1
|
Binder NF, El Amki M, Glück C, Middleham W, Reuss AM, Bertolo A, Thurner P, Deffieux T, Lambride C, Epp R, Handelsmann HL, Baumgartner P, Orset C, Bethge P, Kulcsar Z, Aguzzi A, Tanter M, Schmid F, Vivien D, Wyss MT, Luft A, Weller M, Weber B, Wegener S. Leptomeningeal collaterals regulate reperfusion in ischemic stroke and rescue the brain from futile recanalization. Neuron 2024; 112:1456-1472.e6. [PMID: 38412858 DOI: 10.1016/j.neuron.2024.01.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 11/18/2023] [Accepted: 01/30/2024] [Indexed: 02/29/2024]
Abstract
Recanalization is the mainstay of ischemic stroke treatment. However, even with timely clot removal, many stroke patients recover poorly. Leptomeningeal collaterals (LMCs) are pial anastomotic vessels with yet-unknown functions. We applied laser speckle imaging, ultrafast ultrasound, and two-photon microscopy in a thrombin-based mouse model of stroke and fibrinolytic treatment to show that LMCs maintain cerebral autoregulation and allow for gradual reperfusion, resulting in small infarcts. In mice with poor LMCs, distal arterial segments collapse, and deleterious hyperemia causes hemorrhage and mortality after recanalization. In silico analyses confirm the relevance of LMCs for preserving perfusion in the ischemic region. Accordingly, in stroke patients with poor collaterals undergoing thrombectomy, rapid reperfusion resulted in hemorrhagic transformation and unfavorable recovery. Thus, we identify LMCs as key components regulating reperfusion and preventing futile recanalization after stroke. Future therapeutic interventions should aim to enhance collateral function, allowing for beneficial reperfusion after stroke.
Collapse
Affiliation(s)
- Nadine Felizitas Binder
- Department of Neurology, University Hospital and University of Zurich, Zürich, Switzerland; Neuroscience Center Zurich, University of Zurich, ETH Zurich, Zurich, Switzerland
| | - Mohamad El Amki
- Department of Neurology, University Hospital and University of Zurich, Zürich, Switzerland; Neuroscience Center Zurich, University of Zurich, ETH Zurich, Zurich, Switzerland
| | - Chaim Glück
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, 8057 Zürich, Switzerland; Neuroscience Center Zurich, University of Zurich, ETH Zurich, Zurich, Switzerland
| | - William Middleham
- Department of Neurology, University Hospital and University of Zurich, Zürich, Switzerland; Neuroscience Center Zurich, University of Zurich, ETH Zurich, Zurich, Switzerland
| | - Anna Maria Reuss
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Schmelzbergstrasse 12, 8091 Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich, ETH Zurich, Zurich, Switzerland
| | - Adrien Bertolo
- Iconeus, 6 rue Jean Calvin, Paris, France; Physics for Medicine, INSERM U1273, ESPCI Paris, CNRS UMR 8063, PSL Research University, 17 rue Moreau, Paris, France
| | - Patrick Thurner
- Department of Neuroradiology, University Hospital and University of Zurich, Zürich, France
| | - Thomas Deffieux
- Physics for Medicine, INSERM U1273, ESPCI Paris, CNRS UMR 8063, PSL Research University, 17 rue Moreau, Paris, France
| | - Chryso Lambride
- Department of Neurology, University Hospital and University of Zurich, Zürich, Switzerland; ARTORG Center for Biomedical Engineering Research, University of Bern, Bern, Switzerland; Neuroscience Center Zurich, University of Zurich, ETH Zurich, Zurich, Switzerland
| | - Robert Epp
- Institute of Fluid Dynamics, ETH Zurich, Zurich, Switzerland
| | - Hannah-Lea Handelsmann
- Department of Neurology, University Hospital and University of Zurich, Zürich, Switzerland
| | - Philipp Baumgartner
- Department of Neurology, University Hospital and University of Zurich, Zürich, Switzerland
| | - Cyrille Orset
- Normandie University, UNICAEN, INSERM, Unité Mixte de Recherche-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Institute Blood and Brain @ Caen Normandie, GIP Cyceron, Caen, France
| | - Philipp Bethge
- Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich, ETH Zurich, Zurich, Switzerland
| | - Zsolt Kulcsar
- Department of Neuroradiology, University Hospital and University of Zurich, Zürich, France
| | - Adriano Aguzzi
- Institute of Neuropathology, University Hospital Zurich, University of Zurich, Schmelzbergstrasse 12, 8091 Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich, ETH Zurich, Zurich, Switzerland
| | - Mickael Tanter
- Physics for Medicine, INSERM U1273, ESPCI Paris, CNRS UMR 8063, PSL Research University, 17 rue Moreau, Paris, France
| | - Franca Schmid
- ARTORG Center for Biomedical Engineering Research, University of Bern, Bern, Switzerland
| | - Denis Vivien
- Normandie University, UNICAEN, INSERM, Unité Mixte de Recherche-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), Institute Blood and Brain @ Caen Normandie, GIP Cyceron, Caen, France; Department of Clinical Research, Caen Normandie University Hospital, Caen, France
| | - Matthias Tasso Wyss
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, 8057 Zürich, Switzerland; Neuroscience Center Zurich, University of Zurich, ETH Zurich, Zurich, Switzerland
| | - Andreas Luft
- Department of Neurology, University Hospital and University of Zurich, Zürich, Switzerland; Neuroscience Center Zurich, University of Zurich, ETH Zurich, Zurich, Switzerland
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, Zürich, Switzerland; Neuroscience Center Zurich, University of Zurich, ETH Zurich, Zurich, Switzerland
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of Zurich, Winterthurerstrasse 190, 8057 Zürich, Switzerland; Neuroscience Center Zurich, University of Zurich, ETH Zurich, Zurich, Switzerland
| | - Susanne Wegener
- Department of Neurology, University Hospital and University of Zurich, Zürich, Switzerland; Neuroscience Center Zurich, University of Zurich, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
2
|
Chen YL, Wang Y, Fang QY, Wang T, Chen C, Gao TY, Wu M, Zhang WP, Lu YB. PARP-1 inhibitor alleviates cerebral ischemia/reperfusion injury by reducing PARylation of HK-1 and LDH in mice. Eur J Pharmacol 2024; 967:176377. [PMID: 38346469 DOI: 10.1016/j.ejphar.2024.176377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 01/06/2024] [Accepted: 01/31/2024] [Indexed: 02/18/2024]
Abstract
Poly (ADP-ribose) polymerase-1 (PARP-1) activity significantly increases during cerebral ischemia/reperfusion. PARP-1 is an NAD+-consumption enzyme. PARP-1 hyperactivity causes intracellular NAD+ deficiency and bioenergetic collapse, contributing to neuronal death. Besides, the powerful trigger of PARP-1 causes the catalyzation of poly (ADP-ribosyl)ation (PARylation), a posttranslational modification of proteins. Here, we found that PARP-1 was activated in the ischemic brain tissue during middle-cerebral-artery occlusion and reperfusion (MCAO/R) for 24 h, and PAR accumulated in the neurons in mice. Using immunoprecipitation, Western blotting, liquid chromatography-mass spectrometry, and 3D-modeling analysis, we revealed that the activation of PARP-1 caused PARylation of hexokinase-1 and lactate dehydrogenase-B, which, therefore, caused the inhibition of these enzyme activities and the resulting cell energy metabolism collapse. PARP-1 inhibition significantly reversed the activity of hexokinase and lactate dehydrogenase, decreased infarct volume, and improved neuronal deficiency. PARP-1 inhibitor combined with pyruvate further alleviated MCAO/R-induced ischemic brain injury in mice. As such, we conclude that PARP-1 inhibitor alleviates neuronal death partly by inhibiting the PARylation of metabolic-related enzymes and reversing metabolism reprogramming during cerebral ischemia/reperfusion injury in mice. PARP-1 inhibitor combined with pyruvate might be a promising therapeutic approach against brain ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Ya-Ling Chen
- Department of Pharmacology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Yi Wang
- Department of Pharmacology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Qiu-Yu Fang
- Department of Pharmacology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China; Department of Thoracic Surgery, The Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
| | - Tong Wang
- Department of Pharmacology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Cong Chen
- Department of Pharmacology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Tong-Yao Gao
- Department of Pharmacology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Ming Wu
- Department of Thoracic Surgery, The Second Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310009, China
| | - Wei-Ping Zhang
- Department of Pharmacology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| | - Yun-Bi Lu
- Department of Pharmacology, School of Basic Medical Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| |
Collapse
|
3
|
Liu C, Xie J, Sun S, Li H, Li T, Jiang C, Chen X, Wang J, Le A, Wang J, Li Z, Wang J, Wang W. Hemorrhagic Transformation After Tissue Plasminogen Activator Treatment in Acute Ischemic Stroke. Cell Mol Neurobiol 2022; 42:621-646. [PMID: 33125600 PMCID: PMC11441267 DOI: 10.1007/s10571-020-00985-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 10/22/2020] [Indexed: 12/17/2022]
Abstract
Hemorrhagic transformation (HT) is a common complication after thrombolysis with recombinant tissue-type plasminogen activator (rt-PA) in ischemic stroke. In this article, recent research progress of HT in vivo and in vitro studies was reviewed. We have discussed new potential mechanisms and possible experimental models of HT development, as well as possible biomarkers and treatment methods. Meanwhile, we compared and analyzed rodent models, large animal models and in vitro BBB models of HT, and the limitations of these models were discussed. The molecular mechanism of HT was investigated in terms of BBB disruption, rt-PA neurotoxicity and the effect of neuroinflammation, matrix metalloproteinases, reactive oxygen species. The clinical features to predict HT were represented including blood biomarkers and clinical factors. Recent progress in neuroprotective strategies to improve HT after stroke treated with rt-PA is outlined. Further efforts need to be made to reduce the risk of HT after rt-PA therapy and improve the clinical prognosis of patients with ischemic stroke.
Collapse
Affiliation(s)
- Chengli Liu
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Jie Xie
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Shanshan Sun
- Department of Ultrasound Imaging, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Hui Li
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Tianyu Li
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Chao Jiang
- Department of Neurology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, People's Republic of China
| | - Xuemei Chen
- Department of Anatomy, College of Basic Medical Sciences, Zhengzhou University, Henan, 450000, People's Republic of China
| | - Junmin Wang
- Department of Anatomy, College of Basic Medical Sciences, Zhengzhou University, Henan, 450000, People's Republic of China
| | - Anh Le
- Washington University in St. Louis, Saint Louis, MO, 63130, USA
| | - Jiarui Wang
- The Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Zhanfei Li
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Jian Wang
- Department of Anatomy, College of Basic Medical Sciences, Zhengzhou University, Henan, 450000, People's Republic of China.
| | - Wei Wang
- Department of Traumatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
4
|
El Amki M, Glück C, Binder N, Middleham W, Wyss MT, Weiss T, Meister H, Luft A, Weller M, Weber B, Wegener S. Neutrophils Obstructing Brain Capillaries Are a Major Cause of No-Reflow in Ischemic Stroke. Cell Rep 2021; 33:108260. [PMID: 33053341 DOI: 10.1016/j.celrep.2020.108260] [Citation(s) in RCA: 141] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 08/18/2020] [Accepted: 09/21/2020] [Indexed: 12/29/2022] Open
Abstract
Despite successful clot retrieval in large vessel occlusion stroke, ∼50% of patients have an unfavorable clinical outcome. The mechanisms underlying this functional reperfusion failure remain unknown, and therapeutic options are lacking. In the thrombin-model of middle cerebral artery (MCA) stroke in mice, we show that, despite successful thrombolytic recanalization of the proximal MCA, cortical blood flow does not fully recover. Using in vivo two-photon imaging, we demonstrate that this is due to microvascular obstruction of ∼20%-30% of capillaries in the infarct core and penumbra by neutrophils adhering to distal capillary segments. Depletion of circulating neutrophils using an anti-Ly6G antibody restores microvascular perfusion without increasing the rate of hemorrhagic complications. Strikingly, infarct size and functional deficits are smaller in mice treated with anti-Ly6G. Thus, we propose neutrophil stalling of brain capillaries to contribute to reperfusion failure, which offers promising therapeutic avenues for ischemic stroke.
