1
|
Power E, Mongan D, Healy C, Susai SR, Föcking M, Zammit S, Cannon M, Cotter D. Cannabis use in youth is associated with chronic inflammation. Psychol Med 2024:1-11. [PMID: 39648682 DOI: 10.1017/s0033291724002848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
BACKGROUND Markers of inflammation and cannabis exposure are associated with an increased risk of mental disorders. In the current study, we investigated associations between cannabis use and biomarkers of inflammation. METHODS Utilizing a sample of 914 participants from the Avon Longitudinal Study of Parents and Children, we investigated whether interleukin-6 (IL-6), tumor necrosis factor α (TNFα), C-reactive protein (CRP), and soluble urokinase plasminogen activator receptor (suPAR) measured at age 24 were associated with past year daily cannabis use, less frequent cannabis use, and no past year cannabis use. We adjusted for a number of covariates including sociodemographic measures, body mass index, childhood trauma, and tobacco smoking. We found evidence of a strong association between daily or near daily cannabis use and suPAR. RESULTS We did not find any associations between less frequent cannabis use and suPAR. We did not find evidence of an association between IL-6, TNFα or CRP, and cannabis use. CONCLUSIONS Our finding that frequent cannabis use is strongly associated with suPAR, a biomarker of systemic chronic inflammation implicated in neurodevelopmental and neurodegenerative processes is novel. These findings may provide valuable insights into biological mechanisms by which cannabis affects the brain and impacts the risk of serious mental disorders.
Collapse
Affiliation(s)
- Emmet Power
- Department of Psychiatry, Royal College of Surgeons in Ireland, Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
- Department of Liaison Psychiatry, Children's Health Ireland, Dublin 1, Ireland
| | - David Mongan
- Department of Psychiatry, Royal College of Surgeons in Ireland, Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
- Centre for Public Health, Queens University Belfast, Belfast, UK
| | - Colm Healy
- Department of Psychiatry, Royal College of Surgeons in Ireland, Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
- Centre for Clinical Brain Sciences, Division of Psychiatry, University of Edinburgh, Edinburgh, UK
- Department of Child and Adolescent Psychiatry, School of Medicine, University College Dublin, Dublin, Ireland
| | - Subash Raj Susai
- Department of Psychiatry, Royal College of Surgeons in Ireland, Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Melanie Föcking
- Department of Psychiatry, Royal College of Surgeons in Ireland, Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - Stanley Zammit
- Centre for Academic Mental Health, Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
- Division of Psychological Medicine and Clinical Neurosciences, MRC Centre for Neuropsychiatric Genetics and Genomics, Cardiff University School of Medicine, Cardiff, UK
| | - Mary Cannon
- Department of Psychiatry, Royal College of Surgeons in Ireland, Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
- Department of Liaison Psychiatry, Beaumont Hospital, Dublin 9, Ireland
- FutureNeuro Research Ireland Centre, Royal College of Surgeons in Ireland, Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| | - David Cotter
- Department of Psychiatry, Royal College of Surgeons in Ireland, Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
- Department of Liaison Psychiatry, Beaumont Hospital, Dublin 9, Ireland
- FutureNeuro Research Ireland Centre, Royal College of Surgeons in Ireland, Smurfit Building, Beaumont Hospital, Dublin 9, Ireland
| |
Collapse
|
2
|
Daga KR, Larey AM, Morfin MG, Chen K, Bitarafan S, Carpenter JM, Hynds HM, Hines KM, Wood LB, Marklein RA. Microglia morphological response to mesenchymal stromal cell extracellular vesicles demonstrates EV therapeutic potential for modulating neuroinflammation. J Biol Eng 2024; 18:58. [PMID: 39420399 PMCID: PMC11488223 DOI: 10.1186/s13036-024-00449-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Mesenchymal stromal cell derived extracellular vesicles (MSC-EVs) are a promising therapeutic for neuroinflammation. MSC-EVs can interact with microglia, the resident immune cells of the brain, to exert their immunomodulatory effects. In response to inflammatory cues, such as cytokines, microglia undergo phenotypic changes indicative of their function e.g. morphology and secretion. However, these changes in response to MSC-EVs are not well understood. Additionally, no disease-relevant screening tools to assess MSC-EV bioactivity exist, which has further impeded clinical translation. Here, we developed a quantitative, high throughput morphological profiling approach to assess the response of microglia to neuroinflammation- relevant signals and whether this morphological response can be used to indicate the bioactivity of MSC-EVs. RESULTS Using an immortalized human microglia cell-line, we observed increased size (perimeter, major axis length) and complexity (form factor) upon stimulation with interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α). Upon treatment with MSC-EVs, the overall morphological score (determined using principal component analysis) shifted towards the unstimulated morphology, indicating that MSC-EVs are bioactive and modulate microglia. The morphological effects of MSC-EVs in TNF-α /IFN-γ stimulated cells were concomitant with reduced secretion of 14 chemokines/cytokines (e.g. CXCL6, CXCL9) and increased secretion of 12 chemokines/cytokines (e.g. CXCL8, CXCL10). Proteomic analysis of cell lysates revealed significant increases in 192 proteins (e.g. HIBADH, MEAK7, LAMC1) and decreases in 257 proteins (e.g. PTEN, TOM1, MFF) with MSC-EV treatment. Of note, many of these proteins are involved in regulation of cell morphology and migration. Gene Set Variation Analysis revealed upregulation of pathways associated with immune response, such as regulation of cytokine production, immune cell infiltration (e.g. T cells, NK cells) and morphological changes (e.g. Semaphorin, RHO/Rac signaling). Additionally, changes in microglia mitochondrial morphology were measured suggesting that MSC-EV modulate mitochondrial metabolism. CONCLUSION This study comprehensively demonstrates the effects of MSC-EVs on human microglial morphology, cytokine secretion, cellular proteome, and mitochondrial content. Our high-throughput, rapid, low-cost morphometric approach enables screening of MSC-EV batches and manufacturing conditions to enhance EV function and mitigate EV functional heterogeneity in a disease relevant manner. This approach is highly generalizable and can be further adapted and refined based on selection of the disease-relevant signal, target cell, and therapeutic product.
Collapse
Affiliation(s)
- Kanupriya R Daga
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA
| | - Andrew M Larey
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA
| | - Maria G Morfin
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA
| | - Kailin Chen
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA
- Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA
| | - Sara Bitarafan
- George W. Woodruff School of Mechanical Engineering and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | | | - Hannah M Hynds
- Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Kelly M Hines
- Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Levi B Wood
- George W. Woodruff School of Mechanical Engineering and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Ross A Marklein
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA.
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA.
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20903, USA.
| |
Collapse
|
3
|
Liu Y, Chen J. Senescence-related genes and proteins in the development of Alzheimer's disease: evidence from transcriptomic and Mendelian randomization analysis. Front Aging Neurosci 2024; 16:1423725. [PMID: 39156738 PMCID: PMC11327092 DOI: 10.3389/fnagi.2024.1423725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 07/23/2024] [Indexed: 08/20/2024] Open
Abstract
Purpose Alzheimer's disease (AD) is a common neurodegenerative disease, which can lead to cognitive impairment and dementia. Since AD is tightly associated with aging and cellular senescence, objective of this study was to investigate the association between senescence-related genes and proteins (SRGs and SRPs) and the development of AD. Design The whole study was based on transcriptomic analysis of control and AD brain tissues and Mendelian randomization (MR) analysis. Methods For transcriptomic analysis, GSE5281 dataset from GEO database contains the transcriptomic data of human brain tissues (n = 161) from control group and AD patients. The expression of SRGs in control and AD brain tissues were compared by Student's t test. For MR analysis, the instrumental single-nucleotide polymorphisms (SNPs) associated with 110 SRPs were filtered and selected from a large genome-wide association study (GWAS) for plasma proteome. The causality between plasma levels of SRPs and AD was explored using GWAS data of AD from Lambert et al. (17,008 cases and 37,154 controls) and further validated by using data from FinnGen consortium (6,489 patients and 170,489 controls). MR estimate was performed using the inverse-variance weighted (IVW) method and the heterogeneity and pleiotropy of results were tested. Results Transcriptomic analysis identified 36 up-regulated (including PLAUR) and 8 down-regulated SRGs in AD brain tissues. In addition, the MR results at both discovery and validation stages supported the causality between plasma levels of PLAUR (IVW-p = 3.04E-2, odds ratio [OR] = 1.15), CD55 (IVW-p = 1.56E-3, OR = 0.86), and SERPINE2 (IVW-p = 2.74E-2, OR = 0.91) and the risk of AD. Conclusion Our findings identified that PLAUR, as an SRG, may take part in the development of AD and found that high plasma levels of PLAUR was associated with increased risk of AD, indicating that this gene was a risk factor for this disease and providing the rationale of existing drugs or new preventative and therapeutic strategies.
Collapse
Affiliation(s)
| | - Jiao Chen
- Department of Gerontology and Geriatrics, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
4
|
Daga KR, Larey AM, Morfin MG, Chen K, Bitarafan S, Carpenter JM, Hynds HM, Hines KM, Wood LB, Marklein RA. Microglia Morphological Response to Mesenchymal Stromal Cell Extracellular Vesicles Demonstrates EV Therapeutic Potential for Modulating Neuroinflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.01.601612. [PMID: 39005342 PMCID: PMC11245023 DOI: 10.1101/2024.07.01.601612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Background Mesenchymal stromal cell derived extracellular vesicles (MSC-EVs) are a promising therapeutic for neuroinflammation. MSC-EVs can interact with microglia, the resident immune cells of the brain, to exert their immunomodulatory effects. In response to inflammatory cues, such as cytokines, microglia undergo phenotypic changes indicative of their function e.g. morphology and secretion. However, these changes in response to MSC-EVs are not well understood. Additionally, no disease-relevant screening tools to assess MSC-EV bioactivity exist, which has further impeded clinical translation. Here, we developed a quantitative, high throughput morphological profiling approach to assess the response of microglia to neuroinflammation-relevant signals and whether this morphological response can be used to indicate the bioactivity of MSC-EVs. Results Using an immortalized human microglia cell-line, we observed increased size (perimeter, major axis length) and complexity (form factor) upon stimulation with interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α). Upon treatment with MSC-EVs, the overall morphological score (determined using principal component analysis) shifted towards the unstimulated morphology, indicating that MSC-EVs are bioactive and modulate microglia. The morphological effects of MSC-EVs in TNF-γ/IFN-α stimulated cells were concomitant with reduced secretion of 14 chemokines/cytokines (e.g. CXCL6, CXCL9) and increased secretion of 12 chemokines/cytokines (e.g. CXCL8, CXCL10). Proteomic analysis of cell lysates revealed significant increases in 192 proteins (e.g. HIBADH, MEAK7, LAMC1) and decreases in 257 proteins (e.g. PTEN, TOM1, MFF) with MSC-EV treatment. Of note, many of these proteins are involved in regulation of cell morphology and migration. Gene Set Variation Analysis revealed upregulation of pathways associated with immune response, such as regulation of cytokine production, immune cell infiltration (e.g. T cells, NK cells) and morphological changes (e.g. Semaphorin, RHO/Rac signaling). Additionally, changes in microglia mitochondrial morphology were measured suggesting that MSC-EV modulate mitochondrial metabolism. Conclusion This study comprehensively demonstrates the effects of MSC-EVs on human microglial morphology, cytokine secretion, cellular proteome, and mitochondrial content. Our high-throughput, rapid, low-cost morphological approach enables screening of MSC-EV batches and manufacturing conditions to enhance EV function and mitigate EV functional heterogeneity in a disease relevant manner. This approach is highly generalizable and can be further adapted and refined based on selection of the disease-relevant signal, target cell, and therapeutic product.
Collapse
Affiliation(s)
- Kanupriya R Daga
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA
| | - Andrew M Larey
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA
| | - Maria G Morfin
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA
| | - Kailin Chen
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA
- Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA
| | - Sara Bitarafan
- George W. Woodruff School of Mechanical Engineering and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | | | - Hannah M Hynds
- Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Kelly M Hines
- Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Levi B Wood
- George W. Woodruff School of Mechanical Engineering and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Ross A Marklein
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
5
|
Hamada M, Varkoly KS, Riyadh O, Beladi R, Munuswamy-Ramanujam G, Rawls A, Wilson-Rawls J, Chen H, McFadden G, Lucas AR. Urokinase-Type Plasminogen Activator Receptor (uPAR) in Inflammation and Disease: A Unique Inflammatory Pathway Activator. Biomedicines 2024; 12:1167. [PMID: 38927374 PMCID: PMC11201033 DOI: 10.3390/biomedicines12061167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 04/24/2024] [Accepted: 05/10/2024] [Indexed: 06/28/2024] Open
Abstract
The urokinase-type plasminogen activator receptor (uPAR) is a unique protease binding receptor, now recognized as a key regulator of inflammation. Initially, uPA/uPAR was considered thrombolytic (clot-dissolving); however, recent studies have demonstrated its predominant immunomodulatory functions in inflammation and cancer. The uPA/uPAR complex has a multifaceted central role in both normal physiological and also pathological responses. uPAR is expressed as a glycophosphatidylinositol (GPI)-linked receptor interacting with vitronectin, integrins, G protein-coupled receptors, and growth factor receptors within a large lipid raft. Through protein-to-protein interactions, cell surface uPAR modulates intracellular signaling, altering cellular adhesion and migration. The uPA/uPAR also modifies extracellular activity, activating plasminogen to form plasmin, which breaks down fibrin, dissolving clots and activating matrix metalloproteinases that lyse connective tissue, allowing immune and cancer cell invasion and releasing growth factors. uPAR is now recognized as a biomarker for inflammatory diseases and cancer; uPAR and soluble uPAR fragments (suPAR) are increased in viral sepsis (COVID-19), inflammatory bowel disease, and metastasis. Here, we provide a comprehensive overview of the structure, function, and current studies examining uPAR and suPAR as diagnostic markers and therapeutic targets. Understanding uPAR is central to developing diagnostic markers and the ongoing development of antibody, small-molecule, nanogel, and virus-derived immune-modulating treatments that target uPAR.
