1
|
Rajab HA, Al-Kuraishy HM, Shokr MM, Al-Gareeb AI, Al-Harchan NA, Alruwaili M, Papadakis M, Alexiou A, Batiha GES. Statins for vascular dementia: A hype or hope. Neuroscience 2025; 567:45-55. [PMID: 39746645 DOI: 10.1016/j.neuroscience.2024.12.059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/28/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
Vascular dementia (VaD) is a second most common type of dementia subsequent to Alzheimer disease (AD). VaD is characterized by cognitive impairment and memory loss that may progress due to the development of cerebral amyloid angiopathy (CAA) a hallmark of AD. CAA triggers the progression of ischemic and hemorrhagic strokes with the subsequent the development of VaD and mixed dementia. Early diagnosis of patients with appropriate use of anti-inflammatory can prevent CAA-related inflammation and VaD development. Currently, there are no effective drugs in the management of VaD. Of note, cholesterol-lowering agent statins which are commonly used in patients with vascular diseases and dyslipidemia may affect the progression of VaD. Many previous studies highlighted the potential therapeutic efficacy of statins in treating VaD. Though, the underlying mechanisms of statins in prevention and treatment of VaD are not fully clarified. Consequently, this review aims to discuss the mechanistic role of statins in the management of VaD, and how statins may adversely affect the cognitive function in VaD patients.
Collapse
Affiliation(s)
- Hussein A Rajab
- Endocrinology Consultant, Medical School, Najran University, Saudi Arabia
| | - Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Mustansiriyah University, Baghdad, Iraq
| | - Mustafa M Shokr
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University- Arish Branch, Arish, 45511, Egypt
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, Jabir Ibn Hayyan Medical University, Kufa, Iraq
| | - Nasser A Al-Harchan
- Department of Clinical Pharmacology, College of Dentistry, Al-Rasheed University, Baghdad, Iraq
| | - Mubarak Alruwaili
- Department of Internal Medicine, College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, Wuppertal, 42283, Germany.
| | - Athanasios Alexiou
- University Centre for Research & Development, Chandigarh University, Mohali, India; Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, New South Wales, Australia; Department of Research & Development, Funogen, Athens, Greece
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, AlBeheira, Egypt
| |
Collapse
|
2
|
Puy L, Kuchcinski G, Leboullenger C, Auger F, Cordonnier C, Bérézowski V. Multimodal and serial MRI monitors brain peri-hematomal injury and repair mechanisms after experimental intracerebral hemorrhage. J Cereb Blood Flow Metab 2025; 45:140-152. [PMID: 39113403 PMCID: PMC11571976 DOI: 10.1177/0271678x241270198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 05/27/2024] [Accepted: 06/16/2024] [Indexed: 11/20/2024]
Abstract
The peri-hematomal area (PHA) emerges as a key but puzzling interface where edematous and neuroinflammatory events co-occur after intracerebral hemorrhage (ICH), while being considered either as deleterious or protective. We aimed at unraveling the pathogeny and natural history of PHA over time after experimental ICH. Male and female rats were longitudinally followed up to day 7 using multimodal brain MRI. MRI measures were compared to neuropathological and behavioural results. While the peak of PHA volume at day 3 was predictive for spontaneous locomotor deficit without sex-effect, its drop at day 7 fitted with locomotor recovery and hematoma resorption. The PHA highest water density was observed at onset despite microvascular hypoperfusion, taken over by blood-brain barrier (BBB) leakage at day 3. Water density dropped at day 7, when vascular integrity was normalized, and the highest number of reactive astrocytes, microglial cells, and siderophages found. This study shows that the PHA with edematous component is hematoma-driven at onset and BBB-driven at day 3, but this excess neuroinflammation enabled PHA volume reduction and significant hematoma resorption as soon as day 7. Therapeutic interventions should consider this pathogeny, and be monitored by multimodal MRI in preclinical ICH models.
Collapse
Affiliation(s)
- Laurent Puy
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Gregory Kuchcinski
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Clémence Leboullenger
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
- Univ. Lille, CNRS, Inserm, CHU Lille, Institut Pasteur de Lille, Lille, France
| | - Florent Auger
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Charlotte Cordonnier
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
| | - Vincent Bérézowski
- Univ. Lille, Inserm, CHU Lille, U1172 - LilNCog - Lille Neuroscience & Cognition, Lille, France
- UArtois, Lens, France
| |
Collapse
|
3
|
Liu JC, Lei SY, Zhang DH, He QY, Sun YY, Zhu HJ, Qu Y, Zhou SY, Yang Y, Li C, Guo ZN. The pleiotropic effects of statins: a comprehensive exploration of neurovascular unit modulation and blood-brain barrier protection. Mol Med 2024; 30:256. [PMID: 39707228 DOI: 10.1186/s10020-024-01025-0] [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: 09/12/2024] [Accepted: 12/03/2024] [Indexed: 12/23/2024] Open
Abstract
The blood-brain barrier (BBB) is the most central component of the neurovascular unit (NVU) and is crucial for the maintenance of the internal environment of the central nervous system and the regulation of homeostasis. A multitude of neuroprotective agents have been developed to exert neuroprotective effects and improve the prognosis of patients with ischemic stroke. These agents have been designed to maintain integrity and promote BBB repair. Statins are widely used as pharmacological agents for the treatment and prevention of ischemic stroke, making them a cornerstone in the pharmacological armamentarium for this condition. The primary mechanism of action is the reduction of serum cholesterol through the inhibition of 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase, which results in a decrease in low-density lipoprotein cholesterol (LDL-C) and an increase in cholesterol clearance. Nevertheless, basic and clinical research has indicated that statins may exert additional pleiotropic effects beyond LDL-C reduction. Previous studies on ischemic stroke have demonstrated that statins can enhance neurological function, reduce inflammation, and promote angiogenic and synaptic processes following ischemic stroke. The BBB has been increasingly recognized for its role in the development and progression of ischemic stroke. Statins have also been found to play a potential BBB protective role by affecting members of the NVU. This review aimed to provide a comprehensive theoretical basis for the clinical application of statins by systematically detailing how statins influence the BBB, particularly focusing on the regulation of the function of each member of the NVU.
Collapse
Affiliation(s)
- Jia-Cheng Liu
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Shuang-Yin Lei
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Dian-Hui Zhang
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Qian-Yan He
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Ying-Ying Sun
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Hong-Jing Zhu
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Yang Qu
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Sheng-Yu Zhou
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
| | - Yi Yang
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, Xinmin Street 1#, Changchun, 130021, China
| | - Chao Li
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China.
| | - Zhen-Ni Guo
- Stroke Center, Department of Neurology, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China.
- Jilin Provincial Key Laboratory of Cerebrovascular Disease, Xinmin Street 1#, Changchun, 130021, China.