Collapse
Affiliation(s)
- Mohamad El Amki
- Department of Neurology, University Hospital and University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland
| | - Chaim Glück
- Experimental Imaging and Neuroenergetics, Institute of Pharmacology and Toxicology, University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland
| | - Nadine Binder
- Department of Neurology, University Hospital and University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland
| | - William Middleham
- Department of Neurology, University Hospital and University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland
| | - Matthias T Wyss
- Experimental Imaging and Neuroenergetics, Institute of Pharmacology and Toxicology, University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland
| | - Tobias Weiss
- Department of Neurology, University Hospital and University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland
| | - Hanna Meister
- Department of Neurology, University Hospital and University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland
| | - Andreas Luft
- Department of Neurology, University Hospital and University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland
| | - Michael Weller
- Department of Neurology, University Hospital and University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland
| | - Bruno Weber
- Experimental Imaging and Neuroenergetics, Institute of Pharmacology and Toxicology, University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland.
| | - Susanne Wegener
- Department of Neurology, University Hospital and University of Zurich, and Zurich Neuroscience Center, Zurich, Switzerland.
| |
Collapse
|
5
|
Poly(ADP-Ribose) Polymerase Inhibitor PJ34 Reduces Brain Damage after Stroke in the Neonatal Mouse Brain. Curr Issues Mol Biol 2021; 43:301-312. [PMID: 34200155 PMCID: PMC8929080 DOI: 10.3390/cimb43010025] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 12/12/2022] Open
Abstract
The poly(ADP-ribose) polymerase inhibitor PJ34 has recently been reported to increase cerebral blood flow, via the endothelial NO synthase, in the naive mouse brain throughout life. We addressed here the benefits of PJ34 after neonatal ischemia on hemodynamics and components of the neurovascular unit including the blood-brain barrier (BBB), microglia, and astrocytes. Nine-day-old mice were subjected to permanent MCA occlusion (pMCAo), and treated with either PBS or PJ34 (10 mg/kg). Mean blood-flow velocities (mBFV) were measured in both internal carotid arteries (ICA) and basilar trunk (BT) using Doppler-ultrasonography. BBB opening was assessed through somatostatin-receptor type-2 internalization and immunohistochemistry at 24 and 48 h. Lesion areas were measured 8 days after ischemia. In PBS-treated mice, pMCAo involved a drop in mBFV in the left ICA (p < 0.001 vs. basal), whereas mBFV remained stable in both right ICA and BT. PJ34 prevented this drop in the left ICA (NS vs. basal) and increased mBFV in the right ICA (p = 0.0038 vs. basal). No modification was observed in the BT. In contrast to PBS, BBB disruption extent and astrocyte demise were reduced in PJ34 mice only in the rostral brain at 48 h and 8 days post-pMCAo, respectively. Accordingly, 8 days after pMCAo, affected areas were reduced in the rostral brain (Bregma +0.86 and +0.14 mm), whereas total tissue loss was not reduced after PJ34 (4.0 ± 3.1%) vs. PBS (5.8 ± 3.4%). These results show that PJ34 reduced BBB permeability, astrocyte demise, and tissue loss (particularly in the rostral territories), suggesting that collateral supply mainly proceeds from the anterior ICA’s branches in the ischemic neonatal mouse brain.
Collapse
|
6
|
Koehler RC, Dawson VL, Dawson TM. Targeting Parthanatos in Ischemic Stroke. Front Neurol 2021; 12:662034. [PMID: 34025565 PMCID: PMC8131834 DOI: 10.3389/fneur.2021.662034] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/01/2021] [Indexed: 12/14/2022] Open
Abstract
Parthanatos is a cell death signaling pathway in which excessive oxidative damage to DNA leads to over-activation of poly(ADP-ribose) polymerase (PARP). PARP then generates the formation of large poly(ADP-ribose) polymers that induce the release of apoptosis-inducing factor from the outer mitochondrial membrane. In the cytosol, apoptosis-inducing factor forms a complex with macrophage migration inhibitory factor that translocates into the nucleus where it degrades DNA and produces cell death. In a review of the literature, we identified 24 publications from 13 laboratories that support a role for parthanatos in young male mice and rats subjected to transient and permanent middle cerebral artery occlusion (MCAO). Investigators base their conclusions on the use of nine different PARP inhibitors (19 studies) or PARP1-null mice (7 studies). Several studies indicate a therapeutic window of 4-6 h after MCAO. In young female rats, two studies using two different PARP inhibitors from two labs support a role for parthanatos, whereas two studies from one lab do not support a role in young female PARP1-null mice. In addition to parthanatos, a body of literature indicates that PARP inhibitors can reduce neuroinflammation by interfering with NF-κB transcription, suppressing matrix metaloproteinase-9 release, and limiting blood-brain barrier damage and hemorrhagic transformation. Overall, most of the literature strongly supports the scientific premise that a PARP inhibitor is neuroprotective, even when most did not report behavior outcomes or address the issue of randomization and treatment concealment. Several third-generation PARP inhibitors entered clinical oncology trials without major adverse effects and could be repurposed for stroke. Evaluation in aged animals or animals with comorbidities will be important before moving into clinical stroke trials.
Collapse
Affiliation(s)
- Raymond C Koehler
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University, Baltimore, MD, United States
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, The Institute of Cell Engineering, The Johns Hopkins University, Baltimore, MD, United States.,Department of Neurology, The Johns Hopkins University, Baltimore, MD, United States.,Department of Neuroscience, The Johns Hopkins University, Baltimore, MD, United States.,Department of Physiology, The Johns Hopkins University, Baltimore, MD, United States
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, The Institute of Cell Engineering, The Johns Hopkins University, Baltimore, MD, United States.,Department of Neurology, The Johns Hopkins University, Baltimore, MD, United States.,Department of Neuroscience, The Johns Hopkins University, Baltimore, MD, United States.,Department of Pharmacology and Molecular Sciences, The Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
7
|
Chen H, He Y, Chen S, Qi S, Shen J. Therapeutic targets of oxidative/nitrosative stress and neuroinflammation in ischemic stroke: Applications for natural product efficacy with omics and systemic biology. Pharmacol Res 2020; 158:104877. [PMID: 32407958 DOI: 10.1016/j.phrs.2020.104877] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/11/2022]
Abstract
Oxidative/nitrosative stress and neuroinflammation are critical pathological processes in cerebral ischemia-reperfusion injury, and their intimate interactions mediate neuronal damage, blood-brain barrier (BBB) damage and hemorrhagic transformation (HT) during ischemic stroke. We review current progress towards understanding the interactions of oxidative/nitrosative stress and inflammatory responses in ischemic brain injury. The interactions between reactive oxygen species (ROS)/reactive nitrogen species (RNS) and innate immune receptors such as TLR2/4, NOD-like receptor, RAGE, and scavenger receptors are crucial pathological mechanisms that amplify brain damage during cerebral ischemic injury. Furthermore, we review the current progress of omics and systematic biology approaches for studying complex network regulations related to oxidative/nitrosative stress and inflammation in the pathology of ischemic stroke. Targeting oxidative/nitrosative stress and neuroinflammation could be a promising therapeutic strategy for ischemic stroke treatment. We then review recent advances in discovering compounds from medicinal herbs with the bioactivities of simultaneously regulating oxidative/nitrosative stress and pro-inflammatory molecules for minimizing ischemic brain injury. These compounds include sesamin, baicalin, salvianolic acid A, 6-paradol, silymarin, apocynin, 3H-1,2-Dithiole-3-thione, (-)-epicatechin, rutin, Dl-3-N-butylphthalide, and naringin. We finally summarize recent developments of the omics and systematic biology approaches for exploring the molecular mechanisms and active compounds of Traditional Chinese Medicine (TCM) formulae with the properties of antioxidant and anti-inflammation for neuroprotection. The comprehensive omics and systematic biology approaches provide powerful tools for exploring therapeutic principles of TCM formulae and developing precision medicine for stroke treatment.
Collapse
Affiliation(s)
- Hansen Chen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong Special Administrative Region; The University of Hong Kong-Shenzhen Institute of Research and Innovation (HKU-SIRI), China
| | - Yacong He
- School of Chinese Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Shuang Chen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong Special Administrative Region
| | - Suhua Qi
- School of Medical Technology, Xuzhou Medical University, Xuzhou, 221002, China
| | - Jiangang Shen
- School of Chinese Medicine, The University of Hong Kong, Hong Kong Special Administrative Region; The University of Hong Kong-Shenzhen Institute of Research and Innovation (HKU-SIRI), China; School of Medical Technology, Xuzhou Medical University, Xuzhou, 221002, China.
| |
Collapse
|
8
|
Mangin G, Poittevin M, Charriaut-Marlangue C, Giannesini C, Merkoulova-Rainon T, Kubis N. Glatiramer acetate reduces infarct volume in diabetic mice with cerebral ischemia and prevents long-term memory loss. Brain Behav Immun 2019; 80:315-327. [PMID: 30953775 DOI: 10.1016/j.bbi.2019.04.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2018] [Revised: 02/18/2019] [Accepted: 04/02/2019] [Indexed: 12/15/2022] Open
Abstract
Stroke is currently the second leading cause of death in industrialized countries and the second cause of dementia after Alzheimer's disease. Diabetes is an independent risk factor for stroke that exacerbates the severity of lesions, disability and cognitive decline. There is increasing evidence that sustained brain inflammation may account for this long-term prejudicial outcome in diabetic patients in particular. We sought to demonstrate that experimental permanent middle cerebral artery occlusion (pMCAo) in the diabetic mouse aggravates stroke, induces cognitive decline, and is associated with exacerbated brain inflammation, and that these effects can be alleviated and/or prevented by the immunomodulator, glatiramer acetate (GA). Male diabetic C57Bl6 mice (streptozotocin IP) subjected to permanent middle cerebral artery occlusion (pMCAo), were treated by the immunomodulator, GA (Copaxone®) (1 mg/kg daily, sc) until 3 or 7 days post stroke. Infarct volume, brain pro- and anti-inflammatory mediators, microglial/macrophage density, and neurogenesis were monitored during the first week post stroke. Neurological sensorimotor deficit, spatial memory and brain deposits of Aβ40 and Aβ42 were assessed until six weeks post stroke. In diabetic mice with pMCAo, proinflammatory mediators (IL-1β, MCP1, TNFα and CD68) were significantly higher than in non-diabetic mice. In GA-treated mice, the infarct volume was reduced by 30% at D3 and by 40% at D7 post stroke (P < 0.05), sensorimotor recovery was accelerated as early as D3, and long-term memory loss was prevented. Moreover, proinflammatory mediators significantly decreased between D3 (COX2) and D7 (CD32, TNFα, IL-1β), and neurogenesis was significantly increased at D7. Moreover, GA abrogates the accumulation of insoluble Aβ40. This work is the first one to evidence that the immunomodulatory drug GA reduces infarct volume and proinflammatory mediators, enhances early neurogenesis, accelerates sensorimotor recovery, and prevents long-term memory loss in diabetic mice with pMCAo.
