1
|
Mao N, Zhang M, Shen M, Yuan J, Lin Z. Research progress on ferroptosis in cerebral hemorrhage. Biomed Pharmacother 2025; 185:117932. [PMID: 40015051 DOI: 10.1016/j.biopha.2025.117932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 02/06/2025] [Accepted: 02/15/2025] [Indexed: 03/01/2025] Open
Abstract
The pathophysiology of intracerebral hemorrhage (ICH) is complex and can cause variable degrees of cell death. Recently, ferroptosis, an emerging cell death mechanism, has garnered significant attention in cerebral hemorrhage disorder. This study aimed to examine iron mortality after cerebral hemorrhage and current targets for potential therapeutic interventions. We specifically focused on iron metabolism abnormalities, lipid peroxidation, and related neuroinflammation and introduced molecular mechanisms, including transcription factors, to gain a better understanding of the underlying mechanisms of ferroptosis and investigate possible therapeutic options for ICH.
Collapse
Affiliation(s)
- Niping Mao
- Department of Neonatology, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Min Zhang
- Department of Neonatology, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ming Shen
- Department of Neonatology, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Junhui Yuan
- Department of Neonatology, Wenling maternal and child health care hospital, Wenling, Zhejiang, China.
| | - Zhenlang Lin
- Department of Neonatology, the Second School of Medicine, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Perinatal Medicine of Wenzhou, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China; Zhejiang Provincial Clinical Research Center for Pediatric Disease, the Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China.
| |
Collapse
|
2
|
Wang X, Tian W, Zhao Y, Yang Y, Deng L. Systemic immune inflammation index and system inflammation response index on the third postoperative day predict poor prognosis of aneurysmal subarachnoid hemorrhage patients. Medicine (Baltimore) 2024; 103:e37818. [PMID: 38640265 PMCID: PMC11030023 DOI: 10.1097/md.0000000000037818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 03/09/2024] [Accepted: 03/15/2024] [Indexed: 04/21/2024] Open
Abstract
The inflammatory response is involved in the progression of aneurysmal subarachnoid hemorrhage (aSAH). We sought to investigate the relationships of inflammatory indicators including blood cell counts and the ratios of different blood cells counts with the prognosis of aSAH patients. We performed a retrospective study including 140 patients with aSAH and aneurysm surgeries. The relationships of neutrophils, lymphocytes, monocytes, platelets, systemic immune inflammation index (SII), system inflammation response index (SIRI), neutrophil-lymphocyte ratio and platelet-lymphocyte ratio with prognosis were investigated by univariable analysis and multivariable logistic regression model. The patient with Modified Rankin Scale (mRS) score<3 was defined as having a good prognosis, while with mRS score ≥3 was defined as having a poor prognosis. Among 140 patients included, there were 108 cases with good prognosis and 32 cases with poor prognosis after follow-up. On the 3rd postoperative day, the neutrophils counts, SIRI level and SII level in cases with poor prognosis were significantly higher than cases with good prognosis, P < .05. After adjusting for baseline differences in Hunt-Hess grade, Glasgow Coma Scale score, combination with intraventricular hemorrhage and maximum diameter of aneurysm, the levels of SIRI (odds ratio = 3.968, 95% CI: 1.432-10.992, P = .008) and SII (odds ratio = 3.313, 95% CI: 1.029-10.665, P = .045) on the 3rd postoperative day could predict poor prognosis. SII and SIRI on the 3rd postoperative day could independently predict the poor prognosis in aSAH. However, the cutoff values for predicting prognosis needs to be validated in larger-sample studies.
Collapse
Affiliation(s)
- Xian Wang
- Department of Pharmacy, The First Affiliated Hospital of Yangtze University, Jingzhou, China
| | - Wei Tian
- The Neurointensive Care Unit, The First Affiliated Hospital of Yangtze University, Jingzhou, China
| | - Yongfeng Zhao
- Department of Hematology, The First Affiliated Hospital of Yangtze University, Jingzhou, China
| | - Yong Yang
- The Neurointensive Care Unit, The First Affiliated Hospital of Yangtze University, Jingzhou, China
| | - Li Deng
- Medical Department, The First Affiliated Hospital of Yangtze University, Jingzhou, China
| |
Collapse
|
3
|
Jin J, Duan J, Du L, Xing W, Peng X, Zhao Q. Inflammation and immune cell abnormalities in intracranial aneurysm subarachnoid hemorrhage (SAH): Relevant signaling pathways and therapeutic strategies. Front Immunol 2022; 13:1027756. [PMID: 36505409 PMCID: PMC9727248 DOI: 10.3389/fimmu.2022.1027756] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 10/31/2022] [Indexed: 11/25/2022] Open
Abstract
Intracranial aneurysm subarachnoid hemorrhage (SAH) is a cerebrovascular disorder associated with high overall mortality. Currently, the underlying mechanisms of pathological reaction after aneurysm rupture are still unclear, especially in the immune microenvironment, inflammation, and relevant signaling pathways. SAH-induced immune cell population alteration, immune inflammatory signaling pathway activation, and active substance generation are associated with pro-inflammatory cytokines, immunosuppression, and brain injury. Crosstalk between immune disorders and hyperactivation of inflammatory signals aggravated the devastating consequences of brain injury and cerebral vasospasm and increased the risk of infection. In this review, we discussed the role of inflammation and immune cell responses in the occurrence and development of aneurysm SAH, as well as the most relevant immune inflammatory signaling pathways [PI3K/Akt, extracellular signal-regulated kinase (ERK), hypoxia-inducible factor-1α (HIF-1α), STAT, SIRT, mammalian target of rapamycin (mTOR), NLRP3, TLR4/nuclear factor-κB (NF-κB), and Keap1/nuclear factor (erythroid-derived 2)-like 2 (Nrf2)/ARE cascades] and biomarkers in aneurysm SAH. In addition, we also summarized potential therapeutic drugs targeting the aneurysm SAH immune inflammatory responses, such as nimodipine, dexmedetomidine (DEX), fingolimod, and genomic variation-related aneurysm prophylactic agent sunitinib. The intervention of immune inflammatory responses and immune microenvironment significantly reduces the secondary brain injury, thereby improving the prognosis of patients admitted to SAH. Future studies should focus on exploring potential immune inflammatory mechanisms and developing additional therapeutic strategies for precise aneurysm SAH immune inflammatory regulation and genomic variants associated with aneurysm formation.
Collapse
Affiliation(s)
- Jing Jin
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China,Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jian Duan
- Department of Cerebrovascular Disease, Suining Central Hospital, Suining, Sichuan, China
| | - Leiya Du
- 4Department of Oncology, The Second People Hospital of Yibin, Yibin, Sichuan, China
| | - Wenli Xing
- Department of Cerebrovascular Disease, Suining Central Hospital, Suining, Sichuan, China
| | - Xingchen Peng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China,*Correspondence: Qijie Zhao, ; Xingchen Peng,
| | - Qijie Zhao
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan, China,*Correspondence: Qijie Zhao, ; Xingchen Peng,
| |
Collapse
|
4
|
Molecular, Pathological, Clinical, and Therapeutic Aspects of Perihematomal Edema in Different Stages of Intracerebral Hemorrhage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:3948921. [PMID: 36164392 PMCID: PMC9509250 DOI: 10.1155/2022/3948921] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 08/17/2022] [Accepted: 09/03/2022] [Indexed: 02/07/2023]
Abstract
Acute intracerebral hemorrhage (ICH) is a devastating type of stroke worldwide. Neuronal destruction involved in the brain damage process caused by ICH includes a primary injury formed by the mass effect of the hematoma and a secondary injury induced by the degradation products of a blood clot. Additionally, factors in the coagulation cascade and complement activation process also contribute to secondary brain injury by promoting the disruption of the blood-brain barrier and neuronal cell degeneration by enhancing the inflammatory response, oxidative stress, etc. Although treatment options for direct damage are limited, various strategies have been proposed to treat secondary injury post-ICH. Perihematomal edema (PHE) is a potential surrogate marker for secondary injury and may contribute to poor outcomes after ICH. Therefore, it is essential to investigate the underlying pathological mechanism, evolution, and potential therapeutic strategies to treat PHE. Here, we review the pathophysiology and imaging characteristics of PHE at different stages after acute ICH. As illustrated in preclinical and clinical studies, we discussed the merits and limitations of varying PHE quantification protocols, including absolute PHE volume, relative PHE volume, and extension distance calculated with images and other techniques. Importantly, this review summarizes the factors that affect PHE by focusing on traditional variables, the cerebral venous drainage system, and the brain lymphatic drainage system. Finally, to facilitate translational research, we analyze why the relationship between PHE and the functional outcome of ICH is currently controversial. We also emphasize promising therapeutic approaches that modulate multiple targets to alleviate PHE and promote neurologic recovery after acute ICH.
Collapse
|
5
|
Zhang W, Li Y, Li F, Ling L. Sphingosine-1-phosphate receptor modulators in stroke treatment. J Neurochem 2022; 162:390-403. [PMID: 35943290 DOI: 10.1111/jnc.15685] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2022] [Revised: 06/30/2022] [Accepted: 08/02/2022] [Indexed: 11/30/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a bioactive lysophospholipid that can influence a broad range of biological processes through its binding to five distinct G protein-coupled receptors. S1P receptor modulators are a new group of immunosuppressive agents currently used in the immunotherapy of multiple sclerosis. Inflammation following stroke may exacerbate injury. Given that S1P signaling is linked to multiple immune processes, therapies targeting the S1P axis may be suitable for treating stroke. In this review, we outline S1P metabolism and S1P receptors, discuss the mechanisms of action of S1P receptor modulators in lymphocyte migration and their direct action on cells of the central nervous system, and provide a concise summary of the efficacy of S1P receptor modulators in animal studies and clinical trials on treatments for stroke.
Collapse
Affiliation(s)
- Wanzhou Zhang
- Department of Neurology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Yudi Li
- Department of Neurology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China.,The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Fangming Li
- Department of Neurology, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen, Guangdong, China
| | - Li Ling
- Department of Neurology, Shenzhen Hospital, Southern Medical University, Shenzhen, Guangdong, China
| |
Collapse
|
6
|
Diaz Diaz AC, Malone K, Shearer JA, Moore AC, Waeber C. Preclinical Evaluation of Fingolimod in Rodent Models of Stroke With Age or Atherosclerosis as Comorbidities. Front Pharmacol 2022; 13:920449. [PMID: 35910379 PMCID: PMC9326401 DOI: 10.3389/fphar.2022.920449] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/23/2022] [Indexed: 01/22/2023] Open
Abstract
Preclinical data indicate that fingolimod improves outcome post-ischaemia. This study used a rigorous study design in normal male C57BL/6JOlaHsd mice and in mice with common stroke comorbidities to further evaluate the translational potential of fingolimod. Stroke was induced via middle cerebral artery electrocoagulation in 8–9-week old mice (young mice), 18 month old mice (aged mice), and in high-fat diet-fed 22-week old ApoE−/− mice (hyperlipidaemic mice). Recovery was evaluated using motor behavioural tests 3 and 7 days after stroke. Tissue damage was evaluated at 7 days. A lower dose of fingolimod, 0.5 mg/kg, but not 1 mg/kg, increased lesion size but decreased ipsilateral brain atrophy in younger mice, without an effect on behavioural outcomes. Fingolimod-treated aged mice showed a significant improvement over saline-treated mice in the foot fault test at 7 days. Fingolimod-treated hyperlipidaemic mice showed a decreased infarct size but no difference in behavioural performance. Increasing fingolimod treatment time to 10 days showed no benefit in young mice. Pooled data showed that fingolimod improved performance in the foot fault test. Flow cytometry studies showed that fingolimod had marked effects on T cell frequencies in various tissues. The results show that the effects of fingolimod in stroke are less robust than the existing literature might indicate and may depend on the inflammatory status of the animals.
Collapse
Affiliation(s)
- Andrea C. Diaz Diaz
- School of Pharmacy, University College Cork, Cork, Ireland
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | - Kyle Malone
- School of Pharmacy, University College Cork, Cork, Ireland
- Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| | | | - Anne C. Moore
- Department of Pharmacology, University College Cork, Cork, Ireland
| | - Christian Waeber
- Department of Pharmacology, University College Cork, Cork, Ireland
- *Correspondence: Christian Waeber,
| |
Collapse
|
7
|
Kuai F, Zhou J, Qiu Y, Gao Y. FTY720 Attenuates Cerebral Vasospasm After Subarachnoid Hemorrhage Through the PI3K/AKT/eNOS and NF- κB Pathways in Rats. Neurol India 2022; 70:1517-1524. [PMID: 36076653 DOI: 10.4103/0028-3886.355128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Cerebral vasospasm (CVS) is a major complication of subarachnoid hemorrhage (SAH). Inflammation and nitric oxide (NO) have become increasingly recognized as key pathogenic contributors to brain injury in this condition. We aimed to examine the role of FTY720 in CVS after SAH. Endovascular perforation was used to establish an SAH model. Seventy-five male Sprague-Dawley rats were randomly divided into five groups: sham, sham + FTY720, SAH + saline, and two SAH + FTY720 (0.5 and 1 mg/kg) groups. The results showed that FTY720 treatment in both the surgery and nonsurgery groups decreased the counts of leukocytes and lymphocytes 72 hours after SAH. TNF-α (tumor necrosis factor alpha) and IL-1β (interleukin 1 beta) in both the cerebrospinal fluid (CSF) and the hippocampus were decreased, and the NF-κB (nuclear factor kappa B) pathway was inhibited. The levels of apoptotic proteins were downregulated. FTY720 promoted NO generation by activating the PI3K/AKT/eNOS pathway. CVS and neurological deficits in the SAH rats were ameliorated after FTY720 treatment. Compared with the sham-only animals, FTY720 treatment in the nonsurgery group did not increase mortality. These results indicated that FTY720 could alleviate CVS due to its anti-inflammatory and antiapoptosis effects and the promotion of NO generation. FTY720 may be effective in the clinical treatment of SAH patients.