- Neuroscience Research Center, The First Hospital of Jilin University, Xinmin Street 1#, Changchun, 130021, China.
| |
Collapse
|
4
|
Kumar Nelson V, Jha NK, Nuli MV, Gupta S, Kanna S, Gahtani RM, Hani U, Singh AK, Abomughaid MM, Abomughayedh AM, Almutary AG, Iqbal D, Al Othaim A, Begum SS, Ahmad F, Mishra PC, Jha SK, Ojha S. Unveiling the impact of aging on BBB and Alzheimer's disease: Factors and therapeutic implications. Ageing Res Rev 2024; 98:102224. [PMID: 38346505 DOI: 10.1016/j.arr.2024.102224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 02/01/2024] [Accepted: 02/03/2024] [Indexed: 05/12/2024]
Abstract
Alzheimer's disease (AD) is a highly prevalent neurodegenerative condition that has devastating effects on individuals, often resulting in dementia. AD is primarily defined by the presence of extracellular plaques containing insoluble β-amyloid peptide (Aβ) and neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau protein (P-tau). In addition, individuals afflicted by these age-related illnesses experience a diminished state of health, which places significant financial strain on their loved ones. Several risk factors play a significant role in the development of AD. These factors include genetics, diet, smoking, certain diseases (such as cerebrovascular diseases, obesity, hypertension, and dyslipidemia), age, and alcohol consumption. Age-related factors are key contributors to the development of vascular-based neurodegenerative diseases such as AD. In general, the process of aging can lead to changes in the immune system's responses and can also initiate inflammation in the brain. The chronic inflammation and the inflammatory mediators found in the brain play a crucial role in the dysfunction of the blood-brain barrier (BBB). Furthermore, maintaining BBB integrity is of utmost importance in preventing a wide range of neurological disorders. Therefore, in this review, we discussed the role of age and its related factors in the breakdown of the blood-brain barrier and the development of AD. We also discussed the importance of different compounds, such as those with anti-aging properties, and other compounds that can help maintain the integrity of the blood-brain barrier in the prevention of AD. This review builds a strong correlation between age-related factors, degradation of the BBB, and its impact on AD.
Collapse
Affiliation(s)
- Vinod Kumar Nelson
- Raghavendra Institute of Pharmaceutical Education and Research, Anantapur, India.
| | - Niraj Kumar Jha
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Centre of Research Impact and Outcome, Chitkara University, Rajpura 140401, Punjab, India; School of Bioengineering & Biosciences, Lovely Professional University, Phagwara 144411, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, India.
| | - Mohana Vamsi Nuli
- Raghavendra Institute of Pharmaceutical Education and Research, Anantapur, India
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - Sandeep Kanna
- Department of pharmaceutics, Chalapathi Institute of Pharmaceutical Sciences, Chalapathi Nagar, Guntur 522034, India
| | - Reem M Gahtani
- Departement of Clinical Laboratory Sciences, King Khalid University, Abha, Saudi Arabia
| | - Umme Hani
- Department of pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Arun Kumar Singh
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology BHU, Varanasi, Uttar Pradesh, India
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| | - Ali M Abomughayedh
- Pharmacy Department, Aseer Central Hospital, Ministry of Health, Saudi Arabia
| | - Abdulmajeed G Almutary
- Department of Biomedical Sciences, College of Health Sciences, Abu Dhabi University, Abu Dhabi, P.O. Box 59911, United Arab Emirates
| | - Danish Iqbal
- Department of Health Information Management, College of Applied Medical Sciences, Buraydah Private Colleges, Buraydah 51418, Saudi Arabia
| | - Ayoub Al Othaim
- Department of Medical Laboratory Sciences, College of Applied Medical Science, Majmaah University, Al-Majmaah 11952, Saudi Arabia.
| | - S Sabarunisha Begum
- Department of Biotechnology, P.S.R. Engineering College, Sivakasi 626140, India
| | - Fuzail Ahmad
- Respiratory Care Department, College of Applied Sciences, Almaarefa University, Diriya, Riyadh, 13713, Saudi Arabia
| | - Prabhu Chandra Mishra
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
| | - Saurabh Kumar Jha
- Department of Zoology, Kalindi College, University of Delhi, 110008, India.
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, P.O. Box 15551, United Arab Emirates
| |
Collapse
|
5
|
Thomas RG, Kim JH, Kim JH, Yoon J, Choi KH, Jeong YY. Treatment of Ischemic Stroke by Atorvastatin-Loaded PEGylated Liposome. Transl Stroke Res 2024; 15:388-398. [PMID: 36639607 DOI: 10.1007/s12975-023-01125-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 12/16/2022] [Accepted: 01/04/2023] [Indexed: 01/15/2023]
Abstract
There is insufficient evidence on the effect of nanoparticles, particularly liposomes loaded with a statin, on acute ischemic stroke. We investigated the impact of atorvastatin-loaded PEG (polyethylene glycol) conjugated liposomes (LipoStatin) on the outcomes in rats with cerebral ischemia-reperfusion. PEGylated liposome loaded with atorvastatin was developed as a nanoparticle to specifically accumulate in an ischemic region and release the drug to ameliorate the harmful effects of the stroke. LipoStatin was administered to rats with transient middle cerebral artery occlusion through the tail vein immediately after reperfusion (LipoStatin group). LipoStatin efficiently accumulated at the cerebral ischemic injury site of the rat. The LipoStatin group showed a significantly reduced infarct volume (p < 0.01) in brain micro-MR imaging and improved neurological function recovery compared to the control group (p < 0.05). In addition, markedly improved brain metabolism using fluorine-18 fluorodeoxyglucose micro-PET/CT imaging was demonstrated in the LipoStatin group compared with the control group (p < 0.01). Mechanistically, as a result of evaluation through IL-1 beta, TNF-alpha, ICAM-1, and Iba-1 mRNA expression levels at 5 days after cerebral ischemia, LipoStatin showed significant anti-inflammatory effects. Protein expression of occludin, JAM-A, Caveolin-1, and eNOS by western blot at 3 days and fluorescent images at 7 days showed considerable recovery of blood-brain barrier breakdown and endothelial dysfunction. PEGylated LipoStatin can be more effectively delivered to the ischemic brain and may have significant neuroprotective effects. Thus, PEGylated LipoStatin can be further developed as a promising targeted therapy for ischemic stroke and other major vascular diseases.
