1
|
Lei K, Wu R, Wang J, Lei X, Zhou E, Fan R, Gong L. Sirtuins as Potential Targets for Neuroprotection: Mechanisms of Early Brain Injury Induced by Subarachnoid Hemorrhage. Transl Stroke Res 2024; 15:1017-1034. [PMID: 37779164 PMCID: PMC11522081 DOI: 10.1007/s12975-023-01191-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/26/2023] [Accepted: 08/31/2023] [Indexed: 10/03/2023]
Abstract
Subarachnoid hemorrhage (SAH) is a prevalent cerebrovascular disease with significant global mortality and morbidity rates. Despite advancements in pharmacological and surgical approaches, the quality of life for SAH survivors has not shown substantial improvement. Traditionally, vasospasm has been considered a primary contributor to death and disability following SAH, but anti-vasospastic therapies have not demonstrated significant benefits for SAH patients' prognosis. Emerging studies suggest that early brain injury (EBI) may play a crucial role in influencing SAH prognosis. Sirtuins (SIRTs), a group of NAD + -dependent deacylases comprising seven mammalian family members (SIRT1 to SIRT7), have been found to be involved in neural tissue development, plasticity, and aging. They also exhibit vital functions in various central nervous system (CNS) processes, including cognition, pain perception, mood, behavior, sleep, and circadian rhythms. Extensive research has uncovered the multifaceted roles of SIRTs in CNS disorders, offering insights into potential markers for pathological processes and promising therapeutic targets (such as SIRT1 activators and SIRT2 inhibitors). In this article, we provide an overview of recent research progress on the application of SIRTs in subarachnoid hemorrhage and explore their underlying mechanisms of action.
Collapse
Affiliation(s)
- Kunqian Lei
- Department of Neurosurgery, Affiliated Hospital of Zunyi Medical University CN, Zunyi, China
| | - Rui Wu
- Department of Neurosurgery, Affiliated Hospital of Zunyi Medical University CN, Zunyi, China
| | - Jin Wang
- Department of Neurology, Affiliated Hospital of Zunyi Medical University CN, Zunyi, China
| | - Xianze Lei
- Department of Neurology, Affiliated Hospital of Zunyi Medical University CN, Zunyi, China
| | - Erxiong Zhou
- Department of Neurosurgery, Affiliated Hospital of Zunyi Medical University CN, Zunyi, China
| | - Ruiming Fan
- Department of Neurosurgery, Affiliated Hospital of Zunyi Medical University CN, Zunyi, China.
| | - Lei Gong
- Department of Pharmacy, Institute of Medical Biotechnology, Affiliated Hospital of Zunyi Medical University CN, Zunyi, China.
| |
Collapse
|
2
|
Zhang H, Xu L, He Y, Zhang Z, Zhang J, Yu Q, Liu Y, Wang X, Zhang A, Wang K, Fang Y, Chen S. Tat-NR2B9c attenuates oxidative stress via inhibition of PSD95-NR2B-nNOS complex after subarachnoid hemorrhage in rats. Neuropharmacology 2024; 251:109905. [PMID: 38521229 DOI: 10.1016/j.neuropharm.2024.109905] [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: 10/02/2023] [Revised: 12/18/2023] [Accepted: 03/07/2024] [Indexed: 03/25/2024]
Abstract
Oxidative stress plays important roles in the pathogenesis of early brain injury (EBI) after subarachnoid hemorrhage (SAH). Tat-NR2B9c has shown efficacy as a neuroprotective agent in several studies. Here, we identified the neuroprotective role of Tat-NR2B9c after SAH and its related mechanisms. The results showed that Tat-NR2B9c treatment attenuated oxidative stress, therefore alleviated neuronal apoptosis and neurological deficits after SAH. Tat-NR2B9c treatment could alleviate mitochondrial vacuolization induced by SAH. Compared to SAH + vehicle group, Tat-NR2B9c resulted in the decrease of Acetylated superoxide dismutase2 (Ac-SOD2), Bcl-2-associated X protein (Bax) and cleaved-caspase3 (CC3) protein expression, and the up-regulation of Sirtunin 3 (Sirt3) and Bcl-2 protein level. Moreover, Tat-NR2B9c attenuated excitotoxicity by inhibiting the interaction of PSD95-NR2B-nNOS. Our results demonstrated that Tat-NR2B9c inhibited oxidative stress via inhibition of PSD95-NR2B-nNOS complex formation after SAH. Tat-NR2B9c may serve as a potential treatment for SAH induced brain injury.
Collapse
Affiliation(s)
- Haocheng Zhang
- Department of Neurosurgery, School of Medicine, The Second Affiliated Hospital, Zhejiang University, Zhejiang Province, Hangzhou, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Longbiao Xu
- Department of Neurosurgery, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Yezhao He
- Department of Neurosurgery, School of Medicine, The Second Affiliated Hospital, Zhejiang University, Zhejiang Province, Hangzhou, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Zeyu Zhang
- Department of Neurosurgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Jiahao Zhang
- Department of Neurosurgery, School of Medicine, The Second Affiliated Hospital, Zhejiang University, Zhejiang Province, Hangzhou, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Qian Yu
- Department of Neurosurgery, School of Medicine, The Second Affiliated Hospital, Zhejiang University, Zhejiang Province, Hangzhou, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Yibo Liu
- Department of Neurosurgery, School of Medicine, The Second Affiliated Hospital, Zhejiang University, Zhejiang Province, Hangzhou, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Xiaoyu Wang
- Department of Neurosurgery, School of Medicine, The Second Affiliated Hospital, Zhejiang University, Zhejiang Province, Hangzhou, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Anke Zhang
- Department of Neurosurgery, School of Medicine, The Second Affiliated Hospital, Zhejiang University, Zhejiang Province, Hangzhou, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Kaikai Wang
- Department of Neurosurgery, School of Medicine, The Second Affiliated Hospital, Zhejiang University, Zhejiang Province, Hangzhou, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China
| | - Yuanjian Fang
- Department of Neurosurgery, School of Medicine, The Second Affiliated Hospital, Zhejiang University, Zhejiang Province, Hangzhou, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
| | - Sheng Chen
- Department of Neurosurgery, School of Medicine, The Second Affiliated Hospital, Zhejiang University, Zhejiang Province, Hangzhou, China; Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
| |
Collapse
|
3
|
Busse TL, Munthe S, Ketharanathan B, Bülow K, Jóhannsson B, Diaz A, Nielsen TH. Perfusion Computed Tomography as a Screening Tool for Pending Delayed Cerebral Ischemia in Comatose Patients After Aneurysmal Subarachnoid Hemorrhage: A Retrospective Cohort Study. Neurocrit Care 2024; 40:964-975. [PMID: 37821720 PMCID: PMC11147906 DOI: 10.1007/s12028-023-01855-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 09/05/2023] [Indexed: 10/13/2023]
Abstract
BACKGROUND Aneurysmal subarachnoid hemorrhage (aSAH) is frequently complicated by delayed cerebral ischemia (DCI), leading to poor outcomes. Early diagnosis of DCI is crucial for improving survival and outcomes but remains challenging in comatose patients. In this study, we aimed to evaluate computed tomography with angiography and perfusion (P-CT) as a screening modality on postictal days four and eight for impending DCI after aSAH in comatose patients using vasospasm with hypoperfusion (hVS) as a surrogate and DCI-related infarction as an outcome measure. Two objectives were set: (1) to evaluate the screening's ability to accurately risk stratify patients and (2) to assess the validity of P-CT screening. METHODS We conducted a retrospective review of the records of comatose patients with aSAH from January 2019 to December 2021 who were monitored with P-CT scans on days four and eight. The event rates of DCI-related infarction, hVS, and endovascular rescue therapy (ERT) were analyzed, and the sensitivity, specificity, negative predictive value (NPV), and positive predictive value (PPV) for DCI were calculated. DCI-related infarction was defined as new secondary cerebral infarction > 48 h < 6 weeks post aSAH not attributable to other causes, and hVS was defined as arterial narrowing with corresponding hypoperfusion on P-CT. RESULTS Fifty-six comatose patients were included, and 98 P-CT scans were performed. The incidence of DCI-related infarction was 40%. Screening P-CT on days four and eight found vasospasm in 23% of all patients, including 11% with hVS. A positive hVS on day four or eight revealed a relative risk of 2.4 [95% confidence interval (CI) 1.13-5.11, p = 0.03], sensitivity of 23% (95% CI 8-45, p = 0.03), specificity of 95% (95% CI 36-100, p = 0.03), PPV of 0.83 (95% CI 0.36-1.00, p = 0.03), and NPV of 0.65 (95% CI 0.50-0.78). Six positive P-CT scans led to digital subtraction angiography in five patients, three of whom received ERT. All ERT-intervened patients developed DCI-related infarction. CONCLUSIONS P-CT resulted in few interventions and often resulted in late detection of DCI at an irreversible stage. Although a positive P-CT result accurately predicts impending DCI-related infarction, screening on days four and eight alone in comatose patients with aSAH often fails to timely detect impending DCI. Based on our analysis, we cannot recommend P-CT as a screening modality. P-CT is likely best used as a confirmatory test prior to invasive interventions when guided by continuous multimodal monitoring; however, prospective studies with comparison groups are warranted. The need for a reliable continuous screening modality is evident because of the high rate of deterioration and narrow treatment window.
