1
|
Alaqel SI, Dlamini S, Almarghalani DA, Shettigar A, Alhadidi Q, Kodithuwakku SH, Stary C, Tillekeratne LMV, Shah ZA. Synthesis and Development of a Novel First-in-Class Cofilin Inhibitor for Neuroinflammation in Hemorrhagic Brain Injury. ACS Chem Neurosci 2022; 13:1014-1029. [PMID: 35302736 PMCID: PMC9996837 DOI: 10.1021/acschemneuro.2c00010] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is devastating among stroke types with high mortality. To date, not a single therapeutic intervention has been successful. Cofilin plays a critical role in inflammation and cell death. In the current study, we embarked on designing and synthesizing a first-in-class small-molecule inhibitor of cofilin to target secondary complications of ICH, mainly neuroinflammation. A series of compounds were synthesized, and two lead compounds SZ-3 and SK-1-32 were selected for further studies. Neuronal and microglial viabilities were assessed by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) assay using neuroblastoma (SHSY-5Y) and human microglial (HMC-3) cell lines, respectively. Lipopolysaccharide (LPS)-induced inflammation in HMC-3 cells was used for neurotoxicity assay. Other assays include nitric oxide (NO) by Griess reagent, cofilin inhibition by F-actin depolymerization, migration by scratch wound assay, tumor necrosis factor (TNF-α) by enzyme-linked immunosorbent assay (ELISA), protease-activated receptor-1 (PAR-1) by immunocytochemistry and Western blotting (WB), and protein expression levels of several proteins by WB. SK-1-32 increased neuronal/microglial survival, reduced NO, and prevented neurotoxicity. However, SZ-3 showed no effect on neuronal/microglial survival but prevented microglia from LPS-induced inflammation by decreasing NO and preventing neurotoxicity. Therefore, we selected SZ-3 for further molecular studies, as it showed potent anti-inflammatory activities. SZ-3 decreased cofilin severing activity, and its treatment of LPS-activated HMC-3 cells attenuated microglial activation and suppressed migration and proliferation. HMC-3 cells subjected to thrombin, as an in vitro model for hemorrhagic stroke, and treated with SZ-3 after 3 h showed significantly decreased NO and TNF-α, significantly increased protein expression of phosphocofilin, and decreased PAR-1. In addition, SZ-3-treated SHSY-5Y showed a significant increase in cell viability by significantly reducing nuclear factor-κ B (NF-κB), caspase-3, and high-temperature requirement (HtrA2). Together, our results support the novel idea of targeting cofilin to counter neuroinflammation during secondary injury following ICH.
Collapse
Affiliation(s)
- Saleh I. Alaqel
- Department of Medicinal and Biological Chemistry, The University of Toledo, Toledo, OH, USA 43614
| | - Samkeliso Dlamini
- Department of Medicinal and Biological Chemistry, The University of Toledo, Toledo, OH, USA 43614
| | - Daniyah A. Almarghalani
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, The University of Toledo, Toledo, OH, USA 43614
| | - Arjun Shettigar
- Department of Medicinal and Biological Chemistry, The University of Toledo, Toledo, OH, USA 43614
| | - Qasim Alhadidi
- Department of Medicinal and Biological Chemistry, The University of Toledo, Toledo, OH, USA 43614
| | - Sinali H. Kodithuwakku
- Department of Medicinal and Biological Chemistry, The University of Toledo, Toledo, OH, USA 43614
| | - Creed Stary
- Department of Anesthesiology, Perioperative and Pain Medicine, Stanford University School of Medicine, Stanford, CA, USA 94305
| | | | - Zahoor A. Shah
- Department of Medicinal and Biological Chemistry, The University of Toledo, Toledo, OH, USA 43614
| |
Collapse
|
2
|
Yao X, Song Y, Wang Z, Bai S, Yu H, Wang Y, Guan Y. Proteinase-activated receptor-1 antagonist attenuates brain injury via regulation of FGL2 and TLR4 after intracerebral hemorrhage in mice. Neuroscience 2022; 490:193-205. [PMID: 35182700 DOI: 10.1016/j.neuroscience.2022.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 02/07/2022] [Accepted: 02/10/2022] [Indexed: 11/25/2022]
Abstract