Collapse
Affiliation(s)
- Reju George Thomas
- Department of Radiology, Chonnam National University Medical School and Hwasun Hospital, 322 Seoyang-Ro, Hwasun-Eup, Hwasun-Gun, Jeollanam-Do, 58128, South Korea
| | - Ja-Hae Kim
- Department of Nuclear Medicine, Chonnam National University Medical School and Hospital, Gwangju, South Korea
| | - Ji-Hye Kim
- Department of Neurology, Chonnam National University Medical School and Hwasun Hospital, 322 Seoyang-Ro, Hwasun-Eup, Hwasun-Gun, Jeollanam-Do, 58128, South Korea
| | - Jungwon Yoon
- School of Integrated Technology, Gwangju Institute of Science and Technology, Gwangju, South Korea
| | - Kang-Ho Choi
- Department of Neurology, Chonnam National University Medical School and Hwasun Hospital, 322 Seoyang-Ro, Hwasun-Eup, Hwasun-Gun, Jeollanam-Do, 58128, South Korea.
| | - Yong-Yeon Jeong
- Department of Radiology, Chonnam National University Medical School and Hwasun Hospital, 322 Seoyang-Ro, Hwasun-Eup, Hwasun-Gun, Jeollanam-Do, 58128, South Korea.
| |
Collapse
|
6
|
Lee KH, Carvalho F, Lioutas VA, Heistand E, Das AS, Marchina S, Shoamanesh A, Katsanos AH, Shehadah A, Incontri D, Selim M. Relationship between prior statin therapy and radiological features and clinical outcomes of intracerebral hemorrhage. J Stroke Cerebrovasc Dis 2023; 32:107378. [PMID: 37837803 DOI: 10.1016/j.jstrokecerebrovasdis.2023.107378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 09/16/2023] [Indexed: 10/16/2023] Open
Abstract
OBJECTIVES A post-hoc analysis of the ICH Deferoxamine (i-DEF) trial was performed to examine any associations pre-ICH statin use may have with ICH volume, PHE volume, and clinical outcomes. MATERIALS AND METHODS Baseline characteristics were assessed. Various ICH and PHE parameters were measured via a quantitative, semi-automated method at baseline and follow-up CT scans 72-96 h later. A multivariable logistic regression model was created, adjusting for the variables that were significantly different on univariable analyses (p < 0.05), to assess any associations between pre-ICH statin use and measures of ICH and PHE, as well as good clinical outcome (mRS ≤2), at 90 and 180 days. RESULTS 262 of 291 i-DEF participants had complete data available for analysis. 69 (26.3 %) used statins prior to ICH onset. Pre-ICH statin users had higher prevalences of hypertension, diabetes, and prior ischemic stroke; higher concomitant use of antihypertensives and antiplatelets; and higher blood glucose level at baseline. On univariable analyses, pre-ICH statin users had smaller baseline ICH volume and PHE volume on repeat scan, as well as smaller changes in relative PHE (rPHE) volume and edema extension distance (EED) between the baseline and repeat scans. In the multivariable analysis, none of the ICH and PHE measures or good clinical outcome was significantly associated with pre-ICH statin use. CONCLUSION Pre-ICH statin use was not associated with measures of ICH or PHE, their growth, or clinical outcomes. These findings do not lend support to either overall protective or deleterious effects from statin use before or after ICH.
Collapse
Affiliation(s)
- Kun He Lee
- Department of Neurology, Stroke Division, Temple University Hospital, 3401 N Broad St, Parkinson Pavillion Suite C527, Philadelphia, PA 19140, USA.
| | - Filipa Carvalho
- Department of Neurology, Stroke Division, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA, USA
| | - Vasileios-Arsenios Lioutas
- Department of Neurology, Stroke Division, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA, USA
| | - Elizabeth Heistand
- Department of Neurology, Stroke Division, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA, USA
| | - Alvin S Das
- Department of Neurology, Stroke Division, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA, USA
| | - Sarah Marchina
- Department of Neurology, Stroke Division, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA, USA
| | - Ashkan Shoamanesh
- Division of Neurology, McMaster University / Population Health Research Institute, Hamilton, ON, Canada
| | - Aristeidis H Katsanos
- Division of Neurology, McMaster University / Population Health Research Institute, Hamilton, ON, Canada
| | - Amjad Shehadah
- Department of Neurology, Stroke Division, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA, USA
| | - Diego Incontri
- Department of Neurology, Stroke Division, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA, USA
| | - Magdy Selim
- Department of Neurology, Stroke Division, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, MA, USA
| |
Collapse
|
7
|
Liu J, Zhang S, Jing Y, Zou W. Neutrophil extracellular traps in intracerebral hemorrhage: implications for pathogenesis and therapeutic targets. Metab Brain Dis 2023; 38:2505-2520. [PMID: 37486436 DOI: 10.1007/s11011-023-01268-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 07/19/2023] [Indexed: 07/25/2023]
Abstract
Intracerebral hemorrhage is a common neurological disease, and its pathological mechanism is complex. As the first recruited leukocyte subtype after intracerebral hemorrhage, neutrophils play an important role in tissue damage. In the past, it was considered that neutrophils performed their functions through phagocytosis, chemotaxis, and degranulation. In recent years, studies have found that neutrophils also have the function of secreting extracellular traps. Extracellular traps are fibrous structure composed of chromatin and granular proteins, which plays an important role in innate immunity. Studies have shown a large number of neutrophil extracellular traps in hematoma samples, plasma samples, and drainage samples after intracerebral hemorrhage. In this paper, we summarized the related mechanisms of neutrophil external traps and injury after intracerebral hemorrhage. Neutrophil extracellular traps are involved in the process of brain injury after intracerebral hemorrhage. The application of related inhibitors to inhibit the formation of neutrophil external traps or promote their dissolution can effectively alleviate the pathological damage caused by intracerebral hemorrhage.
Collapse
Affiliation(s)
- Jiawei Liu
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Shuang Zhang
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yunnan Jing
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Wei Zou
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China.
| |
Collapse
|
8
|
Deng X, Yang J, Qing R, Yuan H, Yue P, Tian S. Suppressive role of lovastatin in intracerebral hemorrhage through repression of autophagy. Metab Brain Dis 2023; 38:361-372. [PMID: 36306000 DOI: 10.1007/s11011-022-01101-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 10/08/2022] [Indexed: 02/03/2023]
Abstract
Statins possess critical function in the brain. Here, we intended to investigate the role of lovastatin in brain damage after intracerebral hemorrhage (ICH). A collagenase-induced ICH rat model was established followed by lovastatin treatment. Then, the effect of lovastatin on ICH-induced brain damage was explored with cognitive function, learning and memory abilities, and neurological damage of rats analyzed. Besides, brain water content, number of degenerate neurons, Nissl's body, and apoptosis of neurons were detected. Oxidative stress levels, inflammation, and autophagy levels in ICH were measured after treatment of lovastatin. Lovastatin improved the cognitive impairment of rats, enhanced their spatial learning and memory abilities, reduced nervous system damage, lesion area, and brain water content after ICH. Lovastatin was capable of reducing the number of degenerated neurons, the apoptosis level, autophagy level, and increasing the number of Nissl's body. Lovastatin inhibited the oxidative stress response and inflammatory factors in the brain tissue after ICH, and increased the expression of anti-inflammatory factor IL-10. Lovastatin inhibited AMPK/mTOR signaling pathway after ICH. Our study highlighted the suppressive role of lovastatin in ICH-induced brain damage.
