1
|
Eftekhari R, Ewanchuk BW, Rawji KS, Yates RM, Noorbakhsh F, Kuipers HF, Hollenberg MD. Blockade of Proteinase-Activated Receptor 2 (PAR2) Attenuates Neuroinflammation in Experimental Autoimmune Encephalomyelitis. J Pharmacol Exp Ther 2024; 388:12-22. [PMID: 37699708 DOI: 10.1124/jpet.123.001685] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/17/2023] [Accepted: 08/08/2023] [Indexed: 09/14/2023] Open
Abstract
Proteinase-activated receptor-2 (PAR2), which modulates inflammatory responses, is elevated in the central nervous system in multiple sclerosis (MS) and in its murine model, experimental autoimmune encephalomyelitis (EAE). In PAR2-null mice, disease severity of EAE is markedly diminished. We therefore tested whether inhibiting PAR2 activation in vivo might be a viable strategy for the treatment of MS. Using the EAE model, we show that a PAR2 antagonist, the pepducin palmitoyl-RSSAMDENSEKKRKSAIK-amide (P2pal-18S), attenuates EAE progression by affecting immune cell function. P2pal-18S treatment markedly diminishes disease severity and reduces demyelination, as well as the infiltration of T-cells and macrophages into the central nervous system. Moreover, P2pal-18S decreases granulocyte-macrophage colony-stimulating factor (GM-CSF) production and T-cell activation in cultured splenocytes and prevents macrophage polarization in vitro. We conclude that PAR2 plays a key role in regulating neuroinflammation in EAE and that PAR2 antagonists represent promising therapeutic agents for treating MS and other neuroinflammatory diseases. SIGNIFICANCE STATEMENT: Proteinase-activated receptor-2 modulates inflammatory responses and is increased in multiple sclerosis lesions. We show that the proteinase-activated receptor-2 antagonist palmitoyl-RSSAMDENSEKKRKSAIK-amide reduces disease in the murine experimental autoimmune encephalomyelitis model of multiple sclerosis by inhibiting T-cell and macrophage activation and infiltration into the central nervous system, making it a potential treatment for multiple sclerosis.
Collapse
Affiliation(s)
- Rahil Eftekhari
- Department of Physiology & Pharmacology (R.E., M.D.H.), Department of Medicine (R.E., M.D.H.), Department of Clinical Neurosciences (R.E., K.S.R., H.F.K.), Department of Biochemistry and Molecular Biology (B.W.E., R.M.Y.), Department of Comparative Biology and Experimental Medicine (B.W.E., R.M.Y.), and Department of Cell Biology and Anatomy (H.F.K.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran (R.E., F.N.)
| | - Benjamin W Ewanchuk
- Department of Physiology & Pharmacology (R.E., M.D.H.), Department of Medicine (R.E., M.D.H.), Department of Clinical Neurosciences (R.E., K.S.R., H.F.K.), Department of Biochemistry and Molecular Biology (B.W.E., R.M.Y.), Department of Comparative Biology and Experimental Medicine (B.W.E., R.M.Y.), and Department of Cell Biology and Anatomy (H.F.K.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran (R.E., F.N.)
| | - Khalil S Rawji
- Department of Physiology & Pharmacology (R.E., M.D.H.), Department of Medicine (R.E., M.D.H.), Department of Clinical Neurosciences (R.E., K.S.R., H.F.K.), Department of Biochemistry and Molecular Biology (B.W.E., R.M.Y.), Department of Comparative Biology and Experimental Medicine (B.W.E., R.M.Y.), and Department of Cell Biology and Anatomy (H.F.K.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran (R.E., F.N.)
| | - Robin M Yates
- Department of Physiology & Pharmacology (R.E., M.D.H.), Department of Medicine (R.E., M.D.H.), Department of Clinical Neurosciences (R.E., K.S.R., H.F.K.), Department of Biochemistry and Molecular Biology (B.W.E., R.M.Y.), Department of Comparative Biology and Experimental Medicine (B.W.E., R.M.Y.), and Department of Cell Biology and Anatomy (H.F.K.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran (R.E., F.N.)
| | - Farshid Noorbakhsh
- Department of Physiology & Pharmacology (R.E., M.D.H.), Department of Medicine (R.E., M.D.H.), Department of Clinical Neurosciences (R.E., K.S.R., H.F.K.), Department of Biochemistry and Molecular Biology (B.W.E., R.M.Y.), Department of Comparative Biology and Experimental Medicine (B.W.E., R.M.Y.), and Department of Cell Biology and Anatomy (H.F.K.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran (R.E., F.N.)
| | - Hedwich F Kuipers
- Department of Physiology & Pharmacology (R.E., M.D.H.), Department of Medicine (R.E., M.D.H.), Department of Clinical Neurosciences (R.E., K.S.R., H.F.K.), Department of Biochemistry and Molecular Biology (B.W.E., R.M.Y.), Department of Comparative Biology and Experimental Medicine (B.W.E., R.M.Y.), and Department of Cell Biology and Anatomy (H.F.K.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran (R.E., F.N.)
| | - Morley D Hollenberg
- Department of Physiology & Pharmacology (R.E., M.D.H.), Department of Medicine (R.E., M.D.H.), Department of Clinical Neurosciences (R.E., K.S.R., H.F.K.), Department of Biochemistry and Molecular Biology (B.W.E., R.M.Y.), Department of Comparative Biology and Experimental Medicine (B.W.E., R.M.Y.), and Department of Cell Biology and Anatomy (H.F.K.), Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada; and Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran (R.E., F.N.)
