal surface and fibrinogen chain γ increased by 3 times (p < 0.01) on the ventral surface compared to sham animals. Human cultured astrocytes and neurons constitutively expressed all three fibrinogen chains. Their expression changed differentially when exposed for 24 h to biologically significant stimuli: TNFα, NO or ATP. Western blot and RT-qPCR confirmed presence of the products of the appropriate molecular weight and respective mRNA. We demonstrate for the first time that mouse and human astrocytes and neurons express fibrinogen chains suggesting potential presence of endogenous to the brain fibrinogen chains differentially changing to biologically significant stimuli. SAH is followed by increased expression of fibrinogen chains associated with glia limitans remote from the hemorrhage. We conclude that brain astrocytes and neurons are capable of production of fibrinogen chains, which may be involved in various normal and pathological processes.
Collapse
Affiliation(s)
- Eugene V Golanov
- Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, United States
| | - Martyn A Sharpe
- Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, United States
| | | | - Gregory J Del Zoppo
- Division of Hematology, University of Washington School of Medicine, Seattle, WA, United States
| | - David S Baskin
- Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, United States
| | - Gavin W Britz
- Department of Neurosurgery, Houston Methodist Hospital, Houston, TX, United States
| |
Collapse
|
123
|
Huang LJ, Li G, Ding Y, Sun JH, Wu TT, Zhao W, Zeng YS. LINGO-1 deficiency promotes nerve regeneration through reduction of cell apoptosis, inflammation, and glial scar after spinal cord injury in mice. Exp Neurol 2019; 320:112965. [PMID: 31132364 DOI: 10.1016/j.expneurol.2019.112965] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 05/14/2019] [Accepted: 05/23/2019] [Indexed: 12/16/2022]
Abstract
Leucine-rich repeat and immunoglobulin domain-containing protein 1 (LINGO-1) is a transmembrane protein that negatively regulates neural regeneration in the central nervous system. LINGO-1 expression is up-regulated after central nerve injury, and is accompanied by cell death. Both LINGO-1 and cell death in the injury microenvironment are thought to limit neural regeneration, but the relationship between LINGO-1 and cell death has not been characterized. To investigate whether LINGO-1 deletion improves the spinal cord microenvironment after spinal cord injury (SCI) and contributes to cell survival, we generated LINGO-1 knockout (KO) mice. These mice and wild-type control mice were subjected to spinal cord transection. Fourteen days after spinal cord transection, cell apoptosis, inflammation, glial scar, and growth of nerve fibers were evaluated by immunostaining. The results showed that LINGO-1 KO mice demonstrated a profound reduction in expression of caspase-3, transferase-mediated deoxyuridine triphosphate biotin nick end labeling (TUNEL), ionized calcium binding adapter molecule 1 (IBA1), glial fibrillary acidic protein (GFAP), and chondroitin sulfate proteoglycans (CSPGs) compared to controls. In contrast, expression of neurofilament (NF) at the SCI site in LINGO-1 KO mice was markedly increased compared to that in wild-type mice. These results suggested that LINGO-1 plays a critical role in the injury microenvironment in processes such as cell death, inflammatory response, and glial scar formation. Importantly, LINGO-1 deletion and a positive microenvironment may exert synergistic effects to promote nerve fiber regeneration. Therefore, inhibition of LINGO-1 may be a therapeutic strategy to promote neural regeneration following SCI.
Collapse
Affiliation(s)
- Li-Jun Huang
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ge Li
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ying Ding
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China
| | - Jia-Hui Sun
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Ting-Ting Wu
- Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Wei Zhao
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Yuan-Shan Zeng
- Key Laboratory for Stem Cells and Tissue Engineering (Sun Yat-sen University), Ministry of Education, Guangzhou 510080, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China; Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou 510120, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong 226001, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.
| |
Collapse
|
124
|
McConnell HL, Li Z, Woltjer RL, Mishra A. Astrocyte dysfunction and neurovascular impairment in neurological disorders: Correlation or causation? Neurochem Int 2019; 128:70-84. [PMID: 30986503 DOI: 10.1016/j.neuint.2019.04.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/08/2019] [Accepted: 04/09/2019] [Indexed: 12/14/2022]
Abstract
The neurovascular unit, consisting of neurons, astrocytes, and vascular cells, has become the focus of much discussion in the last two decades and emerging literature now suggests an association between neurovascular dysfunction and neurological disorders. In this review, we synthesize the known and suspected contributions of astrocytes to neurovascular dysfunction in disease. Throughout the brain, astrocytes are centrally positioned to dynamically mediate interactions between neurons and the cerebral vasculature, and play key roles in blood-brain barrier maintenance and neurovascular coupling. It is increasingly apparent that the changes in astrocytes in response to a variety of insults to brain tissue -collectively referred to as "reactive astrogliosis" - are not just an epiphenomenon restricted to morphological alterations, but comprise functional changes in astrocytes that contribute to the phenotype of neurological diseases with both beneficial and detrimental effects. In the context of the neurovascular unit, astrocyte dysfunction accompanies, and may contribute to, blood-brain barrier impairment and neurovascular dysregulation, highlighting the need to determine the exact nature of the relationship between astrocyte dysfunction and neurovascular impairments. Targeting astrocytes may represent a new strategy in combinatorial therapeutics for preventing the mismatch of energy supply and demand that often accompanies neurological disorders.
Collapse
Affiliation(s)
- Heather L McConnell
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, United States
| | - Zhenzhou Li
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, United States; Department of Anesthesiology, General Hospital of Ningxia Medical University, Yinchuan City, China
| | - Randall L Woltjer
- Department of Neuropathology, Oregon Health & Science University, Portland, OR, United States
| | - Anusha Mishra
- Knight Cardiovascular Institute, Oregon Health & Science University, Portland, OR, United States.
| |
Collapse
|
125
|
Alizadeh A, Dyck SM, Karimi-Abdolrezaee S. Traumatic Spinal Cord Injury: An Overview of Pathophysiology, Models and Acute Injury Mechanisms. Front Neurol 2019; 10:282. [PMID: 30967837 PMCID: PMC6439316 DOI: 10.3389/fneur.2019.00282] [Citation(s) in RCA: 635] [Impact Index Per Article: 127.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 03/05/2019] [Indexed: 12/11/2022] Open
Abstract
Traumatic spinal cord injury (SCI) is a life changing neurological condition with substantial socioeconomic implications for patients and their care-givers. Recent advances in medical management of SCI has significantly improved diagnosis, stabilization, survival rate and well-being of SCI patients. However, there has been small progress on treatment options for improving the neurological outcomes of SCI patients. This incremental success mainly reflects the complexity of SCI pathophysiology and the diverse biochemical and physiological changes that occur in the injured spinal cord. Therefore, in the past few decades, considerable efforts have been made by SCI researchers to elucidate the pathophysiology of SCI and unravel the underlying cellular and molecular mechanisms of tissue degeneration and repair in the injured spinal cord. To this end, a number of preclinical animal and injury models have been developed to more closely recapitulate the primary and secondary injury processes of SCI. In this review, we will provide a comprehensive overview of the recent advances in our understanding of the pathophysiology of SCI. We will also discuss the neurological outcomes of human SCI and the available experimental model systems that have been employed to identify SCI mechanisms and develop therapeutic strategies for this condition.