Collapse
Affiliation(s)
- Gabrielle Mangin
- Université Paris Diderot, Sorbonne Paris Cité & CART, INSERM U965, F-75475 Paris, France
| | - Marine Poittevin
- Université Paris Diderot, Sorbonne Paris Cité & CART, INSERM U965, F-75475 Paris, France
| | | | - Claire Giannesini
- Service de Neurologie, AP-HP, Hôpital Saint Antoine, 75012 Paris, France
| | | | - Nathalie Kubis
- Université Paris Diderot, Sorbonne Paris Cité & CART, INSERM U965, F-75475 Paris, France; Service de Physiologie Clinique, AP-HP, Hôpital Lariboisière, 75475 Paris, France.
| |
Collapse
|
9
|
Pétrault M, Ouk T, Pétrault O, Bastide M, Bordet R, Bérézowski V. Safety of oral anticoagulants on experimental brain microbleeding and cognition. Neuropharmacology 2019; 155:162-172. [PMID: 31132437 DOI: 10.1016/j.neuropharm.2019.05.030] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Revised: 05/22/2019] [Accepted: 05/23/2019] [Indexed: 12/24/2022]
Abstract
This study aims at determining the ability of clinical-based doses of four oral anticoagulants to transform the onset of a cerebral microhemorrhages (CMH) burden into a symptomatic intracerebral hemorrhage (ICH) in the healthy brain, and precipitate cognitive impairment. Wild-type mice were anticoagulated for 10 days using apixaban, rivaroxaban or dabigatran as direct oral anticoagulants (DOACs), or warfarin as vitamin K-antagonist. Meanwhile, a burden of ∼20 CMHs was induced in the Sylvian territory by intra-carotid injection of cyclodextrin nanoparticles. At bleeding onset, only warfarin provoked deadly hematoma, and dramatically increased mortality (+45%). All the DOACs enhanced CMH burden through a greater number of intermediate-sized microhemorrhages (+80% to +180%). Although silent at onset, both baseline- and anticoagulant-enhanced CMH burdens increased mortality (+11% to +58%) along the following year without statistical difference among groups, and despite cessation of anticoagulation and absence of CMH progression or transformation into ICH. All survivor mice exhibited reduction in visual recognition memory from 9 months. In the healthy brain, DOACs preserve the onset of microhemorrhages from transformation into ICH, and do not precipitate cognitive impairment despite enhancement of CMH burden. High CMH burdens should however be considered for early detection and preventive memory care apart from anticoagulation decisions.
Collapse
Affiliation(s)
- Maud Pétrault
- Univ. Lille, Inserm, CHU Lille, U1171, Degenerative and Vascular Cognitive Disorders, F-59000, Lille, France
| | - Thavarak Ouk
- Univ. Lille, Inserm, CHU Lille, U1171, Degenerative and Vascular Cognitive Disorders, F-59000, Lille, France
| | - Olivier Pétrault
- Univ. Lille, Inserm, CHU Lille, U1171, Degenerative and Vascular Cognitive Disorders, F-59000, Lille, France; UArtois, F-62300, Lens, France
| | - Michèle Bastide
- Univ. Lille, Inserm, CHU Lille, U1171, Degenerative and Vascular Cognitive Disorders, F-59000, Lille, France
| | - Régis Bordet
- Univ. Lille, Inserm, CHU Lille, U1171, Degenerative and Vascular Cognitive Disorders, F-59000, Lille, France
| | - Vincent Bérézowski
- Univ. Lille, Inserm, CHU Lille, U1171, Degenerative and Vascular Cognitive Disorders, F-59000, Lille, France; UArtois, F-62300, Lens, France.
| |
Collapse
|
10
|
Fréchou M, Margaill I, Marchand-Leroux C, Beray-Berthat V. Behavioral tests that reveal long-term deficits after permanent focal cerebral ischemia in mouse. Behav Brain Res 2018; 360:69-80. [PMID: 30500429 DOI: 10.1016/j.bbr.2018.11.040] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 11/26/2018] [Accepted: 11/26/2018] [Indexed: 01/10/2023]
Abstract
Efforts are still needed regarding the research of therapeutics for ischemic stroke. While in experimental studies the protective effect of pharmacological agents is often highlighted by a reduction of the lesion size evaluated in the short term (days), in clinical studies a functional recovery of patients suffering from stroke is expected on the long-term (months and years). Long-term functional preclinical studies are highly recommended to evaluate potential neuroprotective agents for stroke, rather than an assessment of the infarction size at a short time point. The present study thus aimed to select among various behavioral tests those able to highlight long-term deficits (3 months) after cerebral ischemia in mice. Permanent focal cerebral ischemia was carried out in male Swiss mice by intraluminal occlusion of the left middle cerebral artery (MCA). Fourteen behavioral tests were assessed from 7 days to 90 days after ischemia (locomotor activity, neurological score, exit circle test, grip and string tests, chimney test, adhesive removal test, pole test, beam-walking tests, elevated plus maze, marble burying test, forced swimming test, novel object recognition test). The present study clearly identified a battery of behavioral tests able to highlight deficits up to 3 months in our mouse model of permanent MCA occlusion (locomotor activity, neurological score, adhesive removal test, pole test, beam-walking tests, elevated plus maze, marble burying test, forced swimming test and novel object recognition test). This battery of behavioral tests highlighting long-term deficits is useful to study future neuroprotective strategies for stroke treatment.
Collapse
Affiliation(s)
- Magalie Fréchou
- Equipe de recherche "Pharmacologie de la Circulation Cérébrale" EA 4475, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Pharmacie de Paris, 75006 Paris, France.
| | - Isabelle Margaill
- Equipe de recherche "Pharmacologie de la Circulation Cérébrale" EA 4475, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Pharmacie de Paris, 75006 Paris, France.
| | - Catherine Marchand-Leroux
- Equipe de recherche "Pharmacologie de la Circulation Cérébrale" EA 4475, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Pharmacie de Paris, 75006 Paris, France.
| | - Virginie Beray-Berthat
- Equipe de recherche "Pharmacologie de la Circulation Cérébrale" EA 4475, Université Paris Descartes, Sorbonne Paris Cité, Faculté de Pharmacie de Paris, 75006 Paris, France.
| |
Collapse
|
11
|
Chen H, Chen X, Luo Y, Shen J. Potential molecular targets of peroxynitrite in mediating blood–brain barrier damage and haemorrhagic transformation in acute ischaemic stroke with delayed tissue plasminogen activator treatment. Free Radic Res 2018; 52:1220-1239. [PMID: 30468092 DOI: 10.1080/10715762.2018.1521519] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Hansen Chen
- School of Chinese Medicine, the University of Hong Kong, PR China
- Shenzhen Institute of Research and Innovation (HKU-SIRI), University of Hong Kong, Hong Kong, PR China
| | - Xi Chen
- Department of Core Facility, the People’s Hospital of Bao-an Shenzhen, Shenzhen, PR China
- The 8th People’s Hospital of Shenzhen, the Affiliated Bao-an Hospital of Southern Medical University, Shenzhen, PR China
| | - Yunhao Luo
- School of Chinese Medicine, the University of Hong Kong, PR China
| | - Jiangang Shen
- School of Chinese Medicine, the University of Hong Kong, PR China
- Shenzhen Institute of Research and Innovation (HKU-SIRI), University of Hong Kong, Hong Kong, PR China
| |
Collapse
|
12
|
Noh MY, Lee WM, Lee SJ, Kim HY, Kim SH, Kim YS. Regulatory T cells increase after treatment with poly (ADP-ribose) polymerase-1 inhibitor in ischemic stroke patients. Int Immunopharmacol 2018; 60:104-110. [PMID: 29709770 DOI: 10.1016/j.intimp.2018.04.043] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 04/04/2018] [Accepted: 04/24/2018] [Indexed: 01/26/2023]
Abstract
BACKGROUND Regulatory T cells (Tregs) are thought to play a modulatory role in immune responses and to improve outcomes after ischemic stroke. Thus, various strategies for increasing Tregs in animal models of ischemic stroke have yielded successful results. The aim of this study was to examine the potential effect of poly (ADP-ribose) polymerase-1 (PARP-1) inhibitor on Treg proportion in stroke patients. METHODS Peripheral blood samples were collected from 12 ischemic stroke patients (within 72 h of stroke onset) and 5 healthy control subjects. Flow cytometry analyses and quantitative reverse transcription polymerase chain reactions (qRT-PCR) were performed on peripheral blood mononuclear cells (PBMCs) before and after treating them with PARP-1 inhibitor (3-AB; JPI-289 1 μm, JPI-289 10 μm) for 24 h. RESULTS Treg proportions were significantly higher in healthy controls (median 2.8%, IQR 2.6-5.0%) than ischemic stroke patients (median 1.6%, IQR 1.25-2.2%) (p < 0.001). In the latter, Treg proportions were positively correlated with age (r = 0.595, p = 0.041), but not with infarct volume (r = 0.367, p = 0.241). After PARP-1 inhibitor treatment, Treg proportions among PBMCs increased in response to high dose (10 μm) JPI-289 (median 2.3%, IQR 2.0-2.9%) as did Treg-associated transcription factors such as FoxP3 and CTLA-4 mRNA. PARP-1 inhibitor treatment also decreased pro-inflammatory cytokines (IFN-γ, TNF-α, and IL-17) and increased anti-inflammatory cytokines (IL-4, IL-10, and TGF-β1). CONCLUSION Treg proportions are reduced in ischemic stroke patients and increased by treatment with high-dose PARP-1 inhibitor JPI-289. The PARP-1 inhibitor also had a possible anti-inflammatory effect on cytokine levels, and may ameliorate the outcome of ischemic stroke by up-regulating Tregs.
Collapse
Affiliation(s)
- Min-Young Noh
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Won Moo Lee
- Department of Obstetrics and Gynecology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Su-Jung Lee
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Hyun Young Kim
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Seung Hyun Kim
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea
| | - Young Seo Kim
- Department of Neurology, College of Medicine, Hanyang University, Seoul, Republic of Korea.
| |
Collapse
|
13
|
El Amki M, Lerouet D, Garraud M, Teng F, Beray-Berthat V, Coqueran B, Barsacq B, Abbou C, Palmier B, Marchand-Leroux C, Margaill I. Improved Reperfusion and Vasculoprotection by the Poly(ADP-Ribose)Polymerase Inhibitor PJ34 After Stroke and Thrombolysis in Mice. Mol Neurobiol 2018; 55:9156-9168. [PMID: 29651748 DOI: 10.1007/s12035-018-1063-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 04/03/2018] [Indexed: 10/17/2022]
Abstract
Benefits from thrombolysis with recombinant tissue plasminogen activator (rt-PA) after ischemic stroke remain limited due to a narrow therapeutic window, low reperfusion rates, and increased risk of hemorrhagic transformations (HT). Experimental data showed that rt-PA enhances the post-ischemic activation of poly(ADP-ribose)polymerase (PARP) which in turn contributes to blood-brain barrier injury. The aim of the present study was to evaluate whether PJ34, a potent PARP inhibitor, improves poor reperfusion induced by delayed rt-PA administration, exerts vasculoprotective effects, and finally increases the therapeutic window of rt-PA. Stroke was induced by thrombin injection (0.75 UI in 1 μl) in the left middle cerebral artery (MCA) of male Swiss mice. Administration of rt-PA (0.9 mg kg-1) or saline was delayed for 4 h after ischemia onset. Saline or PJ34 (3 mg kg-1) was given intraperitoneally twice, just after thrombin injection and 3 h later, or once, 3 h after ischemia onset. Reperfusion was evaluated by laser Doppler, vascular inflammation by immunohistochemistry of vascular cell adhesion molecule-1 (VCAM-1) expression, and vasospasm by morphometric measurement of the MCA. Edema, cortical lesion, and sensorimotor deficit were evaluated. Treatment with PJ34 improved rt-PA-induced reperfusion and promoted vascular protection including reduction in vascular inflammation (decrease in VCAM-1 expression), HT, and MCA vasospasm. Additionally, the combined treatment significantly reduced brain edema, cortical lesion, and sensorimotor deficit. In conclusion, the combination of the PARP inhibitor PJ34 with rt-PA after cerebral ischemia may be of particular interest in order to improve thrombolysis with an extended therapeutic window.