Proteinase-activated receptor-1 (PAR1) antagonist plays a protective effect in brain injury. We investigated the potential function and mechanisms of PAR1 antagonist in ICH-induced brain injury. Results showed that PAR1 antagonist protected against neurobehavior deficits, brain edema and BBB integrity in ICH mice via activating JNK/ERK/p38 MAPK signaling pathway at 24h after ICH. In addition, ICH resulted in the increase of FGL2 and TLR4 expression over time, and phosphorylated JNK, ERK and p38 MAPK expression. Suppression of FGL2 and TLR4 alleviated brain injury and decreased the expression of p-JNK, p-ERK, p-p38 MAPK and p-IKKα at 24 h after ICH; while overexpression of them showed the opposite result. Moreover, the protective effect of PAR1 antagonist on ICH-induced brain injury was blocked by FGL2 or TLR4 overexpression, and the levels of p-JNK, p-ERK and p-p38 MAPK were inhibited. Furthermore, PAR1 antagonist combined with TLR4 antagonist markedly alleviated brain injury after ICH at 72h. Overall, PAR1 antagonist protected against short-term brain injury, and the effect of PAR1 antagonist on ICH-induced brain injury was mediated by FGL2 or TLR4.
Collapse
Affiliation(s)
- Xiaoying Yao
- Department of Neurology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yaying Song
- Department of Neurology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Ze Wang
- Department of Neurology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Shuwei Bai
- Department of Neurology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Haojun Yu
- Department of Neurology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yishu Wang
- Department of Neurology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Yangtai Guan
- Department of Neurology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China.
| |
Collapse
|
3
|
Scimemi A. Astrocytes and the Warning Signs of Intracerebral Hemorrhagic Stroke. Neural Plast 2018; 2018:7301623. [PMID: 29531526 PMCID: PMC5817320 DOI: 10.1155/2018/7301623] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 12/13/2017] [Indexed: 12/11/2022] Open
Abstract
Two decades into the two thousands, intracerebral hemorrhagic stroke (ICH) continues to reap lives across the globe. In the US, nearly 12,000 people suffer from ICH every year. Half of them survive, but many are left with permanent physical and cognitive disabilities, the severity of which depends on the location and broadness of the brain region affected by the hemorrhage. The ongoing efforts to identify risk factors for hemorrhagic stroke have been instrumental for the development of new medical practices to prevent, aid the recovery and reduce the risk of recurring ICH. Recent efforts approach the study of ICH from a different angle, providing information on how we can limit brain damage by manipulating astrocyte receptors. These results provide a novel understanding of how astrocytes contribute to brain injury and recovery from small ICH. Here, we discuss current knowledge on the risk factors and molecular pathology of ICH and the functional properties of astrocytes and their role in ICH. Last, we discuss candidate astrocyte receptors that may prove to be valuable therapeutic targets to treat ICH. Together, these findings provide basic and clinical scientists useful information for the future development of strategies to improve the detection of small ICH, limit brain damage, and prevent the onset of more severe episodes of brain hemorrhage.
Collapse
Affiliation(s)
- Annalisa Scimemi
- SUNY Albany, Department of Biology, 1400 Washington Avenue, Albany, NY 12222, USA
| |
Collapse
|
4
|
Bao L, Zu J, He Q, Zhao H, Zhou S, Ye X, Yang X, Zan K, Zhang Z, Shi H, Cui G. Thrombin-induced apoptosis in neurons through activation of c-Jun-N-terminal kinase. Toxicol Mech Methods 2016; 27:18-23. [PMID: 27841083 DOI: 10.3109/15376516.2016.1172691] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
CONTEXT Studies have shown that thrombin activation played a central role in cell injuries associated with intracerebral hemorrhage (ICH). OBJECTIVE Here, our study investigated the cytotoxicity of thrombin on neurons, and determined the involvement of JNK pathways in thrombin-induced neuronal apoptosis. MATERIALS AND METHODS Primary cultured neurons were treated with different doses of thrombin. Some neurons were given either SP600125 or vehicle. LDH release assay and flow cytometry were used to measure neuronal apoptosis caused by thrombin. The activation of JNK and capases-3 were measured by Western blot. RESULTS Our results showed large doses of thrombin that increased the LDH release, the level of cleaved caspase-3 and apoptosis rate of neurons. JNK was activated by thrombin in a time-dependent manner. Administration of SP600125 protects neurons from thrombin-induced apoptosis. CONCLUSION These data indicate that the activation of JNK is crucial for thrombin-induced neuronal apoptosis, and inhibition of JNK may be a potential therapeutic target for ICH.