Collapse
Affiliation(s)
- Arsalan Alizadeh
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Spinal Cord Research Center, University of Manitoba, Winnipeg, MB, Canada
| | - Scott Matthew Dyck
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Spinal Cord Research Center, University of Manitoba, Winnipeg, MB, Canada
| | - Soheila Karimi-Abdolrezaee
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Spinal Cord Research Center, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
126
|
Albayar AA, Roche A, Swiatkowski P, Antar S, Ouda N, Emara E, Smith DH, Ozturk AK, Awad BI. Biomarkers in Spinal Cord Injury: Prognostic Insights and Future Potentials. Front Neurol 2019; 10:27. [PMID: 30761068 PMCID: PMC6361789 DOI: 10.3389/fneur.2019.00027] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 01/10/2019] [Indexed: 12/17/2022] Open
Abstract
Spinal Cord Injury (SCI) is a major challenge in Neurotrauma research. Complex pathophysiological processes take place immediately after the injury and later on as the chronic injury develops. Moreover, SCI is usually accompanied by traumatic injuries because the most common modality of injury is road traffic accidents and falls. Patients develop significant permanent neurological deficits that depend on the extent and the location of the injury itself and in time they develop further neurological and body changes that may risk their mere survival. In our review, we explored the recent updates with regards to SCI biomarkers. We observed two methods that may lead to the appearance of biomarkers for SCI. First, during the first few weeks following the injury the Blood Spinal Cord Barrier (BSCB) disruption that releases several neurologic structure components from the injured tissue. These components find their way to Cerebrospinal Fluid (CSF) and the systemic circulation. Also, as the injury develops several components of the pathological process are expressed or released such as in neuroinflammation, apoptosis, reactive oxygen species, and excitotoxicity sequences. Therefore, there is a growing interest in examining any correlations between these components and the degrees or the outcomes of the injury. Additionally, some of the candidate biomarkers are theorized to track the progressive changes of SCI which offers an insight on the patients' prognoses, potential-treatments-outcomes assessment, and monitoring the progression of the complications of chronic SCI such as Pressure Ulcers and urinary dysfunction. An extensive literature review was performed covering literature, published in English, until February 2018 using the Medline/PubMed database. Experimental and human studies were included and titles, PMID, publication year, authors, biomarkers studies, the method of validation, relationship to SCI pathophysiology, and concluded correlation were reported. Potential SCI biomarkers need further validation using clinical studies. The selection of the appropriate biomarker group should be made based on the stage of the injuries, the accompanying trauma and with regards to any surgical, or medical interference that might have been done. Additionally, we suggest testing multiple biomarkers related to the several pathological changes coinciding to offer a more precise prediction of the outcome.
Collapse
Affiliation(s)
- Ahmed A. Albayar
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, PA, United States
| | - Abigail Roche
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, PA, United States
| | - Przemyslaw Swiatkowski
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, PA, United States
| | - Sarah Antar
- Department of Medical Biochemistry, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Nouran Ouda
- Department of Neurosurgery, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Eman Emara
- Department of Neurosurgery, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Douglas H. Smith
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, PA, United States
| | - Ali K. Ozturk
- Department of Neurosurgery, Penn Center for Brain Injury and Repair, University of Pennsylvania, Philadelphia, PA, United States
| | - Basem I. Awad
- Department of Neurosurgery, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
127
|
Luo D, Zhang Y, Yuan X, Pan Y, Yang L, Zhao Y, Zhuo R, Chen C, Peng L, Li W, Jin X, Zhou Y. Oleoylethanolamide inhibits glial activation via moudulating PPARα and promotes motor function recovery after brain ischemia. Pharmacol Res 2019; 141:530-540. [PMID: 30660821 DOI: 10.1016/j.phrs.2019.01.027] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 12/29/2018] [Accepted: 01/14/2019] [Indexed: 02/06/2023]
Abstract
Glial activation and scar formation impede the neurological function recovery after cerebral ischemia. Oleoylethanolamide (OEA), a bioactive lipid mediator, shows neuroprotection against acute brain ischemia, however, its long-term effect, especially on glial scar formation, has not been characterized. In this research, we investigate the effect of OEA on glial activation and scar formation after cerebral ischemia in vitro and in vivo experiments. Glial scar formation in vitro model was induced by transforming growth factor β1 (TGF-β1) in C6 glial cell culture, and experiment model in vivo was induced by middle cerebral artery occlusion (MCAO) in mice. The protein expressions of the markers of glial activation (S100β, GFAP, or pSmads) and glial scar (neurocan) were detected by Western blot and/or immunofluorescence staining; To evaluate the role of PPARɑ in the effect of OEA on glial activation, the PPARɑ antagonist GW6471 was used. Behavior tests were used to assay the effect of OEA on motor function recovery 14 days after brain ischemia in mice. Our results show that OEA (10-50 μM) concentration-dependently inhibited the upregulation of S100β, GFAP, pSmads and neurocan induced by TGF-β1 in C6 glial cells. At the same time, OEA promoted the protein expression and nuclear transportation of PPARɑ in glial cells. PPARα antagonist GW6471 abolished the effect of OEA on glial activation. In addition, we found that delay administration of OEA inhibited the astrocyte activation and promoted the recovery of motor function after brain ischemia in mice. These results indicate that OEA may be developed into a new candidate for attenuating astrocytic scar formation and improving motor function after ischemic stroke.
Collapse
Affiliation(s)
- Doudou Luo
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China
| | - Yali Zhang
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China; Medical College, Xuchang University, Xuchang, 461000, PR China
| | - Xiaoqian Yuan
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China
| | - Yilin Pan
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China
| | - Lichao Yang
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China
| | - Yun Zhao
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China
| | - Rengong Zhuo
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China
| | - Caixia Chen
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China
| | - Lu Peng
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China
| | - Wenjun Li
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China
| | - Xin Jin
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China.
| | - Yu Zhou
- Department of Basic Medical Science, School of Medicine, Xiamen University, Xiamen, 361102, PR China; Key Laboratory of Chiral Drugs, Xiamen, 361102, PR China.
| |
Collapse
|
128
|
Pavlou MAS, Grandbarbe L, Buckley NJ, Niclou SP, Michelucci A. Transcriptional and epigenetic mechanisms underlying astrocyte identity. Prog Neurobiol 2018; 174:36-52. [PMID: 30599178 DOI: 10.1016/j.pneurobio.2018.12.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 11/20/2018] [Accepted: 12/28/2018] [Indexed: 12/14/2022]
Abstract
Astrocytes play a significant role in coordinating neural development and provide critical support for the function of the CNS. They possess important adaptation capacities that range from their transition towards reactive astrocytes to their ability to undergo reprogramming, thereby revealing their potential to retain latent features of neural progenitor cells. We propose that the mechanisms underlying reactive astrogliosis or astrocyte reprogramming provide an opportunity for initiating neuronal regeneration, a process that is notably reduced in the mammalian nervous system throughout evolution. Conversely, this plasticity may also affect normal astrocytic functions resulting in pathologies ranging from neurodevelopmental disorders to neurodegenerative diseases and brain tumors. We postulate that epigenetic mechanisms linking extrinsic cues and intrinsic transcriptional programs are key factors to maintain astrocyte identity and function, and critically, to control the balance of regenerative and degenerative activity. Here, we will review the main evidences supporting this concept. We propose that unravelling the epigenetic and transcriptional mechanisms underlying the acquisition of astrocyte identity and plasticity, as well as understanding how these processes are modulated by the local microenvironment under specific threatening or pathological conditions, may pave the way to new therapeutic avenues for several neurological disorders including neurodegenerative diseases and brain tumors of astrocytic lineage.
Collapse
Affiliation(s)
- Maria Angeliki S Pavlou
- Life Sciences Research Unit, University of Luxembourg, Esch-sur-Alzette, Luxembourg; NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg
| | - Luc Grandbarbe
- Life Sciences Research Unit, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Noel J Buckley
- Department of Psychiatry, Warneford Hospital, University of Oxford, Oxford, United Kingdom
| | - Simone P Niclou
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg; KG Jebsen Brain Tumour Research Center, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Alessandro Michelucci
- NORLUX Neuro-Oncology Laboratory, Department of Oncology, Luxembourg Institute of Health, Luxembourg, Luxembourg; Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-Belval, Luxembourg.
| |
Collapse
|
129
|
Piers TM, East E, Villegas-Llerena C, Sevastou IG, Matarin M, Hardy J, Pocock JM. Soluble Fibrinogen Triggers Non-cell Autonomous ER Stress-Mediated Microglial-Induced Neurotoxicity. Front Cell Neurosci 2018; 12:404. [PMID: 30524237 PMCID: PMC6257202 DOI: 10.3389/fncel.2018.00404] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 10/19/2018] [Indexed: 01/08/2023] Open
Abstract
Aberrant or chronic microglial activation is strongly implicated in neurodegeneration, where prolonged induction of classical inflammatory pathways may lead to a compromised blood-brain barrier (BBB) or vasculature, features of many neurodegenerative disorders and implicated in the observed cognitive decline. BBB disruption or vascular disease may expose the brain parenchyma to “foreign” plasma proteins which subsequently impact on neuronal network integrity through neurotoxicity, synaptic loss and the potentiation of microglial inflammation. Here we show that the blood coagulation factor fibrinogen (FG), implicated in the pathogenesis of dementias such as Alzheimer’s disease (AD), induces an inflammatory microglial phenotype as identified through genetic microarray analysis of a microglial cell line, and proteome cytokine profiling of primary microglia. We also identify a FG-mediated induction of non-cell autonomous ER stress-associated neurotoxicity via a signaling pathway that can be blocked by pharmacological inhibition of microglial TNFα transcription or neuronal caspase-12 activity, supporting a disease relevant role for plasma components in neuronal dysfunction.