Collapse
Affiliation(s)
- Mohamad El Amki
- EA4475 - "Pharmacologie de la Circulation Cérébrale", Faculté de Pharmacie de Paris, Université Paris Descartes, Université Sorbonne Paris Cité, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Dominique Lerouet
- EA4475 - "Pharmacologie de la Circulation Cérébrale", Faculté de Pharmacie de Paris, Université Paris Descartes, Université Sorbonne Paris Cité, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Marie Garraud
- EA4475 - "Pharmacologie de la Circulation Cérébrale", Faculté de Pharmacie de Paris, Université Paris Descartes, Université Sorbonne Paris Cité, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Fei Teng
- EA4475 - "Pharmacologie de la Circulation Cérébrale", Faculté de Pharmacie de Paris, Université Paris Descartes, Université Sorbonne Paris Cité, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Virginie Beray-Berthat
- EA4475 - "Pharmacologie de la Circulation Cérébrale", Faculté de Pharmacie de Paris, Université Paris Descartes, Université Sorbonne Paris Cité, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Bérard Coqueran
- EA4475 - "Pharmacologie de la Circulation Cérébrale", Faculté de Pharmacie de Paris, Université Paris Descartes, Université Sorbonne Paris Cité, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Benoît Barsacq
- EA4475 - "Pharmacologie de la Circulation Cérébrale", Faculté de Pharmacie de Paris, Université Paris Descartes, Université Sorbonne Paris Cité, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Charlotte Abbou
- EA4475 - "Pharmacologie de la Circulation Cérébrale", Faculté de Pharmacie de Paris, Université Paris Descartes, Université Sorbonne Paris Cité, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Bruno Palmier
- EA4475 - "Pharmacologie de la Circulation Cérébrale", Faculté de Pharmacie de Paris, Université Paris Descartes, Université Sorbonne Paris Cité, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Catherine Marchand-Leroux
- EA4475 - "Pharmacologie de la Circulation Cérébrale", Faculté de Pharmacie de Paris, Université Paris Descartes, Université Sorbonne Paris Cité, 4 avenue de l'Observatoire, 75006, Paris, France
| | - Isabelle Margaill
- EA4475 - "Pharmacologie de la Circulation Cérébrale", Faculté de Pharmacie de Paris, Université Paris Descartes, Université Sorbonne Paris Cité, 4 avenue de l'Observatoire, 75006, Paris, France.
| |
Collapse
|
14
|
Matrix Metalloproteinase-9 and Recovery of Acute Ischemic Stroke. J Stroke Cerebrovasc Dis 2017; 26:733-740. [PMID: 28063771 DOI: 10.1016/j.jstrokecerebrovasdis.2016.09.043] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 08/17/2016] [Accepted: 09/24/2016] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Stroke outcome can be predicted by clinical features, biochemical parameters, and some risk factors. Matrix metalloproteinase-9 (MMP-9) is involved in various stages of stroke pathology. MMP-9 inhibitors are potential stroke therapeutic agents. Little is known about the relation between MMP-9-after the acute stage-and clinical recovery. OBJECTIVE The study aimed to investigate the serum level of MMP-9 at stroke onset as predictor of stroke outcome and the relation between the level of MMP-9 after 30 days and stroke recovery. METHODS The National Institutes of Health Stroke Scale, modified Rankin Scale, and serum level of MMP-9 were assessed in 30 patients with acute ischemic stroke during the first 24 hours of onset and then a month later. None of the patients received thrombolytic therapy. Thirty normal volunteers of matched age and sex were included in the control group. RESULTS The serum level of MMP-9 at stroke onset was independently positively correlated with stroke outcome. The serum level of MMP-9 30 days after stroke onset was positively correlated with initial stroke severity and outcome, as well as with clinical recovery. CONCLUSION Higher serum level of MMP-9 at stroke onset can be a predictor of poor stroke outcome. However, beyond the acute stage, MMP-9 may play beneficial role in stroke recovery.
Collapse
|
15
|
Zhang S, Kojic L, Tsang M, Grewal P, Liu J, Namjoshi D, Wellington CL, Tetzlaff W, Cynader MS, Jia W. Distinct roles for metalloproteinases during traumatic brain injury. Neurochem Int 2016; 96:46-55. [PMID: 26939762 DOI: 10.1016/j.neuint.2016.02.013] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Revised: 02/16/2016] [Accepted: 02/25/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND Significant protease activations have been reported after traumatic brain injury (TBI). These proteases are responsible for cleavage of transmembrane proteins in neurons, glial, and endothelial cells and this results in the release of their extracellular domains (ectodomains). METHODS Two TBI models were employed here, representing both closed head injury (CHI) and open head injury (OHI). In situ zymography, immunohistochemistry, bright field and confocal microscopy, quantification of immunopositive cells and statistical analysis were applied. RESULTS We found, using in situ zymography, that gelatinase activity of matrix metalloproteinases (MMP)-2 and MMP-9 was upregulated in cortex of both injury models. Using immunohistochemistry for several MPPs (Matrix metalloproteinases) and ADAMs (disintegrin and metalloproteinases), including MMP-2, -9, ADAM-10, -17, distinct patterns of induction were observed in the two TBI models. In closed head injury, an early increase in protein expression of MMP-2, -9 and ADAM-17 was found as early as 10 min post injury in cortex and peaked at 1 h for all 4 proteases examined. In contrast, after OHI the maximal expression was observed locally neighboring the impact site, at a later time-point, as long as 24 h after the injury for MMP-2 and MMP-9. Confocal microscopy revealed colocalization of the 4 proteases with the neuronal marker NeuN in CHI, but only MMP2 colocalized with NeuN in OHI. CONCLUSIONS The findings may lead to a trauma-induced therapeutic strategy triggered soon after a primary insult to improve survival and to reduce brain damage following TBI.
Collapse
Affiliation(s)
- Si Zhang
- Brain Research Center, University of British Columbia, Vancouver, BC, Canada.
| | - Luba Kojic
- Brain Research Center, University of British Columbia, Vancouver, BC, Canada.
| | - Michelle Tsang
- Brain Research Center, University of British Columbia, Vancouver, BC, Canada.
| | - Parampal Grewal
- Brain Research Center, University of British Columbia, Vancouver, BC, Canada.
| | - Jie Liu
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada.
| | - Dhananjay Namjoshi
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.
| | - Cheryl L Wellington
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.
| | - Wolfram Tetzlaff
- International Collaboration on Repair Discoveries (ICORD), University of British Columbia, Vancouver, BC, Canada.
| | - Max S Cynader
- Brain Research Center, University of British Columbia, Vancouver, BC, Canada.
| | - William Jia
- Brain Research Center, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
16
|
Ingberg E, Dock H, Theodorsson E, Theodorsson A, Ström JO. Method parameters' impact on mortality and variability in mouse stroke experiments: a meta-analysis. Sci Rep 2016; 6:21086. [PMID: 26876353 PMCID: PMC4753409 DOI: 10.1038/srep21086] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 01/13/2016] [Indexed: 12/17/2022] Open
Abstract
Although hundreds of promising substances have been tested in clinical trials,
thrombolysis currently remains the only specific pharmacological treatment for
ischemic stroke. Poor quality, e.g. low statistical power, in the preclinical
studies has been suggested to play an important role in these failures. Therefore,
it would be attractive to use animal models optimized to minimize unnecessary
mortality and outcome variability, or at least to be able to power studies more
exactly by predicting variability and mortality given a certain experimental setup.
The possible combinations of methodological parameters are innumerous, and an
experimental comparison of them all is therefore not feasible. As an alternative
approach, we extracted data from 334 experimental mouse stroke articles and, using a
hypothesis-driven meta-analysis, investigated the method parameters’
impact on infarct size variability and mortality. The use of Swiss and C57BL6 mice
as well as permanent occlusion of the middle cerebral artery rendered the lowest
variability of the infarct size while the emboli methods increased variability. The
use of Swiss mice increased mortality. Our study offers guidance for researchers
striving to optimize mouse stroke models.
Collapse
Affiliation(s)
- Edvin Ingberg
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden
| | - Hua Dock
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden
| | - Elvar Theodorsson
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden
| | - Annette Theodorsson
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden.,Division of Neuro and Inflammation Science, Department of Clinical and Experimental Medicine, Linköping University, Department of Neurosurgery, Anaesthetics, Operations and Specialty Surgery Center, Region Östergötland, Sweden
| | - Jakob O Ström
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden.,Vårdvetenskapligt Forskningscentrum/Centre for Health Sciences, Örebro University Hospital, County Council of Örebro, Örebro, Sweden.,School of Health and Medical Sciences, Örebro University, Örebro, Sweden
| |
Collapse
|
17
|
Teng F, Zhu L, Su J, Zhang X, Li N, Nie Z, Jin L. Neuroprotective Effects of Poly(ADP-ribose)polymerase Inhibitor Olaparib in Transient Cerebral Ischemia. Neurochem Res 2016; 41:1516-26. [PMID: 26869042 DOI: 10.1007/s11064-016-1864-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2015] [Revised: 01/30/2016] [Accepted: 02/04/2016] [Indexed: 10/22/2022]
Abstract
Olaparib was the first poly(ADP-ribose)polymerase inhibitor approved by Food and Drug Administration for oncology treatment. However, its neuroprotective effects have not been elucidated. This study aimed to evaluate the effects of olaparib in transient cerebral ischemia. A mouse model of transient middle cerebral artery occlusion was used. Reperfusion was performed at 2 h after ischemia. Different doses of olaparib (1, 3, 5, 10 and 25 mg/kg) were administered intraperitoneally immediately after reperfusion. Twenty-four hours after ischemia, the neurological score was assessed, and grip and string tests were performed to evaluate the behavioral deficits in the mice. Cresyl violet staining was used to assess cerebral edema and the lesion volume. Immunohistochemistry was performed to evaluate the expression of blood-brain barrier proteins collagen IV and claudin-5, as well as extravasation of IgG. Ischemia induced a neurological deficit, which was significantly ameliorated by olaparib at 3 and 5 mg/kg. However, this neuroprotective effect was not observed in mice treated with either low-dose or high-dose olaparib. Both 3 and 5 mg/kg olaparib markedly reduced cerebral infarction volume, but not cerebral edema. The expression of collagen IV decreased after cerebral ischemia, which was improved by olaparib at 3 and 5 mg/kg. These results were confirmed by the reduction of IgG extravasation with olaparib. Olaparib showed clear neuroprotective effects in transient ischemic mice mainly through the reduction of cerebral infarction and blood-brain barrier damage.