Collapse
Affiliation(s)
- Thor Löwe Busse
- Department of Neurosurgery, Odense University Hospital, Odense, Denmark.
| | - Sune Munthe
- Department of Neurosurgery, Odense University Hospital, Odense, Denmark
| | | | - Karsten Bülow
- Department of Anaesthesiology and Intensive Care, Odense University Hospital, Odense, Denmark
| | - Bjarni Jóhannsson
- Department of Neurosurgery, Odense University Hospital, Odense, Denmark
| | - Anabel Diaz
- Department of Radiology, Odense University Hospital, Odense, Denmark
| | | |
Collapse
|
4
|
Zhang J, Zhang Z, Wang X, Liu Y, Yu Q, Wang K, Fang Y, Lenahan C, Chen M, Chen S. Connection between oxidative stress and subcellular organelle in subarachnoid hemorrhage: Novel mechanisms and therapeutic implications. CNS Neurosci Ther 2023; 29:3672-3683. [PMID: 37408392 PMCID: PMC10651993 DOI: 10.1111/cns.14348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 06/12/2023] [Accepted: 06/24/2023] [Indexed: 07/07/2023] Open
Abstract
Spontaneous subarachnoid hemorrhage (SAH) is one of the most devastating forms of stroke, with limited treatment modalities and poor patient outcomes. Previous studies have proposed multiple prognostic factors; however, relative research on treatment has not yet yielded favorable clinical outcomes. Moreover, recent studies have suggested that early brain injury (EBI) occurring within 72 h after SAH may contribute to its poor clinical outcomes. Oxidative stress is recognized as one of the main mechanisms of EBI, which causes damage to various subcellular organelles, including the mitochondria, nucleus, endoplasmic reticulum (ER), and lysosomes. This could lead to significant impairment of numerous cellular functions, such as energy supply, protein synthesis, and autophagy, which may directly contribute to the development of EBI and poor long-term prognostic outcomes. In this review, the mechanisms underlying the connection between oxidative stress and subcellular organelles after SAH are discussed, and promising therapeutic options based on these mechanisms are summarized.
Collapse
Affiliation(s)
- Jiahao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Clinical Research Center for Neurological Diseases of Zhejiang ProvinceHangzhouChina
| | - Zeyu Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Clinical Research Center for Neurological Diseases of Zhejiang ProvinceHangzhouChina
- Department of Neurosurgery, Renji Hospital, School of MedicineShanghai Jiao Tong UniversityShanghaiChina
| | - Xiaoyu Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Clinical Research Center for Neurological Diseases of Zhejiang ProvinceHangzhouChina
| | - Yibo Liu
- Department of Neurosurgery, The Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Clinical Research Center for Neurological Diseases of Zhejiang ProvinceHangzhouChina
| | - Qian Yu
- Department of Neurosurgery, The Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Clinical Research Center for Neurological Diseases of Zhejiang ProvinceHangzhouChina
| | - Kaikai Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Clinical Research Center for Neurological Diseases of Zhejiang ProvinceHangzhouChina
| | - Yuanjian Fang
- Department of Neurosurgery, The Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Clinical Research Center for Neurological Diseases of Zhejiang ProvinceHangzhouChina
| | - Cameron Lenahan
- Center for Neuroscience ResearchLoma Linda University School of MedicineLoma LindaCaliforniaUSA
| | - Maohua Chen
- Department of Neurosurgery, Wenzhou Central HospitalAffiliated Dingli Clinical Institute of Wenzhou Medical UniversityWenzhouChina
| | - Sheng Chen
- Department of Neurosurgery, The Second Affiliated Hospital, School of MedicineZhejiang UniversityHangzhouChina
- Clinical Research Center for Neurological Diseases of Zhejiang ProvinceHangzhouChina
| |
Collapse
|
5
|
Zhang T, Zuo G, Zhang H. GPR18 Agonist Resolvin D2 Reduces Early Brain Injury in a Rat Model of Subarachnoid Hemorrhage by Multiple Protective Mechanisms. Cell Mol Neurobiol 2022; 42:2379-2392. [PMID: 34089427 DOI: 10.1007/s10571-021-01114-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Accepted: 05/31/2021] [Indexed: 10/21/2022]
Abstract
Early brain injury (EBI) is the early phase of secondary complications arising from subarachnoid hemorrhage (SAH). G protein-coupled receptor 18 (GPR18) can exert neuroprotective effects during ischemia. In this study, we investigated the roles of GPR18 in different brain regions during EBI using a GPR18 agonist, resolvin D2 (RvD2). Location and dynamics of GPR18 expression were assessed by immunohistochemistry and western blotting in a rat model of SAH based on endovascular perforation. RvD2 was given intranasally at 1 h after SAH, and SAH grade, brain water content and behavior were assayed before sacrifice. TUNEL and dihydroethidium staining of the cortex were performed at 24 h after SAH. Selected brain regions were also examined for pathway related proteins using immunofluorescence and Western blotting. We found that GPR18 was expressed in meninges, hypothalamus, cortex and white matter before EBI. After SAH, GPR18 expression was increased in meninges and hypothalamus but decreased in cortex and white matter. RvD2 improved neurological scores and brain edema after SAH. RvD2 attenuated mast cell degranulation and reduced expression of chymase and tryptase expression in the meninges. In the hypothalamus, RvD2 attenuated inflammation, increased expression of proopiomelanocortin and interleukin-10, as well as decreased expression of nerve peptide Y and tumor necrosis factor-α. In cortex, RvD2 alleviated oxidative stress and apoptosis, and protected the blood-brain barrier. RvD2 also ameliorated white matter injury by elevating myelin basic protein and suppressing amyloid precursor protein. Our results suggest that GPR18 may help protect multiple brain regions during EBI, particularly in the cortex and hypothalamus. Upregulating GPR18 by RvD2 may improve neurological functions in different brain regions via multiple mechanisms.
Collapse
Affiliation(s)
- Tongyu Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun St., Beijing, 100053, China
| | - Gang Zuo
- Department of Neurosurgery, The Affiliated Taicang Hospital, Soochow University, Taicang, Suzhou, 215400, Jiangsu, China
| | - Hongqi Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, 45 Changchun St., Beijing, 100053, China.
| |
Collapse
|
6
|
Zhang Z, Zhang A, Liu Y, Hu X, Fang Y, Wang X, Luo Y, Lenahan C, Chen S. New Mechanisms and Targets of Subarachnoid Hemorrhage: A Focus on Mitochondria. Curr Neuropharmacol 2022; 20:1278-1296. [PMID: 34720082 PMCID: PMC9881073 DOI: 10.2174/1570159x19666211101103646] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 10/06/2021] [Accepted: 10/28/2021] [Indexed: 11/22/2022] Open
Abstract
Spontaneous subarachnoid hemorrhage (SAH) accounts for 5-10% of all strokes and is a subtype of hemorrhagic stroke that places a heavy burden on health care. Despite great progress in surgical clipping and endovascular treatment for ruptured aneurysms, cerebral vasospasm (CVS) and delayed cerebral ischemia (DCI) threaten the long-term outcomes of patients with SAH. Moreover, there are limited drugs available to reduce the risk of DCI and adverse outcomes in SAH patients. New insight suggests that early brain injury (EBI), which occurs within 72 h after the onset of SAH, may lay the foundation for further DCI development and poor outcomes. The mechanisms of EBI mainly include excitotoxicity, oxidative stress, neuroinflammation, blood-brain barrier (BBB) destruction, and cellular death. Mitochondria are a double-membrane organelle, and they play an important role in energy production, cell growth, differentiation, apoptosis, and survival. Mitochondrial dysfunction, which can lead to mitochondrial membrane potential (Δψm) collapse, overproduction of reactive oxygen species (ROS), release of apoptogenic proteins, disorders of mitochondrial dynamics, and activation of mitochondria-related inflammation, is considered a novel mechanism of EBI related to DCI as well as post-SAH outcomes. In addition, mitophagy is activated after SAH. In this review, we discuss the latest perspectives on the role of mitochondria in EBI and DCI after SAH. We emphasize the potential of mitochondria as therapeutic targets and summarize the promising therapeutic strategies targeting mitochondria for SAH.
