1
|
Islam R, Choudhary HH, Mehta H, Zhang F, Jovin TG, Hanafy KA. Development of a 3D Brain Model to Study Sex-Specific Neuroinflammation After Hemorrhagic Stroke. Transl Stroke Res 2025; 16:655-671. [PMID: 38558012 PMCID: PMC12045812 DOI: 10.1007/s12975-024-01243-y] [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: 02/06/2024] [Revised: 03/12/2024] [Accepted: 03/16/2024] [Indexed: 04/04/2024]
Abstract
Subarachnoid hemorrhage (SAH) accounts for 5% of stroke, with women having a decreased inflammatory response compared to men; however, this mechanism has yet to be identified. One hurdle in SAH research is the lack of human brain models. Studies in murine models are helpful, but human models should be used in conjunction for improved translatability. These observations lead us to develop a 3D system to study the sex-specific microglial and neuroglial function in a novel in vitro human SAH model and compare it to our validated in vivo SAH model. Our lab has developed a 3D, membrane-based in vitro cell culture system with human astrocytes, microglia, and neurons from both sexes. The 3D cultures were incubated with male and female cerebrospinal fluid from SAH patients in the Neuro-ICU. Furthermore, microglial morphology, erythrophagocytosis, microglial inflammatory cytokine production, and neuronal apoptosis were studied and compared with our murine SAH models. The human 3D system demonstrated intercellular interactions and proportions of the three cell types similar to the adult human brain. In vitro and in vivo models of SAH showed concordance in male microglia being more inflammatory than females via morphology and flow cytometry. On the contrary, both in vitro and in vivo models revealed that female microglia were more phagocytic and less prone to damaging neurons than males. One possible explanation for the increased phagocytic ability of female microglia was the increased expression of CD206 and MerTK. Our in vitro, human, 3D cell culture SAH model showed similar results to our in vivo murine SAH model with respect to microglial morphology, inflammation, and phagocytosis when comparing the sexes. A human 3D brain model of SAH may be a useful adjunct to murine models to improve translation to SAH patients.
Collapse
Affiliation(s)
- Rezwanul Islam
- Department of Biomedical Sciences, Cooper Medical School at Rowan University, Camden, NJ, USA
- Cooper Neurological Institute, Cooper University Health Care, Camden, NJ, USA
| | - Hadi Hasan Choudhary
- Department of Biomedical Sciences, Cooper Medical School at Rowan University, Camden, NJ, USA
| | - Hritik Mehta
- Department of Biomedical Sciences, Cooper Medical School at Rowan University, Camden, NJ, USA
- Cooper Neurological Institute, Cooper University Health Care, Camden, NJ, USA
| | - Feng Zhang
- Department of Biomedical Sciences, Cooper Medical School at Rowan University, Camden, NJ, USA
- Cooper Neurological Institute, Cooper University Health Care, Camden, NJ, USA
| | - Tudor G Jovin
- Department of Biomedical Sciences, Cooper Medical School at Rowan University, Camden, NJ, USA
- Cooper Neurological Institute, Cooper University Health Care, Camden, NJ, USA
| | - Khalid A Hanafy
- Department of Biomedical Sciences, Cooper Medical School at Rowan University, Camden, NJ, USA.
- Cooper Neurological Institute, Cooper University Health Care, Camden, NJ, USA.
- Center for Neuroinflammation, Cooper Medical School at Rowan University, Camden, NJ, USA.
| |
Collapse
|
2
|
Gao Q, Su Z, Pang X, Chen J, Luo R, Li X, Zhang C, Zhao Y. Overexpression of Heme Oxygenase 1 Enhances the Neuroprotective Effects of Exosomes in Subarachnoid Hemorrhage by Suppressing Oxidative Stress and Endoplasmic Reticulum Stress. Mol Neurobiol 2025; 62:6088-6101. [PMID: 39710823 DOI: 10.1007/s12035-024-04651-1] [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: 04/11/2024] [Accepted: 11/28/2024] [Indexed: 12/24/2024]
Abstract
AIMS This study aims to elucidate the therapeutic effects and underlying mechanisms of exosomes derived from Heme oxygenase 1 (HO-1)-overexpressing human umbilical cord mesenchymal stem cells (ExoHO-1) in a subarachnoid hemorrhage (SAH) mouse model. METHODS In this study, exosomes were identified using Western blotting, particle analysis, and transmission electron microscopy. The effect of ExoHO-1 and ExoCtrl on the neurological function of SAH mice was assessed using the Garcia scoring system, Beam balance, Rotarod test, and Morris water maze test. Neuronal apoptosis and survival were evaluated through TUNEL and Nissl staining. Levels of oxidative and endoplasmic reticulum stress were measured via immunofluorescence, Western blotting, DHE staining, enzyme-linked immunosorbent assay, and commercial kits. RESULTS HO-1-overexpressing human umbilical cord mesenchymal stem cells encapsulated HO-1 into their exosomes. ExoHO-1 significantly enhanced both short-term and long-term neurological function protection. By reducing the activation of the PERK/CHOP/Caspase12 pathway and decreasing oxidative stress levels, ExoHO-1 effectively inhibited neuronal apoptosis in the ipsilateral temporal cortex. CONCLUSION ExoHO-1 enhances the therapeutic efficacy of exosomes in SAH mice by countering neuronal apoptosis, primarily through the suppression of oxidative and endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Qiongqiong Gao
- Department of neurology, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
- Central Laboratory, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Zhumin Su
- Department of neurology, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Xiangxiong Pang
- Translational Medicine Research Center, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, China
| | - Jinshuo Chen
- Central Laboratory, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Ruixiang Luo
- Central Laboratory, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Xiaoyang Li
- Central Laboratory, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Chi Zhang
- Central Laboratory, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China.
| | - Yun Zhao
- Department of neurology, The Third Affiliated Hospital, Sun Yat-sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China.
- Translational Medicine Research Center, Zhujiang Hospital of Southern Medical University, Guangzhou, 510282, China.
| |
Collapse
|
3
|
Kanamaru H, Suzuki H. Therapeutic potential of stem cells in subarachnoid hemorrhage. Neural Regen Res 2025; 20:936-945. [PMID: 38989928 PMCID: PMC11438332 DOI: 10.4103/nrr.nrr-d-24-00124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Accepted: 04/27/2024] [Indexed: 07/12/2024] Open
Abstract
Aneurysm rupture can result in subarachnoid hemorrhage, a condition with potentially severe consequences, such as disability and death. In the acute stage, early brain injury manifests as intracranial pressure elevation, global cerebral ischemia, acute hydrocephalus, and direct blood-brain contact due to aneurysm rupture. This may subsequently cause delayed cerebral infarction, often with cerebral vasospasm, significantly affecting patient outcomes. Chronic complications such as brain volume loss and chronic hydrocephalus can further impact outcomes. Investigating the mechanisms of subarachnoid hemorrhage-induced brain injury is paramount for identifying effective treatments. Stem cell therapy, with its multipotent differentiation capacity and anti-inflammatory effects, has emerged as a promising approach for treating previously deemed incurable conditions. This review focuses on the potential application of stem cells in subarachnoid hemorrhage pathology and explores their role in neurogenesis and as a therapeutic intervention in preclinical and clinical subarachnoid hemorrhage studies.
Collapse
Affiliation(s)
- Hideki Kanamaru
- Department of Neurosurgery, Mie University Graduate School of Medicine, Tsu, Japan
| | | |
Collapse
|
4
|
Ge S, Jing Z, Wang L, Cui X, Zhang X, Wang X. Iron Metabolism and Ferroptosis in Early Brain Injury after Subarachnoid Haemorrhage. Mol Neurobiol 2024; 61:10736-10746. [PMID: 38777982 PMCID: PMC11584420 DOI: 10.1007/s12035-024-04218-0] [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: 07/04/2023] [Accepted: 05/02/2024] [Indexed: 05/25/2024]
Abstract
At present, it appears that the prognosis for subarachnoid haemorrhage (SAH), which has a high death and disability rate, cannot be greatly improved by medication or other treatment. Recent research suggests that different types of cell death are implicated in early brain injury (EBI) after SAH, and this has been recognised as a major factor impacting the prognosis of SAH. Ferroptosis, which is a recently identified imbalance of iron metabolism and programmed cell death triggered by phospholipid peroxidation, has been shown to be involved in EBI after SAH and is thought to have a significant impact on EBI. The decomposition of cleaved haemoglobin during SAH involves the release of enormous amounts of free iron, resulting in iron metabolism disorders. Potential therapeutic targets for the signalling pathways of iron metabolism disorders and ferroptosis after SAH are constantly being discovered. To serve as a guide for research into other possible therapeutic targets, this paper will briefly describe the mechanisms of dysregulated iron metabolism and ferroptosis in the pathogenesis of SAH and highlight how they are involved in the development and promotion of EBI in SAH.
Collapse
Affiliation(s)
- Shihao Ge
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China
| | - Ziwen Jing
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China
| | - Lele Wang
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China
| | - Xiaocong Cui
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China
| | - Xin Zhang
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China
| | - Xiaopeng Wang
- Department of Neurology, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, Hebei, China.
| |
Collapse
|
5
|
Huang Y, Bai J. Ferroptosis in the neurovascular unit after spinal cord injury. Exp Neurol 2024; 381:114943. [PMID: 39242069 DOI: 10.1016/j.expneurol.2024.114943] [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: 06/10/2024] [Revised: 08/27/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024]
Abstract
The mechanisms of secondary injury following spinal cord injury are complicated. The role of ferroptosis, which is a newly discovered form of regulated cell death in the neurovascular unit(NVU), is increasingly important. Ferroptosis inhibitors have been shown to improve neurovascular homeostasis and attenuate secondary spinal cord injury(SCI). This review focuses on the mechanisms of ferroptosis in NVU cells and NVU-targeted therapeutic strategies according to the stages of SCI, and analyzes possible future research directions.
Collapse
Affiliation(s)
- Yushan Huang
- School of Rehabilitation, Capital Medical University, Beijing, China
| | - Jinzhu Bai
- School of Rehabilitation, Capital Medical University, Beijing, China; Department of Spine and Spinal Cord Surgery, Beijing Boai Hospital, China Rehabilitation Research Center, Beijing, China; Department of Orthopedics, Capital Medical University, Beijing, China.
| |
Collapse
|
6
|
Islam R, Choudhary H, Rajan R, Vrionis F, Hanafy KA. An overview on microglial origin, distribution, and phenotype in Alzheimer's disease. J Cell Physiol 2024; 239:e30829. [PMID: 35822939 PMCID: PMC9837313 DOI: 10.1002/jcp.30829] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 06/12/2022] [Accepted: 07/04/2022] [Indexed: 01/17/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease that is responsible for about one-third of dementia cases worldwide. It is believed that AD is initiated with the deposition of Ab plaques in the brain. Genetic studies have shown that a high number of AD risk genes are expressed by microglia, the resident macrophages of brain. Common mode of action by microglia cells is neuroinflammation and phagocytosis. Moreover, it has been discovered that inflammatory marker levels are increased in AD patients. Recent studies advocate that neuroinflammation plays a major role in AD progression. Microglia have different activation profiles depending on the region of brain and stimuli. In different activation, profile microglia can generate either pro-inflammatory or anti-inflammatory responses. Microglia defend brain cells from pathogens and respond to injuries; also, microglia can lead to neuronal death along the way. In this review, we will bring the different roles played by microglia and microglia-related genes in the progression of AD.
Collapse
Affiliation(s)
- Rezwanul Islam
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL
| | - Hadi Choudhary
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL
| | - Robin Rajan
- Marcus Neuroscience Institute, Boca Raton Medical Center, Boca Raton, FL
| | - Frank Vrionis
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL
- Marcus Neuroscience Institute, Boca Raton Medical Center, Boca Raton, FL
| | - Khalid A. Hanafy
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL
- Marcus Neuroscience Institute, Boca Raton Medical Center, Boca Raton, FL
| |
Collapse
|
7
|
Lauzier DC, Athiraman U. Role of microglia after subarachnoid hemorrhage. J Cereb Blood Flow Metab 2024; 44:841-856. [PMID: 38415607 PMCID: PMC11318405 DOI: 10.1177/0271678x241237070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 01/30/2024] [Accepted: 02/18/2024] [Indexed: 02/29/2024]
Abstract
Subarachnoid hemorrhage is a devastating sequela of aneurysm rupture. Because it disproportionately affects younger patients, the population impact of hemorrhagic stroke from subarachnoid hemorrhage is substantial. Secondary brain injury is a significant contributor to morbidity after subarachnoid hemorrhage. Initial hemorrhage causes intracranial pressure elevations, disrupted cerebral perfusion pressure, global ischemia, and systemic dysfunction. These initial events are followed by two characterized timespans of secondary brain injury: the early brain injury period and the delayed cerebral ischemia period. The identification of varying microglial phenotypes across phases of secondary brain injury paired with the functions of microglia during each phase provides a basis for microglia serving a critical role in both promoting and attenuating subarachnoid hemorrhage-induced morbidity. The duality of microglial effects on outcomes following SAH is highlighted by the pleiotropic features of these cells. Here, we provide an overview of the key role of microglia in subarachnoid hemorrhage-induced secondary brain injury as both cytotoxic and restorative effectors. We first describe the ontogeny of microglial populations that respond to subarachnoid hemorrhage. We then correlate the phenotypic development of secondary brain injury after subarachnoid hemorrhage to microglial functions, synthesizing experimental data in this area.