Collapse
Affiliation(s)
- Thomas M Piers
- Cell Signalling Laboratory, Department of Neuroinflammation, Institute of Neurology, University College London, London, United Kingdom
| | - Emma East
- Cell Signalling Laboratory, Department of Neuroinflammation, Institute of Neurology, University College London, London, United Kingdom
| | - Claudio Villegas-Llerena
- Cell Signalling Laboratory, Department of Neuroinflammation, Institute of Neurology, University College London, London, United Kingdom.,Department of Molecular Neuroscience, Institute of Neurology, University College London, London, United Kingdom
| | - Ioanna G Sevastou
- Cell Signalling Laboratory, Department of Neuroinflammation, Institute of Neurology, University College London, London, United Kingdom
| | - Mar Matarin
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, United Kingdom.,Department of Neuropsychology, National Hospital for Neurology and Neurosurgery, University College London Hospitals, London, United Kingdom
| | - John Hardy
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, United Kingdom
| | - Jennifer M Pocock
- Cell Signalling Laboratory, Department of Neuroinflammation, Institute of Neurology, University College London, London, United Kingdom
| |
Collapse
|
130
|
Wang F, Cao Y, Ma L, Pei H, Rausch WD, Li H. Dysfunction of Cerebrovascular Endothelial Cells: Prelude to Vascular Dementia. Front Aging Neurosci 2018; 10:376. [PMID: 30505270 PMCID: PMC6250852 DOI: 10.3389/fnagi.2018.00376] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 10/29/2018] [Indexed: 12/19/2022] Open
Abstract
Vascular dementia (VaD) is the second most common type of dementia after Alzheimer's disease (AD), characterized by progressive cognitive impairment, memory loss, and thinking or speech problems. VaD is usually caused by cerebrovascular disease, during which, cerebrovascular endothelial cells (CECs) are vulnerable. CEC dysfunction occurs before the onset of VaD and can eventually lead to dysregulation of cerebral blood flow and blood-brain barrier damage, followed by the activation of glia and inflammatory environment in the brain. White matter, neuronal axons, and synapses are compromised in this process, leading to cognitive impairment. The present review summarizes the mechanisms underlying CEC impairment during hypoperfusion and pathological role of CECs in VaD. Through the comprehensive examination and summarization, endothelial nitric oxide synthase (eNOS)/nitric oxide (NO) signaling pathway, Ras homolog gene family member A (RhoA) signaling pathway, and CEC-derived caveolin-1 (CAV-1) are proposed to serve as targets of new drugs for the treatment of VaD.
Collapse
Affiliation(s)
- Feixue Wang
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Yu Cao
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Lina Ma
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Hui Pei
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Wolf Dieter Rausch
- Department for Biomedical Sciences, Institute of Medical Biochemistry, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Hao Li
- Department of Geriatrics, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
131
|
Tran AP, Warren PM, Silver J. The Biology of Regeneration Failure and Success After Spinal Cord Injury. Physiol Rev 2018. [PMID: 29513146 DOI: 10.1152/physrev.00017.2017] [Citation(s) in RCA: 513] [Impact Index Per Article: 85.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Since no approved therapies to restore mobility and sensation following spinal cord injury (SCI) currently exist, a better understanding of the cellular and molecular mechanisms following SCI that compromise regeneration or neuroplasticity is needed to develop new strategies to promote axonal regrowth and restore function. Physical trauma to the spinal cord results in vascular disruption that, in turn, causes blood-spinal cord barrier rupture leading to hemorrhage and ischemia, followed by rampant local cell death. As subsequent edema and inflammation occur, neuronal and glial necrosis and apoptosis spread well beyond the initial site of impact, ultimately resolving into a cavity surrounded by glial/fibrotic scarring. The glial scar, which stabilizes the spread of secondary injury, also acts as a chronic, physical, and chemo-entrapping barrier that prevents axonal regeneration. Understanding the formative events in glial scarring helps guide strategies towards the development of potential therapies to enhance axon regeneration and functional recovery at both acute and chronic stages following SCI. This review will also discuss the perineuronal net and how chondroitin sulfate proteoglycans (CSPGs) deposited in both the glial scar and net impede axonal outgrowth at the level of the growth cone. We will end the review with a summary of current CSPG-targeting strategies that help to foster axonal regeneration, neuroplasticity/sprouting, and functional recovery following SCI.
Collapse
Affiliation(s)
- Amanda Phuong Tran
- Department of Neurosciences, Case Western Reserve University , Cleveland, Ohio ; and School of Biomedical Sciences, University of Leeds , Leeds , United Kingdom
| | - Philippa Mary Warren
- Department of Neurosciences, Case Western Reserve University , Cleveland, Ohio ; and School of Biomedical Sciences, University of Leeds , Leeds , United Kingdom
| | - Jerry Silver
- Department of Neurosciences, Case Western Reserve University , Cleveland, Ohio ; and School of Biomedical Sciences, University of Leeds , Leeds , United Kingdom
| |
Collapse
|
132
|
Clark VD, Layson A, Charkviani M, Muradashvili N, Lominadze D. Hyperfibrinogenemia-mediated astrocyte activation. Brain Res 2018; 1699:158-165. [PMID: 30153459 DOI: 10.1016/j.brainres.2018.08.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 08/10/2018] [Accepted: 08/22/2018] [Indexed: 12/26/2022]
Abstract
Fibrinogen (Fg)-containing plaques are associated with memory loss during various inflammatory neurodegenerative diseases such as Alzheimer's disease, multiple sclerosis, stroke, and traumatic brain injury. However, mechanisms of its action in neurovascular unit are not clear. As Fg is a high molecular weight blood protein and cannot translocate far from the vessel after extravasation, we hypothesized that it may interact with astrocytes first causing their activation. Cultured mouse cortical astrocytes were treated with Fg in the presence or absence of function-blocking anti-mouse intercellular adhesion molecule 1 (ICAM-1) antibody, or with medium alone (control). Expressions of ICAM-1 and tyrosine receptor kinase B (TrkB) as markers of astrocyte activation, and phosphorylation of TrkB (pTrkB) were assessed. Fg dose-dependently increased activation of astrocytes defined by their shape change, retraction of processes, and enhanced expressions of ICAM-1 and TrkB, and increased pTrkB. Blocking of ICAM-1 function ameliorated these Fg effects. Data suggest that Fg interacts with astrocytes causing overexpression of ICAM-1 and TrkB, and TrkB phosphorylation, and thus, astrocyte activation. Since TrkB is known to be involved in neurodegeneration, interaction of Fg with astrocytes and the resultant activation of TrkB can be a possible mechanism involved in memory reduction, which were observed in previous studies and were associated with formation of complexes of Fg deposited in extravascular space with proteins such as Amyloid beta or prion, the proteins involved in development of dementia.