Collapse
Affiliation(s)
- Fei Teng
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, 389, Xincun Road, Shanghai, 200065, China
| | - Ling Zhu
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, 389, Xincun Road, Shanghai, 200065, China.,Department of Neurology, Shanghai Liqun Hospital, Shanghai, China
| | - Junhui Su
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, 389, Xincun Road, Shanghai, 200065, China
| | - Xi Zhang
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, 389, Xincun Road, Shanghai, 200065, China
| | - Ning Li
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, 389, Xincun Road, Shanghai, 200065, China
| | - Zhiyu Nie
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, 389, Xincun Road, Shanghai, 200065, China
| | - Lingjing Jin
- Department of Neurology, Shanghai Tongji Hospital, Tongji University School of Medicine, 389, Xincun Road, Shanghai, 200065, China.
| |
Collapse
|
18
|
Poittevin M, Bonnin P, Pimpie C, Rivière L, Sebrié C, Dohan A, Pocard M, Charriaut-Marlangue C, Kubis N. Diabetic microangiopathy: impact of impaired cerebral vasoreactivity and delayed angiogenesis after permanent middle cerebral artery occlusion on stroke damage and cerebral repair in mice. Diabetes 2015; 64:999-1010. [PMID: 25288671 DOI: 10.2337/db14-0759] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Diabetes increases the risk of stroke by three, increases related mortality, and delays recovery. We aimed to characterize functional and structural alterations in cerebral microvasculature before and after experimental cerebral ischemia in a mouse model of type 1 diabetes. We hypothesized that preexisting brain microvascular disease in patients with diabetes might partly explain increased stroke severity and impact on outcome. Diabetes was induced in 4-week-old C57Bl/6J mice by intraperitoneal injections of streptozotocin (60 mg/kg). After 8 weeks of diabetes, the vasoreactivity of the neurovascular network to CO2 was abolished and was not reversed by nitric oxide (NO) donor administration; endothelial NO synthase (eNOS) and neuronal NO synthase (nNOS) mRNA, phospho-eNOS protein, nNOS, and phospho-nNOS protein were significantly decreased; angiogenic and vessel maturation factors (vascular endothelial growth factor a [VEGFa], angiopoietin 1 (Ang1), Ang2, transforming growth factor-β [TGF-β], and platelet-derived growth factor-β [PDGF-β]) and blood-brain barrier (BBB) occludin and zona occludens 1 (ZO-1) expression were significantly decreased; and microvessel density was increased without changes in ultrastructural imaging. After permanent focal cerebral ischemia induction, infarct volume and neurological deficit were significantly increased at D1 and D7, and neuronal death (TUNEL+ / NeuN+ cells) and BBB permeability (extravasation of Evans blue) at D1. At D7, CD31+ / Ki67+ double-immunolabeled cells and VEGFa and Ang2 expression were significantly increased, indicating delayed angiogenesis. We show that cerebral microangiopathy thus partly explains stroke severity in diabetes.
Collapse
Affiliation(s)
- Marine Poittevin
- Université Paris Diderot, Sorbonne Paris Cité, Angiogenesis and Translational Research Center, INSERM U965, Paris, France
| | - Philippe Bonnin
- Université Paris Diderot, Sorbonne Paris Cité, Angiogenesis and Translational Research Center, INSERM U965, Paris, France Service de Physiologie Clinique, AP-HP, Hôpital Lariboisière, Paris, France
| | - Cynthia Pimpie
- Université Paris Diderot, Sorbonne Paris Cité, Angiogenesis and Translational Research Center, INSERM U965, Paris, France
| | - Léa Rivière
- Université Paris Diderot, Sorbonne Paris Cité, Angiogenesis and Translational Research Center, INSERM U965, Paris, France
| | | | - Anthony Dohan
- Université Paris Diderot, Sorbonne Paris Cité, Angiogenesis and Translational Research Center, INSERM U965, Paris, France
| | - Marc Pocard
- Université Paris Diderot, Sorbonne Paris Cité, Angiogenesis and Translational Research Center, INSERM U965, Paris, France
| | | | - Nathalie Kubis
- Université Paris Diderot, Sorbonne Paris Cité, Angiogenesis and Translational Research Center, INSERM U965, Paris, France Service de Physiologie Clinique, AP-HP, Hôpital Lariboisière, Paris, France
| |
Collapse
|
19
|
Protective actions of PJ34, a poly(ADP-ribose)polymerase inhibitor, on the blood-brain barrier after traumatic brain injury in mice. Neuroscience 2015; 291:26-36. [PMID: 25668593 DOI: 10.1016/j.neuroscience.2015.01.070] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 01/30/2015] [Accepted: 01/30/2015] [Indexed: 01/12/2023]
Abstract
Poly(ADP-ribose) polymerase (PARP) is activated by oxidative stress and plays an important role in traumatic brain injury (TBI). The objective of this study was to investigate whether PARP activation participated in the blood-brain barrier (BBB) disruption and edema formation in a mouse model of controlled cortical impact (CCI). N-(6-oxo-5,6-dihydrophenanthridin-2-yl)-N,N-dimethylacetamide (PJ34) (10 mg/kg), a selective PARP inhibitor, was administered intraperitoneally at 5 min and 8 h after experimental CCI. After 6 h and 24 h of CCI, the permeability of the cortical BBB was determined after Evans Blue administration. The water content of the brain was also measured. Treatment with PJ34 markedly attenuated the permeability of the BBB and decreased the brain edema at 6 h and 24 h after CCI. Our data showed the up-regulation of nuclear factor-κB in cytosolic fractions and nuclear fractions in the injured cortex, and these changes were reversed by PJ34. Moreover, PJ34 significantly lessened the activities of myeloperoxidase and the levels of matrix metalloproteinase-9, enhanced the levels of occludin, laminin, collagen IV and integrin β1, reduced neurological deficits, decreased the contusion volume, and attenuated the necrotic and apoptotic neuronal cell death. These data suggest the protective effects of PJ34 on BBB integrity and cell death during acute TBI.
Collapse
|
20
|
The poly(adenosine diphosphate-ribose) polymerase inhibitor PJ34 reduces pulmonary ischemia-reperfusion injury in rats. Transplantation 2015; 98:618-24. [PMID: 25221900 PMCID: PMC4160283 DOI: 10.1097/tp.0000000000000305] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Supplemental digital content is available in the text. Background Ischemia-reperfusion (I/R) injury after lung transplantation causes alveolar damage, lung edema, and acute rejection. Poly(adenosine diphosphate-ribose) polymerase (PARP) is a single-stranded DNA repair enzyme that induces apoptosis and necrosis after DNA damage caused by reactive oxygen species. We evaluated tissue protective effects of the PARP inhibitor (PARP-i) PJ34 against pulmonary I/R injury. Methods Rats (total n=45) underwent a thoracotomy with left hilar isolation and saline administration (sham group) or thoracotomy with hilar clamping and saline administration (I/R group) or PJ34 administration (PARP-i group). Parameters were measured for 7 days after reperfusion. Results Pathologic analysis revealed that reperfusion injury was drastically suppressed in the PARP-i group 2 days after reperfusion. Terminal deoxynucleotide transferase-mediated deoxyuridine triphosphate nick-end labeling–positive cells were significantly decreased in the PARP-i group compared to the I/R group (P<0.05). Accordingly, the wet-to-dry lung ratio in the I/R group was significantly higher compared with the PARP-i group (P=0.025). Four hours after reperfusion, serum tissue necrosis factor-α and interleukin-6 were significantly suppressed in the PARP-i group compared with the I/R group (P<0.05). Serum derivatives of reactive oxygen metabolites increased quickly and remained high in the I/R and PARP-i groups from 4 hr until 7 days after reperfusion. Interestingly, the serum biologic antioxidant potential in the PARP-i group was significantly higher than that in the I/R group from day 2 until day 7. Conclusion The PARP-i decreased inflammation and tissue damage caused by pulmonary I/R injury. These beneficial effects of the PARP-i may be correlated with its antioxidative efficacy.
Collapse
|
21
|
Lechaftois M, Dreano E, Palmier B, Margaill I, Marchand-Leroux C, Bachelot-Loza C, Lerouet D. Another "string to the bow" of PJ34, a potent poly(ADP-Ribose)polymerase inhibitor: an antiplatelet effect through P2Y12 antagonism? PLoS One 2014; 9:e110776. [PMID: 25329809 PMCID: PMC4203827 DOI: 10.1371/journal.pone.0110776] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 09/16/2014] [Indexed: 11/25/2022] Open
Abstract
Background Neuro- and vasoprotective effects of poly(ADP-ribose)polymerase (PARP) inhibition have been largely documented in models of cerebral ischemia, particularly with the potent PARP inhibitor PJ34. Furthermore, after ischemic stroke, physicians are faced with incomplete tissue reperfusion and reocclusion, in which platelet activation/aggregation plays a key role. Data suggest that certain PARP inhibitors could act as antiplatelet agents. In that context, the present in vitro study investigated on human blood the potential antiplatelet effect of PJ34 and two structurally different PARP inhibitors, DPQ and INO-1001. Methods and results ADP concentrations were chosen to induce a biphasic aggregation curve resulting from the successive activation of both its receptors P2Y1 and P2Y12. In these experimental conditions, PJ34 inhibited the second phase of aggregation; this effect was reduced by incremental ADP concentrations. In addition, in line with a P2Y12 pathway inhibitory effect, PJ34 inhibited the dephosphorylation of the vasodilator stimulated phosphoprotein (VASP) in a concentration-dependent manner. Besides, PJ34 had no effect on platelet aggregation induced by collagen or PAR1 activating peptide, used at concentrations inducing a strong activation independent on secreted ADP. By contrast, DPQ and INO-1001 were devoid of any effect whatever the platelet agonist used. Conclusions We showed that, in addition to its already demonstrated beneficial effects in in vivo models of cerebral ischemia, the potent PARP inhibitor PJ34 exerts in vitro an antiplatelet effect. Moreover, this is the first study to report that PJ34 could act via a competitive P2Y12 antagonism. Thus, this antiplatelet effect could improve post-stroke reperfusion and/or prevent reocclusion, which reinforces the interest of this drug for stroke treatment.
Collapse
Affiliation(s)
- Marie Lechaftois
- EA4475-“Pharmacologie de la Circulation Cérébrale”, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Comue Sorbonne Paris Cité, Paris, France
| | - Elise Dreano
- Inserm UMR S1140, Paris, France
- Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Comue Sorbonne Paris Cité, Paris, France
| | - Bruno Palmier
- EA4475-“Pharmacologie de la Circulation Cérébrale”, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Comue Sorbonne Paris Cité, Paris, France
| | - Isabelle Margaill
- EA4475-“Pharmacologie de la Circulation Cérébrale”, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Comue Sorbonne Paris Cité, Paris, France
| | - Catherine Marchand-Leroux
- EA4475-“Pharmacologie de la Circulation Cérébrale”, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Comue Sorbonne Paris Cité, Paris, France
| | - Christilla Bachelot-Loza
- Inserm UMR S1140, Paris, France
- Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Comue Sorbonne Paris Cité, Paris, France
| | - Dominique Lerouet
- EA4475-“Pharmacologie de la Circulation Cérébrale”, Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Comue Sorbonne Paris Cité, Paris, France
- * E-mail:
| |
Collapse
|
22
|
Greco R, Tassorelli C, Mangione AS, Levandis G, Certo M, Nappi G, Bagetta G, Blandini F, Amantea D. Neuroprotection by the PARP inhibitor PJ34 modulates cerebral and circulating RAGE levels in rats exposed to focal brain ischemia. Eur J Pharmacol 2014; 744:91-7. [PMID: 25446913 DOI: 10.1016/j.ejphar.2014.10.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 10/03/2014] [Accepted: 10/06/2014] [Indexed: 12/30/2022]
Abstract
The receptor for advanced glycation end products (RAGE) has a potential role as a damage-sensing molecule; however, to date, its involvement in the pathophysiology of stroke and its modulation following neuroprotective treatment are not completely understood. We have previously demonstrated that expression of distinct RAGE isoforms, recognized by different antibodies, is differentially modulated in the brain of rats subjected to focal cerebral ischemia. Here, we focus on the full-length membrane-bound RAGE isoform, showing that its expression is significantly elevated in the striatum, whereas it is reduced in the cortex of rats subjected to transient middle cerebral artery occlusion (MCAo). Notably, the reduction of cortical levels of full-length RAGE detected 24 h after reperfusion is abolished by systemic administration of a neuroprotective dose of the poly(ADP-ribose) polymerase (PARP) inhibitor, N-(6-oxo-5,6-dihydrophenanthridin-2-yl)-N,N-dimethylacetamide (PJ34). More interestingly, a significant reduction of plasma soluble RAGE (sRAGE) occurs 24 h after reperfusion and this effect is reverted by a neuroprotective dose of PJ34. Soluble forms of RAGE, generated either by alternative splicing or by proteolysis of the full-length form, effectively bind advanced glycation end products, thereby competing with the cell surface full-length RAGE, thus providing a 'decoy' function that may counteract the adverse effects of receptor signaling in neurons and may possibly exert cytoprotective effects. Thus, our data confirm the important role of RAGE in ischemic cerebral damage and, more interestingly, suggest the potential use of sRAGE as a blood biomarker of stroke severity and of neuroprotective treatment efficacy.