Collapse
Affiliation(s)
- Xiong Deng
- Department of Neurosurgery, the First Affiliated Hospital of Shaoyang University, No. 39, Tongheng Street, Shuangqing District, Shaoyang, Hunan, 422001, People's Republic of China
| | - Jinmei Yang
- Department of Nursing, the First Affiliated Hospital of Shaoyang University, Shaoyang, Hunan, 422001, People's Republic of China
| | - Ruqi Qing
- Department of Neurosurgery, the First Affiliated Hospital of Shaoyang University, No. 39, Tongheng Street, Shuangqing District, Shaoyang, Hunan, 422001, People's Republic of China
| | - Heying Yuan
- Health Management Center, the First Affiliated Hospital of Shaoyang University, Shaoyang, Hunan, 422001, People's Republic of China
| | - Pinhua Yue
- Department of Neurosurgery, the First Affiliated Hospital of Shaoyang University, No. 39, Tongheng Street, Shuangqing District, Shaoyang, Hunan, 422001, People's Republic of China
| | - Song Tian
- Department of Neurosurgery, the First Affiliated Hospital of Shaoyang University, No. 39, Tongheng Street, Shuangqing District, Shaoyang, Hunan, 422001, People's Republic of China.
| |
Collapse
|
9
|
Simani L, Ramezani M, Ahmadi N, Abazari F, Raminfard S, Shojaei M, Zoghi A, Karimialavijeh E, Hossein Aghamiri S, Pakdaman H. The effect of atorvastatin on the blood-brain barrier biomarkers in acute intracerebral hemorrhage, a pilot clinical trial. BRAIN HEMORRHAGES 2022. [DOI: 10.1016/j.hest.2022.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
10
|
Li G, Wang S, Xiong Y, Gu H, Yang K, Yang X, Wang C, Wang C, Li Z, Zhao X. Prior statin and short-term outcomes of primary intracerebral hemorrhage: From a large-scale nationwide longitudinal registry. CNS Neurosci Ther 2022; 28:1240-1248. [PMID: 35603937 PMCID: PMC9253784 DOI: 10.1111/cns.13868] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 04/28/2022] [Accepted: 04/30/2022] [Indexed: 12/16/2022] Open
Abstract
Introduction The relationship between statins and intracerebral hemorrhage outcomes is unclear. Aim We aimed to compare the in‐hospital mortality and evacuation of intracranial hematoma rates in patients with primary intracerebral hemorrhage between prior statin users and nonusers. Results The final study population included 66,263 patients. Multivariable logistics analyses showed that prior statin use was not associated with in‐hospital mortality for primary intracerebral hemorrhage (adjusted odd ratio 0.78, 95% CI 0.61–1.01), but reduced the proportion of patients undergoing evacuation of intracranial hematoma (adjusted odd ratio 0.70, 95% CI 0.61–0.82). Propensity score matching analyses yielded similar results. Conclusion Prior statin use was not associated with in‐hospital mortality but did reduce evacuation of intracranial hematoma rates.
Collapse
Affiliation(s)
- Guangshuo Li
- Vascular Neurology, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Shang Wang
- Neurocardiology Center, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Yunyun Xiong
- Vascular Neurology, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Chinese Institute of Brain Research, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Hongqiu Gu
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Kaixuan Yang
- China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Xin Yang
- China National Clinical Research Center for Neurological Diseases, Beijing, China.,National Center for Healthcare Quality Management in Neurological Diseases, Beijing, China
| | - Chunjuan Wang
- China National Clinical Research Center for Neurological Diseases, Beijing, China.,National Center for Healthcare Quality Management in Neurological Diseases, Beijing, China
| | - Chuanying Wang
- Vascular Neurology, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zixiao Li
- Vascular Neurology, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,Chinese Institute of Brain Research, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Beijing Key Laboratory of Translational Medicine for Cerebrovascular Disease, Beijing, China.,Center for Stroke, Beijing Institute for Brain Disorders, Beijing, China
| | - Xingquan Zhao
- Vascular Neurology, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| |
Collapse
|
11
|
Beyond Lipid-Lowering: Effects of Statins on Cardiovascular and Cerebrovascular Diseases and Cancer. Pharmaceuticals (Basel) 2022; 15:ph15020151. [PMID: 35215263 PMCID: PMC8877351 DOI: 10.3390/ph15020151] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/21/2022] [Accepted: 01/24/2022] [Indexed: 12/15/2022] Open
Abstract
The 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors, also known as statins, are administered as first-line therapy for hypercholesterolemia, both as primary and secondary prevention. Besides the lipid-lowering effect, statins have been suggested to inhibit the development of cardiovascular disease through anti-inflammatory, antioxidant, vascular endothelial function-improving, plaque-stabilizing, and platelet aggregation-inhibiting effects. The preventive effect of statins on atherothrombotic stroke has been well established, but statins can influence other cerebrovascular diseases. This suggests that statins have many neuroprotective effects in addition to lowering cholesterol. Furthermore, research suggests that statins cause pro-apoptotic, growth-inhibitory, and pro-differentiation effects in various malignancies. Preclinical and clinical evidence suggests that statins inhibit tumor growth and induce apoptosis in specific cancer cell types. The pleiotropic effects of statins on cardiovascular and cerebrovascular diseases have been well established; however, the effects of statins on cancer patients have not been fully elucidated and are still controversial. This review discusses the recent evidence on the effects of statins on cardiovascular and cerebrovascular diseases and cancer. Additionally, this study describes the pharmacological action of statins, focusing on the aspect of ‘beyond lipid-lowering’.
Collapse
|
12
|
Trigiani LJ, Bourourou M, Lacalle-Aurioles M, Lecrux C, Hynes A, Spring S, Fernandes DJ, Sled JG, Lesage F, Schwaninger M, Hamel E. A functional cerebral endothelium is necessary to protect against cognitive decline. J Cereb Blood Flow Metab 2022; 42:74-89. [PMID: 34515549 PMCID: PMC8721775 DOI: 10.1177/0271678x211045438] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/23/2021] [Accepted: 07/28/2021] [Indexed: 12/30/2022]
Abstract
A vascular insult occurring early in disease onset may initiate cognitive decline leading to dementia, while pharmacological and lifestyle interventions can prevent this progression. Mice with a selective, tamoxifen-inducible deletion of NF-κB essential modulator (Nemo) in brain endothelial cells were studied as a model of vascular cognitive impairment. Groups included NemoFl controls and three NemobeKO groups: One untreated, and two treated with simvastatin or exercise. Social preference and nesting were impaired in NemobeKO mice and were not countered by treatments. Cerebrovascular function was compromised in NemobeKO groups regardless of treatment, with decreased changes in sensory-evoked cerebral blood flow and total hemoglobin levels, and impaired endothelium-dependent vasodilation. NemobeKO mice had increased string vessel pathology, blood-brain barrier disruption, neuroinflammation, and reduced cortical somatostatin-containing interneurons. These alterations were reversed when endothelial function was recovered. Findings strongly suggest that damage to the cerebral endothelium can trigger pathologies associated with dementia and its functional integrity should be an effective target in future therapeutic efforts.