Collapse
Affiliation(s)
- David C Lauzier
- Department of Neurological Surgery, Washington University School of Medicine, St. Louis, MO, USA
| | - Umeshkumar Athiraman
- Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
8
|
LaMonica Ostrem BE, Domínguez-Iturza N, Stogsdill JA, Faits T, Kim K, Levin JZ, Arlotta P. Fetal brain response to maternal inflammation requires microglia. Development 2024; 151:dev202252. [PMID: 38775708 PMCID: PMC11190434 DOI: 10.1242/dev.202252] [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: 08/10/2023] [Accepted: 04/09/2024] [Indexed: 06/23/2024]
Abstract
In utero infection and maternal inflammation can adversely impact fetal brain development. Maternal systemic illness, even in the absence of direct fetal brain infection, is associated with an increased risk of neuropsychiatric disorders in affected offspring. The cell types mediating the fetal brain response to maternal inflammation are largely unknown, hindering the development of novel treatment strategies. Here, we show that microglia, the resident phagocytes of the brain, highly express receptors for relevant pathogens and cytokines throughout embryonic development. Using a rodent maternal immune activation (MIA) model in which polyinosinic:polycytidylic acid is injected into pregnant mice, we demonstrate long-lasting transcriptional changes in fetal microglia that persist into postnatal life. We find that MIA induces widespread gene expression changes in neuronal and non-neuronal cells; importantly, these responses are abolished by selective genetic deletion of microglia, indicating that microglia are required for the transcriptional response of other cortical cell types to MIA. These findings demonstrate that microglia play a crucial durable role in the fetal response to maternal inflammation, and should be explored as potential therapeutic cell targets.
Collapse
Affiliation(s)
- Bridget Elaine LaMonica Ostrem
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Nuria Domínguez-Iturza
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Jeffrey A. Stogsdill
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Tyler Faits
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kwanho Kim
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Joshua Z. Levin
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Paola Arlotta
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| |
Collapse
|
9
|
Huang L, Wang X, Zheng Y, Lang D, Wang J, Yan S, Chen Y. EGCG-NPs inhibition HO-1-mediated reprogram iron metabolism against ferroptosis after subarachnoid hemorrhage. Redox Biol 2024; 70:103075. [PMID: 38364686 PMCID: PMC10878112 DOI: 10.1016/j.redox.2024.103075] [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: 12/20/2023] [Revised: 01/31/2024] [Accepted: 02/04/2024] [Indexed: 02/18/2024] Open
Abstract
Subarachnoid hemorrhage (SAH), a devastating disease with a high mortality rate and poor outcomes, tightly associated with the dysregulation of iron metabolism and ferroptosis. (-)-Epigallocatechin-3-gallate (EGCG) is one of major bioactive compounds of tea catechin because of its well-known iron-chelating and antioxidative activities. However, the findings of iron-induced cell injuries after SAH remain controversial and the underlying therapeutic mechanisms of EGCG in ferroptosis is limited. Here, the ability of EGCG to inhibit iron-induced cell death following the alleviation of neurological function deficits was investigated by using in vivo SAH models. As expected, EGCG inhibited oxyhemoglobin (OxyHb)-induced the over-expression of HO-1, which mainly distributed in astrocytes and microglial cells. Subsequently, EGCG blocked ferrous iron accumulation through HO-1-mediated iron metabolic reprogramming. Therefore, oxidative stress and mitochondrial dysfunction was rescued by EGCG, which resulted in the downregulation of ferroptosis and ferritinophagy rather than apoptosis after SAH. As a result, EGCG exerted the superior therapeutic effects in the maintenance of iron homeostasis in glial cells, such as astrocytes and microglial cells, as well as in the improvement of functional outcomes after SAH. These findings highlighted that glial cells were not only the iron-rich cells in the brain but also susceptible to ferroptosis and ferritinophagy after SAH. The detrimental role of HO-1-mediated ferroptosis in glial cells can be regarded as an effective therapeutic target of EGCG in the prevention and treatment of SAH.
Collapse
Affiliation(s)
- Liyong Huang
- Henan Key Laboratory of Neurorestoratology, The First Affiliated Hospital of Xinxiang Medical University, Henan, China; Department of Neurosurgery, The First Affiliated Hospital of Xinxiang Medical University, Henan, China
| | - Xue Wang
- College of Life Sciences, Henan Normal University, Xinxiang, Henan, China
| | - Yanning Zheng
- College of Life Sciences, Henan Normal University, Xinxiang, Henan, China
| | - Dongcen Lang
- College of Life Sciences, Henan Normal University, Xinxiang, Henan, China
| | - Jian Wang
- College of Life Sciences, Henan Normal University, Xinxiang, Henan, China
| | - Shuaiguo Yan
- College of Life Sciences, Henan Normal University, Xinxiang, Henan, China
| | - Ying Chen
- College of Life Sciences, Henan Normal University, Xinxiang, Henan, China.
| |
Collapse
|
10
|
Wang Y, Wang R, Zhu J, Chen L. Identification of mitophagy and ferroptosis-related hub genes associated with intracerebral haemorrhage through bioinformatics analysis. Ann Hum Biol 2024; 51:2334719. [PMID: 38863372 DOI: 10.1080/03014460.2024.2334719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 03/21/2024] [Indexed: 06/13/2024]
Abstract
BACKGROUND Mitophagy and ferroptosis occur in intracerebral haemorrhage (ICH) but our understanding of mitophagy and ferroptosis-related genes remains incomplete. AIM This study aims to identify shared ICH genes for both processes. METHODS ICH differentially expressed mitophagy and ferroptosis-related genes (DEMFRGs) were sourced from the GEO database and literature. Enrichment analysis elucidated functions. Hub genes were selected via STRING, MCODE, and MCC algorithms in Cytoscape. miRNAs targeting hubs were predicted using miRWalk 3.0, forming a miRNA-hub gene network. Immune microenvironment variances were assessed with MCP and TIMER. Potential small molecules for ICH were forecasted via CMap database. RESULTS 64 DEMFRGs and ten hub genes potentially involved in various processes like ferroptosis, TNF signalling pathway, MAPK signalling pathway, and NF-kappa B signalling pathway were discovered. Several miRNAs were identified as shared targets of hub genes. The ICH group showed increased infiltration of monocytic lineage and myeloid dendritic cells compared to the Healthy group. Ten potential small molecule drugs (e.g. Zebularine, TWS-119, CG-930) were predicted via CMap. CONCLUSION Several shared genes between mitophagy and ferroptosis potentially drive ICH progression via TNF, MAPK, and NF-kappa B pathways. These results offer valuable insights for further exploring the connection between mitophagy, ferroptosis, and ICH.
Collapse
Affiliation(s)
- Yan Wang
- Department of Basic Medicine, Cangzhou Medical College, Cangzhou, China
| | - Rufeng Wang
- Department of Basic Medicine, Cangzhou Medical College, Cangzhou, China
| | - Jianzhong Zhu
- Department of Basic Medicine, Cangzhou Medical College, Cangzhou, China
| | - Ling Chen
- Department of Gynaecology, People's Hospital Affiliated to Cangzhou Medical College, Cangzhou, China
| |
Collapse
|
11
|
Henrich L, Kiessling I, Steimer M, Frase S, Kaiser S, Schallner N. Circadian dependency of microglial heme oxygenase-1 expression and inflammation determine neuronal injury in hemorrhagic stroke. J Inflamm (Lond) 2023; 20:43. [PMID: 38104143 PMCID: PMC10725034 DOI: 10.1186/s12950-023-00371-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 12/13/2023] [Indexed: 12/19/2023] Open
Abstract
BACKGROUND The heme oxygenase-1 (HO-1) enzyme pathway is of crucial importance in the removal of toxic blood components and regulation of neuroinflammation following hemorrhagic stroke. Although a circadian pattern dependency in the incidence and severity of hemorrhagic stroke exists, it is unknown whether the activity of the HO-1 system in the context of hemorrhagic injury also exhibits circadian dependency. We hypothesized that the circadian regulation of microglial HO-1 would determine the extent of neuroinflammation and neuronal injury in a murine model of subarachnoid hemorrhage (SAH). METHODS In vitro expression patterns of HO-1 and circadian rhythm genes were analyzed in the microglial BV-2 cell line and primary microglia (PMG) using Western blot and qPCR. PMG isolated from Hmox1fl/fl and LyzM-Cre-Hmox1fl/fl mice were used to evaluate the role of microglial HO-1. We further investigated the in vivo relevance in a murine subarachnoid hemorrhage (SAH) model using Hmox1fl/fl and LyzM-Cre-Hmox1fl/fl mice with myeloid cell HO-1 deficiency, inducing SAH at different zeitgeber (ZT) times and analyzing the expression of HO-1 and the circadian control gene Period-2 (Per-2), respectively. Furthermore, we measured the inflammatory cytokine Monocyte Chemoattractant Protein-1 (MCP-1) in the cerebrospinal fluid of SAH patients in correlation with clinical outcome. RESULTS HO-1 baseline expression and response to CO with blood exposure depended on ZT. In vitro expression of circadian control genes was de-synchronized in LyzM-Cre-Hmox1fl/fl PMG and did not respond to exogenous CO exposure. We found that circadian rhythm plays a crucial role in brain damage after SAH. At ZT2, we observed less phagocytic function, more vasospasm and increased microglial activation. CO reduced mortality at ZT12 in HO-1 deficient mice and reduced the difference between ZT2 and ZT12 in the inflammatory response. Induction of MCP-1 in the CSF from SAH patients was time-dependent and correlated with the expression of circadian control genes, SAH severity, functional impairment and delirium. CONCLUSIONS Our data point towards a crucial role for the HO-1 enzyme system and circadian control in neuronal injury after a hemorrhagic stroke.
Collapse
Affiliation(s)
- Luise Henrich
- Department of Anesthesiology & Critical Care, Medical Center, University of Freiburg, Hugstetter Str. 55, Freiburg, 79106, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Iva Kiessling
- Department of Anesthesiology & Critical Care, Medical Center, University of Freiburg, Hugstetter Str. 55, Freiburg, 79106, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Matti Steimer
- Department of Anesthesiology & Critical Care, Medical Center, University of Freiburg, Hugstetter Str. 55, Freiburg, 79106, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sibylle Frase
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Neurology and Neuroscience, Medical Center, University of Freiburg, Freiburg, Germany
| | - Sandra Kaiser
- Department of Anesthesiology & Critical Care, Medical Center, University of Freiburg, Hugstetter Str. 55, Freiburg, 79106, Germany
- Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nils Schallner
- Department of Anesthesiology & Critical Care, Medical Center, University of Freiburg, Hugstetter Str. 55, Freiburg, 79106, Germany.
- Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
12
|
Becker K. Animal Welfare Aspects in Planning and Conducting Experiments on Rodent Models of Subarachnoid Hemorrhage. Cell Mol Neurobiol 2023; 43:3965-3981. [PMID: 37861870 PMCID: PMC11407738 DOI: 10.1007/s10571-023-01418-5] [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: 07/31/2023] [Accepted: 09/18/2023] [Indexed: 10/21/2023]
Abstract
Subarachnoid hemorrhage is an acute life-threatening cerebrovascular disease with high socio-economic impact. The most frequent cause, the rupture of an intracerebral aneurysm, is accompanied by abrupt changes in intracerebral pressure, cerebral perfusion pressure and, consequently, cerebral blood flow. As aneurysms rupture spontaneously, monitoring of these parameters in patients is only possible with a time delay, upon hospitalization. To study alterations in cerebral perfusion immediately upon ictus, animal models are mandatory. This article addresses the points necessarily to be included in an animal project proposal according to EU directive 2010/63/EU for the protection of animals used for scientific purposes and herewith offers an insight into animal welfare aspects of using rodent models for the investigation of cerebral perfusion after subarachnoid hemorrhage. It compares surgeries, model characteristics, advantages, and drawbacks of the most-frequently used rodent models-the endovascular perforation model and the prechiasmatic and single or double cisterna magna injection model. The topics of discussing anesthesia, advice on peri- and postanesthetic handling of animals, assessing the severity of suffering the animals undergo during the procedure according to EU directive 2010/63/EU and weighing the use of these in vivo models for experimental research ethically are also presented. In conclusion, rodent models of subarachnoid hemorrhage display pathophysiological characteristics, including changes of cerebral perfusion similar to the clinical situation, rendering the models suited to study the sequelae of the bleeding. A current problem is low standardization of the models, wherefore reporting according to the ARRIVE guidelines is highly recommended. Animal welfare aspects of rodent models of subarachnoid hemorrhage. Rodent models for investigation of cerebral perfusion after subarachnoid hemorrhage are compared regarding surgeries and model characteristics, and 3R measures are suggested. Anesthesia is discussed, and advice given on peri- and postanesthetic handling. Severity of suffering according to 2010/63/EU is assessed and use of these in vivo models weighed ethically.
