1
|
Demeter F, Németh Z, Kajdácsi E, Bihari G, Dobó J, Gál P, Cervenak L. Detrimental interactions of hypoxia and complement MASP-1 in endothelial cells as a model for atherosclerosis-related diseases. Sci Rep 2024; 14:14882. [PMID: 38937560 PMCID: PMC11211410 DOI: 10.1038/s41598-024-64479-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 06/10/2024] [Indexed: 06/29/2024] Open
Abstract
Both hypoxia and the complement lectin pathway (CLP) are involved in atherosclerosis and atherosclerosis-related stroke and acute myocardial infarction (AMI). We have previously shown that mannose-binding lectin-associated serine protease-1 (MASP-1), the most abundant enzyme of CLP, induces an inflammatory phenotype of endothelial cells (ECs) by cleaving protease activated receptors (PARs). In the absence of data, we aimed to investigate whether hypoxia and MASP-1 interact at the level of ECs, to better understand their role in atherosclerosis-related diseases. Hypoxia attenuated the wound healing ability of ECs, increased ICAM-1 and decreased ICAM-2 expression and upregulated PAR2 gene expression. Hypoxia and MASP-1 increased GROα and IL-8 production, and endothelial permeability without potentiating each other's effects, whereas they cooperatively disrupted vascular network integrity, activated the Ca2+, CREB and NFκB signaling pathways, and upregulated the expression of E-selectin, a crucial adhesion molecule in neutrophil homing. VCAM-1 expression was not influenced either by hypoxia, or by MASP-1. In summary, hypoxia potentiates the effect of MASP-1 on ECs, at least partially by increasing PAR expression, resulting in interaction at several levels, which may altogether exacerbate stroke and AMI progression. Our findings suggest that MASP-1 is a potential drug target in the acute phase of atherosclerosis-related diseases.
Collapse
Affiliation(s)
- Flóra Demeter
- Research Laboratory, Department of Internal Medicine and Haematology, Semmelweis University, Szentkirályi U. 46, Budapest, 1088, Hungary
| | - Zsuzsanna Németh
- Research Laboratory, Department of Internal Medicine and Haematology, Semmelweis University, Szentkirályi U. 46, Budapest, 1088, Hungary
| | - Erika Kajdácsi
- Research Laboratory, Department of Internal Medicine and Haematology, Semmelweis University, Szentkirályi U. 46, Budapest, 1088, Hungary
- Research Group for Immunology and Hematology, Semmelweis University-HUN-REN-SU (Office for Supported Research Groups), Budapest, Hungary
| | - György Bihari
- Research Laboratory, Department of Internal Medicine and Haematology, Semmelweis University, Szentkirályi U. 46, Budapest, 1088, Hungary
| | - József Dobó
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Hungarian Research Network, Budapest, Hungary
| | - Péter Gál
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Hungarian Research Network, Budapest, Hungary
| | - László Cervenak
- Research Laboratory, Department of Internal Medicine and Haematology, Semmelweis University, Szentkirályi U. 46, Budapest, 1088, Hungary.
| |
Collapse
|
2
|
Connelly JA, Zhang X, Chen Y, Chao Y, Shi Y, Jacob TC, Wang QJ. Protein kinase D2 confers neuroprotection by promoting AKT and CREB activation in ischemic stroke. Neurobiol Dis 2023; 187:106305. [PMID: 37730136 PMCID: PMC10836334 DOI: 10.1016/j.nbd.2023.106305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 09/12/2023] [Accepted: 09/16/2023] [Indexed: 09/22/2023] Open
Abstract
Ischemic stroke, constituting 80-90% of all strokes, is a leading cause of death and long-term disability in adults. There is an urgent need to discover new targets and therapies for this devastating condition. Protein kinase D (PKD), as a key target of diacylglycerol involved in ischemic responses, has not been well studied in ischemic stroke, particularly PKD2. In this study, we found that PKD2 expression and activity were significantly upregulated in the ipsilateral side of the brain after transient focal cerebral ischemia, which coincides with the upregulation of PKD2 in primary neurons in response to in vitro ischemia, implying a potential role of PKD2 in neuronal survival in ischemic stroke. Using kinase-dead PKD2 knock-in (PKD2-KI) mice, we examined whether loss of PKD2 activity affected stroke outcomes in mice subjected to 1 h of transient middle cerebral artery occlusion (tMCAO) and 24 h of reperfusion. Our data demonstrated that PKD2-KI mice exhibited larger infarction volumes and worsened neurological scores, indicative of increased brain injury, as compared to the wild-type (WT) mice, confirming a neuroprotective role of PKD2 in ischemia/reperfusion (I/R) injury. Mouse primary neurons obtained from PKD2-KI mice also exhibited increased cell death as compared to the WT neurons when subjected to in vitro ischemia. We have further identified AKT and CREB as two main signaling nodes through which PKD2 regulates neuronal survival during I/R injury. In summary, PKD2 confers neuroprotection in ischemic stroke by promoting AKT and CREB activation and targeted activation of PKD2 may benefit neuronal survival in ischemic stroke.
Collapse
Affiliation(s)
- Jaclyn A Connelly
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, USA
| | - Xuejing Zhang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, USA
| | - Yuzhou Chen
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, USA
| | - Yapeng Chao
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, USA
| | - Yejie Shi
- Department of Neurology, University of Pittsburgh, Pittsburgh, USA
| | - Tija C Jacob
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, USA
| | - Q Jane Wang
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, USA.
| |
Collapse
|
3
|
Diet Supplementation with Polyphenol-Rich Salicornia ramosissima Extracts Protects against Tissue Damage in Experimental Models of Cerebral Ischemia. Nutrients 2022; 14:nu14235077. [PMID: 36501107 PMCID: PMC9735563 DOI: 10.3390/nu14235077] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/22/2022] [Accepted: 11/25/2022] [Indexed: 12/05/2022] Open
Abstract
Strokes are the second most common cause of death worldwide and a leading cause of disability. Regular consumption of polyphenols has been shown to reduce the risk of suffering a cardiovascular event. For this reason, we have investigated the protective effect of Salicornia ramosissima, a seasonal halophyte that synthetizes high amounts of bioactive compounds, including polyphenols, in response to environmental stress. Aqueous, hydroalcoholic, and ethanolic extracts were prepared to investigate if dietary supplementation prior to ischemic challenge can prevent subsequent damage using two animal models. First, we screened the protective effect against hypoxia-reoxygenation in Drosophila melanogaster and observed that both ethanolic and hydroalcoholic extracts protected flies from the deleterious effects of hypoxia. Second, we confirmed the protective effect of S. ramosissima ethanolic extract against brain ischemia using the transient middle cerebral artery occlusion mice model. Four weeks of oral supplementation with the ethanolic extract before artery occlusion reduced infarct volume and lowered the plasma levels of the DNA peroxidant product 8-hydroxydeoxyguanosine. Phytochemical profiling of S. ramosissima ethanolic extract revealed 50 compounds. Thus, it represents a valuable source of bioactive compounds that show promising disease-modifying activities and could be further developed as an effective food supplement for the prevention or treatment of neurovascular disorders.
Collapse
|
4
|
Pinto R, Magalhães A, Sousa M, Melo L, Lobo A, Barros P, Gomes JR. Bridging the Transient Intraluminal Stroke Preclinical Model to Clinical Practice: From Improved Surgical Procedures to a Workflow of Functional Tests. Front Neurol 2022; 13:846735. [PMID: 35359638 PMCID: PMC8963503 DOI: 10.3389/fneur.2022.846735] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/07/2022] [Indexed: 12/18/2022] Open
Abstract
Acute ischemic stroke (AIS) remains a leading cause of mortality, despite significant advances in therapy (endovascular thrombectomy). Failure in developing novel effective therapies is associated with unsuccessful translation from preclinical studies to clinical practice, associated to inconsistent and highly variable infarct areas and lack of relevant post-stroke functional evaluation in preclinical research. To outreach these limitations, we optimized the intraluminal transient middle cerebral occlusion, a widely used mouse stroke model, in two key parameters, selection of appropriate occlusion filaments and time of occlusion, which show a significant variation in the literature. We demonstrate that commercially available filaments with short coating length (1–2 mm), together with 45-min occlusion, results in a consistent affected brain region, similar to what is observed in most patients with AIS. Importantly, a dedicated post-stroke care protocol, based on clinical practice applied to patients who had stroke, resulted in lower mortality and improved mice welfare. Finally, a battery of tests covering relevant fine motor skills, sensory functions, and learning/memory behaviors revealed a significant effect of tMCAO brain infarction, which is parallel to patient symptomatology as measured by relevant clinical scales (NIH Stroke Scale, NIHSS and modified Rankin Scale, mRS). Thus, in order to enhance translation to clinical practice, future preclinical stroke research must consider the methodology described in this study, which includes improved reproducible surgical procedure, postoperative care, and the battery of functional tests. This will be a major step s closing the gap from bench to bedside, rendering the development of novel effective therapeutic approaches.
Collapse
Affiliation(s)
- Raquel Pinto
- Molecular Neurobiology Unit, IBMC-Instituto de Biologia Molecular e Celular, Porto, Portugal.,I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Ana Magalhães
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Addiction Biology Unit, IBMC-Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Mafalda Sousa
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Advanced Light Microscopy Unit, I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Lúcia Melo
- Molecular Neurobiology Unit, IBMC-Instituto de Biologia Molecular e Celular, Porto, Portugal.,I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Andrea Lobo
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Addiction Biology Unit, IBMC-Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Pedro Barros
- Neurology Department, Centro Hospitalar de Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal.,Stroke Unit, Centro Hospitalar de Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
| | - João R Gomes
- Molecular Neurobiology Unit, IBMC-Instituto de Biologia Molecular e Celular, Porto, Portugal.,I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| |
Collapse
|
5
|
Simats A, Ramiro L, Valls R, de Ramón H, García-Rodríguez P, Orset C, Artigas L, Sardon T, Rosell A, Montaner J. Ceruletide and Alpha-1 Antitrypsin as a Novel Combination Therapy for Ischemic Stroke. Neurotherapeutics 2022; 19:513-527. [PMID: 35226340 PMCID: PMC9226209 DOI: 10.1007/s13311-022-01203-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2022] [Indexed: 12/29/2022] Open
Abstract
Ischemic stroke is a primary cause of morbidity and mortality worldwide. Beyond the approved thrombolytic therapies, there is no effective treatment to mitigate its progression. Drug repositioning combinational therapies are becoming promising approaches to identify new uses of existing drugs to synergically target multiple disease-response mechanisms underlying complex pathologies. Here, we used a systems biology-based approach based on artificial intelligence and pattern recognition tools to generate in silico mathematical models mimicking the ischemic stroke pathology. Combinational treatments were acquired by screening these models with more than 5 million two-by-two combinations of drugs. A drug combination (CA) formed by ceruletide and alpha-1 antitrypsin showing a predicted value of neuroprotection of 92% was evaluated for their synergic neuroprotective effects in a mouse pre-clinical stroke model. The administration of both drugs in combination was safe and effective in reducing by 39.42% the infarct volume 24 h after cerebral ischemia. This neuroprotection was not observed when drugs were given individually. Importantly, potential incompatibilities of the drug combination with tPA thrombolysis were discarded in vitro and in vivo by using a mouse thromboembolic stroke model with t-PA-induced reperfusion, revealing an improvement in the forepaw strength 72 h after stroke in CA-treated mice. Finally, we identified the predicted mechanisms of action of ceruletide and alpha-1 antitrypsin and we demonstrated that CA modulates EGFR and ANGPT-1 levels in circulation within the acute phase after stroke. In conclusion, we have identified a promising combinational treatment with neuroprotective effects for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Alba Simats
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Hospital Universitari Vall d'Hebron, Pg. Vall d'Hebron 119-129, Barcelona, 08035, Spain
| | - Laura Ramiro
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Hospital Universitari Vall d'Hebron, Pg. Vall d'Hebron 119-129, Barcelona, 08035, Spain
| | | | - Helena de Ramón
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Hospital Universitari Vall d'Hebron, Pg. Vall d'Hebron 119-129, Barcelona, 08035, Spain
| | - Paula García-Rodríguez
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Hospital Universitari Vall d'Hebron, Pg. Vall d'Hebron 119-129, Barcelona, 08035, Spain
| | - Cyrille Orset
- Inserm UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, Université Caen-Normandie, GIP Cyceron, Caen, France
| | | | | | - Anna Rosell
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Hospital Universitari Vall d'Hebron, Pg. Vall d'Hebron 119-129, Barcelona, 08035, Spain
| | - Joan Montaner
- Neurovascular Research Laboratory, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Hospital Universitari Vall d'Hebron, Pg. Vall d'Hebron 119-129, Barcelona, 08035, Spain.
- Stroke Research Program, Institute of Biomedicine of Seville, IBiS/Hospital Universitario Virgen del Rocío/CSIC, University of Seville, Seville, Spain.
- Department of Neurology, Hospital Universitario Virgen Macarena, Seville, Spain.
| |
Collapse
|
6
|
Grayston A, Zhang Y, Garcia-Gabilondo M, Arrúe M, Martin A, Kopcansky P, Timko M, Kovac J, Strbak O, Castellote L, Belloli S, Moresco RM, Picchio M, Roig A, Rosell A. Endovascular administration of magnetized nanocarriers targeting brain delivery after stroke. J Cereb Blood Flow Metab 2022; 42:237-252. [PMID: 34229512 PMCID: PMC9122522 DOI: 10.1177/0271678x211028816] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The increasing use of mechanical thrombectomy in stroke management has opened the window to local intraarterial brain delivery of therapeutic agents. In this context, the use of nanomedicine could further improve the delivery of new treatments for specific brain targeting, tracking and guidance. In this study we take advantage of this new endovascular approach to deliver biocompatible poly(D-L-lactic-co-glycolic acid) (PLGA) nanocapsules functionalized with superparamagnetic iron oxide nanoparticles and Cy7.5 for magnetic targeting, magnetic resonance and fluorescent molecular imaging. A complete biodistribution study in naïve (n = 59) and ischemic (n = 51) mice receiving intravenous or intraarterial nanocapsules, with two different magnet devices and imaged from 30 min to 48 h, showed an extraordinary advantage of the intraarterial route for brain delivery with a specific improvement in cortical targeting when using a magnetic device in both control and ischemic conditions. Safety was evaluated in ischemic mice (n = 69) showing no signs of systemic toxicity nor increasing mortality, infarct lesions or hemorrhages. In conclusion, the challenging brain delivery of therapeutic nanomaterials could be efficiently and safely overcome with a controlled endovascular administration and magnetic targeting, which could be considered in the context of endovascular interventions for the delivery of multiple treatments for stroke.