Collapse
Affiliation(s)
- Vincent D Clark
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Ailey Layson
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Mariam Charkviani
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Nino Muradashvili
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA; Department of Basic Medicine, Caucasus International University, Tbilisi, Georgia
| | - David Lominadze
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA; Kentucky Spinal Cord Research Center, University of Louisville, School of Medicine, Louisville, KY, USA.
| |
Collapse
|
133
|
NG2/CSPG4 and progranulin in the posttraumatic glial scar. Matrix Biol 2018; 68-69:571-588. [DOI: 10.1016/j.matbio.2017.10.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 10/05/2017] [Accepted: 10/06/2017] [Indexed: 12/17/2022]
|
134
|
Liu W, Quan P, Li Q, Tang P, Chen J, Jiang T, Cai W. Dextran-based biodegradable nanoparticles: an alternative and convenient strategy for treatment of traumatic spinal cord injury. Int J Nanomedicine 2018; 13:4121-4132. [PMID: 30038493 PMCID: PMC6049602 DOI: 10.2147/ijn.s171925] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Introduction After traumatic spinal cord injury (SCI), an inhibitory environment that contains chondroitin sulfate proteoglycans (CSPGs) is formed that prevents axonal regeneration and growth. Materials and methods As previously reported, local administration of Taxol® at a low concentration has shown promising abilities to promote axonal regeneration and downregulate inhibitory molecules after acute SCI. However, the application of an invasive miniosmotic pump to deliver Taxol and the Cremophor-related toxicity caused by Taxol limits the administration of Taxol. Results In this study, the sustained release of paclitaxel (PTX) for 7 days was achieved by incorporating PTX into acetalated dextran (Ac-DEX) nanoparticles, and the prepared PTX-loaded Ac-DEX (PTX@Ac-DEX) nanoparticles promoted neurite extension in the presence of CSPGs. In a rat SCI model, both PTX@Ac-DEX and Taxol enhanced neural regeneration, inhibited CSPGs, protected the injured spinal cord, and improved locomotor recovery. Because of the sustained release of PTX, single administration of PTX@Ac-DEX showed equal therapeutic effect with Taxol, which need to be administered for seven days using a surgically implanted miniosmotic pump. Conclusion Overall, this study provides an effective and convenient strategy for SCI therapy, which can improve neurite extension across an inhibitory environment and avoid Cremophor-related toxicity caused by Taxol.
Collapse
Affiliation(s)
- Wei Liu
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China,
| | - Peng Quan
- Department of Pharmaceutical Science, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Qingqing Li
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China,
| | - Pengyu Tang
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China,
| | - Jian Chen
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China,
| | - Tao Jiang
- Department of Orthopaedics, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu 214023, China
| | - Weihua Cai
- Department of Orthopaedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu 210029, China,
| |
Collapse
|
135
|
Role of Damage Associated Molecular Pattern Molecules (DAMPs) in Aneurysmal Subarachnoid Hemorrhage (aSAH). Int J Mol Sci 2018; 19:ijms19072035. [PMID: 30011792 PMCID: PMC6073937 DOI: 10.3390/ijms19072035] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 07/01/2018] [Accepted: 07/09/2018] [Indexed: 12/27/2022] Open
Abstract
Aneurysmal subarachnoid hemorrhage (aSAH) represents only a small portion of all strokes, but accounts for almost half of the deaths caused by stroke worldwide. Neurosurgical clipping and endovascular coiling can successfully obliterate the bleeding aneurysms, but ensuing complications such as cerebral vasospasm, acute and chronic hydrocephalus, seizures, cortical spreading depression, delayed ischemic neurological deficits, and delayed cerebral ischemia lead to poor clinical outcomes. The mechanisms leading to these complications are complex and poorly understood. Early brain injury resulting from transient global ischemia can release molecules that may be critical to initiate and sustain inflammatory response. Hence, the events during early brain injury can influence the occurrence of delayed brain injury. Since the damage associated molecular pattern molecules (DAMPs) might be the initiators of inflammation in the pathophysiology of aSAH, so the aim of this review is to highlight their role in the context of aSAH from diagnostic, prognostic, therapeutic, and drug therapy monitoring perspectives. DAMPs represent a diverse and a heterogenous group of molecules derived from different compartments of cells upon injury. Here, we have reviewed the most important DAMPs molecules including high mobility group box-1 (HMGB1), S100B, hemoglobin and its derivatives, extracellular matrix components, IL-1α, IL-33, and mitochondrial DNA in the context of aSAH and their role in post-aSAH complications and clinical outcome after aSAH.
Collapse
|
136
|
Boas SEM, Carvalho J, van den Broek M, Weijers EM, Goumans MJ, Koolwijk P, Merks RMH. A local uPAR-plasmin-TGFβ1 positive feedback loop in a qualitative computational model of angiogenic sprouting explains the in vitro effect of fibrinogen variants. PLoS Comput Biol 2018; 14:e1006239. [PMID: 29979675 PMCID: PMC6072121 DOI: 10.1371/journal.pcbi.1006239] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 08/02/2018] [Accepted: 05/28/2018] [Indexed: 11/19/2022] Open
Abstract
In experimental assays of angiogenesis in three-dimensional fibrin matrices, a temporary scaffold formed during wound healing, the type and composition of fibrin impacts the level of sprouting. More sprouts form on high molecular weight (HMW) than on low molecular weight (LMW) fibrin. It is unclear what mechanisms regulate the number and the positions of the vascular-like structures in cell cultures. To address this question, we propose a mechanistic simulation model of endothelial cell migration and fibrin proteolysis by the plasmin system. The model is a hybrid, cell-based and continuum, computational model based on the cellular Potts model and sets of partial-differential equations. Based on the model results, we propose that a positive feedback mechanism between uPAR, plasmin and transforming growth factor β1 (TGFβ1) selects cells in the monolayer for matrix invasion. Invading cells releases TGFβ1 from the extracellular matrix through plasmin-mediated fibrin degradation. The activated TGFβ1 further stimulates fibrin degradation and keeps proteolysis active as the sprout invades the fibrin matrix. The binding capacity for TGFβ1 of LMW is reduced relative to that of HMW. This leads to reduced activation of proteolysis and, consequently, reduced cell ingrowth in LMW fibrin compared to HMW fibrin. Thus our model predicts that endothelial cells in LMW fibrin matrices compared to HMW matrices show reduced sprouting due to a lower bio-availability of TGFβ1. Therapies for a range of medical conditions, including cancer, wound healing and diabetic retinopathy can benefit from a better control over the growth of blood vessels. The chemical properties of fibrin, the material that forms scabs in wounds and can also occur in large concentrations in tumors, can regulate the degree of blood vessel growth (angiogenesis). Angiogenesis can be mimicked in cell cultures. These allow us to modulate the chemical properties of fibrin and study the effect on angiogenesis. Fibrin occurs in high molecular weight (HMW) and in low molecular weight (LMW) forms. Interestingly, there is more ingrowth of angiogenic-like structures into HMW than in LMW fibrin, but the mechanisms are poorly understood. To get more insight into these, we constructed a computational model. Using the model, we propose and analyse a hypothetical mechanism for sprouting that could explain the differences in endothelial cell sprouting in LMW and HMW fibrin matrices. Our model suggests that cells digest fibrin, thus creating space for ingrowth. At the same time, digestion frees growth factors bound to fibrin, that activates further secretion of digestive enzymes by the cells. We propose that the resulting positive feedback loop spontaneously selects cells in the endothelial monolayer for ingrowth and helps the blood vessel sprout move deeper into the fibrin. This could be a complementary mechanism to lateral-inhibition by Delta-Notch for the selection of leader cells, also called ‘tip cells’. Our model predicts that endothelial cells in LMW fibrin compared to HMW fibrin show reduced sprouting due to a lower bio-availability of TGFβ1.