Collapse
Affiliation(s)
- Katrin Becker
- Institute for Translational Neurosurgery, Medical Faculty, RWTH Aachen University, 52074, Aachen, Germany.
- Institute for Cardiovascular Sciences, University Hospital Bonn, 53127, Bonn, Germany.
| |
Collapse
|
13
|
Qu Y, Zhang N, Zhao Y. Resveratrol Inhibits Abdominal Aortic Aneurysm Progression by Reducing Extracellular Matrix Degradation, Apoptosis, Autophagy, and Inflammation of Vascular Smooth Muscle Cells via Upregulation of HMOX1. J Endovasc Ther 2023:15266028231202727. [PMID: 37789605 DOI: 10.1177/15266028231202727] [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: 10/05/2023]
Abstract
OBJECTIVE This study aimed to explore the therapeutic effect of resveratrol (RES) against abdominal aortic aneurysm (AAA) and the role of HMOX1 underlying this effect. METHODS Vascular smooth muscle cells (VSMCs) were induced by angiotensin II (Ang II) to construct the microenvironment of AAA. HMOX1 expression was downregulated by the short hairpin ribonucleic acid (RNA) specific to HMOX1 in RES-pretreated VSMCs. The levels of matrix metalloproteinase (MMP)-2, MMP-9, and elastin were measured by quantitative reverse transcription polymerase chain reaction (qRT-PCR) and western blot. Apoptosis rate was detected. The levels of apoptosis-related proteins (caspase-3 and Bax/Bcl-2), inflammatory cytokines (interleukin [IL]-6, tumor necrosis factor [TNF]-α, and IL-1β), and autophagy-related proteins (Beclin 1, light chain 3 [LC3] II/I, and p62) were detected by western blot. The secretion of inflammatory factors in cell supernatant was detected by enzyme-linked immunosorbent assay (ELISA). The number of autophagic vesicles in VSMCs was observed and analyzed by transmission electron microscopy. A rat model of pancreatic elastase-induced AAA was established to verify the effect and action mechanism of RES. RESULTS Stimulation of Ang II increased the messenger RNA (mRNA) and protein levels of MMP-2 and MMP-9, decreased elastin expression, and enhanced apoptosis, secretion of inflammatory factors, and autophagy in VSMCs, whereas RES pretreatment ameliorated Ang II-induced VSMC dysfunction. In addition, HMOX1 mRNA and heme oxygenase-1 (HO-1) protein levels were significantly increased in VSMCs pretreated with RES compared with Ang II treatment alone. Silencing of HMOX1 abolished the effects of RES on VSMC dysfunction. Consistently, RES suppressed the development of AAA in rats by increasing the expression of HMOX1. CONCLUSION Resveratrol protects against AAA by inhibiting extracellular matrix degradation, apoptosis, autophagy, and inflammation of VSMCs via HMOX1 upregulation. CLINICAL IMPACT Our study found that angiotensin II (Ang II) stimulated increased the levels of MMP-2 and MMP-9 in vascular smooth muscle cells (VSMCs), decreased elastin expression, and promoted apoptosis, autophagy occurrence, and secretion of inflammatory factors, while resveratrol (RES) pretreatment improved this effect. In addition, downregulation of HMOX1 expression eliminated the effect of RES on the function of VSMCs. Our study elucidates that RES improves AAA progression through HMOX1 at both cellular and animal levels. This work can help doctors better understand the pathological mechanism of the occurrence and development of AAA, and provide a theoretical basis for clinicians to find better treatment options.
Collapse
Affiliation(s)
- Yunfei Qu
- Department of Vascular Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| | - Ning Zhang
- General Practice, Chongqing University Three Gorges Hospital, Chongqing, P.R. China
| | - Yu Zhao
- Department of Vascular Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, P.R. China
| |
Collapse
|
14
|
Zhang A, Liu Y, Wang X, Xu H, Fang C, Yuan L, Wang K, Zheng J, Qi Y, Chen S, Zhang J, Shao A. Clinical Potential of Immunotherapies in Subarachnoid Hemorrhage Treatment: Mechanistic Dissection of Innate and Adaptive Immune Responses. Aging Dis 2023; 14:1533-1554. [PMID: 37196120 PMCID: PMC10529760 DOI: 10.14336/ad.2023.0126] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 01/26/2023] [Indexed: 05/19/2023] Open
Abstract
Subarachnoid hemorrhage (SAH), classified as a medical emergency, is a devastating and severe subtype of stroke. SAH induces an immune response, which further triggers brain injury; however, the underlying mechanisms need to be further elucidated. The current research is predominantly focused on the production of specific subtypes of immune cells, especially innate immune cells, post-SAH onset. Increasing evidence suggests the critical role of immune responses in SAH pathophysiology; however, studies on the role and clinical significance of adaptive immunity post-SAH are limited. In this present study, we briefly review the mechanistic dissection of innate and adaptive immune responses post-SAH. Additionally, we summarized the experimental studies and clinical trials of immunotherapies for SAH treatment, which may form the basis for the development of improved therapeutic approaches for the clinical management of SAH in the future.
Collapse
Affiliation(s)
- Anke Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
| | - Yibo Liu
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
| | - Xiaoyu Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
| | - Houshi Xu
- Department of Neurosurgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Chaoyou Fang
- Department of Neurosurgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Ling Yuan
- Department of Neurosurgery, Shanghai General Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - KaiKai Wang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
| | - Jingwei Zheng
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
| | - Yangjian Qi
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
| | - Sheng Chen
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, School of Medicine, Zhejiang University, China.
- Key Laboratory of Precise Treatment and Clinical Translational Research of Neurological Diseases, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
| |
Collapse
|
15
|
Ramagiri S, Pan S, DeFreitas D, Yang PH, Raval DK, Wozniak DF, Esakky P, Strahle JM. Deferoxamine Prevents Neonatal Posthemorrhagic Hydrocephalus Through Choroid Plexus-Mediated Iron Clearance. Transl Stroke Res 2023; 14:704-722. [PMID: 36308676 PMCID: PMC10147846 DOI: 10.1007/s12975-022-01092-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Revised: 09/20/2022] [Accepted: 09/21/2022] [Indexed: 12/14/2022]
Abstract
Posthemorrhagic hydrocephalus occurs in up to 30% of infants with high-grade intraventricular hemorrhage and is associated with the worst neurocognitive outcomes in preterm infants. The mechanisms of posthemorrhagic hydrocephalus after intraventricular hemorrhage are unknown; however, CSF levels of iron metabolic pathway proteins including hemoglobin have been implicated in its pathogenesis. Here, we develop an animal model of intraventricular hemorrhage using intraventricular injection of hemoglobin at post-natal day 4 that results in acute and chronic hydrocephalus, pathologic choroid plexus iron accumulation, and subsequent choroid plexus injury at post-natal days 5, 7, and 15. This model also results in increased expression of aquaporin-1, Na+/K+/Cl- cotransporter 1, and Na+/K+/ATPase on the apical surface of the choroid plexus 24 h post-intraventricular hemorrhage. We use this model to evaluate a clinically relevant treatment strategy for the prevention of neurological sequelae after intraventricular hemorrhage using intraventricular administration of the iron chelator deferoxamine at the time of hemorrhage. Deferoxamine treatment prevented posthemorrhagic hydrocephalus for up to 11 days after intraventricular hemorrhage and prevented the development of sensorimotor gating deficits. In addition, deferoxamine treatment facilitated acute iron clearance through the choroid plexus and subsequently reduced choroid plexus iron levels at 24 h with reversal of hemoglobin-induced aquaporin-1 upregulation on the apical surface of the choroid plexus. Intraventricular administration of deferoxamine at the time of intraventricular hemorrhage may be a clinically relevant treatment strategy for preventing posthemorrhagic hydrocephalus and likely acts through promoting iron clearance through the choroid plexus to prevent hemoglobin-induced injury.
Collapse
Affiliation(s)
- Sruthi Ramagiri
- Department of Neurosurgery, Washington University School of Medicine, MO, 63110, St. Louis, USA
| | - Shelei Pan
- Department of Neurosurgery, Washington University School of Medicine, MO, 63110, St. Louis, USA
| | - Dakota DeFreitas
- Department of Neurosurgery, Washington University School of Medicine, MO, 63110, St. Louis, USA
| | - Peter H Yang
- Department of Neurosurgery, Washington University School of Medicine, MO, 63110, St. Louis, USA
| | - Dhvanii K Raval
- Department of Neurosurgery, Washington University School of Medicine, MO, 63110, St. Louis, USA
| | - David F Wozniak
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110-1093, USA
- Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, St. Louis, MO, 63110-1093, USA
- Taylor Family Institute for Innovative Psychiatric Research, Washington University School of Medicine, St. Louis, MO, 63110-1093, USA
| | - Prabagaran Esakky
- Department of Neurosurgery, Washington University School of Medicine, MO, 63110, St. Louis, USA
| | - Jennifer M Strahle
- Department of Neurosurgery, Washington University School of Medicine, MO, 63110, St. Louis, USA.
- Department of Orthopedic Surgery, Washington University School of Medicine, St. Louis, MO, 63110, USA.
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, 63110, USA.
| |
Collapse
|
16
|
Liu S, Gao X, Zhou S. New Target for Prevention and Treatment of Neuroinflammation: Microglia Iron Accumulation and Ferroptosis. ASN Neuro 2022; 14:17590914221133236. [PMID: 36285433 PMCID: PMC9607999 DOI: 10.1177/17590914221133236] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Microglia play an important role in maintaining central nervous system homeostasis and are the major immune cells in the brain. In response to internal or external inflammatory stimuli, microglia are activated and release numerous inflammatory factors, thus leading to neuroinflammation. Inflammation and microglia iron accumulation promote each other and jointly promote the progression of neuroinflammation. Inhibiting microglia iron accumulation prevents neuroinflammation. Ferroptosis is an iron-dependent phospholipid peroxidation-driven type of cell death regulation. Cell iron accumulation causes the peroxidation of cell membrane phospholipids and damages the cell membrane. Ultimately, this process leads to cell ferroptosis. Iron accumulation or phospholipid peroxidation in microglia releases a large number of inflammatory factors. Thus, inhibiting microglia ferroptosis may be a new target for the prevention and treatment of neuroinflammation.
Collapse
Affiliation(s)
- Shunfeng Liu
- College of Pharmacy, Guilin Medical College, Guilin, China
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical College, Guilin, China
| | - Xue Gao
- College of Pharmacy, Guilin Medical College, Guilin, China
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical College, Guilin, China
| | - Shouhong Zhou
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical College, Guilin, China
- Basic Medical College, Guilin Medical College, Guilin, China
- Shouhong Zhou, Guilin Medical College, No.1, Zhiyuan Road, Lingui District, Guilin City, Guangxi Province, China.
| |
Collapse
|
17
|
Islam R, Vrionis F, Hanafy KA. Microglial TLR4 is Critical for Neuronal Injury and Cognitive Dysfunction in Subarachnoid Hemorrhage. Neurocrit Care 2022; 37:761-769. [PMID: 35778649 PMCID: PMC9672010 DOI: 10.1007/s12028-022-01552-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/06/2022] [Indexed: 11/30/2022]
Abstract
Background Toll-like receptor 4 (TLR4) activation causes excessive production of proinflammatory mediators and an increased expression of costimulatory molecules that leads to neuroinflammation after subarachnoid hemorrhage (SAH). Although TLR4-mediated inflammatory pathways have long been studied in neuroinflammation, the specific glia implicated in initiation and propagation of neuroinflammation in SAH have not been well elucidated. In this study, we investigated the involvement of glial TLR4 including microglia and astrocytes in brain damage and poor neurological outcome. Methods In this study, global TLR4 knockout, cell-specific TLR4 knockout, and floxxed control male and female mice were used. The mice were injected with 60 μl autologous blood near the mesencephalon to induce SAH; animals were euthanized on postoperative day 7 for immunohistochemistry of glia and apoptotic cells. Microglial morphology was evaluated by using immunofluorescence density quantification to determine correlations between morphology and neuroinflammation. Microglial depletion was accomplished with the intracerebroventricular administration of clodronate liposomes. Cognitive function was assessed with Barnes maze. Results On postoperative day 7 after SAH induction, neuronal apoptosis was markedly reduced in the clodronate liposome group compared with phosphate-buffered saline control liposomes, and cognitive performance in the clodronate group was improved, as well. Differences in microglial activation, assessed by morphometric analysis, and neuronal apoptosis were significantly greater in wildtype knockouts compared with cell-specific and global TLR4 knockouts. The mice lacking TLR4 on astrocytes and neurons showed no differences compared with wildtype mice on any end points. Conclusions Our data suggest that microglial depletion with the intracerebroventricular administration of clodronate can improve the cognitive function in an SAH mouse model, and TLR4 is critical for microglial activation and neuronal injury. Only microglial TLR4 is necessary for brain damage and poor cognitive outcome rather than astrocyte or neuronal TLR4. Thus, microglial TLR4 could be a potent therapeutic target to treat SAH-associated neuronal injury and protect against cognitive dysfunction.