Collapse
Affiliation(s)
- Alba Grayston
- Neurovascular Research Laboratory, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona (VHIR-UAB), Barcelona, Spain
| | - Yajie Zhang
- Nanoparticles and Nanocomposites Group, Institut de Ciència de Materials de Barcelona (ICMAB-CSIC), Bellaterra, Spain
| | - Miguel Garcia-Gabilondo
- Neurovascular Research Laboratory, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona (VHIR-UAB), Barcelona, Spain
| | - Mercedes Arrúe
- Neurovascular Research Laboratory, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona (VHIR-UAB), Barcelona, Spain
| | - Abraham Martin
- Achucarro Basque Center for Neuroscience, Laboratory of Neuroimaging and Biomarkers of Inflammation, Leioa, Spain.,Ikerbasque Basque Foundation for Science, Bilbao, Spain
| | - Peter Kopcansky
- Department of Magnetism, Institute of Experimental Physics, SAS, Kosice, Slovakia
| | - Milan Timko
- Department of Magnetism, Institute of Experimental Physics, SAS, Kosice, Slovakia
| | - Jozef Kovac
- Department of Magnetism, Institute of Experimental Physics, SAS, Kosice, Slovakia
| | - Oliver Strbak
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, Martin, Slovakia
| | - Laura Castellote
- Department of Clinical Biochemistry, Clinical Laboratories, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Sara Belloli
- Nuclear Medicine Department, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Institute of Molecular Bioimaging and Physiology (IBFM), CNR, Segrate, Italy
| | - Rosa M Moresco
- Nuclear Medicine Department, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Department of Medicine and Surgery, University of Milano - Bicocca, Monza (MB), Italy
| | - Maria Picchio
- Nuclear Medicine Department, IRCCS San Raffaele Scientific Institute, Milan, Italy.,School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
| | - Anna Roig
- Nanoparticles and Nanocomposites Group, Institut de Ciència de Materials de Barcelona (ICMAB-CSIC), Bellaterra, Spain
| | - Anna Rosell
- Neurovascular Research Laboratory, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona (VHIR-UAB), Barcelona, Spain
| |
Collapse
|
7
|
Sorby-Adams AJ, Learoyd AE, Bath PM, Burrows F, Farr TD, Leonard AV, Schiessl I, Allan SM, Turner RJ, Trueman RC. Glyceryl trinitrate for the treatment of ischaemic stroke: Determining efficacy in rodent and ovine species for enhanced clinical translation. J Cereb Blood Flow Metab 2021; 41:3248-3259. [PMID: 34039053 PMCID: PMC8669202 DOI: 10.1177/0271678x211018901] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Hypertension is a leading risk factor for death and dependency after ischaemic stroke. However, administering anti-hypertensive medications post-stroke remains contentious with concerns regarding deleterious effects on cerebral blood flow and infarct expansion. This study sought to determine the effect of glyceryl trinitrate (GTN) treatment in both lissencephalic and gyrencephalic pre-clinical stroke models. Merino sheep underwent middle cerebral artery occlusion (MCAO) followed by GTN or control patch administration (0.2 mg/h). Monitoring of numerous physiologically relevant measures over 24 h showed that GTN administration was associated with decreased intracranial pressure, infarct volume, cerebral oedema and midline shift compared to vehicle treatment (p < 0.05). No significant changes in blood pressure or cerebral perfusion pressure were observed. Using optical imaging spectroscopy and laser speckle imaging, the effect of varying doses of GTN (0.69-50 µg/h) on cerebral blood flow and tissue oxygenation was examined in mice. No consistent effect was found. Additional mice undergoing MCAO followed by GTN administration (doses varying from 0-60 µg/h) also showed no improvement in infarct volume or neurological score within 24 h post-stroke. GTN administration significantly improved numerous stroke-related physiological outcomes in sheep but was ineffective in mice. This suggests that, whilst GTN administration could potentially benefit patients, further research into mechanisms of action are required.
Collapse
Affiliation(s)
- Annabel J Sorby-Adams
- Adelaide Medical School and Adelaide Centre for Neuroscience Research, The University of Adelaide, Adelaide, SA, Australia
| | - Annastazia E Learoyd
- School of Life Sciences, University of Nottingham Medical School, Nottingham, UK
| | - Philip M Bath
- Stroke Trials Unit, Division of Clinical Neuroscience, University of Nottingham, Nottingham, UK
| | - Fiona Burrows
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Tracy D Farr
- School of Life Sciences, University of Nottingham Medical School, Nottingham, UK
| | - Anna V Leonard
- Adelaide Medical School and Adelaide Centre for Neuroscience Research, The University of Adelaide, Adelaide, SA, Australia
| | - Ingo Schiessl
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.,Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester, UK
| | - Stuart M Allan
- Division of Neuroscience and Experimental Psychology, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.,Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester, UK
| | - Renée J Turner
- Adelaide Medical School and Adelaide Centre for Neuroscience Research, The University of Adelaide, Adelaide, SA, Australia
| | - Rebecca C Trueman
- School of Life Sciences, University of Nottingham Medical School, Nottingham, UK
| |
Collapse
|
8
|
Pericyte hypoxia-inducible factor-1 (HIF-1) drives blood-brain barrier disruption and impacts acute ischemic stroke outcome. Angiogenesis 2021; 24:823-842. [PMID: 34046769 PMCID: PMC8487886 DOI: 10.1007/s10456-021-09796-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/04/2021] [Indexed: 12/19/2022]
Abstract
Pericytes play essential roles in blood-brain barrier integrity and their dysfunction is implicated in neurological disorders such as stroke although the underlying mechanisms remain unknown. Hypoxia-inducible factor-1 (HIF-1), a master regulator of injury responses, has divergent roles in different cells especially during stress scenarios. On one hand HIF-1 is neuroprotective but on the other it induces vascular permeability. Since pericytes are critical for barrier stability, we asked if pericyte HIF-1 signaling impacts barrier integrity and injury severity in a mouse model of ischemic stroke. We show that pericyte HIF-1 loss of function (LoF) diminishes ischemic damage and barrier permeability at 3 days reperfusion. HIF-1 deficiency preserved barrier integrity by reducing pericyte death thereby maintaining vessel coverage and junctional protein organization, and suppressing vascular remodeling. Importantly, considerable improvements in sensorimotor function were observed in HIF-1 LoF mice indicating that better vascular functionality post stroke improves outcome. Thus, boosting vascular integrity by inhibiting pericytic HIF-1 activation and/or increasing pericyte survival may be a lucrative option to accelerate recovery after severe brain injury.
Collapse
|
9
|
Increased Mortality and Vascular Phenotype in a Knock-In Mouse Model of Retinal Vasculopathy With Cerebral Leukoencephalopathy and Systemic Manifestations. Stroke 2020; 51:300-307. [DOI: 10.1161/strokeaha.119.025176] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Background and Purpose—
Retinal vasculopathy with cerebral leukoencephalopathy and systemic manifestations (RVCL-S) is an autosomal dominant small vessel disease caused by C-terminal frameshift mutations in the
TREX1
gene that encodes the major mammalian 3′ to 5′ DNA exonuclease. RVCL-S is characterized by vasculopathy, especially in densely vascularized organs, progressive retinopathy, cerebral microvascular disease, white matter lesions, and migraine, but the underlying mechanisms are unknown.
Methods—
Homozygous transgenic RVCL-S knock-in mice expressing a truncated Trex1 (three prime repair exonuclease 1) protein (similar to what is seen in patients) and wild-type littermates, of various age groups, were subjected to (1) a survival analysis, (2) in vivo postocclusive reactive hyperemia and ex vivo Mulvany myograph studies to characterize the microvascular and macrovascular reactivity, and (3) experimental stroke after transient middle cerebral artery occlusion with neurological deficit assessment.
Results—
The mutant mice show increased mortality starting at midlife (
P
=0.03 with hazard ratio, 3.14 [95% CI, 1.05–9.39]). The mutants also show a vascular phenotype as evidenced by attenuated postocclusive reactive hyperemia responses (across all age groups; F[1, 65]=5.7,
P
=0.02) and lower acetylcholine-induced relaxations in aortae (in 20- to 24-month-old mice; RVCL-S knock-in: E
max
: 37±8% versus WT: E
max
: 65±6%,
P
=0.01). A vascular phenotype is also suggested by the increased infarct volume seen in 12- to 14-month-old mutant mice at 24 hours after infarct onset (RVCL-S knock-in: 75.4±2.7 mm
3
versus WT: 52.9±5.6 mm
3
,
P
=0.01).
Conclusions—
Homozygous RVCL-S knock-in mice show increased mortality, signs of abnormal vascular function, and increased sensitivity to experimental stroke and can be instrumental to investigate the pathology seen in patients with RVCL-S.
Collapse
|
10
|
Schartz ND, Sommer AL, Colin SA, Mendez LB, Brewster AL. Early treatment with C1 esterase inhibitor improves weight but not memory deficits in a rat model of status epilepticus. Physiol Behav 2019; 212:112705. [PMID: 31628931 PMCID: PMC6879103 DOI: 10.1016/j.physbeh.2019.112705] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 09/20/2019] [Accepted: 10/07/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Status epilepticus (SE) is a prolonged and continuous seizure that lasts for at least 5 min. An episode of SE in a healthy system can lead to the development of spontaneous seizures and cognitive deficits which may be accompanied by hippocampal injury and microgliosis. Although the direct mechanisms underlying the SE-induced pathophysiology remain unknown, a candidate mechanism is hyperactivation of the classical complement pathway (C1q-C3 signaling). We recently reported that SE triggered an increase in C1q-C3 signaling in the hippocampus that closely paralleled cognitive decline. Thus, we hypothesized that blocking activation of the classical complement pathway immediately after SE may prevent the development of SE-induced hippocampal-dependent learning and memory deficits. METHODS Because C1 esterase inhibitor (C1-INH) negatively regulates activation of the classical complement pathway, we used this drug to test our hypothesis. Two groups of male rats were subjected to 1 hr of SE with pilocarpine (280-300 mg/kg, i.p.), and treated with either C1-INH (SE+C1-INH, 20 U/kg, s.c.) or vehicle (SE+veh) at 4, 24, and 48 h after SE. Control rats were treated with saline. Body weight was recorded for up to 23 days after SE. At two weeks post SE, recognition and spatial memory were determined using Novel Object Recognition (NOR) and Barnes maze (BM), respectively, as well as locomotion and anxiety-like behaviors using Open Field (OF). Histological and biochemical methods were used to measure hippocampal injury including cell death, microgliosis, and inflammation. RESULTS One day after SE, both SE groups had a significant loss of body weight compared to controls (p<0.05). By day 14, the weight of SE+C1-INH rats was significantly higher than SE+veh rats (p<0.05), and was not different from controls (p>0.05). At 14 days post-SE, SE+C1-INH rats displayed higher mobility (distance travelled and average speed, p<0.05) and had reduced anxiety-like behaviors (outer duration, p<0.05) than control or SE+veh rats. In NOR, control rats spent significantly more time exploring the novel object vs. the familiar (p<0.05), while rats in both SE groups spent similar amount of time exploring both objects. During days 1-4 of BM training, the escape latency of the control group significantly decreased over time (p<0.05), whereas that of the SE groups did not improve (p>0.05). Compared to vehicle-treated SE rats, SE+C1-INH rats had increased levels of C3 and microglia in the hippocampus, but lower levels of caspase-3 and synaptic markers. CONCLUSIONS These findings suggest that acute treatment with C1-INH after SE may have some protective, albeit limited, effects on the physiological recovery of rats' weight and some anxiolytic effects, but does not attenuate SE-induced deficits in hippocampal-dependent learning and memory. Reduced levels of caspase-3 suggest that treatment with C1-INH may protect against cell death, perhaps by regulating inflammatory pathways and promoting phagocytic/clearance pathways.
Collapse
Affiliation(s)
- Nicole D Schartz
- Department of Psychological Sciences, Purdue University, West Lafayette, IN 47907, USA.
| | - Alexandra L Sommer
- Department of Psychological Sciences, Purdue University, West Lafayette, IN 47907, USA.
| | - Samantha A Colin
- Department of Psychological Sciences, Purdue University, West Lafayette, IN 47907, USA.
| | - Loyda B Mendez
- School of Science & Technology, Ana G. Méndez University, Carolina, PR 00984, USA.
| | - Amy L Brewster
- Department of Psychological Sciences, Purdue University, West Lafayette, IN 47907, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN 47907, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
11
|
Lee JD, Coulthard LG, Woodruff TM. Complement dysregulation in the central nervous system during development and disease. Semin Immunol 2019; 45:101340. [PMID: 31708347 DOI: 10.1016/j.smim.2019.101340] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 10/15/2019] [Accepted: 10/24/2019] [Indexed: 12/14/2022]
Abstract
The complement cascade is an important arm of the immune system that plays a key role in protecting the central nervous system (CNS) from infection. Recently, it has also become clear that complement proteins have fundamental roles in the developing and aging CNS that are distinct from their roles in immunity. During neurodevelopment, complement signalling is involved in diverse processes including neural tube closure, neural progenitor proliferation and differentiation, neuronal migration, and synaptic pruning. In acute neurotrauma and ischamic brain injury, complement drives inflammation and neuronal death, but also neuroprotection and regeneration. In diseases of the aging CNS including dementias and motor neuron disease, chronic complement activation is associated with glial activation, and synapse and neuron loss. Proper regulation of complement is thus essential to allow for an appropriately developed CNS and prevention of excessive damage following neurotrauma or during neurodegeneration. This review provides a comprehensive overview of the evidence for functional roles of complement in brain formation, and its dysregulation during acute and chronic disease. We also provide working models for how complement can lead to neurodevelopmental disorders such as schizophrenia and autism, and either protect, or propagate neurodegenerative diseases including Alzheimer's disease and amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- John D Lee
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Liam G Coulthard
- Royal Brisbane and Women's Hospital, Herston, Australia; School of Clinical Medicine, Faculty of Medicine, The University of Queensland, Brisbane, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, Faculty of Medicine, The University of Queensland, Brisbane, Australia.
| |
Collapse
|
12
|
C1 Esterase Inhibitor Reduces BBB Leakage and Apoptosis in the Hypoxic Developing Mouse Brain. Neuromolecular Med 2019; 22:31-44. [DOI: 10.1007/s12017-019-08560-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 07/31/2019] [Indexed: 12/18/2022]
|
13
|
Kobylarek D, Iwanowski P, Lewandowska Z, Limphaibool N, Szafranek S, Labrzycka A, Kozubski W. Advances in the Potential Biomarkers of Epilepsy. Front Neurol 2019; 10:685. [PMID: 31312171 PMCID: PMC6614180 DOI: 10.3389/fneur.2019.00685] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 06/12/2019] [Indexed: 12/11/2022] Open
Abstract
Epilepsy is a group of chronic neurological disorders characterized by recurrent, spontaneous, and unpredictable seizures. It is one of the most common neurological disorders, affecting tens of millions of people worldwide. Comprehensive studies on epilepsy in recent decades have revealed the complexity of epileptogenesis, in which immunological processes, epigenetic modifications, and structural changes in neuronal tissues have been identified as playing a crucial role. This review discusses the recent advances in the biomarkers of epilepsy. We evaluate the possible molecular background underlying the clinical changes observed in recent studies, focusing on therapeutic investigations, and the evidence of their safety and efficacy in the human population. This article reviews the pathophysiology of epilepsy, including recent reports on the effects of oxidative stress and hypoxia, and focuses on specific biomarkers and their clinical implications, along with further perspectives in epilepsy research.