Collapse
Affiliation(s)
- Mostafa Hamada
- College of Medicine, Kansas City University, 1750 Independence Ave, Kansas City, MO 64106, USA; (M.H.); (O.R.)
| | - Kyle Steven Varkoly
- Department of Internal Medicine, McLaren Macomb Hospital, Michigan State University College of Human Medicine, 1000 Harrington St., Mt Clemens, MI 48043, USA
| | - Omer Riyadh
- College of Medicine, Kansas City University, 1750 Independence Ave, Kansas City, MO 64106, USA; (M.H.); (O.R.)
| | - Roxana Beladi
- Department of Neurosurgery, Ascension Providence Hospital, Michigan State University College of Human Medicine, 16001 W Nine Mile Rd, Southfield, MI 48075, USA;
| | - Ganesh Munuswamy-Ramanujam
- Molecular Biology and Immunobiology Division, Interdisciplinary Institute of Indian System of Medicine, SRM Institute of Science and Technology, Kattankulathur 603203, India;
| | - Alan Rawls
- School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ 85281, USA; (A.R.); (J.W.-R.)
| | - Jeanne Wilson-Rawls
- School of Life Sciences, Arizona State University, 427 E Tyler Mall, Tempe, AZ 85281, USA; (A.R.); (J.W.-R.)
| | - Hao Chen
- Department of Tumor Center, Lanzhou University Second Hospital, Lanzhou 730030, China;
| | - Grant McFadden
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, 727 E Tyler St., Tempe, AZ 85287, USA;
| | - Alexandra R. Lucas
- Center for Personalized Diagnostics, Biodesign Institute, Arizona State University, 727 E Tyler St., Tempe, AZ 85287, USA;
| |
Collapse
|
6
|
Proteome integral solubility alteration high-throughput proteomics assay identifies Collectin-12 as a non-apoptotic microglial caspase-3 substrate. Cell Death Dis 2023; 14:192. [PMID: 36906641 PMCID: PMC10008626 DOI: 10.1038/s41419-023-05714-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 02/23/2023] [Accepted: 02/27/2023] [Indexed: 03/13/2023]
Abstract
Caspases are a family of proteins mostly known for their role in the activation of the apoptotic pathway leading to cell death. In the last decade, caspases have been found to fulfill other tasks regulating the cell phenotype independently to cell death. Microglia are the immune cells of the brain responsible for the maintenance of physiological brain functions but can also be involved in disease progression when overactivated. We have previously described non-apoptotic roles of caspase-3 (CASP3) in the regulation of the inflammatory phenotype of microglial cells or pro-tumoral activation in the context of brain tumors. CASP3 can regulate protein functions by cleavage of their target and therefore could have multiple substrates. So far, identification of CASP3 substrates has been performed mostly in apoptotic conditions where CASP3 activity is highly upregulated and these approaches do not have the capacity to uncover CASP3 substrates at the physiological level. In our study, we aim at discovering novel substrates of CASP3 involved in the normal regulation of the cell. We used an unconventional approach by chemically reducing the basal level CASP3-like activity (by DEVD-fmk treatment) coupled to a Mass Spectrometry screen (PISA) to identify proteins with different soluble amounts, and consequently, non-cleaved proteins in microglia cells. PISA assay identified several proteins with significant change in their solubility after DEVD-fmk treatment, including a few already known CASP3 substrates which validated our approach. Among them, we focused on the Collectin-12 (COLEC12 or CL-P1) transmembrane receptor and uncovered a potential role for CASP3 cleavage of COLEC12 in the regulation of the phagocytic capacity of microglial cells. Taken together, these findings suggest a new way to uncover non-apoptotic substrates of CASP3 important for the modulation of microglia cell physiology.
Collapse
|
7
|
Altered Extracellular Matrix as an Alternative Risk Factor for Epileptogenicity in Brain Tumors. Biomedicines 2022; 10:biomedicines10102475. [PMID: 36289737 PMCID: PMC9599244 DOI: 10.3390/biomedicines10102475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/23/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022] Open
Abstract
Seizures are one of the most common symptoms of brain tumors. The incidence of seizures differs among brain tumor type, grade, location and size, but paediatric-type diffuse low-grade gliomas/glioneuronal tumors are often highly epileptogenic. The extracellular matrix (ECM) is known to play a role in epileptogenesis and tumorigenesis because it is involved in the (re)modelling of neuronal connections and cell-cell signaling. In this review, we discuss the epileptogenicity of brain tumors with a focus on tumor type, location, genetics and the role of the extracellular matrix. In addition to functional problems, epileptogenic tumors can lead to increased morbidity and mortality, stigmatization and life-long care. The health advantages can be major if the epileptogenic properties of brain tumors are better understood. Surgical resection is the most common treatment of epilepsy-associated tumors, but post-surgery seizure-freedom is not always achieved. Therefore, we also discuss potential novel therapies aiming to restore ECM function.
Collapse
|
8
|
Dzhauari S, Litvinova S, Efimenko A, Aleksandrushkina N, Basalova N, Abakumov M, Danilova N, Malkov P, Balabanyan V, Bezuglova T, Balayants V, Mnikhovich M, Gulyaev M, Skryabina M, Popov V, Stambolsky D, Voronina T, Tkachuk V, Karagyaur M. Urokinase-Type Plasminogen Activator Enhances the Neuroprotective Activity of Brain-Derived Neurotrophic Factor in a Model of Intracerebral Hemorrhage. Biomedicines 2022; 10:biomedicines10061346. [PMID: 35740368 PMCID: PMC9220139 DOI: 10.3390/biomedicines10061346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 05/31/2022] [Accepted: 06/06/2022] [Indexed: 11/30/2022] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a classic neuroprotective and pro-regenerative factor in peripheral and central nervous tissue. Its ability to stimulate the restoration of damaged nerve and brain tissue after ischemic stroke and intraventricular hemorrhage has been demonstrated. However, the current concept of regeneration allows us to assert that one factor, even if essential, cannot be the sole contributor to this complex biological process. We have previously shown that urokinase-type plasminogen activator (uPA) complements BDNF activity and stimulates restoration of nervous tissue. Using a model of intracerebral hemorrhage in rats, we investigated the neurotrophic and neuroprotective effect of BDNF combined with uPA. The local simultaneous administration of BDNF and uPA provided effective neuroprotection of brain tissue after intracerebral hemorrhage, promoted survival of experimental animals and their neurological recovery, and decreased lesion volume. The study of cellular mechanisms of the observed neurotrophic effect of BDNF and uPA combination revealed both known mechanisms (neuronal survival and neurite growth) and new ones (microglial activation) that had not been shown for BDNF and uPA. Our findings support the concept of using combinations of biological factors with diverse but complementary mechanisms of action as a promising regenerative approach.
Collapse
Affiliation(s)
- Stalik Dzhauari
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovsky Ave., 119192 Moscow, Russia; (S.D.); (A.E.); (N.A.); (N.B.); (V.B.); (M.G.); (M.S.); (V.P.); (V.T.)
| | - Svetlana Litvinova
- Federal State Budgetary Institution “Research Zakusov Institute of Pharmacology”, 8, Baltiyskaya Str., 125315 Moscow, Russia; (S.L.); (T.V.)
| | - Anastasia Efimenko
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovsky Ave., 119192 Moscow, Russia; (S.D.); (A.E.); (N.A.); (N.B.); (V.B.); (M.G.); (M.S.); (V.P.); (V.T.)
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, 27/10, Lomonosovsky Ave., 119192 Moscow, Russia
| | - Natalia Aleksandrushkina
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovsky Ave., 119192 Moscow, Russia; (S.D.); (A.E.); (N.A.); (N.B.); (V.B.); (M.G.); (M.S.); (V.P.); (V.T.)
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, 27/10, Lomonosovsky Ave., 119192 Moscow, Russia
| | - Nataliya Basalova
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovsky Ave., 119192 Moscow, Russia; (S.D.); (A.E.); (N.A.); (N.B.); (V.B.); (M.G.); (M.S.); (V.P.); (V.T.)
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, 27/10, Lomonosovsky Ave., 119192 Moscow, Russia
| | - Maxim Abakumov
- Department of Medical Nanobiotechnology, National University of Science and Technology MISiS, 4, Leninskiy Ave., 119049 Moscow, Russia;
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, 1, Ostrovityanova Str., 117997 Moscow, Russia
| | - Natalia Danilova
- Medical Research and Education Center, Lomonosov Moscow State University, 27/10, Lomonosovsky Ave., 119192 Moscow, Russia; (N.D.); (P.M.); (D.S.)
| | - Pavel Malkov
- Medical Research and Education Center, Lomonosov Moscow State University, 27/10, Lomonosovsky Ave., 119192 Moscow, Russia; (N.D.); (P.M.); (D.S.)
| | - Vadim Balabanyan
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovsky Ave., 119192 Moscow, Russia; (S.D.); (A.E.); (N.A.); (N.B.); (V.B.); (M.G.); (M.S.); (V.P.); (V.T.)
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, 27/10, Lomonosovsky Ave., 119192 Moscow, Russia
| | - Tatiana Bezuglova
- Research Institute of Human Morphology, 3, Tsyurupy Str., 117418 Moscow, Russia; (T.B.); (V.B.); (M.M.)
| | - Viktor Balayants
- Research Institute of Human Morphology, 3, Tsyurupy Str., 117418 Moscow, Russia; (T.B.); (V.B.); (M.M.)
| | - Maxim Mnikhovich
- Research Institute of Human Morphology, 3, Tsyurupy Str., 117418 Moscow, Russia; (T.B.); (V.B.); (M.M.)
| | - Mikhail Gulyaev
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovsky Ave., 119192 Moscow, Russia; (S.D.); (A.E.); (N.A.); (N.B.); (V.B.); (M.G.); (M.S.); (V.P.); (V.T.)
| | - Mariya Skryabina
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovsky Ave., 119192 Moscow, Russia; (S.D.); (A.E.); (N.A.); (N.B.); (V.B.); (M.G.); (M.S.); (V.P.); (V.T.)
| | - Vladimir Popov
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovsky Ave., 119192 Moscow, Russia; (S.D.); (A.E.); (N.A.); (N.B.); (V.B.); (M.G.); (M.S.); (V.P.); (V.T.)
| | - Dmitry Stambolsky
- Medical Research and Education Center, Lomonosov Moscow State University, 27/10, Lomonosovsky Ave., 119192 Moscow, Russia; (N.D.); (P.M.); (D.S.)
| | - Tatiana Voronina
- Federal State Budgetary Institution “Research Zakusov Institute of Pharmacology”, 8, Baltiyskaya Str., 125315 Moscow, Russia; (S.L.); (T.V.)
| | - Vsevolod Tkachuk
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovsky Ave., 119192 Moscow, Russia; (S.D.); (A.E.); (N.A.); (N.B.); (V.B.); (M.G.); (M.S.); (V.P.); (V.T.)
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, 27/10, Lomonosovsky Ave., 119192 Moscow, Russia
| | - Maxim Karagyaur
- Faculty of Medicine, Lomonosov Moscow State University, 27/1, Lomonosovsky Ave., 119192 Moscow, Russia; (S.D.); (A.E.); (N.A.); (N.B.); (V.B.); (M.G.); (M.S.); (V.P.); (V.T.)
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, 27/10, Lomonosovsky Ave., 119192 Moscow, Russia
- Correspondence:
| |
Collapse
|
9
|
De Almeida SM, Rotta I, Tang B, Umlauf A, Vaida F, Cherner M, Franklin D, Letendre S, Ellis RJ. Higher Cerebrospinal Fluid Soluble Urokinase-type Plasminogen Activator Receptor, But Not Interferon γ-inducible Protein 10, Correlate With Higher Working Memory Deficits. J Acquir Immune Defic Syndr 2022; 90:106-114. [PMID: 35090158 PMCID: PMC8986587 DOI: 10.1097/qai.0000000000002924] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Accepted: 01/18/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND We hypothesized that the induction of monocyte activation biomarkers, especially soluble urokinase-type plasminogen activator receptor (suPAR) and interferon γ-inducible protein 10 (IP-10), is lower in HIV-1C than HIV-1B, owing to a defective Tat cysteine dimotif (C30S). METHODS A total of 68 paired cerebrospinal fluid (CSF) and blood samples from people with HIV (PWH), free of CNS opportunistic infections, from a Southern Brazil outpatient HIV clinic were evaluated such as HIV-1B subtype (n = 27), HIV-1C (n = 26), other (n = 15), and 19 HIV-negative controls. The levels of suPAR, IP-10, neopterin, and β2 microglobulin (β2m) in the CSF and serum were quantified using different immunoassays. RESULTS Overall, in PWH, increases in CSF suPAR, CSF/serum suPAR, and CSF/serum β2m correlated with worse working memory deficits (r = 0.303, 0.353, and 0.289, respectively, all P < 0.05). The medians of IP-10, suPAR, neopterin, and β2m in CSF and serum and the CSF/serum ratio and suPAR index were comparable between the HIV-1B and HIV-1C subtypes. CSF IP-10 and neopterin and serum IP-10 and suPAR levels were higher in PWH than the HIV-negative controls (P = 0.015, P = 0.001, P < 0.0001, and P < 0.001, respectively). The serum β2m level was higher in HIV-associated dementia than neuropsychologically normal or asymptomatic (P = 0.024). DISCUSSION We observed that higher levels of CSF suPAR and the suPAR quotient correlated with worse working memory deficit. Elevated levels of monocyte activation were similar in both HIV-1 B and C subtypes, providing no evidence of reduced neuropathogenicity of HIV-1 subtype C Tat compared with subtype B.
Collapse
Affiliation(s)
- Sergio M De Almeida
- Neuroinfection Unity and Virology Laboratory, Universidade Federal Do Paraná, Curitiba, Paraná, Brazil
| | - Indianara Rotta
- Neuroinfection Unity and Virology Laboratory, Universidade Federal Do Paraná, Curitiba, Paraná, Brazil
| | - Bin Tang
- Department of Psychiatry, University of California, San Diego, CA
| | - Anya Umlauf
- Department of Psychiatry, University of California, San Diego, CA
| | - Florin Vaida
- Division of Biostatistics and Bioinformatics, Department of Family Medicine and Public Health, University of California, San Diego, CA
| | - Mariana Cherner
- Department of Psychiatry, University of California, San Diego, CA
- HIV Neurobehavioral Research Center, University of California, San Diego, CA
| | - Donald Franklin
- HIV Neurobehavioral Research Center, University of California, San Diego, CA
| | - Scott Letendre
- HIV Neurobehavioral Research Center, University of California, San Diego, CA
- Division of Infectious Diseases, Department of Medicine, University of California, San Diego, CA; and
| | - Ronald J Ellis
- HIV Neurobehavioral Research Center, University of California, San Diego, CA
- Department of Neurosciences, University of California, San Diego, CA
| |
Collapse
|
10
|
Patel T, Carnwath TP, Wang X, Allen M, Lincoln SJ, Lewis‐Tuffin L, Quicksall ZS, Lin S, Tutor‐New FQ, Ho CC, Min Y, Malphrus KG, Nguyen TT, Martin E, Garcia CA, Alkharboosh RM, Grewal S, Chaichana K, Wharen R, Guerrero‐Cazares H, Quinones‐Hinojosa A, Ertekin‐Taner N. Transcriptional landscape of human microglia implicates age, sex, and APOE-related immunometabolic pathway perturbations. Aging Cell 2022; 21:e13606. [PMID: 35388616 PMCID: PMC9124307 DOI: 10.1111/acel.13606] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/19/2022] [Accepted: 03/21/2022] [Indexed: 12/20/2022] Open
Abstract
Microglia have fundamental roles in health and disease; however, effects of age, sex, and genetic factors on human microglia have not been fully explored. We applied bulk and single-cell approaches to comprehensively characterize human microglia transcriptomes and their associations with age, sex, and APOE. We identified a novel microglial signature, characterized its expression in bulk tissue and single-cell microglia transcriptomes. We discovered microglial co-expression network modules associated with age, sex, and APOE-ε4 that are enriched for lipid and carbohydrate metabolism genes. Integrated analyses of modules with single-cell transcriptomes revealed significant overlap between age-associated module genes and both pro-inflammatory and disease-associated microglial clusters. These modules and clusters harbor known neurodegenerative disease genes including APOE, PLCG2, and BIN1. Meta-analyses with published bulk and single-cell microglial datasets further supported our findings. Thus, these data represent a well-characterized human microglial transcriptome resource and highlight age, sex, and APOE-related microglial immunometabolism perturbations with potential relevance in neurodegeneration.