Collapse
Affiliation(s)
- Zeyu Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; ,These authors contributed equally to this work.
| | - Anke Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; ,These authors contributed equally to this work.
| | - Yibo Liu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; ,These authors contributed equally to this work.
| | - Xiaoming Hu
- Department of Neurosurgery, Taizhou Hospital, Taizhou, Zhejiang Province, China;
| | - Yuanjian Fang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China;
| | - Xiaoyu Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China;
| | - Yujie Luo
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China;
| | - Cameron Lenahan
- Center for Neuroscience Research, Loma Linda University School of Medicine, Loma Linda, CA, USA
| | - Sheng Chen
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China; ,Address correspondence to this author at the Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang Province, China; Tel: +86-571-87784815; Fax: +86-571-87784755; E-mail:
| |
Collapse
|
7
|
The role of autophagy and apoptosis in early brain injury after subarachnoid hemorrhage: an updated review. Mol Biol Rep 2022; 49:10775-10782. [PMID: 35819555 DOI: 10.1007/s11033-022-07756-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 06/29/2022] [Indexed: 12/11/2022]
Abstract
Subarachnoid hemorrhage (SAH) is a worldwide devastating type of stroke with high mortality and morbidity. Accumulating evidence show early brain injury (EBI) as the leading cause of mortality after SAH. The pathological processes involved in EBI include decreased cerebral blood flow, increased intracranial pressure, vasospasm, and disruption of the blood-brain barrier. In addition, neuroinflammation, oxidative stress, apoptosis, and autophagy have also been proposed to contribute to EBI. Among the various processes involved in EBI, neuronal apoptosis has been proven to be a key factor contributing to the poor prognosis of SAH patients. Meanwhile, as another important catabolic process maintaining the cellular and tissue homeostasis, autophagy has been shown to be neuroprotective after SAH. Studies have shown that enhancing autophagy reduced apoptosis, whereas inhibiting autophagy aggravate neuronal apoptosis after SAH. The physiological substrates and mechanisms of neuronal autophagy and apoptosis by which defects in neuronal function are largely unknown. In this review, we summarize and discuss the role of autophagy and apoptosis after SAH and contribute to further study for investigation of the means to control the balance between them.
Collapse
|
8
|
Solár P, Zamani A, Lakatosová K, Joukal M. The blood-brain barrier and the neurovascular unit in subarachnoid hemorrhage: molecular events and potential treatments. Fluids Barriers CNS 2022; 19:29. [PMID: 35410231 PMCID: PMC8996682 DOI: 10.1186/s12987-022-00312-4] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 01/24/2022] [Indexed: 12/12/2022] Open
Abstract
The response of the blood-brain barrier (BBB) following a stroke, including subarachnoid hemorrhage (SAH), has been studied extensively. The main components of this reaction are endothelial cells, pericytes, and astrocytes that affect microglia, neurons, and vascular smooth muscle cells. SAH induces alterations in individual BBB cells, leading to brain homeostasis disruption. Recent experiments have uncovered many pathophysiological cascades affecting the BBB following SAH. Targeting some of these pathways is important for restoring brain function following SAH. BBB injury occurs immediately after SAH and has long-lasting consequences, but most changes in the pathophysiological cascades occur in the first few days following SAH. These changes determine the development of early brain injury as well as delayed cerebral ischemia. SAH-induced neuroprotection also plays an important role and weakens the negative impact of SAH. Supporting some of these beneficial cascades while attenuating the major pathophysiological pathways might be decisive in inhibiting the negative impact of bleeding in the subarachnoid space. In this review, we attempt a comprehensive overview of the current knowledge on the molecular and cellular changes in the BBB following SAH and their possible modulation by various drugs and substances.
Collapse
Affiliation(s)
- Peter Solár
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic
- Department of Neurosurgery, Faculty of Medicine, Masaryk University and St. Anne's University Hospital Brno, Pekařská 53, 656 91, Brno, Czech Republic
| | - Alemeh Zamani
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic
| | - Klaudia Lakatosová
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic
| | - Marek Joukal
- Department of Anatomy, Cellular and Molecular Neurobiology Research Group, Faculty of Medicine, Masaryk University, 625 00, Brno, Czech Republic.
| |
Collapse
|
9
|
Kalra P, Khan H, Kaur A, Singh TG. Mechanistic Insight on Autophagy Modulated Molecular Pathways in Cerebral Ischemic Injury: From Preclinical to Clinical Perspective. Neurochem Res 2022; 47:825-843. [PMID: 34993703 DOI: 10.1007/s11064-021-03500-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 12/26/2022]
Abstract
Cerebral ischemia is one of the most devastating brain injuries and a primary cause of acquired and persistent disability worldwide. Despite ongoing therapeutic interventions at both the experimental and clinical levels, options for stroke-related brain injury are still limited. Several evidence suggests that autophagy is triggered in response to cerebral ischemia, therefore targeting autophagy-related signaling pathways can provide a new direction for the therapeutic implications in the ischemic injury. Autophagy is a highly conserved lysosomal-dependent pathway that degrades and recycles damaged or non-essential cellular components to maintain neuronal homeostasis. But, whether autophagy activation promotes cell survival against ischemic injury or, on the contrary, causes neuronal death is still under debate. We performed an extensive literature search from PubMed, Bentham and Elsevier for various aspects related to molecular mechanisms and pathobiology involved in autophagy and several pre-clinical studies justifiable further in the clinical trials. Autophagy modulates various downstream molecular cascades, i.e., mTOR, NF-κB, HIF-1, PPAR-γ, MAPK, UPR, and ROS pathways in cerebral ischemic injury. In this review, the various approaches and their implementation in the translational research in ischemic injury into practices has been covered. It will assist researchers in finding a way to cross the unbridgeable chasm between the pre-clinical and clinical studies.
Collapse
Affiliation(s)
- Palak Kalra
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Heena Khan
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Amarjot Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
10
|
Camara R, Matei N, Camara J, Enkhjargal B, Tang J, Zhang JH. Hydrogen gas therapy improves survival rate and neurological deficits in subarachnoid hemorrhage rats: a pilot study. Med Gas Res 2020; 9:74-79. [PMID: 31249255 PMCID: PMC6607870 DOI: 10.4103/2045-9912.260648] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
The high morbidity, high mortality, and significant shortage of effective therapies for subarachnoid hemorrhage (SAH) have created an urgency to discover novel therapies. Human studies in Asia have established the safety of hydrogen gas in the treatment of hepatic, renal, pulmonary, and cardiac diseases. Mechanistically, hydrogen gas has been shown to affect oxidative stress, inflammation, and apoptosis. We hypothesized that hydrogen therapy would improve neurological function and increase survival rate in SAH. High dose hydrogen gas (66% at 3 L/min) was administered for 2 hours at 0.5, 8, and 18 hours after SAH. This treatment increased 72-hour survival rate and provided 24-hour neuroprotection after SAH in rats. To our knowledge, this is the first report demonstrating that high dose hydrogen gas therapy reduces mortality and improves outcome after SAH. Our results correlate well with the proposed mechanisms of hydrogen gas therapy within the literature. We outline four pathways and downstream targets of hydrogen gas potentially responsible for our results. A potentially complex network of pathways responsible for the efficacy of hydrogen gas therapy, along with a limited mechanistic understanding of these pathways, justifies further investigation to provide a basis for clinical trials and the advancement of hydrogen gas therapy in humans. This study was approved by the Institutional Animal Care and Use Committee of Loma Linda University, USA (Approval No. 8160016) in May 2016.
Collapse
Affiliation(s)
- Richard Camara
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Nathanael Matei
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Justin Camara
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Budbazar Enkhjargal
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - John H Zhang
- Department of Physiology and Pharmacology; Department of Anesthesiology; Department of Neurosurgery, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
11
|
Zheng ZV, Lam PK, Poon WS, Wong KCG. The Time Course of Cognitive Deficits in Experimental Subarachnoid Hemorrhage. ACTA NEUROCHIRURGICA. SUPPLEMENT 2019; 127:121-125. [PMID: 31407072 DOI: 10.1007/978-3-030-04615-6_18] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Subarachnoid hemorrhage (SAH) is a devastating stroke type. Approximately 50% of survivors suffer from the permanent disability, caused by the cognitive deficits. To enrich the pre-clinical research on the neurological deficits after SAH, we investigate the temporal cognitive deficits and the longitudinal course of cognitive recovery in endovascular perforation SAH murine model. The SAH mice show reproducible body weakness and headache-symbolized moaning symptoms, which is closed to clinical patients. SAH mice exhibit significantly impaired cognitive function in domains of learning ability, short-term and long-term memory. The cognitive deficits occur mostly in the early phase and recover gradually till day 10 after SAH. The systematical assessments of cognitive function after experimental aneurysmal SAH elucidate the time course of cognitive deficits and provide the time window of potential interventions.