Collapse
Affiliation(s)
- Dominik Kobylarek
- Department of Neurology, Poznan University of Medical Sciences, Poznan, Poland
| | | | | | | | | | | | | |
Collapse
|
14
|
Ma Y, Liu Y, Zhang Z, Yang GY. Significance of Complement System in Ischemic Stroke: A Comprehensive Review. Aging Dis 2019; 10:429-462. [PMID: 31011487 PMCID: PMC6457046 DOI: 10.14336/ad.2019.0119] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Accepted: 01/19/2019] [Indexed: 12/14/2022] Open
Abstract
The complement system is an essential part of innate immunity, typically conferring protection via eliminating pathogens and accumulating debris. However, the defensive function of the complement system can exacerbate immune, inflammatory, and degenerative responses in various pathological conditions. Cumulative evidence indicates that the complement system plays a critical role in the pathogenesis of ischemic brain injury, as the depletion of certain complement components or the inhibition of complement activation could reduce ischemic brain injury. Although multiple candidates modulating or inhibiting complement activation show massive potential for the treatment of ischemic stroke, the clinical availability of complement inhibitors remains limited. The complement system is also involved in neural plasticity and neurogenesis during cerebral ischemia. Thus, unexpected side effects could be induced if the systemic complement system is inhibited. In this review, we highlighted the recent concepts and discoveries of the roles of different kinds of complement components, such as C3a, C5a, and their receptors, in both normal brain physiology and the pathophysiology of brain ischemia. In addition, we comprehensively reviewed the current development of complement-targeted therapy for ischemic stroke and discussed the challenges of bringing these therapies into the clinic. The design of future experiments was also discussed to better characterize the role of complement in both tissue injury and recovery after cerebral ischemia. More studies are needed to elucidate the molecular and cellular mechanisms of how complement components exert their functions in different stages of ischemic stroke to optimize the intervention of targeting the complement system.
Collapse
Affiliation(s)
- Yuanyuan Ma
- 1Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,2Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yanqun Liu
- 3Department of Neurology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Zhijun Zhang
- 2Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Guo-Yuan Yang
- 1Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,2Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
15
|
Farfara D, Feierman E, Richards A, Revenko AS, MacLeod RA, Norris EH, Strickland S. Knockdown of circulating C1 inhibitor induces neurovascular impairment, glial cell activation, neuroinflammation, and behavioral deficits. Glia 2019; 67:1359-1373. [PMID: 30882931 DOI: 10.1002/glia.23611] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 02/11/2019] [Accepted: 02/19/2019] [Indexed: 12/20/2022]
Abstract
The cross-talk between blood proteins, immune cells, and brain function involves complex mechanisms. Plasma protein C1 inhibitor (C1INH) is an inhibitor of vascular inflammation that is induced by activation of the kallikrein-kinin system (KKS) and the complement system. Knockout of C1INH was previously correlated with peripheral vascular permeability via the bradykinin pathway, yet there was no evidence of its correlation with blood-brain barrier (BBB) integrity and brain function. In order to understand the effect of plasma C1INH on brain pathology via the vascular system, we knocked down circulating C1INH in wild-type (WT) mice using an antisense oligonucleotide (ASO), without affecting C1INH expression in peripheral immune cells or the brain, and examined brain pathology. Long-term elimination of endogenous C1INH in the plasma induced the activation of the KKS and peritoneal macrophages but did not activate the complement system. Bradykinin pathway proteins were elevated in the periphery and the brain, resulting in hypotension. BBB permeability, extravasation of plasma proteins into the brain parenchyma, activation of glial cells, and elevation of pro-inflammatory response mediators were detected. Furthermore, infiltrating innate immune cells were observed entering the brain through the lateral ventricle walls and the neurovascular unit. Mice showed normal locomotion function, yet cognition was impaired and depressive-like behavior was evident. In conclusion, our results highlight the important role of regulated plasma C1INH as it acts as a gatekeeper to the brain via the neurovascular system. Thus, manipulation of C1INH in neurovascular disorders might be therapeutically beneficial.
Collapse
Affiliation(s)
- Dorit Farfara
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, New York
| | - Emily Feierman
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, New York
| | - Allison Richards
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, New York
| | - Alexey S Revenko
- Department of Antisense Drug Discovery, IONIS Pharmaceuticals Inc., Carlsbad, California
| | - Robert A MacLeod
- Department of Antisense Drug Discovery, IONIS Pharmaceuticals Inc., Carlsbad, California
| | - Erin H Norris
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, New York
| | - Sidney Strickland
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, New York
| |
Collapse
|
16
|
Valerieva A, Caccia S, Cicardi M. Recombinant human C1 esterase inhibitor (Conestat alfa) for prophylaxis to prevent attacks in adult and adolescent patients with hereditary angioedema. Expert Rev Clin Immunol 2018; 14:707-718. [DOI: 10.1080/1744666x.2018.1503055] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Anna Valerieva
- Medical University of Sofia, Clinical Center of Allergology, University Hospital “Alexandrovska”, Sofia, Bulgaria
| | - Sonia Caccia
- Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, University of Milan, Milan, Italy
| | - Marco Cicardi
- Department of Biomedical and Clinical Sciences, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, University of Milan, Milan, Italy
| |
Collapse
|
17
|
Lin Z, Lin H, Li W, Huang Y, Dai H. Complement Component C3 Promotes Cerebral Ischemia/Reperfusion Injury Mediated by TLR2/NFκB Activation in Diabetic Mice. Neurochem Res 2018; 43:1599-1607. [PMID: 29948726 DOI: 10.1007/s11064-018-2574-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Revised: 05/16/2018] [Accepted: 06/11/2018] [Indexed: 12/20/2022]
Abstract
Complement component C3 (C3), a key factor in the complement system, is heavily involved in various inflammation-associated diseases. However, it remains obscure for its role in the pathogenesis of cerebral ischemia/reperfusion (I/R) injury in diabetes. A transient middle cerebral artery occlusion (tMCAO) model was used for cerebral I/R injury in streptozotocin-induced diabetic mice. Cerebral infarct volume and neurological function were measured at different times of reperfusion. Complement C3 was measured by ELISA and western blotting. It was observed that complement C3 expression was increased in cerebral I/R injury of diabetic mice, whereas complement C3 deficiency abrogated the activation and injury. Furthermore, activating complement C3 promotes TLR2/NFκB activation after I/R injury in diabetic mice, which is inhibited by of the silencing of TLR2. Taken together, our data demonstrate that complement C3 promotes cerebral I/R injury via the TLR2/NFκB pathway in diabetic mice, and regulating the complement C3/TLR2/NFκB pathway may be a novel target for therapeutic intervention in diabetic stroke.
Collapse
Affiliation(s)
- Zheng Lin
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Haoran Lin
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Wenlu Li
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Yuwen Huang
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Haibin Dai
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
18
|
Nokkari A, Abou-El-Hassan H, Mechref Y, Mondello S, Kindy MS, Jaffa AA, Kobeissy F. Implication of the Kallikrein-Kinin system in neurological disorders: Quest for potential biomarkers and mechanisms. Prog Neurobiol 2018; 165-167:26-50. [PMID: 29355711 PMCID: PMC6026079 DOI: 10.1016/j.pneurobio.2018.01.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Accepted: 01/15/2018] [Indexed: 01/06/2023]
Abstract
Neurological disorders represent major health concerns in terms of comorbidity and mortality worldwide. Despite a tremendous increase in our understanding of the pathophysiological processes involved in disease progression and prevention, the accumulated knowledge so far resulted in relatively moderate translational benefits in terms of therapeutic interventions and enhanced clinical outcomes. Aiming at specific neural molecular pathways, different strategies have been geared to target the development and progression of such disorders. The kallikrein-kinin system (KKS) is among the most delineated candidate systems due to its ubiquitous roles mediating several of the pathophysiological features of these neurological disorders as well as being implicated in regulating various brain functions. Several experimental KKS models revealed that the inhibition or stimulation of the two receptors of the KKS system (B1R and B2R) can exhibit neuroprotective and/or adverse pathological outcomes. This updated review provides background details of the KKS components and their functions in different neurological disorders including temporal lobe epilepsy, traumatic brain injury, stroke, spinal cord injury, Alzheimer's disease, multiple sclerosis and glioma. Finally, this work will highlight the putative roles of the KKS components as potential neurotherapeutic targets and provide future perspectives on the possibility of translating these findings into potential clinical biomarkers in neurological disease.
Collapse
Affiliation(s)
- Amaly Nokkari
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon
| | - Hadi Abou-El-Hassan
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, USA
| | - Stefania Mondello
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Mark S Kindy
- Department of Pharmaceutical Science, College of Pharmacy, University of South Florida, Tampa, FL, USA; James A. Haley VA Medical Center, Tampa, FL, USA
| | - Ayad A Jaffa
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon; Department of Medicine, Medical University of South, Charleston, SC, USA.
| | - Firas Kobeissy
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Lebanon; Center for Neuroproteomics & Biomarkers Research, Department of Psychiatry, McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
19
|
Combination Therapy with Low-Dose IVIG and a C1-esterase Inhibitor Ameliorates Brain Damage and Functional Deficits in Experimental Ischemic Stroke. Neuromolecular Med 2018; 20:63-72. [PMID: 29299869 DOI: 10.1007/s12017-017-8474-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 12/29/2017] [Indexed: 01/08/2023]
Abstract
Acute ischemic stroke causes a high rate of deaths and permanent neurological deficits in survivors. Current interventional treatment, in the form of enzymatic thrombolysis, benefits only a small percentage of patients. Brain ischemia triggers mobilization of innate immunity, specifically the complement system and Toll-like receptors (TLRs), ultimately leading to an exaggerated inflammatory response. Here we demonstrate that intravenous immunoglobulin (IVIG), a scavenger of potentially harmful complement fragments, and C1-esterase inhibitor (C1-INH), an inhibitor of complement activation, exert a beneficial effect on the outcome of experimental brain ischemia (I) and reperfusion (R) injury induced by transient occlusion of middle cerebral artery in mice. Both IVIG and C1-INH significantly and in a dose-responsive manner reduced brain infarction size, neurological deficit and mortality when administered to male mice 30 min before ischemia or up to 6 h after the onset of reperfusion. When combined, suboptimal doses of IVIG and C1-INH potentiated each other's neuroprotective therapeutic effects. Complement C3 and TLR2 signals were colocalized and significantly greater in brain cells adjacent to infracted brain lesions when compared to the corresponding regions of the contralateral hemisphere and to control (sham) mice. Treatment with IVIG and C1-INH effectively reduced deposition of C3b and downregulated excessive TLR2 and p-JNK1 expression at the site of I/R injury. Taken together, these results provide a rationale for potential use of IVIG and C1-INH, alone or in combination with ischemic stroke and other neurological conditions that involve inappropriately activated components of the innate immune system.
Collapse
|
20
|
Status epilepticus triggers long-lasting activation of complement C1q-C3 signaling in the hippocampus that correlates with seizure frequency in experimental epilepsy. Neurobiol Dis 2017; 109:163-173. [PMID: 29074125 DOI: 10.1016/j.nbd.2017.10.012] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/30/2017] [Accepted: 10/19/2017] [Indexed: 01/01/2023] Open
Abstract
Status epilepticus (SE) triggers a myriad of neurological alterations that include unprovoked seizures, temporal lobe epilepsy (TLE), and cognitive deficits. Although SE-induced loss of hippocampal dendritic structures and synaptic remodeling are often associated with this pathophysiology, the underlying mechanisms remain elusive. Recent evidence points to the classical complement pathway as a potential mechanism. Signaling through the complement protein C1q to C3, which is cleaved into smaller biologically active fragments including C3b and iC3b, contributes to the elimination of synaptic structures in the normal developing brain and in models of neurodegenerative disorders. We recently found increased protein levels of C1q and iC3b fragments in human drug-resistant epilepsy. Thus, to identify a potential role for C1q-C3 in SE-induced epilepsy, we performed a temporal analysis of C1q protein levels and C3 cleavage in the hippocampus along with their association to seizures and hippocampal-dependent cognitive functions in a rat model of SE and acquired TLE. We found significant increases in the levels of C1q, C3, and iC3b in the hippocampus at 2-, 3- and 5-weeks after SE relative to controls (p<0.05). In the SE group, greater iC3b levels were significantly correlated with higher seizure frequency (p<0.05). Together, these data support that hyperactivation of the classical complement pathway after SE parallels the progression of epilepsy. Future studies will determine whether C1q-C3 signaling contributes to epileptogenic synaptic remodeling in the hippocampus.