Collapse
Affiliation(s)
- Sonja E. M. Boas
- Centrum Wiskunde & Informatica (CWI), Amsterdam, The Netherlands
- Mathematical Institute, Leiden University, Leiden, The Netherlands
| | - Joao Carvalho
- Centrum Wiskunde & Informatica (CWI), Amsterdam, The Netherlands
- CFisUC, Department of Physics, University of Coimbra, Coimbra, Portugal
| | - Marloes van den Broek
- Amsterdam Cardiovascular Sciences, VU University medical Center, Dept. of Physiology, Amsterdam, The Netherlands
| | - Ester M. Weijers
- Amsterdam Cardiovascular Sciences, VU University medical Center, Dept. of Physiology, Amsterdam, The Netherlands
| | - Marie-José Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Pieter Koolwijk
- Amsterdam Cardiovascular Sciences, VU University medical Center, Dept. of Physiology, Amsterdam, The Netherlands
| | - Roeland M. H. Merks
- Centrum Wiskunde & Informatica (CWI), Amsterdam, The Netherlands
- Mathematical Institute, Leiden University, Leiden, The Netherlands
- * E-mail:
| |
Collapse
|
137
|
To Be or Not to Be: Environmental Factors that Drive Myelin Formation during Development and after CNS Trauma. ACTA ACUST UNITED AC 2018. [DOI: 10.3390/neuroglia1010007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Oligodendrocytes are specialized glial cells that myelinate central nervous system (CNS) axons. Historically, it was believed that the primary role of myelin was to compactly ensheath axons, providing the insulation necessary for rapid signal conduction. However, mounting evidence demonstrates the dynamic importance of myelin and oligodendrocytes, including providing metabolic support to neurons and regulating axon protein distribution. As such, the development and maintenance of oligodendrocytes and myelin are integral to preserving CNS homeostasis and supporting proper functioning of widespread neural networks. Environmental signals are critical for proper oligodendrocyte lineage cell progression and their capacity to form functional compact myelin; these signals are markedly disturbed by injury to the CNS, which may compromise endogenous myelin repair capabilities. This review outlines some key environmental factors that drive myelin formation during development and compares that to the primary factors that define a CNS injury milieu. We aim to identify developmental factors disrupted after CNS trauma as well as pathogenic factors that negatively impact oligodendrocyte lineage cells, as these are potential therapeutic targets to promote myelin repair after injury or disease.
Collapse
|
138
|
Xiong Y, Mahmood A, Chopp M. Current understanding of neuroinflammation after traumatic brain injury and cell-based therapeutic opportunities. Chin J Traumatol 2018; 21:137-151. [PMID: 29764704 PMCID: PMC6034172 DOI: 10.1016/j.cjtee.2018.02.003] [Citation(s) in RCA: 130] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 03/02/2018] [Accepted: 03/05/2018] [Indexed: 02/04/2023] Open
Abstract
Traumatic brain injury (TBI) remains a major cause of death and disability worldwide. Increasing evidence indicates that TBI is an important risk factor for neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, and chronic traumatic encephalopathy. Despite improved supportive and rehabilitative care of TBI patients, unfortunately, all late phase clinical trials in TBI have yet to yield a safe and effective neuroprotective treatment. The disappointing clinical trials may be attributed to variability in treatment approaches and heterogeneity of the population of TBI patients as well as a race against time to prevent or reduce inexorable cell death. TBI is not just an acute event but a chronic disease. Among many mechanisms involved in secondary injury after TBI, emerging preclinical studies indicate that posttraumatic prolonged and progressive neuroinflammation is associated with neurodegeneration which may be treatable long after the initiating brain injury. This review provides an overview of recent understanding of neuroinflammation in TBI and preclinical cell-based therapies that target neuroinflammation and promote functional recovery after TBI.
Collapse
Affiliation(s)
- Ye Xiong
- Department of Neurosurgery Henry Ford Health System, 2799 West Grand Boulevard, Detroit, MI, 48202, USA.
| | - Asim Mahmood
- Department of Neurosurgery Henry Ford Health System, 2799 West Grand Boulevard, Detroit, MI, 48202, USA
| | - Michael Chopp
- Department of Neurology, Henry Ford Health System, 2799 West Grand Boulevard, Detroit, MI, 48202, USA; Department of Physics, Oakland University, Rochester, MI, 48309, USA
| |
Collapse
|
139
|
Modulation of Receptor Protein Tyrosine Phosphatase Sigma Increases Chondroitin Sulfate Proteoglycan Degradation through Cathepsin B Secretion to Enhance Axon Outgrowth. J Neurosci 2018; 38:5399-5414. [PMID: 29760175 DOI: 10.1523/jneurosci.3214-17.2018] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 05/03/2018] [Accepted: 05/09/2018] [Indexed: 12/22/2022] Open
Abstract
Severed axon tips reform growth cones following spinal cord injury that fail to regenerate, in part, because they become embedded within an inhibitory extracellular matrix. Chondroitin sulfate proteoglycans (CSPGs) are the major axon inhibitory matrix component that is increased within the lesion scar and in perineuronal nets around deafferented neurons. We have recently developed a novel peptide modulator (intracellular sigma peptide) of the cognate receptor of CSPGs, protein tyrosine phosphatase σ (RPTPσ), which has been shown to markedly improve sensorimotor function, micturition, and coordinated locomotor behavior in spinal cord contused rats. However, the mechanism(s) underlying how modulation of RPTPσ mediates axon outgrowth through inhibitory CSPGs remain unclear. Here, we describe how intracellular sigma peptide modulation of RPTPσ induces enhanced protease Cathepsin B activity. Using DRG neurons from female Sprague Dawley rats cultured on an aggrecan/laminin spot assay and a combination of biochemical techniques, we provide evidence suggesting that modulation of RPTPσ regulates secretion of proteases that, in turn, relieves CSPG inhibition through its digestion to allow axon migration though proteoglycan barriers. Understanding the mechanisms underlying RPTPσ modulation elucidates how axon regeneration is impaired by proteoglycans but can then be facilitated following injury.SIGNIFICANCE STATEMENT Following spinal cord injury, chondroitin sulfate proteoglycans (CSPGs) upregulate and potently inhibit axon regeneration and functional recovery. Protein tyrosine phosphatase σ (RPTPσ) has been identified as a critical cognate receptor of CSPGs. We have previously characterized a synthetic peptide (intracellular sigma peptide) that targets the regulatory intracellular domain of the receptor to allow axons to regenerate despite the presence of CSPGs. Here, we have found that one important mechanism by which peptide modulation of the receptor enhances axon outgrowth is through secretion of a protease, Cathepsin B, which enables digestion of CSPGs. This work links protease secretion to the CSPG receptor RPTPσ for the first time with implications for understanding the molecular mechanisms underlying neural regeneration and plasticity.
Collapse
|
140
|
Johnson VE, Weber MT, Xiao R, Cullen DK, Meaney DF, Stewart W, Smith DH. Mechanical disruption of the blood-brain barrier following experimental concussion. Acta Neuropathol 2018; 135:711-726. [PMID: 29460006 DOI: 10.1007/s00401-018-1824-0] [Citation(s) in RCA: 95] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/09/2018] [Accepted: 02/10/2018] [Indexed: 12/14/2022]
Abstract
Although concussion is now recognized as a major health issue, its non-lethal nature has limited characterization of the underlying pathophysiology. In particular, potential neuropathological changes have typically been inferred from non-invasive techniques or post-mortem examinations of severe traumatic brain injury (TBI). Here, we used a swine model of head rotational acceleration based on human concussion to examine blood-brain barrier (BBB) integrity after injury in association with diffuse axonal injury and glial responses. We then determined the potential clinical relevance of the swine concussion findings through comparisons with pathological changes in human severe TBI, where post-mortem examinations are possible. At 6-72 h post-injury in swine, we observed multifocal disruption of the BBB, demonstrated by extravasation of serum proteins, fibrinogen and immunoglobulin-G, in the absence of hemorrhage or other focal pathology. BBB disruption was observed in a stereotyped distribution consistent with biomechanical insult. Specifically, extravasated serum proteins were frequently observed at interfaces between regions of tissue with differing material properties, including the gray-white boundary, periventricular and subpial regions. In addition, there was substantial overlap of BBB disruption with regions of axonal pathology in the white matter. Acute perivascular cellular uptake of blood-borne proteins was observed to be prominent in astrocytes (GFAP-positive) and neurons (MAP-2-positive), but not microglia (IBA1-positive). Parallel examination of human severe TBI revealed similar patterns of serum extravasation and glial uptake of serum proteins, but to a much greater extent than in the swine model, attributed to the higher injury severity. These data suggest that BBB disruption represents a new and important pathological feature of concussion.