Collapse
Affiliation(s)
- Zhiyuan Vera Zheng
- Division of Neurosurgery, Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Ping Kuen Lam
- Department of Surgery, Chow Tai Fook-Cheng Yu Tung Surgical Stem cell Research Centre, The Chinese University of Hong Kong, Hong Kong, China
| | - Wai Sang Poon
- Division of Neurosurgery, Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China
| | - Kwok Chu George Wong
- Division of Neurosurgery, Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Hong Kong, China.
| |
Collapse
|
12
|
You ZQ, Wu Q, Zhou XM, Zhang XS, Yuan B, Wen LL, Xu WD, Cui S, Tang XL, Zhang X. Receptor-Mediated Delivery of Astaxanthin-Loaded Nanoparticles to Neurons: An Enhanced Potential for Subarachnoid Hemorrhage Treatment. Front Neurosci 2019; 13:989. [PMID: 31619957 PMCID: PMC6759683 DOI: 10.3389/fnins.2019.00989] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 09/02/2019] [Indexed: 12/13/2022] Open
Abstract
Astaxanthin (ATX) is a carotenoid that exerts strong anti-oxidant and anti-inflammatory property deriving from its highly unsaturated molecular structures. However, the low stability and solubility of ATX results in poor bioavailability, which markedly hampers its application as therapeutic agent in clinic advancement. This study investigated a promising way of transferrin conjugated to poly (ethylene glycol) (PEG)-encapsulated ATX nanoparticles (ATX-NPs) on targeted delivery and evaluated the possible mechanism underlying neuroprotection capability. As a result, the ATX integrated into nanocarrier presented both well water-dispersible and biocompatible, primely conquering its limitations. More than that, the transferrin-containing ATX-NPs exhibited enhanced cellular uptake efficiency than that of ATX-NPs without transferrin conjugated in primary cortical neurons. Additionally, compared to free ATX, transferrin-containing ATX-NPs with lower ATX concentration showed powerful neuroprotective effects on OxyHb-induced neuronal damage. Taken together, the improved bioavailability and enhanced neuroprotective effects enabled ATX-NPs as favorable candidates for targeted delivery and absorption of ATX. We believe that these in vitro findings will provide insights for advancement of subarachnoid hemorrhage therapy.
Collapse
Affiliation(s)
- Zong-Qi You
- Department of Neurosurgery, School of Medicine, Jinling Hospital, Jiangsu University, Zhenjiang, China
| | - Qi Wu
- Department of Neurosurgery, School of Medicine, Jinling Hospital, Nanjing University, Nanjing, China
| | - Xiao-Ming Zhou
- Department of Neurosurgery, School of Medicine, Jinling Hospital, Second Military Medical University, Shanghai, China
| | - Xiang-Sheng Zhang
- Department of Neurosurgery, School of Medicine, Jinling Hospital, Nanjing University, Nanjing, China
| | - Bin Yuan
- Department of Neurosurgery, School of Medicine, Jinling Hospital, Nanjing Medical University, Nanjing, China
| | - Li-Li Wen
- Department of Neurosurgery, School of Medicine, Jinling Hospital, Nanjing Medical University, Nanjing, China
| | - Wei-Dong Xu
- Department of Neurosurgery, School of Medicine, Jinling Hospital, Southern Medical University (Guangzhou), Nanjing, China
| | - Sheng Cui
- College of Material Sciences and Engineering, Nanjing Tech University, Nanjing, China
| | - Xiang-Long Tang
- College of Material Sciences and Engineering, Nanjing Tech University, Nanjing, China
| | - Xin Zhang
- Department of Neurosurgery, School of Medicine, Jinling Hospital, Jiangsu University, Zhenjiang, China.,Department of Neurosurgery, School of Medicine, Jinling Hospital, Nanjing University, Nanjing, China.,Department of Neurosurgery, School of Medicine, Jinling Hospital, Nanjing Medical University, Nanjing, China
| |
Collapse
|
13
|
Zhang T, Wu P, Budbazar E, Zhu Q, Sun C, Mo J, Peng J, Gospodarev V, Tang J, Shi H, Zhang JH. Mitophagy Reduces Oxidative Stress Via Keap1 (Kelch-Like Epichlorohydrin-Associated Protein 1)/Nrf2 (Nuclear Factor-E2-Related Factor 2)/PHB2 (Prohibitin 2) Pathway After Subarachnoid Hemorrhage in Rats. Stroke 2019; 50:978-988. [PMID: 30890112 PMCID: PMC6433519 DOI: 10.1161/strokeaha.118.021590] [Citation(s) in RCA: 112] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 01/30/2019] [Indexed: 11/16/2022]
Abstract
Background and Purpose- Mitoquinone has been reported as a mitochondria-targeting antioxidant to promote mitophagy in various chronic diseases. Here, our aim was to study the role of mitoquinone in mitophagy activation and oxidative stress-induced neuronal death reduction after subarachnoid hemorrhage (SAH) in rats. Methods- Endovascular perforation was used for SAH model of male Sprague-Dawley rats. Exogenous mitoquinone was injected intraperitoneally 1 hour after SAH. ML385, an inhibitor of Nrf2 (nuclear factor-E2-related factor 2), was given intracerebroventricularly 24 hours before SAH. Small interfering RNA for PHB2 (prohibitin 2) was injected intracerebroventricularly 48 hours before SAH. Nuclear, mitochondrial, and cytoplasmic fractions were gathered using nucleus and mitochondria isolation kits. SAH grade evaluation, short- and long- term neurological function tests, oxidative stress, and apoptosis measurements were performed. Pathway related proteins were investigated with Western blot and immunofluorescence staining. Results- Expression of Keap1 (Kelch-like epichlorohydrin-associated protein 1, 2.84× at 24 hours), Nrf2 (2.78× at 3 hours), and LC3II (light chain 3-II; 1.94× at 24 hours) increased, whereas PHB2 (0.46× at 24 hours) decreased after SAH compared with sham group. Mitoquinone treatment attenuated oxidative stress and neuronal death, both short-term and long-term. Administration of mitoquinone resulted in a decrease in expression of Keap1 (0.33×), Romo1 (reactive oxygen species modulator 1; 0.24×), Bax (B-cell lymphoma-2 associated X protein; 0.31×), Cleaved Caspase-3 (0.29×) and an increase in Nrf2 (2.13×), Bcl-xl (B-cell lymphoma-extra large; 1.67×), PINK1 (phosphatase and tensin-induced kinase 1; 1.67×), Parkin (1.49×), PHB2 (1.60×), and LC3II (1.67×) proteins compared with SAH+vehicle group. ML385 abolished the treatment effects of mitoquinone on behavior and protein levels. PHB2 small interfering RNA reversed the outcomes of mitoquinone administration through reduction in protein expressions downstream of PHB2. Conclusions- Mitoquinone inhibited oxidative stress-related neuronal death by activating mitophagy via Keap1/Nrf2/PHB2 pathway after SAH. Mitoquinone may serve as a potential treatment to relieve brain injury after SAH.
Collapse
Affiliation(s)
- Tongyu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Pei Wu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Enkhjargal Budbazar
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Qiquan Zhu
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Chengmei Sun
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Jun Mo
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Jianhua Peng
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Vadim Gospodarev
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Huaizhang Shi
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - John H. Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
14
|
Yang C, Li T, Xue H, Wang L, Deng L, Xie Y, Bai X, Xin D, Yuan H, Qiu J, Wang Z, Li G. Inhibition of Necroptosis Rescues SAH-Induced Synaptic Impairments in Hippocampus via CREB-BDNF Pathway. Front Neurosci 2019; 12:990. [PMID: 30666179 PMCID: PMC6330293 DOI: 10.3389/fnins.2018.00990] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 12/10/2018] [Indexed: 12/13/2022] Open
Abstract
Subarachnoid hemorrhage (SAH) is a devastating form of stroke that leads to incurable outcomes. Increasing evidence has proved that early brain injury (EBI) contributes mostly to unfavorable outcomes after SAH. A previously unknown mechanism of regulated cell death known as necroptosis has recently been reported. Necrostatin-1 (nec-1), a specific and potent inhibitor of necroptosis, can attenuate brain impairments after SAH. However, the effect of nec-1 on the hippocampus and its neuroprotective impact on synapses after SAH is not well understood. Our present study was designed to investigate the potential effects of nec-1 administration on synapses and its relevant signal pathway in EBI after SAH. Nec-1 was administrated in a rat model via intracerebroventricular injection after SAH. Neurobehavior scores and brain edema were detected at 24 h after SAH occurred. The expression of the receptor-interacting proteins 1 and 3 (RIP1and3) was examined as a marker of necroptosis. We used hematoxylin and eosin staining, Nissl staining, silver staining and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) to observe the morphological changes in hippocampus. The protective effect of nec-1 on synapses was evaluated using western blotting and electron microscopy and Western blotting was used to detect the cAMP responsive element binding (CREB) protein and brain-derived neurotrophic factor (BDNF), and we used transmission electron microscopy and TUNEL to detect the protective effects of nec-1 when a specific inhibitor of CREB, known as 666-15, was used. Our results showed that in the SAH group, RIP1, and RIP3 significantly increased in the hippocampus. Additionally, injection of nec-1 alleviated brain edema and improved neurobehavior scores, compared with those in the SAH group. The damage to neurons was attenuated, and synaptic structure also improved in the Sham+nec-1 group. Furthermore, nec-1 treatment significantly enhanced the levels of phospho-CREB and BDNF compared with those in the SAH group. The protective effect of nec-1 could hindered by 666-15. Thus, nec-1 mitigated SAH-induced synaptic impairments in the hippocampus through the inhibition of necroptosis in connection with the CREB-BDNF pathway. This study may provide a new strategy for SAH patients in clinical practice.