Collapse
|
21
|
Emmrich JV, Neher JJ, Boehm-Sturm P, Endres M, Dirnagl U, Harms C. Stage 1 Registered Report: Effect of deficient phagocytosis on neuronal survival and neurological outcome after temporary middle cerebral artery occlusion (tMCAo). F1000Res 2017; 6:1827. [PMID: 29152223 PMCID: PMC5664978 DOI: 10.12688/f1000research.12537.3] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/14/2018] [Indexed: 01/29/2023] Open
Abstract
Stroke is a major cause of death and disability worldwide. In addition to neuronal death resulting directly from energy depletion due to lack of blood supply, inflammation and microglial activation following ischemic brain injury has been increasingly recognized to be a key contributor to the pathophysiology of cerebrovascular disease. However, our understanding of the cross talk between the ischemic brain and the immune system is limited. Recently, we demonstrated that following focal ischemia, death of mature viable neurons can be executed through phagocytosis by microglial cells or recruited macrophages, i.e. through phagoptosis. It was shown that inhibition of phagocytic signaling pathways following endothelin-1 induced focal cerebral ischemia leads to increased neuronal survival and neurological recovery. This suggests that inhibition of specific phagocytic pathways may prevent neuronal death during cerebral ischemia. To further explore this potential therapeutic target, we propose to assess the role of phagocytosis in an established model of temporary (45min) middle cerebral artery occlusion (tMCAo), and to evaluate neuronal survival and neurological recovery in mice with deficient phagocytosis. The primary outcome of this study will be forelimb function assessed with the staircase test. Secondary outcomes constitute Rotarod performance, stroke volume (quantified on MR imaging or brain sections, respectively), diffusion tensor imaging (DTI) connectome mapping, and histological analyses to measure neuronal and microglial densities, and phagocytic activity. Male mice aged 10-12 weeks will be used for experiments.
Collapse
Affiliation(s)
- Julius V Emmrich
- Department of Experimental Neurology and Center for Stroke Research, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jonas J Neher
- Department of Cellular Neurology, Hertie Institute for Clinical Brain Research, University of Tübingen, Tübingen, Germany.,German Center for Neurodegenerative Diseases (DZNE), Tübingen, Germany
| | - Philipp Boehm-Sturm
- Department of Experimental Neurology and Center for Stroke Research, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Matthias Endres
- Department of Experimental Neurology and Center for Stroke Research, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
| | - Ulrich Dirnagl
- Department of Experimental Neurology and Center for Stroke Research, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany.,QUEST - Center for Transforming Biomedical Research, Berlin Institute of Health (BIH), Berlin, Germany
| | - Christoph Harms
- Department of Experimental Neurology and Center for Stroke Research, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,QUEST - Center for Transforming Biomedical Research, Berlin Institute of Health (BIH), Berlin, Germany
| |
Collapse
|
22
|
Impact of aging immune system on neurodegeneration and potential immunotherapies. Prog Neurobiol 2017; 157:2-28. [PMID: 28782588 DOI: 10.1016/j.pneurobio.2017.07.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 07/25/2017] [Accepted: 07/28/2017] [Indexed: 12/19/2022]
Abstract
The interaction between the nervous and immune systems during aging is an area of avid interest, but many aspects remain unclear. This is due, not only to the complexity of the aging process, but also to a mutual dependency and reciprocal causation of alterations and diseases between both the nervous and immune systems. Aging of the brain drives whole body systemic aging, including aging-related changes of the immune system. In turn, the immune system aging, particularly immunosenescence and T cell aging initiated by thymic involution that are sources of chronic inflammation in the elderly (termed inflammaging), potentially induces brain aging and memory loss in a reciprocal manner. Therefore, immunotherapeutics including modulation of inflammation, vaccination, cellular immune therapies and "protective autoimmunity" provide promising approaches to rejuvenate neuroinflammatory disorders and repair brain injury. In this review, we summarize recent discoveries linking the aging immune system with the development of neurodegeneration. Additionally, we discuss potential rejuvenation strategies, focusing aimed at targeting the aging immune system in an effort to prevent acute brain injury and chronic neurodegeneration during aging.
Collapse
|
23
|
Sammali E, Alia C, Vegliante G, Colombo V, Giordano N, Pischiutta F, Boncoraglio GB, Barilani M, Lazzari L, Caleo M, De Simoni MG, Gaipa G, Citerio G, Zanier ER. Intravenous infusion of human bone marrow mesenchymal stromal cells promotes functional recovery and neuroplasticity after ischemic stroke in mice. Sci Rep 2017; 7:6962. [PMID: 28761170 PMCID: PMC5537246 DOI: 10.1038/s41598-017-07274-w] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 06/27/2017] [Indexed: 12/13/2022] Open
Abstract
Transplantation of human bone marrow mesenchymal stromal cells (hBM-MSC) promotes functional recovery after stroke in animal models, but the mechanisms underlying these effects remain incompletely understood. We tested the efficacy of Good Manufacturing Practices (GMP) compliant hBM-MSC, injected intravenously 3.5 hours after injury in mice subjected to transient middle cerebral artery occlusion (tMCAo). We addressed whether hBM-MSC are efficacious and if this efficacy is associated with cortical circuit reorganization using neuroanatomical analysis of GABAergic neurons (parvalbumin; PV-positive cells) and perineuronal nets (PNN), a specialized extracellular matrix structure which acts as an inhibitor of neural plasticity. tMCAo mice receiving hBM-MSC, showed early and lasting improvement of sensorimotor and cognitive functions compared to control tMCAo mice. Furthermore, 5 weeks post-tMCAo, hBM-MSC induced a significant rescue of ipsilateral cortical neurons; an increased proportion of PV-positive neurons in the perilesional cortex, suggesting GABAergic interneurons preservation; and a lower percentage of PV-positive cells surrounded by PNN, indicating an enhanced plastic potential of the perilesional cortex. These results show that hBM-MSC improve functional recovery and stimulate neuroprotection after stroke. Moreover, the downregulation of “plasticity brakes” such as PNN suggests that hBM-MSC treatment stimulates plasticity and formation of new connections in the perilesional cortex.
Collapse
Affiliation(s)
- Eliana Sammali
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa,19, 20156, Milano, Italy.,Department of Cerebrovascular Diseases, Fondazione IRCCS - Istituto Neurologico Carlo Besta, Milano, Italy
| | - Claudia Alia
- Neuroscience Institute, CNR, Pisa, Italy.,Scuola Normale Superiore, Pisa, Italy
| | - Gloria Vegliante
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa,19, 20156, Milano, Italy
| | - Valentina Colombo
- Laboratory for Cell and Gene Therapy "Stefano Verri", ASST-Monza, San Gerardo Hospital, Monza, Italy.,Tettamanti Research Center, Pediatric Department, University of Milano-Bicocca, Monza, Italy
| | - Nadia Giordano
- Neuroscience Institute, CNR, Pisa, Italy.,Scuola Normale Superiore, Pisa, Italy
| | - Francesca Pischiutta
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa,19, 20156, Milano, Italy
| | - Giorgio B Boncoraglio
- Department of Cerebrovascular Diseases, Fondazione IRCCS - Istituto Neurologico Carlo Besta, Milano, Italy
| | - Mario Barilani
- Cell Factory, Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milano, Italy
| | - Lorenza Lazzari
- Cell Factory, Unit of Cell Therapy and Cryobiology, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Via F. Sforza 35, 20122, Milano, Italy
| | | | - Maria-Grazia De Simoni
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa,19, 20156, Milano, Italy
| | - Giuseppe Gaipa
- Laboratory for Cell and Gene Therapy "Stefano Verri", ASST-Monza, San Gerardo Hospital, Monza, Italy.,Tettamanti Research Center, Pediatric Department, University of Milano-Bicocca, Monza, Italy
| | - Giuseppe Citerio
- School of Medicine and Surgery, University of Milano-Bicocca, Milano, Italy.,Neurointensive Care, ASST-Monza, San Gerardo Hospital, Monza, Italy
| | - Elisa R Zanier
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Via La Masa,19, 20156, Milano, Italy.
| |
Collapse
|
24
|
Interaction of ARC and Daxx: A Novel Endogenous Target to Preserve Motor Function and Cell Loss after Focal Brain Ischemia in Mice. J Neurosci 2017; 36:8132-48. [PMID: 27488634 DOI: 10.1523/jneurosci.4428-15.2016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 06/07/2016] [Indexed: 12/26/2022] Open
Abstract
UNLABELLED The aim of this study was to explore the signaling and neuroprotective effect of transactivator of transcription (TAT) protein transduction of the apoptosis repressor with CARD (ARC) in in vitro and in vivo models of cerebral ischemia in mice. In mice, transient focal cerebral ischemia reduced endogenous ARC protein in neurons in the ischemic striatum at early reperfusion time points, and in primary neuronal cultures, RNA interference resulted in greater neuronal susceptibility to oxygen glucose deprivation (OGD). TAT.ARC protein delivery led to a dose-dependent better survival after OGD. Infarct sizes 72 h after 60 min middle cerebral artery occlusion (MCAo) were on average 30 ± 8% (mean ± SD; p = 0.005; T2-weighted MRI) smaller in TAT.ARC-treated mice (1 μg intraventricularly during MCAo) compared with controls. TAT.ARC-treated mice showed better performance in the pole test compared with TAT.β-Gal-treated controls. Importantly, post-stroke treatment (3 h after MCAo) was still effective in affording reduced lesion volume by 20 ± 7% (mean ± SD; p < 0.05) and better functional outcome compared with controls. Delayed treatment in mice subjected to 30 min MCAo led to sustained neuroprotection and functional behavior benefits for at least 28 d. Functionally, TAT.ARC treatment inhibited DAXX-ASK1-JNK signaling in the ischemic brain. ARC interacts with DAXX in a CARD-dependent manner to block DAXX trafficking and ASK1-JNK activation. Our work identifies for the first time ARC-DAXX binding to block ASK1-JNK activation as an ARC-specific endogenous mechanism that interferes with neuronal cell death and ischemic brain injury. Delayed delivery of TAT.ARC may present a promising target for stroke therapy. SIGNIFICANCE STATEMENT Up to now, the only successful pharmacological target of human ischemic stroke is thrombolysis. Neuroprotective pharmacological strategies are needed to accompany therapies aiming to achieve reperfusion. We describe that apoptosis repressor with CARD (ARC) interacts and inhibits DAXX and proximal signals of cell death. In a murine stroke model mimicking human malignant infarction in the territory of the middle cerebral artery, TAT.ARC salvages brain tissue when given during occlusion or 3 h delayed with sustained functional benefits (28 d). This is a promising novel therapeutic approach because it appears to be effective in a model producing severe injury by interfering with an array of proximal signals and effectors of the ischemic cascade, upstream of JNK, caspases, and BIM and BAX activation.
Collapse
|
25
|
Trotman-Lucas M, Kelly ME, Janus J, Fern R, Gibson CL. An alternative surgical approach reduces variability following filament induction of experimental stroke in mice. Dis Model Mech 2017; 10:931-938. [PMID: 28550100 PMCID: PMC5536906 DOI: 10.1242/dmm.029108] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 05/24/2017] [Indexed: 01/11/2023] Open
Abstract
Animal models are essential for understanding the pathology of stroke and investigating potential treatments. However, in vivo stroke models are associated, particularly in mice, with high variability in lesion volume. We investigated whether a surgical refinement where reperfusion is not reliant on the Circle of Willis reduced outcome variability. Mice underwent 60 min of transient middle cerebral artery occlusion avoiding ligation of the external carotid artery. During reperfusion, the common carotid artery was either ligated (standard approach), or it was repaired to allow re-establishment of blood flow through the common carotid artery. All mice underwent MRI scanning for assessment of infarct volume, apparent diffusion coefficient and fractional anisotropy, along with terminal assessment of infarct volume by 2,3,5-triphenyltetrazolium chloride (TTC) staining. Repairing the common carotid artery following middle cerebral artery occlusion enhanced reperfusion (P<0.01) and reduced the variability seen in both total (histological analysis, P=0.008; T2-weighted MRI, P=0.015) and core (diffusion tensor MRI, P=0.043) lesion volume. Avoiding external carotid artery ligation may improve animal wellbeing, through reduced weight loss, while using an alternative surgical approach that enabled reperfusion through the common carotid artery decreased the variability in lesion volume seen within groups. Summary: An alternative surgical approach following middle cerebral artery occlusion, which allows reperfusion through the common carotid artery, decreases the variability in lesion volume seen within groups and reduces the number of animals required to detect a treatment effect.
Collapse
Affiliation(s)
- Melissa Trotman-Lucas
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester LE1 9HN, UK
| | - Michael E Kelly
- Preclinical Imaging Facility, Core Biotechnology Services, University of Leicester, Leicester LE1 9HN, UK
| | - Justyna Janus
- Preclinical Imaging Facility, Core Biotechnology Services, University of Leicester, Leicester LE1 9HN, UK
| | - Robert Fern
- Peninsula School of Medicine and Dentistry, University of Plymouth, Plymouth PL6 8BU, UK
| | - Claire L Gibson
- Department of Neuroscience, Psychology and Behaviour, University of Leicester, Leicester LE1 9HN, UK
| |
Collapse
|
26
|
Stokowska A, Atkins AL, Morán J, Pekny T, Bulmer L, Pascoe MC, Barnum SR, Wetsel RA, Nilsson JA, Dragunow M, Pekna M. Complement peptide C3a stimulates neural plasticity after experimental brain ischaemia. Brain 2016; 140:353-369. [PMID: 27956400 DOI: 10.1093/brain/aww314] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 10/19/2016] [Accepted: 10/20/2016] [Indexed: 11/12/2022] Open
Abstract
Ischaemic stroke induces endogenous repair processes that include proliferation and differentiation of neural stem cells and extensive rewiring of the remaining neural connections, yet about 50% of stroke survivors live with severe long-term disability. There is an unmet need for drug therapies to improve recovery by promoting brain plasticity in the subacute to chronic phase after ischaemic stroke. We previously showed that complement-derived peptide C3a regulates neural progenitor cell migration and differentiation in vitro and that C3a receptor signalling stimulates neurogenesis in unchallenged adult mice. To determine the role of C3a-C3a receptor signalling in ischaemia-induced neural plasticity, we subjected C3a receptor-deficient mice, GFAP-C3a transgenic mice expressing biologically active C3a in the central nervous system, and their respective wild-type controls to photothrombotic stroke. We found that C3a overexpression increased, whereas C3a receptor deficiency decreased post-stroke expression of GAP43 (P < 0.01), a marker of axonal sprouting and plasticity, in the peri-infarct cortex. To verify the translational potential of these findings, we used a pharmacological approach. Daily intranasal treatment of wild-type mice with C3a beginning 7 days after stroke induction robustly increased synaptic density (P < 0.01) and expression of GAP43 in peri-infarct cortex (P < 0.05). Importantly, the C3a treatment led to faster and more complete recovery of forepaw motor function (P < 0.05). We conclude that C3a-C3a receptor signalling stimulates post-ischaemic neural plasticity and intranasal treatment with C3a receptor agonists is an attractive approach to improve functional recovery after ischaemic brain injury.