Collapse
Affiliation(s)
- Tulsi Patel
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | | | - Xue Wang
- Department of Quantitative Health SciencesMayo ClinicJacksonvilleFloridaUSA
| | - Mariet Allen
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | | | | | | | - Shu Lin
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | | | | | - Yuhao Min
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | | | - Thuy T. Nguyen
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
| | | | | | - Rawan M. Alkharboosh
- Department of NeurosurgeryMayo ClinicJacksonvilleFloridaUSA
- Neuroscience Graduate ProgramMayo Clinic Graduate School of Biomedical SciencesMayo ClinicRochesterMinnesotaUSA
- Regenerative Sciences Training ProgramCenter for Regenerative MedicineMayo ClinicRochesterMinnesotaUSA
| | - Sanjeet Grewal
- Department of NeurosurgeryMayo ClinicJacksonvilleFloridaUSA
| | | | - Robert Wharen
- Department of NeurosurgeryMayo ClinicJacksonvilleFloridaUSA
| | | | | | - Nilüfer Ertekin‐Taner
- Department of NeuroscienceMayo ClinicJacksonvilleFloridaUSA
- Department of NeurologyMayo ClinicJacksonvilleFloridaUSA
| |
Collapse
|
11
|
Hyperglycemia Promotes Endothelial Cell Senescence through AQR/PLAU Signaling Axis. Int J Mol Sci 2022; 23:ijms23052879. [PMID: 35270021 PMCID: PMC8911151 DOI: 10.3390/ijms23052879] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/23/2022] [Accepted: 03/02/2022] [Indexed: 02/06/2023] Open
Abstract
Hyperglycemia is reported to accelerate endothelial cell senescence that contributes to diabetic complications. The underlying mechanism, however, remains elusive. We previously demonstrated AQR as a susceptibility gene for type 2 diabetes mellitus (T2DM) and showed that it was increased in multiple tissues in models with T2DM or metabolic syndrome. This study aimed to investigate the role of AQR in hyperglycemia-induced senescence and its underlying mechanism. Here, we retrieved several datasets of the aging models and found the expression of AQR was increased by high glucose and by aging across species, including C. elegans (whole-body), rat (cardiac tissues), and monkey (blood). we validated the increased AQR expression in senescent human umbilical vein endothelial cells (HUVECs). When overexpressed, AQR promoted the endothelial cell senescence, confirmed by an increased number of cells stained with senescence-associated beta-galactosidase and upregulation of CDKN1A (P21) as well as the prohibited cellular colony formation and G2/M phase arrest. To explore the mechanism by which AQR regulated the cellular senescence, transcriptomic analyses of HUVECs with the overexpression and knockdown of the AQR were performed. We identified 52 co-expressed genes that were enriched, in the terms of plasminogen activation, innate immunity, immunity, and antiviral defense. Among co-expressed genes, PLAU was selected to evaluate its contribution to senescence for its highest strength in the enrichment of the biological process. We demonstrated that the knockdown of PLAU rescued senescence-related phenotypes, endothelial cell activation, and inflammation in models induced by AQR or TNF-α. These findings, for the first time, indicate that AQR/PLAU is a critical signaling axis in the modulation of endothelial cell senescence, revealing a novel link between hyperglycemia and vascular dysfunction. The study may have implications in the prevention of premature vascular aging associated with T2DM.
Collapse
|
12
|
Karunia J, Niaz A, Mandwie M, Thomas Broome S, Keay KA, Waschek JA, Al-Badri G, Castorina A. PACAP and VIP Modulate LPS-Induced Microglial Activation and Trigger Distinct Phenotypic Changes in Murine BV2 Microglial Cells. Int J Mol Sci 2021; 22:ijms222010947. [PMID: 34681607 PMCID: PMC8535941 DOI: 10.3390/ijms222010947] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/01/2021] [Accepted: 10/04/2021] [Indexed: 01/01/2023] Open
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal peptide (VIP) are two structurally related immunosuppressive peptides. However, the underlying mechanisms through which these peptides regulate microglial activity are not fully understood. Using lipopolysaccharide (LPS) to induce an inflammatory challenge, we tested whether PACAP or VIP differentially affected microglial activation, morphology and cell migration. We found that both peptides attenuated LPS-induced expression of the microglial activation markers Iba1 and iNOS (### p < 0.001), as well as the pro-inflammatory mediators IL-1β, IL-6, Itgam and CD68 (### p < 0.001). In contrast, treatment with PACAP or VIP exerted distinct effects on microglial morphology and migration. PACAP reversed LPS-induced soma enlargement and increased the percentage of small-sized, rounded cells (54.09% vs. 12.05% in LPS-treated cells), whereas VIP promoted a phenotypic shift towards cell subpopulations with mid-sized, spindle-shaped somata (48.41% vs. 31.36% in LPS-treated cells). Additionally, PACAP was more efficient than VIP in restoring LPS-induced impairment of cell migration and the expression of urokinase plasminogen activator (uPA) in BV2 cells compared with VIP. These results suggest that whilst both PACAP and VIP exert similar immunosuppressive effects in activated BV2 microglia, each peptide triggers distinctive shifts towards phenotypes of differing morphologies and with differing migration capacities.
Collapse
Affiliation(s)
- Jocelyn Karunia
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (J.K.); (A.N.); (M.M.); (S.T.B.); (G.A.-B.)
| | - Aram Niaz
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (J.K.); (A.N.); (M.M.); (S.T.B.); (G.A.-B.)
| | - Mawj Mandwie
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (J.K.); (A.N.); (M.M.); (S.T.B.); (G.A.-B.)
| | - Sarah Thomas Broome
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (J.K.); (A.N.); (M.M.); (S.T.B.); (G.A.-B.)
| | - Kevin A. Keay
- School of Medical Science, [Neuroscience] and Brain and Mind Centre, The University of Sydney, Sydney, NSW 2006, Australia;
| | - James A. Waschek
- Intellectual Development and Disabilities Research Centre, Semel Institute for Neuroscience and Human Behaviour/Neuropsychiatric Institute, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA 90095, USA;
| | - Ghaith Al-Badri
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (J.K.); (A.N.); (M.M.); (S.T.B.); (G.A.-B.)
| | - Alessandro Castorina
- Laboratory of Cellular and Molecular Neuroscience (LCMN), School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (J.K.); (A.N.); (M.M.); (S.T.B.); (G.A.-B.)
- School of Medical Science, [Neuroscience] and Brain and Mind Centre, The University of Sydney, Sydney, NSW 2006, Australia;
- Correspondence:
| |
Collapse
|
13
|
Ziliotto N, Bernardi F, Piazza F. Hemostasis components in cerebral amyloid angiopathy and Alzheimer's disease. Neurol Sci 2021; 42:3177-3188. [PMID: 34041636 DOI: 10.1007/s10072-021-05327-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Accepted: 05/15/2021] [Indexed: 01/17/2023]
Abstract
Increased cerebrovascular amyloid-β (Aβ) deposition represents the main pathogenic mechanisms characterizing Alzheimer's disease (AD) and cerebral amyloid angiopathy (CAA). Whereas an increasing number of studies define the contribution of fibrin(ogen) to neurodegeneration, how other hemostasis factors might be pleiotropically involved in the AD and CAA remains overlooked. Although traditionally regarded as pertaining to hemostasis, these proteins are also modulators of inflammation and angiogenesis, and exert cytoprotective functions. This review discusses the contribution of hemostasis components to Aβ cerebrovascular deposition, which settle the way to endothelial and blood-brain barrier dysfunction, vessel fragility, cerebral bleeding, and the associated cognitive changes. From the primary hemostasis, the process that refers to platelet aggregation, we discuss evidence regarding the von Willebrand factor (vWF) and its regulator ADAMTS13. Then, from the secondary hemostasis, we focus on tissue factor, which triggers the extrinsic coagulation cascade, and on the main inhibitors of coagulation, i.e., tissue factor pathway inhibitor (TFPI), and the components of protein C pathway. Last, from the tertiary hemostasis, we discuss evidence on FXIII, involved in fibrin cross-linking, and on components of fibrinolysis, including tissue-type plasminogen activator (tPA), urokinase-type plasminogen activator (uPA) and its receptor uPA(R), and plasminogen activator inhibitor-1 (PAI-1). Increased knowledge on contributors of Aβ-related disease progression may favor new therapeutic approaches for early modifiable risk factors.
Collapse
Affiliation(s)
- Nicole Ziliotto
- CAA and AD Translational Research and Biomarkers Laboratory, School of Medicine and Surgery, University of Milano - Bicocca, Via Cadore 48, 20900, Monza, Italy.
| | - Francesco Bernardi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Fabrizio Piazza
- CAA and AD Translational Research and Biomarkers Laboratory, School of Medicine and Surgery, University of Milano - Bicocca, Via Cadore 48, 20900, Monza, Italy
| |
Collapse
|
14
|
Plasminogen Activators in Neurovascular and Neurodegenerative Disorders. Int J Mol Sci 2021; 22:ijms22094380. [PMID: 33922229 PMCID: PMC8122722 DOI: 10.3390/ijms22094380] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 04/14/2021] [Accepted: 04/20/2021] [Indexed: 12/14/2022] Open
Abstract
The neurovascular unit (NVU) is a dynamic structure assembled by endothelial cells surrounded by a basement membrane, pericytes, astrocytes, microglia and neurons. A carefully coordinated interplay between these cellular and non-cellular components is required to maintain normal neuronal function, and in line with these observations, a growing body of evidence has linked NVU dysfunction to neurodegeneration. Plasminogen activators catalyze the conversion of the zymogen plasminogen into the two-chain protease plasmin, which in turn triggers a plethora of physiological events including wound healing, angiogenesis, cell migration and inflammation. The last four decades of research have revealed that the two mammalian plasminogen activators, tissue-type plasminogen activator (tPA) and urokinase-type plasminogen activator (uPA), are pivotal regulators of NVU function during physiological and pathological conditions. Here, we will review the most relevant data on their expression and function in the NVU and their role in neurovascular and neurodegenerative disorders.
Collapse
|
15
|
Marogianni C, Sokratous M, Dardiotis E, Hadjigeorgiou GM, Bogdanos D, Xiromerisiou G. Neurodegeneration and Inflammation-An Interesting Interplay in Parkinson's Disease. Int J Mol Sci 2020; 21:E8421. [PMID: 33182554 PMCID: PMC7697354 DOI: 10.3390/ijms21228421] [Citation(s) in RCA: 190] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 10/30/2020] [Accepted: 11/06/2020] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder, caused by, so far, unknown pathogenetic mechanisms. There is no doubt that pro-inflammatory immune-mediated mechanisms are pivotal to the pathogenicity and progression of the disease. In this review, we highlight the binary role of microglia activation in the pathophysiology of the disorder, both neuroprotective and neuromodulatory. We present how the expression of several cytokines implicated in dopaminergic neurons (DA) degeneration could be used as biomarkers for PD. Viral infections have been studied and correlated to the disease progression, usually operating as trigger factors for the inflammatory process. The gut-brain axis and the possible contribution of the peripheral bowel inflammation to neuronal death, mainly dopaminergic neurons, seems to be a main contributor of brain neuroinflammation. The role of the immune system has also been analyzed implicating a-synuclein in the activation of innate and adaptive immunity. We also discuss therapeutic approaches concerning PD and neuroinflammation, which have been studied in experimental and in vitro models and data stemming from epidemiological studies.
Collapse
Affiliation(s)
- Chrysoula Marogianni
- Department of Neurology, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41110 Larissa, Greece; (C.M.); (M.S.); (E.D.)
| | - Maria Sokratous
- Department of Neurology, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41110 Larissa, Greece; (C.M.); (M.S.); (E.D.)
| | - Efthimios Dardiotis
- Department of Neurology, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41110 Larissa, Greece; (C.M.); (M.S.); (E.D.)
| | | | - Dimitrios Bogdanos
- Department of Internal Medicine, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41110 Larissa, Greece;
| | - Georgia Xiromerisiou
- Department of Neurology, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41110 Larissa, Greece; (C.M.); (M.S.); (E.D.)
| |
Collapse
|
16
|
Robust Dopaminergic Differentiation and Enhanced LPS-Induced Neuroinflammatory Response in Serum-Deprived Human SH-SY5Y Cells: Implication for Parkinson's Disease. J Mol Neurosci 2020; 71:565-582. [PMID: 32789724 DOI: 10.1007/s12031-020-01678-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 08/03/2020] [Indexed: 01/07/2023]
Abstract
Parkinson's disease (PD) is a chronic neurodegenerative condition characterized by motor symptoms such as bradykinesia, resting tremor, and rigidity. PD diagnosis is based on medical history, review of signs, symptoms, neurological and physical examinations. Unfortunately, by the time the disease is diagnosed, dopamine (DA) neuronal loss is often extended, thereby resulting in ineffective therapies. Recent evidence suggests that neuroinflammation may be pivotal during PD onset and progression. However, suitable cellular models and biomarkers to detect early signs of neuroinflammation are still missing. In this study, we developed a well-differentiated DAergic neuronal cell line where we triggered a neuroinflammatory response to assess the temporal expression of the tissue- and urokinase plasminogen activators (tPA and uPA) and their endogenous inhibitor (PAI-1) along with that of pro-inflammatory mediators and the neuronal marker nNOS. Human neuroblastoma cells SH-SY5Y were differentiated into DAergic neuronal-like cells using a combination of 12-O-tetradecanoylphorbol-13-acetate (TPA) and serum depletion. Terminally-differentiated neurons were then exposed to lipopolysaccharide (LPS) for short (up to 24 h) or long term (up to 10 days) to mimic acute or chronic inflammation. Results demonstrated that uPA protein expression was stably upregulated during chronic inflammation, whereas the expression of nNOS protein better reflected the cellular response to acute inflammation. Additional studies revealed that the temporal induction of uPA was associated with increased AKT phosphorylation, but did not seem to involve cAMP-responsive element-binding protein (CREB) activation, nor the mitogen-activated protein kinase (MAPK) pathway. In conclusion, our in vitro data suggests that nNOS and uPA may serve as viable candidate biomarkers of acute and chronic neuroinflammation.