Collapse
Affiliation(s)
- Feng Kuai
- Department of Geriatrics, The First People's Hospital of Yancheng, The Fourth Affiliated Hospital of Nantong University, Yancheng, China
| | - Jianping Zhou
- Department of Geriatrics, The First People's Hospital of Yancheng, The Fourth Affiliated Hospital of Nantong University, Yancheng, China
| | - Yuchen Qiu
- Department of Geriatrics, The First People's Hospital of Yancheng, The Fourth Affiliated Hospital of Nantong University, Yancheng, China
| | - Yang Gao
- Neurology, The First People's Hospital of Yancheng, The Fourth Affiliated Hospital of Nantong University, Yancheng, China
| |
Collapse
|
8
|
Pournajaf S, Dargahi L, Javan M, Pourgholami MH. Molecular Pharmacology and Novel Potential Therapeutic Applications of Fingolimod. Front Pharmacol 2022; 13:807639. [PMID: 35250559 PMCID: PMC8889014 DOI: 10.3389/fphar.2022.807639] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/31/2022] [Indexed: 12/14/2022] Open
Abstract
Fingolimod is a well-tolerated, highly effective disease-modifying therapy successfully utilized in the management of multiple sclerosis. The active metabolite, fingolimod-phosphate, acts on sphingosine-1-phosphate receptors (S1PRs) to bring about an array of pharmacological effects. While being initially recognized as a novel agent that can profoundly reduce T-cell numbers in circulation and the CNS, thereby suppressing inflammation and MS, there is now rapidly increasing knowledge on its previously unrecognized molecular and potential therapeutic effects in diverse pathological conditions. In addition to exerting inhibitory effects on sphingolipid pathway enzymes, fingolimod also inhibits histone deacetylases, transient receptor potential cation channel subfamily M member 7 (TRMP7), cytosolic phospholipase A2α (cPLA2α), reduces lysophosphatidic acid (LPA) plasma levels, and activates protein phosphatase 2A (PP2A). Furthermore, fingolimod induces apoptosis, autophagy, cell cycle arrest, epigenetic regulations, macrophages M1/M2 shift and enhances BDNF expression. According to recent evidence, fingolimod modulates a range of other molecular pathways deeply rooted in disease initiation or progression. Experimental reports have firmly associated the drug with potentially beneficial therapeutic effects in immunomodulatory diseases, CNS injuries, and diseases including Alzheimer's disease (AD), Parkinson's disease (PD), epilepsy, and even cancer. Attractive pharmacological effects, relative safety, favorable pharmacokinetics, and positive experimental data have collectively led to its testing in clinical trials. Based on the recent reports, fingolimod may soon find its way as an adjunct therapy in various disparate pathological conditions. This review summarizes the up-to-date knowledge about molecular pharmacology and potential therapeutic uses of fingolimod.
Collapse
Affiliation(s)
- Safura Pournajaf
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Leila Dargahi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Javan
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | | |
Collapse
|
9
|
Zhang L, Sui R, Zhang L. Fingolimod protects against cerebral ischemia reperfusion injury in rats by reducing inflammatory cytokines and inhibiting the activation of p38 MAPK and NF-κB signaling pathways. Neurosci Lett 2021; 771:136413. [PMID: 34942319 DOI: 10.1016/j.neulet.2021.136413] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 11/29/2021] [Accepted: 12/17/2021] [Indexed: 11/26/2022]
Abstract
Fingolimod (FTY720) is a sphingosine 1-phosphate (S1P) receptor agonist. Here, to understand biological activity of FTY720 pretreatment and post-treatment on cerebral ischemia reperfusion injury (CIRI), rat transient middle cerebral artery occlusion/reperfusion (tMCAO/R) model was generated. Neurological deficit scoring was assessed after tMCAO/R. Four groups were established including sham-operated control group, operated group, and two FTY720-treated groups. Neuron damage was observed by Nissl staining. Gene expression was measured using qPCR and western blot analysis. Tumor necrosis factor α (TNF-α), interleukin 1β (IL-1β) and interleukin 6 (IL-6) levels were evaluated by enzyme-linked immunosorbent assay (ELISA). We uncovered that neurological score in two FTY720-treated groups was significantly lower than that in the operated group. FTY720 pretreatment or posttreatment groups had a significantly increased number of Nissl bodies in cerebral cortex as compared with the operated group, indicating that FTY720 administration reduced neuronal damage. Besides, FTY720 posttreatment improved memory impairment induced by tMCAO/R. In addition, IL-1β, IL-6, and TNF-α levels in the cerebral cortex and hippocampus of two FTY720-treated groups were significantly decreased in comparison to the operated group, showing that FTY720 could reduce the release of inflammatory cytokines in brain tissue. Furthermore, phosphorylation of p38MAPK and NF-κB pathway-related molecules in ischemic brain tissues of FTY720 group were markedly down-regulated compared to the operated group. Together, FTY720 pretreatment or posttreatment improved the neurological deficit of middle cerebral ischemia/reperfusion rat model and reduced neuronal damage by decreasing the levels of inflammatory cytokines and attenuating the phosphorylation levels of p38MAPK and NF-κB pathway-associated molecules. FTY720 exhibits neuroprotective effects against ischemic reperfusion injury in rats.
Collapse
Affiliation(s)
- Ling Zhang
- Department of Neurology, The First Affiliated Hospital of Jinzhou Medical University, PR China
| | - Rubo Sui
- Department of Neurology, The First Affiliated Hospital of Jinzhou Medical University, PR China
| | - Lei Zhang
- Nursing College of Jinzhou Medical University, Jinzhou, Liaoning 121001, PR China.
| |
Collapse
|
10
|
Jha RM, Raikwar SP, Mihaljevic S, Casabella AM, Catapano JS, Rani A, Desai S, Gerzanich V, Simard JM. Emerging therapeutic targets for cerebral edema. Expert Opin Ther Targets 2021; 25:917-938. [PMID: 34844502 PMCID: PMC9196113 DOI: 10.1080/14728222.2021.2010045] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 11/20/2021] [Indexed: 01/04/2023]
Abstract
INTRODUCTION Cerebral edema is a key contributor to death and disability in several forms of brain injury. Current treatment options are limited, reactive, and associated with significant morbidity. Targeted therapies are emerging based on a growing understanding of the molecular underpinnings of cerebral edema. AREAS COVERED We review the pathophysiology and relationships between different cerebral edema subtypes to provide a foundation for emerging therapies. Mechanisms for promising molecular targets are discussed, with an emphasis on those advancing in clinical trials, including ion and water channels (AQP4, SUR1-TRPM4) and other proteins/lipids involved in edema signaling pathways (AVP, COX2, VEGF, and S1P). Research on novel treatment modalities for cerebral edema [including recombinant proteins and gene therapies] is presented and finally, insights on reducing secondary injury and improving clinical outcome are offered. EXPERT OPINION Targeted molecular strategies to minimize or prevent cerebral edema are promising. Inhibition of SUR1-TRPM4 (glyburide/glibenclamide) and VEGF (bevacizumab) are currently closest to translation based on advances in clinical trials. However, the latter, tested in glioblastoma multiforme, has not demonstrated survival benefit. Research on recombinant proteins and gene therapies for cerebral edema is in its infancy, but early results are encouraging. These newer modalities may facilitate our understanding of the pathobiology underlying cerebral edema.
Collapse
Affiliation(s)
- Ruchira M. Jha
- Department of Neurology, Barrow Neurological Institute and St. Joseph’s Hospital and Medical Center, Phoenix, AZ, USA
- Department of Neurobiology, Barrow Neurological Institute and St. Joseph’s Hospital and Medical Center, Phoenix, AZ, USA
- Department of Neurosurgery, Barrow Neurological Institute and St. Joseph’s Hospital and Medical Center, Phoenix, AZ, USA
| | - Sudhanshu P. Raikwar
- Department of Neurobiology, Barrow Neurological Institute and St. Joseph’s Hospital and Medical Center, Phoenix, AZ, USA
| | - Sandra Mihaljevic
- Department of Neurobiology, Barrow Neurological Institute and St. Joseph’s Hospital and Medical Center, Phoenix, AZ, USA
| | | | - Joshua S. Catapano
- Department of Neurosurgery, Barrow Neurological Institute and St. Joseph’s Hospital and Medical Center, Phoenix, AZ, USA
| | - Anupama Rani
- Department of Neurobiology, Barrow Neurological Institute and St. Joseph’s Hospital and Medical Center, Phoenix, AZ, USA
| | - Shashvat Desai
- Department of Neurology, Barrow Neurological Institute and St. Joseph’s Hospital and Medical Center, Phoenix, AZ, USA
| | - Volodymyr Gerzanich
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore MD, USA
| | - J. Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore MD, USA
- Department of Pathology, University of Maryland School of Medicine, Baltimore MD, USA
- Department of Physiology, University of Maryland School of Medicine, Baltimore MD, USA
| |
Collapse
|
11
|
Molecular Mechanisms of Neuroimmune Crosstalk in the Pathogenesis of Stroke. Int J Mol Sci 2021; 22:ijms22179486. [PMID: 34502395 PMCID: PMC8431165 DOI: 10.3390/ijms22179486] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/26/2021] [Accepted: 08/28/2021] [Indexed: 12/21/2022] Open
Abstract
Stroke disrupts the homeostatic balance within the brain and is associated with a significant accumulation of necrotic cellular debris, fluid, and peripheral immune cells in the central nervous system (CNS). Additionally, cells, antigens, and other factors exit the brain into the periphery via damaged blood–brain barrier cells, glymphatic transport mechanisms, and lymphatic vessels, which dramatically influence the systemic immune response and lead to complex neuroimmune communication. As a result, the immunological response after stroke is a highly dynamic event that involves communication between multiple organ systems and cell types, with significant consequences on not only the initial stroke tissue injury but long-term recovery in the CNS. In this review, we discuss the complex immunological and physiological interactions that occur after stroke with a focus on how the peripheral immune system and CNS communicate to regulate post-stroke brain homeostasis. First, we discuss the post-stroke immune cascade across different contexts as well as homeostatic regulation within the brain. Then, we focus on the lymphatic vessels surrounding the brain and their ability to coordinate both immune response and fluid homeostasis within the brain after stroke. Finally, we discuss how therapeutic manipulation of peripheral systems may provide new mechanisms to treat stroke injury.
Collapse
|
12
|
Diaz Diaz AC, Shearer JA, Malone K, Waeber C. Acute Treatment With Fingolimod Does Not Confer Long-Term Benefit in a Mouse Model of Intracerebral Haemorrhage. Front Pharmacol 2021; 11:613103. [PMID: 33488389 PMCID: PMC7821021 DOI: 10.3389/fphar.2020.613103] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 11/23/2020] [Indexed: 01/01/2023] Open
Abstract
Intracerebral haemorrhage (ICH) has no specific treatment, but accounts for up to 15% of all strokes and has the highest mortality. Fingolimod (FTY720) is an immunomodulator approved for the management of multiple sclerosis, with abundant evidence of efficacy in experimental ischemic stroke, and more limited evidence in experimental ICH. The goal of this study was to confirm the efficacy of fingolimod in experimental ICH using rigorous and statistically well-powered studies. ICH was induced in C57BL/6JOlaHsd male and female mice by intrastriatal bacterial collagenase injection. Fingolimod (0.5 mg/kg) or saline was administered intraperitoneally after 0.5, 24 and 72 h, in a randomized and blinded manner. Functional improvement with cylinder, wire hanging, and foot fault tests was evaluated one and two weeks later. Lesion volume and hemispheric atrophy were quantified at the 14-day endpoint. There was a higher mortality in saline-treated females compared to fingolimod-treated females and saline-treated males. There was no treatment- or gender-related difference in the behavioural tests. Histological outcome measures did not differ between any of the groups. These results, contrasting with those of previous studies of fingolimod in experimental ICH, emphasize the importance of rigorous testing of this agent in models more representative of the clinical situation.
Collapse
Affiliation(s)
| | | | - Kyle Malone
- School of Pharmacy, University College Cork, Cork, Ireland
| | - Christian Waeber
- School of Pharmacy, University College Cork, Cork, Ireland.,Department of Pharmacology and Therapeutics, University College Cork, Cork, Ireland
| |
Collapse
|
13
|
Chua XY, Ho LTY, Xiang P, Chew WS, Lam BWS, Chen CP, Ong WY, Lai MKP, Herr DR. Preclinical and Clinical Evidence for the Involvement of Sphingosine 1-Phosphate Signaling in the Pathophysiology of Vascular Cognitive Impairment. Neuromolecular Med 2020; 23:47-67. [PMID: 33180310 DOI: 10.1007/s12017-020-08632-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Accepted: 11/03/2020] [Indexed: 02/07/2023]
Abstract
Sphingosine 1-phosphates (S1Ps) are bioactive lipids that mediate a diverse range of effects through the activation of cognate receptors, S1P1-S1P5. Scrutiny of S1P-regulated pathways over the past three decades has identified important and occasionally counteracting functions in the brain and cerebrovascular system. For example, while S1P1 and S1P3 mediate proinflammatory effects on glial cells and directly promote endothelial cell barrier integrity, S1P2 is anti-inflammatory but disrupts barrier integrity. Cumulatively, there is significant preclinical evidence implicating critical roles for this pathway in regulating processes that drive cerebrovascular disease and vascular dementia, both being part of the continuum of vascular cognitive impairment (VCI). This is supported by clinical studies that have identified correlations between alterations of S1P and cognitive deficits. We review studies which proposed and evaluated potential mechanisms by which such alterations contribute to pathological S1P signaling that leads to VCI-associated chronic neuroinflammation and neurodegeneration. Notably, S1P receptors have divergent but overlapping expression patterns and demonstrate complex interactions. Therefore, the net effect produced by S1P represents the cumulative contributions of S1P receptors acting additively, synergistically, or antagonistically on the neural, vascular, and immune cells of the brain. Ultimately, an optimized therapeutic strategy that targets S1P signaling will have to consider these complex interactions.
Collapse
Affiliation(s)
- Xin Ying Chua
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Leona T Y Ho
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119260, Singapore
| | - Ping Xiang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Wee Siong Chew
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Brenda Wan Shing Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Christopher P Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Kent Ridge, Singapore
| | - Wei-Yi Ong
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119260, Singapore
- Neurobiology Programme, Life Sciences Institute, National University of Singapore, Singapore, 119260, Singapore
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Memory Aging and Cognition Centre, National University Health System, Kent Ridge, Singapore.
| | - Deron R Herr
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Department of Biology, San Diego State University, San Diego, CA, USA.