Collapse
Affiliation(s)
- Anna Stokowska
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Alison L Atkins
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Javier Morán
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Tulen Pekny
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Linda Bulmer
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Michaela C Pascoe
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Scott R Barnum
- Department of Microbiology, University of Alabama, Birmingham, Alabama, USA
| | - Rick A Wetsel
- Research Center for Immunology and Autoimmune Diseases, Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas-Houston, Houston, Texas, USA
| | - Jonas A Nilsson
- Department of Surgery, Institute of Clinical Sciences, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Mike Dragunow
- Department of Pharmacology and Centre for Brain Research, Faculty of Medical and Health Sciences, The University of Auckland, Auckland, New Zealand
| | - Marcela Pekna
- Center for Brain Repair and Rehabilitation, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden .,Florey Institute of Neuroscience and Mental Health, Parkville, Victoria, Australia.,Hunter Medical Research Institute, University of Newcastle, New South Wales, Australia
| |
Collapse
|
27
|
Macrophages are essential for maintaining a M2 protective response early after ischemic brain injury. Neurobiol Dis 2016; 96:284-293. [DOI: 10.1016/j.nbd.2016.09.017] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 09/12/2016] [Accepted: 09/26/2016] [Indexed: 01/01/2023] Open
|
28
|
Orsini F, Chrysanthou E, Dudler T, Cummings WJ, Takahashi M, Fujita T, Demopulos G, De Simoni MG, Schwaeble W. Mannan binding lectin-associated serine protease-2 (MASP-2) critically contributes to post-ischemic brain injury independent of MASP-1. J Neuroinflammation 2016; 13:213. [PMID: 27577570 PMCID: PMC5006610 DOI: 10.1186/s12974-016-0684-6] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 08/18/2016] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Complement activation via the lectin activation pathway (LP) has been identified as the key mechanism behind post-ischemic tissue inflammation causing ischemia-reperfusion injury (IRI) which can significantly impact the clinical outcome of ischemic disease. This work defines the contributions of each of the three LP-associated enzymes-mannan-binding lectin-associated serine protease (MASP)-1, MASP-2, and MASP-3-to ischemic brain injury in experimental mouse models of stroke. METHODS Focal cerebral ischemia was induced in wild-type (WT) mice or mice deficient for defined complement components by transient middle cerebral artery occlusion (tMCAO) or three-vessel occlusion (3VO). The inhibitory MASP-2 antibody was administered systemically 7 and 3.5 days before and at reperfusion in WT mice in order to assure an effective MASP-2 inhibition throughout the study. Forty-eight hours after ischemia, neurological deficits and infarct volumes were assessed. C3 deposition and microglia/macrophage morphology were detected by immunohistochemical, immunofluorescence, and confocal analyses. RESULTS MASP-2-deficient mice (MASP-2(-/-)) and WT mice treated with an antibody that blocks MASP-2 activity had significantly reduced neurological deficits and histopathological damage after transient ischemia and reperfusion compared to WT or control-treated mice. Surprisingly, MASP-1/3(-/-) mice were not protected, while mice deficient in factor B (fB(-/-)) showed reduced neurological deficits compared to WT mice. Consistent with behavioral and histological data, MASP-2(-/-) had attenuated C3 deposition and presented with a significantly higher proportion of ramified, surveying microglia in contrast to the hypertrophic pro-inflammatory microglia/macrophage phenotype seen in the ischemic brain tissue of WT mice. CONCLUSIONS This work demonstrates the essential role of the low-abundant MASP-2 in the mediation of cerebral ischemia-reperfusion injury and demonstrates that targeting MASP-2 by an inhibitory therapeutic antibody markedly improved the neurological and histopathological outcome after focal cerebral ischemia. These results contribute to identifying the key lectin pathway component driving brain tissue injury following cerebral ischemia and call for a revision of the presently widely accepted view that MASP-1 is an essential activator of the lectin pathway effector component MASP-2.
Collapse
Affiliation(s)
- Franca Orsini
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, via La Masa, 19-20156, Milan, Italy
| | - Elvina Chrysanthou
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, via La Masa, 19-20156, Milan, Italy.,Department of Infection, Immunity and Inflammation, University of Leicester, MSB, University Road, Leicester, LE1 9HN, UK.,MRC Toxicology Unit, Leicester, LE1 9HN, UK
| | - Thomas Dudler
- OMEROS Corporation, 201 Elliott Ave W, Seattle, WA, 98119, USA
| | | | - Minoru Takahashi
- Fukushima Prefectural General Hygiene Institute and Department of Immunology, Fukushima Medical University, 1 Hikariga-oka, Fukushima City, 960-1295, Japan
| | - Teizo Fujita
- Fukushima Prefectural General Hygiene Institute and Department of Immunology, Fukushima Medical University, 1 Hikariga-oka, Fukushima City, 960-1295, Japan
| | | | - Maria-Grazia De Simoni
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri, via La Masa, 19-20156, Milan, Italy.
| | - Wilhelm Schwaeble
- Department of Infection, Immunity and Inflammation, University of Leicester, MSB, University Road, Leicester, LE1 9HN, UK.
| |
Collapse
|
29
|
|
30
|
Pires AO, Mendes-Pinheiro B, Teixeira FG, Anjo SI, Ribeiro-Samy S, Gomes ED, Serra SC, Silva NA, Manadas B, Sousa N, Salgado AJ. Unveiling the Differences of Secretome of Human Bone Marrow Mesenchymal Stem Cells, Adipose Tissue-Derived Stem Cells, and Human Umbilical Cord Perivascular Cells: A Proteomic Analysis. Stem Cells Dev 2016; 25:1073-83. [PMID: 27226274 DOI: 10.1089/scd.2016.0048] [Citation(s) in RCA: 158] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The use of human mesenchymal stem cells (hMSCs) has emerged as a possible therapeutic strategy for CNS-related conditions. Research in the last decade strongly suggests that MSC-mediated benefits are closely related with their secretome. Studies published in recent years have shown that the secretome of hMSCs isolated from different tissue sources may present significant variation. With this in mind, the present work performed a comparative proteomic-based analysis through mass spectrometry on the secretome of hMSCs derived from bone marrow (BMSCs), adipose tissue (ASCs), and human umbilical cord perivascular cells (HUCPVCs). The results revealed that BMSCs, ASCs, and HUCPVCs differed in their secretion of neurotrophic, neurogenic, axon guidance, axon growth, and neurodifferentiative proteins, as well as proteins with neuroprotective actions against oxidative stress, apoptosis, and excitotoxicity, which have been shown to be involved in several CNS disorder/injury processes. Although important changes were observed within the secretome of the cell populations that were analyzed, all cell populations shared the capability of secreting important neuroregulatory molecules. The difference in their secretion pattern may indicate that their secretome is specific to a condition of the CNS. Nevertheless, the confirmation that the secretome of MSCs isolated from different tissue sources is rich in neuroregulatory molecules represents an important asset not only for the development of future neuroregenerative strategies but also for their use as a therapeutic option for human clinical trials.
Collapse
Affiliation(s)
- Ana O Pires
- 1 Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho , Braga, Portugal .,2 ICVS/3B's-PT Government Associate Laboratory , Braga/Guimarães, Portugal
| | - Barbara Mendes-Pinheiro
- 1 Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho , Braga, Portugal .,2 ICVS/3B's-PT Government Associate Laboratory , Braga/Guimarães, Portugal
| | - Fábio G Teixeira
- 1 Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho , Braga, Portugal .,2 ICVS/3B's-PT Government Associate Laboratory , Braga/Guimarães, Portugal
| | - Sandra I Anjo
- 3 Department of Life Sciences, Faculty of Sciences and Technology, University of Coimbra , Coimbra, Portugal .,4 CNC-Center for Neurosciences and Cell Biology, University of Coimbra , Coimbra, Portugal
| | - Silvina Ribeiro-Samy
- 1 Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho , Braga, Portugal .,2 ICVS/3B's-PT Government Associate Laboratory , Braga/Guimarães, Portugal
| | - Eduardo D Gomes
- 1 Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho , Braga, Portugal .,2 ICVS/3B's-PT Government Associate Laboratory , Braga/Guimarães, Portugal
| | - Sofia C Serra
- 1 Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho , Braga, Portugal .,2 ICVS/3B's-PT Government Associate Laboratory , Braga/Guimarães, Portugal
| | - Nuno A Silva
- 1 Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho , Braga, Portugal .,2 ICVS/3B's-PT Government Associate Laboratory , Braga/Guimarães, Portugal
| | - Bruno Manadas
- 4 CNC-Center for Neurosciences and Cell Biology, University of Coimbra , Coimbra, Portugal
| | - Nuno Sousa
- 1 Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho , Braga, Portugal .,2 ICVS/3B's-PT Government Associate Laboratory , Braga/Guimarães, Portugal
| | - Antonio J Salgado
- 1 Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho , Braga, Portugal .,2 ICVS/3B's-PT Government Associate Laboratory , Braga/Guimarães, Portugal
| |
Collapse
|
31
|
Brennan FH, Lee JD, Ruitenberg MJ, Woodruff TM. Therapeutic targeting of complement to modify disease course and improve outcomes in neurological conditions. Semin Immunol 2016; 28:292-308. [PMID: 27049459 DOI: 10.1016/j.smim.2016.03.015] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 03/17/2016] [Accepted: 03/18/2016] [Indexed: 12/14/2022]
Abstract
The recognition that complement proteins are abundantly present and can have pathological roles in neurological conditions offers broad scope for therapeutic intervention. Accordingly, an increasing number of experimental investigations have explored the potential of harnessing the unique activation pathways, proteases, receptors, complexes, and natural inhibitors of complement, to mitigate pathology in acute neurotrauma and chronic neurodegenerative diseases. Here, we review mechanisms of complement activation in the central nervous system (CNS), and explore the effects of complement inhibition in cerebral ischemic-reperfusion injury, traumatic brain injury, spinal cord injury, Alzheimer's disease, amyotrophic lateral sclerosis, Parkinson's disease and Huntington's disease. We consider the challenges and opportunities arising from these studies. As complement therapies approach clinical translation, we provide perspectives on how promising complement-targeted therapeutics could become part of novel and effective future treatment options to improve outcomes in the initiation and progression stages of these debilitating CNS disorders.
Collapse
Affiliation(s)
- Faith H Brennan
- School of Biomedical Sciences, The University of Queensland, Brisbane 4072, Australia
| | - John D Lee
- School of Biomedical Sciences, The University of Queensland, Brisbane 4072, Australia
| | - Marc J Ruitenberg
- School of Biomedical Sciences, The University of Queensland, Brisbane 4072, Australia; Queensland Brain Institute, The University of Queensland, Brisbane 4072, Australia; Trauma, Critical Care and Recovery, Brisbane Diamantina Health Partners, The University of Queensland, Brisbane 4072, Australia
| | - Trent M Woodruff
- School of Biomedical Sciences, The University of Queensland, Brisbane 4072, Australia.
| |
Collapse
|
32
|
Silverman SM, Kim BJ, Howell GR, Miller J, John SWM, Wordinger RJ, Clark AF. C1q propagates microglial activation and neurodegeneration in the visual axis following retinal ischemia/reperfusion injury. Mol Neurodegener 2016; 11:24. [PMID: 27008854 PMCID: PMC4806521 DOI: 10.1186/s13024-016-0089-0] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 03/18/2016] [Indexed: 12/03/2022] Open
Abstract
Background C1q represents the initiating protein of the classical complement cascade, however recent findings indicate pathway independent roles such as developmental pruning of retinal ganglion cell (RGC) axons. Furthermore, chronic neuroinflammation, including increased expression of C1q and activation of microglia and astrocytes, appears to be a common finding among many neurodegenerative disease models. Here we compare the effects of a retinal ischemia/reperfusion (I/R) injury on glial activation and neurodegeneration in wild type (WT) and C1qa-deficient mice in the retina and superior colliculus (SC). Retinal I/R was induced in mice through elevation of intraocular pressure to 120 mmHg for 60 min followed by reperfusion. Glial cell activation and population changes were assessed using immunofluorescence. Neuroprotection was determined using histological measurements of retinal layer thickness, RGC counts, and visual function by flash electroretinography (ERG). Results Retinal I/R injury significantly upregulated C1q expression in the retina as early as 72 h and within 7 days in the superficial SC, and was sustained as long as 28 days. Accompanying increased C1q expression was activation of microglia and astrocytes as well as a significantly increased glial population density observed in the retina and SC. Microglial activation and changes in density were completely ablated in C1qa-deficient mice, interestingly however there was no effect on astrocytes. Furthermore, loss of C1qa significantly rescued I/R-induced loss of RGCs and protected against retinal layer thinning in comparison to WT mice. ERG assessment revealed early preservation of b-wave amplitude deficits from retinal I/R injury due to C1qa-deficiency that was lost by day 28. Conclusions Our results for the first time demonstrate the spatiotemporal changes in the neuroinflammatory response following retinal I/R injury at both local and distal sites of injury. In addition, we have shown a role for C1q as a primary mediator of microglial activation and pathological damage. This suggests developmental mechanisms of C1q may be re-engaged during injury response, modulation of which may be beneficial for neuroprotection. Electronic supplementary material The online version of this article (doi:10.1186/s13024-016-0089-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sean M Silverman
- North Texas Eye Research Institute, University of North Texas Health Science Center, CBH-441, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA
| | - Byung-Jin Kim
- North Texas Eye Research Institute, University of North Texas Health Science Center, CBH-441, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA
| | | | | | - Simon W M John
- The Jackson Laboratory, Bar Harbor, 04609, ME, USA.,Howard Hughes Medical Institute, Bar Harbor, ME, 04609, USA
| | - Robert J Wordinger
- North Texas Eye Research Institute, University of North Texas Health Science Center, CBH-441, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA
| | - Abbot F Clark
- North Texas Eye Research Institute, University of North Texas Health Science Center, CBH-441, 3500 Camp Bowie Boulevard, Fort Worth, TX, 76107, USA.
| |
Collapse
|
33
|
Dugue R, Barone FC. Ischemic, traumatic and neurodegenerative brain inflammatory changes. FUTURE NEUROLOGY 2016. [DOI: 10.2217/fnl.16.5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
This review serves to link the role of the immune system in the neuropathology of acute ischemic stroke, traumatic brain injury and neurodegenerative disease. The blood–brain barrier delineates the CNS from the peripheral immune system. However, the blood–cerebrospinal fluid barrier acts as a gate between the periphery and the brain, permitting immune activity crosstalk and modulation. In acute ischemic stroke, traumatic brain injury and other neurodegenerative diseases, the blood–brain barrier is compromised and an influx of inflammatory cells and plasma proteins occurs, resulting in edema, demyelination, cell dysfunction and death, and neurobehavioral changes. The role of the complement system, key cytokines, microglia and other neuroglia in brain degenerative pathology will be discussed.
