1
|
Bailey ZS, Scultetus AH, Korotcov A, Wang P, Yang X, Cardiff K, Yang F, Ahlers ST, Shear DA, Bell RS. Supra-Prophylactic Doses of Enoxaparin Reduces Fibrin Deposition Without Exacerbation of Intracerebral Hemorrhage in a Rat Model of Penetrating Traumatic Brain Injury. J Neurotrauma 2024. [PMID: 39523877 DOI: 10.1089/neu.2023.0060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Deep vein thrombosis and pulmonary embolism prophylaxis is an important part of trauma care. Despite an increased risk of thrombotic complications, the use of venous thrombosis chemoprophylaxis in penetrating traumatic brain injury (pTBI) patients is met with reluctance from neurosurgeons because of concern for the exacerbation of intracerebral hemorrhage. The objective of this study was to provide initial pre-clinical evidence of the effects of Lovenox (LVX) administration following pTBI with significant intracerebral hemorrhage. Sprague-Dawley rats received a penetrating ballistic-like brain injury. Animals were randomly divided into two groups following injury: LVX (25 mg/kg) or vehicle (VEH, saline). LVX or vehicle was administered subcutaneously beginning 24 h after the injury and continued daily for 7 days post-injury. A neurological assessment was performed daily and magnetic resonance imaging (MRI) was performed at baseline, 1, 2, 3, and 7 days post-injury. Following the final MRI, brains were isolated and prepared for histological analysis. Thromboelastography demonstrated dramatic anticoagulation effects which were confirmed by significant increases in partial thromboplastin time (p < 0.001). Daily neurological assessment revealed no worsening of functional deficits following LVX treatment. MRI analysis demonstrated no differences in cerebral edema or intracranial hemorrhage volumes between treatment groups at any tested post-injury time points. However, LVX elicited a significant reduction in fibrin deposition in the ipsilateral striatum and lesion site at 7 days post-injury (p < 0.05). Serum levels of beta-amyloid were decreased at 7 days following LVX treatment (p < 0.05) which may indicate neuroprotective effects but was not correlated to brain levels. The results presented indicate that administration of LVX at a dose capable of inducing anticoagulation is safe in a rodent model of pTBI without exacerbation of intracerebral hemorrhage within the first 7 days of injury.
Collapse
Affiliation(s)
- Zachary S Bailey
- Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Anke H Scultetus
- Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
- Naval Medical Research Center, Silver Spring, Maryland, USA
- Department of Radiology and Radiological Services, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Alexandru Korotcov
- Department of Radiology and Radiological Services, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, Maryland, USA
| | - Ping Wang
- Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Xiaofang Yang
- Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Katherine Cardiff
- Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Fangzhou Yang
- Naval Medical Research Center, Silver Spring, Maryland, USA
| | - Stephen T Ahlers
- Naval Medical Research Center, Silver Spring, Maryland, USA
- Department of Radiology and Radiological Services, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Deborah A Shear
- Walter Reed Army Institute of Research, Silver Spring, Maryland, USA
| | - Randy S Bell
- Department of Radiology and Radiological Services, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
- Avera McKennan Hospital and University Health Center, Sioux Falls, South Dakota, USA
| |
Collapse
|
2
|
Xing G, Mu L, Han B, Zhu R. The silent information regulator 1 agonist SRT1720 reduces experimental intracerebral hemorrhagic brain injury by regulating the blood-brain barrier integrity. Neuroreport 2024; 35:679-686. [PMID: 38874950 DOI: 10.1097/wnr.0000000000002052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Intracerebral hemorrhage (ICH) is a significant public health matter that has no effective treatment. ICH-induced destruction of the blood-brain barrier (BBB) leads to neurological deterioration. Astrocytic sonic hedgehog (SHH) alleviates brain injury by maintaining the integrity of the BBB after ICH. Silent information regulator 1 (SIRT1) is neuroprotective in several central nervous system diseases via BBB regulation. It is also a possible influential factor of the SHH signaling pathway. Nevertheless, the role of SIRT1 on BBB and the underlying pathological process associated with the SHH signaling pathway after ICH remain unclear. We established an intracerebral hemorrhagic mouse model by collagenase injection. SRT1720 (a selective agonist of SIRT1) was used to evaluate the effect of SIRT1 on BBB integrity after ICH. SIRT1 expression was reduced in the mouse brain after ICH. SRT1720 attenuated neurobehavioral impairments and brain edema of ICH mouse. After ICH induction, SRT1720 improved BBB integrity and tight junction expressions in the mouse brain. The SHH signaling pathway-related factors smoothened and glioma-associated oncogene homolog-1 were increased with the intervention of SRT1720, while cyclopamine (a specific inhibitor of the SHH signaling pathway) reversed these effects. These findings suggest that SIRT1 protects from ICH by altering BBB permeability and tight junction expression levels. This process is associated with the SHH signaling pathway, suggesting that SIRT1 may be a potential therapeutic target for ICH.
Collapse
Affiliation(s)
- Gebeili Xing
- Departments of Neurology, Inner Mongolia People's Hospital
| | - Lei Mu
- Geriatrics, Inner Mongolia People's Hospital, Hohhot, Inner Mongolia, China
| | - Bing Han
- Departments of Neurology, Inner Mongolia People's Hospital
| | - Runxiu Zhu
- Departments of Neurology, Inner Mongolia People's Hospital
| |
Collapse
|
3
|
He Q, Lin FX, Su JY, Zhuo LY, Zhu Q, Sun XC, Jiang RC, Yao ZG, Wang L, Dang YW, Liu DZ, Liu Y, Fang WH, Wang FY, Lin YX, Wang AX, Wang DL, Kang DZ. Naoxueshu Oral Liquid Accelerates Post-Craniotomy Hematoma Absorption in Patients: An Open-Label, Multicenter, and Randomized Controlled Trial. Chin J Integr Med 2024; 30:675-683. [PMID: 38570473 DOI: 10.1007/s11655-024-3902-6] [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] [Accepted: 11/15/2023] [Indexed: 04/05/2024]
Abstract
OBJECTIVE To investigate whether Naoxueshu Oral Liquid (NXS) could promote hematoma absorption in post-craniotomy hematoma (PCH) patients. METHODS This is an open-label, multicenter, and randomized controlled trial conducted at 9 hospitals in China. Patients aged 18-80 years with post-craniotomy supratentorial hematoma volume ranging from 10 to 30 mL or post-craniotomy infratentorial hematoma volume less than 10 mL, or intraventricular hemorrhage following cranial surgery were enrolled. They were randomly assigned at a 1:1 ratio to the NXS (10 mL thrice daily for 15 days) or control groups using a randomization code table. Standard medical care was administered in both groups. The primary outcome was the percentage reduction in hematoma volume from day 1 to day 15. The secondary outcomes included the percentage reduction in hematoma volume from day 1 to day 7, the absolute reduction in hematoma volume from day 1 to day 7 and 15, and the change in neurological function from day 1 to day 7 and 15. The safety was closely monitored throughout the study. Moreover, subgroup analysis was performed based on age, gender, history of diabetes, and etiology of intracerebral hemorrhage (ICH). RESULTS A total of 120 patients were enrolled and randomly assigned between March 30, 2018 and April 15, 2020. One patient was lost to follow-up in the control group. Finally, there were 119 patients (60 in the NXS group and 59 in the control group) included in the analysis. In the full analysis set (FAS) analysis, the NXS group had a greater percentage reduction in hematoma volume from day 1 to day 15 than the control group [median (Q1, Q3): 85% (71%, 97%) vs. 76% (53%, 93%), P<0.05]. The secondary outcomes showed no statistical significance between two groups, either in FAS or per-protocol set (P>0.05). Furthermore, no adverse events were reported during the study. In the FAS analysis, the NXS group exhibited a higher percentage reduction in hematoma volume on day 15 in the following subgroups: male patients, patients younger than 65 years, patients without diabetes, or those with initial cranial surgery due to ICH (all P<0.05). CONCLUSIONS The administration of NXS demonstrated the potential to promote the percentage reduction in hematoma volume from day 1 to day 15. This intervention was found to be safe and feasible. The response to NXS may be influenced by patient characteristics. (Registration No. ChiCTR1800017981).