Collapse
Affiliation(s)
- Lei Bao
- a Department of Neurology , The Affiliated Hospital of Xuzhou Medical College , Xuzhou , Jiangsu , China
| | - Jie Zu
- a Department of Neurology , The Affiliated Hospital of Xuzhou Medical College , Xuzhou , Jiangsu , China
| | - Qianqian He
- a Department of Neurology , The Affiliated Hospital of Xuzhou Medical College , Xuzhou , Jiangsu , China
| | - Hui Zhao
- b Department of Neurology , Xuzhou Central Hospital , Xuzhou , Jiangsu , China
| | - Su Zhou
- a Department of Neurology , The Affiliated Hospital of Xuzhou Medical College , Xuzhou , Jiangsu , China
| | - Xinchun Ye
- a Department of Neurology , The Affiliated Hospital of Xuzhou Medical College , Xuzhou , Jiangsu , China
| | - Xinxin Yang
- a Department of Neurology , The Affiliated Hospital of Xuzhou Medical College , Xuzhou , Jiangsu , China
| | - Kun Zan
- a Department of Neurology , The Affiliated Hospital of Xuzhou Medical College , Xuzhou , Jiangsu , China
| | - Zuohui Zhang
- a Department of Neurology , The Affiliated Hospital of Xuzhou Medical College , Xuzhou , Jiangsu , China
| | - Hongjuan Shi
- a Department of Neurology , The Affiliated Hospital of Xuzhou Medical College , Xuzhou , Jiangsu , China
| | - Guiyun Cui
- a Department of Neurology , The Affiliated Hospital of Xuzhou Medical College , Xuzhou , Jiangsu , China
| |
Collapse
|
5
|
Electroacupuncture Exerts Neuroprotection through Caveolin-1 Mediated Molecular Pathway in Intracerebral Hemorrhage of Rats. Neural Plast 2016; 2016:7308261. [PMID: 27725888 PMCID: PMC5048052 DOI: 10.1155/2016/7308261] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 08/25/2016] [Indexed: 01/08/2023] Open
Abstract
Spontaneous intracerebral hemorrhage (ICH) is one of the most devastating types of stroke. Here, we aim to demonstrate that electroacupuncture on Baihui (GV20) exerts neuroprotection for acute ICH possibly via the caveolin-1/matrix metalloproteinase/blood-brain barrier permeability pathway. The model of ICH was established by using collagenase VII. Rats were randomly divided into three groups: Sham-operation group, Sham electroacupuncture group, and electroacupuncture group. Each group was further divided into 4 subgroups according to the time points of 6 h, 1 d, 3 d, and 7 d after ICH. The methods were used including examination of neurological deficit scores according to Longa's scale, measurement of blood-brain barrier permeability through Evans Blue content, in situ immunofluorescent detection of caveolin-1 in brains, western blot analysis of caveolin-1 in brains, and in situ zymography for measuring matrix metalloproteinase-2/9 activity in brains. Compared with Sham electroacupuncture group, electroacupuncture group has resulted in a significant improvement in neurological deficit scores and in a reduction in Evans Blue content, expression of caveolin-1, and activity of matrix metalloproteinase-2/9 at 6 h, 1 d, 3 d, and 7 d after ICH (P < 0.05). In conclusion, the present results suggested that electroacupuncture on GV20 can improve neurological deficit scores and reduce blood-brain barrier permeability after ICH, and the mechanism possibly targets caveolin-1/matrix metalloproteinase/blood-brain barrier permeability pathway.