Collapse
Affiliation(s)
- Rachelle Dugue
- Departments of Neurology & Physiology & Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY 11203, USA
| | - Frank C Barone
- Departments of Neurology & Physiology & Pharmacology, SUNY Downstate Medical Center, Brooklyn, NY 11203, USA
| |
Collapse
|
34
|
Alawieh A, Elvington A, Tomlinson S. Complement in the Homeostatic and Ischemic Brain. Front Immunol 2015; 6:417. [PMID: 26322048 PMCID: PMC4533015 DOI: 10.3389/fimmu.2015.00417] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 07/30/2015] [Indexed: 11/29/2022] Open
Abstract
The complement system is a component of the immune system involved in both recognition and response to pathogens, and it is implicated in an increasing number of homeostatic and disease processes. It is well documented that reperfusion of ischemic tissue results in complement activation and an inflammatory response that causes post-reperfusion injury. This occurs following cerebral ischemia and reperfusion and triggers secondary damage that extends beyond the initial infarcted area, an outcome that has rationalized the use of complement inhibitors as candidate therapeutics after stroke. In the central nervous system, however, recent studies have revealed that complement also has essential roles in synaptic pruning, neurogenesis, and neuronal migration. In the context of recovery after stroke, these apparent divergent functions of complement may account for findings that the protective effect of complement inhibition in the acute phase after stroke is not always maintained in the subacute and chronic phases. The development of effective stroke therapies based on modulation of the complement system will require a detailed understanding of complement-dependent processes in both early neurodegenerative events and delayed neuro-reparatory processes. Here, we review the role of complement in normal brain physiology, the events initiating complement activation after cerebral ischemia-reperfusion injury, and the contribution of complement to both injury and recovery. We also discuss how the design of future experiments may better characterize the dual role of complement in recovery after ischemic stroke.
Collapse
Affiliation(s)
- Ali Alawieh
- Neuroscience Institute, Department of Neurosciences, Medical University of South Carolina , Charleston, SC , USA
| | - Andrew Elvington
- Department of Pathology and Immunology, Washington University School of Medicine , St. Louis, MO , USA
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Ralph H. Johnson Veteran Affairs Medical Center, Medical University of South Carolina , Charleston, SC , USA
| |
Collapse
|
35
|
Zanier ER, Fumagalli S, Perego C, Pischiutta F, De Simoni MG. Shape descriptors of the "never resting" microglia in three different acute brain injury models in mice. Intensive Care Med Exp 2015. [PMID: 26215806 PMCID: PMC4513020 DOI: 10.1186/s40635-015-0039-0] [Citation(s) in RCA: 95] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The study of microglia and macrophage (M/M) morphology represents a key tool to understand the functional activation state and the pattern of distribution of these cells in acute brain injury. The identification of reliable quantitative morphological parameters is urgently needed to understand these cell roles in brain injury and to explore strategies aimed at therapeutically manipulating the inflammatory response. METHODS We used three different clinically relevant murine models of focal injury, namely, controlled cortical impact brain injury (traumatic brain injury (TBI)) and transient and permanent occlusion of middle cerebral artery (tMCAo and pMCAo, respectively). Twenty-four hours after injury, M/M cells were labeled by CD11b, and ×40 photomicrographs were acquired by unbiased sampling of the lesion core using a motorized stage microscope. Images were processed with Fiji software to obtain shape descriptors. RESULTS We validated several parameters, including area, perimeter, Feret's diameter (caliper), circularity, aspect ratio, and solidity, providing quantitative information on M/M morphology over wide tissue portions. We showed that the shape descriptors that best represent M/M ramification/elongation are area and perimeter, while circularity and solidity provide information on the ameboid shape. We also provide evidence of the involvement of different populations in local inflammatory events, with macrophages replacing microglia into the lesion core when reperfusion does not occur. Analysis of CD45(high)+ cell morphology, whose shape does not change, did not yield any difference, thus confirming the reliability of the approach. CONCLUSIONS We have defined specific morphological features that M/M acquire in response to different acute insults by applying a sensitive and readily applicable approach to cell morphological analysis in the brain tissue. Potential application of this method can be extended to all cell types able to change shape following activation, e.g., astrocytes, or to different disease states, including chronic pathologies.
Collapse
Affiliation(s)
- Elisa R Zanier
- IRCCS - Istituto di Ricerche Farmacologiche Mario Negri, Department of Neuroscience, Via La Masa 19, 20156, Milan, Italy,
| | | | | | | | | |
Collapse
|
36
|
Orsini F, De Blasio D, Zangari R, Zanier ER, De Simoni MG. Versatility of the complement system in neuroinflammation, neurodegeneration and brain homeostasis. Front Cell Neurosci 2014; 8:380. [PMID: 25426028 PMCID: PMC4224073 DOI: 10.3389/fncel.2014.00380] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 10/22/2014] [Indexed: 01/30/2023] Open
Abstract
The immune response after brain injury is highly complex and involves both local and systemic events at the cellular and molecular level. It is associated to a dramatic over-activation of enzyme systems, the expression of proinflammatory genes and the activation/recruitment of immune cells. The complement system represents a powerful component of the innate immunity and is highly involved in the inflammatory response. Complement components are synthesized predominantly by the liver and circulate in the bloodstream primed for activation. Moreover, brain cells can produce complement proteins and receptors. After acute brain injury, the rapid and uncontrolled activation of the complement leads to massive release of inflammatory anaphylatoxins, recruitment of cells to the injury site, phagocytosis and induction of blood brain barrier (BBB) damage. Brain endothelial cells are particularly susceptible to complement-mediated effects, since they are exposed to both circulating and locally synthesized complement proteins. Conversely, during neurodegenerative disorders, complement factors play distinct roles depending on the stage and degree of neuropathology. In addition to the deleterious role of the complement, increasing evidence suggest that it may also play a role in normal nervous system development (wiring the brain) and adulthood (either maintaining brain homeostasis or supporting regeneration after brain injury). This article represents a compendium of the current knowledge on the complement role in the brain, prompting a novel view that complement activation can result in either protective or detrimental effects in brain conditions that depend exquisitely on the nature, the timing and the degree of the stimuli that induce its activation. A deeper understanding of the acute, subacute and chronic consequences of complement activation is needed and may lead to new therapeutic strategies, including the ability of targeting selective step in the complement cascade.
Collapse
Affiliation(s)
- Franca Orsini
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri Milan, Italy
| | - Daiana De Blasio
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri Milan, Italy ; Department of Experimental and Clinical Sciences, University of Chieti Pescara, Italy
| | - Rosalia Zangari
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri Milan, Italy ; Department of Anesthesia and Critical Care Medicine, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan Milan, Italy
| | - Elisa R Zanier
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri Milan, Italy
| | - Maria-Grazia De Simoni
- Department of Neuroscience, IRCCS - Istituto di Ricerche Farmacologiche Mario Negri Milan, Italy
| |
Collapse
|
37
|
de la Rosa X, Cervera A, Kristoffersen AK, Valdés CP, Varma HM, Justicia C, Durduran T, Chamorro Á, Planas AM. Mannose-binding lectin promotes local microvascular thrombosis after transient brain ischemia in mice. Stroke 2014; 45:1453-9. [PMID: 24676774 DOI: 10.1161/strokeaha.113.004111] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
BACKGROUND AND PURPOSE Several lines of evidence support the involvement of mannose-binding lectin (MBL) in stroke brain damage. The lectin pathway of the complement system facilitates thrombin activation and clot formation under certain experimental conditions. In the present study, we examine whether MBL promotes thrombosis after ischemia/reperfusion and influences the course and prognosis of ischemic stroke. METHODS Middle cerebral artery occlusion/reperfusion was performed in MBL-deficient (n=85) and wild-type (WT; n=83) mice, and the brain lesion was assessed by MRI at days 1 and 7. Relative cerebral blood flow was monitored up to 6 hours after middle cerebral artery occlusion with laser speckle contrast imaging. Fibrin(ogen) was analyzed in the brain vasculature and plasma, and the effects of thrombin inhibitor argatroban were evaluated to assess the role of MBL in thrombin activation. RESULTS Infarct volumes and neurological deficits were smaller in MBL knockout mice than in WT mice. Relative cerebral blood flow values during middle cerebral artery occlusion and at reperfusion were similar in both groups, but decreased during the next 6 hours in the WT group only. Also, the WT mice showed more fibrin(ogen) in brain vessels and a better outcome after argatroban treatment. In contrast, argatroban did not improve the outcome in MBL knockout mice. CONCLUSIONS MBL promotes brain damage and functional impairment after brain ischemia/reperfusion in mice. These effects are secondary to intravascular thrombosis and impaired relative cerebral blood flow during reperfusion. Argatroban protects WT mice, but not MBL knockout mice, emphasizing a role of MBL in local thrombus formation in acute ischemia/reperfusion.
Collapse
Affiliation(s)
- Xavier de la Rosa
- From the Department of Brain Ischemia and Neurodegeneration, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain (X.d.l.R., C.J., A.M.P.); Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain (A.C., C.J., Á.C., A.M.P.); ICFO-Institut de Ciències Fotòniques, Castelldefels, Spain (A.K.K., C.P.V., H.M.V., T.D.); and Functional Stroke Unit, Hospital Clínic, Barcelona, Spain (A.C., Á.C.)
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
C1 esterase inhibitor reduces lower extremity ischemia/reperfusion injury and associated lung damage. PLoS One 2013; 8:e72059. [PMID: 23991040 PMCID: PMC3753343 DOI: 10.1371/journal.pone.0072059] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 07/05/2013] [Indexed: 12/13/2022] Open
Abstract
Background Ischemia/reperfusion injury of lower extremities and associated lung damage may result from thrombotic occlusion, embolism, trauma, or surgical intervention with prolonged ischemia and subsequent restoration of blood flow. This clinical entity is characterized by high morbidity and mortality. Deprivation of blood supply leads to molecular and structural changes in the affected tissue. Upon reperfusion inflammatory cascades are activated causing tissue injury. We therefore tested preoperative treatment for prevention of reperfusion injury by using C1 esterase inhibitor (C1 INH). Methods and Findings Wistar rats systemically pretreated with C1 INH (n = 6), APT070 (a membrane-targeted myristoylated peptidyl construct derived from human complement receptor 1, n = 4), vehicle (n = 7), or NaCl (n = 8) were subjected to 3h hind limb ischemia and 24h reperfusion. The femoral artery was clamped and a tourniquet placed under maintenance of a venous return. C1 INH treated rats showed significantly less edema in muscle (P<0.001) and lung and improved muscle viability (P<0.001) compared to controls and APT070. C1 INH prevented up-regulation of bradykinin receptor b1 (P<0.05) and VE-cadherin (P<0.01), reduced apoptosis (P<0.001) and fibrin deposition (P<0.01) and decreased plasma levels of pro-inflammatory cytokines, whereas deposition of complement components was not significantly reduced in the reperfused muscle. Conclusions C1 INH reduced edema formation locally in reperfused muscle as well as in lung, and improved muscle viability. C1 INH did not primarily act via inhibition of the complement system, but via the kinin and coagulation cascade. APT070 did not show beneficial effects in this model, despite potent inhibition of complement activation. Taken together, C1 INH might be a promising therapy to reduce peripheral ischemia/reperfusion injury and distant lung damage in complex and prolonged surgical interventions requiring tourniquet application.
Collapse
|
39
|
Stokowska A, Olsson S, Holmegaard L, Jood K, Blomstrand C, Jern C, Pekna M. Cardioembolic and small vessel disease stroke show differences in associations between systemic C3 levels and outcome. PLoS One 2013; 8:e72133. [PMID: 23977229 PMCID: PMC3748011 DOI: 10.1371/journal.pone.0072133] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2013] [Accepted: 07/05/2013] [Indexed: 01/24/2023] Open
Abstract
Background Activation of the complement system has been proposed to play a role in the pathophysiology of stroke. As the specific involvement of the complement proteins may be influenced by stroke etiology, we compared plasma C3 and C3a levels in patients with cardioembolic (CE) and small vessel disease (SVD) subtypes of ischemic stroke and control subjects and evaluated their association to outcome at three months and two years. Methodology/Principal Findings Plasma C3 and C3a levels in 79 CE and 79 SVD stroke patients, sampled within 10 days and at three months after stroke, and age- and sex-matched control subjects from The Sahlgrenska Academy Study on Ischemic Stroke were measured by ELISA. Functional outcome was assesed with modified Rankin Scale. In the CE group, plasma C3 levels were elevated only in the acute phase, whereas C3a was elevated at both time points. The follow-up phase plasma C3 levels in the upper third were associated with an increased risk of unfavorable outcome at three months (OR 7.12, CI 1.72–29.46, P = 0.007) as well as after two years (OR 8.25, CI 1.61–42.28, P = 0.011) after stroke. These associations withstand adjustment for age and sex. Conversely, three-month follow-up plasma C3a/C3 level ratios in the middle third were associated with favorable outcome after two years both in the univariate analysis (OR 0.19, CI 0.05–0.82, P = 0.026) and after adjustment for age and sex (OR 0.19, CI 0.04–0.88, P = 0.033). In the SVD group, plasma C3 and C3a levels were elevated at both time points but showed no significant associations with outcome. Conclusions Plasma C3 and C3a levels are elevated after CE and SVD stroke but show associations with outcome only in CE stroke.