| |
Collapse
|
2
|
Kipp M. Astrocytes: Lessons Learned from the Cuprizone Model. Int J Mol Sci 2023; 24:16420. [PMID: 38003609 PMCID: PMC10671869 DOI: 10.3390/ijms242216420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/06/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
A diverse array of neurological and psychiatric disorders, including multiple sclerosis, Alzheimer's disease, and schizophrenia, exhibit distinct myelin abnormalities at both the molecular and histological levels. These aberrations are closely linked to dysfunction of oligodendrocytes and alterations in myelin structure, which may be pivotal factors contributing to the disconnection of brain regions and the resulting characteristic clinical impairments observed in these conditions. Astrocytes, which significantly outnumber neurons in the central nervous system by a five-to-one ratio, play indispensable roles in the development, maintenance, and overall well-being of neurons and oligodendrocytes. Consequently, they emerge as potential key players in the onset and progression of a myriad of neurological and psychiatric disorders. Furthermore, targeting astrocytes represents a promising avenue for therapeutic intervention in such disorders. To gain deeper insights into the functions of astrocytes in the context of myelin-related disorders, it is imperative to employ appropriate in vivo models that faithfully recapitulate specific aspects of complex human diseases in a reliable and reproducible manner. One such model is the cuprizone model, wherein metabolic dysfunction in oligodendrocytes initiates an early response involving microglia and astrocyte activation, culminating in multifocal demyelination. Remarkably, following the cessation of cuprizone intoxication, a spontaneous process of endogenous remyelination occurs. In this review article, we provide a historical overview of studies investigating the responses and putative functions of astrocytes in the cuprizone model. Following that, we list previously published works that illuminate various aspects of the biology and function of astrocytes in this multiple sclerosis model. Some of the studies are discussed in more detail in the context of astrocyte biology and pathology. Our objective is twofold: to provide an invaluable overview of this burgeoning field, and, more importantly, to inspire fellow researchers to embark on experimental investigations to elucidate the multifaceted functions of this pivotal glial cell subpopulation.
Collapse
Affiliation(s)
- Markus Kipp
- Institute of Anatomy, Rostock University Medical Center, 18057 Rostock, Germany
| |
Collapse
|
3
|
Liang H, Zhang X, Hou Y, Zheng K, Hao H, He B, Li H, Sun C, Yang T, Song H, Cai R, Wang Y, Jiang H, Qi L, Wang Y. Super-high procoagulant activity of gecko thrombin: A gift from sky dragon. CNS Neurosci Ther 2023; 29:3081-3093. [PMID: 37144588 PMCID: PMC10493662 DOI: 10.1111/cns.14250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 04/06/2023] [Accepted: 04/17/2023] [Indexed: 05/06/2023] Open
Abstract
AIMS Gecko, the "sky dragon" named by Traditional Chinese Medicine, undergoes rapid coagulation and scarless regeneration following tail amputation in the natural ecology, providing a perfect opportunity to develop the efficient and safe drug for blood clotting. Here, gecko thrombin (gthrombin) was recombinantly prepared and comparatively studied on its procoagulant activity. METHODS The 3D structure of gthrombin was constructed using the homology modeling method of I-TASSER. The active gthrombin was prepared by the expression of gecko prethrombin-2 in 293 T cells, followed by purification with Ni2+ -chelating column chromatography prior to activation by snake venom-derived Ecarin. The enzymatic activities of gthrombin were assayed by hydrolysis of synthetic substrate S-2238 and the fibrinogen clotting. The vulnerable nerve cells were used to evaluate the toxicity of gthrombin at molecular and cellular levels. RESULTS The active recombinant gthrombin showed super-high catalytic and fibrinogenolytic efficiency than those of human under different temperatures and pH conditions. In addition, gthrombin made nontoxic effects on the central nerve cells including neurons, contrary to those of mammalian counterparts, which contribute to neuronal damage, astrogliosis, and demyelination. CONCLUSIONS A super-high activity but safe procoagulant candidate drug was identified from reptiles, which provided a promising perspective for clinical application in rapid blood clotting.
Collapse
Affiliation(s)
- Hao Liang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| | - Xingyuan Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| | - Yuxuan Hou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| | - Kang Zheng
- Anti‐aging & Regenerative Medicine Research Institution, School of Life Sciences and MedicineShandong University of TechnologyZiboPR China
| | - Huifei Hao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| | - Bingqiang He
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| | - Hui Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| | - Chunshuai Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| | - Ting Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| | - Honghua Song
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| | - Rixin Cai
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| | - Yingjie Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| | - Haiyan Jiang
- Department of Emergency MedicineAffiliated Hospital of Nantong UniversityNantongPR China
| | - Lei Qi
- Department of Emergency MedicineAffiliated Hospital of Nantong UniversityNantongPR China
| | - Yongjun Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co‐innovation Center of NeuroregenerationNantong UniversityNantongPR China
| |
Collapse
|
4
|
Su WS, Wu CH, Song WS, Chen SF, Yang FY. Low-intensity pulsed ultrasound ameliorates glia-mediated inflammation and neuronal damage in experimental intracerebral hemorrhage conditions. J Transl Med 2023; 21:565. [PMID: 37620888 PMCID: PMC10464049 DOI: 10.1186/s12967-023-04377-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 07/21/2023] [Indexed: 08/26/2023] Open
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) is a condition associated with high morbidity and mortality, and glia-mediated inflammation is a major contributor to neurological deficits. However, there is currently no proven effective treatment for clinical ICH. Recently, low-intensity pulsed ultrasound (LIPUS), a non-invasive method, has shown potential for neuroprotection in neurodegenerative diseases. This study aimed to investigate the neuroprotective effects and potential mechanisms of LIPUS on glia-mediated inflammation in ICH. METHODS This study used 289 mice to investigate the effects of LIPUS on ICH. ICH was induced by injecting bacterial collagenase (type VII-S; 0.0375 U) into the striatum of the mice. LIPUS was applied noninvasively for 3 days, including a 2-h-delayed intervention to mimic clinical usage. The study evaluated neurological function, histology, brain water content, hemoglobin content, MRI, and protein expression of neurotrophic factors, inflammatory molecules, and apoptosis. In vitro studies investigated glia-mediated inflammation by adding thrombin (10 U/mL) or conditioned media to primary and cell line cultures. The PI3K inhibitor LY294002 was used to confirm the effects of PI3K/Akt signaling after LIPUS treatment. RESULTS LIPUS treatment improved neurological deficits and reduced tissue loss, edema, and neurodegeneration after ICH. The protective effects of LIPUS resulted from decreased glia-mediated inflammation by inhibiting PI3K/Akt-NF-κB signaling, which reduced cytokine expression and attenuated microglial activation-induced neuronal damage in vitro. CONCLUSIONS LIPUS treatment improved neurological outcomes and reduced glia-mediated inflammation by inhibiting PI3K/Akt-NF-κB signaling after ICH. LIPUS may provide a non-invasive potential management strategy for ICH.