Collapse
Affiliation(s)
- Lianne J Trigiani
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, Canada
| | - Miled Bourourou
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, Canada
| | - María Lacalle-Aurioles
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, Canada
| | - Clotilde Lecrux
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, Canada
| | - Amy Hynes
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, Canada
| | - Shoshana Spring
- Mouse Imaging Centre (MICe), Hospital for Sick Children, Toronto, Canada
| | - Darren J Fernandes
- Mouse Imaging Centre (MICe), Hospital for Sick Children, Toronto, Canada
| | - John G Sled
- Mouse Imaging Centre (MICe), Hospital for Sick Children, Toronto, Canada
| | - Frédéric Lesage
- Biomedical Engineering Institute, École Polytechnique de Montréal, Montréal, Canada
| | - Markus Schwaninger
- Institute for Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Edith Hamel
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, Montréal, Canada
| |
Collapse
|
13
|
Leal K, Saavedra K, Rebolledo C, Salazar LA. MicroRNAs hsa-miR-618 and hsa-miR-297 Might Modulate the Pleiotropic Effects Exerted by Statins in Endothelial Cells Through the Inhibition of ROCK2 Kinase: in-silico Approach. Front Cardiovasc Med 2021; 8:704175. [PMID: 34485404 PMCID: PMC8415262 DOI: 10.3389/fcvm.2021.704175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 07/20/2021] [Indexed: 01/11/2023] Open
Abstract
Several studies show that statin therapy improves endothelial function by cholesterol-independent mechanisms called “pleiotropic effects.” These are due to the inhibition of the RhoA/ROCK kinase pathway, its inhibition being an attractive atheroprotective treatment. In addition, recent work has shown that microRNAs, posttranscriptional regulators of gene expression, can affect the response of statins and their efficacy. For this reason, the objective of this study was to identify by bioinformatic analysis possible new microRNAs that could modulate the pleiotropic effects exerted by statins through the inhibition of ROCK kinases. A bioinformatic study was performed in which the differential expression of miRNAs in endothelial cells was compared under two conditions: Control and treated with simvastatin at 10 μM for 24 h, using a microarray. Seven miRNAs were differentially expressed, three up and four down. Within the up group, the miRNAs hsa-miR-618 and hsa-miR-297 present as a predicted target to ROCK2 kinase. Also, functional and enriched pathway analysis showed an association with mechanisms associated with atheroprotective effects. This work shows an in-silico approach of how posttranscriptional regulation mediated by miRNAs could modulate the pleiotropic effects exerted by statins on endothelial cells, through the inhibition of ROCK2 kinase and its effects.
Collapse
Affiliation(s)
- Karla Leal
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco, Chile
| | - Kathleen Saavedra
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco, Chile
| | - Camilo Rebolledo
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco, Chile
| | - Luis A Salazar
- Center of Molecular Biology and Pharmacogenetics, Scientific and Technological Bioresource Nucleus, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco, Chile
| |
Collapse
|
14
|
Neuroprotective Therapies for Spontaneous Intracerebral Hemorrhage. Neurocrit Care 2021; 35:862-886. [PMID: 34341912 DOI: 10.1007/s12028-021-01311-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 06/25/2021] [Indexed: 12/15/2022]
Abstract
Patients who survive the initial ictus of spontaneous intracerebral hemorrhage (ICH) remain vulnerable to subsequent injury of the perilesional parenchyma by molecular and cellular responses to the hematoma. Secondary brain injury after ICH, which contributes to long-term functional impairment and mortality, has emerged as an attractive therapeutic target. This review summarizes preclinical and clinical evidence for neuroprotective therapies targeting secondary injury pathways following ICH. A focus on therapies with pleiotropic antiinflammatory effects that target thrombin-mediated chemotaxis and inflammatory cell migration has led to studies investigating statins, anticholinergics, sphingosine-1-phosphate receptor modulators, peroxisome proliferator activated receptor gamma agonists, and magnesium. Attempts to modulate ICH-induced blood-brain barrier breakdown and perihematomal edema formation has prompted studies of nonsteroidal antiinflammatory agents, matrix metalloproteinase inhibitors, and complement inhibitors. Iron chelators, such as deferoxamine and albumin, have been used to reduce the free radical injury that ensues from erythrocyte lysis. Stem cell transplantation has been assessed for its potential to enhance subacute neurogenesis and functional recovery. Despite promising preclinical results of numerous agents, their outcomes have not yet translated into positive clinical trials in patients with ICH. Further studies are necessary to improve our understanding of the molecular events that promote damage and inflammation of the perihematomal parenchyma after ICH. Elucidating the temporal and pathophysiologic features of this secondary brain injury could enhance the clinical efficacy of neuroprotective therapies for ICH.
Collapse
|
15
|
Chen CJ, Ding D, Ironside N, Buell TJ, Elder LJ, Warren A, Adams AP, Ratcliffe SJ, James RF, Naval NS, Worrall BB, Johnston KC, Southerland AM. Statins for neuroprotection in spontaneous intracerebral hemorrhage. Neurology 2019; 93:1056-1066. [PMID: 31712367 DOI: 10.1212/wnl.0000000000008627] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Accepted: 07/24/2019] [Indexed: 12/14/2022] Open
Abstract
Statins, a common drug class for treatment of dyslipidemia, may be neuroprotective for spontaneous intracerebral hemorrhage (ICH) by targeting secondary brain injury pathways in the surrounding brain parenchyma. Statin-mediated neuroprotection may stem from downregulation of mevalonate and its derivatives, targeting key cell signaling pathways that control proliferation, adhesion, migration, cytokine production, and reactive oxygen species generation. Preclinical studies have consistently demonstrated the neuroprotective and recovery enhancement effects of statins, including improved neurologic function, reduced cerebral edema, increased angiogenesis and neurogenesis, accelerated hematoma clearance, and decreased inflammatory cell infiltration. Retrospective clinical studies have reported reduced perihematomal edema, lower mortality rates, and improved functional outcomes in patients who were taking statins before ICH. Several clinical studies have also observed lower mortality rates and improved functional outcomes in patients who were continued or initiated on statins after ICH. Subgroup analysis of a previous randomized trial has raised concerns of a potentially elevated risk of recurrent ICH in patients with previous hemorrhagic stroke who are administered statins. However, most statin trials failed to show an association between statin use and increased hemorrhagic stroke risk. Variable statin dosing, statin use in the pre-ICH setting, and selection biases have limited rigorous investigation of the effects of statins on post-ICH outcomes. Future prospective trials are needed to investigate the association between statin use and outcomes in ICH.