Collapse
|
141
|
Petersen MA, Ryu JK, Akassoglou K. Fibrinogen in neurological diseases: mechanisms, imaging and therapeutics. Nat Rev Neurosci 2018; 19:283-301. [PMID: 29618808 PMCID: PMC6743980 DOI: 10.1038/nrn.2018.13] [Citation(s) in RCA: 293] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The blood coagulation protein fibrinogen is deposited in the brain in a wide range of neurological diseases and traumatic injuries with blood-brain barrier (BBB) disruption. Recent research has uncovered pleiotropic roles for fibrinogen in the activation of CNS inflammation, induction of scar formation in the brain, promotion of cognitive decline and inhibition of repair. Such diverse roles are possible in part because of the unique structure of fibrinogen, which contains multiple binding sites for cellular receptors and proteins expressed in the nervous system. The cellular and molecular mechanisms underlying the actions of fibrinogen are beginning to be elucidated, providing insight into its involvement in neurological diseases, such as multiple sclerosis, Alzheimer disease and traumatic CNS injury. Selective drug targeting to suppress the damaging functions of fibrinogen in the nervous system without affecting its beneficial effects in haemostasis opens a new fibrinogen therapeutics pipeline for neurological disease.
Collapse
Affiliation(s)
- Mark A. Petersen
- Gladstone Institutes, San Francisco, CA USA
- Division of Neonatology, Department of Pediatrics, University of California, San Francisco, CA, USA
| | | | - Katerina Akassoglou
- Gladstone Institutes, San Francisco, CA USA
- Department of Neurology, University of California, San Francisco, CA, USA
| |
Collapse
|
142
|
Frik J, Merl-Pham J, Plesnila N, Mattugini N, Kjell J, Kraska J, Gómez RM, Hauck SM, Sirko S, Götz M. Cross-talk between monocyte invasion and astrocyte proliferation regulates scarring in brain injury. EMBO Rep 2018; 19:embr.201745294. [PMID: 29632244 PMCID: PMC5934774 DOI: 10.15252/embr.201745294] [Citation(s) in RCA: 79] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 03/02/2018] [Accepted: 03/09/2018] [Indexed: 01/02/2023] Open
Abstract
Scar formation after brain injury is still poorly understood. To further elucidate such processes, here, we examine the interplay between astrocyte proliferation taking place predominantly at the vascular interface and monocyte invasion. Using genetic mouse models that decrease or increase reactive astrocyte proliferation, we demonstrate inverse effects on monocyte numbers in the injury site. Conversely, reducing monocyte invasion using CCR2-/- mice causes a strong increase in astrocyte proliferation, demonstrating an intriguing negative cross-regulation between these cell types at the vascular interface. CCR2-/- mice show reduced scar formation with less extracellular matrix deposition, smaller lesion site and increased neuronal coverage. Surprisingly, the GFAP+ scar area in these mice is also significantly decreased despite increased astrocyte proliferation. Proteomic analysis at the peak of increased astrocyte proliferation reveals a decrease in extracellular matrix synthesizing enzymes in the injury sites of CCR2-/- mice, highlighting how early key aspects of scar formation are initiated. Taken together, we provide novel insights into the cross-regulation of juxtavascular proliferating astrocytes and invading monocytes as a crucial mechanism of scar formation upon brain injury.
Collapse
Affiliation(s)
- Jesica Frik
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University Munich, Munich, Germany.,Institute for Stem Cell Research, Helmholtz Center Munich, Munich, Germany.,Instituto de Biotecnología y Biología Molecular, UNLP-CONICET, La Plata, Argentina
| | - Juliane Merl-Pham
- Research Unit for Protein Science, Helmholtz Center Munich, Munich, Germany
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research, Experimental Stroke Research, University of Munich Medical School, Munich, Germany.,SYNERGY, Excellence Cluster Systems Neurology, University of Munich, Munich, Germany
| | - Nicola Mattugini
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University Munich, Munich, Germany.,Institute for Stem Cell Research, Helmholtz Center Munich, Munich, Germany.,Graduate School of Systemic Neurosciences, Biocenter, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Jacob Kjell
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University Munich, Munich, Germany.,Institute for Stem Cell Research, Helmholtz Center Munich, Munich, Germany
| | - Jonas Kraska
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Ricardo M Gómez
- Instituto de Biotecnología y Biología Molecular, UNLP-CONICET, La Plata, Argentina
| | - Stefanie M Hauck
- Research Unit for Protein Science, Helmholtz Center Munich, Munich, Germany
| | - Swetlana Sirko
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University Munich, Munich, Germany .,Institute for Stem Cell Research, Helmholtz Center Munich, Munich, Germany
| | - Magdalena Götz
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians-University Munich, Munich, Germany .,Institute for Stem Cell Research, Helmholtz Center Munich, Munich, Germany.,SYNERGY, Excellence Cluster Systems Neurology, University of Munich, Munich, Germany
| |
Collapse
|
143
|
The extracellular matrix: Focus on oligodendrocyte biology and targeting CSPGs for remyelination therapies. Glia 2018; 66:1809-1825. [DOI: 10.1002/glia.23333] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 03/06/2018] [Accepted: 03/06/2018] [Indexed: 12/31/2022]
|
144
|
Li M, Sirko S. Traumatic Brain Injury: At the Crossroads of Neuropathology and Common Metabolic Endocrinopathies. J Clin Med 2018. [PMID: 29538298 PMCID: PMC5867585 DOI: 10.3390/jcm7030059] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Building on the seminal work by Geoffrey Harris in the 1970s, the neuroendocrinology field, having undergone spectacular growth, has endeavored to understand the mechanisms of hormonal connectivity between the brain and the rest of the body. Given the fundamental role of the brain in the orchestration of endocrine processes through interactions among neurohormones, it is thus not surprising that the structural and/or functional alterations following traumatic brain injury (TBI) can lead to endocrine changes affecting the whole organism. Taking into account that systemic hormones also act on the brain, modifying its structure and biochemistry, and can acutely and chronically affect several neurophysiological endpoints, the question is to what extent preexisting endocrine dysfunction may set the stage for an adverse outcome after TBI. In this review, we provide an overview of some aspects of three common metabolic endocrinopathies, e.g., diabetes mellitus, obesity, and thyroid dysfunction, and how these could be triggered by TBI. In addition, we discuss how the complex endocrine networks are woven into the responses to sudden changes after TBI, as well as some of the potential mechanisms that, separately or synergistically, can influence outcomes after TBI.
Collapse
Affiliation(s)
- Melanie Li
- Physiological Genomics, Biomedical Center (BMC), Institute of Physiology, Medical Faculty of the Ludwig-Maximilian University Munich, 82152 Planegg-Martinsried, Germany.
| | - Swetlana Sirko
- Physiological Genomics, Biomedical Center (BMC), Institute of Physiology, Medical Faculty of the Ludwig-Maximilian University Munich, 82152 Planegg-Martinsried, Germany.
- Institute of Stem Cell Research, Helmholtz Center Munich, Research Center for Environmental Health GmbH, 85764 Neuherberg, Germany.
| |
Collapse
|
145
|
Blanquie O, Bradke F. Cytoskeleton dynamics in axon regeneration. Curr Opin Neurobiol 2018; 51:60-69. [PMID: 29544200 DOI: 10.1016/j.conb.2018.02.024] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 02/22/2018] [Accepted: 02/26/2018] [Indexed: 11/19/2022]
Abstract
Recent years have seen cytoskeleton dynamics emerging as a key player in axon regeneration. The cytoskeleton, in particular microtubules and actin, ensures the growth of neuronal processes and maintains the singular, highly polarized shape of neurons. Following injury, adult central axons are tipped by a dystrophic structure, the retraction bulb, which prevents their regeneration. Abnormal cytoskeleton dynamics are responsible for the formation of this growth-incompetent structure but pharmacologically modulating cytoskeleton dynamics of injured axons can transform this structure into a growth-competent growth cone. The cytoskeleton also drives the migration of scar-forming cells after an injury. Targeting its dynamics modifies the composition of the inhibitory environment formed by scar tissue and renders it more permissive for regenerating axons. Hence, cytoskeleton dynamics represent an appealing target to promote axon regeneration. As some of cytoskeleton-targeting drugs are used in the clinics for other purposes, they hold the promise to be used as a basis for a regenerative therapy after a spinal cord injury.