Collapse
Affiliation(s)
- Wei-Shen Su
- Department of Biomedical Imaging and Radiological Sciences, School of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong Street, Taipei, 11221, Taiwan
| | - Chun-Hu Wu
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Wen-Shin Song
- Division of Neurosurgery, Cheng Hsin General Hospital, Taipei, Taiwan
- Department of Neurological Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Szu-Fu Chen
- Department of Physical Medicine and Rehabilitation, Cheng Hsin General Hospital, No. 45, Cheng Hsin Street, Taipei, 11221, Taiwan.
- Department of Physiology and Biophysics, National Defense Medical Center, Taipei, Taiwan.
| | - Feng-Yi Yang
- Department of Biomedical Imaging and Radiological Sciences, School of Biomedical Science and Engineering, National Yang Ming Chiao Tung University, No. 155, Sec. 2, Li-Nong Street, Taipei, 11221, Taiwan.
| |
Collapse
|
5
|
Leo H, Kipp M. Remyelination in Multiple Sclerosis: Findings in the Cuprizone Model. Int J Mol Sci 2022; 23:ijms232416093. [PMID: 36555733 PMCID: PMC9783537 DOI: 10.3390/ijms232416093] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Remyelination therapies, which are currently under development, have a great potential to delay, prevent or even reverse disability in multiple sclerosis patients. Several models are available to study the effectiveness of novel compounds in vivo, among which is the cuprizone model. This model is characterized by toxin-induced demyelination, followed by endogenous remyelination after cessation of the intoxication. Due to its high reproducibility and ease of use, this model enjoys high popularity among various research and industrial groups. In this review article, we will summarize recent findings using this model and discuss the potential of some of the identified compounds to promote remyelination in multiple sclerosis patients.
Collapse
Affiliation(s)
| | - Markus Kipp
- Correspondence: ; Tel.: +49-(0)-381-494-8400
| |
Collapse
|
6
|
Yoon H, Triplet EM, Simon WL, Choi CI, Kleppe LS, De Vita E, Miller AK, Scarisbrick IA. Blocking Kallikrein 6 promotes developmental myelination. Glia 2022; 70:430-450. [PMID: 34626143 PMCID: PMC8732303 DOI: 10.1002/glia.24100] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 09/11/2021] [Accepted: 09/13/2021] [Indexed: 11/12/2022]
Abstract
Kallikrein related peptidase 6 (Klk6) is a secreted serine protease highly expressed in oligodendrocytes and implicated in demyelinating conditions. To gain insights into the significance of Klk6 to oligodendrocyte biology, we investigated the impact of global Klk6 gene knockout on CNS developmental myelination using the spinal cord of male and female mice as a model. Results demonstrate that constitutive loss of Klk6 expression accelerates oligodendrocyte differentiation developmentally, including increases in the expression of myelin proteins such as MBP, PLP and CNPase, in the number of CC-1+ mature oligodendrocytes, and myelin thickness by the end of the first postnatal week. Co-ordinate elevations in the pro-myelinating signaling pathways ERK and AKT, expression of fatty acid 2-hydroxylase, and myelin regulatory transcription factor were also observed in the spinal cord of 7d Klk6 knockouts. LC/MS/MS quantification of spinal cord lipids showed sphingosine and sphingomyelins to be elevated in Klk6 knockouts at the peak of myelination. Oligodendrocyte progenitor cells (OPCs)-derived from Klk6 knockouts, or wild type OPCs-treated with a Klk6 inhibitor (DFKZ-251), also showed increased MBP and PLP. Moreover, inhibition of Klk6 in OPC cultures enhanced brain derived neurotrophic factor-driven differentiation. Altogether, these findings suggest that oligodendrocyte-derived Klk6 may operate as an autocrine or paracrine rheostat, or brake, on pro-myelinating signaling serving to regulate myelin homeostasis developmentally and in the adult. These findings document for the first time that inhibition of Klk6 globally, or specifically in oligodendrocyte progenitors, is a strategy to increase early stages of oligodendrocyte differentiation and myelin production in the CNS.