Collapse
|
17
|
Baart VM, Houvast RD, de Geus-Oei LF, Quax PHA, Kuppen PJK, Vahrmeijer AL, Sier CFM. Molecular imaging of the urokinase plasminogen activator receptor: opportunities beyond cancer. EJNMMI Res 2020; 10:87. [PMID: 32725278 PMCID: PMC7387399 DOI: 10.1186/s13550-020-00673-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Accepted: 07/14/2020] [Indexed: 02/07/2023] Open
Abstract
The urokinase plasminogen activator receptor (uPAR) plays a multifaceted role in almost any process where migration of cells and tissue-remodeling is involved such as inflammation, but also in diseases as arthritis and cancer. Normally, uPAR is absent in healthy tissues. By its carefully orchestrated interaction with the protease urokinase plasminogen activator and its inhibitor (plasminogen activator inhibitor-1), uPAR localizes a cascade of proteolytic activities, enabling (patho)physiologic cell migration. Moreover, via the interaction with a broad range of cell membrane proteins, like vitronectin and various integrins, uPAR plays a significant, but not yet completely understood, role in differentiation and proliferation of cells, affecting also disease progression. The implications of these processes, either for diagnostics or therapeutics, have received much attention in oncology, but only limited beyond. Nonetheless, the role of uPAR in different diseases provides ample opportunity to exploit new applications for targeting. Especially in the fields of oncology, cardiology, rheumatology, neurology, and infectious diseases, uPAR-targeted molecular imaging could offer insights for new directions in diagnosis, surveillance, or treatment options.
Collapse
Affiliation(s)
- V M Baart
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - R D Houvast
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - L F de Geus-Oei
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, The Netherlands.,Biomedical Photonic Imaging Group, University of Twente, Enschede, The Netherlands
| | - P H A Quax
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - P J K Kuppen
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - A L Vahrmeijer
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - C F M Sier
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands. .,Percuros BV, Leiden, The Netherlands.
| |
Collapse
|
18
|
Ellegaard Nielsen J, Sofie Pedersen K, Vestergård K, Georgiana Maltesen R, Christiansen G, Lundbye-Christensen S, Moos T, Risom Kristensen S, Pedersen S. Novel Blood-Derived Extracellular Vesicle-Based Biomarkers in Alzheimer's Disease Identified by Proximity Extension Assay. Biomedicines 2020; 8:E199. [PMID: 32645971 PMCID: PMC7400538 DOI: 10.3390/biomedicines8070199] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 06/29/2020] [Accepted: 07/03/2020] [Indexed: 12/14/2022] Open
Abstract
Easily accessible biomarkers for Alzheimer's dementia (AD) are lacking and established clinical markers are limited in applicability. Blood is a common biofluid for biomarker discoveries, and extracellular vesicles (EVs) may provide a matrix for exploring AD related biomarkers. Thus, we investigated proteins related to neurological and inflammatory processes in plasma and EVs. By proximity extension assay (PEA), 182 proteins were measured in plasma and EVs from patients with AD (n = 10), Mild Cognitive Impairment (MCI, n = 10), and healthy controls (n = 10). Plasma-derived EVs were enriched by 20,000× g, 1 h, 4 °C, and confirmed using nanoparticle tracking analysis (NTA), western blotting, and transmission electron microscopy with immunolabelling (IEM). Presence of CD9+ EVs was confirmed by western blotting and IEM. No group differences in particle concentration or size were detected by NTA. However, significant protein profiles were observed among subjects, particularly for EVs. Several proteins and their ratios could distinguish cognitively affected from healthy individuals. For plasma TGF-α│CCL20 (AUC = 0.96, 95% CI = 0.88-1.00, p = 0.001) and EVs CLEC1B│CCL11 (AUC = 0.95, 95% CI = 0.86-1.00, p = 0.001) showed diagnostic capabilities. Using PEA, we identified protein profiles capable of distinguishing healthy controls from AD patients. EVs provided additional biological information related to AD not observed in plasma alone.
Collapse
Affiliation(s)
- Jonas Ellegaard Nielsen
- Department of Clinical Medicine, Aalborg University, DK-9000 Aalborg, Denmark; (S.L.-C.); (S.R.K.)
- Department of Clinical Biochemistry, Aalborg University Hospital, DK-9000 Aalborg, Denmark
| | | | - Karsten Vestergård
- Department of Neurology, Aalborg University Hospital, DK-9000 Aalborg, Denmark;
| | - Raluca Georgiana Maltesen
- Department of Anaesthesia and Intensive Care, Aalborg University Hospital, DK-9000 Aalborg, Denmark;
| | - Gunna Christiansen
- Department of Biomedicine, Aarhus University, DK-8000 Aarhus, Denmark;
- Department of Health Science and Technology, Aalborg University, DK-9220 Aalborg, Denmark;
| | - Søren Lundbye-Christensen
- Department of Clinical Medicine, Aalborg University, DK-9000 Aalborg, Denmark; (S.L.-C.); (S.R.K.)
- Unit of Clinical Biostatistics, Aalborg University Hospital, DK-9000 Aalborg, Denmark
| | - Torben Moos
- Department of Health Science and Technology, Aalborg University, DK-9220 Aalborg, Denmark;
| | - Søren Risom Kristensen
- Department of Clinical Medicine, Aalborg University, DK-9000 Aalborg, Denmark; (S.L.-C.); (S.R.K.)
- Department of Clinical Biochemistry, Aalborg University Hospital, DK-9000 Aalborg, Denmark
| | - Shona Pedersen
- Department of Clinical Medicine, Aalborg University, DK-9000 Aalborg, Denmark; (S.L.-C.); (S.R.K.)
- Department of Clinical Biochemistry, Aalborg University Hospital, DK-9000 Aalborg, Denmark
| |
Collapse
|
19
|
Kaesmacher J, Bellwald S, Dobrocky T, Meinel TR, Piechowiak EI, Goeldlin M, Kurmann CC, Heldner MR, Jung S, Mordasini P, Arnold M, Mosimann PJ, Schroth G, Mattle HP, Gralla J, Fischer U. Safety and Efficacy of Intra-arterial Urokinase After Failed, Unsuccessful, or Incomplete Mechanical Thrombectomy in Anterior Circulation Large-Vessel Occlusion Stroke. JAMA Neurol 2020; 77:318-326. [PMID: 31816018 PMCID: PMC6902179 DOI: 10.1001/jamaneurol.2019.4192] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 09/27/2019] [Indexed: 12/29/2022]
Abstract
Importance Achieving complete reperfusion is a key determinant of good outcome in patients treated with mechanical thrombectomy (MT). However, data on treatments geared toward improving reperfusion after incomplete MT are sparse. Objective To determine whether administration of intra-arterial urokinase is safe and improves reperfusion after failed or incomplete MT. Design, Setting, and Participants This observational cohort study included a consecutive sample of patients treated with second-generation MT from January 1, 2010, through August 4, 2017. Data were collected from the prospective registry of a tertiary care stroke center. Of 1274 patients screened, 69 refused to participate, and 993 met the observational studies inclusion criteria of a large vessel occlusion in the anterior circulation. Data were analyzed from September 1, 2017, through September 20, 2019. Intervention One hundred patients received intra-arterial urokinase after failed or incomplete MT using manual microcatheter injections. Main Outcomes and Measures Primary safety outcome was the occurrence of symptomatic intracranial hemorrhage (sICH) according to the Prolyse in Acute Cerebral Thromboembolism II criteria. Secondary end points included 90-day mortality and 90-day functional independence (defined as modified Rankin Scale score of ≤2). Efficacy was evaluated angiographically, applying the Thrombolysis in Cerebral Infarction (TICI) scale. Results After exclusion of patients with posterior circulation strokes and those treated with intra-arterial thrombolytics only, 993 patients were included in the final analyses (median age, 74.6 [interquartile range, 62.6-82.2] years; 505 [50.9%] women). Additional intra-arterial urokinase was administered in 100 patients (10.1%). The most common reason for administering intra-arterial urokinase was incomplete reperfusion (TICI<3) after MT (53 [53.0%]). After adjusting for baseline characteristics underlying case selection, intra-arterial urokinase was not associated with an increased risk of sICH (adjusted odds ratio [aOR], 0.81; 95% CI, 0.31-2.13) or 90-day mortality (aOR, 0.78; 95% CI, 0.43-1.40). Among 53 cases of partial or near-complete reperfusion and treated with intra-arterial urokinase, 32 (60.4%) had early reperfusion improvement, and 18 of 53 (34.0%) had an improvement in TICI grade. Correspondingly, patients treated with intra-arterial urokinase had higher rates of functional independence after adjusting for the selection bias favoring a priori poor TICI grades in the intra-arterial urokinase group (aOR, 1.93; 95% CI, 1.11-3.37). Conclusions and Relevance In selected patients, adjunctive treatment with intra-arterial urokinase during or after MT was safe and improved angiographic reperfusion. Systemic evaluation of this approach in a multicenter prospective registry or a randomized clinical trial seems warranted.
Collapse
Affiliation(s)
- Johannes Kaesmacher
- University Institute of Diagnostic and Interventional Neuroradiology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
- University Institute of Diagnostic, Interventional and Pediatric Radiology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
| | - Sebastian Bellwald
- Department of Neurology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
| | - Tomas Dobrocky
- University Institute of Diagnostic and Interventional Neuroradiology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
| | - Thomas R. Meinel
- Department of Neurology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
| | - Eike I. Piechowiak
- University Institute of Diagnostic and Interventional Neuroradiology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
| | - Martina Goeldlin
- University Institute of Diagnostic and Interventional Neuroradiology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
- Department of Neurology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
| | - Christoph C. Kurmann
- University Institute of Diagnostic and Interventional Neuroradiology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
| | - Mirjam R. Heldner
- Department of Neurology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
| | - Simon Jung
- Department of Neurology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
| | - Pasquale Mordasini
- University Institute of Diagnostic and Interventional Neuroradiology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
| | - Marcel Arnold
- Department of Neurology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
| | - Pascal J. Mosimann
- University Institute of Diagnostic and Interventional Neuroradiology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
| | - Gerhard Schroth
- University Institute of Diagnostic and Interventional Neuroradiology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
| | - Heinrich P. Mattle
- Department of Neurology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
| | - Jan Gralla
- University Institute of Diagnostic and Interventional Neuroradiology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
| | - Urs Fischer
- Department of Neurology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
| |
Collapse
|
20
|
Yepes M. Urokinase-type plasminogen activator is a modulator of synaptic plasticity in the central nervous system: implications for neurorepair in the ischemic brain. Neural Regen Res 2020; 15:620-624. [PMID: 31638083 PMCID: PMC6975136 DOI: 10.4103/1673-5374.266904] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The last two decades have witnessed a rapid decrease in mortality due to acute cerebral ischemia that paradoxically has led to a rapid increase in the number of patients that survive an acute ischemic stroke with various degrees of disability. Unfortunately, the lack of an effective therapeutic strategy to promote neurological recovery among stroke survivors has led to a rapidly growing population of disabled patients. Thus, understanding the mechanisms of neurorepair in the ischemic brain is a priority with wide scientific, social and economic implications. Cerebral ischemia has a harmful effect on synaptic structure associated with the development of functional impairment. In agreement with these observations, experimental evidence indicates that synaptic repair underlies the recovery of neurological function following an ischemic stroke. Furthermore, it has become evident that synaptic plasticity is crucial not only during development and learning, but also for synaptic repair after an ischemic insult. The plasminogen activating system is assembled by a cascade of enzymes and their inhibitors initially thought to be solely involved in the generation of plasmin. However, recent work has shown that in the brain this system has an important function regulating the development of synaptic plasticity via mechanisms that not always require plasmin generation. Urokinase-type plasminogen activator (uPA) is a serine proteinase and one of the plasminogen activators, that upon binding to its receptor (uPAR) not only catalyzes the conversion of plasminogen into plasmin on the cell surface, but also activates cell signaling pathways that promote cell migration, proliferation and survival. The role of uPA is the brain is not fully understood. However, it has been reported while uPA and uPAR are abundantly found in the developing central nervous system, in the mature brain their expression is restricted to a limited group of cells. Remarkably, following an ischemic injury to the mature brain the expression of uPA and uPAR increases to levels comparable to those observed during development. More specifically, neurons release uPA during the recovery phase from an ischemic injury, and astrocytes, axonal boutons and dendritic spines recruit uPAR to their plasma membrane. Here we will review recent evidence indicating that binding of uPA to uPAR promotes the repair of synapses damaged by an ischemic injury, with the resultant recovery of neurological function. Furthermore, we will discuss data indicating that treatment with recombinant uPA is a potential therapeutic strategy to promote neurological recovery among ischemic stroke survivors.