Collapse
Affiliation(s)
- Rezwanul Islam
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
| | - Frank Vrionis
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA
- Marcus Neuroscience Institute, Boca Raton Medical Center, Boca Raton, FL, USA
| | - Khalid A Hanafy
- Department of Biomedical Sciences, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL, USA.
- Marcus Neuroscience Institute, Boca Raton Medical Center, Boca Raton, FL, USA.
| |
Collapse
|
18
|
Trichosanthis Semen Suppresses Lipopolysaccharide-Induced Neuroinflammation by Regulating the NF-κB Signaling Pathway and HO-1 Expression in Microglia. Toxins (Basel) 2021; 13:toxins13120898. [PMID: 34941735 PMCID: PMC8704237 DOI: 10.3390/toxins13120898] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 12/10/2021] [Indexed: 01/08/2023] Open
Abstract
Neuroinflammation, which is mediated by microglia that release various inflammatory cytokines, is a typical feature of neurodegenerative diseases (NDDs), such as Alzheimer’s disease and Parkinson’s disease. Hence, alleviating neuroinflammation by downregulating pro-inflammatory action, and upregulating anti-inflammatory action of microglia is an efficient therapeutic target for NDDs. In this study, we evaluated whether trichosanthis semen (TS), a dried ripe seed of Trichosanthes kirilowii Maximowicz, reduces lipopolysaccharide (LPS)-induced neuroinflammation by regulating microglial responses in vitro and in vivo. Our results presented that TS reduced the release of pro-inflammatory mediators, such as nitric oxide (NO), inducible NO synthase, tumor necrosis factor-α, interleukin-1β, and interleukin-6 via inhibition of the nuclear factor kappa B (NF-κB) signaling pathway in LPS-treated BV2 microglial cells. Moreover, TS induced anti-inflammatory mediators, such as interleukin-10, found in inflammatory zone 1, and chitinase 3-like 3 by the upregulation of heme oxygenase 1 (HO-1). We further confirmed that TS administration suppressed microglial activation, but enhanced HO-1 expression in LPS-injected mice. These results suggest that TS has anti-neuroinflammatory effects via inhibition of NF-κB signaling through the activation of HO-1, and that TS may be a therapeutical candidate for NDDs treatment.
Collapse
|
19
|
Luo Y, Ullah R, Wang J, Du Y, Huang S, Meng L, Gao Y, Gong M, Galaj E, Yin X, Shi H. Exogenous Carbon Monoxide Produces Rapid Antidepressant- and Anxiolytic-Like Effects. Front Pharmacol 2021; 12:757417. [PMID: 34867375 PMCID: PMC8637155 DOI: 10.3389/fphar.2021.757417] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/26/2021] [Indexed: 12/25/2022] Open
Abstract
Carbon monoxide (CO), a byproduct of heme catalyzed by heme oxygenase (HO), has been reported to exert antioxidant and anti-inflammatory actions, and to produce significant neuroprotective effects. The potential effects of CO and even HO on depressive-like behaviors are still poorly understood. Utilizing several approaches including adeno-associated virus (AAV)-mediated overexpression of HO-1, systemic CO-releasing molecules (CO-RMs), CO-rich saline or CO gas treatment procedures in combination with hydrogen peroxide (H2O2)-induced PC12 cell injury model, and lipopolysaccharide (LPS)-induced depression mouse model, the present study aimed to investigate the potential antidepressant- and anxiolytic-like effects of endogenous and exogenous CO administration in vivo and in vitro. The results of in vitro experiments showed that both CO-RM-3 and CO-RM-A1 pretreatment blocked H2O2-induced cellular injuries by increasing cell survival and decreasing cell apoptosis and necrosis. Similar to the effects of CO-RM-3 and CO-RM-A1 pretreatment, AAV-mediated HO-1 overexpression in the dorsal hippocampus produced significant antidepressant-like activities in mice under normal conditions. Further investigation showed that the CO gas treatment significantly blocked LPS-induced depressive- and anxiety-like behaviors in mice. Taken together, our results suggest that the activation of HO-1 and/or exogenous CO administration produces protective effects and exerts antidepressant- and anxiolytic-like effects. These data uncover a novel function of the HO-1/CO system that appears to be a promising therapeutic target for the treatment of depression and anxiety.
Collapse
Affiliation(s)
- Yixiao Luo
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China.,Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, China
| | - Rafi Ullah
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, China
| | - Jinfeng Wang
- Department of Obstetrics and Gynecology, The No.1 Hospital of Yongnian District Handan City, Handan, China
| | - Yuru Du
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, China
| | - Shihao Huang
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Li Meng
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, China
| | - Yuan Gao
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, China.,Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang, China
| | - Miao Gong
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, China
| | - Ewa Galaj
- Neuroscience Program, Department of Psychological and Brain Sciences, Colgate University, Hamilton, NY, United States
| | - Xi Yin
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, China.,Department of Functional Region of Diagnosis, Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| | - Haishui Shi
- Neuroscience Research Center, Institute of Medical and Health Science of HeBMU, Hebei Medical University, Shijiazhuang, China.,Hebei Key Laboratory of Neurophysiology, Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
20
|
Liu H, Schwarting J, Terpolilli NA, Nehrkorn K, Plesnila N. Scavenging Free Iron Reduces Arteriolar Microvasospasms After Experimental Subarachnoid Hemorrhage. Stroke 2021; 52:4033-4042. [PMID: 34749506 DOI: 10.1161/strokeaha.120.033472] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
BACKGROUND AND PURPOSE Subarachnoid hemorrhage (SAH) is associated with acute and delayed cerebral ischemia resulting in high acute mortality and severe chronic neurological deficits. Spasms of the pial and intraparenchymal microcirculation (microvasospasms) contribute to acute cerebral ischemia after SAH; however, the underlying mechanisms remain unknown. We hypothesize that free iron (Fe3+) released from hemolytic red blood cells into the subarachnoid space may be involved in microvasospasms formation. METHODS Male C57BL/6 mice (n=8/group) received 200 mg/kg of the iron scavenger deferoxamine or vehicle intravenously and were then subjected to SAH by filament perforation. Microvasospasms of pial and intraparenchymal vessels were imaged three hours after SAH by in vivo 2-photon microscopy. RESULTS Microvasospasms occurred in all investigated vessel categories down to the capillary level. Deferoxamine significantly reduced the number of microvasospasms after experimental SAH. The effect was almost exclusively observed in larger pial arterioles (>30 µm) covered with blood. CONCLUSIONS These results provide proof-of-principle evidence that Fe3+ is involved in the formation of arteriolar microvasospasms after SAH and that arteriolar and capillary microvasospasms are triggered by different mechanisms. Deciphering the mechanisms of Fe3+-induced microvasospasms may result in novel therapeutic strategies for SAH patients.
Collapse
Affiliation(s)
- Hanhan Liu
- Institute for Stroke and Dementia Research (H.L., J.S., N.A.T., K.N., N.P.), University of Munich Medical Center, Ludwig-Maximilians-University (LMU), Germany
| | - Julian Schwarting
- Institute for Stroke and Dementia Research (H.L., J.S., N.A.T., K.N., N.P.), University of Munich Medical Center, Ludwig-Maximilians-University (LMU), Germany
- Department of Neurosurgery (J.S., N.A.T.), University of Munich Medical Center, Ludwig-Maximilians-University (LMU), Germany
| | - Nicole Angela Terpolilli
- Institute for Stroke and Dementia Research (H.L., J.S., N.A.T., K.N., N.P.), University of Munich Medical Center, Ludwig-Maximilians-University (LMU), Germany
- Munich Cluster of Systems Neurology (Synergy), Germany (N.A.T., K.N., N.P.)
| | - Kathrin Nehrkorn
- Institute for Stroke and Dementia Research (H.L., J.S., N.A.T., K.N., N.P.), University of Munich Medical Center, Ludwig-Maximilians-University (LMU), Germany
- Munich Cluster of Systems Neurology (Synergy), Germany (N.A.T., K.N., N.P.)
| | - Nikolaus Plesnila
- Institute for Stroke and Dementia Research (H.L., J.S., N.A.T., K.N., N.P.), University of Munich Medical Center, Ludwig-Maximilians-University (LMU), Germany
- Munich Cluster of Systems Neurology (Synergy), Germany (N.A.T., K.N., N.P.)
| |
Collapse
|
21
|
Peterson C, Umoye AO, Puglisi CH, Waldau B. Mechanisms of memory impairment in animal models of nontraumatic intracranial hemorrhage: A systematic review of the literature. BRAIN HEMORRHAGES 2021; 3:77-93. [DOI: 10.1016/j.hest.2021.08.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
|
22
|
Wan H, Brathwaite S, Ai J, Hynynen K, Macdonald RL. Role of perivascular and meningeal macrophages in outcome following experimental subarachnoid hemorrhage. J Cereb Blood Flow Metab 2021; 41:1842-1857. [PMID: 33444089 PMCID: PMC8327101 DOI: 10.1177/0271678x20980296] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The distribution and clearance of erythrocytes after subarachnoid hemorrhage (SAH) is poorly understood. We aimed to characterize the distribution of erythrocytes after SAH and the cells involved in their clearance. To visualize erythrocyte distribution, we injected fluorescently-labelled erythrocytes into the prechiasmatic cistern of mice. 10 minutes after injection, we found labelled erythrocytes in the subarachnoid space and ventricular system, and also in the perivascular spaces surrounding large penetrating arterioles. 2 and 5 days after SAH, fluorescence was confined within leptomeningeal and perivascular cells. We identified the perivascular cells as perivascular macrophages based on their morphology, location, Iba-1 immunoreactivity and preferential uptake of FITC-dextran. We subsequently depleted meningeal and perivascular macrophages 2 days before or 3 hours after SAH with clodronate liposomes. At day 5 after SAH, we found increased blood deposition in mice treated prior to SAH, but not those treated after. Treatment post-SAH improved neurological scoring, reduced neuronal cell death and perivascular inflammation, whereas pre-treatment only reduced perivascular inflammation. Our data indicate that after SAH, erythrocytes are distributed throughout the subarachnoid space extending into the perivascular spaces of parenchymal arterioles. Furthermore, meningeal and perivascular macrophages are involved in erythrocyte uptake and play an important role in outcome after SAH.
Collapse
Affiliation(s)
- Hoyee Wan
- Division of Neurosurgery, St. Michael's Hospital, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Canada.,Sunnybrook Health Sciences Research Institute, Sunnybrook Hospital, Toronto, Canada
| | - Shakira Brathwaite
- Division of Neurosurgery, St. Michael's Hospital, Toronto, Canada.,Sunnybrook Health Sciences Research Institute, Sunnybrook Hospital, Toronto, Canada
| | - Jinglu Ai
- Division of Neurosurgery, St. Michael's Hospital, Toronto, Canada.,Barrow Neurological Institute, Phoenix, AZ, USA
| | - Kullervo Hynynen
- Department of Medical Biophysics, University of Toronto, Toronto, Canada.,Sunnybrook Health Sciences Research Institute, Sunnybrook Hospital, Toronto, Canada
| | - R Loch Macdonald
- Division of Neurosurgery, St. Michael's Hospital, Toronto, Canada.,Department of Neurological Surgery, University of California San Francisco, Fresno, CA, USA
| |
Collapse
|
23
|
Wang J, Chen R, Liu C, Wu X, Zhang Y. Antidepressant mechanism of catalpol: Involvement of the PI3K/Akt/Nrf2/HO-1 signaling pathway in rat hippocampus. Eur J Pharmacol 2021; 909:174396. [PMID: 34332921 DOI: 10.1016/j.ejphar.2021.174396] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 07/07/2021] [Accepted: 07/28/2021] [Indexed: 02/06/2023]
Abstract
Catalpol is a major compound in Rehmanniae Radix with outstanding medicinal and nutritional values. Our previous studies have demonstrated catalpol's antidepressant effect, but its mechanisms remain unclear. This study aimed to explore the antidepressant mechanisms of catalpol via the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/nuclear factor E2-related factor 2(Nrf2)/heme oxygenase-1 (HO-1) pathway. Results demonstrated that chronic unpredictable mild stress (CUMS) for 5 consecutive weeks caused significant decreases in the sucrose preference and the horizontal and vertical scores of open-field test, as well as a significant increase in the swimming-immobility time of rats; catalpol administration significantly reversed the abnormality of these indicators. Further real-time fluorescent quantitative polymerase chain reaction and Western blotting results together showed that CUMS significantly downregulated the expression levels of hippocampal genes and proteins, including PI3K, Akt, Nrf2, HO-1, tropomyosin-related kinase B (TrkB), and brain-derived neurotrophic factor; catalpol administration significantly reversed the abnormal expression of these genes and proteins. CUMS also caused a significant decrease in the hippocampal superoxide dismutase, catalase, glutathione peroxidase, glutathione-s transferase, and reduced glutathione levels, as well as a significant increase in thiobarbituric acid reactive substances level in rats; catalpol administration significantly reversed the abnormality of these indicators. Taken together, this study confirmed for the first time that the antidepressant effect of catalpol on CUMS-induced depression involved the upregulation of the PI3K/Akt/Nrf2/HO-1 signaling pathway, thereby improving the hippocampal neurotrophic, neuroprotective, and antioxidant levels. The PI3K/Akt/Nrf2/HO-1 pathway-related molecules may serve as potential new biomarkers and candidate molecular targets for catalpol's antidepressant effects.