Collapse
Affiliation(s)
- Hyesook Yoon
- Department of Physical Medicine and Rehabilitation, Mayo Clinic School of Biomedical Sciences Rochester 55905
| | - Erin M. Triplet
- Regenerative Sciences Program, Mayo Clinic School of Biomedical Sciences Rochester 55905
| | - Whitney L. Simon
- Department of Physical Medicine and Rehabilitation, Mayo Clinic School of Biomedical Sciences Rochester 55905
| | - Chan-Il Choi
- Department of Physical Medicine and Rehabilitation, Mayo Clinic School of Biomedical Sciences Rochester 55905
| | - Laurel S. Kleppe
- Department of Physical Medicine and Rehabilitation, Mayo Clinic School of Biomedical Sciences Rochester 55905
| | - Elena De Vita
- University of Heidelberg, Faculty of Biosciences, 69120 Heidelberg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Aubry K. Miller
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
| | - Isobel A. Scarisbrick
- Department of Physical Medicine and Rehabilitation, Mayo Clinic School of Biomedical Sciences Rochester 55905
- Regenerative Sciences Program, Mayo Clinic School of Biomedical Sciences Rochester 55905
- Department of Physiology and Biomedical Engineering, Minnesota USA 55905
| |
Collapse
|
7
|
Schrader JM, Xu F, Lee H, Barlock B, Benveniste H, Van Nostrand WE. Emergent White Matter Degeneration in the rTg-DI Rat Model of Cerebral Amyloid Angiopathy Exhibits Unique Proteomic Changes. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:426-440. [PMID: 34896071 PMCID: PMC8895424 DOI: 10.1016/j.ajpath.2021.11.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 11/16/2021] [Accepted: 11/23/2021] [Indexed: 12/23/2022]
Abstract
Cerebral amyloid angiopathy (CAA), characterized by cerebral vascular amyloid accumulation, neuroinflammation, microbleeds, and white matter (WM) degeneration, is a common comorbidity in Alzheimer disease and a prominent contributor to vascular cognitive impairment and dementia. WM loss was recently reported in the corpus callosum (CC) in the rTg-DI rat model of CAA. The current study shows that the CC exhibits a much lower CAA burden compared with the adjacent cortex. Sequential Window Acquisition of All Theoretical Mass Spectra tandem mass spectrometry was used to show specific proteomic changes in the CC with emerging WM loss and compare them with the proteome of adjacent cortical tissue in rTg-DI rats. In the CC, annexin A3, heat shock protein β1, and cystatin C were elevated at 4 months (M) before WM loss and at 12M with evident WM loss. Although annexin A3 and cystatin C were also enhanced in the cortex at 12M, annexin A5 and the leukodystrophy-associated astrocyte proteins megalencephalic leukoencephalopathy with subcortical cysts 1 and GlialCAM were distinctly elevated in the CC. Pathway analysis indicated neurodegeneration of axons, reflected by reduced expression of myelin and neurofilament proteins, was common to the CC and cortex; activation of Tgf-β1 and F2/thrombin was restricted to the CC. This study provides new insights into the proteomic changes that accompany WM loss in the CC of rTg-DI rats.
Collapse
Affiliation(s)
- Joseph M Schrader
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island; Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island
| | - Feng Xu
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island; Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island
| | - Hedok Lee
- Department of Anesthesiology, Yale University, New Haven, Connecticut
| | - Benjamin Barlock
- Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island
| | - Helene Benveniste
- Department of Anesthesiology, Yale University, New Haven, Connecticut
| | - William E Van Nostrand
- George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, Rhode Island; Department of Biomedical and Pharmaceutical Sciences, University of Rhode Island, Kingston, Rhode Island.
| |
Collapse
|
8
|
Shavit-Stein E, Berkowitz S, Gofrit SG, Altman K, Weinberg N, Maggio N. Neurocoagulation from a Mechanistic Point of View in the Central Nervous System. Semin Thromb Hemost 2022; 48:277-287. [PMID: 35052009 DOI: 10.1055/s-0041-1741569] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Coagulation mechanisms are critical for maintaining homeostasis in the central nervous system (CNS). Thrombin, an important player of the coagulation cascade, activates protease activator receptors (PARs), members of the G-protein coupled receptor family. PAR1 is located on neurons and glia. Following thrombin activation, PAR1 signals through the extracellular signal-regulated kinase pathway, causing alterations in neuronal glutamate release and astrocytic morphological changes. Similarly, the anticoagulation factor activated protein C (aPC) can cleave PAR1, following interaction with the endothelial protein C receptor. Both thrombin and aPC are expressed on endothelial cells and pericytes in the blood-brain barrier (BBB). Thrombin-induced PAR1 activation increases cytosolic Ca2+ concentration in brain vessels, resulting in nitric oxide release and increasing F-actin stress fibers, damaging BBB integrity. aPC also induces PAR1 activation and preserves BBB vascular integrity via coupling to sphingosine 1 phosphate receptors. Thrombin-induced PAR1 overactivation and BBB disruption are evident in CNS pathologies. During epileptic seizures, BBB disruption promotes thrombin penetration. Thrombin induces PAR1 activation and potentiates N-methyl-D-aspartate receptors, inducing glutamate-mediated hyperexcitability. Specific PAR1 inhibition decreases status epilepticus severity in vivo. In stroke, the elevation of brain thrombin levels further compromises BBB integrity, with direct parenchymal damage, while systemic factor Xa inhibition improves neurological outcomes. In multiple sclerosis (MS), brain thrombin inhibitory capacity correlates with clinical presentation. Both thrombin inhibition by hirudin and the use of recombinant aPC improve disease severity in an MS animal model. This review presents the mechanisms underlying the effects of coagulation on the physiology and pathophysiology of the CNS.