Collapse
Affiliation(s)
- Chunlei Yang
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Tong Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China.,Department of Neurosurgery, Qingdao Municipal Hospital, Qingdao, China
| | - Hao Xue
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Lingxiao Wang
- Department of Physiology, Shandong University School of Basic Medical Sciences, Jinan, China
| | - Lin Deng
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| | - Yunkai Xie
- Department of Physiology, Shandong University School of Basic Medical Sciences, Jinan, China
| | - Xuemei Bai
- Department of Physiology, Shandong University School of Basic Medical Sciences, Jinan, China
| | - Danqing Xin
- Department of Physiology, Shandong University School of Basic Medical Sciences, Jinan, China
| | - Hongtao Yuan
- Department of Physiology, Shandong University School of Basic Medical Sciences, Jinan, China
| | - Jie Qiu
- Department of Physiology, Shandong University School of Basic Medical Sciences, Jinan, China
| | - Zhen Wang
- Department of Physiology, Shandong University School of Basic Medical Sciences, Jinan, China
| | - Gang Li
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Shandong Key Laboratory of Brain Function Remodeling, Jinan, China
| |
Collapse
|
15
|
Fang Y, Chen S, Reis C, Zhang J. The Role of Autophagy in Subarachnoid Hemorrhage: An Update. Curr Neuropharmacol 2018; 16:1255-1266. [PMID: 28382869 PMCID: PMC6251055 DOI: 10.2174/1570159x15666170406142631] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Revised: 03/16/2017] [Accepted: 04/05/2017] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Autophagy is an extensive self-degradation process for the disposition of cytosolic aggregated or misfolded proteins and defective organelles which executes the functions of pro-survival and pro-death to maintain cellular homeostasis. The pathway plays essential roles in several neurological disorders. Subarachnoid Hemorrhage (SAH) is a devastating subtype of hemorrhagic stroke with high risk of neurological deficit and high mortality. Early brain injury (EBI) plays a role in the poor clinical course and outcome after SAH. Recent studies have paid attention on the role of the autophagy pathway in the development of EBI after SAH. We aim to update the multifaceted roles of autophagy pathway in the pathogenesis of SAH, especially in the phase of EBI. METHODS We reviewed early researches related to autophagy and SAH. The following three aspects of contents will be mainly discussed: the process of the autophagy pathway, the role of the autophagy in SAH and the interaction between organelle dysfunction and autophagy pathway after SAH. RESULTS Accumulating evidence shows an increased autophagy reaction in response to early stages of SAH. However, others suggest inadequate or excessive autophagy activation can result in cell injury and death. In addition to autophagy, apoptosis and necrosis can occur in neurons simultaneously after SAH, leading to mixed features of cell death morphologies. And it is also known that there is extensive crosstalk between autophagy and apoptosis pathway. Subcellular organelles of neural cells generally participate in the formation and functional parts of autophagy process. CONCLUSION Autophagy plays an important role in the SAH-induced brain injury. A better understanding of the interrelationship among autophagy, apoptosis, and necrosis might provide us better therapeutic targets for the treatment of SAH.
Collapse
Affiliation(s)
- Yuanjian Fang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Sheng Chen
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Cesar Reis
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, United States.,Department of Preventive Medicine, Loma Linda University Medical Center, Loma Linda, CA, United States
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Brain Research Institute, Zhejiang University, Hangzhou, Zhejiang, China.,Collaborative Innovation Center for Brain Science, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
16
|
Huang L, Hou Y, Wang L, Xu X, Guan Q, Li X, Chen Y, Zhou W. p38 Inhibitor Protects Mitochondrial Dysfunction by Induction of DJ-1 Mitochondrial Translocation After Subarachnoid Hemorrhage. J Mol Neurosci 2018; 66:163-171. [PMID: 30242669 DOI: 10.1007/s12031-018-1131-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2018] [Accepted: 07/19/2018] [Indexed: 12/20/2022]
Abstract
p38 mitogen-activated protein kinase (MAPK) is a major player in mitochondrial dysfunction after subarachnoid hemorrhage (SAH). Moreover, DJ-1, which responds to oxidative stress and translocates to mitochondria, maintains mitochondrial homeostasis. Although a few studies have demonstrated that DJ-1 indirectly regulates p38 activation, the relationship between DJ-1 and p38 in mitochondrial dysfunction after SAH has not been delineated. Using an in vitro SAH model, alterations in p38, p-p38, DJ-1, and autophagic-related protein expression were detected. As expected, p38 inhibitor significantly blocked excessive expression of p38 and p-p38 after SAH, whereas total DJ-1 expression and mitochondrial DJ-1 were up-regulated. Further analysis showed that p38 inhibitor significantly blocked oxyhemoglobin (OxyHb) induced mitochondrial dysfunction, including mitochondrial membrane potential depolarization and reactive oxygen species (ROS) release. In addition, p38 inhibitor restored OxyHb-induced abnormal autophagic flux at the initiation and formation stage by regulating Atg5, beclin-1, the ratio of LC3-II/LC3-I, and p62 expression. This study suggested that overexpression of p38 induced the accumulation of mitochondrial dysfunction partly due to abnormal activation of autophagy, which largely relied on DJ-1 mitochondrial translocation.
Collapse
Affiliation(s)
- Liyong Huang
- Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, China
| | - Yaqing Hou
- Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, China
| | - Lei Wang
- Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, China
| | - Xiahui Xu
- Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, China
| | - Qingkai Guan
- Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, China
| | - Xiangsheng Li
- Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, China
| | - Ying Chen
- College of Life Science, Henan Normal University, Xinxiang, 453007, Henan, China
| | - Wenke Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, Henan, China.
| |
Collapse
|
17
|
Shao Z, Wu P, Wang X, Jin M, Liu S, Ma X, Shi H. Tetramethylpyrazine Protects Against Early Brain Injury and Inhibits the PERK/Akt Pathway in a Rat Model of Subarachnoid Hemorrhage. Neurochem Res 2018; 43:1650-1659. [PMID: 29951731 DOI: 10.1007/s11064-018-2581-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Revised: 06/14/2018] [Accepted: 06/19/2018] [Indexed: 12/25/2022]
Abstract
Neuronal apoptosis is a potentially fatal pathological process that occurs in early brain injury (EBI) after subarachnoid hemorrhage (SAH). There is an urgent need to identify effective therapeutics to alleviate neuronal apoptosis. Tetramethylpyrazine (TMP), as an important component of the Chinese traditional medicinal herb Ligusticum wallichii, has been widely used in China to treat cerebral ischemic injury and confer neuroprotection. In the present work, we investigate whether TMP can reduce EBI following SAH in rats, specifically via inactivating the PERK/Akt signaling cascade. One hundred twenty-five male Sprague-Dawley rats were used in the present study. TMP was administered by intravenous (i.v.) injection, and the Akt inhibitor MK2206 was injected intracerebroventricularly (i.c.v.). SAH grade, neurological scores, and brain water content were measured 24 h after SAH. Neuronal apoptosis was visualized by Fluoro-Jade C (FJC) staining. Western blotting was used to measure the levels of PERK, p-PERK, eIF2α, p-eIF2α, Akt, p-Akt, Bcl-2, Bax, and cleaved caspase-3. Our results showed that TMP effectively reduced neuronal apoptosis and improved neurobehavioral deficits 24 h after SAH. Administration of TMP reduced the abundance of p-PERK and p-eIF2α. In addition, TMP increased the p-Akt level and the Bcl-2/Bax ratio and decreased the level of cleaved caspase-3. The selective Akt inhibitor MK2206 abolished the anti-apoptotic effect of TMP at 24 h after SAH. Collectively, these results indicate that Akt-related anti-apoptosis through the PERK pathway is a major, potent mechanism of EBI. Further investigation of this pathway may provide a basis for the development of TMP as a clinical treatment.