Collapse
Affiliation(s)
- Rosaria Greco
- Laboratory of Neurophysiology of Integrative Autonomic Systems, Headache Science Centre, "C. Mondino" National Neurological Institute, Pavia, Italy.
| | - Cristina Tassorelli
- Laboratory of Neurophysiology of Integrative Autonomic Systems, Headache Science Centre, "C. Mondino" National Neurological Institute, Pavia, Italy; Department of Brain and Behavior, University of Pavia, Italy
| | - Antonina Stefania Mangione
- Laboratory of Neurophysiology of Integrative Autonomic Systems, Headache Science Centre, "C. Mondino" National Neurological Institute, Pavia, Italy
| | - Giovanna Levandis
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, "C. Mondino" National Neurological Institute, Pavia, Italy
| | - Michelangelo Certo
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende (CS), Italy
| | - Giuseppe Nappi
- Laboratory of Neurophysiology of Integrative Autonomic Systems, Headache Science Centre, "C. Mondino" National Neurological Institute, Pavia, Italy
| | - Giacinto Bagetta
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende (CS), Italy
| | - Fabio Blandini
- Laboratory of Functional Neurochemistry, Center for Research in Neurodegenerative Diseases, "C. Mondino" National Neurological Institute, Pavia, Italy
| | - Diana Amantea
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende (CS), Italy
| |
Collapse
|
23
|
Fagan SC, Lapchak PA, Liebeskind DS, Ishrat T, Ergul A. Recommendations for preclinical research in hemorrhagic transformation. Transl Stroke Res 2014; 4:322-7. [PMID: 23730351 DOI: 10.1007/s12975-012-0222-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Hemorrhagic transformation (HT) is an important complication of ischemic stroke and is responsible for most of the mortality associated with acute reperfusion therapy. Although many important publications address the preclinical models of ischemic stroke, there are no current recommendations on the conduct of research aimed at understanding the mechanisms and consequences of HT. The purpose of this review is to present the various models used in HT research, the clinical correlates, and the experimental variables known to influence the quantitation of HT in preclinical investigation. Lastly, recommendations for the conduct of preclinical research in HT are provided.
Collapse
Affiliation(s)
- Susan C Fagan
- Program in Clinical and Experimental Therapeutics, University of Georgia College of Pharmacy, Augusta, GA ; Charlie Norwood VA Medical Center, Augusta, GA ; Department of Neurology, Medical College of Georgia, Augusta, GA
| | | | | | | | | |
Collapse
|
24
|
Sriram CS, Jangra A, Kasala ER, Bodduluru LN, Bezbaruah BK. Targeting poly(ADP-ribose)polymerase1 in neurological diseases: A promising trove for new pharmacological interventions to enter clinical translation. Neurochem Int 2014; 76:70-81. [PMID: 25049175 DOI: 10.1016/j.neuint.2014.07.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2014] [Revised: 07/02/2014] [Accepted: 07/04/2014] [Indexed: 12/22/2022]
Abstract
The highly conserved abundant nuclear protein poly(ADP-ribose)polymerase1 (PARP1) functions at the center of cellular stress response and is mainly implied in DNA damage repair mechanism. Apart from its involvement in DNA damage repair, it does sway multiple vital cellular processes such as cell death pathways, cell aging, insulator function, chromatin modification, transcription and mitotic apparatus function. Since brain is the principal organ vulnerable to oxidative stress and inflammatory responses, upon stress encounters robust DNA damage can occur and intense PARP1 activation may result that will lead to various CNS diseases. In the context of soaring interest towards PARP1 as a therapeutic target for newer pharmacological interventions, here in the present review, we are attempting to give a silhouette of the role of PARP1 in the neurological diseases and the potential of its inhibitors to enter clinical translation, along with its structural and functional aspects.
Collapse
Affiliation(s)
- Chandra Shekhar Sriram
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), III Floor, Guwahati Medical College, Narkachal Hilltop, Bhangagarh, Guwahati, Assam 781032, India.
| | - Ashok Jangra
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), III Floor, Guwahati Medical College, Narkachal Hilltop, Bhangagarh, Guwahati, Assam 781032, India
| | - Eshvendar Reddy Kasala
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), III Floor, Guwahati Medical College, Narkachal Hilltop, Bhangagarh, Guwahati, Assam 781032, India
| | - Lakshmi Narendra Bodduluru
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), III Floor, Guwahati Medical College, Narkachal Hilltop, Bhangagarh, Guwahati, Assam 781032, India
| | - Babul Kumar Bezbaruah
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), III Floor, Guwahati Medical College, Narkachal Hilltop, Bhangagarh, Guwahati, Assam 781032, India; Department of Pharmacology, III Floor, Guwahati Medical College, Narkachal Hilltop, Bhangagarh, Guwahati, Assam 781032, India
| |
Collapse
|
25
|
Flores JJ, Zhang Y, Klebe DW, Lekic T, Fu W, Zhang JH. Small molecule inhibitors in the treatment of cerebral ischemia. Expert Opin Pharmacother 2014; 15:659-80. [PMID: 24491068 DOI: 10.1517/14656566.2014.884560] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Stroke is the world's second leading cause of death. Although recombinant tissue plasminogen activator is an effective treatment for cerebral ischemia, its limitations and ischemic stroke's complex pathophysiology dictate an increased need for the development of new therapeutic interventions. Small molecule inhibitors (SMIs) have the potential to be used as novel therapeutic modalities for stroke, since many preclinical and clinical trials have established their neuroprotective capabilities. AREAS COVERED This paper provides a summary of the pathophysiology of stroke as well as clinical and preclinical evaluations of SMIs as therapeutic interventions for cerebral ischemia. Cerebral ischemia is broken down into four mechanisms in this article: thrombosis, ischemic insult, mitochondrial injury and immune response. Insight is provided into preclinical and current clinical assessments of SMIs targeting each mechanism as well as a summary of reported results. EXPERT OPINION Many studies demonstrated that pre- or post-treatment with certain SMIs significantly ameliorated adverse effects from stroke. Although some of these promising SMIs moved on to clinical trials, they generally failed, possibly due to the poor translation of preclinical to clinical experiments. Yet, there are many steps being taken to improve the quality of experimental research and translation to clinical trials.
Collapse
Affiliation(s)
- Jerry J Flores
- Loma Linda University School of Medicine, Department of Physiology and Pharmacology , Risley Hall, Room 223, Loma Linda, CA 92354 , USA
| | | | | | | | | | | |
Collapse
|
26
|
Hemorrhagic transformation after ischemic stroke in animals and humans. J Cereb Blood Flow Metab 2014; 34:185-99. [PMID: 24281743 PMCID: PMC3915212 DOI: 10.1038/jcbfm.2013.203] [Citation(s) in RCA: 408] [Impact Index Per Article: 37.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 10/10/2013] [Accepted: 10/28/2013] [Indexed: 01/12/2023]
Abstract
Hemorrhagic transformation (HT) is a common complication of ischemic stroke that is exacerbated by thrombolytic therapy. Methods to better prevent, predict, and treat HT are needed. In this review, we summarize studies of HT in both animals and humans. We propose that early HT (<18 to 24 hours after stroke onset) relates to leukocyte-derived matrix metalloproteinase-9 (MMP-9) and brain-derived MMP-2 that damage the neurovascular unit and promote blood-brain barrier (BBB) disruption. This contrasts to delayed HT (>18 to 24 hours after stroke) that relates to ischemia activation of brain proteases (MMP-2, MMP-3, MMP-9, and endogenous tissue plasminogen activator), neuroinflammation, and factors that promote vascular remodeling (vascular endothelial growth factor and high-moblity-group-box-1). Processes that mediate BBB repair and reduce HT risk are discussed, including transforming growth factor beta signaling in monocytes, Src kinase signaling, MMP inhibitors, and inhibitors of reactive oxygen species. Finally, clinical features associated with HT in patients with stroke are reviewed, including approaches to predict HT by clinical factors, brain imaging, and blood biomarkers. Though remarkable advances in our understanding of HT have been made, additional efforts are needed to translate these discoveries to the clinic and reduce the impact of HT on patients with ischemic stroke.
Collapse
|
27
|
Stoica BA, Loane DJ, Zhao Z, Kabadi SV, Hanscom M, Byrnes KR, Faden AI. PARP-1 inhibition attenuates neuronal loss, microglia activation and neurological deficits after traumatic brain injury. J Neurotrauma 2014; 31:758-72. [PMID: 24476502 DOI: 10.1089/neu.2013.3194] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Traumatic brain injury (TBI) causes neuronal cell death as well as microglial activation and related neurotoxicity that contribute to subsequent neurological dysfunction. Poly (ADP-ribose) polymerase (PARP-1) induces neuronal cell death through activation of caspase-independent mechanisms, including release of apoptosis inducing factor (AIF), and microglial activation. Administration of PJ34, a selective PARP-1 inhibitor, reduced cell death of primary cortical neurons exposed to N-Methyl-N'-Nitro-N-Nitrosoguanidine (MNNG), a potent inducer of AIF-dependent cell death. PJ34 also attenuated lipopolysaccharide and interferon-γ-induced activation of BV2 or primary microglia, limiting NF-κB activity and iNOS expression as well as decreasing generation of reactive oxygen species and TNFα. Systemic administration of PJ34 starting as late as 24 h after controlled cortical impact resulted in improved motor function recovery in mice with TBI. Stereological analysis demonstrated that PJ34 treatment reduced the lesion volume, attenuated neuronal cell loss in the cortex and thalamus, and reduced microglial activation in the TBI cortex. PJ34 treatment did not improve cognitive performance in a Morris water maze test or reduce neuronal cell loss in the hippocampus. Overall, our data indicate that PJ34 has a significant, albeit selective, neuroprotective effect after experimental TBI, and its therapeutic effect may be from multipotential actions on neuronal cell death and neuroinflammatory pathways.