- American University of Health Sciences, Long Beach, CA, USA.
| |
Collapse
|
14
|
Kim H, Lee JE, Yoo HJ, Sung JH, Yang SH. Effect of Pioglitazone on Perihematomal Edema in Intracerebral Hemorrhage Mouse Model by Regulating NLRP3 Expression and Energy Metabolism. J Korean Neurosurg Soc 2020; 63:689-697. [PMID: 33105536 PMCID: PMC7671775 DOI: 10.3340/jkns.2020.0056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 05/26/2020] [Indexed: 12/21/2022] Open
Abstract
Objective Cerebral edema is the predominant mechanism of secondary inflammation after intracerebral hemorrhage (ICH). Pioglitazone, peroxisome proliferator-activated receptor gamma agonist has been shown to play a role in regulation of central nervous system inflammation. Here, we examined the pharmacological effects of pioglitazone in an ICH mouse model and investigated its regulation on NLRP3 inflammasome and glucose metabolism.
Methods The ICH model was established in C57 BL/6 mice by the stereotactical inoculation of blood (30 µL) into the right frontal lobe. The treatment group was administered i.p. pioglitazone (20 mg/kg) for 1, 3, and 6 days. The control group was administered i.p. phosphate-buffered saline for 1, 3, and 6 days. We investigated brain water contents, NLRP3 expression, and changes in the metabolites in the ICH model using liquid chromatography-tandem mass spectrometry.
Results On day 3, brain edema in the mice treated with pioglitazone was decreased more than that in the control group. Expression levels of NLRP3 in the ICH model treated with pioglitazone were decreased more than those of the control mice on days 3 and 7. The pioglitazone group showed higher levels of glycolytic metabolites than those in the ICH mice. Lactate production was increased in the ICH mice treated with pioglitazone.
Conclusion Our results demonstrated less brain swelling following ICH in mice treated with pioglitazone. Pioglitazone decreased NLRP3-related brain edema and increased anaerobic glycolysis, resulting in the production of lactate in the ICH mice model. NLRP3 might be a therapeutic target for ICH recovery.
Collapse
Affiliation(s)
- Hoon Kim
- Department of Neurosurgery, Bucheon St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jung Eun Lee
- Department of Neurosurgery, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hyun Ju Yoo
- Biomedical Research Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Jae Hoon Sung
- Department of Neurosurgery, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seung Ho Yang
- Department of Neurosurgery, Cell Death Disease Research Center, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
15
|
Yavuz A, Sezik M, Eris Yalcin S, Asci H, Ozmen O. Immunohistochemical analysis of protective effects of maternal fingolimod on the placenta and fetal lung and brain in chorioamnionitis-induced preterm birth rat model. Immunopharmacol Immunotoxicol 2020; 42:564-571. [PMID: 32892670 DOI: 10.1080/08923973.2020.1818771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
OBJECTIVES Fingolimod (FIN) is used for multiple sclerosis treatment and has potential antiapoptotic and anti-inflammatory effects. We aimed at expanding our knowledge on various immunohistochemical markers for elucidating the possible mechanisms of action of fingolimod in the placenta and fetal lung and brain. METHODS Sixteen pregnant rats were divided into four groups. On gestational day 17, lipopolysaccharide (LPS) was injected intraperitoneally to induce preterm fetal injury followed by intraperitoneal injection of fingolimod. Hysterotomy for preterm delivery was performed 6 h after fingolimod was injected. The study groups included (1) control, (2) LPS (1 mg/kg), (3) FIN (4 mg/kg), and (4) FIN + LPS. Fetal brain and lung and placenta samples were collected for histopathological examination. Moreover, fetal lungs (surfactant protein-A (SP-A), SP-B, SP-D, caspase-3, and caspase-8), fetal brains (interleukin-10, interleukin-1β, TNF-α, caspase-8, glial fibrillary acidic protein, vimentin, myelin basic protein, and receptor activator of nuclear factor kappa), and placenta tissues (interleukin-10, interleukin-1β, TNF-α, caspase-3, and caspase-8) were immunohistochemically evaluated. RESULTS Maternal fingolimod treatment led to attenuation of LPS-induced fetal brain, lung, and placental injury, as indicated by lower immunoexpression of inflammatory markers compared to LPS group (p < .0001 for all comparisons). CONCLUSION The findings of the present study confirm the neuroprotective effects of antenatally administered fingolimod, which also significantly improved preterm fetal lung injury and placental inflammation in LPS-exposed preterm pregnancies by possible antiapoptotic and anti-inflammatory effects.
Collapse
Affiliation(s)
- And Yavuz
- Department of Obstetrics and Gynecology, University of Health Sciences, Antalya Education and Research Hospital, Antalya, Turkey
| | - Mekin Sezik
- Department of Obstetrics and Gynecology, Süleyman Demirel University Faculty of Medicine, Isparta, Turkey
| | - Serenat Eris Yalcin
- Department of Obstetrics and Gynecology, University of Health Sciences, Antalya Education and Research Hospital, Antalya, Turkey
| | - Halil Asci
- Department of Pharmacology, Süleyman Demirel University Faculty of Medicine, Isparta, Turkey
| | - Ozlem Ozmen
- Department of Pathology, Burdur Mehmet Akif Ersoy University Faculty of Veterinary Medicine, Burdur, Turkey
| |
Collapse
|
16
|
Abstract
Rodents are the most widely used experimental animals in stroke research due to their similar vascular anatomy, high reproductive rates, and availability of transgenic models. However, the difficulties in assessing higher brain functions, such as cognition and memory, in rodents decrease the translational potential of these studies. In this review, we summarize commonly used motor/sensorimotor and cognition tests in rodent models of stroke. Specifically, we first briefly introduce the objective and procedure of each behavioral test. Next, we summarize the application of each test in both ischemic stroke and hemorrhagic stroke. Last, the advantages and disadvantages of these tests in assessing stroke outcome are discussed. This review summarizes commonly used behavioral tests in stroke studies and compares their applications in different stroke types.
Collapse
Affiliation(s)
- Jingsong Ruan
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, 240 W Green Street, Athens, GA, USA
| | - Yao Yao
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia, 240 W Green Street, Athens, GA, USA
| |
Collapse
|
17
|
Wang Y, Zhou S, Han Z, Yin D, Luo Y, Tian Y, Wang Z, Zhang J. Fingolimod administration improves neurological functions of mice with subarachnoid hemorrhage. Neurosci Lett 2020; 736:135250. [PMID: 32673690 DOI: 10.1016/j.neulet.2020.135250] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 06/30/2020] [Accepted: 07/12/2020] [Indexed: 12/01/2022]
Abstract
PURPOSE To investigate the brain protective effects of fingolimod on inflammatory response of SAH mice. METHODS We utilized an endovascular mouse perforation model of SAH. Mice were divided into three groups: sham group, SAH group and SAH + Fingolimod group. Mice received either saline or fingolimod (1 mg/kg) intraperitoneally 2 h after sham surgery or SAH. The modified neurological severity score (mNSS) and Morris water maze were respectively used to evaluate the influence of nerve function. Evens blue (EB) extravasation was used to detect the permeability of blood-brain barrier, and water content in brain tissue was also detected. Flow cytometry, ELISA kits and western blotting were used to detect inflammatory factors in brain tissue. RESULTS The results showed that compared with SAH group, after treatment, the delay time of locating the hidden platform was shorter. The mNSS results showed that fingolimod improved the behavior of SAH mice. In addition, fingolimod could reduce the water content in brain. Flow cytometry results showed that after 3 d of treatment, fingolimod significantly increased Treg cells and down-regulated NK cells. Western blotting results showed fingolimod inhibited the expression of inflammatory cytokines in brain tissue. ELISA kit results showed that fingolimod could down-regulate IL-6 and TNF-α and up-regulate IL-10 and TGF-β1 in serum. CONCLUSIONS Fingolimod could regulate the inflammatory response to alleviate SAH-induced brain damage and promote neurological recovery, which provides a new therapeutic strategy for SAH treatment.
Collapse
Affiliation(s)
- Yi Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education & Key Laboratory of Injuries, Variations and Regeneration of Nervous System, 154 Anshan Road, Tianjin, China.
| | - Shuai Zhou
- Department of ICU, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, 300052, China
| | - Zhenfeng Han
- Department of Neurosurgery, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education & Key Laboratory of Injuries, Variations and Regeneration of Nervous System, 154 Anshan Road, Tianjin, China
| | - Dongpei Yin
- Department of Neurosurgery, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education & Key Laboratory of Injuries, Variations and Regeneration of Nervous System, 154 Anshan Road, Tianjin, China
| | - Yuanbo Luo
- Department of Neurosurgery, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education & Key Laboratory of Injuries, Variations and Regeneration of Nervous System, 154 Anshan Road, Tianjin, China
| | - Ye Tian
- Department of Neurosurgery, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education & Key Laboratory of Injuries, Variations and Regeneration of Nervous System, 154 Anshan Road, Tianjin, China
| | - Zengguang Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education & Key Laboratory of Injuries, Variations and Regeneration of Nervous System, 154 Anshan Road, Tianjin, China
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, 154 Anshan Road, Tianjin, 300052, China; Tianjin Neurological Institute, Key Laboratory of Post-trauma Neuro-repair and Regeneration in Central Nervous System, Ministry of Education & Key Laboratory of Injuries, Variations and Regeneration of Nervous System, 154 Anshan Road, Tianjin, China.
| |
Collapse
|
18
|
The S1P-S1PR Axis in Neurological Disorders-Insights into Current and Future Therapeutic Perspectives. Cells 2020; 9:cells9061515. [PMID: 32580348 PMCID: PMC7349054 DOI: 10.3390/cells9061515] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 12/21/2022] Open
Abstract
Sphingosine 1-phosphate (S1P), derived from membrane sphingolipids, is a pleiotropic bioactive lipid mediator capable of evoking complex immune phenomena. Studies have highlighted its importance regarding intracellular signaling cascades as well as membrane-bound S1P receptor (S1PR) engagement in various clinical conditions. In neurological disorders, the S1P–S1PR axis is acknowledged in neurodegenerative, neuroinflammatory, and cerebrovascular disorders. Modulators of S1P signaling have enabled an immense insight into fundamental pathological pathways, which were pivotal in identifying and improving the treatment of human diseases. However, its intricate molecular signaling pathways initiated upon receptor ligation are still poorly elucidated. In this review, the authors highlight the current evidence for S1P signaling in neurodegenerative and neuroinflammatory disorders as well as stroke and present an array of drugs targeting the S1P signaling pathway, which are being tested in clinical trials. Further insights on how the S1P–S1PR axis orchestrates disease initiation, progression, and recovery may hold a remarkable potential regarding therapeutic options in these neurological disorders.
Collapse
|
19
|
Yao S, Li L, Sun X, Hua J, Zhang K, Hao L, Liu L, Shi D, Zhou H. FTY720 Inhibits MPP +-Induced Microglial Activation by Affecting NLRP3 Inflammasome Activation. J Neuroimmune Pharmacol 2019; 14:478-492. [PMID: 31069623 DOI: 10.1007/s11481-019-09843-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 02/25/2019] [Indexed: 12/14/2022]
Abstract
Parkinson's disease (PD) is characterized by the degeneration of dopaminergic neurons and excessive microglial activation in the substantia nigra pars compacta (SNpc). In the present study, we aimed to demonstrate the therapeutic effectiveness of the potent sphingosine-1-phosphate receptor antagonist fingolimod (FTY720) in an animal model of PD induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), and to identify the potential mechanisms underlying these therapeutic effects. C57BL/6J mice were orally administered FTY720 before subcutaneous injection of MPTP. Open-field and rotarod tests were performed to determine the therapeutic effect of FTY720. The damage to dopaminergic neurons and the production of monoamine neurotransmitters were assessed using immunohistochemistry, high-performance liquid chromatography, and flow cytometry. Immunofluorescence (CD68- positive) and enzyme-linked immunosorbent assay were used to analyze the activation of microglia, and the levels of activated signaling molecules were measured using Western blotting. Our findings indicated that FTY720 significantly attenuated MPTP-induced behavioral deficits, reduced the loss of dopaminergic neurons, and increased dopamine release. FTY720 directly inhibited MPTP-induced microglial activation in the SNpc, suppressed the production of interleukin (IL)-6, IL-1β, and tumor necrosis factor-α in BV-2 microglial cells treated with 1-methyl-4-phenylpyridinium (MPP+), and subsequently decreased apoptosis in SH-SY5Y neuroblastoma cells. Moreover, in MPP+-treated BV-2 cells and primary microglia, FTY720 treatment significantly attenuated the increases in the phosphorylation of PI3K/AKT/GSK-3β, reduced ROS generation and p65 activation, and also inhibited the activation of NLRP3 inflammasome and caspase-1. In conclusion, FTY720 may reduce PD progression by inhibiting NLRP3 inflammasome activation via its effects on ROS generation and p65 activation in microglia. These findings provide novel insights into the mechanisms underlying the therapeutic effects of FTY720, suggesting its potential as a novel therapeutic strategy against PD. Graphical Abstract FTY720 may reduce ROS production by inhibiting the PI3K/AKT/GSK-3β signaling pathway, while at the same time reducing p65 phosphorylation, thus decreasing NLRP3 inflammasome activation through these two pathways, ultimately reducing microglia activation-induced neuronal damage.
Collapse
Affiliation(s)
- Shu Yao
- Department of Immunology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, JS, China
| | - Longjun Li
- Department of Immunology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, JS, China
| | - Xin Sun
- Department of Immunology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, JS, China
| | - Jun Hua
- Department of Clinical Pharmacy, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No. 1, Fuhua Road, Futian District, Shenzhen, 518033, Guangdong, China
| | - Keqi Zhang
- Institute of Microscope Science and Technology, Ningbo Yongxin Optics Co. Ltd., 385 Mingzhu Road, Hi-tech Industry Park, Ningbo, 315040, China
| | - Li Hao
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - Lixin Liu
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK, S7N 5E5, Canada
| | - Dongyan Shi
- Department of Immunology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, JS, China.
| | - Hong Zhou
- Department of Immunology, Nanjing Medical University, 101 Longmian Avenue, Nanjing, 211166, JS, China.