Collapse
Affiliation(s)
- Qiu He
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
- Department of Neurosurgery, Binhai Branch of National Regional Medical Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350209, China
| | - Fu-Xin Lin
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
- Department of Neurosurgery, Binhai Branch of National Regional Medical Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350209, China
- Clinical Research and Translation Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
- Fujian Clinical Research Center for Neurological Diseases, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
- Fujian Provincial Institutes of Brain Disorders and Brain Sciences, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
| | - Jin-Ye Su
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
- Department of Neurosurgery, Binhai Branch of National Regional Medical Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350209, China
| | - Ling-Yun Zhuo
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
- Department of Neurosurgery, Binhai Branch of National Regional Medical Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350209, China
- Clinical Research and Translation Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
- Fujian Clinical Research Center for Neurological Diseases, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
- Fujian Provincial Institutes of Brain Disorders and Brain Sciences, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
| | - Qing Zhu
- Department of Neurosurgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, 215000, China
| | - Xiao-Chuan Sun
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400000, China
| | - Rong-Cai Jiang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, 300000, China
| | - Zhi-Gang Yao
- Department of Neurosurgery, The Third Hospital of Shijiazhuang, Shijiazhuang, 050000, China
| | - Lei Wang
- Department of Neurosurgery, The First College of Clinical Medical Science, China Three Gorges University, Yichang, Hubei Province, 443000, China
- Department of Neurosurgery, Yichang Central People's Hospital, Yichang, Hubei Province, 443000, China
| | - Yan-Wei Dang
- Department of Neurosurgery, Xiangyang No. 1 People's Hospital Affiliated to Hubei University of Medicine, Xiangfan, Hubei Province, 441100, China
| | - De-Zhong Liu
- Department of Neurosurgery, Zhoukou Central Hospital, Zhoukou, Henan Province, 466000, China
| | - Yang Liu
- Department of Neurosurgery, The Third Hospital of Mianyang, Mianyang, Sichuan Province, 621000, China
| | - Wen-Hua Fang
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
- Department of Neurosurgery, Binhai Branch of National Regional Medical Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350209, China
- Clinical Research and Translation Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
- Fujian Clinical Research Center for Neurological Diseases, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
- Fujian Provincial Institutes of Brain Disorders and Brain Sciences, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
| | - Fang-Yu Wang
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
- Department of Neurosurgery, Binhai Branch of National Regional Medical Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350209, China
- Clinical Research and Translation Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
- Fujian Clinical Research Center for Neurological Diseases, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
- Fujian Provincial Institutes of Brain Disorders and Brain Sciences, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
| | - Yuan-Xiang Lin
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
- Department of Neurosurgery, Binhai Branch of National Regional Medical Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350209, China
- Clinical Research and Translation Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
- Fujian Clinical Research Center for Neurological Diseases, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
- Fujian Provincial Institutes of Brain Disorders and Brain Sciences, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
| | - An-Xin Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100000, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, 100000, China
- Department of Clinical Epidemiology and Clinical Trial, Capital Medical University, Beijing, 100000, China
- Beijing Municipal Key Laboratory of Clinical Epidemiology, Beijing, 100000, China
| | - Deng-Liang Wang
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
- Department of Neurosurgery, Binhai Branch of National Regional Medical Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350209, China
- Clinical Research and Translation Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
- Fujian Clinical Research Center for Neurological Diseases, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
- Fujian Provincial Institutes of Brain Disorders and Brain Sciences, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China
| | - De-Zhi Kang
- Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China.
- Department of Neurosurgery, Binhai Branch of National Regional Medical Center, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350209, China.
- Clinical Research and Translation Center, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China.
- Fujian Clinical Research Center for Neurological Diseases, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China.
- Fujian Provincial Institutes of Brain Disorders and Brain Sciences, The First Affiliated Hospital of Fujian Medical University, Fuzhou, 350004, China.
| |
Collapse
|
4
|
Tao C, Li Y, An N, Liu H, Liu Z, Sun Y, Qian Y, Li N, Xing Y, Gao Y. Pathological mechanisms and future therapeutic directions of thrombin in intracerebral hemorrhage: a systematic review. Front Pharmacol 2024; 15:1293428. [PMID: 38698822 PMCID: PMC11063263 DOI: 10.3389/fphar.2024.1293428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 03/05/2024] [Indexed: 05/05/2024] Open
Abstract
Intracerebral hemorrhage (ICH), a common subtype of hemorrhagic stroke, often causes severe disability or death. ICH induces adverse events that might lead to secondary brain injury (SBI), and there is currently a lack of specific effective treatment strategies. To provide a new direction for SBI treatment post-ICH, the systematic review discussed how thrombin impacts secondary injury after ICH through several potentially deleterious or protective mechanisms. We included 39 studies and evaluated them using SYRCLE's ROB tool. Subsequently, we explored the potential molecular mechanisms of thrombin-mediated effects on SBI post-ICH in terms of inflammation, iron deposition, autophagy, and angiogenesis. Furthermore, we described the effects of thrombin in endothelial cells, astrocytes, pericytes, microglia, and neurons, as well as the harmful and beneficial effects of high and low thrombin concentrations on ICH. Finally, we concluded the current research status of thrombin therapy for ICH, which will provide a basis for the future clinical application of thrombin in the treatment of ICH.
Collapse
Affiliation(s)
- Chenxi Tao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
| | - Yuanyuan Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
| | - Na An
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Haoqi Liu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Zhenhong Liu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
| | - Yikun Sun
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ying Qian
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Na Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yanwei Xing
- Guang’an Men Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yonghong Gao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
5
|
Wen T, Zhang Z. Cellular mechanisms of fibrin (ogen): insight from neurodegenerative diseases. Front Neurosci 2023; 17:1197094. [PMID: 37529232 PMCID: PMC10390316 DOI: 10.3389/fnins.2023.1197094] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 06/27/2023] [Indexed: 08/03/2023] Open
Abstract
Neurodegenerative diseases are prevalent and currently incurable conditions that progressively impair cognitive, behavioral, and psychiatric functions of the central or peripheral nervous system. Fibrinogen, a macromolecular glycoprotein, plays a crucial role in the inflammatory response and tissue repair in the human body and interacts with various nervous system cells due to its unique molecular structure. Accumulating evidence suggests that fibrinogen deposits in the brains of patients with neurodegenerative diseases. By regulating pathophysiological mechanisms and signaling pathways, fibrinogen can exacerbate the neuro-pathological features of neurodegenerative diseases, while depletion of fibrinogen contributes to the amelioration of cognitive function impairment in patients. This review comprehensively summarizes the molecular mechanisms and biological functions of fibrinogen in central nervous system cells and neurodegenerative diseases, including Alzheimer's disease, Multiple Sclerosis, Parkinson's disease, Vascular dementia, Huntington's disease, and Amyotrophic Lateral Sclerosis. Additionally, we discuss the potential of fibrinogen-related treatments in the management of neurodegenerative disorders.
Collapse
|
6
|
Sheu ML, Pan LY, Yang CN, Sheehan J, Pan LY, You WC, Wang CC, Pan HC. Thrombin-Induced Microglia Activation Modulated through Aryl Hydrocarbon Receptors. Int J Mol Sci 2023; 24:11416. [PMID: 37511175 PMCID: PMC10380349 DOI: 10.3390/ijms241411416] [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: 04/07/2023] [Revised: 07/01/2023] [Accepted: 07/03/2023] [Indexed: 07/30/2023] Open
Abstract
Thrombin is a multifunctional serine protein which is closely related to neurodegenerative disorders. The Aryl hydrocarbon receptor (AhR) is well expressed in microglia cells involving inflammatory disorders of the brain. However, it remains unclear as to how modulation of AhR expression by thrombin is related to the development of neurodegeneration disorders. In this study, we investigated the role of AhR in the development of thrombin-induced neurodegenerative processes, especially those concerning microglia. The primary culture of either wild type or AhR deleted microglia, as well as BV-2 cell lines, was used for an in vitro study. Hippocampal slice culture and animals with either wild type or with AhR deleted were used for the ex vivo and in vivo studies. Simulations of ligand protein docking showed a strong integration between the thrombin and AhR. In thrombin-triggered microglia cells, deleting AhR escalated both the NO release and iNOS expression. Such effects were abolished by the administration of the AhR agonist. In thrombin-activated microglia cells, downregulating AhR increased the following: vascular permeability, pro-inflammatory genetic expression, MMP-9 activity, and the ratio of M1/M2 phenotype. In the in vivo study, thrombin induced the activation of microglia and their volume, thereby contributing to the deterioration of neurobehavior. Deleting AhR furthermore aggravated the response in terms of impaired neurobehavior, increasing brain edema, aggregating microglia, and increasing neuronal death. In conclusion, thrombin caused the activation of microglia through increased vessel permeability, expression of inflammatory response, and phenotype of M1 microglia, as well the MMP activity. Deleting AhR augmented the above detrimental effects. These findings indicate that the modulation of AhR is essential for the regulation of thrombin-induced brain damages and that the AhR agonist may harbor the potentially therapeutic effect in thrombin-induced neurodegenerative disorder.