Collapse
|
6
|
Guan J, Zhang S, Zhou Q, Yuan Z, Lu Z. Effect of thrombin preconditioning on migration of subventricular zone-derived cells after intracerebral hemorrhage in rats. Neurol Res 2016; 38:809-16. [PMID: 27477964 DOI: 10.1080/01616412.2016.1210356] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
OBJECTIVE To investigate the effect of thrombin preconditioning (TPC) on the intracerebral hemorrhage (ICH)-induced proliferation, migration, and function of subventriclular zone (SVZ) cells and to find new strategies that enhance endogenous neurogenesis after ICH. METHODS Male Sprague-Dawley rats were randomly divided into 3 groups (ICH, TPC, and control group). Rats of each group were randomly divided into 5 subgroups (3-d, 7-d, 14-d, 21-d, and 28-d subgroup). ICH was caused by intrastrial stereotactic administration of collagenase type IV. Brdu was used to label newborn SVZ cells. Organotypic brain slices were cultured to dynamically observe the migration of SVZ cells at living brain tissue. Migration of Dil-labeled SVZ cells in living brain slices was traced by time-lapse microscopy. To assess whether SVZ cells migrating to injured striatum had the ability to form synapses with other cells, brain slices from each group were double immunolabeled with Brdu and synapsin I. RESULTS The number of Brdu-positive cells markedly increased in the ipsilateral SVZ and striatum 3 days after TPC, peaked at 14 days (P < 0.01), continued to 21 days, and then gradually decreased at 28 days with significant difference compared to the ICH group at each time point (P < 0.01). Migration of Dil-labeled SVZ cells in brain slices in each group was observed and imaged during a 12-h period. Dil-labeled SVZ cells in the TPC group were observed to migrate laterally toward striatum with time with a faster velocity compared to the ICH group (P < 0.01). Our study also demonstrated that TPC induced strong colocalization of Brdu and synapsin I in the ipsilateral striatum between 3 and 28 days after injury.TPC made colocalization of Brdu and synapsin I appear earlier and continue for a longer time compared to the ICH group. CONCLUSIONS Our results demonstrated that TPC could promote proliferation, migration, and function of SVZ cells after ICH, which may provide a new idea for enhancing endogenous neurogenesis and developing new therapeutic strategies against ICH-induced brain injury.
Collapse
Affiliation(s)
- Jingxia Guan
- a Department of Neurology , Renmin Hospital of Wuhan University , Wuhan , China
| | - Shaofeng Zhang
- a Department of Neurology , Renmin Hospital of Wuhan University , Wuhan , China
| | - Qin Zhou
- a Department of Neurology , Renmin Hospital of Wuhan University , Wuhan , China
| | - Zhenhua Yuan
- a Department of Neurology , Renmin Hospital of Wuhan University , Wuhan , China
| | - Zuneng Lu
- a Department of Neurology , Renmin Hospital of Wuhan University , Wuhan , China
| |
Collapse
|
7
|
Kim H, Edwards NJ, Choi HA, Chang TR, Jo KW, Lee K. Treatment Strategies to Attenuate Perihematomal Edema in Patients With Intracerebral Hemorrhage. World Neurosurg 2016; 94:32-41. [PMID: 27373415 DOI: 10.1016/j.wneu.2016.06.093] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2016] [Revised: 06/20/2016] [Accepted: 06/21/2016] [Indexed: 11/24/2022]
Abstract
Spontaneous intracerebral hemorrhage (SICH) continues to be a significant cause of neurologic morbidity and mortality throughout the world. Although recent advances in the treatment of SICH have significantly decreased mortality rates, functional recovery has not been dramatically improved by any intervention to date. There are 2 predominant mechanisms of brain injury from intracerebral hemorrhage: mechanical injury from the primary hematoma (including growth of that hematoma), and secondary injury from perihematomal inflammation. For instance, in the hours to weeks after SICH as the hematoma is being degraded, thrombin and iron are released and can result in neurotoxicity, free radical damage, dysregulated coagulation, and harmful inflammatory cascades; this can clinically and radiologically manifest as perihematomal edema (PHE). PHE can contribute to mass effect, cause acute neurologic deterioration in patients, and has even been associated with poor long-term functional outcomes. PHE therefore lends itself to being a potential therapeutic target. In this article, we will review 1) the pathogenesis and time course of the development of PHE, and 2) the clinical series and trials exploring various methods, with a focus on minimally invasive surgical techniques, to reduce PHE and minimize secondary brain injury. Promising areas of continued research also will be discussed.