Collapse
Affiliation(s)
- Anna Stokowska
- Institute of Neuroscience and Physiology, Department of Clinical Neuroscience and Rehabilitation, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
- * E-mail:
| | - Sandra Olsson
- Institute of Neuroscience and Physiology, Department of Clinical Neuroscience and Rehabilitation, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Lukas Holmegaard
- Institute of Neuroscience and Physiology, Department of Clinical Neuroscience and Rehabilitation, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Katarina Jood
- Institute of Neuroscience and Physiology, Department of Clinical Neuroscience and Rehabilitation, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Christian Blomstrand
- Institute of Neuroscience and Physiology, Department of Clinical Neuroscience and Rehabilitation, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Christina Jern
- Institute of Neuroscience and Physiology, Department of Clinical Neuroscience and Rehabilitation, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Marcela Pekna
- Institute of Neuroscience and Physiology, Department of Clinical Neuroscience and Rehabilitation, The Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
40
|
Heydenreich N, Nolte MW, Göb E, Langhauser F, Hofmeister M, Kraft P, Albert-Weissenberger C, Brede M, Varallyay C, Göbel K, Meuth SG, Nieswandt B, Dickneite G, Stoll G, Kleinschnitz C. C1-Inhibitor Protects From Brain Ischemia-Reperfusion Injury by Combined Antiinflammatory and Antithrombotic Mechanisms. Stroke 2012; 43:2457-67. [DOI: 10.1161/strokeaha.112.660340] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Nadine Heydenreich
- From the Department of Neurology (N.H., E.G., F.L., P.K., C.A.W., G.S., C.K.), University of Würzburg, Würzburg, Germany; CSL Behring GmbH (M.W.N., M.H., G.D.), Marburg, Germany; Department of Anesthesiology and Critical Care (M.B.), University of Würzburg, Würzburg, Germany; Department of Neuroradiology (C.V.), University of Würzburg, Würzburg, Germany; Department of Neurology–Inflammatory Disorders of the Nervous System and Neurooncology (K.G., S.G.M.), University of Münster, Münster, Germany
| | - Marc W. Nolte
- From the Department of Neurology (N.H., E.G., F.L., P.K., C.A.W., G.S., C.K.), University of Würzburg, Würzburg, Germany; CSL Behring GmbH (M.W.N., M.H., G.D.), Marburg, Germany; Department of Anesthesiology and Critical Care (M.B.), University of Würzburg, Würzburg, Germany; Department of Neuroradiology (C.V.), University of Würzburg, Würzburg, Germany; Department of Neurology–Inflammatory Disorders of the Nervous System and Neurooncology (K.G., S.G.M.), University of Münster, Münster, Germany
| | - Eva Göb
- From the Department of Neurology (N.H., E.G., F.L., P.K., C.A.W., G.S., C.K.), University of Würzburg, Würzburg, Germany; CSL Behring GmbH (M.W.N., M.H., G.D.), Marburg, Germany; Department of Anesthesiology and Critical Care (M.B.), University of Würzburg, Würzburg, Germany; Department of Neuroradiology (C.V.), University of Würzburg, Würzburg, Germany; Department of Neurology–Inflammatory Disorders of the Nervous System and Neurooncology (K.G., S.G.M.), University of Münster, Münster, Germany
| | - Friederike Langhauser
- From the Department of Neurology (N.H., E.G., F.L., P.K., C.A.W., G.S., C.K.), University of Würzburg, Würzburg, Germany; CSL Behring GmbH (M.W.N., M.H., G.D.), Marburg, Germany; Department of Anesthesiology and Critical Care (M.B.), University of Würzburg, Würzburg, Germany; Department of Neuroradiology (C.V.), University of Würzburg, Würzburg, Germany; Department of Neurology–Inflammatory Disorders of the Nervous System and Neurooncology (K.G., S.G.M.), University of Münster, Münster, Germany
| | - Marion Hofmeister
- From the Department of Neurology (N.H., E.G., F.L., P.K., C.A.W., G.S., C.K.), University of Würzburg, Würzburg, Germany; CSL Behring GmbH (M.W.N., M.H., G.D.), Marburg, Germany; Department of Anesthesiology and Critical Care (M.B.), University of Würzburg, Würzburg, Germany; Department of Neuroradiology (C.V.), University of Würzburg, Würzburg, Germany; Department of Neurology–Inflammatory Disorders of the Nervous System and Neurooncology (K.G., S.G.M.), University of Münster, Münster, Germany
| | - Peter Kraft
- From the Department of Neurology (N.H., E.G., F.L., P.K., C.A.W., G.S., C.K.), University of Würzburg, Würzburg, Germany; CSL Behring GmbH (M.W.N., M.H., G.D.), Marburg, Germany; Department of Anesthesiology and Critical Care (M.B.), University of Würzburg, Würzburg, Germany; Department of Neuroradiology (C.V.), University of Würzburg, Würzburg, Germany; Department of Neurology–Inflammatory Disorders of the Nervous System and Neurooncology (K.G., S.G.M.), University of Münster, Münster, Germany
| | - Christiane Albert-Weissenberger
- From the Department of Neurology (N.H., E.G., F.L., P.K., C.A.W., G.S., C.K.), University of Würzburg, Würzburg, Germany; CSL Behring GmbH (M.W.N., M.H., G.D.), Marburg, Germany; Department of Anesthesiology and Critical Care (M.B.), University of Würzburg, Würzburg, Germany; Department of Neuroradiology (C.V.), University of Würzburg, Würzburg, Germany; Department of Neurology–Inflammatory Disorders of the Nervous System and Neurooncology (K.G., S.G.M.), University of Münster, Münster, Germany
| | - Marc Brede
- From the Department of Neurology (N.H., E.G., F.L., P.K., C.A.W., G.S., C.K.), University of Würzburg, Würzburg, Germany; CSL Behring GmbH (M.W.N., M.H., G.D.), Marburg, Germany; Department of Anesthesiology and Critical Care (M.B.), University of Würzburg, Würzburg, Germany; Department of Neuroradiology (C.V.), University of Würzburg, Würzburg, Germany; Department of Neurology–Inflammatory Disorders of the Nervous System and Neurooncology (K.G., S.G.M.), University of Münster, Münster, Germany
| | - Csanad Varallyay
- From the Department of Neurology (N.H., E.G., F.L., P.K., C.A.W., G.S., C.K.), University of Würzburg, Würzburg, Germany; CSL Behring GmbH (M.W.N., M.H., G.D.), Marburg, Germany; Department of Anesthesiology and Critical Care (M.B.), University of Würzburg, Würzburg, Germany; Department of Neuroradiology (C.V.), University of Würzburg, Würzburg, Germany; Department of Neurology–Inflammatory Disorders of the Nervous System and Neurooncology (K.G., S.G.M.), University of Münster, Münster, Germany
| | - Kerstin Göbel
- From the Department of Neurology (N.H., E.G., F.L., P.K., C.A.W., G.S., C.K.), University of Würzburg, Würzburg, Germany; CSL Behring GmbH (M.W.N., M.H., G.D.), Marburg, Germany; Department of Anesthesiology and Critical Care (M.B.), University of Würzburg, Würzburg, Germany; Department of Neuroradiology (C.V.), University of Würzburg, Würzburg, Germany; Department of Neurology–Inflammatory Disorders of the Nervous System and Neurooncology (K.G., S.G.M.), University of Münster, Münster, Germany
| | - Sven G. Meuth
- From the Department of Neurology (N.H., E.G., F.L., P.K., C.A.W., G.S., C.K.), University of Würzburg, Würzburg, Germany; CSL Behring GmbH (M.W.N., M.H., G.D.), Marburg, Germany; Department of Anesthesiology and Critical Care (M.B.), University of Würzburg, Würzburg, Germany; Department of Neuroradiology (C.V.), University of Würzburg, Würzburg, Germany; Department of Neurology–Inflammatory Disorders of the Nervous System and Neurooncology (K.G., S.G.M.), University of Münster, Münster, Germany
| | - Bernhard Nieswandt
- From the Department of Neurology (N.H., E.G., F.L., P.K., C.A.W., G.S., C.K.), University of Würzburg, Würzburg, Germany; CSL Behring GmbH (M.W.N., M.H., G.D.), Marburg, Germany; Department of Anesthesiology and Critical Care (M.B.), University of Würzburg, Würzburg, Germany; Department of Neuroradiology (C.V.), University of Würzburg, Würzburg, Germany; Department of Neurology–Inflammatory Disorders of the Nervous System and Neurooncology (K.G., S.G.M.), University of Münster, Münster, Germany
| | - Gerhard Dickneite
- From the Department of Neurology (N.H., E.G., F.L., P.K., C.A.W., G.S., C.K.), University of Würzburg, Würzburg, Germany; CSL Behring GmbH (M.W.N., M.H., G.D.), Marburg, Germany; Department of Anesthesiology and Critical Care (M.B.), University of Würzburg, Würzburg, Germany; Department of Neuroradiology (C.V.), University of Würzburg, Würzburg, Germany; Department of Neurology–Inflammatory Disorders of the Nervous System and Neurooncology (K.G., S.G.M.), University of Münster, Münster, Germany
| | - Guido Stoll
- From the Department of Neurology (N.H., E.G., F.L., P.K., C.A.W., G.S., C.K.), University of Würzburg, Würzburg, Germany; CSL Behring GmbH (M.W.N., M.H., G.D.), Marburg, Germany; Department of Anesthesiology and Critical Care (M.B.), University of Würzburg, Würzburg, Germany; Department of Neuroradiology (C.V.), University of Würzburg, Würzburg, Germany; Department of Neurology–Inflammatory Disorders of the Nervous System and Neurooncology (K.G., S.G.M.), University of Münster, Münster, Germany
| | - Christoph Kleinschnitz
- From the Department of Neurology (N.H., E.G., F.L., P.K., C.A.W., G.S., C.K.), University of Würzburg, Würzburg, Germany; CSL Behring GmbH (M.W.N., M.H., G.D.), Marburg, Germany; Department of Anesthesiology and Critical Care (M.B.), University of Würzburg, Würzburg, Germany; Department of Neuroradiology (C.V.), University of Würzburg, Würzburg, Germany; Department of Neurology–Inflammatory Disorders of the Nervous System and Neurooncology (K.G., S.G.M.), University of Münster, Münster, Germany
| |
Collapse
|
41
|
Orsini F, Villa P, Parrella S, Zangari R, Zanier ER, Gesuete R, Stravalaci M, Fumagalli S, Ottria R, Reina JJ, Paladini A, Micotti E, Ribeiro-Viana R, Rojo J, Pavlov VI, Stahl GL, Bernardi A, Gobbi M, De Simoni MG. Targeting mannose-binding lectin confers long-lasting protection with a surprisingly wide therapeutic window in cerebral ischemia. Circulation 2012; 126:1484-94. [PMID: 22879370 DOI: 10.1161/circulationaha.112.103051] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND The involvement of the complement system in brain injury has been scarcely investigated. Here, we document the pivotal role of mannose-binding lectin (MBL), one of the recognition molecules of the lectin complement pathway, in brain ischemic injury. METHODS AND RESULTS Focal cerebral ischemia was induced in mice (by permanent or transient middle cerebral artery occlusion) and rats (by 3-vessel occlusion). We first observed that MBL is deposited on ischemic vessels up to 48 hours after injury and that functional MBL/MBL-associated serine protease 2 complexes are increased. Next, we demonstrated that (1) MBL(-/-) mice are protected from both transient and permanent ischemic injury; (2) Polyman2, the newly synthesized mannosylated molecule selected for its binding to MBL, improves neurological deficits and infarct volume when given up to 24 hours after ischemia in mice; (3) anti-MBL-A antibody improves neurological deficits and infarct volume when given up to 18 hours after ischemia, as assessed after 28 days in rats. CONCLUSIONS Our data show an important role for MBL in the pathogenesis of brain ischemic injury and provide a strong support to the concept that MBL inhibition may be a relevant therapeutic target in humans, one with a wide therapeutic window of application.
Collapse
|
42
|
Refined Qingkailing Protects MCAO Mice from Endoplasmic Reticulum Stress-Induced Apoptosis with a Broad Time Window. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2012; 2012:567872. [PMID: 22536287 PMCID: PMC3320064 DOI: 10.1155/2012/567872] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2011] [Revised: 01/02/2012] [Accepted: 01/03/2012] [Indexed: 11/18/2022]
Abstract
In the current study, we are investigating effect of refined QKL on ischemia-reperfusion-induced brain injury in mice. Methods. Mice were employed to induce ischemia-reperfusion injury of brain by middle cerebral artery occlusion (MCAO). RQKL solution was administered with different doses (0, 1.5, 3, and 6 mL/kg body weight) at the same time of onset of ischemia, and with the dose of 1.5 mL/kg at different time points (0, 1.5, 3, 6, and 9 h after MCAO). Neurological function and brain infarction were examined and cell apoptosis and ROS at prefrontal cortex were evaluated 24 h after MCAO, and western blot and intracellular calcium were also researched, respectively. Results. RQKL of all doses can improve neurological function and decrease brain infarction, and it performed significant effect in 0, 1.5, 3, and 6 h groups. Moreover, RQKL was able to reduce apoptotic process by reduction of caspase-3 expression, or restraint of eIF2a phosphorylation and caspase-12 activation. It was also able to reduce ROS and modulate intracellular calcium in the brain. Conclusion. RQKL can prevent ischemic-induced brain injury with a time window of 6 h, and its mechanism might be related to suppress ER stress-mediated apoptotic signaling.