- Department of Biotherapy, Second Affiliated Hospital, Nanjing Medical University, Nanjing, 210011, Jiangsu, China.
| |
Collapse
|
20
|
Clark AR, Ohlmeyer M. Protein phosphatase 2A as a therapeutic target in inflammation and neurodegeneration. Pharmacol Ther 2019; 201:181-201. [PMID: 31158394 PMCID: PMC6700395 DOI: 10.1016/j.pharmthera.2019.05.016] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 05/29/2019] [Indexed: 12/11/2022]
Abstract
Protein phosphatase 2A (PP2A) is a highly complex heterotrimeric enzyme that catalyzes the selective removal of phosphate groups from protein serine and threonine residues. Emerging evidence suggests that it functions as a tumor suppressor by constraining phosphorylation-dependent signalling pathways that regulate cellular transformation and metastasis. Therefore, PP2A-activating drugs (PADs) are being actively sought and investigated as potential novel anti-cancer treatments. Here we explore the concept that PP2A also constrains inflammatory responses through its inhibitory effects on various signalling pathways, suggesting that PADs may be effective in the treatment of inflammation-mediated pathologies.
Collapse
Affiliation(s)
- Andrew R Clark
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom.
| | | |
Collapse
|
21
|
Shao A, Zhu Z, Li L, Zhang S, Zhang J. Emerging therapeutic targets associated with the immune system in patients with intracerebral haemorrhage (ICH): From mechanisms to translation. EBioMedicine 2019; 45:615-623. [PMID: 31208948 PMCID: PMC6642355 DOI: 10.1016/j.ebiom.2019.06.012] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 06/01/2019] [Accepted: 06/08/2019] [Indexed: 12/28/2022] Open
Abstract
Intracranial haemorrhage (ICH) is a life-threatening type of stroke with high mortality, morbidity, and recurrence rates. However, no effective treatment has been established to improve functional outcomes in patients with ICH to date. Strategies targeting secondary brain injury are of great interest in both experimental and translational studies. The immune system is increasingly considered to be a crucial contributor to ICH-induced brain injury because it participates in multiple phases of ICH, from the early vascular rupture events to brain recovery. Various pathobiological processes that contribute to secondary brain injury closely interact with the immune system, such as brain oedema, neuroinflammation, and neuronal damage. Hence, we summarize the immune response to ICH and recent progress in treatments targeting the immune system in this review. The emerging therapeutic strategies that target the immune system after ICH are a particular focus and have been summarized.
Collapse
Affiliation(s)
- Anwen Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China.
| | - Zhiyuan Zhu
- Division of Neurosurgery, Department of Surgery, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong; Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lingfei Li
- Department of Neurology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shizhong Zhang
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China; The National Key Clinic Specialty, The Neurosurgery Institute of Guangdong Province, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Southern Medical University, Guangzhou, China.
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; Brain Research Institute, Zhejiang University, Hangzhou, China; Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, China
| |
Collapse
|
22
|
Salas-Perdomo A, Miró-Mur F, Gallizioli M, Brait VH, Justicia C, Meissner A, Urra X, Chamorro A, Planas AM. Role of the S1P pathway and inhibition by fingolimod in preventing hemorrhagic transformation after stroke. Sci Rep 2019; 9:8309. [PMID: 31165772 PMCID: PMC6549179 DOI: 10.1038/s41598-019-44845-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 05/24/2019] [Indexed: 12/31/2022] Open
Abstract
Hemorrhagic transformation (HT) is a complication of severe ischemic stroke after revascularization. Patients with low platelet counts do not receive reperfusion therapies due to high risk of HT. The immunomodulatory drug fingolimod attenuated HT after tissue plasminogen activator in a thromboembolic stroke model, but the underlying mechanism is unknown. Fingolimod acts on several sphingosine-1-phosphate (S1P) receptors, prevents lymphocyte trafficking to inflamed tissues, and affects brain and vascular cells. This study aimed to investigate changes in S1P-signaling in response to brain ischemia/reperfusion and the effects of the S1P receptor modulator fingolimod on HT. We studied brain expression of S1P signaling components, S1P concentration, and immune cell infiltration after ischemia/reperfusion in mice. We administered fingolimod after ischemia to wild-type mice, lymphocyte-deficient Rag2−/− mice, and mice with low platelet counts. Ischemia increased S1P-generating enzyme SphK1 mRNA, S1P concentration, and S1P receptor-1 (S1P1)+ T-cells in the brain. Fingolimod prevented lymphocyte infiltration, and attenuated the severity of HT in Rag2−/− mice but it was ineffective under thrombocytopenia. Fingolimod prevented β-catenin degradation but not Evans blue extravasation. Ischemia/reperfusion upregulates brain S1P signaling pathway, and fingolimod exerts local effects that attenuate HT. Although fingolimod seems to act on the brain tissue, it did not prevent blood-brain barrier leakage.
Collapse
Affiliation(s)
- Angélica Salas-Perdomo
- Departament d'Isquèmia Cerebral i Neurodegeneració, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain.,Àrea de Neurociències, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Francesc Miró-Mur
- Àrea de Neurociències, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Mattia Gallizioli
- Departament d'Isquèmia Cerebral i Neurodegeneració, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain.,Àrea de Neurociències, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Vanessa H Brait
- Àrea de Neurociències, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,The Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, Victoria, Australia
| | - Carles Justicia
- Departament d'Isquèmia Cerebral i Neurodegeneració, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain.,Àrea de Neurociències, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Anja Meissner
- Àrea de Neurociències, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Department of Experimental Medical Sciences & Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden
| | - Xabier Urra
- Àrea de Neurociències, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Functional Unit of Cerebrovascular Diseases, Hospital Clínic, Barcelona, Spain
| | - Angel Chamorro
- Àrea de Neurociències, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.,Functional Unit of Cerebrovascular Diseases, Hospital Clínic, Barcelona, Spain
| | - Anna M Planas
- Departament d'Isquèmia Cerebral i Neurodegeneració, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain. .,Àrea de Neurociències, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| |
Collapse
|
23
|
Yang Z, Dong S, Zheng Q, Zhang L, Tan X, Zou J, Yan B, Chen Y. FTY720 attenuates iron deposition and glial responses in improving delayed lesion and long-term outcomes of collagenase-induced intracerebral hemorrhage. Brain Res 2019; 1718:91-102. [PMID: 31039342 DOI: 10.1016/j.brainres.2019.04.031] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 04/25/2019] [Accepted: 04/27/2019] [Indexed: 12/23/2022]
Abstract
Most intracerebral hemorrhage (ICH) survivors have poor long-term outcomes, such as cognitive deficits and depression. Delayed lesions of ICH include neuron loss and white matter injury and the pathology of the lesions involves iron deposition and glial responses, which contribute to depressive-like behavior and cognitive impairment in animals. This study aimed to investigate the effects of FTY720 (0.3 mg/kg/day for 4 weeks) on iron deposition, glial responses, histological abnormalities and behavioral dysfunction in mice with ICH. The primary adverse long-term outcomes in our study of ICH mice were depressive-like behavior and impaired recognition memory. We found that FTY720 safely ameliorated depressive-like behavior and impaired recognition without affecting recovery of grip function and locomotor activity 28 days post-ICH. Moreover, we measured neuron loss, white matter lesions, lesion volume and iron deposition at day 28, which were attenuated in the FTY720-treated group compared to the ICH-control group, without changing initial hematoma volume on day 1 post-ICH. Long-term elevation of glial responses, including microglia activity and astrogliosis with tumor necrosis factor alpha (TNFα) expression was demonstrated by Western blot and immunofluorescence staining, which we found was attenuated by FTY720 treatment. Hence, FTY720 could become a novel therapeutic agent for improving long-term outcomes after ICH.
Collapse
Affiliation(s)
- Zhiyong Yang
- The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China; Department of Neurology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu 225001, China
| | - Sisi Dong
- The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Qiuyue Zheng
- Department of Neurology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu 225001, China
| | - Lingling Zhang
- Department of Neurology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu 225001, China
| | - Xinmei Tan
- The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, China
| | - Jun Zou
- Department of Neurology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu 225001, China
| | - Bingchun Yan
- Department of Integrative Traditional & Western Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu 225001, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, Jiangsu 225001, China.
| | - Yingzhu Chen
- Department of Neurology, Clinical Medical College, Yangzhou University, Yangzhou, Jiangsu 225001, China.
| |
Collapse
|
24
|
Napier J, Rose L, Adeoye O, Hooker E, Walsh KB. Modulating acute neuroinflammation in intracerebral hemorrhage: the potential promise of currently approved medications for multiple sclerosis. Immunopharmacol Immunotoxicol 2019; 41:7-15. [PMID: 30702002 DOI: 10.1080/08923973.2019.1566361] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The secondary inflammatory injury following intracerebral hemorrhage (ICH) results in increased morbidity and mortality. White blood cells have been implicated as critical mediators of this inflammatory injury. Currently, no medications have been clinically proven to ameliorate or beneficially modulate inflammation, or to improve outcomes by any mechanism, following ICH. However, other neuroinflammatory conditions, such as multiple sclerosis, have approved pharmacologic therapies that modulate the inflammatory response and minimize the damage caused by inflammatory cells. Thus, there is substantial interest in existing therapies for neuroinflammation and their potential applicability to other acute neurological diseases such as ICH. In this review, we examined the mechanism of action of twelve currently approved medications for multiple sclerosis: alemtuzumab, daclizumab, dimethyl fumarate, fingolimod, glatiramer acetate, interferon beta-1a, interferon beta-1b, mitoxantrone, natalizumab, ocrelizumab, rituximab, teriflunomide. We analyzed the existing literature pertaining to the effects of these medications on various leukocytes and also with emphasis on mechanisms of action during the acute period following initiation of therapy. As a result, we provide a valuable summary of the current body of knowledge regarding these therapies and evidence that supports or refutes their likely promise for treating neuroinflammation following ICH.
Collapse
Affiliation(s)
- Jarred Napier
- a College of Medicine , University of Cincinnati , Cincinnati , OH , USA
| | - Lucas Rose
- a College of Medicine , University of Cincinnati , Cincinnati , OH , USA
| | - Opeolu Adeoye
- b Department of Emergency Medicine , University of Cincinnati , Cincinnati , OH , USA.,c Gardner Neuroscience Institute , University of Cincinnati , Cincinnati , OH , USA
| | - Edmond Hooker
- b Department of Emergency Medicine , University of Cincinnati , Cincinnati , OH , USA
| | - Kyle B Walsh
- b Department of Emergency Medicine , University of Cincinnati , Cincinnati , OH , USA.,c Gardner Neuroscience Institute , University of Cincinnati , Cincinnati , OH , USA
| |
Collapse
|
25
|
Kong W, Qi Z, Xia P, Chang Y, Li H, Qu Y, Pan S, Yang X. Local delivery of FTY720 and NSCs on electrospun PLGA scaffolds improves functional recovery after spinal cord injury. RSC Adv 2019; 9:17801-17811. [PMID: 35520542 PMCID: PMC9064641 DOI: 10.1039/c9ra01717h] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 05/27/2019] [Indexed: 12/23/2022] Open
Abstract
Spinal cord injury (SCI) is a common issue in the clinic that causes severe motor and sensory dysfunction below the lesion level. FTY720, also known as fingolimod, has recently been reported to exert a positive effect on the recovery from a spinal cord injury. Through local delivery to the lesion site, FTY720 effectively integrates with biomaterials, and the systemic adverse effects are alleviated. However, the effects of the proper mass ratio of FTY720 in biomaterials on neural stem cell (NSC) proliferation and differentiation, as well as functional recovery after SCI, have not been thoroughly investigated. In our study, we fabricated electrospun poly (lactide-co-glycolide) (PLGA)/FTY720 scaffolds at different mass ratios (0.1%, 1%, and 10%) and characterized these scaffolds. The effects of electrospun PLGA/FTY720 scaffolds on NSC proliferation and differentiation were measured. Then, a rat model of spinal transection was established to investigate the effects of PLGA/FTY720 scaffolds loaded with NSCs. Notably, 1% PLGA/FTY720 scaffolds exerted the best effects on the proliferation and differentiation of NSCs and 10% PLGA/FTY720 was cytotoxic to NSCs. Based on the Basso, Beattie, and Bresnahan (BBB) score, HE staining and immunofluorescence staining, the PLGA/FTY720 scaffold loaded with NSCs effectively promoted the recovery of spinal cord function. Thus, FTY720 properly integrated with electrospun PLGA scaffolds, and electrospun PLGA/FTY720 scaffolds loaded with NSCs may have potential applications for SCI as a nerve implant. Spinal cord injury (SCI) is a common issue in the clinic that causes severe motor and sensory dysfunction below the lesion level.![]()
Collapse
Affiliation(s)
- Weijian Kong
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- PR China
| | - Zhiping Qi
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- PR China
| | - Peng Xia
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- PR China
| | - Yuxin Chang
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- PR China
| | - Hongru Li
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- PR China
| | - Yunpeng Qu
- Department of Cardiovascular Medicine
- The Second Hospital of Jilin University
- Changchun
- PR China
| | - Su Pan
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- PR China
| | - Xiaoyu Yang
- Department of Orthopedic Surgery
- The Second Hospital of Jilin University
- Changchun
- PR China
| |
Collapse
|
26
|
Sivertseva SA, Prilenskaya AM, Sivertsev MY, Bazhukhin DV, Smirnova NF, Guseva ME, Boyko AN. [The risk of ischemic stroke in patients with multiple sclerosis]. Zh Nevrol Psikhiatr Im S S Korsakova 2018; 118:88-94. [PMID: 30160674 DOI: 10.17116/jnevro201811808288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
A brief literature review on risk factors of ischemic stroke in patients with multiple sclerosis (MS) is presented. A case of ischemic stroke in a MS patient was demonstrated, and risk factors were identified.