Collapse
Affiliation(s)
- Efrat Shavit-Stein
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel.,Department of Neurology and Neurosurgery, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Shani Berkowitz
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel.,Department of Neurology and Neurosurgery, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Shany Guly Gofrit
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Keren Altman
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Nitai Weinberg
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel
| | - Nicola Maggio
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, Israel.,Department of Neurology and Neurosurgery, Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.,Talpiot Medical Leadership Program, The Chaim Sheba Medical Center, Ramat Gan, Israel
| |
Collapse
|
9
|
Pompili E, De Franchis V, Giampietri C, Leone S, De Santis E, Fornai F, Fumagalli L, Fabrizi C. Protease Activated Receptor 1 and Its Ligands as Main Regulators of the Regeneration of Peripheral Nerves. Biomolecules 2021; 11:1668. [PMID: 34827666 PMCID: PMC8615415 DOI: 10.3390/biom11111668] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 10/29/2021] [Accepted: 11/05/2021] [Indexed: 12/16/2022] Open
Abstract
In contrast with the brain and spinal cord, peripheral nerves possess a striking ability to regenerate after damage. This characteristic of the peripheral nervous system is mainly due to a specific population of glial cells, the Schwann cells. Schwann cells promptly activate after nerve injury, dedifferentiate assuming a repair phenotype, and assist axon regrowth. In general, tissue injury determines the release of a variety of proteases which, in parallel with the degradation of their specific targets, also activate plasma membrane receptors known as protease-activated receptors (PARs). PAR1, the prototypical member of the PAR family, is also known as thrombin receptor and is present at the Schwann cell plasma membrane. This receptor is emerging as a possible regulator of the pro-regenerative capacity of Schwann cells. Here, we summarize the most recent literature data describing the possible contribution of PAR1 and PAR1-activating proteases in regulating the regeneration of peripheral nerves.
Collapse
Affiliation(s)
- Elena Pompili
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Via A. Borelli 50, 00161 Rome, Italy; (V.D.F.); (C.G.); (E.D.S.); (L.F.); (C.F.)
| | - Valerio De Franchis
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Via A. Borelli 50, 00161 Rome, Italy; (V.D.F.); (C.G.); (E.D.S.); (L.F.); (C.F.)
| | - Claudia Giampietri
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Via A. Borelli 50, 00161 Rome, Italy; (V.D.F.); (C.G.); (E.D.S.); (L.F.); (C.F.)
| | - Stefano Leone
- Department of Science, Roma Tre University, Viale Guglielmo Marconi 446, 00146 Rome, Italy;
| | - Elena De Santis
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Via A. Borelli 50, 00161 Rome, Italy; (V.D.F.); (C.G.); (E.D.S.); (L.F.); (C.F.)
| | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy;
- I.R.C.C.S. Neuromed, Via Atinense 18, 86077 Pozzilli, Italy
| | - Lorenzo Fumagalli
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Via A. Borelli 50, 00161 Rome, Italy; (V.D.F.); (C.G.); (E.D.S.); (L.F.); (C.F.)
| | - Cinzia Fabrizi
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, Via A. Borelli 50, 00161 Rome, Italy; (V.D.F.); (C.G.); (E.D.S.); (L.F.); (C.F.)
| |
Collapse
|
10
|
Critical Role of Astrocyte NAD + Glycohydrolase in Myelin Injury and Regeneration. J Neurosci 2021; 41:8644-8667. [PMID: 34493542 DOI: 10.1523/jneurosci.2264-20.2021] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 08/12/2021] [Accepted: 08/30/2021] [Indexed: 12/13/2022] Open
Abstract
Western-style diets cause disruptions in myelinating cells and astrocytes within the mouse CNS. Increased CD38 expression is present in the cuprizone and experimental autoimmune encephalomyelitis models of demyelination and CD38 is the main nicotinamide adenine dinucleotide (NAD+)-depleting enzyme in the CNS. Altered NAD+ metabolism is linked to both high fat consumption and multiple sclerosis (MS). Here, we identify increased CD38 expression in the male mouse spinal cord following chronic high fat consumption, after focal toxin [lysolecithin (LL)]-mediated demyelinating injury, and in reactive astrocytes within active MS lesions. We demonstrate that CD38 catalytically inactive mice are substantially protected from high fat-induced NAD+ depletion, oligodendrocyte loss, oxidative damage, and astrogliosis. A CD38 inhibitor, 78c, increased NAD+ and attenuated neuroinflammatory changes induced by saturated fat applied to astrocyte cultures. Conditioned media from saturated fat-exposed astrocytes applied to oligodendrocyte cultures impaired myelin protein production, suggesting astrocyte-driven indirect mechanisms of oligodendrogliopathy. In cerebellar organotypic slice cultures subject to LL-demyelination, saturated fat impaired signs of remyelination effects that were mitigated by concomitant 78c treatment. Significantly, oral 78c increased counts of oligodendrocytes and remyelinated axons after focal LL-induced spinal cord demyelination. Using a RiboTag approach, we identified a unique in vivo brain astrocyte translatome profile induced by 78c-mediated CD38 inhibition in mice, including decreased expression of proinflammatory astrocyte markers and increased growth factors. Our findings suggest that a high-fat diet impairs oligodendrocyte survival and differentiation through astrocyte-linked mechanisms mediated by the NAD+ase CD38 and highlights CD38 inhibitors as potential therapeutic candidates to improve myelin regeneration.SIGNIFICANCE STATEMENT Myelin disturbances and oligodendrocyte loss can leave axons vulnerable, leading to permanent neurologic deficits. The results of this study suggest that metabolic disturbances, triggered by consumption of a diet high in fat, promote oligodendrogliopathy and impair myelin regeneration through astrocyte-linked indirect nicotinamide adenine dinucleotide (NAD+)-dependent mechanisms. We demonstrate that restoring NAD+ levels via genetic inactivation of CD38 can overcome these effects. Moreover, we show that therapeutic inactivation of CD38 can enhance myelin regeneration. Together, these findings point to a new metabolic targeting strategy positioned to improve disease course in multiple sclerosis and other conditions in which the integrity of myelin is a key concern.