Collapse
Affiliation(s)
- Junming Wang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China; Co-construction Collaborative Innovation Center for Chinese Medicine and Respiratory Diseases by Henan & Education Ministry of PR China, Henan University of Chinese Medicine, Zhengzhou, 450046, China.
| | - Rongxing Chen
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Chen Liu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Xiaohui Wu
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Yueyue Zhang
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| |
Collapse
|
24
|
Involvement of Microglia in the Pathophysiology of Intracranial Aneurysms and Vascular Malformations-A Short Overview. Int J Mol Sci 2021; 22:ijms22116141. [PMID: 34200256 PMCID: PMC8201350 DOI: 10.3390/ijms22116141] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/01/2021] [Accepted: 06/04/2021] [Indexed: 12/19/2022] Open
Abstract
Aneurysms and vascular malformations of the brain represent an important source of intracranial hemorrhage and subsequent mortality and morbidity. We are only beginning to discern the involvement of microglia, the resident immune cell of the central nervous system, in these pathologies and their outcomes. Recent evidence suggests that activated proinflammatory microglia are implicated in the expansion of brain injury following subarachnoid hemorrhage (SAH) in both the acute and chronic phases, being also a main actor in vasospasm, considerably the most severe complication of SAH. On the other hand, anti-inflammatory microglia may be involved in the resolution of cerebral injury and hemorrhage. These immune cells have also been observed in high numbers in brain arteriovenous malformations (bAVM) and cerebral cavernomas (CCM), although their roles in these lesions are currently incompletely ascertained. The following review aims to shed a light on the most significant findings related to microglia and their roles in intracranial aneurysms and vascular malformations, as well as possibly establish the course for future research.
Collapse
|
25
|
Zhang Y, Lu X, Tai B, Li W, Li T. Ferroptosis and Its Multifaceted Roles in Cerebral Stroke. Front Cell Neurosci 2021; 15:615372. [PMID: 34149358 PMCID: PMC8209298 DOI: 10.3389/fncel.2021.615372] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Accepted: 04/09/2021] [Indexed: 12/11/2022] Open
Abstract
Ferroptosis is a unique regulated cell death defined by the intracellular iron overload and distinct biological features compared with other well-known programmed cell death. Ferroptosis can be triggered by many causes including decreased expression of glutathione (GSH), inhibition of the function of glutathione-dependent peroxidase 4 (GPX4), and system xc–, all of which finally lead to the over-accumulation of lipid peroxides in the cell. Ferroptosis has been reported to play an important role in the pathophysiological process of various cancers. In recent years, much evidence also proved that ferroptosis is involved in the progress of cerebral stroke. In this review, we summarized the characteristics of ferroptosis and the potential relationship between ferroptosis and ischemic and hemorrhagic stroke, to provide new targets and ideas for the therapy of stroke.
Collapse
Affiliation(s)
- Yongfa Zhang
- Department of Neurosurgery, The First People's Hospital of Yunnan Province, Kunhua Hospital, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Xiaoyang Lu
- Department of Neurosurgery, School of Medicine, The Second Affiliated Hospital, Zhejiang University, Hangzhou, China.,Translational Neurosurgery and Neurobiology, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Bai Tai
- Department of Neurosurgery, The First People's Hospital of Yunnan Province, Kunhua Hospital, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Weijia Li
- Department of Neurosurgery, The First People's Hospital of Yunnan Province, Kunhua Hospital, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Tao Li
- Department of Neurosurgery, The First People's Hospital of Yunnan Province, Kunhua Hospital, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
26
|
Chung DY, Oka F, Jin G, Harriott A, Kura S, Aykan SA, Qin T, Edmiston WJ, Lee H, Yaseen MA, Sakadžić S, Boas DA, Whalen MJ, Ayata C. Subarachnoid hemorrhage leads to early and persistent functional connectivity and behavioral changes in mice. J Cereb Blood Flow Metab 2021; 41:975-985. [PMID: 32936728 PMCID: PMC8054726 DOI: 10.1177/0271678x20940152] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Aneurysmal subarachnoid hemorrhage (SAH) leads to significant long-term cognitive deficits, which can be associated with alterations in resting state functional connectivity (RSFC). However, modalities such as fMRI-which is commonly used to assess RSFC in humans-have practical limitations in small animals. Therefore, we used non-invasive optical intrinsic signal imaging to determine the effect of SAH on RSFC in mice up to three months after prechiasmatic blood injection. We assessed Morris water maze (MWM), open field test (OFT), Y-maze, and rotarod performance from approximately two weeks to three months after SAH. Compared to sham, we found that SAH reduced motor, retrosplenial, and visual seed-based connectivity indices. These deficits persisted in retrosplenial and visual cortex seeds at three months. Seed-to-seed analysis confirmed early attenuation of correlation coefficients in SAH mice, which persisted in predominantly posterior network connections at later time points. Seed-independent global and interhemispheric indices of connectivity revealed decreased correlations following SAH for at least one month. SAH led to MWM hidden platform and OFT deficits at two weeks, and Y-maze deficits for at least three months, without altering rotarod performance. In conclusion, experimental SAH leads to early and persistent alterations both in hemodynamically derived measures of RSFC and in cognitive performance.
Collapse
Affiliation(s)
- David Y Chung
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA.,Division of Neurocritical Care, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Fumiaki Oka
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA.,Department of Neurosurgery, Yamaguchi University School of Medicine, Ube, Japan
| | - Gina Jin
- Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA
| | - Andrea Harriott
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA.,Stroke Service, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| | - Sreekanth Kura
- Neurophotonics Center, Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Sanem A Aykan
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Tao Qin
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - William J Edmiston
- Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA
| | - Hang Lee
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Mohammad A Yaseen
- Department of Bioengineering, Northeastern University, Boston, MA, USA.,Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Sava Sakadžić
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA
| | - David A Boas
- Neurophotonics Center, Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Michael J Whalen
- Department of Pediatrics, Massachusetts General Hospital, Boston, MA, USA
| | - Cenk Ayata
- Neurovascular Research Unit, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA.,Stroke Service, Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
27
|
Deng X, Liang C, Qian L, Zhang Q. miR-24 targets HMOX1 to regulate inflammation and neurofunction in rats with cerebral vasospasm after subarachnoid hemorrhage. Am J Transl Res 2021; 13:1064-1074. [PMID: 33841640 PMCID: PMC8014398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 01/11/2021] [Indexed: 06/12/2023]
Abstract
OBJECTIVE To investigate the effects of miR-24 and HMOX1 on the inflammatory response and neurological function in rats with cerebral vasospasm (CVS) after subarachnoid hemorrhage (SAH). METHODS Fifteen Sprague-Dawley rats were randomly assigned to the sham group (sham operation, treated with normal saline). Rat model of SAH-induced CVS was established in 90 rats, and these rats were randomly divided into the model, miR-24 NC (treated with miR-24-NC vector), miR-24 inhibitor (treated with miR-24 inhibitor vector), HMOX-NC (treated with HMOX1-NC vector), oe-HMOX1 (treated with HMOX1 overexpression vector), and miR-24 inhibitor + si-HMOX1 (treated with miR-24 inhibitor and si-HMOX1 vectors) groups. Adenoviral vectors containing the target sequences were injected into the hippocampus of the rats in the corresponding groups. Dual-luciferase reporter assay was conducted to verify the relationship between miR-24 and HMOX1. The learning and memory abilities, neurological function, cerebral edema, permeability of blood-brain barrier, myeloperoxidase activity, and levels of miR-24, HMOX1, interleukin-6, tumor necrosis factor-α, superoxide dismutase, and malondialdehyde in rats were examined. RESULTS miR-24 could negatively regulate HMOX1 expression. SAH-induced CVS was accompanied with increased miR-24 expression and decreased HMOX1 expression. Inhibiting miR-24 expression or enhancing the expression of its down streaming target, HMOX1, could partly reverse the increased oxidation and inflammation as well as functional deficits in the rats. Moreover, the effects of miR-24 inhibitor could be reversed by inhibiting HMOX1 expression. CONCLUSION miR-24 downregulation can promote HMOX1 expression, thereby decreasing the inflammatory response and improving the neurological function of rats with CVS after SAH.
Collapse
Affiliation(s)
- Xiaodong Deng
- Department of Neurosurgery, The First Affiliated Hospital of China Naval Medical UniversityShanghai City, China
| | - Chong Liang
- Department of Neurosurgery, Jinling Hospital, Nanjing University School of MedicineNanjing, Jiangsu Province, China
| | - Lei Qian
- Department of Neurosurgery, The First Affiliated Hospital of China Naval Medical UniversityShanghai City, China
| | - Qi Zhang
- Department of Cerebrovascular Diseases, Brain Hospital Affiliated to Tongji UniversityShanghai City, China
| |
Collapse
|
28
|
Kosyakovsky J, Fine JM, Frey WH, Hanson LR. Mechanisms of Intranasal Deferoxamine in Neurodegenerative and Neurovascular Disease. Pharmaceuticals (Basel) 2021; 14:ph14020095. [PMID: 33513737 PMCID: PMC7911954 DOI: 10.3390/ph14020095] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/15/2022] Open
Abstract
Identifying disease-modifying therapies for neurological diseases remains one of the greatest gaps in modern medicine. Herein, we present the rationale for intranasal (IN) delivery of deferoxamine (DFO), a high-affinity iron chelator, as a treatment for neurodegenerative and neurovascular disease with a focus on its novel mechanisms. Brain iron dyshomeostasis with iron accumulation is a known feature of brain aging and is implicated in the pathogenesis of a number of neurological diseases. A substantial body of preclinical evidence and early clinical data has demonstrated that IN DFO and other iron chelators have strong disease-modifying impacts in Alzheimer’s disease (AD), Parkinson’s disease (PD), ischemic stroke, and intracranial hemorrhage (ICH). Acting by the disease-nonspecific pathway of iron chelation, DFO targets each of these complex diseases via multifactorial mechanisms. Accumulating lines of evidence suggest further mechanisms by which IN DFO may also be beneficial in cognitive aging, multiple sclerosis, traumatic brain injury, other neurodegenerative diseases, and vascular dementia. Considering its known safety profile, targeted delivery method, robust preclinical efficacy, multiple mechanisms, and potential applicability across many neurological diseases, the case for further development of IN DFO is considerable.
Collapse
Affiliation(s)
- Jacob Kosyakovsky
- School of Medicine, University of Virginia, 200 Jeanette Lancaster Way, Charlottesville, VA 22903, USA;
- HealthPartners Neuroscience Center, HealthPartners Institute, Saint Paul, MN 55130, USA; (W.H.F.II); (L.R.H.)
| | - Jared M. Fine
- HealthPartners Neuroscience Center, HealthPartners Institute, Saint Paul, MN 55130, USA; (W.H.F.II); (L.R.H.)
- Correspondence:
| | - William H. Frey
- HealthPartners Neuroscience Center, HealthPartners Institute, Saint Paul, MN 55130, USA; (W.H.F.II); (L.R.H.)
| | - Leah R. Hanson
- HealthPartners Neuroscience Center, HealthPartners Institute, Saint Paul, MN 55130, USA; (W.H.F.II); (L.R.H.)
| |
Collapse
|
29
|
Fernández-Mendívil C, Luengo E, Trigo-Alonso P, García-Magro N, Negredo P, López MG. Protective role of microglial HO-1 blockade in aging: Implication of iron metabolism. Redox Biol 2020; 38:101789. [PMID: 33212416 PMCID: PMC7680814 DOI: 10.1016/j.redox.2020.101789] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 10/06/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
Heme oxygenase-1 (HO-1) is an inducible enzyme known for its anti-inflammatory, antioxidant and neuroprotective effects. However, increased expression of HO-1 during aging and age-related neurodegenerative diseases have been associated to neurotoxic ferric iron deposits. Being microglia responsible for the brain's innate immune response, the aim of this study was to understand the role of microglial HO-1 under inflammatory conditions in aged mice. For this purpose, aged wild type (WT) and LysMCreHmox1△△ (HMOX1M-KO) mice that lack HO-1 in microglial cells, were used. Aged WT mice showed higher basal expression levels of microglial HO-1 in the brain than adult mice. This increase was even higher when exposed to an inflammatory stimulus (LPS via i.p.) and was accompanied by alterations in different iron-related metabolism proteins, resulting in an increase of iron deposits, oxidative stress, ferroptosis and cognitive decline. Furthermore, microglia exhibited a primed phenotype and increased levels of inflammatory markers such as iNOS, p65, IL-1β, TNF-α, Caspase-1 and NLRP3. Interestingly, all these alterations were prevented in aged HMOX1M-KO and WT mice treated with the HO-1 inhibitor ZnPPIX. In order to determine the effects of microglial HO-1-dependent iron overload, aged WT mice were treated with the iron chelator deferoxamine (DFX). DFX caused major improvements in iron, inflammatory and behavioral alterations found in aged mice exposed to LPS. In conclusion, this study highlights how microglial HO-1 overexpression contributes to neurotoxic iron accumulation providing deleterious effects in aged mice exposed to an inflammatory insult. Microglial HO-1 increases with aging and under an acute inflammatory stimulus. LPS-dependent microglial HO-1 upregulation during aging leads to iron overload. Microglial HO-1-dependent iron accumulation leads to ferroptosis. HO-1-dependent iron alterations lead to neuroinflammation. HO-1 inhibitors/iron chelators reduce iron accumulation and neuroinflammation.