Collapse
Affiliation(s)
- S A Sivertseva
- Tyumen Regional Center of Multiple Sclerosis, Tyumen, Russia
| | - A M Prilenskaya
- Tyumen Regional Center of Multiple Sclerosis, Tyumen, Russia
| | | | - D V Bazhukhin
- Tyumen Regional Center of Multiple Sclerosis, Tyumen, Russia
| | - N F Smirnova
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - M E Guseva
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - A N Boyko
- Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
27
|
Dreikorn M, Milacic Z, Pavlovic V, Meuth SG, Kleinschnitz C, Kraft P. Immunotherapy of experimental and human stroke with agents approved for multiple sclerosis: a systematic review. Ther Adv Neurol Disord 2018; 11:1756286418770626. [PMID: 29774055 PMCID: PMC5949925 DOI: 10.1177/1756286418770626] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 03/23/2018] [Indexed: 12/11/2022] Open
Abstract
Background 'Thromboinflammation' describes a novel concept in stroke pathophysiology that has opened up the possibility of immunotherapeutic approaches which could become promising strategies for targeted stroke therapies in the future. Methods We reviewed current evidence for agents approved for multiple sclerosis in preclinical and clinical stroke studies. A systematic review was performed in accordance with the PRISMA statement, searching MEDLINE, the Cochrane Central Register of Controlled Trials, and reference lists of articles published until 16 October 2017. Results The review included 52 of 629 identified studies, consisting of 5 clinical and 47 preclinical trials. Most of the studies showed beneficial effects of the evaluated immunotherapeutic drugs in terms of reduction in morphological lesion size and improvement in functional outcome. Nevertheless, the significance of these findings is limited due to the high degree of heterogeneity. Conclusions Immunotherapy of stroke might be effective and could become a promising treatment strategy, but larger clinical trials with standardized interventions and outcome measures are needed.
Collapse
Affiliation(s)
- Mirjam Dreikorn
- Department of Neurology, Hospital Main-Spessart, Lohr, Germany
| | - Zeljko Milacic
- Department of Neurology, Hospital Main-Spessart, Lohr, Germany
| | | | - Sven G Meuth
- Department of Neurology, University Hospital Münster, Münster, Germany
| | | | - Peter Kraft
- Department of Neurology, Hospital Main-Spessart, Grafen-von-Rieneck-Str. 5, 97816 Lohr, Germany
| |
Collapse
|
28
|
Chong JR, Xiang P, Wang W, Hind T, Chew WS, Ong WY, Lai MKP, Herr DR. Sphingolipidomics analysis of large clinical cohorts. Part 2: Potential impact and applications. Biochem Biophys Res Commun 2018; 504:602-607. [PMID: 29654757 DOI: 10.1016/j.bbrc.2018.04.075] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 04/10/2018] [Indexed: 11/18/2022]
Abstract
It has been known for decades that the regulation of sphingolipids (SLs) is essential for the proper function of many cellular processes. However, a complete understanding of these processes has been complicated by the structural diversity of these lipids. A well-characterized metabolic pathway is responsible for homeostatic maintenance of hundreds of distinct SL species. This pathway is perturbed in a number of pathological processes, resulting in derangement of the "sphingolipidome." Recently, advances in mass spectrometry (MS) techniques have made it possible to characterize the sphingolipidome in large-scale clinical studies, allowing for the identification of specific SL molecules that mediate pathological processes and/or may serve as biomarkers. This manuscript provides an overview of the functions of SLs, and reviews previous studies that have used MS techniques to identify changes to the sphingolipidome in non-metabolic diseases.
Collapse
Affiliation(s)
- Joyce R Chong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore
| | - Ping Xiang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore
| | - Wei Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore
| | - Tatsuma Hind
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore; Department of Pharmacology, University of British Columbia, Vancouver, BC, Canada
| | - Wee Siong Chew
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore
| | - Wei-Yi Ong
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, 119260, Singapore; Neurobiology and Ageing Research Programme, Life Sciences Institute, National University of Singapore, 119260, Singapore
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore; Neurobiology and Ageing Research Programme, Life Sciences Institute, National University of Singapore, 119260, Singapore
| | - Deron R Herr
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore; Department of Biology, San Diego State University, San Diego, CA, USA.
| |
Collapse
|
29
|
Neutrophil-to-Lymphocyte Ratio Is an Independent Predictor of 30-Day Mortality of Intracerebral Hemorrhage Patients: a Validation Cohort Study. Neurotox Res 2018; 34:347-352. [PMID: 29594812 PMCID: PMC6154212 DOI: 10.1007/s12640-018-9890-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 03/14/2018] [Accepted: 03/16/2018] [Indexed: 12/11/2022]
Abstract
In a previous study in patients with intracranial hemorrhage (ICH), we found an association between high neutrophil-to-lymphocyte ratio (NLR) with poor short-term mortality. In the current study, this preliminary finding was validated using an independent patient cohort. A total of 181 ICH patients (from January 2016 to December 2017) were included. Diagnosis was confirmed using computed tomography (CT) in all cases. Patient survival (up to 30 days) was compared between subjects with high NLR (above the 7.35 cutoff; n = 74) versus low NLR (≤ 7.35; n = 107) using Kaplan-Meier analysis. A multivariate logistic regression was performed to identify factors that influenced the 30-day mortality. Correlation between NLR with other relevant factors (e.g., C-reactive protein (CRP) and fibrinogen) was examined using Spearman correlation analysis. The 30-day mortality was 19.3% (35/181) in the entire sample, 37.8% (28/74) in the high-NLR group, and 6.5% (7/107) in the low-NLR group (P < 0.001). In comparison to the low-NLR group, the high-NLR group had higher rate of intraventricular hemorrhage (29.7 vs. 16.8%), ICH volume (median 23.9 vs. 6.0 cm3) and ICH score (median 1.5 vs. 0), and lower GCS score (9.4 ± 4.5 vs. 12.9 ± 3.2). An analysis that divided the samples into three equal parts based on NLR also showed increasing 30-day mortality with incremental NLR (1.6, 15.0, and 41.7% from lowest to highest NLR tertile, P for trend < 0.001). Kaplan-Meier curve showed higher 30-day mortality in subjects with high NLR than those with low NLR (P < 0.001 vs. low-NLR group, log-rank test). High NLR (> 7.35) is associated with poor short-term survival in acute ICH patients.
Collapse
|
30
|
Duris K, Splichal Z, Jurajda M. The Role of Inflammatory Response in Stroke Associated Programmed Cell Death. Curr Neuropharmacol 2018; 16:1365-1374. [PMID: 29473512 PMCID: PMC6251044 DOI: 10.2174/1570159x16666180222155833] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 07/17/2017] [Accepted: 02/22/2018] [Indexed: 01/13/2023] Open
Abstract
Stroke represents devastating pathology which is associated with a high morbidity and mortality. Initial damage caused directly by the onset of stroke, primary injury, may be eclipsed by secondary injury which may have a much more devastating effect on the brain. Primary injury is predominantly associated with necrotic cell death due to fatal insufficiency of oxygen and glucose. Secondary injury may on the contrary, lead apoptotic cell death due to structural damage which is not compatible with cellular functions or which may even represent the danger of malign transformation. The immune system is responsible for surveillance, defense and healing processes and the immune system plays a major role in triggering programmed cell death. Severe pathologies, such as stroke, are often associated with deregulation of the immune system, resulting in aggravation of secondary brain injury. The goal of this article is to overview the current knowledge about the role of immune system in the pathophysiology of stroke with respect to programmed neuronal cell death as well as to discuss current therapeutic strategies targeting inflammation after stroke.
Collapse
Affiliation(s)
| | | | - M. Jurajda
- Address correspondence to this author at the Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; E-mail:
| |
Collapse
|
31
|
Bonsack F, Alleyne CH, Sukumari-Ramesh S. Resveratrol Attenuates Neurodegeneration and Improves Neurological Outcomes after Intracerebral Hemorrhage in Mice. Front Cell Neurosci 2017; 11:228. [PMID: 28848394 PMCID: PMC5550718 DOI: 10.3389/fncel.2017.00228] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 07/19/2017] [Indexed: 12/17/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a devastating type of stroke with a substantial public health impact. Currently, there is no effective treatment for ICH. The purpose of the study was to evaluate whether the post-injury administration of Resveratrol confers neuroprotection in a pre-clinical model of ICH. To this end, ICH was induced in adult male CD1 mice by collagenase injection method. Resveratrol (10 mg/kg) or vehicle was administered at 30 min post-induction of ICH and the neurobehavioral outcome, neurodegeneration, cerebral edema, hematoma resolution and neuroinflammation were assessed. The Resveratrol treatment significantly attenuated acute neurological deficits, neurodegeneration and cerebral edema after ICH in comparison to vehicle treated controls. Further, Resveratrol treated mice exhibited improved hematoma resolution with a concomitant reduction in the expression of proinflammatory cytokine, IL-1β after ICH. Altogether, the data suggest the efficacy of post-injury administration of Resveratrol in improving acute neurological function after ICH.
Collapse
Affiliation(s)
- Frederick Bonsack
- Department of Neurosurgery, Medical College of Georgia, Augusta UniversityAugusta, GA, United States
| | - Cargill H Alleyne
- Department of Neurosurgery, Medical College of Georgia, Augusta UniversityAugusta, GA, United States
| | - Sangeetha Sukumari-Ramesh
- Department of Neurosurgery, Medical College of Georgia, Augusta UniversityAugusta, GA, United States
| |
Collapse
|
32
|
Lan X, Han X, Li Q, Yang QW, Wang J. Modulators of microglial activation and polarization after intracerebral haemorrhage. Nat Rev Neurol 2017; 13:420-433. [PMID: 28524175 PMCID: PMC5575938 DOI: 10.1038/nrneurol.2017.69] [Citation(s) in RCA: 620] [Impact Index Per Article: 77.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Intracerebral haemorrhage (ICH) is the most lethal subtype of stroke but currently lacks effective treatment. Microglia are among the first non-neuronal cells on the scene during the innate immune response to ICH. Microglia respond to acute brain injury by becoming activated and developing classic M1-like (proinflammatory) or alternative M2-like (anti-inflammatory) phenotypes. This polarization implies as yet unrecognized actions of microglia in ICH pathology and recovery, perhaps involving microglial production of proinflammatory or anti-inflammatory cytokines and chemokines. Furthermore, alternatively activated M2-like microglia might promote phagocytosis of red blood cells and tissue debris, a major contribution to haematoma clearance. Interactions between microglia and other cells modulate microglial activation and function, and are also important in ICH pathology. This Review summarizes key studies on modulators of microglial activation and polarization after ICH, including M1-like and M2-like microglial phenotype markers, transcription factors and key signalling pathways. Microglial phagocytosis, haematoma resolution, and the potential crosstalk between microglia and T lymphocytes, neurons, astrocytes, and oligodendrocytes in the ICH brain are described. Finally, the clinical and translational implications of microglial polarization in ICH are presented, including the evidence that therapeutic approaches aimed at modulating microglial function might mitigate ICH injury and improve brain repair.
Collapse
Affiliation(s)
- Xi Lan
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Building 370B, Baltimore, Maryland 21205, USA
| | - Xiaoning Han
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Building 370B, Baltimore, Maryland 21205, USA
| | - Qian Li
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Building 370B, Baltimore, Maryland 21205, USA
| | - Qing-Wu Yang
- Department of Neurology, Xinqiao Hospital, Third Military Medical University, 183 Xinqiao Main Street, Shapingba District, Chongqing 400037, China
| | - Jian Wang
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Building 370B, Baltimore, Maryland 21205, USA
| |
Collapse
|
33
|
Zhang X, Liu W, Yuan J, Zhu H, Yang Y, Wen Z, Chen Y, Li L, Lin J, Feng H. T lymphocytes infiltration promotes blood-brain barrier injury after experimental intracerebral hemorrhage. Brain Res 2017. [PMID: 28633994 DOI: 10.1016/j.brainres.2017.06.019] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
T lymphocytes migrate into the brain after intracerebral hemorrhage (ICH) and promote cerebral inflammation, thus exacerbating neuronal injury. However, the relationship between of T lymphocytes infiltration and blood-brain barrier (BBB) injury after ICH has not been clarified. In this study, we investigated the spatial-temporal distribution of infiltrating T lymphocytes after ICH in C57BL/6 mice by immunofluorescence and flow cytometry, and the accompanying change rules of BBB permeability were detected by Evans blue dye leakage and tight junction protein expression. Furthermore, T lymphocyte-deficient nude mice and T lymphocyte-decreased C57BL/6 mice treated with fingolimod were used to verify the relationship between T lymphocytes infiltration and BBB leakage after ICH. Here, we reported that brain-infiltrating T lymphocytes in the hemorrhagic hemisphere began to accumulate on the first day and peaked on the fifth day after ICH; BBB leakage also at peaked on the fifth day. Moreover, T lymphocyte-deficient nude mice showed minor BBB leakage after ICH compared with C57BL/6 control mice. Similarly, fingolimod treatment can significantly decrease T lymphocyte infiltration and promote BBB integrity compared with a vehicle control. Overall, our results suggested that suppression of T lymphocyte infiltration may be a novel way to improve BBB integrity after ICH.
Collapse
Affiliation(s)
- Xuan Zhang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Wei Liu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Jichao Yuan
- Department of Neurology, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Haitao Zhu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Yang Yang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Zexian Wen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Yaxing Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Lan Li
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | - Jiangkai Lin
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, China.