Collapse
|
11
|
Kim HN, Triplet EM, Radulovic M, Bouchal S, Kleppe LS, Simon WL, Yoon H, Scarisbrick IA. The thrombin receptor modulates astroglia-neuron trophic coupling and neural repair after spinal cord injury. Glia 2021; 69:2111-2132. [PMID: 33887067 PMCID: PMC8672305 DOI: 10.1002/glia.24012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 04/10/2021] [Accepted: 04/11/2021] [Indexed: 12/15/2022]
Abstract
Excessive activation of the thrombin receptor, protease activated receptor 1 (PAR1) is implicated in diverse neuropathologies from neurodegenerative conditions to neurotrauma. PAR1 knockout mice show improved outcomes after experimental spinal cord injury (SCI), however information regarding the underpinning cellular and molecular mechanisms is lacking. Here we demonstrate that genetic blockade of PAR1 in female mice results in improvements in sensorimotor co-ordination after thoracic spinal cord lateral compression injury. We document improved neuron preservation with increases in Synapsin-1 presynaptic proteins and GAP43, a growth cone marker, after a 30 days recovery period. These improvements were coupled to signs of enhanced myelin resiliency and repair, including increases in the number of mature oligodendrocytes, their progenitors and the abundance of myelin basic protein. These significant increases in substrates for neural recovery were accompanied by reduced astrocyte (Serp1) and microglial/monocyte (CD68 and iNOS) pro-inflammatory markers, with coordinate increases in astrocyte (S100A10 and Emp1) and microglial (Arg1) markers reflective of pro-repair activities. Complementary astrocyte-neuron co-culture bioassays suggest astrocytes with PAR1 loss-of-function promote both neuron survival and neurite outgrowth. Additionally, the pro-neurite outgrowth effects of switching off astrocyte PAR1 were blocked by inhibiting TrkB, the high affinity receptor for brain derived neurotrophic factor. Altogether, these studies demonstrate unique modulatory roles for PAR1 in regulating glial-neuron interactions, including the capacity for neurotrophic factor signaling, and underscore its position at neurobiological intersections critical for the response of the CNS to injury and the capacity for regenerative repair and restoration of function.
Collapse
Affiliation(s)
- Ha Neui Kim
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, Rochester MN 55905
- Department of Physiology and Biomedical Engineering, Rochester MN 55905
| | - Erin M. Triplet
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, Rochester MN 55905
- Department of Physiology and Biomedical Engineering, Rochester MN 55905
- Neuroscience Program, Mayo Clinic Graduate School of Biomedical Sciences, Rochester MN 55905
| | - Maja Radulovic
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, Rochester MN 55905
- Department of Physiology and Biomedical Engineering, Rochester MN 55905
| | - Samantha Bouchal
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, Rochester MN 55905
| | - Laurel S. Kleppe
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, Rochester MN 55905
| | - Whitney L. Simon
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, Rochester MN 55905
| | - Hyesook Yoon
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, Rochester MN 55905
- Department of Physiology and Biomedical Engineering, Rochester MN 55905
| | - Isobel A. Scarisbrick
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, Rochester MN 55905
- Department of Physiology and Biomedical Engineering, Rochester MN 55905
- Neuroscience Program, Mayo Clinic Graduate School of Biomedical Sciences, Rochester MN 55905
| |
Collapse
|
12
|
Triplet EM, Kim HN, Yoon H, Radulovic M, Kleppe L, Simon WL, Choi CI, Walsh PJ, Dutton JR, Scarisbrick IA. The thrombin receptor links brain derived neurotrophic factor to neuron cholesterol production, resiliency and repair after spinal cord injury. Neurobiol Dis 2021; 152:105294. [PMID: 33549720 PMCID: PMC8021459 DOI: 10.1016/j.nbd.2021.105294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 01/13/2021] [Accepted: 02/03/2021] [Indexed: 11/28/2022] Open
Abstract
Despite concerted efforts to identify CNS regeneration strategies, an incomplete understanding of how the needed molecular machinery is regulated limits progress. Here we use models of lateral compression and FEJOTA clip contusion-compression spinal cord injury (SCI) to identify the thrombin receptor (Protease Activated Receptor 1 (PAR1)) as an integral facet of this machine with roles in regulating neurite growth through a growth factor- and cholesterol-dependent mechanism. Functional recovery and signs of neural repair, including expression of cholesterol biosynthesis machinery and markers of axonal and synaptic integrity, were all increased after SCI in PAR1 knockout female mice, while PTEN was decreased. Notably, PAR1 differentially regulated HMGCS1, a gene encoding a rate-limiting enzyme in cholesterol production, across the neuronal and astroglial compartments of the intact versus injured spinal cord. Pharmacologic inhibition of cortical neuron PAR1 using vorapaxar in vitro also decreased PTEN and promoted neurite outgrowth in a cholesterol dependent manner, including that driven by suboptimal brain derived neurotrophic factor (BDNF). Pharmacologic inhibition of PAR1 also augmented BDNF-driven HMGCS1 and cholesterol production by murine cortical neurons and by human SH-SY5Y and iPSC-derived neurons. The link between PAR1, cholesterol and BDNF was further highlighted by demonstrating that the deleterious effects of PAR1 over-activation are overcome by supplementing cultures with BDNF, cholesterol or by blocking an inhibitor of adenylate cyclase, Gαi. These findings document PAR1-linked neurotrophic coupling mechanisms that regulate neuronal cholesterol metabolism as an important component of the machinery regulating CNS repair and point to new strategies to enhance neural resiliency after injury.