Collapse
Affiliation(s)
- Meei-Ling Sheu
- Institute of Biomedical Sciences, National Chung-Hsing University, Taichung 40227, Taiwan;
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 40210, Taiwan
- Ph.D. Program in Translational Medicine, Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 40227, Taiwan
| | - Liang-Yi Pan
- Faculty of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan;
| | - Cheng-Ning Yang
- Department of Dentistry, School of Dentistry, College of Medicine, National Taiwan University, Taipei 106319, Taiwan;
| | - Jason Sheehan
- Department of Neurosurgery, University of Virginia, Charlottesville, VA 22904, USA;
| | - Liang-Yu Pan
- Faculty of Medicine, Poznan University of Medical Sciences, 61-701 Poznań, Poland;
| | - Weir-Chiang You
- Department of Radiation Oncology, Taichung Veterans General Hospital, Taichung 40210, Taiwan;
| | - Chien-Chia Wang
- Department of Life Sciences, National Central University, Taoyuan 32001, Taiwan;
| | - Hung-Chuan Pan
- Department of Medical Research, Taichung Veterans General Hospital, Taichung 40210, Taiwan
- Ph.D. Program in Translational Medicine, Rong Hsing Research Center for Translational Medicine, National Chung Hsing University, Taichung 40227, Taiwan
- Department of Neurosurgery, Taichung Veterans General Hospital, Taichung 40210, Taiwan
| |
Collapse
|
7
|
Bian Z, Hu X, Liu X, Yu H, Bian Y, Sun H, Fukui Y, Morihara R, Ishiura H, Yamashita T. Protective Effects of Rivaroxaban on White Matter Integrity and Remyelination in a Mouse Model of Alzheimer's Disease Combined with Cerebral Hypoperfusion. J Alzheimers Dis 2023; 96:609-622. [PMID: 37840489 DOI: 10.3233/jad-230413] [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] [Indexed: 10/17/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is characterized by cognitive dysfunction and memory loss that is accompanied by pathological changes to white matter. Some clinical and animal research revealed that AD combined with chronic cerebral hypoperfusion (CCH) exacerbates AD progression by inducing blood-brain barrier dysfunction and fibrinogen deposition. Rivaroxaban, an anticoagulant, has been shown to reduce the rates of dementia in atrial fibrillation patients, but its effects on white matter and the underlying mechanisms are unclear. OBJECTIVE The main purpose of this study was to explore the therapeutic effect of rivaroxaban on the white matter of AD+CCH mice. METHODS In this study, the therapeutic effects of rivaroxaban on white matter in a mouse AD+CCH model were investigated to explore the potential mechanisms involving fibrinogen deposition, inflammation, and oxidative stress on remyelination in white matter. RESULTS The results indicate that rivaroxaban significantly attenuated fibrinogen deposition, fibrinogen-related microglia activation, oxidative stress, and enhanced demyelination in AD+CCH mice, leading to improved white matter integrity, reduced axonal damage, and restored myelin loss. CONCLUSIONS These findings suggest that long-term administration of rivaroxaban might reduce the risk of dementia.
Collapse
Affiliation(s)
- Zhihong Bian
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Xinran Hu
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Xia Liu
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haibo Yu
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Yuting Bian
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Hongming Sun
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Yusuke Fukui
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Ryuta Morihara
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Hiroyuki Ishiura
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| | - Toru Yamashita
- Department of Neurology, Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Kita-ku, Okayama, Japan
| |
Collapse
|
8
|
Li Y, Tian C, Wei Y, Liu H, An N, Song K, Sun Y, Gao Y, Gao Y. Exploring the pharmacological mechanism of Naoxueshu oral liquid in the treatment of intracerebral hemorrhage through weighted gene co-expression network analysis, network pharmacological and experimental validation. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 108:154530. [PMID: 36356328 DOI: 10.1016/j.phymed.2022.154530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 10/24/2022] [Accepted: 10/26/2022] [Indexed: 06/16/2023]
Abstract
BACKGROUND Intracerebral hemorrhage (ICH) is a life-threatening stroke subtype with high rates of disability and mortality. Naoxueshu oral liquid is a proprietary Chinese medicine that absorbs hematoma and exhibits neuroprotective effects in patients with ICH. However, the underlying mechanisms remain obscure. PURPOSE Exploring and elucidating the pharmacological mechanism of Naoxueshu oral liquid in the treatment of ICH. STUDY DESIGN AND METHODS The Gene Expression Omnibus (GEO) database was used to download the gene expression data on ICH. ICH-related hub modules were obtained by weighted gene co-expression network analysis (WGCNA) of differentially co-expressed genes (DEGs). The Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were conducted using the obtained key modules to identify the ICH-related signaling pathways. Network pharmacology technology was applied to forecast the targets of Naoxueshu oral liquid and to establish a protein-protein interaction (PPI) network of overlapping targets between Naoxueshu oral liquid and ICH. Functional annotation and enrichment pathway analyses of the intersectional targets were performed using the omicsbean database. Finally, we verified the therapeutic role and mechanism of Naoxueshu oral liquid in ICH through molecular docking and experiments. RESULTS Through the WGCNA analysis, combined with network pharmacology, it was found that immune inflammation was closely related to the early pathological mechanism of ICH. Naoxueshu oral liquid suppressed the inflammatory response; hence, it could be a potential drug for ICH treatment. Molecular docking further confirmed that the effective components of Naoxueshu oral liquid docked well with CD163. Finally, the experimental results showed that Naoxueshu oral liquid treatment in the ICH rat model attenuated neurological deficits and neuronal injury, decreased hematoma volume, and promoted hematoma absorption. In addition, Naoxueshu oral liquid treatment also significantly increased the levels of Arg-1, CD163, Nrf2, and HO-1 around hematoma after ICH. CONCLUSION This study demonstrated that Naoxueshu oral liquid attenuated neurological deficits and accelerated hematoma absorption, possibly by suppressing inflammatory responses, which might be related to the regulation of Nrf2/CD163/HO-1 that interfered with the activation of M2 microglia, thus accelerating the clearance and decomposition of hemoglobin in the hematoma.
Collapse
Affiliation(s)
- Yuanyuan Li
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China; Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, 100700, China; Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Chao Tian
- Beijing University of Chinese Medicine, Beijing, 100029, China; China-Japan Friendship Hospital, Beijing, 100029, China
| | - Yufei Wei
- Department of Internal Neurology, First Affiliated Hospital, Guangxi University of Chinese Medicine, Guangxi, 530000, China
| | - Haoqi Liu
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Na An
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Ke Song
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Yikun Sun
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Yonghong Gao
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China; Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, 100700, China.
| | - Ying Gao
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, 100700, China.