Collapse
Affiliation(s)
- Hoon Kim
- Department of Neurosurgery, College of Medicine, Bucheon St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea
| | - Nancy J Edwards
- Department of Neurosurgery and Neurology, University of Texas Medical School at Houston, Houston, Texas, USA
| | - Huimahn A Choi
- Department of Neurosurgery and Neurology, University of Texas Medical School at Houston, Houston, Texas, USA
| | - Tiffany R Chang
- Department of Neurosurgery and Neurology, University of Texas Medical School at Houston, Houston, Texas, USA
| | - Kwang Wook Jo
- Department of Neurosurgery, College of Medicine, Bucheon St. Mary's Hospital, The Catholic University of Korea, Seoul, Republic of Korea.
| | - Kiwon Lee
- Department of Neurosurgery and Neurology, University of Texas Medical School at Houston, Houston, Texas, USA
| |
Collapse
|
8
|
Askenase MH, Sansing LH. Stages of the Inflammatory Response in Pathology and Tissue Repair after Intracerebral Hemorrhage. Semin Neurol 2016; 36:288-97. [PMID: 27214704 DOI: 10.1055/s-0036-1582132] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Intracerebral hemorrhage (ICH) is a major health concern, with high rates of mortality and morbidity and no highly effective clinical interventions. Basic research in animal models of ICH has provided insight into its complex pathology, in particular revealing the role of inflammation in driving neuronal death and neurologic deficits after hemorrhage. The response to ICH occurs in four distinct phases: (1) initial tissue damage and local activation of inflammatory factors, (2) inflammation-driven breakdown of the blood-brain barrier, (3) recruitment of circulating inflammatory cells and subsequent secondary immunopathology, and (4) engagement of tissue repair responses that promote tissue repair and restoration of neurologic function. The development of CNS inflammation occurs over many days after initial hemorrhage and thus may represent an ideal target for treatment of the disease, but further research is required to identify the mechanisms that promote engagement of inflammatory versus anti-inflammatory pathways. In this review, the authors examine how experimental models of ICH have uncovered critical mediators of pathology in each of the four stages of the inflammatory response, and focus on the role of the immune system in these processes.
Collapse
Affiliation(s)
- Michael H Askenase
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut
| | - Lauren H Sansing
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
9
|
Itsekson-Hayosh Z, Shavit-Stein E, Katzav A, Rubovitch V, Maggio N, Chapman J, Harnof S, Pick CG. Minimal Traumatic Brain Injury in Mice: Protease-Activated Receptor 1 and Thrombin-Related Changes. J Neurotrauma 2016; 33:1848-1854. [PMID: 26537880 DOI: 10.1089/neu.2015.4146] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Minimal traumatic brain injury (mTBI) is partially defined by the existence of retrograde amnesia and is associated with microscopic bleeds containing activated coagulation factors. In a previous study, we have found that mTBI immediately releases thrombin-like activity in the brain, which induces amnesia by activating protease-activated receptor 1 (PAR-1) and blocking long-term potentiation (LTP). In the present study, we assessed the effects of mTBI on thrombin and PAR-1 levels in the brain using the same model. After the immediate elevation, thrombin activity returned to baseline 1 h post-trauma and increased again 72 h later (42% relative to control; p < 0.005). These changes were associated with a significant increase in PAR-1 levels 24 (17%; p < 0.05) and 72 h (20%; p < 0.05) post-trauma. Interestingly, the late elevation in thrombin-like activity was also associated with elevation of the major central nervous system thrombin inhibitor, protease nexin-1, 72 h post-mTBI (10%; p < 0.005). When thrombin was injected into brain ventricles, an increased sensitivity to seizure-like activity was detected at 72 h post-mTBI. The results are compatible with astrocyte activation post-mTBI resulting in increased thrombin secretion, PAR-1 expression, and seizure sensitivity.