Collapse
Affiliation(s)
- Erin M Triplet
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic Alix School of Medicine and the Mayo Clinic Medical Scientist Training Program Sciences Rochester, United States of America
| | - Ha Neui Kim
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, United States of America
| | - Hyesook Yoon
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, United States of America
| | - Maja Radulovic
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, United States of America
| | - Laurel Kleppe
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, United States of America
| | - Whitney L Simon
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, United States of America
| | - Chan-Il Choi
- Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, United States of America
| | - Patrick J Walsh
- Department of Genetics and Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - James R Dutton
- Department of Genetics and Cell Biology and Development, University of Minnesota, Minneapolis, MN 55455, United States of America
| | - Isobel A Scarisbrick
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic Alix School of Medicine and the Mayo Clinic Medical Scientist Training Program Sciences Rochester, United States of America; Department of Physical Medicine and Rehabilitation, Rehabilitation Medicine Research Center, United States of America; Department of Physiology and Biomedical Engineering, Rochester, MN 55905, United States of America.
| |
Collapse
|
13
|
Guo H, Baker G, Hartle K, Fujiwara E, Wang J, Zhang Y, Xing J, Lyu H, Li XM, Chen J. Exploratory study on neurochemical effects of low-intensity pulsed ultrasound in brains of mice. Med Biol Eng Comput 2021; 59:1099-1110. [PMID: 33881705 DOI: 10.1007/s11517-021-02351-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 03/19/2021] [Indexed: 01/25/2023]
Abstract
There is now a relatively large body of evidence suggesting a relationship between dysfunction of myelin and oligodendrocytes and the etiology of several neuropsychiatric disorders, including depression and schizophrenia, and also suggesting that ultrasound methods may alleviate some of the symptoms of depression. We have applied low-intensity pulsed ultrasound (LIPUS) to the brains of mice treated with the demyelinating drug cuprizone, a drug that has been used as the basis for a rodent model relevant to a number of psychiatric and neurologic disorders including depression, schizophrenia, and multiple sclerosis. Prior to conducting the studies in mice, preliminary studies were carried out on the effects of LIPUS in vitro in neuron-like SH-SY5Y cells and primary glial cells. In subsequent studies in mice, female C57BL/6 mice were restrained in plastic tubes for 20 min daily with the ultrasound transducer near the end of the tube directly above the mouse's head. LIPUS was used at an intensity of 25 mW/cm2 once daily for 22 days in control mice and in mice undergoing daily repetitive restraint stress (RRS). Behavioral or neurochemical studies were done on the mice or the brain tissue obtained from them. The studies in vitro indicated that LIPUS stimulation at an intensity of 15 mW/cm2 delivered for 5 min daily for 3 days in an enclosed sterile cell culture plate in an incubator increased the viability of SH-SY5Y and primary glial cells. In the studies in mice, LIPUS elevated levels of doublecortin, a marker for neurogenesis, in the cortex compared to levels in the RRS mice and caused a trend in elevation of brain levels of brain-derived neurotrophic factor in the hippocampus relative to control levels. LIPUS also increased sucrose preference (a measure of the attenuation of anhedonia, a common symptom of several psychiatric disorders) in the RRS model in mice. The ability of LIPUS administered daily to rescue damaged myelin and oligodendrocytes was studied in mice treated chronically with cuprizone for 35 days. LIPUS increased cortex and corpus callosum levels of myelin basic protein, a protein marker for mature oligodendrocytes, and neural/glial antigen 2, a protein marker for oligodendrocyte precursor cells, relative to levels in the cuprizone + sham animals. These results of this exploratory study suggest that future comprehensive time-related studies with LIPUS on brain chemistry and behavior related to neuropsychiatric disorders are warranted. Exploratory Study on Neurochemical Effects of Low Intensity Pulsed Ultrasound in Brains of Mice. Upper part of figure: LIPUS device and in-vitro cell experimental set-up. The center image is the LIPUS generating box; the image in the upper left shows the cell experiment set-up; the image in the upper right shows a zoomed-in sketch for the cell experiment; the image in the lower left shows the set-up of repetitive restraint stress (RRS) with a mouse; the image in the lower middle shows the set-up of LIPUS treatment of a mouse; the image in the lower right shows a zoomed-in sketch for the LIPUS treatment of a mouse.
Collapse
Affiliation(s)
- Huining Guo
- Department of Psychiatry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, TGG 2B7, Canada
| | - Glen Baker
- Department of Psychiatry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, TGG 2B7, Canada.,Neuroscience & Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Kelly Hartle
- Department of Psychiatry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, TGG 2B7, Canada.,Neuroscience & Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Esther Fujiwara
- Department of Psychiatry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, TGG 2B7, Canada.,Neuroscience & Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Junhui Wang
- Department of Psychiatry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, TGG 2B7, Canada
| | - Yanbo Zhang
- Department of Psychiatry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, TGG 2B7, Canada.,Neuroscience & Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Jida Xing
- Department of Electrical and Computer Engineering, Faculty of Engineering, University of Alberta, Edmonton, Canada
| | - Haiyan Lyu
- Department of Pharmacy, Xianyue Hospital, Xiamen, China
| | - Xin-Min Li
- Department of Psychiatry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, TGG 2B7, Canada. .,Neuroscience & Mental Health Institute, University of Alberta, Edmonton, AB, Canada.