Collapse
Affiliation(s)
- Cristina Fernández-Mendívil
- Instituto Teófilo Hernando for Drug Discovery. Department of Pharmacology. School of Medicine. Universidad Autónoma Madrid. Madrid, Spain; Instituto de Investigación Sanitario (IIS-IP), Hospital Universitario de La Princesa, Madrid, Spain
| | - Enrique Luengo
- Instituto Teófilo Hernando for Drug Discovery. Department of Pharmacology. School of Medicine. Universidad Autónoma Madrid. Madrid, Spain; Instituto de Investigación Sanitario (IIS-IP), Hospital Universitario de La Princesa, Madrid, Spain
| | - Paula Trigo-Alonso
- Instituto Teófilo Hernando for Drug Discovery. Department of Pharmacology. School of Medicine. Universidad Autónoma Madrid. Madrid, Spain; Instituto de Investigación Sanitario (IIS-IP), Hospital Universitario de La Princesa, Madrid, Spain
| | - Nuria García-Magro
- Department of Anatomy, Histology and Neuroscience. School of Medicine. Universidad Autónoma de Madrid. Madrid, Spain
| | - Pilar Negredo
- Department of Anatomy, Histology and Neuroscience. School of Medicine. Universidad Autónoma de Madrid. Madrid, Spain
| | - Manuela G López
- Instituto Teófilo Hernando for Drug Discovery. Department of Pharmacology. School of Medicine. Universidad Autónoma Madrid. Madrid, Spain; Instituto de Investigación Sanitario (IIS-IP), Hospital Universitario de La Princesa, Madrid, Spain.
| |
Collapse
|
30
|
Aging and Progression of Beta-Amyloid Pathology in Alzheimer's Disease Correlates with Microglial Heme-Oxygenase-1 Overexpression. Antioxidants (Basel) 2020; 9:antiox9070644. [PMID: 32708329 PMCID: PMC7402118 DOI: 10.3390/antiox9070644] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/13/2020] [Accepted: 07/19/2020] [Indexed: 02/07/2023] Open
Abstract
Neuroinflammation and oxidative stress are being recognized as characteristic hallmarks in many neurodegenerative diseases, especially those that portray proteinopathy, such as Alzheimer’s disease (AD). Heme-oxygenase 1 (HO-1) is an inducible enzyme with antioxidant and anti-inflammatory properties, while microglia are the immune cells in the central nervous system. To elucidate the brain expression profile of microglial HO-1 in aging and AD-progression, we have used the 5xFAD (five familial AD mutations) mouse model of AD and their littermates at different ages (four, eight, 12, and 18 months). Total brain expression of HO-1 was increased with aging and such increase was even higher in 5xFAD animals. In co-localization studies, HO-1 expression was mainly found in microglia vs. other brain cells. The percentage of microglial cells expressing HO-1 and the amount of HO-1 expressed within microglia increased progressively with aging. Furthermore, this upregulation was increased by 2–3-fold in the elder 5xFAD mice. In addition, microglia overexpressing HO-1 was predominately found surrounding beta-amyloid plaques. These results were corroborated using postmortem brain samples from AD patients, where microglial HO-1 was found up-regulated in comparison to brain samples from aged matched non-demented patients. This study demonstrates that microglial HO-1 expression increases with aging and especially with AD progression, highlighting HO-1 as a potential biomarker or therapeutic target for AD.
Collapse
|
31
|
Thomas AJ, Ascanio-Cortez L, Gomez S, Salem M, Maragkos G, Hanafy KA. Defining the Mechanism of Subarachnoid Hemorrhage-Induced Pyrexia. Neurotherapeutics 2020; 17:1160-1169. [PMID: 32372402 PMCID: PMC7609635 DOI: 10.1007/s13311-020-00866-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Fever can affect the majority of patients with subarachnoid hemorrhage (SAH) and many times no identifiable source is found for the fever whether infectious or sterile, like deep vein thrombosis. We hypothesized that fever in SAH is mediated by a NON-cyclo-oxygenase-dependent mechanism, which we neologized as subarachnoid hemorrhage-induced pyrexia (SAHiP). This hypothesis was investigated using genetically modified mice, pharmacological manipulation, cerebrospinal fluid from SAH patients, and a large cohort of SAH patients. Mice with deletions of neuronal prostaglandin EP3 receptor, global toll-like receptor 4 (TLR4), myeloid TLR4, and microglial TLR4 were subjected to SAH after being implanted with thermometers. Pathways necessary for SAHiP were identified. In SAH patients, cerebrospinal fluid was examined by flow cytometry and correlated with SAHiP. From a large cohort of SAH patients, independent associations with SAHiP were determined using logistic regression analysis. In our mouse model of SAH, microglial TLR4 is necessary for SAHiP, but independent of the neuronal prostaglandin EP3 receptor, cyclo-oxygenase, and prostaglandins. Macrophages from the cerebrospinal fluid of SAH patients with SAHiP expressed more TLR4-co-receptor than SAH patients without SAHiP. In a large cohort of SAH patients, SAHiP was found to be independently, yet inversely, associated with acetaminophen administration. SAHiP is independent of the neuronal prostaglandin EP3 receptor, cyclo-oxygenase, and prostaglandins, but dependent on microglial/macrophage TLR4 with evidence from both SAH mouse models and SAH patients.
Collapse
Affiliation(s)
- Ajith J Thomas
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Luis Ascanio-Cortez
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Santiago Gomez
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Mohamed Salem
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - George Maragkos
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Khalid A Hanafy
- Department of Neurology, Division of Neurointensive Care, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| |
Collapse
|
32
|
Kaiser S, Selzner L, Weber J, Schallner N. Carbon monoxide controls microglial erythrophagocytosis by regulating CD36 surface expression to reduce the severity of hemorrhagic injury. Glia 2020; 68:2427-2445. [PMID: 32476210 DOI: 10.1002/glia.23864] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 04/22/2020] [Accepted: 05/15/2020] [Indexed: 12/27/2022]
Abstract
Microglial erythrophagocytosis is crucial in injury response to hemorrhagic stroke. We hypothesized that regulation of microglial erythrophagocytosis via HO-1/CO depends on a pathway involving reactive oxygen species (ROS) and CD36 surface-expression. The microglial BV-2 cell line and primary microglia (PMG) were incubated +/-blood and +/-CO-exposure. PMG isolated from tissue-specific HO-1-deficient (LyzM-Cre-Hmox1 fl/fl ) and CD36 -/- mice or siRNA against AMPK (AMP-activated protein kinase) were used to test our hypothesis. In a murine subarachnoid hemorrhage (SAH) model, we compared neuronal injury in wild-type and CD36 -/- mice. Readouts included vasospasm, microglia activation, neuronal apoptosis, and spatial memory. We observed increased microglial HO-1-expression after blood-exposure. A burst in ROS-production was seen after CO-exposure, which led to increased amounts of phosphorylated AMPK with subsequently enhanced CD36 surface-expression. Naïve PMG from LyzM-Cre-Hmox1 fl/fl mice showed reduced ROS-production and CD36 surface-expression and failed to respond to CO with increased CD36 surface-expression. Lack of HO-1 and CD36 resulted in reduced erythrophagocytosis that could not be rescued with CO. Erythrophagocytosis was enhanced in BV-2 cells in the presence of exogenous CO, which was abolished in cells treated with siRNA to AMPK. CD36 -/- mice subjected to SAH showed enhanced neuronal cell death, which resulted in impaired spatial memory function. We demonstrate that microglial phagocytic function partly depends on a pathway involving HO-1 with changes in ROS-production, phosphorylated AMPK, and surface expression of CD36. CD36 was identified as a crucial component in blood clearance after hemorrhage that ultimately determines neuronal outcome. These results demand further investigations studying the potential neuroprotective properties of CO.
Collapse
Affiliation(s)
- Sandra Kaiser
- Department of Anesthesiology & Critical Care Medicine, Medical Center, University of Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Germany
| | - Lisa Selzner
- Department of Anesthesiology & Critical Care Medicine, Medical Center, University of Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Germany
| | - Janick Weber
- Department of Anesthesiology & Critical Care Medicine, Medical Center, University of Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Germany
| | - Nils Schallner
- Department of Anesthesiology & Critical Care Medicine, Medical Center, University of Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Germany
| |
Collapse
|
33
|
Peng D, Chen CA, Ruhela D, Li Y, Regan RF. Deferoxamine deconditioning increases neuronal vulnerability to hemoglobin. Exp Cell Res 2020; 390:111926. [PMID: 32112801 PMCID: PMC7301423 DOI: 10.1016/j.yexcr.2020.111926] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 02/12/2020] [Accepted: 02/25/2020] [Indexed: 01/23/2023]
Abstract
Concomitant treatment with deferoxamine (DFO) protects neural cells from iron and heme-mediated oxidative injury, but also disrupts cell responses to iron loading that may be protective. We hypothesized that DFO treatment and withdrawal would subsequently increase neuronal vulnerability to hemoglobin. Pretreatment with DFO followed by its washout increased neuronal loss after subsequent hemoglobin exposure by 3-4-fold compared with control vehicle-pretreated cultures. This was associated with reduced ferritin induction by hemoglobin; expression of heme oxygenase-1, which catalyzes iron release from heme, was not altered. Increased neuronal loss was prevented by exogenous apoferritin or by continuing DFO or antioxidants throughout the experimental course. Cell nonheme iron levels after hemoglobin treatment were similar in DFO-pretreated and control cultures. These results indicate that DFO deconditions neurons and subsequently increases their vulnerability to heme-mediated injury. Its net effect after CNS hemorrhage may be highly dependent on the timing and duration of its administration. Withdrawal of DFO while heme or iron levels remain elevated may be deleterious, and may negate any benefit of prior concomitant therapy.
Collapse
Affiliation(s)
- Denggao Peng
- Department of Emergency Medicine, University of Maryland, School of Medicine, USA
| | - Cindy Acon Chen
- Department of Emergency Medicine, University of Maryland, School of Medicine, USA
| | - Deepa Ruhela
- Department of Emergency Medicine, University of Maryland, School of Medicine, USA
| | - Yang Li
- Department of Emergency Medicine, University of Maryland, School of Medicine, USA
| | - Raymond F Regan
- Department of Emergency Medicine, University of Maryland, School of Medicine, USA.
| |
Collapse
|
34
|
Peng D, Chen CA, Ruhela D, Li Y, Regan RF. Deferoxamine deconditioning increases neuronal vulnerability to hemoglobin. Exp Cell Res 2020; 390:111926. [DOI: https:/doi.org/10.1016/j.yexcr.2020.111926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
35
|
Zera KA, Buckwalter MS. The Local and Peripheral Immune Responses to Stroke: Implications for Therapeutic Development. Neurotherapeutics 2020; 17:414-435. [PMID: 32193840 PMCID: PMC7283378 DOI: 10.1007/s13311-020-00844-3] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The immune response to stroke is an exciting target for future stroke therapies. Stroke is a leading cause of morbidity and mortality worldwide, and clot removal (mechanical or pharmacological) to achieve tissue reperfusion is the only therapy currently approved for patient use. Due to a short therapeutic window and incomplete effectiveness, however, many patients are left with infarcted tissue that stimulates inflammation. Although this is critical to promote repair, it can also damage surrounding healthy brain tissue. In addition, acute immunodepression and subsequent infections are common and are associated with worse patient outcomes. Thus, the acute immune response is a major focus of researchers attempting to identify ways to amplify its benefits and suppress its negative effects to improve short-term recovery of patients. Here we review what is known about this powerful process. This includes the role of brain resident cells such as microglia, peripherally activated cells such as macrophages and neutrophils, and activated endothelium. The role of systemic immune activation and subsequent immunodepression in the days after stroke is also discussed, as is the chronic immune responses and its effects on cognitive function. The biphasic role of inflammation, as well as complex timelines of cell production, differentiation, and trafficking, suggests that the relationship between the acute and chronic phases of stroke recovery is complex. Gaining a more complete understanding of this intricate process by which inflammation is initiated, propagated, and terminated may potentially lead to therapeutics that can treat a larger population of stroke patients than what is currently available. The immune response plays a critical role in patient recovery in both the acute and chronic phases after stroke. In patients, the immune response can be beneficial by promoting repair and recovery, and also detrimental by propagating a pro-inflammatory microenvironment. Thus, it is critical to understand the mechanisms of immune activation following stroke in order to successfully design therapeutics.