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, Chongqing 400038, China.
| |
Collapse
|
34
|
Rolland WB, Krafft PR, Lekic T, Klebe D, LeGrand J, Weldon AJ, Xu L, Zhang JH. Fingolimod confers neuroprotection through activation of Rac1 after experimental germinal matrix hemorrhage in rat pups. J Neurochem 2017; 140:776-786. [PMID: 28054340 DOI: 10.1111/jnc.13946] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2016] [Revised: 12/19/2016] [Accepted: 12/21/2016] [Indexed: 01/23/2023]
Abstract
Fingolimod, a sphingosine-1-phosphate receptor (S1PR) agonist, is clinically available to treat multiple sclerosis and is showing promise in treating stroke. We investigated if fingolimod provides long-term protection from experimental neonatal germinal matrix hemorrhage (GMH), aiming to support a potential mechanism of acute fingolimod-induced protection. GMH was induced in P7 rats by infusion of collagenase (0.3 U) into the right ganglionic eminence. Animals killed at 4 weeks post-GMH received low- or high-dose fingolimod (0.25 or 1.0 mg/kg) or vehicle, and underwent neurocognitive testing before histopathological evaluation. Subsequently, a cohort of animals killed at 72 h post-GMH received 1.0 mg/kg fingolimod; the specific S1PR1 agonist, SEW2871; or fingolimod co-administered with the S1PR1/3/4 inhibitor, VPC23019, or the Rac1 inhibitor, EHT1864. All drugs were injected intraperitoneally 1, 24, and 48 h post-surgery. At 72 h post-GMH, brain water content, extravasated Evans blue dye, and hemoglobin were measured as well as the expression levels of phospho-Akt, Akt, GTP-Rac1, Total-Rac1, ZO1, occludin, and claudin-3 determined. Fingolimod significantly improved long-term neurocognitive performance and ameliorated brain tissue loss. At 72 h post-GMH, fingolimod reduced brain water content and Evans blue dye extravasation as well as reversed GMH-induced loss of tight junctional proteins. S1PR1 agonism showed similar protection, whereas S1PR or Rac1 inhibition abolished the protective effect of fingolimod. Fingolimod treatment improved functional and morphological outcomes after GMH, in part, by tempering acute post-hemorrhagic blood-brain barrier disruption via the activation of the S1PR1/Akt/Rac1 pathway.
Collapse
Affiliation(s)
- William B Rolland
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, California, USA
| | - Paul R Krafft
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, California, USA.,Department of Neurosurgery, Loma Linda University Medical Center, Loma Linda, California, USA
| | - Tim Lekic
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, California, USA
| | - Damon Klebe
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, California, USA
| | - Julia LeGrand
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, California, USA
| | - Abby Jones Weldon
- Department of Pharmaceutical and Administrative Sciences, Loma Linda University, Loma Linda, California, USA
| | - Liang Xu
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, California, USA
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, California, USA.,Department of Neurosurgery, Loma Linda University Medical Center, Loma Linda, California, USA
| |
Collapse
|
35
|
Mittal MK, LacKamp A. Intracerebral Hemorrhage: Perihemorrhagic Edema and Secondary Hematoma Expansion: From Bench Work to Ongoing Controversies. Front Neurol 2016; 7:210. [PMID: 27917153 PMCID: PMC5116572 DOI: 10.3389/fneur.2016.00210] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 11/08/2016] [Indexed: 12/30/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a medical emergency, which often leads to severe disability and death. ICH-related poor outcomes are due to primary injury causing structural damage and mass effect and secondary injury in the perihemorrhagic region over several days to weeks. Secondary injury after ICH can be due to hematoma expansion (HE) or a consequence of repair pathway along the continuum of neuroinflammation, neuronal death, and perihemorrhagic edema (PHE). This review article is focused on PHE and HE and will cover the animal studies, related human studies, and clinical trials relating to these mechanisms of secondary brain injury in ICH patients.
Collapse
Affiliation(s)
- Manoj K Mittal
- Department of Neurology, University of Kansas Medical Center , Kansas City, KS , USA
| | - Aaron LacKamp
- Department of Anesthesiology, University of Kansas Medical Center , Kansas City, KS , USA
| |
Collapse
|
36
|
Zhong Q, Zhou K, Liang QL, Lin S, Wang YC, Xiong XY, Meng ZY, Zhao T, Zhu WY, Yang YR, Liao MF, Gong QW, Liu L, Xiong A, Hao J, Wang J, Yang QW. Interleukin-23 Secreted by Activated Macrophages Drives γδT Cell Production of Interleukin-17 to Aggravate Secondary Injury After Intracerebral Hemorrhage. J Am Heart Assoc 2016; 5:JAHA.116.004340. [PMID: 27729335 PMCID: PMC5121525 DOI: 10.1161/jaha.116.004340] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND Neuroinflammation plays a key role in intracerebral hemorrhage (ICH)-induced secondary brain injury, but the specific roles of peripheral inflammatory cells such as macrophages and lymphocytes remain unknown. The purpose of this study was to explore the roles of macrophages, T lymphocytes, and the cytokines they secrete as potential targets for treating secondary brain injury after ICH. METHODS AND RESULTS Our results showed that peripheral macrophages and T lymphocytes successively infiltrated the brain, with macrophage counts peaking 1 day after ICH and T-lymphocyte counts peaking after 4 days. These peaks in cellular infiltration corresponded to increases in interleukin (IL)-23 and IL-17 expression, respectively. We found that hemoglobin from the hematoma activated IL-23 secretion by infiltrating macrophages by inducing the formation of toll-like receptor (TLR) 2/4 heterodimer. This increased IL-23 expression stimulated γδT-cell production of IL-17, which increased brain edema and neurologic deficits in the model mice as a proinflammatory factor. Finally, we found that sparstolonin B (SsnB) could ameliorate brain edema and neurologic deficits in ICH model mice via inhibition of TLR2/TLR4 heterodimer formation, and notably, SsnB interacted with myeloid differentiation factor 88 Arg196. CONCLUSIONS Together, our results reveal the importance of the IL-23/IL-17 inflammatory axis in secondary brain injury after ICH and thus provide a new therapeutic target for ICH treatment.
Collapse
Affiliation(s)
- Qi Zhong
- Department of Neurology, Xinqiao Hospital, the Third Military Medical University, Chongqing, China
| | - Kai Zhou
- Department of Neurology, Xinqiao Hospital, the Third Military Medical University, Chongqing, China
| | - Qiao-Li Liang
- Nanjing University of Chinese Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing, China
| | - Sen Lin
- Department of Anatomy, Histology and Embryology, Chengdu Medical College, Chengdu, China
| | - Yan-Chun Wang
- Department of Neurology, Xinqiao Hospital, the Third Military Medical University, Chongqing, China
| | - Xiao-Yi Xiong
- Department of Neurology, Xinqiao Hospital, the Third Military Medical University, Chongqing, China
| | - Zhao-You Meng
- Department of Neurology, Xinqiao Hospital, the Third Military Medical University, Chongqing, China
| | - Ting Zhao
- Department of Neurology, Xinqiao Hospital, the Third Military Medical University, Chongqing, China
| | - Wen-Yao Zhu
- Department of Neurology, Xinqiao Hospital, the Third Military Medical University, Chongqing, China
| | - Yuan-Rui Yang
- Department of Neurology, Xinqiao Hospital, the Third Military Medical University, Chongqing, China
| | - Mao-Fan Liao
- Department of Neurology, Xinqiao Hospital, the Third Military Medical University, Chongqing, China
| | - Qiu-Wen Gong
- Department of Neurology, Xinqiao Hospital, the Third Military Medical University, Chongqing, China
| | - Liang Liu
- Department of Neurology, Xinqiao Hospital, the Third Military Medical University, Chongqing, China
| | - Ao Xiong
- Basic Medical College, Zhengzhou University, Zhengzhou, China
| | - Junwei Hao
- Department of Neurology, Key Laboratory of Neurorepair and Regeneration, Tianjin and Ministry of Education, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Jian Wang
- Department of Anesthesiology/Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD
| | - Qing-Wu Yang
- Department of Neurology, Xinqiao Hospital, the Third Military Medical University, Chongqing, China
| |
Collapse
|
37
|
Li W, Xu H, Testai FD. Mechanism of Action and Clinical Potential of Fingolimod for the Treatment of Stroke. Front Neurol 2016; 7:139. [PMID: 27617002 PMCID: PMC4999895 DOI: 10.3389/fneur.2016.00139] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 08/15/2016] [Indexed: 01/23/2023] Open
Abstract
Fingolimod (FTY720) is an orally bio-available immunomodulatory drug currently approved by the FDA for the treatment of multiple sclerosis. Currently, there is a significant interest in the potential benefits of FTY720 on stroke outcomes. FTY720 and the sphingolipid signaling pathway it modulates has a ubiquitous presence in the central nervous system and both rodent models and pilot clinical trials seem to indicate that the drug may improve overall functional recovery in different stroke subtypes. Although the precise mechanisms behind these beneficial effects are yet unclear, there is evidence that FTY720 has a role in regulating cerebrovascular responses, blood-brain barrier permeability, and cell survival in the event of cerebrovascular insult. In this article, we critically review the data obtained from the latest laboratory findings and clinical trials involving both ischemic and hemorrhagic stroke, and attempt to form a cohesive picture of FTY720's mechanisms of action in stroke.
Collapse
Affiliation(s)
- Wentao Li
- Department of Neurology and Rehabilitation, University of Illinois College of Medicine , Chicago, IL , USA
| | - Haoliang Xu
- Department of Pathology, University of Illinois College of Medicine , Chicago, IL , USA
| | - Fernando D Testai
- Department of Neurology and Rehabilitation, University of Illinois College of Medicine , Chicago, IL , USA
| |
Collapse
|
38
|
Sun N, Keep RF, Hua Y, Xi G. Critical Role of the Sphingolipid Pathway in Stroke: a Review of Current Utility and Potential Therapeutic Targets. Transl Stroke Res 2016; 7:420-38. [PMID: 27339463 DOI: 10.1007/s12975-016-0477-3] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 06/12/2016] [Accepted: 06/15/2016] [Indexed: 12/16/2022]
Abstract
Sphingolipids are a series of cell membrane-derived lipids which act as signaling molecules and play a critical role in cell death and survival, proliferation, recognition, and migration. Sphingosine-1-phosphate acts as a key signaling molecule and regulates lymphocyte trafficking, glial cell activation, vasoconstriction, endothelial barrier function, and neuronal death pathways which plays a critical role in numerous neurological conditions. Stroke is a second leading cause of death all over the world and effective therapies are still in great demand, including ischemic stroke and hemorrhagic stroke as well as poststroke repair. Significantly, sphingolipid activities change after stroke and correlate with stroke outcome, which has promoted efforts to testify whether the sphingolipid pathway could be a novel therapeutic target in stroke. The sphingolipid metabolic pathway, the connection between the pathway and stroke, as well as therapeutic interventions to manipulate the pathway to reduce stroke-induced brain injury are discussed in this review.
Collapse
Affiliation(s)
- Na Sun
- Department of Neurosurgery, University of Michigan, 5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
- Department of Neurology, Tianjin Medical University General Hospital, Tianjin, China
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, 5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, 5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, 5018 BSRB, 109 Zina Pitcher Place, Ann Arbor, MI, 48109-2200, USA.
| |
Collapse
|
39
|
Sun N, Shen Y, Han W, Shi K, Wood K, Fu Y, Hao J, Liu Q, Sheth KN, Huang D, Shi FD. Selective Sphingosine-1-Phosphate Receptor 1 Modulation Attenuates Experimental Intracerebral Hemorrhage. Stroke 2016; 47:1899-906. [PMID: 27174529 DOI: 10.1161/strokeaha.115.012236] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 04/07/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND PURPOSE Preclinical studies and a proof-of-concept clinical study have shown that sphingosine-1-phosphate receptor (S1PR) modulator, fingolimod, improves the clinical outcome of intracerebral hemorrhage (ICH). However, the specific subtype of the S1PRs through which immune modulation provides protection in ICH remains unclear. In addition, fingolimod-induced adverse effects could limit its use in patients with stroke because of interactions with other S1PR subtypes, particularly with S1PR3. RP101075 is a selective S1PR1 agonist with superior cardiovascular safety profile. In this study, we investigated the impact of RP101075 treatment in a mouse model of ICH. METHODS ICH was induced by injection of autologous blood in 294 male C57BL/6J and Rag2(-/-) mice. ICH mice randomly received vehicle, RP101075, or RP101075 plus S1PR1 antagonist W146 by daily oral gavage for three consecutive days, starting from 30 minutes after surgery. Neurodeficits, brain edema, brain infiltration of immune cells, blood-brain barrier integrity, and cell death were assessed after ICH. RESULTS RP101075 significantly attenuated neurological deficits and reduced brain edema in ICH mice. W146 blocked the effects of RP101075 on neurodeficits and brain edema. RP101075 reduced the counts of brain-infiltrating lymphocytes, neutrophils, and microglia, as well as cytokine expression after ICH. Enhanced blood-brain barrier integrity and alleviated neuronal death were also seen in ICH mice after RP101075 treatment. CONCLUSIONS S1PR1 modulation via RP101075 provides protection in experimental ICH. Together with the advantageous pharmacological features of RP101075, these results warrant further investigations of its mechanisms of action and translational values in ICH patients.