Collapse
Affiliation(s)
- Manuel Yepes
- Division of Neuropharmacology and Neurologic Diseases, Yerkes National Primate Research Center; Department of Neurology, Emory University School of Medicine; Department of Neurology, Veterans Affairs Medical Center, Atlanta, GA, USA
| |
Collapse
|
21
|
Pontecorvi P, Banki MA, Zampieri C, Zalfa C, Azmoon P, Kounnas MZ, Marchese C, Gonias SL, Mantuano E. Fibrinolysis protease receptors promote activation of astrocytes to express pro-inflammatory cytokines. J Neuroinflammation 2019; 16:257. [PMID: 31810478 PMCID: PMC6896679 DOI: 10.1186/s12974-019-1657-3] [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] [Received: 08/20/2019] [Accepted: 11/25/2019] [Indexed: 12/23/2022] Open
Abstract
Background Astrocytes contribute to the crosstalk that generates chronic neuro-inflammation in neurological diseases; however, compared with microglia, astrocytes respond to a more limited continuum of innate immune system stimulants. Recent studies suggest that the fibrinolysis system may regulate inflammation. The goal of this study was to test whether fibrinolysis system components activate astrocytes and if so, elucidate the responsible biochemical pathway. Methods Primary cultures of astrocytes and microglia were prepared from neonatal mouse brains. The ability of purified fibrinolysis system proteins to elicit a pro-inflammatory response was determined by measuring expression of the mRNAs encoding tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), and chemokine (C-C motif) ligand 2 (CCL2). IκBα phosphorylation also was measured. Plasminogen activation in association with cells was detected by chromogenic substrate hydrolysis. The activity of specific receptors was tested using neutralizing antibodies and reagents. Results Astrocytes expressed pro-inflammatory cytokines when treated with plasminogen but not when treated with agonists for Toll-like Receptor-4 (TLR4), TLR2, or TLR9. Microglia also expressed pro-inflammatory cytokines in response to plasminogen; however, in these cells, the response was observed only when tissue-type plasminogen activator (tPA) was added to activate plasminogen. In astrocytes, endogenously produced urokinase-type plasminogen activator (uPA) converted plasminogen into plasmin in the absence of tPA. Plasminogen activation was dependent on the plasminogen receptor, α-enolase, and the uPA receptor, uPAR. Although uPAR is capable of directly activating cell-signaling, the receptor responsible for cytokine expression and IκBα phosphorylation response to plasmin was Protease-activated Receptor-1 (PAR-1). The pathway, by which plasminogen induced astrocyte activation, was blocked by inhibiting any one of the three receptors implicated in this pathway with reagents such as εACA, α-enolase-specific antibody, uPAR-specific antibody, the uPA amino terminal fragment, or a pharmacologic PAR-1 inhibitor. Conclusions Plasminogen may activate astrocytes for pro-inflammatory cytokine expression through the concerted action of at least three distinct fibrinolysis protease receptors. The pathway is dependent on uPA to activate plasminogen, which is expressed endogenously by astrocytes in culture but also may be provided by other cells in the astrocytic cell microenvironment in the CNS.
Collapse
Affiliation(s)
- Paola Pontecorvi
- The Department of Pathology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0612, USA.,The Department of Experimental Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Michael A Banki
- The Department of Pathology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0612, USA
| | - Carlotta Zampieri
- The Department of Pathology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0612, USA.,The Department of Chemical Sciences and Technologies, Tor Vergata University of Rome, 00133, Rome, Italy
| | - Cristina Zalfa
- The Department of Pathology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0612, USA
| | - Pardis Azmoon
- The Department of Pathology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0612, USA
| | - Maria Z Kounnas
- The Department of Pathology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0612, USA
| | - Cinzia Marchese
- The Department of Experimental Medicine, Sapienza University of Rome, 00161, Rome, Italy
| | - Steven L Gonias
- The Department of Pathology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0612, USA.
| | - Elisabetta Mantuano
- The Department of Pathology, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0612, USA.
| |
Collapse
|
22
|
Girard RA, Chauhan PS, Tucker TA, Allen T, Kaur J, Jeffers A, Koenig K, Florova G, Komissarov AA, Gaidenko TA, Chamiso MB, Fowler J, Morris DE, Sarva K, Singh KP, Idell S, Idell RD. Increased expression of plasminogen activator inhibitor-1 (PAI-1) is associated with depression and depressive phenotype in C57Bl/6J mice. Exp Brain Res 2019; 237:3419-3430. [PMID: 31734788 DOI: 10.1007/s00221-019-05682-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2019] [Accepted: 11/07/2019] [Indexed: 02/07/2023]
Abstract
Plasminogen activator inhibitor 1 (PAI-1), which is elevated in numerous disease states, has been implicated as a stress-related protein involved in the pathogenesis of depression. We measured PAI-1 in the plasma of healthy and depressed individuals and assessed plasminogen activator (PA) expression and regulation by PAI-1 in cultured normal human astrocytes (NHA). Elevated plasma PAI-1 levels were found in depressed patients. Brain tissues from depressed individuals also showed stronger expression of hippocampal PAI-1 by confocal imaging in comparison to healthy individuals. Using a lipopolysaccharide-induced inflammatory model of depression in mice, we measured PAI-1 in murine plasma and brain, by ELISA and immunohistochemistry, respectively. Similar elevations were seen in plasma but not in brain homogenates of mice exposed to LPS. We further correlated the findings with depressive behavior. Ex vivo experiments with NHA treated with proinflammatory cytokines implicated in the pathogenesis of depression showed increased PAI-1 expression. Furthermore, these studies suggest that urokinase-type plasminogen activator may serve as an astrocyte PA reservoir, able to promote cleavage of brain-derived neurotrophic factor (BDNF) during stress or inflammation. In summary, our findings confirm that derangements of PAI-1 variably occur in the brain in association with the depressive phenotype. These derangements may impede the availability of active, mature (m)BDNF and thereby promote a depressive phenotype.
Collapse
Affiliation(s)
- René A Girard
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, 75708, USA
| | - Prashant S Chauhan
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, 75708, USA
| | - Torry A Tucker
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, 75708, USA
| | - Tim Allen
- Department of Pathology, The University of Mississippi Medical Center, Jackson, MS, USA
| | - Jaswinder Kaur
- Department of Pathology, The University of Mississippi Medical Center, Jackson, MS, USA
| | - Ann Jeffers
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, 75708, USA
| | - Kathleen Koenig
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, 75708, USA
| | - Galina Florova
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, 75708, USA
| | - Andrey A Komissarov
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, 75708, USA
| | - Tatiana A Gaidenko
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, 75708, USA
| | - Mignote B Chamiso
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, 75708, USA
| | - James Fowler
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, 75708, USA
| | - Danna E Morris
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, 75708, USA
| | - Krishna Sarva
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, 75708, USA
| | - Karan P Singh
- Department of Epidemiology and Biostatistics, School of Rural and Community Health, The University of Texas Health Science Center at Tyler, Tyler, TX, 75708, USA
| | - Steven Idell
- Department of Cellular and Molecular Biology, The University of Texas Health Science Center at Tyler, Tyler, TX, 75708, USA
| | - Richard D Idell
- Department of Behavioral Health, Child and Adolescent Psychiatry, The University of Texas Health Science Center at Tyler, 11937 US Highway 271, Tyler, TX, 75708, USA.
| |
Collapse
|
23
|
Shmakova AA, Rubina KA, Rysenkova KD, Gruzdeva AM, Ivashkina OI, Anokhin KV, Tkachuk VA, Semina EV. Urokinase receptor and tissue plasminogen activator as immediate-early genes in pentylenetetrazole-induced seizures in the mouse brain. Eur J Neurosci 2019; 51:1559-1572. [PMID: 31587391 DOI: 10.1111/ejn.14584] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 08/13/2019] [Accepted: 09/20/2019] [Indexed: 11/30/2022]
Abstract
Epileptogenesis progressively leads to the rearrangement of normal neuronal networks into more excitable ones and can be viewed as a form of neuroplasticity, the molecular mechanisms of which still remain obscure. Here, we studied pentylenetetrazole seizure-induced regulation of genes for plasminogen activator system in the mouse brain. We found that expression of tissue plasminogen activator (tPA) and urokinase receptor (uPAR) mRNA was strongly increased in the mouse cerebral cortex, hippocampus, striatum and amygdala as early as 3 hr after pentylenetetrazole seizures. Such early activity-induced expression of uPAR in the central nervous system has not been demonstrated before. uPAR mRNA accumulation was followed by elevation of uPAR protein, indicating a complete transcription-translation process. Both tPA gene induction and uPAR gene induction were independent of the protein synthesis, suggesting that they are regulated by neural activity as immediate-early genes. In contrast to tPA and uPAR genes, the expression of which returned to the basal level 6 hr following seizures, urokinase and plasminogen activator inhibitor-1 gene expression showed a delayed activation only at 3 days after seizures. In conclusion, our results suggest an important sensitivity of the brain plasminogen activator system to seizure activity which raises the question of its role in activity-dependent neural tissue remodeling in pathological and normal conditions.
Collapse
Affiliation(s)
- Anna A Shmakova
- Laboratory of Gene and Cell Technologies, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russian Federation
| | - Kseniya A Rubina
- Laboratory of Gene and Cell Technologies, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russian Federation
| | - Karina D Rysenkova
- Laboratory of Gene and Cell Technologies, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russian Federation
| | - Anna M Gruzdeva
- Institute for Advanced Brain Studies, Lomonosov Moscow State University, Moscow, Russian Federation.,Laboratory for Neurobiology of Memory, P.K. Anokhin Research Institute of Normal Physiology, Moscow, Russian Federation
| | - Olga I Ivashkina
- Institute for Advanced Brain Studies, Lomonosov Moscow State University, Moscow, Russian Federation.,Laboratory for Neurobiology of Memory, P.K. Anokhin Research Institute of Normal Physiology, Moscow, Russian Federation
| | - Konstantin V Anokhin
- Institute for Advanced Brain Studies, Lomonosov Moscow State University, Moscow, Russian Federation.,Laboratory for Neurobiology of Memory, P.K. Anokhin Research Institute of Normal Physiology, Moscow, Russian Federation
| | - Vsevolod A Tkachuk
- Laboratory of Gene and Cell Technologies, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russian Federation.,Institute of Experimental Cardiology, Federal State Budgetary Organization National Cardiology Research Center Ministry of Health of the Russian Federation, Moscow, Russian Federation
| | - Ekaterina V Semina
- Laboratory of Gene and Cell Technologies, Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russian Federation.,Institute of Experimental Cardiology, Federal State Budgetary Organization National Cardiology Research Center Ministry of Health of the Russian Federation, Moscow, Russian Federation
| |
Collapse
|
24
|
Ziliotto N, Bernardi F, Jakimovski D, Zivadinov R. Coagulation Pathways in Neurological Diseases: Multiple Sclerosis. Front Neurol 2019; 10:409. [PMID: 31068896 PMCID: PMC6491577 DOI: 10.3389/fneur.2019.00409] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 04/04/2019] [Indexed: 12/11/2022] Open
Abstract
Significant progress has been made in understanding the complex interactions between the coagulation system and inflammation and autoimmunity. Increased blood-brain-barrier (BBB) permeability, a key event in the pathophysiology of multiple sclerosis (MS), leads to the irruption into the central nervous system of blood components that include virtually all coagulation/hemostasis factors. Besides their cytotoxic deposition and role as a possible trigger of the coagulation cascade, hemostasis components cause inflammatory response and immune activation, sustaining neurodegenerative events in MS. Early studies showing the contribution of altered hemostasis in the complex pathophysiology of MS have been strengthened by recent studies using methodologies that permitted deeper investigation. Fibrin(ogen), an abundant protein in plasma, has been identified as a key contributor to neuroinflammation. Perturbed fibrinolysis was found to be a hallmark of progressive MS with abundant cortical fibrin(ogen) deposition. The immune-modulatory function of the intrinsic coagulation pathway still remains to be elucidated in MS. New molecular details in key hemostasis components participating in MS pathophysiology, and particularly involved in inflammatory and immune responses, could favor the development of novel therapeutic targets to ameliorate the evolution of MS. This review article introduces essential information on coagulation factors, inhibitors, and the fibrinolytic pathway, and highlights key aspects of their involvement in the immune system and inflammatory response. It discusses how hemostasis components are (dys)regulated in MS, and summarizes histopathological post-mortem human brain evidence, as well as cerebrospinal fluid, plasma, and serum studies of hemostasis and fibrinolytic pathways in MS. Studies of disease-modifying treatments as potential modifiers of coagulation factor levels, and case reports of autoimmunity affecting hemostasis in MS are also discussed.
Collapse
Affiliation(s)
- Nicole Ziliotto
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy.,Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, Buffalo Neuroimaging Analysis Center, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Francesco Bernardi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy
| | - Dejan Jakimovski
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, Buffalo Neuroimaging Analysis Center, University at Buffalo, State University of New York, Buffalo, NY, United States
| | - Robert Zivadinov
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, Buffalo Neuroimaging Analysis Center, University at Buffalo, State University of New York, Buffalo, NY, United States.,Clinical Translational Science Institute, Center for Biomedical Imaging, University at Buffalo, State University of New York, Buffalo, NY, United States
| |
Collapse
|
25
|
Cardoso AL, Fernandes A, Aguilar-Pimentel JA, de Angelis MH, Guedes JR, Brito MA, Ortolano S, Pani G, Athanasopoulou S, Gonos ES, Schosserer M, Grillari J, Peterson P, Tuna BG, Dogan S, Meyer A, van Os R, Trendelenburg AU. Towards frailty biomarkers: Candidates from genes and pathways regulated in aging and age-related diseases. Ageing Res Rev 2018; 47:214-277. [PMID: 30071357 DOI: 10.1016/j.arr.2018.07.004] [Citation(s) in RCA: 301] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2018] [Revised: 07/08/2018] [Accepted: 07/10/2018] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Use of the frailty index to measure an accumulation of deficits has been proven a valuable method for identifying elderly people at risk for increased vulnerability, disease, injury, and mortality. However, complementary molecular frailty biomarkers or ideally biomarker panels have not yet been identified. We conducted a systematic search to identify biomarker candidates for a frailty biomarker panel. METHODS Gene expression databases were searched (http://genomics.senescence.info/genes including GenAge, AnAge, LongevityMap, CellAge, DrugAge, Digital Aging Atlas) to identify genes regulated in aging, longevity, and age-related diseases with a focus on secreted factors or molecules detectable in body fluids as potential frailty biomarkers. Factors broadly expressed, related to several "hallmark of aging" pathways as well as used or predicted as biomarkers in other disease settings, particularly age-related pathologies, were identified. This set of biomarkers was further expanded according to the expertise and experience of the authors. In the next step, biomarkers were assigned to six "hallmark of aging" pathways, namely (1) inflammation, (2) mitochondria and apoptosis, (3) calcium homeostasis, (4) fibrosis, (5) NMJ (neuromuscular junction) and neurons, (6) cytoskeleton and hormones, or (7) other principles and an extensive literature search was performed for each candidate to explore their potential and priority as frailty biomarkers. RESULTS A total of 44 markers were evaluated in the seven categories listed above, and 19 were awarded a high priority score, 22 identified as medium priority and three were low priority. In each category high and medium priority markers were identified. CONCLUSION Biomarker panels for frailty would be of high value and better than single markers. Based on our search we would propose a core panel of frailty biomarkers consisting of (1) CXCL10 (C-X-C motif chemokine ligand 10), IL-6 (interleukin 6), CX3CL1 (C-X3-C motif chemokine ligand 1), (2) GDF15 (growth differentiation factor 15), FNDC5 (fibronectin type III domain containing 5), vimentin (VIM), (3) regucalcin (RGN/SMP30), calreticulin, (4) PLAU (plasminogen activator, urokinase), AGT (angiotensinogen), (5) BDNF (brain derived neurotrophic factor), progranulin (PGRN), (6) α-klotho (KL), FGF23 (fibroblast growth factor 23), FGF21, leptin (LEP), (7) miRNA (micro Ribonucleic acid) panel (to be further defined), AHCY (adenosylhomocysteinase) and KRT18 (keratin 18). An expanded panel would also include (1) pentraxin (PTX3), sVCAM/ICAM (soluble vascular cell adhesion molecule 1/Intercellular adhesion molecule 1), defensin α, (2) APP (amyloid beta precursor protein), LDH (lactate dehydrogenase), (3) S100B (S100 calcium binding protein B), (4) TGFβ (transforming growth factor beta), PAI-1 (plasminogen activator inhibitor 1), TGM2 (transglutaminase 2), (5) sRAGE (soluble receptor for advanced glycosylation end products), HMGB1 (high mobility group box 1), C3/C1Q (complement factor 3/1Q), ST2 (Interleukin 1 receptor like 1), agrin (AGRN), (6) IGF-1 (insulin-like growth factor 1), resistin (RETN), adiponectin (ADIPOQ), ghrelin (GHRL), growth hormone (GH), (7) microparticle panel (to be further defined), GpnmB (glycoprotein nonmetastatic melanoma protein B) and lactoferrin (LTF). We believe that these predicted panels need to be experimentally explored in animal models and frail cohorts in order to ascertain their diagnostic, prognostic and therapeutic potential.