Collapse
Affiliation(s)
- Ching-Jen Chen
- From the Department of Neurological Surgery (C.-J.C., N.I., T.J.B.), University of Virginia Health System, Charlottesville, VA; Department of Neurological Surgery (D.D., R.F.J.), University of Louisville School of Medicine, Louisville, KY; Clinical Trials Office (L.J.E., A.W.), University of Virginia School of Medicine; Department of Pharmacology (A.P.A.), University of Virginia Health System, Charlottesville, VA; Department of Public Health Sciences (S.J.R., B.B.W., A.M.S.), University of Virginia School of Medicine, Charlottesville, VA; Department of Neurosurgery (N.S.N.), Baptist Health, Jacksonville, FL; and Department of Neurology (B.B.W., K.C.J., A.M.S.), University of Virginia Health System, Charlottesville, VA.
| | - Dale Ding
- From the Department of Neurological Surgery (C.-J.C., N.I., T.J.B.), University of Virginia Health System, Charlottesville, VA; Department of Neurological Surgery (D.D., R.F.J.), University of Louisville School of Medicine, Louisville, KY; Clinical Trials Office (L.J.E., A.W.), University of Virginia School of Medicine; Department of Pharmacology (A.P.A.), University of Virginia Health System, Charlottesville, VA; Department of Public Health Sciences (S.J.R., B.B.W., A.M.S.), University of Virginia School of Medicine, Charlottesville, VA; Department of Neurosurgery (N.S.N.), Baptist Health, Jacksonville, FL; and Department of Neurology (B.B.W., K.C.J., A.M.S.), University of Virginia Health System, Charlottesville, VA
| | - Natasha Ironside
- From the Department of Neurological Surgery (C.-J.C., N.I., T.J.B.), University of Virginia Health System, Charlottesville, VA; Department of Neurological Surgery (D.D., R.F.J.), University of Louisville School of Medicine, Louisville, KY; Clinical Trials Office (L.J.E., A.W.), University of Virginia School of Medicine; Department of Pharmacology (A.P.A.), University of Virginia Health System, Charlottesville, VA; Department of Public Health Sciences (S.J.R., B.B.W., A.M.S.), University of Virginia School of Medicine, Charlottesville, VA; Department of Neurosurgery (N.S.N.), Baptist Health, Jacksonville, FL; and Department of Neurology (B.B.W., K.C.J., A.M.S.), University of Virginia Health System, Charlottesville, VA
| | - Thomas J Buell
- From the Department of Neurological Surgery (C.-J.C., N.I., T.J.B.), University of Virginia Health System, Charlottesville, VA; Department of Neurological Surgery (D.D., R.F.J.), University of Louisville School of Medicine, Louisville, KY; Clinical Trials Office (L.J.E., A.W.), University of Virginia School of Medicine; Department of Pharmacology (A.P.A.), University of Virginia Health System, Charlottesville, VA; Department of Public Health Sciences (S.J.R., B.B.W., A.M.S.), University of Virginia School of Medicine, Charlottesville, VA; Department of Neurosurgery (N.S.N.), Baptist Health, Jacksonville, FL; and Department of Neurology (B.B.W., K.C.J., A.M.S.), University of Virginia Health System, Charlottesville, VA
| | - Lori J Elder
- From the Department of Neurological Surgery (C.-J.C., N.I., T.J.B.), University of Virginia Health System, Charlottesville, VA; Department of Neurological Surgery (D.D., R.F.J.), University of Louisville School of Medicine, Louisville, KY; Clinical Trials Office (L.J.E., A.W.), University of Virginia School of Medicine; Department of Pharmacology (A.P.A.), University of Virginia Health System, Charlottesville, VA; Department of Public Health Sciences (S.J.R., B.B.W., A.M.S.), University of Virginia School of Medicine, Charlottesville, VA; Department of Neurosurgery (N.S.N.), Baptist Health, Jacksonville, FL; and Department of Neurology (B.B.W., K.C.J., A.M.S.), University of Virginia Health System, Charlottesville, VA
| | - Amy Warren
- From the Department of Neurological Surgery (C.-J.C., N.I., T.J.B.), University of Virginia Health System, Charlottesville, VA; Department of Neurological Surgery (D.D., R.F.J.), University of Louisville School of Medicine, Louisville, KY; Clinical Trials Office (L.J.E., A.W.), University of Virginia School of Medicine; Department of Pharmacology (A.P.A.), University of Virginia Health System, Charlottesville, VA; Department of Public Health Sciences (S.J.R., B.B.W., A.M.S.), University of Virginia School of Medicine, Charlottesville, VA; Department of Neurosurgery (N.S.N.), Baptist Health, Jacksonville, FL; and Department of Neurology (B.B.W., K.C.J., A.M.S.), University of Virginia Health System, Charlottesville, VA
| | - Amy P Adams
- From the Department of Neurological Surgery (C.-J.C., N.I., T.J.B.), University of Virginia Health System, Charlottesville, VA; Department of Neurological Surgery (D.D., R.F.J.), University of Louisville School of Medicine, Louisville, KY; Clinical Trials Office (L.J.E., A.W.), University of Virginia School of Medicine; Department of Pharmacology (A.P.A.), University of Virginia Health System, Charlottesville, VA; Department of Public Health Sciences (S.J.R., B.B.W., A.M.S.), University of Virginia School of Medicine, Charlottesville, VA; Department of Neurosurgery (N.S.N.), Baptist Health, Jacksonville, FL; and Department of Neurology (B.B.W., K.C.J., A.M.S.), University of Virginia Health System, Charlottesville, VA
| | - Sarah J Ratcliffe
- From the Department of Neurological Surgery (C.-J.C., N.I., T.J.B.), University of Virginia Health System, Charlottesville, VA; Department of Neurological Surgery (D.D., R.F.J.), University of Louisville School of Medicine, Louisville, KY; Clinical Trials Office (L.J.E., A.W.), University of Virginia School of Medicine; Department of Pharmacology (A.P.A.), University of Virginia Health System, Charlottesville, VA; Department of Public Health Sciences (S.J.R., B.B.W., A.M.S.), University of Virginia School of Medicine, Charlottesville, VA; Department of Neurosurgery (N.S.N.), Baptist Health, Jacksonville, FL; and Department of Neurology (B.B.W., K.C.J., A.M.S.), University of Virginia Health System, Charlottesville, VA