Collapse
Affiliation(s)
- Na Sun
- From the Departments of Neurology (N.S., Y.S., K.S., Y.F., J.H., Q.L., F.-D.S.) and Radiology (W.H.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ (N.S., K.W., Q.L., F.-D.S.); Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); and Department of Clinical Research and Multiple Sclerosis Center, Neurology and Neuroscience Associates, Unity Health Network, Akron, OH (D.H.)
| | - Yi Shen
- From the Departments of Neurology (N.S., Y.S., K.S., Y.F., J.H., Q.L., F.-D.S.) and Radiology (W.H.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ (N.S., K.W., Q.L., F.-D.S.); Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); and Department of Clinical Research and Multiple Sclerosis Center, Neurology and Neuroscience Associates, Unity Health Network, Akron, OH (D.H.)
| | - Wei Han
- From the Departments of Neurology (N.S., Y.S., K.S., Y.F., J.H., Q.L., F.-D.S.) and Radiology (W.H.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ (N.S., K.W., Q.L., F.-D.S.); Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); and Department of Clinical Research and Multiple Sclerosis Center, Neurology and Neuroscience Associates, Unity Health Network, Akron, OH (D.H.)
| | - Kaibin Shi
- From the Departments of Neurology (N.S., Y.S., K.S., Y.F., J.H., Q.L., F.-D.S.) and Radiology (W.H.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ (N.S., K.W., Q.L., F.-D.S.); Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); and Department of Clinical Research and Multiple Sclerosis Center, Neurology and Neuroscience Associates, Unity Health Network, Akron, OH (D.H.)
| | - Kristofer Wood
- From the Departments of Neurology (N.S., Y.S., K.S., Y.F., J.H., Q.L., F.-D.S.) and Radiology (W.H.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ (N.S., K.W., Q.L., F.-D.S.); Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); and Department of Clinical Research and Multiple Sclerosis Center, Neurology and Neuroscience Associates, Unity Health Network, Akron, OH (D.H.)
| | - Ying Fu
- From the Departments of Neurology (N.S., Y.S., K.S., Y.F., J.H., Q.L., F.-D.S.) and Radiology (W.H.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ (N.S., K.W., Q.L., F.-D.S.); Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); and Department of Clinical Research and Multiple Sclerosis Center, Neurology and Neuroscience Associates, Unity Health Network, Akron, OH (D.H.)
| | - Junwei Hao
- From the Departments of Neurology (N.S., Y.S., K.S., Y.F., J.H., Q.L., F.-D.S.) and Radiology (W.H.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ (N.S., K.W., Q.L., F.-D.S.); Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); and Department of Clinical Research and Multiple Sclerosis Center, Neurology and Neuroscience Associates, Unity Health Network, Akron, OH (D.H.)
| | - Qiang Liu
- From the Departments of Neurology (N.S., Y.S., K.S., Y.F., J.H., Q.L., F.-D.S.) and Radiology (W.H.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ (N.S., K.W., Q.L., F.-D.S.); Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); and Department of Clinical Research and Multiple Sclerosis Center, Neurology and Neuroscience Associates, Unity Health Network, Akron, OH (D.H.)
| | - Kevin N Sheth
- From the Departments of Neurology (N.S., Y.S., K.S., Y.F., J.H., Q.L., F.-D.S.) and Radiology (W.H.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ (N.S., K.W., Q.L., F.-D.S.); Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); and Department of Clinical Research and Multiple Sclerosis Center, Neurology and Neuroscience Associates, Unity Health Network, Akron, OH (D.H.)
| | - DeRen Huang
- From the Departments of Neurology (N.S., Y.S., K.S., Y.F., J.H., Q.L., F.-D.S.) and Radiology (W.H.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ (N.S., K.W., Q.L., F.-D.S.); Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); and Department of Clinical Research and Multiple Sclerosis Center, Neurology and Neuroscience Associates, Unity Health Network, Akron, OH (D.H.)
| | - Fu-Dong Shi
- From the Departments of Neurology (N.S., Y.S., K.S., Y.F., J.H., Q.L., F.-D.S.) and Radiology (W.H.), Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China; Department of Neurology, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ (N.S., K.W., Q.L., F.-D.S.); Department of Neurology, Yale University School of Medicine, New Haven, CT (K.N.S.); and Department of Clinical Research and Multiple Sclerosis Center, Neurology and Neuroscience Associates, Unity Health Network, Akron, OH (D.H.).
| |
Collapse
|
40
|
Blanc CA, Grist JJ, Rosen H, Sears-Kraxberger I, Steward O, Lane TE. Sphingosine-1-phosphate receptor antagonism enhances proliferation and migration of engrafted neural progenitor cells in a model of viral-induced demyelination. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 185:2819-32. [PMID: 26435414 DOI: 10.1016/j.ajpath.2015.06.009] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 06/22/2015] [Accepted: 06/25/2015] [Indexed: 01/12/2023]
Abstract
The oral drug FTY720 affects sphingosine-1-phosphate (S1P) signaling on targeted cells that bear the S1P receptors S1P1, S1P3, S1P4, and S1P5. We examined the effect of FTY720 treatment on the biology of mouse neural progenitor cells (NPCs) after transplantation in a viral model of demyelination. Intracerebral infection with the neurotropic JHM strain of mouse hepatitis virus (JHMV) resulted in an acute encephalomyelitis, followed by demyelination similar in pathology to the human demyelinating disease, multiple sclerosis. We have previously reported that intraspinal transplantation of mouse NPCs into JHMV-infected animals resulted in selective colonization of demyelinated lesions, preferential differentiation into oligodendroglia accompanied by axonal preservation, and increased remyelination. Cultured NPCs expressed transcripts for S1P receptors S1P1, S1P2, S1P3, S1P4, and S1P5. FTY720 treatment of cultured NPCs resulted in increased mitogen-activated protein kinase phosphorylation and migration after exposure to the chemokine CXCL12. Administration of FTY720 to JHMV-infected mice resulted in enhanced migration and increased proliferation of transplanted NPCs after spinal cord engraftment. FTY720 treatment did not improve clinical disease, diminish neuroinflammation or the severity of demyelination, nor increase remyelination. These findings argue that FTY720 treatment selectively increases NPC proliferation and migration but does not either improve clinical outcome or enhance remyelination after transplantation into animals in which immune-mediated demyelination is initiated by the viral infection of the central nervous system.
Collapse
Affiliation(s)
- Caroline A Blanc
- Department of Molecular Biology and Biochemistry, Reeve-Irvine Research Center Irvine School of Medicine, University of California, Irvine, California
| | - Jonathan J Grist
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah
| | - Hugh Rosen
- Department of Chemical Physiology, The Scripps Research Institute, La Jolla, California
| | - Ilse Sears-Kraxberger
- Departments of Anatomy and Neurobiology and Neurobiology and Behavior, Reeve-Irvine Research Center Irvine School of Medicine, University of California, Irvine, California
| | - Oswald Steward
- Departments of Anatomy and Neurobiology and Neurobiology and Behavior, Reeve-Irvine Research Center Irvine School of Medicine, University of California, Irvine, California
| | - Thomas E Lane
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah.
| |
Collapse
|
41
|
Stiles M, Qi H, Sun E, Tan J, Porter H, Allegood J, Chalfant CE, Yasumura D, Matthes MT, LaVail MM, Mandal NA. Sphingolipid profile alters in retinal dystrophic P23H-1 rats and systemic FTY720 can delay retinal degeneration. J Lipid Res 2016; 57:818-31. [PMID: 26947037 DOI: 10.1194/jlr.m063719] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Indexed: 12/28/2022] Open
Abstract
Retinal degeneration (RD) affects millions of people and is a major cause of ocular impairment and blindness. With a wide range of mutations and conditions leading to degeneration, targeting downstream processes is necessary for developing effective treatments. Ceramide and sphingosine-1-phosphate, a pair of bioactive sphingolipids, are involved in apoptosis and its prevention, respectively. Apoptotic cell death is a potential driver of RD, and in order to understand the mechanism of degeneration and potential treatments, we studied rhodopsin mutant RD model, P23H-1 rats. Investigating this genetic model of human RD allows us to investigate the association of sphingolipid metabolites with the degeneration of the retina in P23H-1 rats and the effects of a specific modulator of sphingolipid metabolism, FTY720. We found that P23H-1 rat retinas had altered sphingolipid profiles that, when treated with FTY720, were rebalanced closer to normal levels. FTY720-treated rats also showed protection from RD compared with their vehicle-treated littermates. Based on these data, we conclude that sphingolipid dysregulation plays a secondary role in retinal cell death, which may be common to many forms of RDs, and that the U.S. Food and Drug Administration-approved drug FTY720 or related compounds that modulate sphingolipid metabolism could potentially delay the cell death.
Collapse
Affiliation(s)
- Megan Stiles
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 Dean McGee Eye Institute, Oklahoma City, OK 73104
| | - Hui Qi
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 Dean McGee Eye Institute, Oklahoma City, OK 73104
| | - Eleanor Sun
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 Dean McGee Eye Institute, Oklahoma City, OK 73104
| | - Jeremy Tan
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 Dean McGee Eye Institute, Oklahoma City, OK 73104
| | - Hunter Porter
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 Dean McGee Eye Institute, Oklahoma City, OK 73104
| | - Jeremy Allegood
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298
| | - Charles E Chalfant
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University, Richmond, VA 23298 Research and Development, Hunter Holmes McGuire Veterans Administration Medical Center, Richmond, VA 23249 Virginia Commonwealth University School of Medicine, Virginia Commonwealth University Massey Cancer Center, Virginia Commonwealth University, Richmond, VA 23298 Virginia Commonwealth University Institute of Molecular Medicine and the Virginia Commonwealth University Johnson Center, Virginia Commonwealth University, Richmond, VA 23298
| | - Douglas Yasumura
- Beckman Vision Center, University of California, San Francisco School of Medicine, San Francisco, CA 94143
| | - Michael T Matthes
- Beckman Vision Center, University of California, San Francisco School of Medicine, San Francisco, CA 94143
| | - Matthew M LaVail
- Beckman Vision Center, University of California, San Francisco School of Medicine, San Francisco, CA 94143
| | - Nawajes A Mandal
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 Dean McGee Eye Institute, Oklahoma City, OK 73104 Department of Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104
| |
Collapse
|
42
|
Artesunate Protected Blood-Brain Barrier via Sphingosine 1 Phosphate Receptor 1/Phosphatidylinositol 3 Kinase Pathway After Subarachnoid Hemorrhage in Rats. Mol Neurobiol 2016; 54:1213-1228. [PMID: 26820677 DOI: 10.1007/s12035-016-9732-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 01/19/2016] [Indexed: 10/22/2022]
Abstract
Blood-brain barrier preservation plays an important role in attenuating vasogenic brain edema after subarachnoid hemorrhage (SAH). This study was designed to investigate the protective effect and mechanism of artesunate, a traditional anti-malaria drug, on blood-brain barrier after SAH. Three hundred and seventy-seven (377) male Sprague-Dawley rats were subjected to endovascular perforation model for SAH. The rats received artesunate alone or in combination with Sphingosine-1-phosphate receptor-1 (S1P1) small interfering RNA (siRNA), antagonist VPC23019, or phosphatidylinositol 3-kinase inhibitor wortmannin after SAH. Modified Garcia score, SAH grades, brain water content, Evans blue leakage, transmission electron microscope, immunohistochemistry staining, Western blot, and cultured endothelial cells were used to investigate the optimum concentration and the therapeutic mechanism of artesunate. We found that artesunate (200 mg/kg) could do better in raising modified Garcia score, reducing brain water content and Evans blue leakage than other groups after SAH. Moreover, artesunate elevated S1P1 expression, enhanced phosphatidylinositol 3-kinase activation, lowered GSK-3β activation, stabilized β-catenin, and improved the expression of Claudin-3 and Claudin-5 after SAH in rats. These effects were eliminated by S1P1 siRNA, VPC23019, and wortmannin. This study revealed that artesunate could preserve blood-brain barrier integrity and improve neurological outcome after SAH, possibly through activating S1P1, enhancing phosphatidylinositol 3-kinase activation, stabilizing β-catenin via GSK-3β inhibition, and then effectively raising the expression of Claudin-3 and Claudin-5. Therefore, artesunate may be favorable for the blood-brain barrier (BBB) protection after SAH and become a potential candidate for the treatment of SAH patients.
Collapse
|
43
|
Klebe D, McBride D, Flores JJ, Zhang JH, Tang J. Modulating the Immune Response Towards a Neuroregenerative Peri-injury Milieu After Cerebral Hemorrhage. J Neuroimmune Pharmacol 2015; 10:576-86. [PMID: 25946986 PMCID: PMC4636976 DOI: 10.1007/s11481-015-9613-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 04/29/2015] [Indexed: 01/06/2023]
Abstract
Cerebral hemorrhages account for 15-20 % of stroke sub-types and have very poor prognoses. The mortality rate for cerebral hemorrhage patients is between 40 and 50 %, of which at least half of the deaths occur within the first 2 days, and 75 % of survivors are incapable of living independently after 1 year. Current emergency interventions involve lowering blood pressure and reducing intracranial pressure by controlled ventilations or, in the worst case scenarios, surgical intervention. Some hemostatic and coagulatherapeutic interventions are being investigated, although a few that were promising in experimental studies have failed in clinical trials. No significant immunomodulatory intervention, however, exists for clinical management of cerebral hemorrhage. The inflammatory response following cerebral hemorrhage is particularly harmful in the acute stage because blood-brain barrier disruption is amplified and surrounding tissue is destroyed by secreted proteases and reactive oxygen species from infiltrated leukocytes. In this review, we discuss both the destructive and regenerative roles the immune response play following cerebral hemorrhage and focus on microglia, macrophages, and T-lymphocytes as the primary agents directing the response. Microglia, macrophages, and T-lymphocytes each have sub-types that significantly influence the over-arching immune response towards either a pro-inflammatory, destructive, or an anti-inflammatory, regenerative, state. Both pre-clinical and clinical studies of cerebral hemorrhages that selectively target these immune cells are reviewed and we suggest immunomodulatory therapies that reduce inflammation, while augmenting neural repair, will improve overall cerebral hemorrhage outcomes.
Collapse
Affiliation(s)
- Damon Klebe
- Department of Physiology & Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Devin McBride
- Department of Physiology & Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Jerry J Flores
- Department of Physiology & Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - John H Zhang
- Department of Physiology & Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
- Departments of Anesthesiology and Neurosurgery, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA
| | - Jiping Tang
- Department of Physiology & Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA, 92350, USA.
| |
Collapse
|
44
|
Abstract
Approaches for the effective management of acute stroke are sparse, and many measures for brain protection fail. However, our ability to modulate the immune system and modify the progression of multiple sclerosis is increasing. As a result, immune interventions are currently being explored as therapeutic interventions in acute stroke. In this Review, we compare the immunological features of acute stroke with those of multiple sclerosis, identify unique immunological features of stroke, and consider the evidence for immune interventions. In patients with acute stroke, microglial activation and cell death products trigger an inflammatory cascade that damages vessels and the parenchyma within minutes to hours of the ischaemia or haemorrhage. Immune interventions that restrict brain inflammation, vascular permeability and tissue oedema must be administered rapidly to reduce acute immune-mediated destruction and to avoid subsequent immunosuppression. Preliminary results suggest that the use of drugs that modify disease in multiple sclerosis might accomplish these goals in ischaemic and haemorrhagic stroke. Further elucidation of the immune mechanisms involved in stroke is likely to lead to successful immune interventions.