Collapse
Affiliation(s)
- Zeev Itsekson-Hayosh
- 1 Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University , Tel Aviv, Israel .,2 Department of Neurology and Joseph Sagol Neuroscience Center, The Chaim Sheba Medical Center , Tel HaShomer, Israel
| | - Efrat Shavit-Stein
- 1 Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University , Tel Aviv, Israel .,2 Department of Neurology and Joseph Sagol Neuroscience Center, The Chaim Sheba Medical Center , Tel HaShomer, Israel
| | - Aviva Katzav
- 2 Department of Neurology and Joseph Sagol Neuroscience Center, The Chaim Sheba Medical Center , Tel HaShomer, Israel
| | - Vardit Rubovitch
- 4 Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University , Tel Aviv, Israel
| | - Nicola Maggio
- 2 Department of Neurology and Joseph Sagol Neuroscience Center, The Chaim Sheba Medical Center , Tel HaShomer, Israel .,3 The Talpiot Medical Leadership Program, The Chaim Sheba Medical Center , Tel HaShomer, Israel .,7 Department of Neurology, Sackler Faculty of Medicine, Tel Aviv University , Tel Aviv, Israel .,8 Sagol School of Neuroscience, Tel Aviv University , Tel Aviv, Israel
| | - Joab Chapman
- 1 Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University , Tel Aviv, Israel .,2 Department of Neurology and Joseph Sagol Neuroscience Center, The Chaim Sheba Medical Center , Tel HaShomer, Israel .,6 Robert and Martha Harden Chair in Mental and Neurological Diseases, Sackler Faculty of Medicine, Tel Aviv University , Tel Aviv, Israel .,8 Sagol School of Neuroscience, Tel Aviv University , Tel Aviv, Israel
| | - Sagi Harnof
- 5 Department of Neurosurgery, The Chaim Sheba Medical Center , Tel HaShomer, Israel
| | - Chaim G Pick
- 4 Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University , Tel Aviv, Israel .,8 Sagol School of Neuroscience, Tel Aviv University , Tel Aviv, Israel
| |
Collapse
|
10
|
Abstract
OPINION STATEMENT Cerebral edema (i.e., "brain swelling") is a common complication following intracerebral hemorrhage (ICH) and is associated with worse clinical outcomes. Perihematomal edema (PHE) accumulates during the first 72 h after hemorrhage, and during this period, patients are at risk of clinical deterioration due to the resulting tissue shifts and brain herniation. First-line medical therapies for patients symptomatic of PHE include osmotic agents, such as mannitol in low- or high-dose bolus form, or boluses of hypertonic saline (HTS) at varied concentrations with or without subsequent continuous infusion. Decompressive craniectomy may be required for symptomatic edema refractory to osmotherapy. Other strategies that reduce PHE such as hypothermia and minimally invasive surgery have shown promise in pilot studies and are currently being evaluated in larger clinical trials. Ongoing basic, translational, and clinical research seek to better elucidate the pathophysiology of PHE to identify novel strategies to prevent edema formation as a next major advance in the treatment of ICH.
Collapse
|
11
|
Urday S, Kimberly WT, Beslow LA, Vortmeyer AO, Selim MH, Rosand J, Simard JM, Sheth KN. Targeting secondary injury in intracerebral haemorrhage--perihaematomal oedema. Nat Rev Neurol 2015; 11:111-22. [PMID: 25623787 DOI: 10.1038/nrneurol.2014.264] [Citation(s) in RCA: 183] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Perihaematomal oedema (PHO) is an important pathophysiological marker of secondary injury in intracerebral haemorrhage (ICH). In this Review, we describe a novel method to conceptualize PHO formation within the framework of Starling's principle of movement of fluid across a capillary wall. We consider progression of PHO through three stages, characterized by ionic oedema (stage 1) and progressive vasogenic oedema (stages 2 and 3). In this context, possible modifiers of PHO volume and their value in identifying patients who would benefit from therapies that target secondary injury are discussed; the practicalities of using neuroimaging to measure PHO volume are also considered. We examine whether PHO can be used as a predictor of neurological outcome following ICH, and we provide an overview of emerging therapies. Our discussion emphasizes that PHO has clinical relevance both as a therapeutic target, owing to its augmentation of the mass effect of a haemorrhage, and as a surrogate marker for novel interventions that target secondary injury.