Collapse
Affiliation(s)
- Kristy A Zera
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Marion S Buckwalter
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA.
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA.
- Department of Neurosurgery, Stanford Univeristy School of Medicine, Stanford, CA, USA.
| |
Collapse
|
36
|
Zhang R, Bai Q, Liu Y, Zhang Y, Sheng Z, Xue M, Yong VW. Intracerebral hemorrhage in translational research. BRAIN HEMORRHAGES 2020. [DOI: 10.1016/j.hest.2020.02.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
37
|
Akamatsu Y, Pagan VA, Hanafy KA. The role of TLR4 and HO-1 in neuroinflammation after subarachnoid hemorrhage. J Neurosci Res 2019; 98:549-556. [PMID: 31468571 PMCID: PMC6980436 DOI: 10.1002/jnr.24515] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/29/2019] [Accepted: 07/29/2019] [Indexed: 02/06/2023]
Abstract
This review on the mechanisms of neuroinflammation following subarachnoid hemorrhage will focus mainly on toll-like receptor 4 (TLR4), Heme Oxygenase-1 (HO-1), and the role of microglia and macrophages in this process. Vasospasm has long been the focus of research in SAH; however, clinical trials have shown that amelioration of vasospasm does not lead to an improved clinical outcome. This necessitates the need for novel avenues of research. Our work has demonstrated that microglial TLR4 and microglial HO-1, not only affects cognitive dysfunction, but also circadian dysrhythmia in a mouse model of SAH. To attempt to translate these findings, we have also begun investigating macrophages in the cerebrospinal fluid of SAH patients. The goal of this review is to provide an update on the role of TLR4, HO-1, and other signal transduction pathways in SAH-induced neuroinflammation.
Collapse
Affiliation(s)
- Yosuke Akamatsu
- Department of Surgery, Division of Neurosurgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Vicente A Pagan
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Khalid A Hanafy
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts.,Division of Neurointensive Care, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
38
|
Heme, Heme Oxygenase, and Endoplasmic Reticulum Stress-A New Insight into the Pathophysiology of Vascular Diseases. Int J Mol Sci 2019; 20:ijms20153675. [PMID: 31357546 PMCID: PMC6695876 DOI: 10.3390/ijms20153675] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/23/2019] [Accepted: 07/24/2019] [Indexed: 02/06/2023] Open
Abstract
The prevalence of vascular disorders continues to rise worldwide. Parallel with that, new pathophysiological pathways have been discovered, providing possible remedies for prevention and therapy in vascular diseases. Growing evidence suggests that endoplasmic reticulum (ER) stress is involved in a number of vasculopathies, including atherosclerosis, vascular brain events, and diabetes. Heme, which is released from hemoglobin or other heme proteins, triggers various pathophysiological consequence, including heme stress as well as ER stress. The potentially toxic free heme is converted by heme oxygenases (HOs) into carbon monoxide (CO), iron, and biliverdin (BV), the latter of which is reduced to bilirubin (BR). Redox-active iron is oxidized and stored by ferritin, an iron sequestering protein which exhibits ferroxidase activity. In recent years, CO, BV, and BR have been shown to control cellular processes such as inflammation, apoptosis, and antioxidant defense. This review covers our current knowledge about how heme induced endoplasmic reticulum stress (HIERS) participates in the pathogenesis of vascular disorders and highlights recent discoveries in the molecular mechanisms of HO-mediated cytoprotection in heme stress and ER stress, as well as crosstalk between ER stress and HO-1. Furthermore, we focus on the translational potential of HIERS and heme oxygenase-1 (HO-1) in atherosclerosis, diabetes mellitus, and brain hemorrhage.
Collapse
|
39
|
Qin Y, Li G, Sun Z, Xu X, Gu J, Gao F. Comparison of the effects of nimodipine and deferoxamine on brain injury in rat with subarachnoid hemorrhage. Behav Brain Res 2019; 367:194-200. [PMID: 30953658 DOI: 10.1016/j.bbr.2019.04.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 03/28/2019] [Accepted: 04/02/2019] [Indexed: 02/05/2023]
Abstract
Subarachnoid hemorrhage (SAH) may lead to brain atrophy and cognitive dysfunction. This study aimed to compare the efficacy of nimodipine and deferoxamine on these sequelae of SAH. A rat model of SAH was established by the double-hemorrhage method. These rats were injected with saline (intraperitoneal, IP), nimodipine (IP), or deferoxamine (IP and intranasal) every 12 h for 5 days after SAH. The MRI scanning, including magnetic resonance angiography, diffusion tensor imaging, T2-weighted imaging, was performed to detect the brain structure. The levels of iron metabolism-related proteins were examined by Western blot analysis. The Morris water maze (MWM) test was used to assess the cognitive function. Then, then neurons in the cortex and hippocampus were counted on hematoxylin and eosin-stained brain sections. Significant cerebral vasospasm (CVS) was found in the saline and deferoxamine groups, but not in the nimodipine group. Cerebral peduncle injury was detected in the saline and nimodipine groups, but not significantly in the deferoxamine group. Compared with nimodipine, deferoxamine reduced transferrin (Tf), Tf receptor, and ferritin levels after SAH. The MWM performances were significantly worse in the saline and nimodipine groups than that in the deferoxamine group. Brain atrophy and neuronal losses were more significant in the saline and nimodipine groups than in the deferoxamine group. Nimodipine significantly ameliorated CVS, but it did not improve the late changes in brain structure and cognitive function. Deferoxamine effectively reduced neuronal cell death and ameliorated cognitive function after SAH.
Collapse
Affiliation(s)
- Yang Qin
- Department of Neurosurgery, The General Hospital of Western Theater Command, Chengdu, China; Department of Geriatrics, The General Hospital of Western Theater Command, Chengdu, China
| | - Gaili Li
- Department of Geriatrics, The General Hospital of Western Theater Command, Chengdu, China
| | - Zhiyong Sun
- Department of Nuclear Medicine, The General Hospital of Western Theater Command, Chengdu, China
| | - Xianhua Xu
- Department of Geriatrics, The General Hospital of Western Theater Command, Chengdu, China
| | - Jianwen Gu
- Department of Neurosurgery, The General Hospital of Western Theater Command, Chengdu, China; Department of Neurosurgery, The 306th Hospital of PLA, Beijing, China.
| | - Fabao Gao
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
40
|
Guo X, Qi X, Li H, Duan Z, Wei Y, Zhang F, Tian M, Ma L, You C. Deferoxamine Alleviates Iron Overload and Brain Injury in a Rat Model of Brainstem Hemorrhage. World Neurosurg 2019; 128:e895-e904. [PMID: 31082547 DOI: 10.1016/j.wneu.2019.05.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/30/2019] [Accepted: 05/02/2019] [Indexed: 02/05/2023]
Abstract
BACKGROUND Brainstem hemorrhage (BSH) is the most dangerous and devastating subtype of intracerebral hemorrhage and is associated with high morbidity and mortality. However, to date, no effective prevention methods or specific therapies have been available to improve its clinical outcomes. We preliminarily explored the efficacy of deferoxamine (DFO), a clinical chelator known for its iron-scavenging activities, in a rat model of BSH induced with collagenase infusion. METHODS DFO or saline was administrated 6 hours after BSH induction and then every 12 hours for ≤7 days. The survival curve of the rats was created, and the neurological scores were examined on days 1, 3, and 7 after BSH. The rats were sacrificed after 1, 3, and 7 days of DFO treatment for histological examination and immunohistochemistry. RESULTS The results showed that administration of DFO delayed erythrocytes lysis, reduced iron deposition, reduced reactive oxygen species generation, reduced heme oxygenase-1 expression, and alleviated brain injury such as neuron degeneration and myelin sheath injury. However, DFO did not improve the survival rate and neurobehavioral outcomes in this model. CONCLUSIONS Administration of DFO had limited therapeutic effects on collagenase-induced brainstem hemorrhage in rats. Some potential explanations were proposed, and more preclinical work is required to clarify the controversial curative effect of DFO in ICH.
Collapse
Affiliation(s)
- Xi Guo
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xin Qi
- Department of Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hao Li
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhongxin Duan
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yang Wei
- Department of Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Fan Zhang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Meng Tian
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China; West China Brain Research Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Lu Ma
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chao You
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Neurosurgery Research Laboratory, West China Hospital, Sichuan University, Chengdu, Sichuan, China; West China Brain Research Centre, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
41
|
Abstract
PURPOSE OF REVIEW To discuss the mechanisms of iron regulation in the brain and the pathophysiological role of deregulation of iron homeostasis following a stroke, and to review existing evidence supporting the potential role of iron chelators in the treatment of ischemic and hemorrhagic stroke. RECENT FINDINGS In recent years, accumulating evidence has highlighted the role of neuroinflammation in neurological injury after ischemic and hemorrhagic stroke, and that free iron is central to this process. Via the Fenton reaction, free iron catalyzes the conversion of superoxide ion and hydrogen peroxide into hydroxyl radicals, which promote oxidative stress. Advances in our understanding of changes in brain iron metabolism and its relationship to neuronal injury in stroke could provide new therapeutic strategies to improve the outcome of stroke patients. Pharmacological agents targeting brain iron regulation hold promise as potentially effective treatments in both ischemic and hemorrhagic stroke.
Collapse
Affiliation(s)
- Khalid A Hanafy
- Beth Israel Deaconess Medical Center, Department of Neurology, Division of Stroke & Cerebrovascular Disease, Harvard Medical School, 330 Brookline Avenue - Palmer 127, Boston, MA, 02215, USA
| | - Joao A Gomes
- Cerebrovascular Center, Cleveland Clinic, Cleveland, OH, USA
| | - Magdy Selim
- Beth Israel Deaconess Medical Center, Department of Neurology, Division of Stroke & Cerebrovascular Disease, Harvard Medical School, 330 Brookline Avenue - Palmer 127, Boston, MA, 02215, USA.
| |
Collapse
|
42
|
DeGregorio-Rocasolano N, Martí-Sistac O, Gasull T. Deciphering the Iron Side of Stroke: Neurodegeneration at the Crossroads Between Iron Dyshomeostasis, Excitotoxicity, and Ferroptosis. Front Neurosci 2019; 13:85. [PMID: 30837827 PMCID: PMC6389709 DOI: 10.3389/fnins.2019.00085] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 01/25/2019] [Indexed: 12/21/2022] Open
Abstract
In general, iron represents a double-edged sword in metabolism in most tissues, especially in the brain. Although the high metabolic demands of brain cells require iron as a redox-active metal for ATP-producing enzymes, the brain is highly vulnerable to the devastating consequences of excessive iron-induced oxidative stress and, as recently found, to ferroptosis as well. The blood-brain barrier (BBB) protects the brain from fluctuations in systemic iron. Under pathological conditions, especially in acute brain pathologies such as stroke, the BBB is disrupted, and iron pools from the blood gain sudden access to the brain parenchyma, which is crucial in mediating stroke-induced neurodegeneration. Each brain cell type reacts with changes in their expression of proteins involved in iron uptake, efflux, storage, and mobilization to preserve its internal iron homeostasis, with specific organelles such as mitochondria showing specialized responses. However, during ischemia, neurons are challenged with excess extracellular glutamate in the presence of high levels of extracellular iron; this causes glutamate receptor overactivation that boosts neuronal iron uptake and a subsequent overproduction of membrane peroxides. This glutamate-driven neuronal death can be attenuated by iron-chelating compounds or free radical scavenger molecules. Moreover, vascular wall rupture in hemorrhagic stroke results in the accumulation and lysis of iron-rich red blood cells at the brain parenchyma and the subsequent presence of hemoglobin and heme iron at the extracellular milieu, thereby contributing to iron-induced lipid peroxidation and cell death. This review summarizes recent progresses made in understanding the ferroptosis component underlying both ischemic and hemorrhagic stroke subtypes.