| | - Jie Chen
- Department of Electrical and Computer Engineering, Faculty of Engineering, University of Alberta, Edmonton, Canada. .,Department of Biomedical Engineering, Faculty of Engineering, University of Alberta, Edmonton, AB, T6G 1H9, Canada.
| |
Collapse
|
14
|
Jordan KR, Parra-Izquierdo I, Gruber A, Shatzel JJ, Pham P, Sherman LS, McCarty OJT, Verbout NG. Thrombin generation and activity in multiple sclerosis. Metab Brain Dis 2021; 36:407-420. [PMID: 33411219 PMCID: PMC7864536 DOI: 10.1007/s11011-020-00652-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/25/2020] [Indexed: 01/19/2023]
Abstract
The coagulation cascade and immune system are intricately linked, highly regulated and respond cooperatively in response to injury and infection. Increasingly, evidence of hyper-coagulation has been associated with autoimmune disorders, including multiple sclerosis (MS). The pathophysiology of MS includes immune cell activation and recruitment to the central nervous system (CNS) where they degrade myelin sheaths, leaving neuronal axons exposed to damaging inflammatory mediators. Breakdown of the blood-brain barrier (BBB) facilitates the entry of peripheral immune cells. Evidence of thrombin activity has been identified within the CNS of MS patients and studies using animal models of experimental autoimmune encephalomyelitis (EAE), suggest increased thrombin generation and activity may play a role in the pathogenesis of MS as well as inhibit remyelination processes. Thrombin is a serine protease capable of cleaving multiple substrates, including protease activated receptors (PARs), fibrinogen, and protein C. Cleavage of all three of these substrates represent pathways through which thrombin activity may exert immuno-regulatory effects and regulate permeability of the BBB during MS and EAE. In this review, we summarize evidence that thrombin activity directly, through PARs, and indirectly, through fibrin formation and activation of protein C influences neuro-immune responses associated with MS and EAE pathology.
Collapse
Affiliation(s)
- Kelley R Jordan
- Department of Biomedical Engineering, Oregon Health and Science University, School of Medicine, 3303 SW Bond Avenue, Portland, OR, 97239, USA.
| | - Ivan Parra-Izquierdo
- Department of Biomedical Engineering, Oregon Health and Science University, School of Medicine, 3303 SW Bond Avenue, Portland, OR, 97239, USA
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Knight Cancer Institute, Portland, OR, USA
| | - András Gruber
- Department of Biomedical Engineering, Oregon Health and Science University, School of Medicine, 3303 SW Bond Avenue, Portland, OR, 97239, USA
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Knight Cancer Institute, Portland, OR, USA
- Aronora Inc, Portland, OR, USA
| | - Joseph J Shatzel
- Department of Biomedical Engineering, Oregon Health and Science University, School of Medicine, 3303 SW Bond Avenue, Portland, OR, 97239, USA
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Knight Cancer Institute, Portland, OR, USA
| | - Peter Pham
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| | - Larry S Sherman
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health and Science University, Beaverton, OR, USA
| | - Owen J T McCarty
- Department of Biomedical Engineering, Oregon Health and Science University, School of Medicine, 3303 SW Bond Avenue, Portland, OR, 97239, USA
- Division of Hematology and Medical Oncology, Oregon Health and Science University, Knight Cancer Institute, Portland, OR, USA
| | - Norah G Verbout
- Department of Biomedical Engineering, Oregon Health and Science University, School of Medicine, 3303 SW Bond Avenue, Portland, OR, 97239, USA
- Aronora Inc, Portland, OR, USA
| |
Collapse
|
15
|
Bhattacharjee A, Ghosh S, Chatterji A, Chakraborty K. Neuron-glia: understanding cellular copper homeostasis, its cross-talk and their contribution towards neurodegenerative diseases. Metallomics 2020; 12:1897-1911. [PMID: 33295934 DOI: 10.1039/d0mt00168f] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Over the years, the mechanism of copper homeostasis in various organ systems has gained importance. This is owing to the involvement of copper in a wide range of genetic disorders, most of them involving neurological symptoms. This highlights the importance of copper and its tight regulation in a complex organ system like the brain. It demands understanding the mechanism of copper acquisition and delivery to various cell types overcoming the limitation imposed by the blood brain barrier. The present review aims to investigate the existing work to understand the mechanism and complexity of cellular copper homeostasis in the two major cell types of the CNS - the neurons and the astrocytes. It investigates the mechanism of copper uptake, incorporation and export by these cell types. Furthermore, it brings forth the common as well as the exclusive aspects of neuronal and glial copper homeostasis including the studies from copper-based sensors. Glia act as a mediator of copper supply between the endothelium and the neurons. They possess all the qualifications of acting as a 'copper-sponge' for supply to the neurons. The neurons, on the other hand, require copper for various essential functions like incorporation as a cofactor for enzymes, synaptogenesis, axonal extension, inhibition of postsynaptic excitotoxicity, etc. Lastly, we also aim to understand the neuronal and glial pathology in various copper homeostasis disorders. The etiology of glial pathology and its contribution towards neuronal pathology and vice versa underlies the complexity of the neuropathology associated with the copper metabolism disorders.
Collapse
Affiliation(s)
- Ashima Bhattacharjee
- Amity Institute of Biotechnology, Amity University, Plot No: 36, 37 & 38, Major Arterial Road, Action Area II, Kadampukur Village, Rajarhat, Newtown, Kolkata, West Bengal 700135, India.
| | | | | | | |
Collapse
|