Collapse
Affiliation(s)
- Zhengkai Shao
- Department of Neurosurgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Pei Wu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Xuefeng Wang
- Department of Neurosurgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Meishan Jin
- Department of Neurosurgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Shuang Liu
- Department of Neurosurgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Xudong Ma
- Department of Neurosurgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China
| | - Huaizhang Shi
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, People's Republic of China.
| |
Collapse
|
18
|
Pawlowska E, Szczepanska J, Wisniewski K, Tokarz P, Jaskólski DJ, Blasiak J. NF-κB-Mediated Inflammation in the Pathogenesis of Intracranial Aneurysm and Subarachnoid Hemorrhage. Does Autophagy Play a Role? Int J Mol Sci 2018; 19:E1245. [PMID: 29671828 PMCID: PMC5979412 DOI: 10.3390/ijms19041245] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 04/13/2018] [Accepted: 04/17/2018] [Indexed: 12/12/2022] Open
Abstract
The rupture of saccular intracranial aneurysms (IA) is the commonest cause of non-traumatic subarachnoid hemorrhage (SAH)—the most serious form of stroke with a high mortality rate. Aneurysm walls are usually characterized by an active inflammatory response, and NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) has been identified as the main transcription factor regulating the induction of inflammation-related genes in IA lesions. This transcription factor has also been related to IA rupture and resulting SAH. We and others have shown that autophagy interacts with inflammation in many diseases, but there is no information of such interplay in IA. Moreover, NF-κB, which is a pivotal factor controlling inflammation, is regulated by autophagy-related proteins, and autophagy is regulated by NF-κB signaling. It was also shown that autophagy mediates the normal functioning of vessels, so its disturbance can be associated with vessel-related disorders. Early brain injury, delayed brain injury, and associated cerebral vasospasm are among the most serious consequences of IA rupture and are associated with impaired function of the autophagy⁻lysosomal system. Further studies on the role of the interplay between autophagy and NF-κB-mediated inflammation in IA can help to better understand IA pathogenesis and to identify IA patients with an increased SAH risk.
Collapse
Affiliation(s)
- Elzbieta Pawlowska
- Department of Orthodontics, Medical University of Lodz, 92-216 Lodz, Poland.
| | - Joanna Szczepanska
- Department of Pediatric Dentistry, Medical University of Lodz, 92-216 Lodz, Poland.
| | - Karol Wisniewski
- Department of Neurosurgery and Neurooncology, Medical University of Lodz, Barlicki University Hospital, Kopcinskiego 22, 90-153 Lodz, Poland.
| | - Paulina Tokarz
- Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland.
| | - Dariusz J Jaskólski
- Department of Neurosurgery and Neurooncology, Medical University of Lodz, Barlicki University Hospital, Kopcinskiego 22, 90-153 Lodz, Poland.
| | - Janusz Blasiak
- Department of Molecular Genetics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland.
| |
Collapse
|
19
|
Li X, Peng J, Pang J, Wu Y, Huang X, Li Y, Zhou J, Gu L, Sun X, Chen L, Vitek MP, Jiang Y. Apolipoprotein E-Mimetic Peptide COG1410 Promotes Autophagy by Phosphorylating GSK-3β in Early Brain Injury Following Experimental Subarachnoid Hemorrhage. Front Neurosci 2018; 12:127. [PMID: 29556174 PMCID: PMC5844970 DOI: 10.3389/fnins.2018.00127] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 02/16/2018] [Indexed: 12/29/2022] Open
Abstract
COG1410, a mimetic peptide derived from the apolipoprotein E (apoE) receptor binding region, exerts positive effect on neurological deficits in early brain injury (EBI) after experimental subarachnoid hemorrhage (SAH). Currently the neuroprotective effect of COG1410 includes inhibiting BBB disruption, reducing neuronal apoptosis, and neuroinflammation. However, the effect and mechanism of COG1410 to subcellular organelles disorder have not been fully investigated. As the main pathway for recycling long-lived proteins and damaged organelles, neuronal autophagy is activated in SAH and exhibits neuroprotective effects by reducing the insults of EBI. Pharmacologically elevated autophagy usually contributes to alleviated brain injury, while few of the agents achieved clinical transformation. In this study, we explored the activation of autophagy during EBI by measuring the Beclin-1 and LC3B-II protein levels. Administration of COG1410 notably elevated the autophagic markers expression in neurons, simultaneously reversed the neurological deficits. Furthermore, the up-regulated autophagy by COG1410 was further promoted by p-GSK-3β agonist, whereas decreased by p-GSK-3β inhibitor. Taken together, these data suggest that the COG1410 might be a promising therapeutic strategy for EBI via promoting autophagy in SAH.
Collapse
Affiliation(s)
- Xinshen Li
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jianhua Peng
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jinwei Pang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yue Wu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xueping Huang
- Department of Neurosurgery, Luzhou People's Hospital, Luzhou, China
| | - Yong Li
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jian Zhou
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Long Gu
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiaochuan Sun
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ligang Chen
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Michael P Vitek
- Medicine, Duke University Medical Center, Durham, NC, United States
| | - Yong Jiang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
20
|
Şekerdağ E, Solaroğlu I, Gürsoy-Özdemir Y. Cell Death Mechanisms in Stroke and Novel Molecular and Cellular Treatment Options. Curr Neuropharmacol 2018; 16:1396-1415. [PMID: 29512465 PMCID: PMC6251049 DOI: 10.2174/1570159x16666180302115544] [Citation(s) in RCA: 212] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Revised: 07/18/2017] [Accepted: 03/01/2018] [Indexed: 02/06/2023] Open
Abstract
As a result of ischemia or hemorrhage, blood supply to neurons is disrupted which subsequently promotes a cascade of pathophysiological responses resulting in cell loss. Many mechanisms are involved solely or in combination in this disorder including excitotoxicity, mitochondrial death pathways, and the release of free radicals, protein misfolding, apoptosis, necrosis, autophagy and inflammation. Besides neuronal cell loss, damage to and loss of astrocytes as well as injury to white matter contributes also to cerebral injury. The core problem in stroke is the loss of neuronal cells which makes recovery difficult or even not possible in the late states. Acute treatment options that can be applied for stroke are mainly targeting re-establishment of blood flow and hence, their use is limited due to the effective time window of thrombolytic agents. However, if the acute time window is exceeded, neuronal loss starts due to the activation of cell death pathways. This review will explore the most updated cellular death mechanisms leading to neuronal loss in stroke. Ischemic and hemorrhagic stroke as well as subarachnoid hemorrhage will be debated in the light of cell death mechanisms and possible novel molecular and cellular treatment options will be discussed.
Collapse
Affiliation(s)
- Emine Şekerdağ
- Address correspondence to this author at the Neuroscience Research Lab, Research Center for Translational Medicine, Koç University, Istanbul, Turkey; Tel: +90 850 250 8250; E-mail:
| | | | | |
Collapse
|
21
|
Wu HJ, Wu C, Niu HJ, Wang K, Mo LJ, Shao AW, Dixon BJ, Zhang JM, Yang SX, Wang YR. Neuroprotective Mechanisms of Melatonin in Hemorrhagic Stroke. Cell Mol Neurobiol 2017; 37:1173-1185. [PMID: 28132129 DOI: 10.1007/s10571-017-0461-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Accepted: 01/05/2017] [Indexed: 12/30/2022]
Abstract
Hemorrhagic stroke which consists of subarachnoid hemorrhage and intracerebral hemorrhage is a dominant cause of death and disability worldwide. Although great efforts have been made, the physiological mechanisms of these diseases are not fully understood and effective pharmacological interventions are still lacking. Melatonin (N-acetyl-5-methoxytryptamine), a neurohormone produced by the pineal gland, is a broad-spectrum antioxidant and potent free radical scavenger. More importantly, there is extensive evidence demonstrating that melatonin confers neuroprotective effects in experimental models of hemorrhagic stroke. Multiple molecular mechanisms such as antioxidant, anti-apoptosis, and anti-inflammation, contribute to melatonin-mediated neuroprotection against brain injury after hemorrhagic stroke. This review article aims to summarize current knowledge regarding the beneficial effects of melatonin in experimental models of hemorrhagic stroke and explores the underlying mechanisms. We propose that melatonin is a promising neuroprotective candidate that is worthy of further evaluation for its potential therapeutic applications in hemorrhagic stroke.