Collapse
Affiliation(s)
- Núria DeGregorio-Rocasolano
- Cellular and Molecular Neurobiology Research Group, Department of Neurosciences, Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
| | - Octavi Martí-Sistac
- Cellular and Molecular Neurobiology Research Group, Department of Neurosciences, Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain.,Department of Cellular Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Teresa Gasull
- Cellular and Molecular Neurobiology Research Group, Department of Neurosciences, Germans Trias i Pujol Research Institute (IGTP), Badalona, Spain
| |
Collapse
|
43
|
Bennett C, Mohammed F, Álvarez-Ciara A, Nguyen MA, Dietrich WD, Rajguru SM, Streit WJ, Prasad A. Neuroinflammation, oxidative stress, and blood-brain barrier (BBB) disruption in acute Utah electrode array implants and the effect of deferoxamine as an iron chelator on acute foreign body response. Biomaterials 2019; 188:144-159. [PMID: 30343257 PMCID: PMC6300159 DOI: 10.1016/j.biomaterials.2018.09.040] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/20/2018] [Accepted: 09/24/2018] [Indexed: 02/06/2023]
Abstract
The use of intracortical microelectrode arrays has gained significant attention in being able to help restore function in paralysis patients and study the brain in various neurological disorders. Electrode implantation in the cortex causes vasculature or blood-brain barrier (BBB) disruption and thus elicits a foreign body response (FBR) that results in chronic inflammation and may lead to poor electrode performance. In this study, a comprehensive insight into the acute molecular mechanisms occurring at the Utah electrode array-tissue interface is provided to understand the oxidative stress, neuroinflammation, and neurovascular unit (astrocytes, pericytes, and endothelial cells) disruption that occurs following microelectrode implantation. Quantitative real time polymerase chain reaction (qRT-PCR) was used to quantify the gene expression at acute time-points of 48-hr, 72-hr, and 7-days for factors mediating oxidative stress, inflammation, and BBB disruption in rats implanted with a non-functional 4 × 4 Utah array in the somatosensory cortex. During vascular disruption, free iron released into the brain parenchyma can exacerbate the FBR, leading to oxidative stress and thus further contributing to BBB degradation. To reduce the free iron released into the brain tissue, the effects of an iron chelator, deferoxamine mesylate (DFX), was also evaluated.
Collapse
Affiliation(s)
- Cassie Bennett
- Department of Biomedical Engineering, University of Miami, FL, USA
| | - Farrah Mohammed
- Department of Biomedical Engineering, University of Miami, FL, USA
| | | | | | | | - Suhrud M Rajguru
- Department of Biomedical Engineering, University of Miami, FL, USA
| | | | - Abhishek Prasad
- Department of Biomedical Engineering, University of Miami, FL, USA.
| |
Collapse
|
44
|
Haschka D, Volani C, Stefani A, Tymoszuk P, Mitterling T, Holzknecht E, Heidbreder A, Coassin S, Sumbalova Z, Seifert M, Dichtl S, Theurl I, Gnaiger E, Kronenberg F, Frauscher B, Högl B, Weiss G. Association of mitochondrial iron deficiency and dysfunction with idiopathic restless legs syndrome. Mov Disord 2018; 34:114-123. [DOI: 10.1002/mds.27482] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 07/11/2018] [Accepted: 07/16/2018] [Indexed: 12/11/2022] Open
Affiliation(s)
- David Haschka
- Department of Internal Medicine II Medical University of Innsbruck Innsbruck Austria
| | - Chiara Volani
- Department of Internal Medicine II Medical University of Innsbruck Innsbruck Austria
| | - Ambra Stefani
- Department of Neurology Medical University of Innsbruck Innsbruck Austria
| | - Piotr Tymoszuk
- Department of Internal Medicine II Medical University of Innsbruck Innsbruck Austria
| | - Thomas Mitterling
- Department of Neurology Medical University of Innsbruck Innsbruck Austria
- Department of Neurology Wagner‐Jauregg Hospital Linz Linz Austria
| | - Evi Holzknecht
- Department of Neurology Medical University of Innsbruck Innsbruck Austria
| | - Anna Heidbreder
- Department of Neurology Medical University of Innsbruck Innsbruck Austria
- Department of Neurology, Division of Sleep Medicine and Neuromuscular Disorders University Hospital Muenster Muenster Germany
| | - Stefan Coassin
- Department of Medical Genetics, Division of Genetic Epidemiology, Molecular and Clinical Pharmacology Medical University of Innsbruck Innsbruck Austria
| | - Zuzana Sumbalova
- Pharmacobiochemical Laboratory of the 3rd Department of Internal Medicine, Faculty of Medicine in Bratislava Comenius University Bratislava Slovakia
| | - Markus Seifert
- Department of Internal Medicine II Medical University of Innsbruck Innsbruck Austria
| | - Stefanie Dichtl
- Department of Internal Medicine II Medical University of Innsbruck Innsbruck Austria
| | - Igor Theurl
- Department of Internal Medicine II Medical University of Innsbruck Innsbruck Austria
| | - Erich Gnaiger
- Department of General and Transplant Surgery, D. Swarovski Research Laboratory Medical University of Innsbruck Innsbruck Austria
| | - Florian Kronenberg
- Department of Medical Genetics, Division of Genetic Epidemiology, Molecular and Clinical Pharmacology Medical University of Innsbruck Innsbruck Austria
| | - Birgit Frauscher
- Department of Neurology Medical University of Innsbruck Innsbruck Austria
| | - Birgit Högl
- Department of Neurology Medical University of Innsbruck Innsbruck Austria
| | - Guenter Weiss
- Department of Internal Medicine II Medical University of Innsbruck Innsbruck Austria
- Christian Doppler Laboratory for Iron Metabolism and Anemia Research Medical University of Innsbruck Innsbruck Austria
| |
Collapse
|
45
|
Suvanish Kumar VS, Pretorius E, Rajanikant GK. The Synergistic Combination of Everolimus and Paroxetine Exerts Post-ischemic Neuroprotection In Vitro. Cell Mol Neurobiol 2018; 38:1383-1397. [PMID: 30062636 PMCID: PMC11481845 DOI: 10.1007/s10571-018-0605-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 07/24/2018] [Indexed: 01/08/2023]
Abstract
Ischemic stroke is a debilitating multi-factorial cerebrovascular disorder, representing an area of tremendous unmet medical need. Combination treatment has been proposed as a promising therapeutic approach towards combating ischemic stroke. The present study employs in vitro oxygen glucose deprivation (OGD) model to evaluate the post-ischemic neuroprotective efficacy of Everolimus and Paroxetine, alone and in combination. Post-OGD treatment with Everolimus and Paroxetine, alone or in combination, significantly improved the cell survival (~ 80%) when compared to the cells subjected to ischemic injury alone. The individual neuroprotective doses of Everolimus and Paroxetine were found to be at 6.25 and 25 nM, respectively. Whereas, the synergistic neuroprotective dose for Everolimus:Paroxetine was 2:10 nM, calculated using the Chou-Talalay combination index and other four mathematical models. The synergistic combination dose downregulated neuroinflammatory genes (Tnf-α, Il1b, Nf-κB, and iNos) and upregulated the neuroprotective genes (Bcl-2, Bcl-xl, Hif-1, and Epo). The mitochondrial functioning and ROS neutralizing ability increased with combination treatment. Further, the active role of nitric oxide synthase and calmodulin were revealed while exploring the bio-activity of Everolimus and Paroxetine through network pharmacology. The present study for the first time demonstrates the synergistic post-ischemic neuroprotective efficacy of combination treatment with Everolimus and Paroxetine in vitro. Taken together, these findings clearly suggest that Everolimus in combination with Paroxetine may represent a promising therapeutic strategy for the treatment of ischemic stroke, further supporting the combination treatment strategy for this debilitating disorder.
Collapse
Affiliation(s)
- V S Suvanish Kumar
- School of Biotechnology, National Institute of Technology Calicut, Calicut, 673601, India
| | - Etheresia Pretorius
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch Private Bag X1, Matieland, 7602, South Africa
| | - G K Rajanikant
- School of Biotechnology, National Institute of Technology Calicut, Calicut, 673601, India.
| |
Collapse
|
46
|
Jing C, Zhang H, Shishido H, Keep RF, Hua Y. Association of Brain CD163 Expression and Brain Injury/Hydrocephalus Development in a Rat Model of Subarachnoid Hemorrhage. Front Neurosci 2018; 12:313. [PMID: 29867324 PMCID: PMC5964168 DOI: 10.3389/fnins.2018.00313] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2018] [Accepted: 04/23/2018] [Indexed: 11/13/2022] Open
Abstract
Hemoglobin contributes to brain cell damage and death following subarachnoid hemorrhage (SAH). While CD163, a hemoglobin scavenger receptor, can mediate the clearance of extracellular hemoglobin it has not been well-studied in SAH. In the current study, a filament perforation SAH model was performed in male rats. T2-weighted and T2*-weighted scans were carried out using a 7.0-Tesla MR scanner 24 h after perforation. T2 lesions and hydrocephalus were determined on T2-weighted images. A grading system based on MRI was used to assess SAH severity. The effects of SAH on CD163 were determined by immunohistochemistry staining and Western blots. SAH led to a marked increase in CD163 levels in cortex, white matter and periventricular regions from days 1 to 7. CD163 stained cells were co-localized with neurons, microglia/macrophages, oligodendrocytes and cleaved caspase-3-positive cells, but not astrocytes. Furthermore, CD163 protein levels were increased in rats with higher SAH grades, the presence of T2 lesions on MRI, or hydrocephalus. In conclusion, CD163 expression is markedly upregulated after SAH. It is associated with more severe hemorrhage, as well as MRI T2 lesion and hydrocephalus development.
Collapse
Affiliation(s)
- Chaohui Jing
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, United States
- Department of Neurosurgery, Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Haining Zhang
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, United States
| | - Hajime Shishido
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, United States
| | - Richard F. Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, United States
| | - Ya Hua
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
47
|
Local Fibrinolytic Therapy for Intraventricular Hemorrhage: A Meta-Analysis of Randomized Controlled trials. World Neurosurg 2017; 107:1016-1024.e1. [PMID: 28778779 DOI: 10.1016/j.wneu.2017.07.135] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Accepted: 07/20/2017] [Indexed: 02/05/2023]
Abstract
BACKGROUND The safety and efficacy of intraventricular fibrinolysis (IVF) in patients with intraventricular hemorrhage (IVH) are unclear. We aimed to determine these issues and to evaluate whether there are differences between recombinant tissue-plasminogen activator (rt-PA) and urokinase according to subgroup analyses. METHODS A meta-analysis was undertaken of randomized controlled trials in patients with IVH that compared the administration of rt-PA or urokinase through extraventricular drainage (EVD) with normal saline through EVD or EVD placement alone. RESULTS Six randomized controlled trials involving 607 patients with IVH were included; 2 trials investigated urokinase and 4 rt-PA. IVF reduced death from any cause at the end of follow-up (risk ratio [RR] 0.63, 95% confidence interval [CI] 0.47-0.83), which was driven mostly by rt-PA (RR 0.65, 95% CI 0.48-0.86). Urokinase did not reduce mortality (RR 0.30, 95% CI 0.06-1.53). However, rt-PA did not reduce the proportion of survivors with poor functional outcome (RR 1.36, 95% CI 1.04-1.77), or the composite endpoint of death and poor functional outcome (RR 0.96, 95% CI 0.83-1.11). IVF neither reduced the need for shunt placement (RR 1.06, 95% CI 0.75-1.49) nor increased ventriculitis (RR 0.57, 95% CI 0.35-0.93) and rebleeding (RR 1.65, 95% CI 0.79-3.45). CONCLUSIONS Although the use of IVF in patients with IVH appears generally safe, its benefit is limited to a reduction in mortality at the expense of an increased number of survivors with moderately-severe to severe disability. Subgroup analyses do not suggest an advantage of IVF with urokinase over rt-PA.
Collapse
|
48
|
Rayasam A, Hsu M, Hernández G, Kijak J, Lindstedt A, Gerhart C, Sandor M, Fabry Z. Contrasting roles of immune cells in tissue injury and repair in stroke: The dark and bright side of immunity in the brain. Neurochem Int 2017; 107:104-116. [PMID: 28245997 DOI: 10.1016/j.neuint.2017.02.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Revised: 02/14/2017] [Accepted: 02/16/2017] [Indexed: 01/09/2023]
Abstract
Despite considerable efforts in research and clinical studies, stroke is still one of the leading causes of death and disability worldwide. Originally, stroke was considered a vascular thrombotic disease without significant immune involvement. However, over the last few decades it has become increasingly obvious that the immune responses can significantly contribute to both tissue injury and protection following stroke. Recently, much research has been focused on the immune system's role in stroke pathology and trying to elucidate the mechanism used by immune cells in tissue injury and protection. Since the discovery of tissue plasminogen activator therapy in 1996, there have been no new treatments for stroke. For this reason, research into understanding how the immune system contributes to stroke pathology may lead to better therapies or enhance the efficacy of current treatments. Here, we discuss the contrasting roles of immune cells to stroke pathology while emphasizing myeloid cells and T cells. We propose that focusing future research on balancing the beneficial-versus-detrimental roles of immunity may lead to the discovery of better and novel stroke therapies.
Collapse
Affiliation(s)
- Aditya Rayasam
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA; Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Martin Hsu
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA; Neuroscience Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Gianna Hernández
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA; Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Julie Kijak
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Anders Lindstedt
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Christian Gerhart
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Matyas Sandor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA; Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Zsuzsanna Fabry
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA; Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|