Collapse
Affiliation(s)
- Sebastian Urday
- Department of Neurology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| | - W Taylor Kimberly
- Department of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
| | - Lauren A Beslow
- Department of Neurology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| | - Alexander O Vortmeyer
- Department of Pathology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| | - Magdy H Selim
- Department of Neurology, Beth Israel Deaconess Medical Centre, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Jonathan Rosand
- Department of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
| | - J Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD 21201-1559, USA
| | - Kevin N Sheth
- Department of Neurology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| |
Collapse
|
12
|
Babu R, Bagley JH, Di C, Friedman AH, Adamson C. Thrombin and hemin as central factors in the mechanisms of intracerebral hemorrhage-induced secondary brain injury and as potential targets for intervention. Neurosurg Focus 2012; 32:E8. [PMID: 22463118 DOI: 10.3171/2012.1.focus11366] [Citation(s) in RCA: 142] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Intracerebral hemorrhage (ICH) is a subtype of stoke that may cause significant morbidity and mortality. Brain injury due to ICH initially occurs within the first few hours as a result of mass effect due to hematoma formation. However, there is increasing interest in the mechanisms of secondary brain injury as many patients continue to deteriorate clinically despite no signs of rehemorrhage or hematoma expansion. This continued insult after primary hemorrhage is believed to be mediated by the cytotoxic, excitotoxic, oxidative, and inflammatory effects of intraparenchymal blood. The main factors responsible for this injury are thrombin and erythrocyte contents such as hemoglobin. Therapies including thrombin inhibitors, N-methyl-D-aspartate antagonists, chelators to bind free iron, and antiinflammatory drugs are currently under investigation for reducing this secondary brain injury. This review will discuss the molecular mechanisms of brain injury as a result of intraparenchymal blood, potential targets for therapeutic intervention, and treatment strategies currently in development.
Collapse
Affiliation(s)
- Ranjith Babu
- Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | | | | | | | | |
Collapse
|
13
|
Yang AL, Zhou HJ, Lin Y, Luo JK, Cui HJ, Tang T, Yang QD. Thrombin promotes the expression of thrombospondin-1 and ‐2 in a rat model of intracerebral hemorrhage. J Neurol Sci 2012; 323:141-6. [DOI: 10.1016/j.jns.2012.09.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2012] [Revised: 09/05/2012] [Accepted: 09/07/2012] [Indexed: 10/27/2022]
|
14
|
Bodmer D, Vaughan KA, Zacharia BE, Hickman ZL, Connolly ES. The Molecular Mechanisms that Promote Edema After Intracerebral Hemorrhage. Transl Stroke Res 2012; 3:52-61. [PMID: 24323861 DOI: 10.1007/s12975-012-0162-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2012] [Revised: 03/19/2012] [Accepted: 03/21/2012] [Indexed: 01/01/2023]
Abstract
Intracerebral hemorrhage (ICH) is a devastating type of stroke with no effective therapies. Clinical advances in ICH treatment are limited by an incomplete understanding of the molecular mechanisms responsible for secondary injury and poor outcome. Increasing evidence suggests that cerebral edema is a major contributor to secondary injury and poor outcome in ICH. ICH activates specific signaling pathways that promote edema and damage neuronal tissue. By increasing our understanding of these pathways, we may be able to target them pharmaceutically to reduce edema in ICH patients. In this review, we focus on three major signaling pathways that promote edema after ICH: (1) the coagulation cascade and thrombin, (2) the inflammatory response and matrix metalloproteinases, and (3) the complement cascade and hemoglobin toxicity. We will describe the experimental evidence that confirms these pathways promote edema in ICH, discuss potential targets for new therapies, and comment on important directions for future research.