| |
Collapse
|
9
|
Wang Q, Tu Y, Huang Y, Chen L, Lin Y, Zhan L, He J. High fibrinogen to albumin ratio is associated with hematoma enlargement in spontaneous intracerebral hemorrhage. J Clin Neurosci 2022; 106:37-42. [DOI: 10.1016/j.jocn.2022.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 08/18/2022] [Accepted: 09/12/2022] [Indexed: 11/15/2022]
|
10
|
Lu J, Tang X, Zhang D, Lan T, Huang Q, Xu P, Liu M, Liu L, Wang J. Didang Tang inhibits intracerebral hemorrhage-induced neuronal injury via ASK1/MKK7/JNK signaling pathway, network pharmacology-based analyses combined with experimental validation. Heliyon 2022; 8:e11407. [PMID: 36387497 PMCID: PMC9647469 DOI: 10.1016/j.heliyon.2022.e11407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/14/2022] [Accepted: 10/31/2022] [Indexed: 11/08/2022] Open
Abstract
Background Intracerebral hemorrhage (ICH) is an acute cerebrovascular disease, which is also a principal consideration for disability. Didang tang (DDT) is a classic traditional Chinese medicine formula for treating ICH. However, its pharmacological mechanism of action has not been elucidated. Materials and methods The TCMSP and BATMAN-TCM databases were used to collect chemical compounds and predict targets of DDT. Protein targets in ICH were identified by GeneCards, OMIM, and DrugBank databases. DDT compounds-ICH targets and protein-protein interaction (PPI) networks were constructed for topological analysis and hub-targets screening. Further, Key biological processes and signaling pathways were identified by GO and KEGG enrichment analyses. Then, an ICH rat model and a Cobaltous Chloride (CoCl2)-induced PC12 cells model were established. Cell viability and lactate dehydrogenase (LDH) release were detected using cck8 and LDH kits. Apoptosis levels were detected by TUNEL assessment and flow cytometry. IL-1β levels were detected by ELISA, while key protein expressions were determined by Western blot. Results A total of 126 active compounds related to DDT and 3,263 therapeutic targets for ICH were predicted. The functional enrichment of the GO and KEGG pathways combined with literature studies suggested that DDT is most likely to influence MAPK and apoptotic signaling pathways for ICH treatment. In vitro and in vivo experiments have shown that DDT remarkably inhibited apoptosis and increased the expression of Bcl-2, while inhibiting Bax and cleaved-Caspase 3. For other enriched core proteins, DDT suppressed the phosphorylation of Src and the expression of c-Myc and IL-1β, and up-regulated the level of MMP-9. The further results showed that, DDT decreased the phosphorylation of ASK1, MKK7, JNK and c-JUN. Conclusion Based on network pharmacology and experimental validation results, our in vivo and in vitro study indicated that ASK1/MKK7/JNK pathway might be the critical target for DDT against ICH.
Collapse
|
11
|
Wang Y, Hu Z, Liu H, Gu Y, Ye M, Lu Q, Lu X, Huang C. Adolescent microglia stimulation produces long-lasting protection against chronic stress-induced behavioral abnormalities in adult male mice. Brain Behav Immun 2022; 105:44-66. [PMID: 35781008 DOI: 10.1016/j.bbi.2022.06.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 06/05/2022] [Accepted: 06/28/2022] [Indexed: 11/18/2022] Open
Abstract
Our previous studies had reported that microglia activation one day before stress exposure prevented the behavioral abnormalities induced by chronic stress in adult mice, and a 10-day interval between microglia stimulation and stress exposure can abolish the prophylactic effect of LPS preinjection on the behavioral abnormalities induced by chronic stress, which, however, could be rescued by repeated LPS injection. This suggests that increased stimulation of microglia results in animals developing a strong ability to prevent deleterious stress stimuli. Because microglia in the adolescent brain exhibit flexible immunological plasticity, we hypothesize that a single low-dose LPS injection during adolescence may provide long-lasting protection against behavioral abnormalities induced by chronic stress in adult mice. As expected, our results showed that a single injection of LPS (100 μg/kg) at post-natal day 28 (PND 28) prevented the development of abnormal behaviors and shifted neuroinflammatory responses toward an anti-inflammatory phenotype in adult mice treated with CSDS at their different stages of the age (PND 56, 140, and 252). Moreover, pretreatment with minocycline or PLX3397 to inhibit microglial activation abolished the prophylactic effect of LPS preinjection after PND 28 on behavioral abnormalities and neuroinflammatory responses induced by CSDS in adult mice at their different stages of the age, PND 56, 140, and 252. These results indicate that stimulation of microglia in adolescence may confer long-lasting protection against neuroinflammatory responses and behavioral abnormalities induced by chronic stress in adult mice. This may offer the potential for the development of a "vaccine-like strategy" to prevent mental disorders.
Collapse
Affiliation(s)
- Yue Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Zhichao Hu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Huijun Liu
- Department of Pharmacy, Yancheng First Hospital, the Fourth Affiliated Hospital of Nantong University, #66 Renmin South Road, Yancheng 224006, Jiangsu, China
| | - Yue Gu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Minxiu Ye
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Qun Lu
- Department of Pharmacy, Nantong Third Hospital Affiliated to Nantong University, #60 Middle Qingnian Road, Nantong 226006, Jiangsu, China
| | - Xu Lu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China.
| |
Collapse
|
12
|
Chen S, Li L, Peng C, Bian C, Ocak PE, Zhang JH, Yang Y, Zhou D, Chen G, Luo Y. Targeting Oxidative Stress and Inflammatory Response for Blood-Brain Barrier Protection in Intracerebral Hemorrhage. Antioxid Redox Signal 2022; 37:115-134. [PMID: 35383484 DOI: 10.1089/ars.2021.0072] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Significance: Blood-brain barrier (BBB) disruption is a major pathological change after intracerebral hemorrhage (ICH) and is both the cause and result of oxidative stress and of the immune response post-ICH. These processes contribute to ICH-induced brain injury. Recent Advances: After the breakdown of cerebral vessels, blood components, including erythrocytes and their metabolites, thrombin, and fibrinogen, can access the cerebral parenchyma through the compromised BBB, triggering oxidative stress and inflammatory cascades. These aggravate BBB disruption and contribute to further infiltration of blood components, resulting in a vicious cycle that exacerbates brain edema and neurological injury after ICH. Experimental and clinical studies have highlighted the role of BBB disruption in ICH-induced brain injury. Critical Issues: In this review, we focus on the strategies to protect the BBB in ICH. Specifically, we summarize the evidence and the underlying mechanisms, including the ICH-induced process of oxidative stress and inflammatory response, and we highlight the potential therapeutic targets to protect BBB integrity after ICH. Future Directions: Future studies should probe the mechanism of ferroptosis as well as oxidative stress-inflammation coupling in BBB disruption after ICH and investigate the effects of antioxidants and immunomodulatory agents in more ICH clinical trials. Antioxid. Redox Signal. 37, 115-134.
Collapse
Affiliation(s)
- Shengpan Chen
- Department of Neurosurgery, Guangdong Provincial People's Hospital, Guangdong Institute of Neuroscience, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Lingzhi Li
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Chao Peng
- Department of Neurosurgery, Guangdong Provincial People's Hospital, Guangdong Institute of Neuroscience, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Chunjing Bian
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Pinar Eser Ocak
- Department of Neurosurgery, Uludag University School of Medicine, Bursa, Turkey
| | - John H Zhang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, California, USA
- Department of Neurosurgery, Loma Linda University, Loma Linda, California, USA
| | - Yong Yang
- Department of Neurosurgery, Guangdong Provincial People's Hospital, Guangdong Institute of Neuroscience, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Dong Zhou
- Department of Neurosurgery, Guangdong Provincial People's Hospital, Guangdong Institute of Neuroscience, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Guangzhong Chen
- Department of Neurosurgery, Guangdong Provincial People's Hospital, Guangdong Institute of Neuroscience, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yumin Luo
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
- Beijing Geriatric Medical Research Center and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing, China
| |
Collapse
|
13
|
Wendimu MY, Hooks SB. Microglia Phenotypes in Aging and Neurodegenerative Diseases. Cells 2022; 11:2091. [PMID: 35805174 PMCID: PMC9266143 DOI: 10.3390/cells11132091] [Citation(s) in RCA: 122] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 02/08/2023] Open
Abstract
Neuroinflammation is a hallmark of many neurodegenerative diseases (NDs) and plays a fundamental role in mediating the onset and progression of disease. Microglia, which function as first-line immune guardians of the central nervous system (CNS), are the central drivers of neuroinflammation. Numerous human postmortem studies and in vivo imaging analyses have shown chronically activated microglia in patients with various acute and chronic neuropathological diseases. While microglial activation is a common feature of many NDs, the exact role of microglia in various pathological states is complex and often contradictory. However, there is a consensus that microglia play a biphasic role in pathological conditions, with detrimental and protective phenotypes, and the overall response of microglia and the activation of different phenotypes depends on the nature and duration of the inflammatory insult, as well as the stage of disease development. This review provides a comprehensive overview of current research on the various microglia phenotypes and inflammatory responses in health, aging, and NDs, with a special emphasis on the heterogeneous phenotypic response of microglia in acute and chronic diseases such as hemorrhagic stroke (HS), Alzheimer's disease (AD), and Parkinson's disease (PD). The primary focus is translational research in preclinical animal models and bulk/single-cell transcriptome studies in human postmortem samples. Additionally, this review covers key microglial receptors and signaling pathways that are potential therapeutic targets to regulate microglial inflammatory responses during aging and in NDs. Additionally, age-, sex-, and species-specific microglial differences will be briefly reviewed.