Collapse
Affiliation(s)
- Hai-Jian Wu
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Cheng Wu
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Huan-Jiang Niu
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Kun Wang
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - Lian-Jie Mo
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China
| | - An-Wen Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Brandon J Dixon
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA, USA
| | - Jian-Min Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shu-Xu Yang
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China.
| | - Yi-Rong Wang
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, 3 East Qingchun Road, Hangzhou, 310016, Zhejiang, China.
| |
Collapse
|
22
|
Thelin EP, Tajsic T, Zeiler FA, Menon DK, Hutchinson PJA, Carpenter KLH, Morganti-Kossmann MC, Helmy A. Monitoring the Neuroinflammatory Response Following Acute Brain Injury. Front Neurol 2017; 8:351. [PMID: 28775710 PMCID: PMC5517395 DOI: 10.3389/fneur.2017.00351] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 07/04/2017] [Indexed: 12/11/2022] Open
Abstract
Traumatic brain injury (TBI) and subarachnoid hemorrhage (SAH) are major contributors to morbidity and mortality. Following the initial insult, patients may deteriorate due to secondary brain damage. The underlying molecular and cellular cascades incorporate components of the innate immune system. There are different approaches to assess and monitor cerebral inflammation in the neuro intensive care unit. The aim of this narrative review is to describe techniques to monitor inflammatory activity in patients with TBI and SAH in the acute setting. The analysis of pro- and anti-inflammatory cytokines in compartments of the central nervous system (CNS), including the cerebrospinal fluid and the extracellular fluid, represent the most common approaches to monitor surrogate markers of cerebral inflammatory activity. Each of these compartments has a distinct biology that reflects local processes and the cross-talk between systemic and CNS inflammation. Cytokines have been correlated to outcomes as well as ongoing, secondary injury progression. Alongside the dynamic, focal assay of humoral mediators, imaging, through positron emission tomography, can provide a global in vivo measurement of inflammatory cell activity, which reveals long-lasting processes following the initial injury. Compared to the innate immune system activated acutely after brain injury, the adaptive immune system is likely to play a greater role in the chronic phase as evidenced by T-cell-mediated autoreactivity toward brain-specific proteins. The most difficult aspect of assessing neuroinflammation is to determine whether the processes monitored are harmful or beneficial to the brain as accumulating data indicate a dual role for these inflammatory cascades following injury. In summary, the inflammatory component of the complex injury cascade following brain injury may be monitored using different modalities. Using a multimodal monitoring approach can potentially aid in the development of therapeutics targeting different aspects of the inflammatory cascade and improve the outcome following TBI and SAH.
Collapse
Affiliation(s)
- Eric Peter Thelin
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom.,Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Tamara Tajsic
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Frederick Adam Zeiler
- Division of Anaesthesia, Department of Medicine, University of Cambridge, Cambridge, United Kingdom.,Rady Faculty of Health Sciences, Department of Surgery, University of Manitoba, Winnipeg, MB, Canada.,Clinician Investigator Program, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - David K Menon
- Division of Anaesthesia, Department of Medicine, University of Cambridge, Cambridge, United Kingdom.,Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Peter J A Hutchinson
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom.,Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Keri L H Carpenter
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom.,Wolfson Brain Imaging Centre, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Maria Cristina Morganti-Kossmann
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden.,Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, VIC, Australia.,Department of Child Health, Barrow Neurological Institute at Phoenix Children's Hospital, University of Arizona College of Medicine, Phoenix, Phoenix, AZ, United States
| | - Adel Helmy
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
23
|
Chou SHY, Lan J, Esposito E, Ning M, Balaj L, Ji X, Lo EH, Hayakawa K. Extracellular Mitochondria in Cerebrospinal Fluid and Neurological Recovery After Subarachnoid Hemorrhage. Stroke 2017; 48:2231-2237. [PMID: 28663512 DOI: 10.1161/strokeaha.117.017758] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 05/22/2017] [Accepted: 05/25/2017] [Indexed: 12/22/2022]
Abstract
BACKGROUND AND PURPOSE Recent studies suggest that extracellular mitochondria may be involved in the pathophysiology of stroke. In this study, we assessed the functional relevance of endogenous extracellular mitochondria in cerebrospinal fluid (CSF) in rats and humans after subarachnoid hemorrhage (SAH). METHODS A standard rat model of SAH was used, where an intraluminal suture was used to perforate a cerebral artery, thus leading to blood extravasation into subarachnoid space. At 24 and 72 hours after SAH, neurological outcomes were measured, and the standard JC1 (5,5',6,6'-tetrachloro-1,1',3,3'-tetraethyl-benzimidazolylcarbocyanineiodide) assay was used to quantify mitochondrial membrane potentials in the CSF. To further support the rat model experiments, CSF samples were obtained from 41 patients with SAH and 27 control subjects. Mitochondrial membrane potentials were measured with the JC1 assay, and correlations with clinical outcomes were assessed at 3 months. RESULTS In the standard rat model of SAH, extracellular mitochondria was detected in CSF at 24 and 72 hours after injury. JC1 assays demonstrated that mitochondrial membrane potentials in CSF were decreased after SAH compared with sham-operated controls. In human CSF samples, extracellular mitochondria were also detected, and JC1 levels were also reduced after SAH. Furthermore, higher mitochondrial membrane potentials in the CSF were correlated with good clinical recovery at 3 months after SAH onset. CONCLUSIONS This proof-of-concept study suggests that extracellular mitochondria may provide a biomarker-like glimpse into brain integrity and recovery after injury.
Collapse
Affiliation(s)
- Sherry H-Y Chou
- From the Neuroprotection Research Laboratories, Departments of Radiology and Neurology (S.H.-Y.C., J.L., E.E., M.N., E.H.L., K.H.) and Clinical Proteomics Research Center, Department of Neurology (M.N., E.H.L.), Massachusetts General Hospital and Harvard Medical School, Boston; Departments of Critical Care Medicine, Neurology, and Neurosurgery, University of Pittsburgh, PA (S.H.-Y.C.); Department of Neurology, Brigham and Women's Hospital, Boston, MA (S.H.-Y.C.); Cerebrovascular Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China (J.L., X.J.); and Department of Neurology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston (L.B.)
| | - Jing Lan
- From the Neuroprotection Research Laboratories, Departments of Radiology and Neurology (S.H.-Y.C., J.L., E.E., M.N., E.H.L., K.H.) and Clinical Proteomics Research Center, Department of Neurology (M.N., E.H.L.), Massachusetts General Hospital and Harvard Medical School, Boston; Departments of Critical Care Medicine, Neurology, and Neurosurgery, University of Pittsburgh, PA (S.H.-Y.C.); Department of Neurology, Brigham and Women's Hospital, Boston, MA (S.H.-Y.C.); Cerebrovascular Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China (J.L., X.J.); and Department of Neurology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston (L.B.)
| | - Elga Esposito
- From the Neuroprotection Research Laboratories, Departments of Radiology and Neurology (S.H.-Y.C., J.L., E.E., M.N., E.H.L., K.H.) and Clinical Proteomics Research Center, Department of Neurology (M.N., E.H.L.), Massachusetts General Hospital and Harvard Medical School, Boston; Departments of Critical Care Medicine, Neurology, and Neurosurgery, University of Pittsburgh, PA (S.H.-Y.C.); Department of Neurology, Brigham and Women's Hospital, Boston, MA (S.H.-Y.C.); Cerebrovascular Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China (J.L., X.J.); and Department of Neurology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston (L.B.)
| | - MingMing Ning
- From the Neuroprotection Research Laboratories, Departments of Radiology and Neurology (S.H.-Y.C., J.L., E.E., M.N., E.H.L., K.H.) and Clinical Proteomics Research Center, Department of Neurology (M.N., E.H.L.), Massachusetts General Hospital and Harvard Medical School, Boston; Departments of Critical Care Medicine, Neurology, and Neurosurgery, University of Pittsburgh, PA (S.H.-Y.C.); Department of Neurology, Brigham and Women's Hospital, Boston, MA (S.H.-Y.C.); Cerebrovascular Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China (J.L., X.J.); and Department of Neurology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston (L.B.)
| | - Leonora Balaj
- From the Neuroprotection Research Laboratories, Departments of Radiology and Neurology (S.H.-Y.C., J.L., E.E., M.N., E.H.L., K.H.) and Clinical Proteomics Research Center, Department of Neurology (M.N., E.H.L.), Massachusetts General Hospital and Harvard Medical School, Boston; Departments of Critical Care Medicine, Neurology, and Neurosurgery, University of Pittsburgh, PA (S.H.-Y.C.); Department of Neurology, Brigham and Women's Hospital, Boston, MA (S.H.-Y.C.); Cerebrovascular Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China (J.L., X.J.); and Department of Neurology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston (L.B.)
| | - Xunming Ji
- From the Neuroprotection Research Laboratories, Departments of Radiology and Neurology (S.H.-Y.C., J.L., E.E., M.N., E.H.L., K.H.) and Clinical Proteomics Research Center, Department of Neurology (M.N., E.H.L.), Massachusetts General Hospital and Harvard Medical School, Boston; Departments of Critical Care Medicine, Neurology, and Neurosurgery, University of Pittsburgh, PA (S.H.-Y.C.); Department of Neurology, Brigham and Women's Hospital, Boston, MA (S.H.-Y.C.); Cerebrovascular Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China (J.L., X.J.); and Department of Neurology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston (L.B.)