Collapse
|
43
|
Füst G, Munthe-Fog L, Illes Z, Széplaki G, Molnar T, Pusch G, Hirschberg K, Szegedi R, Széplaki Z, Prohászka Z, Skjoedt MO, Garred P. Low ficolin-3 levels in early follow-up serum samples are associated with the severity and unfavorable outcome of acute ischemic stroke. J Neuroinflammation 2011; 8:185. [PMID: 22206485 PMCID: PMC3314397 DOI: 10.1186/1742-2094-8-185] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 12/29/2011] [Indexed: 11/13/2022] Open
Abstract
Background A number of data indicate that the lectin pathway of complement activation contributes to the pathophysiology of ischemic stroke. The lectin pathway may be triggered by the binding of mannose-binding lectin (MBL), ficolin-2 or ficolin-3 to different ligands. Although several papers demonstrated the significance of MBL in ischemic stroke, the role of ficolins has not been examined. Methods Sera were obtained within 12 hours after the onset of ischemic stroke (admission samples) and 3-4 days later (follow-up samples) from 65 patients. The control group comprised 100 healthy individuals and 135 patients with significant carotid stenosis (patient controls). The concentrations of ficolin-2 and ficolin-3, initiator molecules of the lectin complement pathway, were measured by ELISA methods. Concentration of C-reactive protein (CRP) was also determined by a particle-enhanced immunturbidimetric assay. Results Concentrations of both ficolin-2 and ficolin-3 were significantly (p < 0.001) decreased in both the admission and in the follow-up samples of patients with definite ischemic stroke as compared to healthy subjects. Concentrations of ficolin-2 and ficolin-3 were even higher in patient controls than in healthy subjects, indicating that the decreased levels in sera during the acute phase of stroke are related to the acute ischemic event. Ficolin-3 levels in the follow-up samples inversely correlated with the severity of stroke indicated by NIH scale on admission. In follow-up samples an inverse correlation was observed between ficolin-3 levels and concentration of S100β, an indicator of the size of cerebral infarct. Patients with low ficolin-3 levels and high CRP levels in the follow up samples had a significantly worse outcome (adjusted ORs 5.6 and 3.9, respectively) as measured by the modified Rankin scale compared to patients with higher ficolin-3 and lower CRP concentrations. High CRP concentrations were similarly predictive for worse outcome, and the effects of low ficolin-3 and high CRP were independent. Conclusions Our findings indicate that ficolin-mediated lectin pathways of complement activation contribute to the pathogenesis of ischemic stroke and may be additive to complement-independent inflammatory processes.
Collapse
Affiliation(s)
- George Füst
- 3rd Department of Internal Medicine, Semmelwies University, Budapest, Hungary.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Perego C, Fumagalli S, De Simoni MG. Temporal pattern of expression and colocalization of microglia/macrophage phenotype markers following brain ischemic injury in mice. J Neuroinflammation 2011; 8:174. [PMID: 22152337 PMCID: PMC3251548 DOI: 10.1186/1742-2094-8-174] [Citation(s) in RCA: 392] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2011] [Accepted: 12/10/2011] [Indexed: 01/18/2023] Open
Abstract
Background Emerging evidence indicates that, similarly to what happens for peripheral macrophages, microglia can express different phenotypes depending on microenvironmental signals. In spite of the large literature on inflammation after ischemia, information on M/M phenotype marker expression, their colocalization and temporal evolution in the injured brain is lacking. The present study investigates the presence of microglia/macrophage phenotype markers, their temporal expression, whether they are concomitantly expressed by the same subpopulation, or they are expressed at distinct phases or locations in relation to the ischemic lesion. Methods Volume of ischemic lesion, neuronal counts and TUNEL staining were assessed in C57Bl/6 mice at 6-12-24-48 h and 7d after permanent occlusion of the middle cerebral artery. At the same time points, the expression, distribution in the lesioned area, association with a definite morphology and coexpression of the microglia/macrophage markers CD11b, CD45, CD68, Ym1, CD206 were assessed by immunostaining and confocal microscopy. Results The results show that: 1) the ischemic lesion induces the expression of selected microglia/macrophage markers that develop over time, each with a specific pattern; 2) each marker has a given localization in the lesioned area with no apparent changes during time, with the exception of CD68 that is confined in the border zone of the lesion at early times but it greatly increases and invades the ischemic core at 7d; 3) while CD68 is expressed in both ramified and globular CD11b cells, Ym1 and CD206 are exclusively expressed by globular CD11b cells. Conclusions These data show that the ischemic lesion is accompanied by activation of specific microglia/macrophage phenotype that presents distinctive spatial and temporal features. These different states of microglia/macrophages reflect the complexity of these cells and their ability to differentiate towards a multitude of phenotypes depending on the surrounding micro-environmental signals that can change over time. The data presented in this study provide a basis for understanding this complex response and for developing strategies resulting in promotion of a protective inflammatory phenotype.
Collapse
Affiliation(s)
- Carlo Perego
- Laboratory of Inflammation and Nervous System Diseases, Department of Neuroscience, Mario Negri Institute for Pharmacological Research, Milan, Italy
| | | | | |
Collapse
|
45
|
Gandhi J, Cashman SM, Kumar-Singh R. Soluble CD59 expressed from an adenovirus in vivo is a potent inhibitor of complement deposition on murine liver vascular endothelium. PLoS One 2011; 6:e21621. [PMID: 21720565 PMCID: PMC3123367 DOI: 10.1371/journal.pone.0021621] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2011] [Accepted: 06/03/2011] [Indexed: 11/19/2022] Open
Abstract
Inappropriate activation of complement on the vascular endothelium of specific organs, or systemically, underlies the etiology of a number of diseases. These disorders include atypical hemolytic uremic syndrome, membranoproliferative glomerulonephritis, atherosclerosis, age-related macular degeneration, diabetic retinopathy, and transplant rejection. Inhibition of the terminal step of complement activation, i.e. formation of the membrane attack complex, using CD59 has the advantage of retaining the upstream processes of the complement cascade necessary for fighting pathogens and retaining complement's crucial role in tissue homeostasis. Previous studies have shown the necessity of membrane targeting of soluble CD59 in order for it to prove an effective inhibitor of complement deposition both in vitro and in vivo. In this study we have generated an in vivo model of human complement activation on murine liver vascular endothelium. This model should prove useful for the development of anti-complement therapies for complement-induced pathologies of vascular endothelium. Using this model, we have demonstrated the viability of a non membrane-targeted soluble CD59 to significantly inhibit complement deposition on the endothelium of murine liver vasculature when expressed in vivo from an adenovirus. This result, unanticipated based on prior studies, suggests that the use of non membrane-targeted sCD59 as an anti-complement therapy be re-visited.
Collapse
Affiliation(s)
- Jarel Gandhi
- Department of Ophthalmology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Siobhan M. Cashman
- Department of Ophthalmology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
| | - Rajendra Kumar-Singh
- Department of Ophthalmology, Tufts University School of Medicine, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
46
|
Gesuete R, Orsini F, Zanier ER, Albani D, Deli MA, Bazzoni G, De Simoni MG. Glial Cells Drive Preconditioning-Induced Blood-Brain Barrier Protection. Stroke 2011; 42:1445-53. [DOI: 10.1161/strokeaha.110.603266] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Raffaella Gesuete
- From the Mario Negri Institute (R.G., F.O., E.R.Z., D.A., G.B., M.G.D.S.), Milan, Italy, and Biological Research Center (M.A.D.), Szeged, Hungary
| | - Franca Orsini
- From the Mario Negri Institute (R.G., F.O., E.R.Z., D.A., G.B., M.G.D.S.), Milan, Italy, and Biological Research Center (M.A.D.), Szeged, Hungary
| | - Elisa R. Zanier
- From the Mario Negri Institute (R.G., F.O., E.R.Z., D.A., G.B., M.G.D.S.), Milan, Italy, and Biological Research Center (M.A.D.), Szeged, Hungary
| | - Diego Albani
- From the Mario Negri Institute (R.G., F.O., E.R.Z., D.A., G.B., M.G.D.S.), Milan, Italy, and Biological Research Center (M.A.D.), Szeged, Hungary
| | - Maria A. Deli
- From the Mario Negri Institute (R.G., F.O., E.R.Z., D.A., G.B., M.G.D.S.), Milan, Italy, and Biological Research Center (M.A.D.), Szeged, Hungary
| | - Gianfranco Bazzoni
- From the Mario Negri Institute (R.G., F.O., E.R.Z., D.A., G.B., M.G.D.S.), Milan, Italy, and Biological Research Center (M.A.D.), Szeged, Hungary
| | - Maria-Grazia De Simoni
- From the Mario Negri Institute (R.G., F.O., E.R.Z., D.A., G.B., M.G.D.S.), Milan, Italy, and Biological Research Center (M.A.D.), Szeged, Hungary
| |
Collapse
|
47
|
Castellano G, Melchiorre R, Loverre A, Ditonno P, Montinaro V, Rossini M, Divella C, Battaglia M, Lucarelli G, Annunziata G, Palazzo S, Selvaggi FP, Staffieri F, Crovace A, Daha MR, Mannesse M, van Wetering S, Paolo Schena F, Grandaliano G. Therapeutic targeting of classical and lectin pathways of complement protects from ischemia-reperfusion-induced renal damage. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 176:1648-59. [PMID: 20150432 DOI: 10.2353/ajpath.2010.090276] [Citation(s) in RCA: 175] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Ischemia-reperfusion injury is the major cause of delayed graft function in transplanted kidneys, an early event significantly affecting long-term graft function and survival. Several studies in rodents suggest that the alternative pathway of the complement system plays a pivotal role in renal ischemia-reperfusion injury. However, limited information is currently available from humans and larger animals. Here we demonstrated that 30 minutes of ischemia resulted in the induction of C4d/C1q, C4d/MLB, and MBL/MASP-2 deposits in a swine model of ischemia-reperfusion injury. The infusion of C1-inhibitor led to a significant reduction in peritubular capillary and glomerular C4d and C5b-9 deposition. Moreover, complement-inhibiting treatment significantly reduced the numbers of infiltrating CD163(+), SWC3a(+), CD4a(+), and CD8a(+) cells. C1-inhibitor administration led to significant inhibition of tubular damage and tubular epithelial cells apoptosis. Interestingly, we report that focal C4d-deposition colocalizes with C1q and MBL at the peritubular and glomerular capillary levels also in patients with delayed graft function. In conclusion, we demonstrated the activation and a pathogenic role of classical and lectin pathways of complement in a swine model of ischemia-reperfusion-induced renal damage. Therefore, inhibition of these two pathways might represent a novel therapeutic approach in the prevention of delayed graft function in kidney transplant recipients.
Collapse
Affiliation(s)
- Giuseppe Castellano
- Department of Emergency and Organ Transplantation, University of Bari, Policlinico, Piazza Giulio Cesare 11, Bari, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Zacharia BE, Hickman ZL, Grobelny BT, DeRosa PA, Ducruet AF, Connolly ES. Complement inhibition as a proposed neuroprotective strategy following cardiac arrest. Mediators Inflamm 2010; 2009:124384. [PMID: 20150958 PMCID: PMC2817500 DOI: 10.1155/2009/124384] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Revised: 10/18/2009] [Accepted: 11/04/2009] [Indexed: 01/08/2023] Open
Abstract
Out-of-hospital cardiac arrest (OHCA) is a devastating disease process with neurological injury accounting for a disproportionate amount of the morbidity and mortality following return of spontaneous circulation. A dearth of effective treatment strategies exists for global cerebral ischemia-reperfusion (GCI/R) injury following successful resuscitation from OHCA. Emerging preclinical as well as recent human clinical evidence suggests that activation of the complement cascade plays a critical role in the pathogenesis of GCI/R injury following OHCA. In addition, it is well established that complement inhibition improves outcome in both global and focal models of brain ischemia. Due to the profound impact of GCI/R injury following OHCA, and the relative lack of effective neuroprotective strategies for this pathologic process, complement inhibition provides an exciting opportunity to augment existing treatments to improve patient outcomes. To this end, this paper will explore the pathophysiology of complement-mediated GCI/R injury following OHCA.
Collapse
Affiliation(s)
- Brad E Zacharia
- Department of Neurological Surgery, Columbia University, New York, NY 10032, USA.
| | | | | | | | | | | |
Collapse
|
49
|
ter Weeme M, Vonk ABA, Kupreishvili K, van Ham M, Zeerleder S, Wouters D, Stooker W, Eijsman L, Van Hinsbergh VWM, Krijnen PAJ, Niessen HWM. Activated complement is more extensively present in diseased aortic valves than naturally occurring complement inhibitors: a sign of ongoing inflammation. Eur J Clin Invest 2010; 40:4-10. [PMID: 19843156 DOI: 10.1111/j.1365-2362.2009.02216.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
BACKGROUND Recent studies indicate a role for complement in the pathogenesis of aortic valve disease. However, the role of naturally occurring anti-complement mediators in this context is unknown. In this study, we have analysed this in three different pathological conditions of the aortic valve: degeneration, atherosclerosis and bacterial endocarditis. MATERIALS AND METHODS Human aortic valves were obtained at autopsy (n = 30): 5 control valves, 10 aortic valves with atherosclerotic changes, 10 aortic valves with degenerative changes and 5 degenerative changed aortic valves with bacterial infection. These valves were analysed immunohistochemically for the presence of activated complement (C3d and C5b9) and the complement inhibitors C1-inh and clusterin. Areas of positivity were then quantified. RESULTS C3d, C5b9 and the complement inhibitors C1-inh and clusterin depositions were mainly found in the endothelium and extracellular matrix in aortic valves. All these mediators were already present in control valves, but the area of positivity increased significantly in response to the different diseases, with the highest increase in response to bacterial endocarditis. Interestingly, in all three aortic diseases, the depositions of complement were significantly more widespread than that of their inhibitors. CONCLUSIONS Our study indicates that anti-complement mediators (C1-inh and clusterin) are deposited in diseased aortic valves together with activated complement, indicating an existing counter response against complement locally in the valve. However, deposition of activated complement is significantly more widespread than that of its inhibitors, which could explain ongoing inflammation in those diseased aortic valves.
Collapse
|
50
|
Lelekov-Boissard T, Chapuisat G, Boissel JP, Grenier E, Dronne MA. Exploration of beneficial and deleterious effects of inflammation in stroke: dynamics of inflammation cells. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2009; 367:4699-4716. [PMID: 19884176 DOI: 10.1098/rsta.2009.0184] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
The inflammatory process during stroke consists of activation of resident brain microglia and recruitment of leucocytes, namely neutrophils and monocytes/macrophages. During inflammation, microglial cells, neutrophils and macrophages secrete inflammatory cytokines and chemokines, and phagocytize dead cells. The recruitment of blood cells (neutrophils and macrophages) is mediated by the leucocyte-endothelium interactions and more specifically by cell adhesion molecules. A mathematical model is proposed to represent the dynamics of various brain cells and of immune cells (neutrophils and macrophages). This model is based on a set of six ordinary differential equations and explores the beneficial and deleterious effects of inflammation, respectively phagocytosis by immune cells and the release of pro-inflammatory mediators and nitric oxide (NO). The results of our simulations are qualitatively consistent with those observed in experiments in vivo and would suggest that the increase of phagocytosis could contribute to the increase of the percentage of living cells. The inhibition of the production of cytokines and NO and the blocking of neutrophil and macrophage infiltration into the brain parenchyma led also to the improvement of brain cell survival. This approach may help to explore the respective contributions of the beneficial and deleterious roles of the inflammatory process in stroke, and to study various therapeutic strategies in order to reduce stroke damage.
Collapse
|