Collapse
Affiliation(s)
- Bogdan A Stoica
- 1 Department of Anesthesiology, Center for Shock, Trauma and Anesthesiology Research (STAR), National Study Center for Trauma and EMS, University of Maryland , School of Medicine, Baltimore, Maryland
| | | | | | | | | | | | | |
Collapse
|
28
|
Curtin N, Szabo C. Therapeutic applications of PARP inhibitors: anticancer therapy and beyond. Mol Aspects Med 2013; 34:1217-56. [PMID: 23370117 PMCID: PMC3657315 DOI: 10.1016/j.mam.2013.01.006] [Citation(s) in RCA: 287] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 01/12/2013] [Accepted: 01/18/2013] [Indexed: 12/21/2022]
Abstract
The aim of this article is to describe the current and potential clinical translation of pharmacological inhibitors of poly(ADP-ribose) polymerase (PARP) for the therapy of various diseases. The first section of the present review summarizes the available preclinical and clinical data with PARP inhibitors in various forms of cancer. In this context, the role of PARP in single-strand DNA break repair is relevant, leading to replication-associated lesions that cannot be repaired if homologous recombination repair (HRR) is defective, and the synthetic lethality of PARP inhibitors in HRR-defective cancer. HRR defects are classically associated with BRCA1 and 2 mutations associated with familial breast and ovarian cancer, but there may be many other causes of HRR defects. Thus, PARP inhibitors may be the drugs of choice for BRCA mutant breast and ovarian cancers, and extend beyond these tumors if appropriate biomarkers can be developed to identify HRR defects. Multiple lines of preclinical data demonstrate that PARP inhibition increases cytotoxicity and tumor growth delay in combination with temozolomide, topoisomerase inhibitors and ionizing radiation. Both single agent and combination clinical trials are underway. The final part of the first section of the present review summarizes the current status of the various PARP inhibitors that are in various stages of clinical development. The second section of the present review summarizes the role of PARP in selected non-oncologic indications. In a number of severe, acute diseases (such as stroke, neurotrauma, circulatory shock and acute myocardial infarction) the clinical translatability of PARP inhibition is supported by multiple lines of preclinical data, as well as observational data demonstrating PARP activation in human tissue samples. In these disease indications, PARP overactivation due to oxidative and nitrative stress drives cell necrosis and pro-inflammatory gene expression, which contributes to disease pathology. Accordingly, multiple lines of preclinical data indicate the efficacy of PARP inhibitors to preserve viable tissue and to down-regulate inflammatory responses. As the clinical trials with PARP inhibitors in various forms of cancer progress, it is hoped that a second line of clinical investigations, aimed at testing of PARP inhibitors for various non-oncologic indications, will be initiated, as well.
Collapse
Affiliation(s)
- Nicola Curtin
- Department of Experimental Cancer Therapy, Northern Institute for Cancer Research, Newcastle University, University of Newcastle Upon Tyne, UK
| | - Csaba Szabo
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|
29
|
Teng F, Beray-Berthat V, Coqueran B, Lesbats C, Kuntz M, Palmier B, Garraud M, Bedfert C, Slane N, Bérézowski V, Szeremeta F, Hachani J, Scherman D, Plotkine M, Doan BT, Marchand-Leroux C, Margaill I. Prevention of rt-PA induced blood-brain barrier component degradation by the poly(ADP-ribose)polymerase inhibitor PJ34 after ischemic stroke in mice. Exp Neurol 2013; 248:416-28. [PMID: 23876515 DOI: 10.1016/j.expneurol.2013.07.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 06/28/2013] [Accepted: 07/12/2013] [Indexed: 01/24/2023]
Abstract
Recombinant tissue plasminogen activator (rt-PA) is the only pharmacological treatment approved for thrombolysis in patients suffering from ischemic stroke, but its administration aggravates the risk of hemorrhagic transformations. Experimental data demonstrated that rt-PA increases the activity of poly(ADP-ribose)polymerase (PARP). The aim of the present study was to investigate whether PJ34, a potent (PARP) inhibitor, protects the blood-brain barrier components from rt-PA toxicity. In our mouse model of cerebral ischemia, administration of rt-PA (10 mg/kg, i.v.) 6h after ischemia aggravated the post-ischemic degradation of ZO-1, claudin-5 and VE-cadherin, increased the hemorrhagic transformations (assessed by brain hemoglobin content and magnetic resonance imaging). Furthermore, rt-PA also aggravated ischemia-induced functional deficits. Combining PJ34 with rt-PA preserved the expression of ZO-1, claudin-5 and VE-cadherin, reduced the hemorrhagic transformations and improved the sensorimotor performances. In vitro studies also demonstrated that PJ34 crosses the blood-brain barrier and may thus exert its protective effect by acting on endothelial and/or parenchymal cells. Thus, co-treatment with a PARP inhibitor seems to be a promising strategy to reduce rt-PA-induced vascular toxicity after stroke.
Collapse
Affiliation(s)
- Fei Teng
- Equipe de recherche "Pharmacologie de la Circulation Cérébrale" EA4475, Université Paris Descartes, Sorbonne Paris Cité, Faculté des Sciences Pharmaceutiques et Biologiques, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
El Amki M, Lerouet D, Coqueran B, Curis E, Orset C, Vivien D, Plotkine M, Marchand-Leroux C, Margaill I. Experimental modeling of recombinant tissue plasminogen activator effects after ischemic stroke. Exp Neurol 2012; 238:138-44. [DOI: 10.1016/j.expneurol.2012.08.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Revised: 07/26/2012] [Accepted: 08/07/2012] [Indexed: 10/28/2022]
|
31
|
Fréchou M, Beray-Berthat V, Raynaud JS, Mériaux S, Gombert F, Lancelot E, Plotkine M, Marchand-Leroux C, Ballet S, Robert P, Louin G, Margaill I. Detection of vascular cell adhesion molecule-1 expression with USPIO-enhanced molecular MRI in a mouse model of cerebral ischemia. CONTRAST MEDIA & MOLECULAR IMAGING 2012; 8:157-64. [DOI: 10.1002/cmmi.1512] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Revised: 09/07/2012] [Accepted: 09/19/2012] [Indexed: 11/06/2022]
Affiliation(s)
| | - V. Beray-Berthat
- Equipe de recherche ‘Pharmacologie de la Circulation Cérébrale’ EA 4475; Université Paris Descartes, Sorbonne Paris Cité, Faculté des Sciences Pharmaceutiques et Biologiques; 75006; Paris; France
| | - J.-S. Raynaud
- Guerbet, Division Recherche - Service Imagerie Expérimentale; 95943; Roissy; CDG; France
| | - S. Mériaux
- NeuroSpin, Centre CEA; 91191; Gif-sur-Yvette; France
| | - F. Gombert
- Guerbet, Division Recherche - Service Imagerie Expérimentale; 95943; Roissy; CDG; France
| | - E. Lancelot
- Guerbet, Division Recherche - Service Imagerie Expérimentale; 95943; Roissy; CDG; France
| | - M. Plotkine
- Equipe de recherche ‘Pharmacologie de la Circulation Cérébrale’ EA 4475; Université Paris Descartes, Sorbonne Paris Cité, Faculté des Sciences Pharmaceutiques et Biologiques; 75006; Paris; France
| | - C. Marchand-Leroux
- Equipe de recherche ‘Pharmacologie de la Circulation Cérébrale’ EA 4475; Université Paris Descartes, Sorbonne Paris Cité, Faculté des Sciences Pharmaceutiques et Biologiques; 75006; Paris; France
| | - S. Ballet
- Guerbet, Division Recherche - Service Imagerie Expérimentale; 95943; Roissy; CDG; France
| | - P. Robert
- Guerbet, Division Recherche - Service Imagerie Expérimentale; 95943; Roissy; CDG; France
| | - G. Louin
- Guerbet, Division Recherche - Service Imagerie Expérimentale; 95943; Roissy; CDG; France
| | - I. Margaill
- Equipe de recherche ‘Pharmacologie de la Circulation Cérébrale’ EA 4475; Université Paris Descartes, Sorbonne Paris Cité, Faculté des Sciences Pharmaceutiques et Biologiques; 75006; Paris; France
| |
Collapse
|
32
|
Poittevin M, Deroide N, Azibani F, Delcayre C, Giannesini C, Levy BI, Pocard M, Kubis N. Glatiramer Acetate administration does not reduce damage after cerebral ischemia in mice. J Neuroimmunol 2012; 254:55-62. [PMID: 23026222 DOI: 10.1016/j.jneuroim.2012.09.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Revised: 08/23/2012] [Accepted: 09/07/2012] [Indexed: 11/15/2022]
Abstract
Inflammation plays a key role in ischemic stroke pathophysiology: microglial/macrophage cells and type-1 helper cells (Th1) seem deleterious, while type-2 helper cells (Th2) and regulatory T cells (Treg) seem protective. CD4 Th0 differentiation is modulated by microglial cytokine secretion. Glatiramer Acetate (GA) is an immunomodulatory drug that has been approved for the treatment of human multiple sclerosis by means of a number of mechanisms: reduced microglial activation and pro-inflammatory cytokine production, Th0 differentiation shifting from Th2 to Th2 and Treg with anti-inflammatory cytokine production and increased neurogenesis. We induced permanent (pMCAo) or transient middle cerebral artery occlusion (tMCAo) and GA (2 mg) or vehicle was injected subcutaneously immediately after cerebral ischemia. Mice were sacrificed at D3 to measure neurological deficit, infarct volume, microglial cell density and qPCR of TNFα and IL-1β (pro-inflammatory microglial cytokines), IFNγ (Th2 cytokine), IL-4 (Th2 cytokine), TGFβ and IL-10 (Treg cytokines), and at D7 to evaluate neurological deficit, infarct volume and neurogenesis assessment. We showed that in GA-treated pMCAo mice, infarct volume, microglial cell density and cytokine secretion were not significantly modified at D3, while neurogenesis was enhanced at D7 without significant infarct volume reduction. In GA-treated tMCAo mice, microglial pro-inflammatory cytokines IL-1β and TNFα were significantly decreased without modification of microglial/macrophage cell density, cytokine secretion, neurological deficit or infarct volume at D3, or modification of neurological deficit, neurogenesis or infarct volume at D7. In conclusion, Glatiramer Acetate administered after cerebral ischemia does not reduce infarct volume or improve neurological deficit in mice despite a significant increase in neurogenesis in pMCAo and a microglial pro-inflammatory cytokine reduction in tMCAo.
Collapse
Affiliation(s)
- Marine Poittevin
- Université Paris Diderot, Sorbonne Paris Cité, Angiogenesis and Translational Research Center, INSERM U965, F-75475 Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Moroni F, Cozzi A, Chiarugi A, Formentini L, Camaioni E, Pellegrini-Giampietro DE, Chen Y, Liang S, Zaleska MM, Gonzales C, Wood A, Pellicciari R. Long-lasting neuroprotection and neurological improvement in stroke models with new, potent and brain permeable inhibitors of poly(ADP-ribose) polymerase. Br J Pharmacol 2012; 165:1487-500. [PMID: 21913897 DOI: 10.1111/j.1476-5381.2011.01666.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSES Thienyl-isoquinolone (TIQ-A) is a relatively potent PARP inhibitor able to reduce post-ischaemic neuronal death in vitro. Here we have studied, in different stroke models in vivo, the neuroprotective properties of DAMTIQ and HYDAMTIQ, two TIQ-A derivatives able to reach the brain and to inhibit PARP-1 and PARP-2. EXPERIMENTAL APPROACH Studies were carried out in (i) transient (2 h) middle cerebral artery occlusion (tMCAO), (ii) permanent MCAO (pMCAO) and (iii) electrocoagulation of the distal portion of MCA in conjunction with transient (90 min) bilateral carotid occlusion (focal cortical ischaemia). KEY RESULTS In male rats with tMCAO, HYDAMTIQ (0.1-10 mg·kg(-1)) injected i.p. three times, starting 4 h after MCAO, reduced infarct volumes by up to 70%, reduced the loss of body weight by up to 60% and attenuated the neurological impairment by up to 40%. In age-matched female rats, HYDAMTIQ also reduced brain damage. Protection, however, was less pronounced than in the male rats. In animals with pMCAO, HYDAMTIQ administered 30 min after MCAO reduced infarct volumes by approximately 40%. In animals with focal cortical ischaemia, HYDAMTIQ treatment decreased post-ischaemic accumulation of PAR (the product of PARP activity) and the presence of OX42-positive inflammatory cells in the ischaemic cortex. It also reduced sensorimotor deficits for up to 90 days after MCAO. CONCLUSION AND IMPLICATIONS Our results show that HYDAMTIQ is a potent PARP inhibitor that conferred robust neuroprotection and long-lasting improvement of post-stroke neurological deficits.