Collapse
|
26
|
Takeyama M, Takeuchi F, Gosho M, Sugita K, Zako M, Iwaki M, Kamei M. Effect of oral tranexamic acid on macular edema associated with retinal vein occlusion or diabetes. Clin Ophthalmol 2018; 12:35-41. [PMID: 29339919 PMCID: PMC5745154 DOI: 10.2147/opth.s149935] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Purpose Tranexamic acid (TXA) is a widely used antifibrinolytic agent that can also cause a decrease in vascular permeability. We hypothesized that TXA could improve macular edema (ME) that is caused by an increase in retinal vascular permeability. The aim of this study is to evaluate the efficacy of oral TXA for ME associated with retinal vein occlusion (RVO) or diabetic ME (DME). Patients and methods Oral TXA (1,500 mg daily for 2 weeks) was administered to patients with persistent ME secondary to RVO (7 eyes) and DME (7 eyes). After 2 weeks (ie, the final day of administration) and 6 weeks (ie, 4 weeks after the final administration), best-corrected visual acuity and central macular thickness (CMT) were measured and compared with baseline. Analyses were performed for RVO and DME cases. No other treatment was performed during the study period. Results In RVO cases, significant improvement in CMT was found between baseline (467.7±121.4 μm) and 2-week measurements after treatment (428.7±110.5 μm, p=0.024). No significant change was found in CMT between measurements taken at baseline and 6 weeks after treatment. In DME cases, no significant change was found in CMT between measurements taken at baseline and 2 or 6 weeks after treatment. In all analyses of best-corrected visual acuity, no significant change was observed. Conclusion The results support the hypothesis that plasmin plays a role in the development of ME associated with RVO, and oral TXA administration may be useful as an adjuvant treatment when combined with other agents such as anti-vascular endothelial growth factor.
Collapse
Affiliation(s)
| | - Fumio Takeuchi
- Department of Biochemistry, Aichi Medical University, Nagakute
| | - Masahiko Gosho
- Department of Clinical Trial and Clinical Epidemiology, Faculty of Medicine, University of Tsukuba, Tsukuba
| | - Keijiro Sugita
- Department of Ophthalmology, Aichi Medical University, Nagakute
| | | | - Masayoshi Iwaki
- Department of Ophthalmology, Yokkaichi, Digestive Disease Center, Komono, Japan
| | - Motohiro Kamei
- Department of Ophthalmology, Aichi Medical University, Nagakute
| |
Collapse
|
27
|
Fekih-Mrissa N, Mansour M, Sayeh A, Bedoui I, Mrad M, Riahi A, Mrissa R, Nsiri B. The Plasminogen Activator Inhibitor 1 4G/5G Polymorphism and the Risk of Alzheimer's Disease. Am J Alzheimers Dis Other Demen 2017; 32:342-346. [PMID: 28466654 PMCID: PMC10852582 DOI: 10.1177/1533317517705223] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/03/2024]
Abstract
OBJECTIVE The aim of this study was to determine whether plasminogen activator inhibitor 1 (PAI-1) is associated with the risk of Alzheimer's disease (AD) in Tunisian patients. DESIGN AND METHODS We analyzed the genotype and allele frequency distribution of the PAI-1 polymorphism in 60 Tunisian patients with AD and 120 healthy controls. RESULTS The results show a significantly increased risk of AD in carriers of the 4G/4G and 4G/5G genotypes versus the wild-type 5G/5G genotype (4G/4G: 28.33% in patients vs 10.0% in controls; P < 10-3; OR = 8.78; 4G/5G: 55.0% in patients vs 38.33% in controls; OR = 4.45; P < 10-3). The 4G allele was also more frequently found in patients compared with controls; P < 10-3; OR = 3.07. For all participants and by gender, homozygotic carriers (4G/4G) were at an increased risk of AD over heterozygotes and women were at an increased risk over their male genotype counterparts. The odds ratio for AD among 4G/4G carriers for any group was approximately twice that of heterozygotes in the same group. Women homozygotes ranked highest for AD risk (OR = 20.8) and, in fact, women heterozygotes (OR = 9.03) ranked higher for risk than male homozygotes (OR = 6.12). CONCLUSION These preliminary exploratory results should be confirmed in a larger study.
Collapse
Affiliation(s)
- Najiba Fekih-Mrissa
- Laboratory of Molecular Biology, Department of Hematology, Military Hospital of Tunisia, Tunis, Tunisia
| | - Malek Mansour
- Department of Psychiatry, Military Hospital of Tunisia, Tunis, Tunisia
| | - Aicha Sayeh
- Laboratory of Molecular Biology, Department of Hematology, Military Hospital of Tunisia, Tunis, Tunisia
| | - Ines Bedoui
- Department of Psychiatry, Military Hospital of Tunisia, Tunis, Tunisia
| | - Meriem Mrad
- Laboratory of Molecular Biology, Department of Hematology, Military Hospital of Tunisia, Tunis, Tunisia
| | - Anis Riahi
- Department of Psychiatry, Military Hospital of Tunisia, Tunis, Tunisia
| | - Ridha Mrissa
- Department of Psychiatry, Military Hospital of Tunisia, Tunis, Tunisia
| | - Brahim Nsiri
- Laboratory of Molecular Biology, Department of Hematology, Military Hospital of Tunisia, Tunis, Tunisia
| |
Collapse
|
28
|
Merino P, Diaz A, Yepes M. Urokinase-type plasminogen activator (uPA) and its receptor (uPAR) promote neurorepair in the ischemic brain. RECEPTORS & CLINICAL INVESTIGATION 2017; 4:e1552. [PMID: 28804736 PMCID: PMC5553903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Despite the fact that ischemic stroke has been considered a leading cause of mortality in the world, recent advances in our understanding of the pathophysiological mechanisms underlying the ischemic injury and the treatment of acute ischemic stroke patients have led to a sharp decrease in the number of stroke deaths. However, this decrease in stroke mortality has also led to an increase in the number of patients that survive the acute ischemic injury with different degrees of disability. Unfortunately, to this date we do not have an effective therapeutic strategy to promote neurological recovery in these growing population of stroke survivors. Cerebral ischemia not only causes the destruction of a large number of axons and synapses but also activates endogenous mechanisms that promote the recovery of those neurons that survive its harmful effects. Here we review experimental evidence indicating that one of these mechanisms of repair is the binding of the serine proteinase urokinase-type plasminogen activator (uPA) to its receptor (uPAR) in the growth cones of injured axons. Indeed, the binding of uPA to uPAR in the periphery of growth cones of injured axons induces the recruitment of β1-integrin to the plasma membrane, β1-integrin-mediated activation of the small Rho GTPase Rac1, and Rac1-induced axonal regeneration. Furthermore, we found that this process is modulated by the low density lipoprotein receptor-related protein (LRP1). The data reviewed here indicate that the uPA-uPAR-LRP1 system is a potential target for the development of therapeutic strategies to promote neurological recovery in acute ischemic stroke patients.
Collapse
Affiliation(s)
- Paola Merino
- Department of Neurology and Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, Georgia 30322, USA,Division of Neurosciences, Yerkes National Primate Research Center, Atlanta, Georgia 30329, USA
| | - Ariel Diaz
- Department of Neurology and Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, Georgia 30322, USA,Division of Neurosciences, Yerkes National Primate Research Center, Atlanta, Georgia 30329, USA
| | - Manuel Yepes
- Department of Neurology and Center for Neurodegenerative Disease, Emory University School of Medicine, Atlanta, Georgia 30322, USA,Division of Neurosciences, Yerkes National Primate Research Center, Atlanta, Georgia 30329, USA,Department of Neurology, Veterans Affairs Medical Center, Atlanta, Georgia 30033, USA
| |
Collapse
|
29
|
Integrative transcriptome network analysis of iPSC-derived neurons from schizophrenia and schizoaffective disorder patients with 22q11.2 deletion. BMC SYSTEMS BIOLOGY 2016. [DOI: 10.1186/s12918-016-0366-0 order by 8029-- awyx] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
30
|
Lin M, Pedrosa E, Hrabovsky A, Chen J, Puliafito BR, Gilbert SR, Zheng D, Lachman HM. Integrative transcriptome network analysis of iPSC-derived neurons from schizophrenia and schizoaffective disorder patients with 22q11.2 deletion. BMC SYSTEMS BIOLOGY 2016. [DOI: 10.1186/s12918-016-0366-0 and 1880=1880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
31
|
Integrative transcriptome network analysis of iPSC-derived neurons from schizophrenia and schizoaffective disorder patients with 22q11.2 deletion. BMC SYSTEMS BIOLOGY 2016. [DOI: 10.1186/s12918-016-0366-0 order by 8029-- #] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
32
|
Lin M, Pedrosa E, Hrabovsky A, Chen J, Puliafito BR, Gilbert SR, Zheng D, Lachman HM. Integrative transcriptome network analysis of iPSC-derived neurons from schizophrenia and schizoaffective disorder patients with 22q11.2 deletion. BMC SYSTEMS BIOLOGY 2016. [DOI: 10.1186/s12918-016-0366-0 order by 8029-- -] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
33
|
Lin M, Pedrosa E, Hrabovsky A, Chen J, Puliafito BR, Gilbert SR, Zheng D, Lachman HM. Integrative transcriptome network analysis of iPSC-derived neurons from schizophrenia and schizoaffective disorder patients with 22q11.2 deletion. BMC SYSTEMS BIOLOGY 2016. [DOI: 10.1186/s12918-016-0366-0 order by 1-- -] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
34
|
Lin M, Pedrosa E, Hrabovsky A, Chen J, Puliafito BR, Gilbert SR, Zheng D, Lachman HM. Integrative transcriptome network analysis of iPSC-derived neurons from schizophrenia and schizoaffective disorder patients with 22q11.2 deletion. BMC SYSTEMS BIOLOGY 2016. [DOI: 10.1186/s12918-016-0366-0 order by 1-- gadu] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
35
|
Lin M, Pedrosa E, Hrabovsky A, Chen J, Puliafito BR, Gilbert SR, Zheng D, Lachman HM. Integrative transcriptome network analysis of iPSC-derived neurons from schizophrenia and schizoaffective disorder patients with 22q11.2 deletion. BMC SYSTEMS BIOLOGY 2016. [DOI: 10.1186/s12918-016-0366-0 order by 1-- #] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
36
|
Lin M, Pedrosa E, Hrabovsky A, Chen J, Puliafito BR, Gilbert SR, Zheng D, Lachman HM. Integrative transcriptome network analysis of iPSC-derived neurons from schizophrenia and schizoaffective disorder patients with 22q11.2 deletion. BMC SYSTEMS BIOLOGY 2016; 10:105. [PMID: 27846841 PMCID: PMC5111260 DOI: 10.1186/s12918-016-0366-0] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 11/06/2016] [Indexed: 11/10/2022]
Abstract
BACKGROUND Individuals with 22q11.2 Deletion Syndrome (22q11.2 DS) are a specific high-risk group for developing schizophrenia (SZ), schizoaffective disorder (SAD) and autism spectrum disorders (ASD). Several genes in the deleted region have been implicated in the development of SZ, e.g., PRODH and DGCR8. However, the mechanistic connection between these genes and the neuropsychiatric phenotype remains unclear. To elucidate the molecular consequences of 22q11.2 deletion in early neural development, we carried out RNA-seq analysis to investigate gene expression in early differentiating human neurons derived from induced pluripotent stem cells (iPSCs) of 22q11.2 DS SZ and SAD patients. METHODS Eight cases (ten iPSC-neuron samples in total including duplicate clones) and seven controls (nine in total including duplicate clones) were subjected to RNA sequencing. Using a systems level analysis, differentially expressed genes/gene-modules and pathway of interests were identified. Lastly, we related our findings from in vitro neuronal cultures to brain development by mapping differentially expressed genes to BrainSpan transcriptomes. RESULTS We observed ~2-fold reduction in expression of almost all genes in the 22q11.2 region in SZ (37 genes reached p-value < 0.05, 36 of which reached a false discovery rate < 0.05). Outside of the deleted region, 745 genes showed significant differences in expression between SZ and control neurons (p < 0.05). Function enrichment and network analysis of the differentially expressed genes uncovered converging evidence on abnormal expression in key functional pathways, such as apoptosis, cell cycle and survival, and MAPK signaling in the SZ and SAD samples. By leveraging transcriptome profiles of normal human brain tissues across human development into adulthood, we showed that the differentially expressed genes converge on a sub-network mediated by CDC45 and the cell cycle, which would be disrupted by the 22q11.2 deletion during embryonic brain development, and another sub-network modulated by PRODH, which could contribute to disruption of brain function during adolescence. CONCLUSIONS This study has provided evidence for disruption of potential molecular events in SZ patient with 22q11.2 deletion and related our findings from in vitro neuronal cultures to functional perturbations that can occur during brain development in SZ.