| | - Robert F James
- From the Department of Neurological Surgery (C.-J.C., N.I., T.J.B.), University of Virginia Health System, Charlottesville, VA; Department of Neurological Surgery (D.D., R.F.J.), University of Louisville School of Medicine, Louisville, KY; Clinical Trials Office (L.J.E., A.W.), University of Virginia School of Medicine; Department of Pharmacology (A.P.A.), University of Virginia Health System, Charlottesville, VA; Department of Public Health Sciences (S.J.R., B.B.W., A.M.S.), University of Virginia School of Medicine, Charlottesville, VA; Department of Neurosurgery (N.S.N.), Baptist Health, Jacksonville, FL; and Department of Neurology (B.B.W., K.C.J., A.M.S.), University of Virginia Health System, Charlottesville, VA
| | - Neeraj S Naval
- From the Department of Neurological Surgery (C.-J.C., N.I., T.J.B.), University of Virginia Health System, Charlottesville, VA; Department of Neurological Surgery (D.D., R.F.J.), University of Louisville School of Medicine, Louisville, KY; Clinical Trials Office (L.J.E., A.W.), University of Virginia School of Medicine; Department of Pharmacology (A.P.A.), University of Virginia Health System, Charlottesville, VA; Department of Public Health Sciences (S.J.R., B.B.W., A.M.S.), University of Virginia School of Medicine, Charlottesville, VA; Department of Neurosurgery (N.S.N.), Baptist Health, Jacksonville, FL; and Department of Neurology (B.B.W., K.C.J., A.M.S.), University of Virginia Health System, Charlottesville, VA
| | - Bradford B Worrall
- From the Department of Neurological Surgery (C.-J.C., N.I., T.J.B.), University of Virginia Health System, Charlottesville, VA; Department of Neurological Surgery (D.D., R.F.J.), University of Louisville School of Medicine, Louisville, KY; Clinical Trials Office (L.J.E., A.W.), University of Virginia School of Medicine; Department of Pharmacology (A.P.A.), University of Virginia Health System, Charlottesville, VA; Department of Public Health Sciences (S.J.R., B.B.W., A.M.S.), University of Virginia School of Medicine, Charlottesville, VA; Department of Neurosurgery (N.S.N.), Baptist Health, Jacksonville, FL; and Department of Neurology (B.B.W., K.C.J., A.M.S.), University of Virginia Health System, Charlottesville, VA
| | - Karen C Johnston
- From the Department of Neurological Surgery (C.-J.C., N.I., T.J.B.), University of Virginia Health System, Charlottesville, VA; Department of Neurological Surgery (D.D., R.F.J.), University of Louisville School of Medicine, Louisville, KY; Clinical Trials Office (L.J.E., A.W.), University of Virginia School of Medicine; Department of Pharmacology (A.P.A.), University of Virginia Health System, Charlottesville, VA; Department of Public Health Sciences (S.J.R., B.B.W., A.M.S.), University of Virginia School of Medicine, Charlottesville, VA; Department of Neurosurgery (N.S.N.), Baptist Health, Jacksonville, FL; and Department of Neurology (B.B.W., K.C.J., A.M.S.), University of Virginia Health System, Charlottesville, VA
| | - Andrew M Southerland
- From the Department of Neurological Surgery (C.-J.C., N.I., T.J.B.), University of Virginia Health System, Charlottesville, VA; Department of Neurological Surgery (D.D., R.F.J.), University of Louisville School of Medicine, Louisville, KY; Clinical Trials Office (L.J.E., A.W.), University of Virginia School of Medicine; Department of Pharmacology (A.P.A.), University of Virginia Health System, Charlottesville, VA; Department of Public Health Sciences (S.J.R., B.B.W., A.M.S.), University of Virginia School of Medicine, Charlottesville, VA; Department of Neurosurgery (N.S.N.), Baptist Health, Jacksonville, FL; and Department of Neurology (B.B.W., K.C.J., A.M.S.), University of Virginia Health System, Charlottesville, VA
| |
Collapse
|
16
|
|
17
|
Zhang J, Shi X, Hao N, Chen Z, Wei L, Tan L, Chen Y, Feng H, Chen Q, Zhu G. Simvastatin Reduces Neutrophils Infiltration Into Brain Parenchyma After Intracerebral Hemorrhage via Regulating Peripheral Neutrophils Apoptosis. Front Neurosci 2018; 12:977. [PMID: 30631264 PMCID: PMC6315192 DOI: 10.3389/fnins.2018.00977] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 12/06/2018] [Indexed: 01/13/2023] Open
Abstract
Statins, known for their lipid-lowering effects, also have immunomodulatory properties. This study aims to examine whether systematic simvastatin administration could decrease polymorphonuclear neutrophils (PMNs) infiltration into brain tissue, as well as alleviate neuroinflammation in a rat model of intracerebral hemorrhage (ICH). The ICH model was induced in adult male Sprague-Dawley rats by an injection of autologous blood. Animals randomly received simvastatin (i.p. 2 mg/kg) or vehicle daily from 5 days before ICH until sacrificed. Routine blood counts, brain water content, neurological scoring, immunofluorescence and RT-PCR were conducted to evaluate the anti-inflammatory effect of simvastatin following ICH. Furthermore, flow cytometric and western blotting analysis were implemented for elucidating the mechanisms involved in simvastatin-induced reduction of neutrophil brain-invading. Elevated PMNs count and neutrophil-to-lymphocyte ratio in circulation were detected in rat model of ICH, which was reversed by using simvastatin. Simvastatin effectively alleviated PMNs infiltration and proinflammatory factors release in perihematomal area, as well as attenuated ICH-induced brain edema and neurological deficits. Simvastatin significantly downregulated the expression of antiapoptotic protein-Mcl-1 while increased the level of proapoptotic protein-Bax and cleaved caspase 3 in PMNs. Simvastatin treatment significantly alleviated PMNs brain-infiltrating and subsequent neuroinflammatory reaction after ICH, in part by accelerating peripheral PMNs apoptosis through disorganized the expression of apoptotic related proteins. Our data provided new evidence for simvastatin application on patients with ICH.