Collapse
Affiliation(s)
| | - Shelley B. Hooks
- Hooks Lab, Department of Pharmaceutical and Biomedical Sciences, College of Pharmacy, University of Georgia, Athens, GA 30602, USA;
| |
Collapse
|
14
|
Li WQ, Qin ZS, Chen S, Cheng D, Yang SC, Choi YMM, Chu B, Zhou WH, Zhang ZJ. Hirudin alleviates acute ischemic stroke by inhibiting NLRP3 inflammasome-mediated neuroinflammation: In vivo and in vitro approaches. Int Immunopharmacol 2022; 110:108967. [PMID: 35724604 DOI: 10.1016/j.intimp.2022.108967] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/21/2022] [Accepted: 06/13/2022] [Indexed: 01/06/2023]
Abstract
Acute ischemic stroke is a severe condition that a vessel supplying blood to the brain is abruptly blocked mostly due to cerebral thrombosis and embolism. There is a dearth of the effective prevention and early intervention strategies. NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome-mediated neuroinflammation plays a crucial role in the pathophysiology of ischemic stroke. Hirudin is a secretion from the salivary glands of the leech Hirudo medicinalis and has a role in regulating inflammation. In this study, hirudin with a dose of 10-40 mg/kg was given to middle cerebral artery occlusion/reperfusion mice. Hirudin markedly constrained cerebral infarct area in a dose-dependent manner, and significantly improved locomotor disability at 40 mg/kg dose. Similar to MCC950, a selective NLRP3 inflammasome inhibitor, hirudin inhibited M1 polarization and promoted M2 polarization. It also strikingly suppressed the ischemia-induced overexpression of NLRP3 and its downstream components, caspase-1, apoptosis-associated speck-like protein (ASC), and interleukin-1β (IL-1β). Hirudin and MCC950 equivalently protected viability and death of BV-2 microglia cells against oxygen-glucose deprivation/reperfusion (OGD/R), an in vitro cell model of brain ischemia. Both agents had similar effects in normalizing the OGD/R-evoked aberrant microglial profiles and NLRP3 pathway dysregulation as observed in the mice. These results demonstrated anti-ischemic effects of hirudin and its association with the inhibition of microglial NLRP3 inflammasome-mediated neuroinflammation. Hirudin is a promising agent for the early intervention of acute ischemic stroke.
Collapse
Affiliation(s)
- Wen-Qi Li
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Department of Chinese Medicine, The University of Hong Kong-Shenzhen Hospital (HKU-SZH), Shenzhen, China
| | - Zong-Shi Qin
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Shuang Chen
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Dan Cheng
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Si-Chang Yang
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | | | - Buggic Chu
- JINKANGDAOFU Bio-Technology Limited Co., Hong Kong, China
| | - Wei-Hai Zhou
- Guangxi KeyKen Research Institute of Natural Hirudin, Nanning, Guangxi, China
| | - Zhang-Jin Zhang
- School of Chinese Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China; Department of Chinese Medicine, The University of Hong Kong-Shenzhen Hospital (HKU-SZH), Shenzhen, China.
| |
Collapse
|
15
|
Magid-Bernstein J, Girard R, Polster S, Srinath A, Romanos S, Awad IA, Sansing LH. Cerebral Hemorrhage: Pathophysiology, Treatment, and Future Directions. Circ Res 2022; 130:1204-1229. [PMID: 35420918 PMCID: PMC10032582 DOI: 10.1161/circresaha.121.319949] [Citation(s) in RCA: 180] [Impact Index Per Article: 90.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Intracerebral hemorrhage (ICH) is a devastating form of stroke with high morbidity and mortality. This review article focuses on the epidemiology, cause, mechanisms of injury, current treatment strategies, and future research directions of ICH. Incidence of hemorrhagic stroke has increased worldwide over the past 40 years, with shifts in the cause over time as hypertension management has improved and anticoagulant use has increased. Preclinical and clinical trials have elucidated the underlying ICH cause and mechanisms of injury from ICH including the complex interaction between edema, inflammation, iron-induced injury, and oxidative stress. Several trials have investigated optimal medical and surgical management of ICH without clear improvement in survival and functional outcomes. Ongoing research into novel approaches for ICH management provide hope for reducing the devastating effect of this disease in the future. Areas of promise in ICH therapy include prognostic biomarkers and primary prevention based on disease pathobiology, ultra-early hemostatic therapy, minimally invasive surgery, and perihematomal protection against inflammatory brain injury.
Collapse
Affiliation(s)
| | - Romuald Girard
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Sean Polster
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Abhinav Srinath
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Sharbel Romanos
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Issam A. Awad
- Neurovascular Surgery Program, Department of Neurological Surgery, University of Chicago Medicine and Biological Sciences, Chicago, Illinois, USA
| | - Lauren H. Sansing
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
16
|
Prognostic Significance of the Combined Score of Plasma Fibrinogen and Neutrophil-Lymphocyte Ratio in Patients with Spontaneous Intracerebral Hemorrhage. DISEASE MARKERS 2022; 2021:7055101. [PMID: 35003392 PMCID: PMC8731292 DOI: 10.1155/2021/7055101] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 12/01/2021] [Indexed: 02/05/2023]
Abstract
Background The combination of plasma fibrinogen and neutrophil to lymphocyte ratio (F-NLR) score is a novel inflammatory marker constituted by peripheral blood fibrinogen concentration and neutrophil to lymphocyte ratio. In the current study, we aim to explore the relationship between admission F-NLR score and intracerebral hemorrhage (ICH) and assess its prognostic predictive ability in ICH patients. Methods The original cohort was consecutively recruited from August 2014 to September 2017, and the validation cohort was consecutively recruited between October 2018 and March 2020. The primary outcomes were 3-month functional outcome and 1-month mortality. All statistical analyses were performed using SPSS and R software. Results A total of 431 and 251 ICH patients were included in original cohort and validation cohort, respectively. In the original cohort, F-NLR score could independently predict the 3-month functional outcome (adjusted OR 2.013, 95% CI 1.316-3.078, p = 0.001) and 1-month mortality (adjusted OR 3.036, 95% CI 1.965-4.693, p < 0.001). Receiver operation characteristic (ROC) analyses and predictive model comparison indicated that F-NLR score had a stronger predictive ability in the 3-month outcome and 1-month mortality. Validation cohort verified the results. Conclusion F-NLR score was an independent indicator for both the 3-month functional outcome and 1-month mortality, and its prognostic predictive ability was superior to fibrinogen and NLR in both the original and the validation cohort.
Collapse
|
17
|
Jiao X, Xing Y, Wang H, Jin X, Zhang T, Peng X, Li R, Liang L, Liu R, Han L, Li Z. A strategy based on gene sequencing and molecular docking for analysis and prediction of bioactive peptides in Shuxuetong injection. Biophys Chem 2021; 282:106749. [PMID: 34971853 DOI: 10.1016/j.bpc.2021.106749] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 12/19/2021] [Accepted: 12/21/2021] [Indexed: 12/15/2022]
Abstract
Peptides are a class of protein fragments with relatively high biological activity and intense specificity, which play crucial role in the treatment of Shuxuetong injection (SXT). However, the extraordinary complexity of Chinese medicinal formulates and the lack of systematic identification methods are primary challenges for study of pharmacodynamic peptides. In addition, infinitesimal peptides contents further hinder the identification and structural characterization of polypeptide by traditional means. In this paper, we described a strategy that LC-MS combined with molecular docking to systematically illustrate the peptide components of SXT. The key to this research was used of gene sequencing to establish a SXT protein database to further achieve the separation and enrichment of chemical methods. Moreover, the ADRA2A, PAR4 and DRD3 were precisely docked with the identified peptides. The result indicated that 12 compounds had stable binding ability and were speculated to be the latent bioactive monomers for the treatment of stroke. Additionally, 12 peptides were verified by cell-based experiment. The results showed that only YLKTT could indeed protect astrocytes from oxygen glucose deprivation/reoxygenation (OGD/R). The YLKTT showed higher activity than the others in vitro. It might be a completely new compound that has never been reported before, providing the basis for further research and a new paradigm for stroke.