Collapse
Affiliation(s)
- Oriane Blanquie
- Laboratory for Axon Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Strasse 27, 53127 Bonn, Germany.
| | - Frank Bradke
- Laboratory for Axon Growth and Regeneration, German Center for Neurodegenerative Diseases (DZNE), Sigmund-Freud-Strasse 27, 53127 Bonn, Germany.
| |
Collapse
|
146
|
Masgutov R, Masgutova G, Mukhametova L, Garanina E, Arkhipova SS, Zakirova E, Mukhamedshina YO, Margarita Z, Gilazieva Z, Syromiatnikova V, Mullakhmetova A, Kadyrova G, Nigmetzyanova M, Mikhail S, Igor P, Yagudin R, Rizvanov A. Allogenic Adipose Derived Stem Cells Transplantation Improved Sciatic Nerve Regeneration in Rats: Autologous Nerve Graft Model. Front Pharmacol 2018; 9:86. [PMID: 29559908 PMCID: PMC5845725 DOI: 10.3389/fphar.2018.00086] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 01/25/2018] [Indexed: 01/19/2023] Open
Abstract
We examined the effect of transplantation of allogenic adipose-derived stem cells (ADSCs) with properties of mesenchymal stem cells (MSCs) on posttraumatic sciatic nerve regeneration in rats. We suggested an approach to rat sciatic nerve reconstruction using the nerve from the other leg as a graft. The comparison was that of a critical 10 mm nerve defect repaired by means of autologous nerve grafting versus an identical lesion on the contralateral side. In this experimental model, the same animal acts simultaneously as a test model, and control. Regeneration of the left nerve was enhanced by the use of ADSCs, whereas the right nerve healed under natural conditions. Thus the effects of individual differences were excluded and a result closer to clinical practice obtained. We observed significant destructive changes in the sciatic nerve tissue after surgery which resulted in the formation of combined contractures in knee and ankle joints of both limbs and neurotrophic ulcers only on the right limb. The stimulation of regeneration by ADSCs increased the survival of spinal L5 ganglia neurons by 26.4%, improved sciatic nerve vascularization by 35.68% and increased the number of myelin fibers in the distal nerve by 41.87%. Moreover, we have demonstrated that S100, PMP2, and PMP22 gene expression levels are suppressed in response to trauma as compared to intact animals. We have shown that ADSC-based therapy contributes to significant improvement in the regeneration.
Collapse
Affiliation(s)
- Ruslan Masgutov
- OpenLab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.,Republican Clinical Hospital, Kazan, Russia
| | - Galina Masgutova
- OpenLab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Liliya Mukhametova
- OpenLab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Ekaterina Garanina
- OpenLab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Svetlana S Arkhipova
- OpenLab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Elena Zakirova
- OpenLab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Yana O Mukhamedshina
- OpenLab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.,Department of Histology, Cytology and Embryology, Kazan State Medical University, Kazan, Russia
| | - Zhuravleva Margarita
- OpenLab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Zarema Gilazieva
- OpenLab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Valeriia Syromiatnikova
- OpenLab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Adelya Mullakhmetova
- OpenLab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Gulnaz Kadyrova
- OpenLab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Mariya Nigmetzyanova
- OpenLab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | | | - Pankov Igor
- Department of Traumatology and Orthopedics, Kazan State Medical Academy, Kazan, Russia
| | | | - Albert Rizvanov
- OpenLab "Gene and Cell Technologies", Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| |
Collapse
|
147
|
Montagne A, Nikolakopoulou AM, Zhao Z, Sagare AP, Si G, Lazic D, Barnes SR, Daianu M, Ramanathan A, Go A, Lawson EJ, Wang Y, Mack WJ, Thompson PM, Schneider JA, Varkey J, Langen R, Mullins E, Jacobs RE, Zlokovic BV. Pericyte degeneration causes white matter dysfunction in the mouse central nervous system. Nat Med 2018; 24:326-337. [PMID: 29400711 PMCID: PMC5840035 DOI: 10.1038/nm.4482] [Citation(s) in RCA: 273] [Impact Index Per Article: 45.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 01/04/2018] [Indexed: 02/07/2023]
Abstract
Diffuse white-matter disease associated with small-vessel disease and dementia is prevalent in the elderly. The biological mechanisms, however, remain elusive. Using pericyte-deficient mice, magnetic resonance imaging, viral-based tract-tracing, and behavior and tissue analysis, we found that pericyte degeneration disrupted white-matter microcirculation, resulting in an accumulation of toxic blood-derived fibrin(ogen) deposits and blood-flow reductions, which triggered a loss of myelin, axons and oligodendrocytes. This disrupted brain circuits, leading to white-matter functional deficits before neuronal loss occurs. Fibrinogen and fibrin fibrils initiated autophagy-dependent cell death in oligodendrocyte and pericyte cultures, whereas pharmacological and genetic manipulations of systemic fibrinogen levels in pericyte-deficient, but not control mice, influenced the degree of white-matter fibrin(ogen) deposition, pericyte degeneration, vascular pathology and white-matter changes. Thus, our data indicate that pericytes control white-matter structure and function, which has implications for the pathogenesis and treatment of human white-matter disease associated with small-vessel disease.
Collapse
Affiliation(s)
- Axel Montagne
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Angeliki M. Nikolakopoulou
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Zhen Zhao
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Abhay P. Sagare
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Gabriel Si
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Divna Lazic
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
- Department of Neurobiology, Institute for Biological Research, University of Belgrade, Belgrade, Republic of Serbia
| | - Samuel R. Barnes
- Biological Imaging Center, Beckman Institute, California Institute of Technology, Pasadena, CA 91101, USA
| | - Madelaine Daianu
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, University of Southern California, Marina del Rey, CA 90292, USA
| | - Anita Ramanathan
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Ariel Go
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Erica J. Lawson
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Yaoming Wang
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - William J. Mack
- Department of Neurological Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Paul M. Thompson
- Imaging Genetics Center, Mark & Mary Stevens Neuroimaging & Informatics Institute, University of Southern California, Marina del Rey, CA 90292, USA
| | - Julie A. Schneider
- Alzheimer's Disease Center, Rush University Medical Center, Chicago, IL 60612, USA
| | - Jobin Varkey
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Ralf Langen
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| | - Eric Mullins
- Division of Hematology and Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229-3039
| | - Russell E. Jacobs
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
- Biological Imaging Center, Beckman Institute, California Institute of Technology, Pasadena, CA 91101, USA
| | - Berislav V. Zlokovic
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
148
|
BMP-Responsive Protease HtrA1 Is Differentially Expressed in Astrocytes and Regulates Astrocytic Development and Injury Response. J Neurosci 2018; 38:3840-3857. [PMID: 29483282 DOI: 10.1523/jneurosci.2031-17.2018] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 01/23/2018] [Accepted: 01/29/2018] [Indexed: 11/21/2022] Open
Abstract
Astrocytes perform a wide array of physiological functions, including structural support, ion exchange, and neurotransmitter uptake. Despite this diversity, molecular markers that label subpopulations of astrocytes are limited, and mechanisms that generate distinct astrocyte subtypes remain unclear. Here we identified serine protease high temperature requirement A 1 (HtrA1), a bone morphogenetic protein 4 signaling regulated protein, as a novel marker of forebrain astrocytes, but not of neural stem cells, in adult mice of both sexes. Genetic deletion of HtrA1 during gliogenesis accelerates astrocyte differentiation. In addition, ablation of HtrA1 in cultured astrocytes leads to altered chondroitin sulfate proteoglycan expression and inhibition of neurite extension, along with elevated levels of transforming growth factor-β family proteins. Brain injury induces HtrA1 expression in reactive astrocytes, and loss of HtrA1 leads to an impairment in wound closure accompanied by increased proliferation of endothelial and immune cells. Our findings demonstrate that HtrA1 is differentially expressed in adult mouse forebrain astrocytes, and that HtrA1 plays important roles in astrocytic development and injury response.SIGNIFICANCE STATEMENT Astrocytes, an abundant cell type in the brain, perform a wide array of physiological functions. Although characterized as morphologically and functionally diverse, molecular markers that label astrocyte subtypes or signaling pathways that lead to their diversity remain limited. Here, after examining the expression profile of astrocytes generated in response to bone morphogenetic protein signaling, we identify high temperature requirement A 1 (HtrA1) as an astrocyte-specific marker that is differentially expressed in distinct adult mouse brain regions. HtrA1 is a serine protease that has been linked to cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy, a small blood vessel disease in humans. Understanding the role of HtrA1 during development and after injury will provide insights into how distinct astrocyte populations are generated and their unique roles in injury and disease.