Collapse
Affiliation(s)
- Mingyan Lin
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY USA
| | - Erika Pedrosa
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY USA
| | - Anastasia Hrabovsky
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY USA
| | - Jian Chen
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY USA
| | - Benjamin R. Puliafito
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY USA
| | - Stephanie R. Gilbert
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY USA
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY USA
- Department of Neurology, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY USA
- Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY USA
| | - Herbert M. Lachman
- Department of Genetics, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY USA
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY USA
- Department of Neuroscience, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY USA
- Department of Medicine, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY USA
| |
Collapse
|
37
|
Tavares E, Antequera D, López-González I, Ferrer I, Miñano FJ, Carro E. Potential Role of Aminoprocalcitonin in the Pathogenesis of Alzheimer Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:2723-35. [PMID: 27497681 DOI: 10.1016/j.ajpath.2016.06.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 04/25/2016] [Accepted: 06/09/2016] [Indexed: 11/15/2022]
Abstract
Increasing evidence suggests that inflammatory responses cause brain atrophy and play a prominent and early role in the progression of Alzheimer disease. Recent findings show that the neuroendocrine peptide aminoprocalcitonin (NPCT) plays a critical role in the development of systemic inflammatory response; however, the presence, possible function, regulation, and mechanisms by which NPCT may be involved in Alzheimer disease neuropathology remain unknown. We explored the expression of NPCT and its interaction with amyloid-β (Aβ), and proinflammatory and neurogenic effects. By using brain samples of Alzheimer disease patients and APP/PS1 transgenic mice, we evaluated the potential role of NPCT on Aβ-related pathology. We found that NPCT is expressed in hippocampal and cortical neurons and Aβ-induced up-regulation of NPCT expression. Peripherally administered antibodies against NPCT decreased microglial activation, decreased circulating levels of proinflammatory cytokines, and prevented Aβ-induced neurotoxicity in experimental models of Alzheimer disease. Remarkably, anti-NPTC therapy resulted in a significant improvement in the behavioral status of APP/PS1 mice. Our results indicate a central role of NPCT in Alzheimer disease pathogenesis and suggest NPCT as a potential biomarker and therapeutic target.
Collapse
Affiliation(s)
- Eva Tavares
- Clinical and Experimental Pharmacology Research Unit, Valme University Hospital, Seville, Spain.
| | - Desiree Antequera
- Group of Neurodegenerative Diseases, Instituto de Investigacion Hospital 12 de Octubre (i+12), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Neurodegenerative Diseases Biomedical Research Center (CIBERNED), Madrid, Spain
| | - Irene López-González
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Neurodegenerative Diseases Biomedical Research Center (CIBERNED), Madrid, Spain; Institut de Neuropatologia, IDIBELL-Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Hospitalet de Llobregat, Spain; Universitat de Barcelona, Hospitalet de Llobregat, Barcelona, Spain
| | - Isidro Ferrer
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Neurodegenerative Diseases Biomedical Research Center (CIBERNED), Madrid, Spain; Institut de Neuropatologia, IDIBELL-Hospital Universitari de Bellvitge, Hospitalet de Llobregat, Hospitalet de Llobregat, Spain; Universitat de Barcelona, Hospitalet de Llobregat, Barcelona, Spain
| | - Francisco J Miñano
- Clinical and Experimental Pharmacology Research Unit, Valme University Hospital, Seville, Spain; Department of Pharmacology, Pediatrics and Radiology, Faculty of Medicine, University of Seville, Seville, Spain
| | - Eva Carro
- Group of Neurodegenerative Diseases, Instituto de Investigacion Hospital 12 de Octubre (i+12), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas, Neurodegenerative Diseases Biomedical Research Center (CIBERNED), Madrid, Spain.
| |
Collapse
|
38
|
Bolkvadze T, Puhakka N, Pitkänen A. Epileptogenesis after traumatic brain injury in Plaur-deficient mice. Epilepsy Behav 2016; 60:187-196. [PMID: 27208924 DOI: 10.1016/j.yebeh.2016.04.038] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 04/17/2016] [Accepted: 04/18/2016] [Indexed: 11/16/2022]
Abstract
Binding of the extracellular matrix proteinase urokinase-type plasminogen activator (uPA) to its receptor, uPAR, regulates tissue remodeling during development and after injury in different organs, including the brain. Accordingly, mutations in the Plaur gene, which encodes uPAR, have been linked to language deficits, autism, and epilepsy, both in mouse and human. Whether uPAR deficiency modulates epileptogenesis and comorbidogenesis after brain injury, however, is unknown. To address this question, we induced traumatic brain injury (TBI) by controlled cortical impact (CCI) in 10 wild-type (Wt-CCI) and 16 Plaur-deficient (uPAR-CCI) mice. Sham-operated mice served as controls (10 Wt-sham, 10 uPAR-sham). During the 4-month follow-up, the mice were neurophenotyped by assessing the somatomotor performance with the composite neuroscore test, emotional learning and memory with fear conditioning to tone and context, and epileptogenesis with videoelectroencephalography monitoring and the pentylenetetrazol (PTZ) seizure susceptibility test. At the end of the testing, the mice were perfused for histology to analyze cortical and hippocampal neurodegeneration and mossy fiber sprouting. Fourteen percent (1/7) of the mice in the Wt-CCI and 0% in the uPAR-CCI groups developed spontaneous seizures (p>0.05; chi-square). Both the Wt-CCI and uPAR-CCI groups showed increased seizure susceptibility in the PTZ test (p<0.05), impaired recovery of motor function (p<0.001), and neurodegeneration in the hippocampus and cortex (p<0.05) compared with the corresponding sham-operated controls. Motor recovery and emotional learning showed a genotype effect, being more impaired in uPAR-CCI than in Wt-CCI mice (p<0.05). The findings of the present study indicate that uPAR deficiency does not increase susceptibility to epileptogenesis after CCI injury but has an unfavorable comorbidity-modifying effect after TBI.
Collapse
Affiliation(s)
- Tamuna Bolkvadze
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FI-70211 Kuopio, Finland
| | - Noora Puhakka
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FI-70211 Kuopio, Finland
| | - Asla Pitkänen
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FI-70211 Kuopio, Finland.
| |
Collapse
|
39
|
Toman E, Harrisson S, Belli T. Biomarkers in traumatic brain injury: a review. J ROY ARMY MED CORPS 2015; 162:103-8. [PMID: 26527607 DOI: 10.1136/jramc-2015-000517] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Accepted: 10/11/2015] [Indexed: 12/23/2022]
Abstract
Biomarkers allow physiological processes to be monitored, in both health and injury. Multiple attempts have been made to use biomarkers in traumatic brain injury (TBI). Identification of such biomarkers could allow improved understanding of the pathological processes involved in TBI, diagnosis, prognostication and development of novel therapies. This review article aims to cover both established and emerging TBI biomarkers along with their benefits and limitations. It then discusses the potential value of TBI biomarkers to military, civilian and sporting populations and the future hopes for developing a role for biomarkers in head injury management.
Collapse
Affiliation(s)
- Emma Toman
- Major Trauma Service, Queen Elizabeth Hospital, Birmingham, UK
| | - S Harrisson
- Academic Department of Military Surgery and Trauma, Royal Centre for Defence Medicine, Birmingham, UK
| | - T Belli
- National Institute for Health Research Surgical Reconstruction and Microbiology Research Centre, Birmingham, UK University of Birmingham, Birmingham, UK
| |
Collapse
|
40
|
The plasminogen activation system in neuroinflammation. Biochim Biophys Acta Mol Basis Dis 2015; 1862:395-402. [PMID: 26493446 DOI: 10.1016/j.bbadis.2015.10.011] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 10/08/2015] [Accepted: 10/15/2015] [Indexed: 01/30/2023]
Abstract
The plasminogen activation (PA) system consists in a group of proteases and protease inhibitors regulating the activation of the zymogen plasminogen into its proteolytically active form, plasmin. Here, we give an update of the current knowledge about the role of the PA system on different aspects of neuroinflammation. These include modification in blood-brain barrier integrity, leukocyte diapedesis, removal of fibrin deposits in nervous tissues, microglial activation and neutrophil functions. Furthermore, we focus on the molecular mechanisms (some of them independent of plasmin generation and even of proteolysis) and target receptors responsible for these effects. The description of these mechanisms of action may help designing new therapeutic strategies targeting the expression, activity and molecular mediators of the PA system in neurological disorders involving neuroinflammatory processes. This article is part of a Special Issue entitled: Neuro Inflammation edited by Helga E. de Vries and Markus Schwaninger.
Collapse
|
41
|
Rantala J, Kemppainen S, Ndode-Ekane XE, Lahtinen L, Bolkvadze T, Gurevicius K, Tanila H, Pitkänen A. Urokinase-type plasminogen activator deficiency has little effect on seizure susceptibility and acquired epilepsy phenotype but reduces spontaneous exploration in mice. Epilepsy Behav 2015; 42:117-28. [PMID: 25506794 DOI: 10.1016/j.yebeh.2014.11.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 11/03/2014] [Accepted: 11/08/2014] [Indexed: 01/05/2023]
Abstract
Urokinase-type plasminogen activator (uPA), a serine protease, converts plasminogen to plasmin. Activation of plasmin leads to degradation of the extracellular matrix, which is critical for tissue recovery, angiogenesis, cell migration, and axonal and synaptic plasticity. We hypothesized that uPA deficiency would cause an abnormal neurophenotype and would lead to exacerbated epileptogenesis after brain injury. Wild-type (Wt) and uPA-/- mice underwent a battery of neurologic behavioral tests evaluating general reactivity, spontaneous exploratory activity, motor coordination, pain threshold, fear and anxiety, and memory. We placed particular emphasis on the effect of uPA deficiency on seizure susceptibility, including the response to convulsants (pentylenetetrazol, kainate, or pilocarpine) and kainate-induced epileptogenesis and epilepsy. The uPA-/- mice showed no motor or sensory impairment compared with the Wt mice. Hippocampus-dependent spatial memory also remained intact. The uPA-/- mice, however, exhibited reduced exploratory activity and an enhanced response to a tone stimulus (p<0.05 compared with the Wt mice). The urokinase-type plasminogen activator deficient mice showed no increase in spontaneous or evoked epileptiform electrographic activity. Rather, the response to pilocarpine administration was reduced compared with the Wt mice (p<0.05). Also, the epileptogenesis and the epilepsy phenotype after intrahippocampal kainate injection were similar to those in the Wt mice. Taken together, uPA deficiency led to diminished interest in the environmental surroundings and enhanced emotional reactivity to unexpected aversive stimuli. Urokinase-type plasminogen activator deficiency was not associated with enhanced seizure susceptibility or worsened poststatus epilepticus epilepsy phenotype.
Collapse
Affiliation(s)
- J Rantala
- Epilepsy Research Laboratory, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland
| | - S Kemppainen
- Neurobiology of Memory Laboratory, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland
| | - X E Ndode-Ekane
- Epilepsy Research Laboratory, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland
| | - L Lahtinen
- Epilepsy Research Laboratory, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland
| | - Tamuna Bolkvadze
- Epilepsy Research Laboratory, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland
| | - K Gurevicius
- Neurobiology of Memory Laboratory, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland
| | - H Tanila
- Neurobiology of Memory Laboratory, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland; Department of Neurology, Kuopio University Hospital, PO Box 1777, FIN-70211 Kuopio, Finland
| | - A Pitkänen
- Epilepsy Research Laboratory, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland; Department of Neurology, Kuopio University Hospital, PO Box 1777, FIN-70211 Kuopio, Finland.
| |
Collapse
|
42
|
Sasi SP, Rahimi L, Yan X, Silver M, Qin G, Losordo DW, Kishore R, Goukassian DA. Genetic deletion of TNFR2 augments inflammatory response and blunts satellite-cell-mediated recovery response in a hind limb ischemia model. FASEB J 2014; 29:1208-19. [PMID: 25466901 DOI: 10.1096/fj.14-249813] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 11/12/2014] [Indexed: 01/09/2023]
Abstract
We have previously shown that TNF-tumor necrosis factor receptor-2/p75 (TNFR2/p75) signaling plays a critical role in ischemia-induced neovascularization in skeletal muscle and heart tissues. To determine the role of TNF-TNFR2/p75 signaling in ischemia-induced inflammation and muscle regeneration, we subjected wild-type (WT) and TNFR2/p75 knockout (p75KO) mice to hind limb ischemia (HLI) surgery. Ischemia induced significant and long-lasting inflammation associated with considerable decrease in satellite-cell activation in p75KO muscle tissue up to 10 d after HLI surgery. To determine the possible additive negative roles of tissue aging and the absence of TNFR2/p75, either in the tissue or in the bone marrow (BM), we generated 2 chimeric BM transplantation (BMT) models where both young green fluorescent protein (GFP)-positive p75KO and WT BM-derived cells were transplanted into adult p75KO mice. HLI surgery was performed 1 mo after BMT, after confirming complete engraftment of the recipient BM with GFP donor cells. In adult p75KO with the WT-BMT, proliferative (Ki67(+)) cells were detected only by d 28 and were exclusively GFP(+), suggesting significantly delayed contribution of young WT-BM cell to adult p75KO ischemic tissue recovery. No GFP(+) young p75KO BM cells survived in adult p75KO tissue, signifying the additive negative roles of tissue aging combined with decreased/absent TNFR2/p75 signaling in postischemic recovery.
Collapse
Affiliation(s)
- Sharath P Sasi
- Cardiovascular Research Center, GeneSys Research Institute, Boston, Massachusetts, USA
| | - Layla Rahimi
- Cardiovascular Research Center, GeneSys Research Institute, Boston, Massachusetts, USA
| | - Xinhua Yan
- Cardiovascular Research Center, GeneSys Research Institute, Boston, Massachusetts, USA; Tufts University School of Medicine, Boston, Massachusetts, USA
| | - Marcy Silver
- Cardiovascular Research Center, GeneSys Research Institute, Boston, Massachusetts, USA
| | - Gangjian Qin
- Feinberg Cardiovascular Institute, Feinberg School of Medicine Northwestern University, Chicago, Illinois, USA; and
| | - Douglas W Losordo
- Feinberg Cardiovascular Institute, Feinberg School of Medicine Northwestern University, Chicago, Illinois, USA; and
| | - Raj Kishore
- Center for Translational Medicine, Temple University School of Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - David A Goukassian
- Cardiovascular Research Center, GeneSys Research Institute, Boston, Massachusetts, USA; Tufts University School of Medicine, Boston, Massachusetts, USA;
| |
Collapse
|
43
|
Soluble urokinase plasminogen activator receptor as a marker for use of antidepressants. PLoS One 2014; 9:e110555. [PMID: 25329298 PMCID: PMC4203805 DOI: 10.1371/journal.pone.0110555] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2013] [Accepted: 09/24/2014] [Indexed: 12/22/2022] Open
Abstract
Objectives Inflammation is involved in the pathogenesis of depression. A few cross-sectional population-based studies have found that depression is associated with increased levels of inflammatory markers. Soluble urokinase plasminogen activation receptor (suPAR) is known to be a stable marker for inflammation. We investigated the bidirectional association between suPAR levels and use of antidepressants. Methods suPAR level was measured in 9305 blood donors and analysed in relation to 5-years follow-up data on purchase of antidepressants and hospital diagnoses of depression from a nationwide Danish register. Results For men and women without prior use of antidepressants we found a significantly higher risk for incident use of antidepressants with higher suPAR values. For men, the risk of first use of antidepressants increased by 72% from the 1st to the 4th quartile (HR = 1.72, 95% CI: 1.11–2.69). For women, it increased by 108% from the 1st to the 4th quartile (HR = 2.08, 95% CI: 1.45–2.98). Previous use of antidepressants was also significantly associated with higher suPAR levels (p = 0.002). Conclusions High suPAR levels are associated with an increased risk for both previous and future use of antidepressants in healthy men and women. High suPAR are also associated with increased risk for a hospital diagnosis of depression.