Collapse
Affiliation(s)
- Xinyi Jiao
- State Key Laboratory of Component-based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, West Zone of Tuanbo New City, Jinghai District, Tianjin 301617, China
| | - Yanchao Xing
- State Key Laboratory of Component-based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, West Zone of Tuanbo New City, Jinghai District, Tianjin 301617, China
| | - Haitao Wang
- State Key Laboratory of Component-based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, West Zone of Tuanbo New City, Jinghai District, Tianjin 301617, China
| | - Xin Jin
- Military Medicine Section, Logistics University of Chinese People's Armed Police Force, 1 Huizhihuan Road, Dongli District, Tianjin 300309, China
| | - Tingting Zhang
- State Key Laboratory of Component-based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, West Zone of Tuanbo New City, Jinghai District, Tianjin 301617, China
| | - Xingru Peng
- State Key Laboratory of Component-based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, West Zone of Tuanbo New City, Jinghai District, Tianjin 301617, China
| | - Rui Li
- State Key Laboratory of Component-based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, West Zone of Tuanbo New City, Jinghai District, Tianjin 301617, China
| | - Liuyi Liang
- State Key Laboratory of Component-based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, West Zone of Tuanbo New City, Jinghai District, Tianjin 301617, China
| | - Rui Liu
- State Key Laboratory of Component-based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, West Zone of Tuanbo New City, Jinghai District, Tianjin 301617, China.
| | - Lifeng Han
- State Key Laboratory of Component-based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, West Zone of Tuanbo New City, Jinghai District, Tianjin 301617, China.
| | - Zheng Li
- State Key Laboratory of Component-based Chinese Medicine, Haihe Laboratory of Modern Chinese Medicine, College of Pharmaceutical Engineering of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, 10 Poyang Lake Road, West Zone of Tuanbo New City, Jinghai District, Tianjin 301617, China.
| |
Collapse
|
18
|
Bi R, Fang Z, You M, He Q, Hu B. Microglia Phenotype and Intracerebral Hemorrhage: A Balance of Yin and Yang. Front Cell Neurosci 2021; 15:765205. [PMID: 34720885 PMCID: PMC8549831 DOI: 10.3389/fncel.2021.765205] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 09/23/2021] [Indexed: 11/15/2022] Open
Abstract
Intracerebral hemorrhage (ICH) features extremely high rates of morbidity and mortality, with no specific and effective therapy. And local inflammation caused by the over-activated immune cells seriously damages the recovery of neurological function after ICH. Fortunately, immune intervention to microglia has provided new methods and ideas for ICH treatment. Microglia, as the resident immune cells in the brain, play vital roles in both tissue damage and repair processes after ICH. The perihematomal activated microglia not only arouse acute inflammatory responses, oxidative stress, excitotoxicity, and cytotoxicity to cause neuron death, but also show another phenotype that inhibit inflammation, clear hematoma and promote tissue regeneration. The proportion of microglia phenotypes determines the progression of brain tissue damage or repair after ICH. Therefore, microglia may be a promising and imperative therapeutic target for ICH. In this review, we discuss the dual functions of microglia in the brain after an ICH from immunological perspective, elaborate on the activation mechanism of perihematomal microglia, and summarize related therapeutic drugs researches.
Collapse
Affiliation(s)
- Rentang Bi
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhi Fang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mingfeng You
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Quanwei He
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
19
|
Yu HX, Lin W, Yang K, Wei LJ, Chen JL, Liu XY, Zhong K, Chen X, Pei M, Yang HT. Transcriptome-Based Network Analysis Reveals Hirudin Potentiates Anti-Renal Fibrosis Efficacy in UUO Rats. Front Pharmacol 2021; 12:741801. [PMID: 34621173 PMCID: PMC8490886 DOI: 10.3389/fphar.2021.741801] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/07/2021] [Indexed: 12/17/2022] Open
Abstract
Background: Hirudin has been widely used in the treatment of antifibrosis. Previous studies have shown that hirudin can effectively improve the clinical remission rate of chronic kidney disease. However, the mechanism of its renal protection has not been systematically investigated. Methods: In this study, the reliability of UUO-induced renal interstitial fibrosis was evaluated by histopathological verification. High-throughput transcriptome sequencing was used to elucidate the molecular mechanism of hirudin, differentially expressed mRNAs were identified, and their functions were analyzed by GO analysis and GSEA. In addition, the RNA-seq results were validated by in vitro and vivo experiments. Results: We found 322 identical differential expressed genes (IDEs) in the UUO hirudin-treated group compared with the sham group. Functional enrichment analysis indicated that cellular amino acid metabolic processes were the most obvious enrichment pathways in biological processes. In terms of molecular functional enrichment analysis, IDEs were mainly enriched in coenzyme binding, pyridoxal phosphate binding and other pathways. In addition, microbody is the most obvious pathway for cellular components. A total of 115 signaling pathways were enriched, and AMPK, JAK-STAT, and PI3K-Akt signaling pathways were the important signaling pathways enriched. We found that PI3K, p-Akt, and mTOR expression were significantly reduced by hirudin treatment. In particular, our results showed that hirudin could induce a decrease in the expression of autophagy-related proteins such as P62, LC3, Beclin-1 in TGF-β1-induced NRK-52E cells. Conclusion: Our results suggest that hirudin may protect the kidney by ameliorating renal autophagy impairment through modulating the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Hang-Xing Yu
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Wei Lin
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Kang Yang
- Kidney Disease Treatment Center, The First Affiliated Hospital of Henan University of CM, Zhengzhou, China
| | - Li-Juan Wei
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Jun-Li Chen
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xin-Yue Liu
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Ke Zhong
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Xin Chen
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Ming Pei
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Hong-Tao Yang
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.,National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
20
|
Central Nervous System Tissue Regeneration after Intracerebral Hemorrhage: The Next Frontier. Cells 2021; 10:cells10102513. [PMID: 34685493 PMCID: PMC8534252 DOI: 10.3390/cells10102513] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/13/2021] [Accepted: 09/17/2021] [Indexed: 12/11/2022] Open
Abstract
Despite marked advances in surgical techniques and understanding of secondary brain injury mechanisms, the prognosis of intracerebral hemorrhage (ICH) remains devastating. Harnessing and promoting the regenerative potential of the central nervous system may improve the outcomes of patients with hemorrhagic stroke, but approaches are still in their infancy. In this review, we discuss the regenerative phenomena occurring in animal models and human ICH, provide results related to cellular and molecular mechanisms of the repair process including by microglia, and review potential methods to promote tissue regeneration in ICH. We aim to stimulate research involving tissue restoration after ICH.
Collapse
|
21
|
Zou M, Feng Y, Xiu Y, Li Y, Zhang Y, Fan J, Li H, Cao J, He W, Jin WN. Pertussis toxin-induced inflammatory response exacerbates intracerebral haemorrhage and ischaemic stroke in mice. Stroke Vasc Neurol 2021; 7:29-37. [PMID: 34341068 PMCID: PMC8899681 DOI: 10.1136/svn-2021-000987] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Accepted: 07/01/2021] [Indexed: 12/03/2022] Open
Abstract
Background Stroke is a devastating disease, including intracerebral haemorrhage (ICH) and ischaemic stroke. Emerging evidences indicate that systemic inflammatory cascades after stroke contribute to brain damage. However, the direct effects and features of systemic inflammation on brain injury, especially comparing between ischaemic and haemorrhagic stroke, are still obscure. Methods Pertussis toxin (PT) was used to build a pro-inflammatory milieu after ICH and ischaemic stroke in mouse model. The neurodeficits, stroke lesion, immune response and blood–brain barrier (BBB) destruction were assessed. Results In ICH mouse model, PT-induced systemic inflammation exacerbated neurological deficits, and enlarged haemorrhage lesion and perihaematomal oedema. We also found promoted leucocyte infiltration and inflammatory cytokine release into the brain after PT treatment. Moreover, the integrity of the BBB was further disrupted after receiving PT. Furthermore, we demonstrated that PT enhanced brain inflammation and aggravated stroke severity in middle cerebral artery occlusion mouse model. Conclusions Our results suggest that PT increases inflammatory response that exacerbates brain injury after ICH or ischaemic stroke in mouse model.