Collapse
Affiliation(s)
- Daniel Bodmer
- Department of Neurological Surgery, The Neurological Institute, Columbia University College of Physicians and Surgeons, New York, NY, 10032, USA
| | | | | | | | | |
Collapse
|
15
|
Zhou ZH, Qu F, Zhang CD. Systemic administration of argatroban inhibits protease-activated receptor-1 expression in perihematomal tissue in rats with intracerebral hemorrhage. Brain Res Bull 2011; 86:235-8. [PMID: 21803126 DOI: 10.1016/j.brainresbull.2011.07.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2011] [Revised: 07/13/2011] [Accepted: 07/14/2011] [Indexed: 10/18/2022]
Abstract
The present study investigated the role of thrombin in the expression of protease-activated receptor-1 (PAR-1), and the effect of argatroban (Arg) a direct thrombin inhibitor, on PAR-1 expression in perihematomal tissue with intracerebral hemorrhage (ICH). For these experiments 90 rats were divided into 5 groups: sham, ICH, argatroban-treated ICH (ICH+Arg), thrombin (TM) and argatroban-treated thrombin (TM+Arg). The ICH model or thrombin injection models were established by injecting autologous blood or thrombin, respectively. Rats in TM+Arg and ICH+Arg groups were administered argatroban (0.9mg/kg) after models were established for 3h and 12h, intraperitoneally. All rats were killed to harvest brains after models were established for 24h. The levels of PAR-1 protein and PAR-1 mRNA expression were detected by Western blot and RT-PCR, respectively. Brain water content was also measured. Our results showed that the levels of PAR-1 protein or PAR-1 mRNA in ICH and TM groups were up-regulated compared to that observed for the sham group; while the levels observed in ICH+Arg group and TM+Arg group were significantly lower than that observed for the ICH group and TM group (P<0.01 or P<0.05). The intraperitoneal administration argatroban also significantly reduced edema in ICH or TM group (P<0.05). Our observations suggested that the production of thrombin following ICH play a key role in the up-regulation of PAR-1 and anti-PAR-1 by systemic administration of argatroban, and may be a potential strategy for ICH therapy.
Collapse
Affiliation(s)
- Zhong-He Zhou
- Department of Neurology, General Hospital of Shenyang Military Command, Shenyang, China
| | | | | |
Collapse
|
16
|
Wu H, Zhang Z, Li Y, Zhao R, Li H, Song Y, Qi J, Wang J. Time course of upregulation of inflammatory mediators in the hemorrhagic brain in rats: correlation with brain edema. Neurochem Int 2010; 57:248-53. [PMID: 20541575 DOI: 10.1016/j.neuint.2010.06.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2010] [Revised: 05/24/2010] [Accepted: 06/02/2010] [Indexed: 11/29/2022]
Abstract
Intracerebral hemorrhage (ICH) can cause secondary brain damage through inflammation-related pathways. Thrombin and one of its receptors, protease activated receptor-1 (PAR-1); matrix metalloproteinase (MMP)-9; and aquaporin (AQP)-4 are stroke-related inflammatory mediators that have been implicated in ICH pathology. To further characterize the inflammatory response after ICH, we studied the temporal profile of the expression of these inflammatory mediators and assessed their potential correlation with brain edema formation after brain hemorrhage in rats. ICH was modeled by infusing autologous blood into the striatum. Then mRNA and protein expression was assessed over the course of 5 days. We found that the mRNA and/or protein expression of thrombin, PAR-1, AQP-4, and MMP-9 was upregulated between 2h and 5 days after ICH. Each reached a maximal level at day 2, except for AQP-4 protein, which peaked at day 5. Brain water content after ICH presented a similar trend; it was increased at 2h, peaked at day 2, and then decreased but remained elevated at day 5. Our data provide novel evidence that upregulation of these selected inflammatory mediators occurs very early and persists for several days after ICH, and that temporal patterns of expression of thrombin and AQP-4 are associated with brain edema formation. These findings have important implications for efforts to reduce secondary brain damage after ICH.
Collapse
Affiliation(s)
- He Wu
- Department of Pathology, First Clinical Hospital, Harbin Medical University, Harbin 150001, PR China.
| | | | | | | | | | | | | | | |
Collapse
|