Collapse
|
149
|
Caja L, Tzavlaki K, Dadras MS, Tan EJ, Hatem G, Maturi NP, Morén A, Wik L, Watanabe Y, Savary K, Kamali-Moghaddan M, Uhrbom L, Heldin CH, Moustakas A. Snail regulates BMP and TGFβ pathways to control the differentiation status of glioma-initiating cells. Oncogene 2018; 37:2515-2531. [PMID: 29449696 PMCID: PMC5945579 DOI: 10.1038/s41388-018-0136-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 12/12/2017] [Accepted: 12/28/2017] [Indexed: 12/31/2022]
Abstract
Glioblastoma multiforme is a brain malignancy characterized by high heterogeneity, invasiveness, and resistance to current therapies, attributes related to the occurrence of glioma stem cells (GSCs). Transforming growth factor β (TGFβ) promotes self-renewal and bone morphogenetic protein (BMP) induces differentiation of GSCs. BMP7 induces the transcription factor Snail to promote astrocytic differentiation in GSCs and suppress tumor growth in vivo. We demonstrate that Snail represses stemness in GSCs. Snail interacts with SMAD signaling mediators, generates a positive feedback loop of BMP signaling and transcriptionally represses the TGFB1 gene, decreasing TGFβ1 signaling activity. Exogenous TGFβ1 counteracts Snail function in vitro, and in vivo promotes proliferation and re-expression of Nestin, confirming the importance of TGFB1 gene repression by Snail. In conclusion, novel insight highlights mechanisms whereby Snail differentially regulates the activity of the opposing BMP and TGFβ pathways, thus promoting an astrocytic fate switch and repressing stemness in GSCs.
Collapse
Affiliation(s)
- Laia Caja
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 582, Biomedical Center, Uppsala University, SE-75123, Uppsala, Sweden. .,Ludwig Cancer Research, Science for Life Laboratory, Box 595, Biomedical Center, Uppsala University, SE-75124, Uppsala, Sweden.
| | - Kalliopi Tzavlaki
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 582, Biomedical Center, Uppsala University, SE-75123, Uppsala, Sweden.,Ludwig Cancer Research, Science for Life Laboratory, Box 595, Biomedical Center, Uppsala University, SE-75124, Uppsala, Sweden
| | - Mahsa S Dadras
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 582, Biomedical Center, Uppsala University, SE-75123, Uppsala, Sweden.,Ludwig Cancer Research, Science for Life Laboratory, Box 595, Biomedical Center, Uppsala University, SE-75124, Uppsala, Sweden
| | - E-Jean Tan
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, SE-75185, Uppsala, Sweden
| | - Gad Hatem
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 582, Biomedical Center, Uppsala University, SE-75123, Uppsala, Sweden.,Ludwig Cancer Research, Science for Life Laboratory, Box 595, Biomedical Center, Uppsala University, SE-75124, Uppsala, Sweden
| | - Naga P Maturi
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 582, Biomedical Center, Uppsala University, SE-75123, Uppsala, Sweden.,Ludwig Cancer Research, Science for Life Laboratory, Box 595, Biomedical Center, Uppsala University, SE-75124, Uppsala, Sweden.,Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, SE-75185, Uppsala, Sweden
| | - Anita Morén
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 582, Biomedical Center, Uppsala University, SE-75123, Uppsala, Sweden.,Ludwig Cancer Research, Science for Life Laboratory, Box 595, Biomedical Center, Uppsala University, SE-75124, Uppsala, Sweden
| | - Lotta Wik
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Box 815, Biomedical Center, Uppsala University, SE-75108, Uppsala, Sweden
| | - Yukihide Watanabe
- Ludwig Cancer Research, Science for Life Laboratory, Box 595, Biomedical Center, Uppsala University, SE-75124, Uppsala, Sweden.,Department of Experimental Pathology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Katia Savary
- Ludwig Cancer Research, Science for Life Laboratory, Box 595, Biomedical Center, Uppsala University, SE-75124, Uppsala, Sweden.,UMR CNRS 7369 MEDyC, Université de Reims Champagne Ardenne, Reims, France
| | - Masood Kamali-Moghaddan
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Box 815, Biomedical Center, Uppsala University, SE-75108, Uppsala, Sweden
| | - Lene Uhrbom
- Department of Immunology, Genetics and Pathology, Science for Life Laboratory, Rudbeck Laboratory, Uppsala University, SE-75185, Uppsala, Sweden
| | - Carl-Henrik Heldin
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 582, Biomedical Center, Uppsala University, SE-75123, Uppsala, Sweden.,Ludwig Cancer Research, Science for Life Laboratory, Box 595, Biomedical Center, Uppsala University, SE-75124, Uppsala, Sweden
| | - Aristidis Moustakas
- Department of Medical Biochemistry and Microbiology, Science for Life Laboratory, Box 582, Biomedical Center, Uppsala University, SE-75123, Uppsala, Sweden. .,Ludwig Cancer Research, Science for Life Laboratory, Box 595, Biomedical Center, Uppsala University, SE-75124, Uppsala, Sweden.
| |
Collapse
|
150
|
RGMa mediates reactive astrogliosis and glial scar formation through TGFβ1/Smad2/3 signaling after stroke. Cell Death Differ 2018; 25:1503-1516. [PMID: 29396549 PMCID: PMC6113216 DOI: 10.1038/s41418-018-0058-y] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Revised: 12/22/2017] [Accepted: 12/27/2017] [Indexed: 01/11/2023] Open
Abstract
In response to stroke, astrocytes become reactive astrogliosis and are a major component of a glial scar. This results in the formation of both a physical and chemical (production of chondroitin sulfate proteoglycans) barrier, which prevent neurite regeneration that, in turn, interferes with functional recovery. However, the mechanisms of reactive astrogliosis and glial scar formation are poorly understood. In this work, we hypothesized that repulsive guidance molecule a (RGMa) regulate reactive astrogliosis and glial scar formation. We first found that RGMa was strongly expressed by reactive astrocytes in the glial scar in a rat model of middle cerebral artery occlusion/reperfusion. Genetic or pharmacologic inhibition of RGMa in vivo resulted in a strong reduction of reactive astrogliosis and glial scarring as well as in a pronounced improvement in functional recovery. Furthermore, we showed that transforming growth factor β1 (TGFβ1) stimulated RGMa expression through TGFβ1 receptor activin-like kinase 5 (ALK5) in primary cultured astrocytes. Knockdown of RGMa abrogated key steps of reactive astrogliosis and glial scar formation induced by TGFβ1, including cellular hypertrophy, glial fibrillary acidic protein upregulation, cell migration, and CSPGs secretion. Finally, we demonstrated that RGMa co-immunoprecipitated with ALK5 and Smad2/3. TGFβ1-induced ALK5-Smad2/3 interaction and subsequent phosphorylation of Smad2/3 were impaired by RGMa knockdown. Taken together, we identified that after stroke, RGMa promotes reactive astrogliosis and glial scar formation by forming a complex with ALK5 and Smad2/3 to promote ALK5-Smad2/3 interaction to facilitate TGFβ1/Smad2/3 signaling, thereby inhibiting neurological functional recovery. RGMa may be a new therapeutic target for stroke.
Collapse
|