Collapse
Affiliation(s)
- Ming Zou
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan Feng
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuwhen Xiu
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan Li
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Ying Zhang
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Junwan Fan
- China National Clinical Research Center for Neurological Diseases; Advanced Innovation Center for Human Brain Protection, Beijing Tiantan Hospital, Beijing, China
| | - Haowen Li
- China National Clinical Research Center for Neurological Diseases; Advanced Innovation Center for Human Brain Protection, Beijing Tiantan Hospital, Beijing, China
| | - Jingli Cao
- China National Clinical Research Center for Neurological Diseases; Advanced Innovation Center for Human Brain Protection, Beijing Tiantan Hospital, Beijing, China
| | - Wenyan He
- China National Clinical Research Center for Neurological Diseases; Advanced Innovation Center for Human Brain Protection, Beijing Tiantan Hospital, Beijing, China
| | - Wei-Na Jin
- China National Clinical Research Center for Neurological Diseases; Advanced Innovation Center for Human Brain Protection, Beijing Tiantan Hospital, Beijing, China
| |
Collapse
|
22
|
Li M, Li X, Wang D, Gao X, Li S, Cheng X, Shen Y, Li S, Jia Q, Liu Q. Inhibition of exosome release augments neuroinflammation following intracerebral hemorrhage. FASEB J 2021; 35:e21617. [PMID: 33982343 DOI: 10.1096/fj.202002766r] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 04/03/2021] [Accepted: 04/09/2021] [Indexed: 12/21/2022]
Abstract
Intracerebral hemorrhage (ICH) is a severe stroke subtype without effective pharmacological treatment. Following ICH, peripheral leukocytes infiltrate into the brain and contribute to neuroinflammation and brain edema. However, the intercellular machinery controlling the initiation and propagation of leukocyte infiltration remains elusive. Exosomes are small extracellular vesicles released from donor cells and bridge intercellular communication. In this study, we investigated the effects of inhibition of exosome release on neuroinflammation and ICH injury. Using a mouse model of ICH induced by collagenase injection, we found that ICH induced an increase of exosome level in the brain. Inhibition of exosome release using GW4869 augmented neurological deficits and brain edema after ICH. The exacerbation of ICH injury was accompanied by increased barrier disruption and brain infiltration of leukocytes. The detrimental effects of GW4869 were ablated in ICH mice receiving antibody depletion of Gr-1+ myeloid cells. Extracted exosomes from the ICH brains suppressed the production of inflammatory factors by splenocytes. Additionally, exosomes extracted from brain tissues of donor ICH mice reduced ICH injury in recipient mice. These results demonstrate that inhibition of exosome release augments neuroinflammation and ICH injury. The impact of exosomes released from the ICH brain on the immune system deserves further investigation.
Collapse
Affiliation(s)
- Minshu Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, China
| | - Xiuping Li
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Dan Wang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaolin Gao
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Shiyao Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaojing Cheng
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yiming Shen
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin, China.,Preclinical Multimodal Molecular Imaging Center, Tianjin Medical University General Hospital, Tianjin, China
| | - Shenghui Li
- Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, China.,Preclinical Multimodal Molecular Imaging Center, Tianjin Medical University General Hospital, Tianjin, China
| | - Qiang Jia
- Department of Nuclear Medicine, Tianjin Medical University General Hospital, Tianjin, China.,Preclinical Multimodal Molecular Imaging Center, Tianjin Medical University General Hospital, Tianjin, China
| | - Qiang Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Key Laboratory of Post-neurotrauma Neuro-repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education, Tianjin, China.,Preclinical Multimodal Molecular Imaging Center, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
23
|
Gao L, Shi H, Sherchan P, Tang H, Peng L, Xie S, Liu R, Hu X, Tang J, Xia Y, Zhang JH. Inhibition of lysophosphatidic acid receptor 1 attenuates neuroinflammation via PGE2/EP2/NOX2 signalling and improves the outcome of intracerebral haemorrhage in mice. Brain Behav Immun 2021; 91:615-626. [PMID: 33035633 DOI: 10.1016/j.bbi.2020.09.032] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 08/01/2020] [Accepted: 09/28/2020] [Indexed: 12/16/2022] Open
Abstract
Lysophosphatidic acid receptor 1 (LPA1) plays a critical role in proinflammatory processes in the central nervous system by modulating microglia activation. The aim of this study was to explore the anti-inflammatory effects and neurological function improvement of LPA1 inhibition after intracerebral haemorrhage (ICH) in mice and to determine whether prostaglandin E2 (PGE2), E-type prostaglandin receptor 2 (EP2), and NADPH oxidase 2 (NOX2) signalling are involved in LPA1-mediated neuroinflammation. ICH was induced in CD1 mice by autologous whole blood injection. AM966, a selective LPA1 antagonist, was administered by oral gavage 1 h and 12 h after ICH. The LPA1 endogenous ligand, LPA was administered to verify the effect of LPA1 activation. To elucidate potential inflammatory mechanisms of LPA1, the selective EP2 activator butaprost was administered by intracerebroventricular injection with either AM966 or LPA1 CRISPR knockout (KO). Water content of the brain, neurobehavior, immunofluorescence staining, and western blot were performed. After ICH, EP2 was expressed in microglia whereas LPA1 was expressed in microglia, neurons, and astrocytes, which peaked after 24 h. AM966 inhibition of LPA1 improved neurologic function, reduced brain oedema, and suppressed perihematomal inflammatory cells after ICH. LPA administration aggravated neurological deficits after ICH. AM966 treatment and LPA1 CRISPR KO both decreased the expressions of PGE2, EP2, NOX2, NF-κB, TNF-α, IL-6, and IL-1β expressions after ICH, which was reversed by butaprost. This study demonstrated that inhibition of LPA1 attenuated neuroinflammation caused by ICH via PGE2/EP2/NOX2 signalling pathway in mice, which consequently improved neurobehavioral functions and alleviated brain oedema. LPA1 may be a promising therapeutic target to attenuate ICH-induced secondary brain injury.
Collapse
Affiliation(s)
- Ling Gao
- Department of Neurosurgery, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, Haikou 570208, China; Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Hui Shi
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; Department of Neurosurgery, Affiliated Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Prativa Sherchan
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Hong Tang
- Department of Neurosurgery, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, Haikou 570208, China; Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Li Peng
- Department of Neurosurgery, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, Haikou 570208, China; Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Shucai Xie
- Department of Neurosurgery, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, Haikou 570208, China; Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Rui Liu
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; Department of Neurology, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Xiao Hu
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; Department of Neurology, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Jiping Tang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA
| | - Ying Xia
- Department of Neurosurgery, Affiliated Haikou Hospital, Xiangya School of Medicine, Central South University, Haikou 570208, China.
| | - John H Zhang
- Department of Physiology and Pharmacology, School of Medicine, Loma Linda University, Loma Linda, CA 92354, USA; Department of Neurosurgery and Anesthesiology, Loma Linda University Medical Center, Loma Linda, CA 92354, USA.
| |
Collapse
|
24
|
High CSF thrombin concentration and activity is associated with an unfavorable outcome in patients with intracerebral hemorrhage. PLoS One 2020; 15:e0241565. [PMID: 33175864 PMCID: PMC7657554 DOI: 10.1371/journal.pone.0241565] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/18/2020] [Indexed: 11/19/2022] Open
Abstract
Background The cerebral thrombin system is activated in the early stage after intracerebral hemorrhage (ICH). Expression of thrombin leads to concentration dependent secondary neuronal damage and detrimental neurological outcome. In this study we aimed to investigate the impact of thrombin concentration and activity in the cerebrospinal fluid (CSF) of patients with ICH on clinical outcome. Methods Patients presenting with space-occupying lobar supratentorial hemorrhage requiring extra-ventricular drainage (EVD) were included in our study. The CSF levels of thrombin, its precursor prothrombin and the Thrombin-Antithrombin complex (TAT) were measured using enzyme linked immune sorbent assays (ELISA). The oxidative stress marker Superoxide dismutase (SOD) was assessed in CSF. Initial clot size and intraventricular hemorrhage (IVH) volume was calculated based on by computerized tomography (CT) upon admission to our hospital. Demographic data, clinical status at admission and neurological outcome were assessed using the modified Rankin Scale (mRS) at 6-weeks and 6-month after ICH. Results Twenty-two consecutive patients (9 females, 11 males) with supratentorial hemorrhage were included in this study. CSF concentrations of prothrombin (p < 0.005), thrombin (p = 0.005) and TAT (p = 0.046) were statistical significantly different in patients with ICH compared to non-hemorrhagic CSF samples. CSF concentrations of thrombin 24h after ICH correlated with the mRS index after 6 weeks (r2 = 0.73; < 0.005) and 6 months (r2 = 0.63; < 0.005) after discharge from hospital. Thrombin activity, measured via TAT as surrogate parameter of coagulation, likewise correlated with the mRS at 6 weeks (r2 = 0.54; < 0.01) and 6 months (r2 = 0.66; < 0.04). High thrombin concentrations coincide with higher SOD levels 24h after ICH (p = 0.01). Conclusion In this study we found that initial thrombin concentration and activity in CSF of ICH patients did not correlate with ICH and IVH volume but are associated with a poorer functional neurological outcome. These findings support mounting evidence of the role of thrombin as a contributor to secondary injury formation after ICH.