Collapse
|
44
|
Pitkänen A, Ndode-Ekane XE, Łukasiuk K, Wilczynski GM, Dityatev A, Walker MC, Chabrol E, Dedeurwaerdere S, Vazquez N, Powell EM. Neural ECM and epilepsy. PROGRESS IN BRAIN RESEARCH 2014; 214:229-62. [DOI: 10.1016/b978-0-444-63486-3.00011-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
45
|
Skelly DT, Hennessy E, Dansereau MA, Cunningham C. A systematic analysis of the peripheral and CNS effects of systemic LPS, IL-1β, [corrected] TNF-α and IL-6 challenges in C57BL/6 mice. PLoS One 2013; 8:e69123. [PMID: 23840908 PMCID: PMC3698075 DOI: 10.1371/journal.pone.0069123] [Citation(s) in RCA: 184] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2013] [Accepted: 06/05/2013] [Indexed: 11/18/2022] Open
Abstract
It is increasingly clear that systemic inflammation has both adaptive and deleterious effects on the brain. However, detailed comparisons of brain effects of systemic challenges with different pro-inflammatory cytokines are lacking. In the present study, we challenged female C57BL/6 mice intraperitoneally with LPS (100 µg/kg), IL-1β (15 or 50 µg/kg), TNF-α (50 or 250 µg/kg) or IL-6 (50 or 125 µg/kg). We investigated effects on core body temperature, open field activity and plasma levels of inflammatory markers at 2 hours post injection. We also examined levels of hepatic, hypothalamic and hippocampal inflammatory cytokine transcripts. Hypothermia and locomotor hypoactivity were induced by LPS>IL-1β>TNF-α>>IL-6. Systemic LPS, IL-1β and TNF-α challenges induced robust and broadly similar systemic and central inflammation compared to IL-6, which showed limited effects, but did induce a hepatic acute phase response. Important exceptions included IFNβ, which could only be induced by LPS. Systemic IL-1β could not induce significant blood TNF-α, but induced CNS TNF-α mRNA, while systemic TNF-α could induce IL-1β in blood and brain. Differences between IL-1β and TNF-α-induced hippocampal profiles, specifically for IL-6 and CXCL1 prompted a temporal analysis of systemic and central responses at 1, 2, 4, 8 and 24 hours, which revealed that IL-1β and TNF-α both induced the chemokines CXCL1 and CCL2 but only IL-1β induced the pentraxin PTX3. Expression of COX-2, CXCL1 and CCL2, with nuclear localisation of the p65 subunit of NFκB, in the cerebrovasculature was demonstrated by immunohistochemistry. Furthermore, we used cFOS immunohistochemistry to show that LPS, IL-1β and to a lesser degree, TNF-α activated the central nucleus of the amygdala. Given the increasing attention in the clinical literautre on correlating specific systemic inflammatory mediators with neurological or neuropsychiatric conditions and complications, these data will provide a useful resource on the likely CNS inflammatory profiles resulting from systemic elevation of particular cytokines.
Collapse
Affiliation(s)
- Donal T. Skelly
- School of Biochemistry and Immunology and Trinity College Institute of Neuroscience, Trinity College, Dublin, Republic of Ireland
| | - Edel Hennessy
- School of Biochemistry and Immunology and Trinity College Institute of Neuroscience, Trinity College, Dublin, Republic of Ireland
| | - Marc-Andre Dansereau
- School of Biochemistry and Immunology and Trinity College Institute of Neuroscience, Trinity College, Dublin, Republic of Ireland
| | - Colm Cunningham
- School of Biochemistry and Immunology and Trinity College Institute of Neuroscience, Trinity College, Dublin, Republic of Ireland
| |
Collapse
|
46
|
Lamoureux L, Simon SLR, Plews M, Ruddat V, Brunet S, Graham C, Czub S, Knox JD. Urine proteins identified by two-dimensional differential gel electrophoresis facilitate the differential diagnoses of scrapie. PLoS One 2013; 8:e64044. [PMID: 23704971 PMCID: PMC3660319 DOI: 10.1371/journal.pone.0064044] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 04/11/2013] [Indexed: 01/23/2023] Open
Abstract
The difficulty in developing a diagnostic assay for Creutzfeldt - Jakob disease (CJD) and other transmissible spongiform encephalopathies (TSEs) stems in part from the fact that the infectious agent is an aberrantly folded form of an endogenous cellular protein. This precludes the use of the powerful gene based technologies currently applied to the direct detection of other infectious agents. To circumvent this problem our research objective has been to identify a set of proteins exhibiting characteristic differential abundance in response to TSE infection. The objective of the present study was to assess the disease specificity of differentially abundant urine proteins able to identify scrapie infected mice. Two-dimensional differential gel electrophoresis was used to analyze longitudinal collections of urine samples from both prion-infected mice and a transgenic mouse model of Alzheimer's disease. The introduction of fluorescent dyes, that allow multiple samples to be co-resolved and visualized on one two dimensional gel, have increased the accuracy of this methodology for the discovery of robust protein biomarkers for disease. The accuracy of a small panel of differentially abundant proteins to correctly classify an independent naïve sample set was determined. The results demonstrated that at the time of clinical presentation the differential abundance of urine proteins were capable of identifying the prion infected mice with 87% sensitivity and 93% specificity. The identity of the diagnostic differentially abundant proteins was investigated by mass spectrometry.
Collapse
Affiliation(s)
- Lise Lamoureux
- Prion Laboratory Services Section, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Sharon L. R. Simon
- Prion Laboratory Services Section, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Margot Plews
- Prion Laboratory Services Section, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Viola Ruddat
- GE Healthcare, San Francisco, California, United States of America
| | - Simone Brunet
- Prion Laboratory Services Section, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
| | - Catherine Graham
- National Centres for Animal Disease, Canadian Food Inspection Agency, Lethbridge, Alberta, Canada
| | - Stefanie Czub
- National Centres for Animal Disease, Canadian Food Inspection Agency, Lethbridge, Alberta, Canada
| | - J. David Knox
- Prion Laboratory Services Section, Public Health Agency of Canada, Winnipeg, Manitoba, Canada
- Department of Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
- * E-mail:
| |
Collapse
|
47
|
Ndode-Ekane XE, Pitkänen A. Urokinase-type plasminogen activator receptor modulates epileptogenesis in mouse model of temporal lobe epilepsy. Mol Neurobiol 2012; 47:914-37. [PMID: 23263886 DOI: 10.1007/s12035-012-8386-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 12/04/2012] [Indexed: 11/24/2022]
Abstract
Mutation in Plaur gene encoding urokinase-type plasminogen activator receptor (uPAR) results in epilepsy and autistic phenotype in mice. In humans, a single nucleotide polymorphism in PLAUR gene represents a risk for autism spectrum disorders. Importantly, the expression of uPAR is elevated in the brain after various epileptogenic insults like traumatic brain injury and status epilepticus. So far, the consequences of altered uPAR expression on brain networks are poorly known. We tested a hypothesis that uPAR regulates post-injury neuronal reorganization and consequent functional outcome, particularly epileptogenesis. Epileptogenesis was induced by intrahippocampal injection of kainate in adult male wild type (Wt) or uPAR knockout (uPAR-/-) mice, and animals were monitored with continuous (24/7) video-electroencephalogram for 30 days. The severity of status epilepticus did not differ between the genotypes. The spontaneous electrographic seizures which developed were, however, longer and their behavioral manifestations were more severe in uPAR-/- than Wt mice. The more severe epilepsy phenotype in uPAR-/- mice was associated with delayed but augmented inflammatory response and more severe neurodegeneration in the hippocampus. Also, the distribution of newly born cells in the dentate gyrus was more scattered, and the recovery of hippocampal blood vessel length from status epilepticus-induced damage was compromised in uPAR-/- mice as compared to Wt mice. Our data demonstrate that a deficiency in uPAR represents a mechanisms which results in the development of a more severe epilepsy phenotype and progressive brain pathology after status epilepticus. We suggest that uPAR represents a rational target for disease-modifying treatments after epileptogenic brain insults.
Collapse
Affiliation(s)
- Xavier Ekolle Ndode-Ekane
- Department of Neurobiology, A. I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, P. O. Box 1627, 70 211 Kuopio, Finland.
| | | |
Collapse
|
48
|
Gonzalez-Gronow M, Ray R, Wang F, Pizzo SV. The voltage-dependent anion channel (VDAC) binds tissue-type plasminogen activator and promotes activation of plasminogen on the cell surface. J Biol Chem 2012; 288:498-509. [PMID: 23161549 DOI: 10.1074/jbc.m112.412502] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The voltage-dependent anion channel (VDAC), a major pore-forming protein in the outer membrane of mitochondria, is also found in the plasma membrane of a large number of cells where in addition to its role in regulating cellular ATP release and volume control it is important for maintaining redox homeostasis. Cell surface VDAC is a receptor for plasminogen kringle 5, which promotes partial closure of the channel. In this study, we demonstrate that VDAC binds tissue-type plasminogen activator (t-PA) on human neuroblastoma SK-N-SH cells. Binding of t-PA to VDAC induced a decrease in K(m) and an increase in the V(max) for activation of its substrate, plasminogen (Pg). This resulted in accelerated Pg activation when VDAC, t-PA, and Pg were bound together. VDAC is also a substrate for plasmin; hence, it mimics fibrin activity. Binding of t-PA to VDAC occurs between a t-PA fibronectin type I finger domain located between amino acids Ile(5) and Asn(37) and a VDAC region including amino acids (20)GYGFG(24). These VDAC residues correspond to a GXXXG repeat motif commonly found in amyloid β peptides that is necessary for aggregation when these peptides form fibrillar deposits on the cell surface. Furthermore, we also show that Pg kringle 5 is a substrate for the NADH-dependent reductase activity of VDAC. This ternary complex is an efficient proteolytic complex that may facilitate removal of amyloid β peptide deposits from the normal brain and cell debris from injured brain tissue.
Collapse
Affiliation(s)
- Mario Gonzalez-Gronow
- Department of Pathology, Duke University Medical Center, Durham, North Carolina 27710, USA.
| | | | | | | |
Collapse
|
49
|
Beynon SB, Walker FR. Microglial activation in the injured and healthy brain: what are we really talking about? Practical and theoretical issues associated with the measurement of changes in microglial morphology. Neuroscience 2012; 225:162-71. [PMID: 22824429 DOI: 10.1016/j.neuroscience.2012.07.029] [Citation(s) in RCA: 133] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2012] [Revised: 07/12/2012] [Accepted: 07/12/2012] [Indexed: 12/14/2022]
Abstract
Recently it has become apparent that microglia play a role not only in responding to insults within the central nervous system but also in responding to changes in synaptic activity and potentially modulating synaptic function. This has led to an enormous expansion of interest in how microglia respond to both pathological and nonpathological challenges, with activities that are associated with unique morphological transformations. Examining changes in microglial morphology can provide direct insight into the cells' functional activities, as morphological status is recognized to be tightly coupled with function. Despite these advances in knowledge, many of the image-based morphometric procedures used to investigate changes in microglial morphology have not kept pace. This has created a situation in which morphometric approaches that have been extensively employed in the past can no longer provide accurate information on the complex transformations that microglia can undergo, particularly under non-pathological conditions. This review critically examines the strengths and weaknesses of existing morphometric analysis procedures. This review further examines efforts to improve the utility of existing approaches and discusses new developments, such as digital reconstruction, that yield more accurate and specific information on how microglia remodel themselves. Ultimately, an improved understanding of the strengths and limitations of existing, and emerging, morphometric approaches will greatly facilitate efforts to understand how microglia remodel themselves in response to the full spectrum of challenges that they are known to encounter.
Collapse
Affiliation(s)
- S B Beynon
- School of Biomedical Sciences and Pharmacy, University of Newcastle, Callaghan, NSW 2308, Australia
| | | |
Collapse
|
50
|
Jeon H, Kim JH, Kim JH, Lee WH, Lee MS, Suk K. Plasminogen activator inhibitor type 1 regulates microglial motility and phagocytic activity. J Neuroinflammation 2012; 9:149. [PMID: 22747686 PMCID: PMC3418576 DOI: 10.1186/1742-2094-9-149] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2012] [Accepted: 06/29/2012] [Indexed: 01/05/2023] Open
Abstract
Background Plasminogen activator inhibitor type 1 (PAI-1) is the primary inhibitor of urokinase type plasminogen activators (uPA) and tissue type plasminogen activators (tPA), which mediate fibrinolysis. PAI-1 is also involved in the innate immunity by regulating cell migration and phagocytosis. However, little is known about the role of PAI-1 in the central nervous system. Methods In this study, we identified PAI-1 in the culture medium of mouse mixed glial cells by liquid chromatography and tandem mass spectrometry. Secretion of PAI-1 from glial cultures was detected by ELISA and western blotting analysis. Cell migration was evaluated by in vitro scratch-wound healing assay or Boyden chamber assay and an in vivo stab wound injury model. Phagocytic activity was measured by uptake of zymosan particles. Results The levels of PAI-1 mRNA and protein expression were increased by lipopolysaccharide and interferon-γ stimulation in both microglia and astrocytes. PAI-1 promoted the migration of microglial cells in culture via the low-density lipoprotein receptor-related protein (LRP) 1/Janus kinase (JAK)/signal transducer and activator of transcription (STAT)1 axis. PAI-1 also increased microglial migration in vivo when injected into mouse brain. PAI-1-mediated microglial migration was independent of protease inhibition, because an R346A mutant of PAI-1 with impaired PA inhibitory activity also promoted microglial migration. Moreover, PAI-1 was able to modulate microglial phagocytic activity. PAI-1 inhibited microglial engulfment of zymosan particles in a vitronectin- and Toll-like receptor 2/6-dependent manner. Conclusion Our results indicate that glia-derived PAI-1 may regulate microglial migration and phagocytosis in an autocrine or paracrine manner. This may have important implications in the regulation of brain microglial activities in health and disease.
Collapse
Affiliation(s)
- Hyejin Jeon
- Department of Pharmacology, Brain Science & Engineering Institute, CMRI, Kyungpook National University School of Medicine, 101 Dong-In, Daegu, Joong-gu, 700-422, South Korea
| | | | | | | | | | | |
Collapse
|