Collapse
Affiliation(s)
- Ying Fu
- Departments of Neurology and Immunology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China (Y.F., Q.L., F.-D.S.); Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, 525 East 68th Street, PO Box 117, New York, NY 10065, USA. (J.A.)
| | - Qiang Liu
- Departments of Neurology and Immunology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China (Y.F., Q.L., F.-D.S.); Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, 525 East 68th Street, PO Box 117, New York, NY 10065, USA. (J.A.)
| | - Josef Anrather
- Departments of Neurology and Immunology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China (Y.F., Q.L., F.-D.S.); Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, 525 East 68th Street, PO Box 117, New York, NY 10065, USA. (J.A.)
| | - Fu-Dong Shi
- Departments of Neurology and Immunology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin 300052, China (Y.F., Q.L., F.-D.S.); Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, 525 East 68th Street, PO Box 117, New York, NY 10065, USA. (J.A.)
| |
Collapse
|
45
|
Kathirvelu B, Carmichael ST. Intracerebral hemorrhage in mouse models: therapeutic interventions and functional recovery. Metab Brain Dis 2015; 30:449-59. [PMID: 24810632 PMCID: PMC4226812 DOI: 10.1007/s11011-014-9559-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 04/28/2014] [Indexed: 12/11/2022]
Abstract
There has been strong pre-clinical research on mechanisms of initial cell death and tissue injury in intracerebral hemorrhage (ICH). This data has led to the evaluation of several therapeutics for neuroprotection or the mitigation of early tissue damage. Most of these studies have been done in the rat. Also, there has been little study of the mechanisms of tissue repair and recovery. This review examines the testing of candidate therapeutics in mouse models of ICH for their effect on tissue protection and repair. This review will help the readers compare it to the extensively researched rat model of ICH and thus enhance work that are pending in mouse model.
Collapse
Affiliation(s)
- Balachandar Kathirvelu
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA,
| | | |
Collapse
|
46
|
Xu H, Testai FD, Valyi-Nagy T, N Pavuluri M, Zhai F, Nanegrungsunk D, Paisansathan C, Pelligrino DA. VAP-1 blockade prevents subarachnoid hemorrhage-associated cerebrovascular dilating dysfunction via repression of a neutrophil recruitment-related mechanism. Brain Res 2015; 1603:141-9. [PMID: 25662771 DOI: 10.1016/j.brainres.2015.01.047] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2014] [Revised: 01/19/2015] [Accepted: 01/27/2015] [Indexed: 11/19/2022]
Abstract
Our previous findings indicated that in rats subjected to subarachnoid hemorrhage (SAH), suppression of post-SAH neuroinflammation via vascular adhesion protein-1 (VAP-1) blockade provides significant neuroprotection. We and others have reported that neuroinflammation contributes to cerebral microvascular impairment. Thus, in the present study, we tested the hypotheses that: (1) treatment with LJP-1586, a selective VAP-1 blocker, prevents SAH-associated pial arteriolar dilating dysfunction; and (2) the vasculoprotective effect of LJP-1586 arises from inhibiting SAH-elicited neutrophil recruitment. We utilized an endovascular perforation model of SAH. Rats subjected to SAH were either treated with LJP-1586 or rendered neutropenic via anti-neutrophil-antibody treatment. Findings from these groups were compared to their respective control groups. At 48 h post-SAH, rats were evaluated for neurobehavioral function, pial venular leukocyte trafficking, and pial arteriolar reactivity to topically-applied acetylcholine (ACh) and S-nitroso-N-acetyl penicillamine (SNAP). Pial arteriolar responses decreased at 48 h post-SAH. However, in the presence of LJP-1586, those responses were significantly preserved. Neutrophil-depletion yielded a substantial suppression of SAH-associated leukocyte adhesion and infiltration. This was accompanied by a significant preservation of pial arteriolar dilating function, suggesting a direct link between neutrophil recruitment and the loss of cerebral microvascular reactivity. Moreover, neutrophil depletion also was associated with significant protection of neurobehavioral function. The present findings suggest that attenuating SAH-linked elevation in neutrophil trafficking will protect against the development of microvascular dysfunction and subsequent neurological impairment.
Collapse
Affiliation(s)
- Haoliang Xu
- Neuroanesthesia Research Laboratory, Department of Anesthesiology, University of Illinois-Chicago, Chicago, IL, United States.
| | - Fernando D Testai
- Department of Neurology and Rehabilitation, University of Illinois-Chicago, Chicago, IL, United States
| | - Tibor Valyi-Nagy
- Department of Pathology, University of Illinois-Chicago, Chicago, IL, United States
| | - Mani N Pavuluri
- Department of Psychiatry, University of Illinois-Chicago, Chicago, IL, United States
| | - Fengguo Zhai
- Neuroanesthesia Research Laboratory, Department of Anesthesiology, University of Illinois-Chicago, Chicago, IL, United States
| | - Danop Nanegrungsunk
- Neuroanesthesia Research Laboratory, Department of Anesthesiology, University of Illinois-Chicago, Chicago, IL, United States
| | - Chanannait Paisansathan
- Neuroanesthesia Research Laboratory, Department of Anesthesiology, University of Illinois-Chicago, Chicago, IL, United States
| | - Dale A Pelligrino
- Neuroanesthesia Research Laboratory, Department of Anesthesiology, University of Illinois-Chicago, Chicago, IL, United States
| |
Collapse
|
47
|
Xu HL, Pelligrino DA, Paisansathan C, Testai FD. Protective role of fingolimod (FTY720) in rats subjected to subarachnoid hemorrhage. J Neuroinflammation 2015; 12:16. [PMID: 25622980 PMCID: PMC4324852 DOI: 10.1186/s12974-015-0234-7] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Accepted: 01/01/2015] [Indexed: 12/20/2022] Open
Abstract
Background Subarachnoid hemorrhage (SAH) is a neurological emergency with limited pharmacological treatment options. Inflammation is increasingly recognized as a key pathogenic contributor to brain injury in this condition. In the present study, we examined the neuroprotective effects of the immunomodulatory agent, fingolimod, in rats subjected to SAH. Methods We utilized an endovascular rat perforation model of SAH. Animals were divided into four groups: (1) sham-vehicle; (2) sham-fingolimod; (3) SAH-vehicle; and (4) SAH-fingolimod. Rats received either vehicle solution or fingolimod (0.5 mg/kg) intraperitoneally 3 hours after sham surgery or SAH. A closed cranial window and intravital microscope system was used at 48 hours to assess neuroinflammation, which was represented by rhodamine-6G-labeled leukocyte trafficking in pial venules, and pial arteriolar dilating responses to a variety of vasodilators, including hypercapnia, and topically-applied acetylcholine, adenosine, and S-nitroso-N-acetyl penicillamine. In addition, motor-sensory function was evaluated. Results Compared to sham-vehicle rats, SAH-vehicle animals displayed a four-times greater increase in pial venular intraluminal leukocyte adhesion. Treatment with fingolimod largely reduced the intravascular leukocyte adhesion. Vehicle-treated SAH animals displayed a significant decrease in pial arteriolar responses to all the vasodilators tested and vascular reactivity was preserved, to a significant degree, in the presence of fingolimod. In addition, neurological scores obtained at 48 hours post-SAH indicated significant neurological deficits in the vehicle-treated group (versus sham-vehicle surgical control). Those deficiencies were partially reduced by fingolimod (P < 0.0001 compared to the vehicle-treated SAH group). Conclusions Treatment of rats with fingolimod was associated with a marked limitation in the intravascular adhesion of leukocytes to pial venules, preserved pial arteriolar dilating function, and improved neurological outcome in rats subjected to SAH.
Collapse
Affiliation(s)
- Hao-Liang Xu
- Neuroanesthesia Research Laboratory, University of Illinois College of Medicine, Chicago, IL, USA.
| | - Dale A Pelligrino
- Neuroanesthesia Research Laboratory, University of Illinois College of Medicine, Chicago, IL, USA.
| | - Chanannait Paisansathan
- Department of Anesthesiology of the University of Illinois College of Medicine, Chicago, IL, USA.
| | - Fernando D Testai
- Department of Neurology and Rehabilitation of the University of Illinois College of Medicine, 912 S Wood Street, Rm 855 N NPI (MC 796), Chicago, IL, 60612-7330, USA.
| |
Collapse
|
48
|
Testai FD, Xu HL, Kilkus J, Suryadevara V, Gorshkova I, Berdyshev E, Pelligrino DA, Dawson G. Changes in the metabolism of sphingolipids after subarachnoid hemorrhage. J Neurosci Res 2015; 93:796-805. [PMID: 25597763 DOI: 10.1002/jnr.23542] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 11/19/2014] [Accepted: 11/20/2014] [Indexed: 12/31/2022]
Abstract
We previously described how ceramide (Cer), a mediator of cell death, increases in the cerebrospinal fluid (CSF) of subarachnoid hemorrhage (SAH) patients. This study investigates the alterations of biochemical pathways involved in Cer homeostasis in SAH. Cer, dihydroceramide (DHC), sphingosine-1-phosphate (S1P), and the activities of acid sphingomyelinase (ASMase), neutral sphingomyelinase (NSMase), sphingomyelinase synthase (SMS), S1P-lyase, and glucosylceramide synthase (GCS) were determined in the CSF of SAH subjects and in brain homogenate of SAH rats. Compared with controls (n = 8), SAH patients (n = 26) had higher ASMase activity (10.0 ± 3.5 IF/µl· min vs. 15.0 ± 4.6 IF/µl • min; P = 0.009) and elevated levels of Cer (11.4 ± 8.8 pmol/ml vs. 33.3 ± 48.3 pmol/ml; P = 0.001) and DHC (1.3 ± 1.1 pmol/ml vs. 3.8 ± 3.4 pmol/ml; P = 0.001) in the CSF. The activities of GCS, NSMase, and SMS in the CSF were undetectable. Brain homogenates from SAH animals had increased ASMase activity (control: 9.7 ± 1.2 IF/µg • min; SAH: 16.8 ± 1.6 IF/µg • min; P < 0.05) and Cer levels (control: 3,422 ± 26 fmol/nmol of total lipid P; SAH: 7,073 ± 2,467 fmol/nmol of total lipid P; P < 0.05) compared with controls. In addition, SAH was associated with a reduction of 60% in S1P levels, a 40% increase in S1P-lyase activity, and a twofold increase in the activity of GCS. In comparison, NSMase and SMS activities were similar to controls and SMS activities similar to controls. In conclusion, our results show an activation of ASMase, S1P-lyase, and GCS resulting in a shift in the production of protective (S1P) in favor of deleterious (Cer) sphingolipids after SAH. Additional studies are needed to determine the effect of modulators of the pathways described here in SAH.
Collapse
Affiliation(s)
- Fernando D Testai
- Department of Neurology and Rehabilitation, University of Illinois at Chicago, Chicago, Illinois
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Mracsko E, Veltkamp R. Neuroinflammation after intracerebral hemorrhage. Front Cell Neurosci 2014; 8:388. [PMID: 25477782 PMCID: PMC4238323 DOI: 10.3389/fncel.2014.00388] [Citation(s) in RCA: 268] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Accepted: 10/31/2014] [Indexed: 12/15/2022] Open
Abstract
Spontaneous intracerebral hemorrhage (ICH) is a particularly severe type of stroke for which no specific treatment has been established yet. Although preclinical models of ICH have substantial methodological limitations, important insight into the pathophysiology has been gained. Mounting evidence suggests an important contribution of inflammatory mechanisms to brain damage and potential repair. Neuroinflammation evoked by intracerebral blood involves the activation of resident microglia, the infiltration of systemic immune cells and the production of cytokines, chemokines, extracellular proteases and reactive oxygen species (ROS). Previous studies focused on innate immunity including microglia, monocytes and granulocytes. More recently, the role of adaptive immune cells has received increasing attention. Little is currently known about the interactions among different immune cell populations in the setting of ICH. Nevertheless, immunomodulatory strategies are already being explored in ICH. To improve the chances of translation from preclinical models to patients, a better characterization of the neuroinflammation in patients is desirable.
Collapse
Affiliation(s)
- Eva Mracsko
- Department of Neurology, University Heidelberg Heidelberg, Germany
| | - Roland Veltkamp
- Department of Neurology, University Heidelberg Heidelberg, Germany ; Division of Brain Sciences, Imperial College London, UK
| |
Collapse
|
50
|
Urra X, Miró F, Chamorro A, Planas AM. Antigen-specific immune reactions to ischemic stroke. Front Cell Neurosci 2014; 8:278. [PMID: 25309322 PMCID: PMC4162361 DOI: 10.3389/fncel.2014.00278] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 08/22/2014] [Indexed: 12/24/2022] Open
Abstract
Brain proteins are detected in the cerebrospinal fluid (CSF) and blood of stroke patients and their concentration is related to the extent of brain damage. Antibodies against brain antigens develop after stroke, suggesting a humoral immune response to the brain injury. Furthermore, induced immune tolerance is beneficial in animal models of cerebral ischemia. The presence of circulating T cells sensitized against brain antigens, and antigen presenting cells (APCs) carrying brain antigens in draining lymphoid tissue of stroke patients support the notion that stroke might induce antigen-specific immune responses. After stroke, brain proteins that are normally hidden from the periphery, inflammatory mediators, and danger signals can exit the brain through several efflux routes. They can reach the blood after leaking out of the damaged blood-brain barrier (BBB) or following the drainage of interstitial fluid to the dural venous sinus, or reach the cervical lymph nodes through the nasal lymphatics following CSF drainage along the arachnoid sheaths of nerves across the nasal submucosa. The route and mode of access of brain antigens to lymphoid tissue could influence the type of response. Central and peripheral tolerance prevents autoimmunity, but the actual mechanisms of tolerance to brain antigens released into the periphery in the presence of inflammation, danger signals, and APCs, are not fully characterized. Stroke does not systematically trigger autoimmunity, but under certain circumstances, such as pronounced systemic inflammation or infection, autoreactive T cells could escape the tolerance controls. Further investigation is needed to elucidate whether antigen-specific immune events could underlie neurological complications impairing recovery from stroke.
Collapse
Affiliation(s)
- Xabier Urra
- Functional Unit of Cerebrovascular Diseases, Hospital Clínic Barcelona, Spain ; August Pi i Sunyer Biomedical Research Institute (IDIBAPS) Barcelona, Spain
| | - Francesc Miró
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS) Barcelona, Spain
| | - Angel Chamorro
- Functional Unit of Cerebrovascular Diseases, Hospital Clínic Barcelona, Spain ; August Pi i Sunyer Biomedical Research Institute (IDIBAPS) Barcelona, Spain
| | - Anna M Planas
- August Pi i Sunyer Biomedical Research Institute (IDIBAPS) Barcelona, Spain ; Department of Brain Ischemia and Neurodegeneration, Instituto de Investigaciones Biomédicas de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC) Barcelona, Spain
| |
Collapse
|