Collapse
Affiliation(s)
- F Moroni
- Department of Preclinical and Clinical Pharmacology, University of Florence, Florence, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Haddad M, Beray-Berthat V, Coqueran B, Plotkine M, Marchand-Leroux C, Margaill I. Combined therapy with PJ34, a poly(ADP-ribose)polymerase inhibitor, reduces tissue plasminogen activator-induced hemorrhagic transformations in cerebral ischemia in mice. Fundam Clin Pharmacol 2012; 27:393-401. [DOI: 10.1111/j.1472-8206.2012.01036.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
35
|
Cavone L, Aldinucci A, Ballerini C, Biagioli T, Moroni F, Chiarugi A. PARP-1 inhibition prevents CNS migration of dendritic cells during EAE, suppressing the encephalitogenic response and relapse severity. Mult Scler 2011; 17:794-807. [PMID: 21343230 DOI: 10.1177/1352458511399113] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Pharmacological inhibitors of poly(ADP-ribose) polymerase-1 (PARP-1) are currently evaluated in clinical trials for various malignancies but, interestingly, also proved of remarkable efficacy in preclinical models of autoimmune disorders including experimental autoimmune encephalomyelitis (EAE). OBJECTIVES The objectives of the study were to determine molecular mechanisms underlying suppression of the encephalitogenic response by these drugs; likewise, whether clinically-relevant post-treatment paradigms with PARP-1 inhibitors could prevent EAE relapses. METHODS Adopted both in vitro techniques (bone marrow-derived cultured DC) as well as in vivo models of chronic or relapsing-remitting (RR) EAE. RESULTS We report that two structurally unrelated PARP-1 inhibitors negatively regulated NFκB activation, as well as maturation, cytokine production and APC function of cultured mouse bone marrow-derived dendritic cells (DCs). PARP-1 inhibitors also reduced the number and APC function of DCs migrating in the draining lymph nodes of ovalbumin-immunized mice. In C57Bl mice with chronic EAE or SJL mice with RR EAE, pharmacological inhibition of PARP-1 reduced CNS DC migration and demyelination as well as neurological impairment to an extent similar to that achieved with the potent immunosuppressant cyclosporine A. Remarkably, PARP-1 inhibitors injected after the first phase of disease reduced relapse incidence and severity, as well as the spinal cord number of autoreactive Th17 cells. Under this clinically-relevant treatment paradigm, PARP inhibitors also suppressed epitope spreading of the encephalitogenic response. CONCLUSIONS Overall, data underscore the potential relevance of PARP-1 inhibitors to MS therapy and suppression of autoimmunity.
Collapse
Affiliation(s)
- Leonardo Cavone
- Department of Pharmacology, University of Florence, Florence, Italy.
| | | | | | | | | | | |
Collapse
|
36
|
Lescot T, Fulla-Oller L, Palmier B, Po C, Beziaud T, Puybasset L, Plotkine M, Gillet B, Meric P, Marchand-Leroux C. Effect of Acute Poly(ADP-Ribose) Polymerase Inhibition by 3-AB on Blood–Brain Barrier Permeability and Edema Formation after Focal Traumatic Brain Injury in Rats. J Neurotrauma 2010; 27:1069-79. [DOI: 10.1089/neu.2009.1188] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Thomas Lescot
- Equipe de recherche “Pharmacologie de la Circulation Cérébrale” (EA 2510), Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Paris, France
- Réanimation neurochirurgicale–Département d'Anesthésie Réanimation, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP) et Université Pierre et Marie Curie, Paris, France
| | - Laurence Fulla-Oller
- Réanimation neurochirurgicale–Département d'Anesthésie Réanimation, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP) et Université Pierre et Marie Curie, Paris, France
- Laboratoire de Résonance Magnétique Nucléaire Biologique, Institut de Chimie des Substances Naturelles (ICSN) et Centre National de la Recherche Scientifique (CNRS), Gif sur Yvette, France
| | - Bruno Palmier
- Equipe de recherche “Pharmacologie de la Circulation Cérébrale” (EA 2510), Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Paris, France
| | - Christelle Po
- Laboratoire de Résonance Magnétique Nucléaire Biologique, Institut de Chimie des Substances Naturelles (ICSN) et Centre National de la Recherche Scientifique (CNRS), Gif sur Yvette, France
| | - Tiphaine Beziaud
- Equipe de recherche “Pharmacologie de la Circulation Cérébrale” (EA 2510), Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Paris, France
| | - Louis Puybasset
- Réanimation neurochirurgicale–Département d'Anesthésie Réanimation, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris (AP-HP) et Université Pierre et Marie Curie, Paris, France
| | - Michel Plotkine
- Equipe de recherche “Pharmacologie de la Circulation Cérébrale” (EA 2510), Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Paris, France
| | - Brigitte Gillet
- Laboratoire de Résonance Magnétique Nucléaire Biologique, Institut de Chimie des Substances Naturelles (ICSN) et Centre National de la Recherche Scientifique (CNRS), Gif sur Yvette, France
| | - Philippe Meric
- Laboratoire de Résonance Magnétique Nucléaire Biologique, Institut de Chimie des Substances Naturelles (ICSN) et Centre National de la Recherche Scientifique (CNRS), Gif sur Yvette, France
| | - Catherine Marchand-Leroux
- Equipe de recherche “Pharmacologie de la Circulation Cérébrale” (EA 2510), Faculté des Sciences Pharmaceutiques et Biologiques, Université Paris Descartes, Paris, France
| |
Collapse
|
37
|
NAD+ depletion is necessary and sufficient for poly(ADP-ribose) polymerase-1-mediated neuronal death. J Neurosci 2010; 30:2967-78. [PMID: 20181594 DOI: 10.1523/jneurosci.5552-09.2010] [Citation(s) in RCA: 355] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Poly(ADP-ribose)-1 (PARP-1) is a key mediator of cell death in excitotoxicity, ischemia, and oxidative stress. PARP-1 activation leads to cytosolic NAD(+) depletion and mitochondrial release of apoptosis-inducing factor (AIF), but the causal relationships between these two events have been difficult to resolve. Here, we examined this issue by using extracellular NAD(+) to restore neuronal NAD(+) levels after PARP-1 activation. Exogenous NAD(+) was found to enter neurons through P2X(7)-gated channels. Restoration of cytosolic NAD(+) by this means prevented the glycolytic inhibition, mitochondrial failure, AIF translocation, and neuron death that otherwise results from extensive PARP-1 activation. Bypassing the glycolytic inhibition with the metabolic substrates pyruvate, acetoacetate, or hydroxybutyrate also prevented mitochondrial failure and neuron death. Conversely, depletion of cytosolic NAD(+) with NAD(+) glycohydrolase produced a block in glycolysis inhibition, mitochondrial depolarization, AIF translocation, and neuron death, independent of PARP-1 activation. These results establish NAD(+) depletion as a causal event in PARP-1-mediated cell death and place NAD(+) depletion and glycolytic failure upstream of mitochondrial AIF release.
Collapse
|
38
|
Dong X, Song YN, Liu WG, Guo XL. Mmp-9, a potential target for cerebral ischemic treatment. Curr Neuropharmacol 2009; 7:269-75. [PMID: 20514206 PMCID: PMC2811860 DOI: 10.2174/157015909790031157] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2009] [Revised: 09/22/2009] [Accepted: 10/05/2009] [Indexed: 11/27/2022] Open
Abstract
Matrix metalloproteinase-9 (MMP-9) which is a member of matrix metalloproteinases family that normally remodel the extracellular matrix, has been shown to play an important role in both animal models of cerebral ischemia and human stroke. The expression of MMP-9 is elevated after cerebral ischemia which is involved in accelerating matrix degradation, disrupting the blood-brain barrier, increasing the infarct size and relating to hemorrhagic transformation. Recently, many drugs, such as tetracycline derivatives, cyclooxygenase inhibitors, ACEI inhibitors and AT1 receptor blockers, etc., have been found to attenuate the elevated expression levels of MMP-9 after ischemia and to reduce the damage of cerebral ischemic. This article reviews the physiological features of MMP-9 and its important role in the genesis, propagation, and therapeutics of cerebral ischemic diseases.
Collapse
Affiliation(s)
- Xue Dong
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, No. 44 WenHuaXi Road, Jinan 250012, P.R. China
| | - Yu-Ning Song
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, No. 44 WenHuaXi Road, Jinan 250012, P.R. China
| | - Wei-Guo Liu
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, No. 44 WenHuaXi Road, Jinan 250012, P.R. China
- Qianfoshan Hospital, Jinan 250014, P.R. China
| | - Xiu-Li Guo
- Department of Pharmacology, School of Pharmaceutical Sciences, Shandong University, No. 44 WenHuaXi Road, Jinan 250012, P.R. China
| |
Collapse
|
39
|
Kauppinen TM, Suh SW, Berman AE, Hamby AM, Swanson RA. Inhibition of poly(ADP-ribose) polymerase suppresses inflammation and promotes recovery after ischemic injury. J Cereb Blood Flow Metab 2009; 29:820-9. [PMID: 19190653 DOI: 10.1038/jcbfm.2009.9] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The brain inflammatory response induced by stroke contributes to cell death and impairs neurogenesis. Poly(ADP-ribose) polymerase-1 (PARP-1) is a coactivator of the transcription factor NF-kappaB and required for NF-kappaB-mediated inflammatory responses. Here we evaluated PARP inhibition as a means of suppressing post-stroke inflammation and improving outcome after stroke. Rats were subjected to bilateral carotid occlusion-reperfusion, and treatment with the PARP inhibitor N-(6-oxo-5,6-dihydrophenanthridin-2-yl)-N,N-dimethylacetamide (PJ34) was begun 48 h later. PJ34 was found to rapidly suppress the ischemia-induced microglial activation and astrogliosis. Behavioral tests performed 6 to 8 weeks after ischemia showed deficits in spatial memory and learning that were lessened by the PJ34 treatment. Immunohistochemical evaluation of hippocampus at 8 weeks after ischemia showed increased neuronal density in CA1 layer of PJ34-treated animals relative to vehicle-treated animals. Bromodeoxyuridine labeling showed formation of new neurons in hippocampal CA1 area in PJ34-treated animals, but not in vehicle-treated animals. Together, these results suggest that treatment with a PARP inhibitor for several days after ischemia enhances long-term neuronal survival and neurogenesis by reducing inflammation.
Collapse
Affiliation(s)
- Tiina M Kauppinen
- Department of Neurology, University of California San Francisco, San Francisco, California 94121, USA.
| | | | | | | | | |
Collapse
|
40
|
Abstract
The mitochondrial pathway to apoptosis is a major pathway of physiological cell death in vertebrates. The mitochondrial cell death pathway commences when apoptogenic molecules present between the outer and inner mitochondrial membranes are released into the cytosol by mitochondrial outer membrane permeabilization (MOMP). BCL-2 family members are the sentinels of MOMP in the mitochondrial apoptotic pathway; the pro-apoptotic B cell lymphoma (BCL)-2 proteins, BCL-2 associated x protein and BCL-2 antagonist killer 1 induce MOMP whereas the anti-apoptotic BCL-2 proteins, BCL-2, BCL-xl and myeloid cell leukaemia 1 prevent MOMP from occurring. The release of pro-apoptotic factors such as cytochrome c from mitochondria leads to formation of a multimeric complex known as the apoptosome and initiates caspase activation cascades. These pathways are important for normal cellular homeostasis and play key roles in the pathogenesis of many diseases. In this review, we will provide a brief overview of the mitochondrial death pathway and focus on a selection of diseases whose pathogenesis involves the mitochondrial death pathway and we will examine the various pharmacological approaches that target this pathway.
Collapse
|