| | - Eng H Lo
- From the Neuroprotection Research Laboratories, Departments of Radiology and Neurology (S.H.-Y.C., J.L., E.E., M.N., E.H.L., K.H.) and Clinical Proteomics Research Center, Department of Neurology (M.N., E.H.L.), Massachusetts General Hospital and Harvard Medical School, Boston; Departments of Critical Care Medicine, Neurology, and Neurosurgery, University of Pittsburgh, PA (S.H.-Y.C.); Department of Neurology, Brigham and Women's Hospital, Boston, MA (S.H.-Y.C.); Cerebrovascular Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China (J.L., X.J.); and Department of Neurology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston (L.B.)
| | - Kazuhide Hayakawa
- From the Neuroprotection Research Laboratories, Departments of Radiology and Neurology (S.H.-Y.C., J.L., E.E., M.N., E.H.L., K.H.) and Clinical Proteomics Research Center, Department of Neurology (M.N., E.H.L.), Massachusetts General Hospital and Harvard Medical School, Boston; Departments of Critical Care Medicine, Neurology, and Neurosurgery, University of Pittsburgh, PA (S.H.-Y.C.); Department of Neurology, Brigham and Women's Hospital, Boston, MA (S.H.-Y.C.); Cerebrovascular Research Center, Xuanwu Hospital, Capital Medical University, Beijing, China (J.L., X.J.); and Department of Neurology, Massachusetts General Hospital and Program in Neuroscience, Harvard Medical School, Boston (L.B.).
| |
Collapse
|
24
|
van Lieshout JH, Dibué-Adjei M, Cornelius JF, Slotty PJ, Schneider T, Restin T, Boogaarts HD, Steiger HJ, Petridis AK, Kamp MA. An introduction to the pathophysiology of aneurysmal subarachnoid hemorrhage. Neurosurg Rev 2017; 41:917-930. [PMID: 28215029 DOI: 10.1007/s10143-017-0827-y] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 01/24/2017] [Accepted: 01/31/2017] [Indexed: 02/06/2023]
Abstract
Pathophysiological processes following subarachnoid hemorrhage (SAH) present survivors of the initial bleeding with a high risk of morbidity and mortality during the course of the disease. As angiographic vasospasm is strongly associated with delayed cerebral ischemia (DCI) and clinical outcome, clinical trials in the last few decades focused on prevention of these angiographic spasms. Despite all efforts, no new pharmacological agents have shown to improve patient outcome. As such, it has become clear that our understanding of the pathophysiology of SAH is incomplete and we need to reevaluate our concepts on the complex pathophysiological process following SAH. Angiographic vasospasm is probably important. However, a unifying theory for the pathophysiological changes following SAH has yet not been described. Some of these changes may be causally connected or present themselves as an epiphenomenon of an associated process. A causal connection between DCI and early brain injury (EBI) would mean that future therapies should address EBI more specifically. If the mechanisms following SAH display no causal pathophysiological connection but are rather evoked by the subarachnoid blood and its degradation production, multiple treatment strategies addressing the different pathophysiological mechanisms are required. The discrepancy between experimental and clinical SAH could be one reason for unsuccessful translational results.
Collapse
Affiliation(s)
- Jasper H van Lieshout
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany.
| | - Maxine Dibué-Adjei
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Jan F Cornelius
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Philipp J Slotty
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Toni Schneider
- Institute for Neurophysiology, Medical Faculty, University of Cologne, Robert-Koch-Str. 39, 50931, Köln, Germany
| | - Tanja Restin
- Zurich Centre for Integrative Human Physiology, Institute of Physiology, University of Zurich, Winterthurerstrasse 190, 8057, Zurich, Switzerland.,Institute of Anesthesiology, Medical Faculty, University Hospital Zurich, Rämistrasse 100, 8091, Zurich, Switzerland
| | - Hieronymus D Boogaarts
- Department of Neurosurgery, Medical Faculty, Radboud University Nijmegen, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, the Netherlands
| | - Hans-Jakob Steiger
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Athanasios K Petridis
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| | - Marcel A Kamp
- Department of Neurosurgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Moorenstraße 5, 40225, Düsseldorf, Germany
| |
Collapse
|
25
|
Galluzzi L, Bravo-San Pedro JM, Blomgren K, Kroemer G. Autophagy in acute brain injury. Nat Rev Neurosci 2016; 17:467-84. [PMID: 27256553 DOI: 10.1038/nrn.2016.51] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Autophagy is an evolutionarily ancient mechanism that ensures the lysosomal degradation of old, supernumerary or ectopic cytoplasmic entities. Most eukaryotic cells, including neurons, rely on proficient autophagic responses for the maintenance of homeostasis in response to stress. Accordingly, autophagy mediates neuroprotective effects following some forms of acute brain damage, including methamphetamine intoxication, spinal cord injury and subarachnoid haemorrhage. In some other circumstances, however, the autophagic machinery precipitates a peculiar form of cell death (known as autosis) that contributes to the aetiology of other types of acute brain damage, such as neonatal asphyxia. Here, we dissect the context-specific impact of autophagy on non-infectious acute brain injury, emphasizing the possible therapeutic application of pharmacological activators and inhibitors of this catabolic process for neuroprotection.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Equipe 11 Labellisée Ligue Contre le Cancer, Centre de Recherche des Cordeliers, 75006 Paris, France.,INSERM, U1138, 75006 Paris, France.,Université Paris Descartes/Paris V, Sorbonne Paris Cité, 75006 Paris, France.,Université Pierre et Marie Curie/Paris VI, 75006 Paris, France.,Gustave Roussy Comprehensive Cancer Institute, 94805 Villejuif, France
| | - José Manuel Bravo-San Pedro
- Equipe 11 Labellisée Ligue Contre le Cancer, Centre de Recherche des Cordeliers, 75006 Paris, France.,INSERM, U1138, 75006 Paris, France.,Université Paris Descartes/Paris V, Sorbonne Paris Cité, 75006 Paris, France.,Université Pierre et Marie Curie/Paris VI, 75006 Paris, France.,Gustave Roussy Comprehensive Cancer Institute, 94805 Villejuif, France
| | - Klas Blomgren
- Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital Q2:07, 17176 Stockholm, Sweden
| | - Guido Kroemer
- Equipe 11 Labellisée Ligue Contre le Cancer, Centre de Recherche des Cordeliers, 75006 Paris, France.,INSERM, U1138, 75006 Paris, France.,Université Paris Descartes/Paris V, Sorbonne Paris Cité, 75006 Paris, France.,Université Pierre et Marie Curie/Paris VI, 75006 Paris, France.,Karolinska Institute, Department of Women's and Children's Health, Karolinska University Hospital Q2:07, 17176 Stockholm, Sweden.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, 94805 Villejuif, France.,Pôle de Biologie, Hopitâl Européen George Pompidou, AP-HP, 75015 Paris, France
| |
Collapse
|
26
|
Wu H, Niu H, Wu C, Li Y, Wang K, Zhang J, Wang Y, Yang S. The autophagy-lysosomal system in subarachnoid haemorrhage. J Cell Mol Med 2016; 20:1770-8. [PMID: 27027405 PMCID: PMC4988275 DOI: 10.1111/jcmm.12855] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2015] [Accepted: 02/25/2016] [Indexed: 12/17/2022] Open
Abstract
The autophagy–lysosomal pathway is a self‐catabolic process by which dysfunctional or unnecessary intracellular components are degraded by lysosomal enzymes. Proper function of this pathway is critical for maintaining cell homeostasis and survival. Subarachnoid haemorrhage (SAH) is one of the most devastating forms of stroke. Multiple pathogenic mechanisms, such as inflammation, apoptosis, and oxidative stress, are all responsible for brain injury and poor outcome after SAH. Most recently, accumulating evidence has demonstrated that the autophagy–lysosomal pathway plays a crucial role in the pathophysiological process after SAH. Appropriate activity of autophagy–lysosomal pathway acts as a pro‐survival mechanism in SAH, while excessive self‐digestion results in cell death after SAH. Consequently, in this review article, we will give an overview of the pathophysiological roles of autophagy–lysosomal pathway in the pathogenesis of SAH. And approaching the molecular mechanisms underlying this pathway in SAH pathology is anticipated, which may ultimately allow development of effective therapeutic strategies for SAH patients through regulating the autophagy–lysosomal machinery.
Collapse
Affiliation(s)
- Haijian Wu
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Huanjiang Niu
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Cheng Wu
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yong Li
- Department of Neurosurgery, School of Medicine, Ningbo University, Ningbo, China
| | - Kun Wang
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Yirong Wang
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shuxu Yang
- Department of Neurosurgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
27
|
Chagla A, Mathai KI. Correlation of adiposity and muscle catabolism with clinical vasospasm and mortality after subarachnoid hemorrhage. Neurol India 2015; 63:489-90. [DOI: 10.4103/0028-3886.161984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|