Collapse
|
25
|
Sulimai N, Lominadze D. Fibrinogen and Neuroinflammation During Traumatic Brain Injury. Mol Neurobiol 2020; 57:4692-4703. [PMID: 32776201 DOI: 10.1007/s12035-020-02012-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 07/08/2020] [Indexed: 12/14/2022]
Abstract
Many neurodegenerative diseases such as Alzheimer's disease (AD), multiple sclerosis, and traumatic brain injury (TBI) are associated with systemic inflammation. Inflammation itself results in increased blood content of fibrinogen (Fg), called hyperfibrinogenemia (HFg). Fg is not only considered an acute phase protein and a marker of inflammation, but has been shown that it can cause inflammatory responses. Fibrin deposits have been associated with memory reduction in neuroinflammatory diseases such as AD and TBI. Reduction in short-term memory has been seen during the most common form of TBI, mild-to-moderate TBI. Fibrin deposits have been found in brains of patients with mild-to-moderate TBI. The vast majority of the literature emphasizes the role of fibrin-activated microglia as the mediator in the neuroinflammation pathway. However, the recent discovery that astrocytes, which constitute approximately 30% of the cells in the mammalian central nervous system, manifest different reactive states warrants further investigations in the causative role of HFg in astrocyte-mediated neuroinflammation. Our previous study showed that Fg deposited in the vasculo-astrocyte interface-activated astrocytes. However, little is known of how Fg directly affects astrocytes and neurons. In this review, we summarize studies that show the effect of Fg on different types of cells in the vasculo-neuronal unit. We will also discuss the possible mechanism of HFg-induced neuroinflammation during TBI.
Collapse
Affiliation(s)
- Nurul Sulimai
- Departments of Surgery, University of South Florida Morsani College of Medicine, MDC-4024, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA
| | - David Lominadze
- Departments of Surgery, University of South Florida Morsani College of Medicine, MDC-4024, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA.
- Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA.
| |
Collapse
|
26
|
Zhou L, Wang D, Qiu X, Zhang W, Gong Z, Wang Y, Xu X. DHZCP Modulates Microglial M1/M2 Polarization via the p38 and TLR4/NF-κB Signaling Pathways in LPS-Stimulated Microglial Cells. Front Pharmacol 2020; 11:1126. [PMID: 32848745 PMCID: PMC7406685 DOI: 10.3389/fphar.2020.01126] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 07/10/2020] [Indexed: 12/12/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a disease with a significantly high rate of morbidity, mortality and disability. Inhibition of inflammation is considered a potential strategy for improving the clinical symptoms induced by ICH. The hallmark of neuroinflammation is microglial activation. Microglia can polarize into either the classically activated M1 (proinflammatory) phenotype, exacerbating neuronal damage, or the alternatively activated M2 (antiinflammatory) phenotype, exerting neuroprotection and promoting neuronal recovery. Promoting microglial polarization to the M2 phenotype may be a viable strategy for treating neuroinflammation. Several studies have indicated that promoting blood circulation and removing blood stasis exhibits therapeutic effects on intracerebral hemorrhage. Dahuang Zhechong Pill (DHZCP), a classical recipe that promotes blood circulation and removes blood stasis, has been reported to improve the clinical outcome of ICH. DHZCP has been shown to exert antiinflammatory effects. However, the detailed antiinflammatory mechanism of DHZCP in ICH has rarely been investigated. In this study, DHZCP inhibited lipopolysaccharide (LPS)-induced M1 microglial activation. DHZCP exerted antiinflammatory effects, by inhibiting LPS-induced M1 proinflammatory cytokine (TNF-α and IL-6), and iNOS production and increasing M2 antiinflammatory cytokine (IL-10) production. DHZCP also switched microglial polarization from M1 to M2, as indicated by significantly increased expression of M2 polarization markers (CD209, and CD206) and markedly decreased expression of an M1 polarization marker (CD54). In addition, DHZCP inhibited p38 and TLR4/NF-κB signaling activation, as demonstrated by inhibition of LPS-induced increases in p-p38, TLR4 and nuclear factor kappa B p-65 (NF-κB p-65) protein expression. Taken together, DHZCP modulates microglial M1/M2 polarization via the p38 and TLR4/NF-κB signaling pathways to confer antiinflammatory effects.
Collapse
Affiliation(s)
- Lu Zhou
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Dongsheng Wang
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Xinjian Qiu
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Weiru Zhang
- Department of General Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Zhicheng Gong
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Yang Wang
- Department of Integrated Traditional Chinese and Western Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Xia Xu
- Department of General Medicine, Xiangya Hospital, Central South University, Changsha, China.,Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
27
|
Ploplis VA, Castellino FJ. Host Pathways of Hemostasis that Regulate Group A Streptococcus pyogenes Pathogenicity. Curr Drug Targets 2020; 21:193-201. [PMID: 31556853 PMCID: PMC7670306 DOI: 10.2174/1389450120666190926152914] [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: 05/17/2019] [Revised: 08/02/2019] [Accepted: 09/06/2019] [Indexed: 11/22/2022]
Abstract
A hallmark feature of severe Group A Streptococcus pyogenes (GAS) infection is dysregulated hemostasis. Hemostasis is the primary pathway for regulating blood flow through events that contribute towards clot formation and its dissolution. However, a number of studies have identified components of hemostasis in regulating survival and dissemination of GAS. Several proteins have been identified on the surface of GAS and they serve to either facilitate invasion to host distal sites or regulate inflammatory responses to the pathogen. GAS M-protein, a surface-exposed virulence factor, appears to be a major target for interactions with host hemostasis proteins. These interactions mediate biochemical events both on the surface of GAS and in the solution when M-protein is released into the surrounding environment through shedding or regulated proteolytic processes that dictate the fate of this pathogen. A thorough understanding of the mechanisms associated with these interactions could lead to novel approaches for altering the course of GAS pathogenicity.
Collapse
Affiliation(s)
- Victoria A. Ploplis
- University of Notre Dame, W.M. Keck Center for Transgene Research, 230 Raclin-Carmichael Hall, Notre Dame, IN 46556 USA
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, USA
| | - Francis J. Castellino
- University of Notre Dame, W.M. Keck Center for Transgene Research, 230 Raclin-Carmichael Hall, Notre Dame, IN 46556 USA
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana, USA
| |
Collapse
|
28
|
Dong X, Nao J, Shi J, Zheng D. Predictive Value of Routine Peripheral Blood Biomarkers in Alzheimer's Disease. Front Aging Neurosci 2019; 11:332. [PMID: 31866854 PMCID: PMC6906180 DOI: 10.3389/fnagi.2019.00332] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 11/18/2019] [Indexed: 12/16/2022] Open
Abstract
Background Biomarker screening is of major significance for the early diagnosis and prevention of Alzheimer’s disease (AD). Routine peripheral blood parameters are easy to collect and detect, making them ideal potential biomarkers. Thus, we aimed to evaluate the parameters from routine blood as potential biomarkers for AD. Methods We enrolled 56 AD patients, 57 mild cognitive impairment (MCI) patients, and 59 healthy elderly controls. Receiver operating characteristic (ROC) curves were used to assess the diagnostic values of routine blood biomarkers in patients with cognitive impairment. Results There were significant differences in eight parameters between the groups. Logistic regression revealed that the neutrophil% (odds ratio (OR) 1.34, 95% confidence interval [CI] 1.03–1.75, p = 0.031) and neutrophil-to-lymphocyte ratio (NLR; OR 6.27, 95% CI 3.98–9.82, p = 0.003) differentiated AD patients and controls (areas under the curve [AUCs], 0.728 and 0.721) and that the NLR (OR 1.93, 95% CI 1.07–3.47, p = 0.028) and mean platelet volume (OR 1.67, 95% CI 1.04–2.70, p = 0.036) differentiated MCI patients and controls (AUCs, 0.60 and 0.638). There were no effective diagnostic biomarkers to distinguish AD from MCI. Conclusion Some routine blood biomarkers may correlate with cognitive impairment. Analysis of these biomarkers, such as the NLR, may be useful for the identification of patients with cognitive impairment.
Collapse
Affiliation(s)
- Xiaoyu Dong
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jianfei Nao
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jile Shi
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Dongming Zheng
- Department of Neurology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|