Collapse
Affiliation(s)
- Jianbo Zhang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xia Shi
- Department of Nutrition, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Na Hao
- Department of Orthopedics, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Zhi Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Linjie Wei
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Liang Tan
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yujie Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qianwei Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Department of Neurosurgery, The 452 Hospital of Western Air Force, Chengdu, China
| | - Gang Zhu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
18
|
Chung CM, Lin MS, Liu CH, Lee TH, Chang ST, Yang TY, Pan KL, Lin YS. Discontinuing or continuing statin following intracerebral hemorrhage from the view of a national cohort study. Atherosclerosis 2018; 278:15-22. [PMID: 30236866 DOI: 10.1016/j.atherosclerosis.2018.08.049] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2018] [Revised: 07/19/2018] [Accepted: 08/30/2018] [Indexed: 01/17/2023]
Abstract
BACKGROUND AND AIMS Statins improve clinical outcomes in patients with ischemic stroke but there is no evidence of the effect of continuing long-term statin therapy in patients with intracerebral hemorrhage (ICH). The aim of this study was to evaluate the impact of continuing statin after ICH. METHODS Data on patients with ICH was retrieved from the National Health Insurance Research Database of Taiwan. The final population was separated into two groups according to those who continued and those who discontinued statin treatment. All-cause mortality and cardiovascular outcomes were analyzed after a 3 year follow-up after propensity score matching (PSM). RESULTS Of the 114,101 patients with ICH, who were initially enrolled, 2468 patients with dyslipidemia and ICH were included. After PSM, the benefit of statin therapy on mortality appeared from 1 year to the end of the 3-year follow-up period after discharge (statin group versus non-statin group: 4.9% vs.12.3% at 1 year (hazard ratio [HR], 0.38; 95% confidence interval [CI], 0.26-0.57) and 12.9% vs. 25.3% at the end of the 3 year follow-up period (HR, 0.45; 95% CI, 0.35-0.58). Compared with the patients using lipophilic statins, those using hydrophilic statins had a significantly lower incidence of all-cause mortality (HR = 0.65, 95% CI = 0.43-0.99). There were no differences between those prescribed moderate-intensity statins and those prescribed high-intensity statins in terms of stroke and all-cause mortality (HR = 0.76; 95% CI = 0.40-1.46). CONCLUSIONS There was a lower risk of all-cause mortality following ICH in patients who continued statin treatment compared with those without statin treatment, especially in those treated with hydrophilic statins.
Collapse
Affiliation(s)
- Chang-Min Chung
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan County, Taiwan; Division of Cardiology, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Ming-Shyan Lin
- Division of Cardiology, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Chi-Hung Liu
- Stroke Center and Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, Taiwan
| | - Tsong-Hai Lee
- Stroke Center and Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center, Taiwan
| | - Shih-Tai Chang
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan County, Taiwan; Division of Cardiology, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Teng-Yao Yang
- Division of Cardiology, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Kuo-Li Pan
- School of Traditional Chinese Medicine, College of Medicine, Chang Gung University, Taoyuan County, Taiwan; Division of Cardiology, Chang Gung Memorial Hospital, Chiayi, Taiwan
| | - Yu-Sheng Lin
- Division of Cardiology, Chang Gung Memorial Hospital, Chiayi, Taiwan.
| |
Collapse
|
19
|
Carter CJ. Genetic, Transcriptome, Proteomic, and Epidemiological Evidence for Blood-Brain Barrier Disruption and Polymicrobial Brain Invasion as Determinant Factors in Alzheimer's Disease. J Alzheimers Dis Rep 2017; 1:125-157. [PMID: 30480234 PMCID: PMC6159731 DOI: 10.3233/adr-170017] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Diverse pathogens are detected in Alzheimer's disease (AD) brains. A bioinformatics survey showed that AD genome-wide association study (GWAS) genes (localized in bone marrow, immune locations and microglia) relate to multiple host/pathogen interactomes (Candida albicans, Cryptococcus neoformans, Bornavirus, Borrelia burgdorferri, cytomegalovirus, Ebola virus, HSV-1, HERV-W, HIV-1, Epstein-Barr, hepatitis C, influenza, Chlamydia pneumoniae, Porphyrymonas gingivalis, Helicobacter pylori, Toxoplasma gondii, Trypanosoma cruzi). These interactomes also relate to the AD hippocampal transcriptome and to plaque or tangle proteins. Upregulated AD hippocampal genes match those upregulated by multiple bacteria, viruses, fungi, or protozoa in immunocompetent cells. AD genes are enriched in GWAS datasets reflecting pathogen diversity, suggesting selection for pathogen resistance, as supported by the old age of AD patients, implying resistance to earlier infections. APOE4 is concentrated in regions of high parasitic burden and protects against childhood tropical infections and hepatitis C. Immune/inflammatory gain of function applies to APOE4, CR1, and TREM2 variants. AD genes are also expressed in the blood-brain barrier (BBB), which is disrupted by AD risk factors (age, alcohol, aluminum, concussion, cerebral hypoperfusion, diabetes, homocysteine, hypercholesterolemia, hypertension, obesity, pesticides, pollution, physical inactivity, sleep disruption, smoking) and by pathogens, directly or via olfactory routes to basal-forebrain BBB control centers. The BBB benefits from statins, NSAIDs, estrogen, melatonin, memantine, and the Mediterranean diet. Polymicrobial involvement is supported by upregulation of bacterial, viral, and fungal sensors/defenders in the AD brain, blood, or cerebrospinal fluid. AD serum amyloid-β autoantibodies may attenuate its antimicrobial effects favoring microbial survival and cerebral invasion leading to activation of neurodestructive immune/inflammatory processes, which may also be augmented by age-related immunosenescence. AD may thus respond to antibiotic, antifungal, or antiviral therapy.
Collapse
|
20
|
Siddiqui FM, Langefeld CD, Moomaw CJ, Comeau ME, Sekar P, Rosand J, Kidwell CS, Martini S, Osborne JL, Stutzman S, Hall C, Woo D. Use of Statins and Outcomes in Intracerebral Hemorrhage Patients. Stroke 2017; 48:2098-2104. [PMID: 28663510 PMCID: PMC5659292 DOI: 10.1161/strokeaha.117.017358] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 05/05/2017] [Accepted: 06/02/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND AND PURPOSE Statin use may be associated with improved outcome in intracerebral hemorrhage patients. However, the topic remains controversial. Our analysis examined the effect of prior, continued, or new statin use on intracerebral hemorrhage outcomes using the ERICH (Ethnic/Racial Variations of Intracerebral Hemorrhage) data set. METHODS We analyzed ERICH (a multicenter study designed to examine ethnic variations in the risk, presentation, and outcomes of intracerebral hemorrhage) to explore the association of statin use and hematoma growth, mortality, and 3-month disability. We computed subset analyses with respect to 3 statin categories (prior, continued, or new use). RESULTS Two thousand four hundred and fifty-seven enrolled cases (mean age, 62 years; 42% females) had complete data on mortality and 3-month disability (modified Rankin Scale). Among those, 1093 cases were on statins (prior, n=268; continued, n=423; new, n=402). Overall, statin use was associated with reduced mortality and disability without any effect on hematoma growth. This association was primarily driven by continued/new statin use. A multivariate analysis adjusted for age and major predictors for poor outcome showed that continued/new statins users had good outcomes compared with prior users. However, statins may have been continued/started more frequently among less severe patients. When a propensity score was developed based on factors that could influence a physician's decision in prescribing statins and used as a covariate, continued/new statin use was no longer a significant predictor of good outcome. CONCLUSIONS Although statin use, especially continued/new use, was associated with improved intracerebral hemorrhage outcomes, this effect may merely reflect the physician's view of a patient's prognosis rather than a predictor of survival.
Collapse
Affiliation(s)
- Fazeel M Siddiqui
- From the Department of Neurology, Southern Illinois University School of Medicine, Springfield (F.M.S.); Center for Public Health Genomics and Department of Biostatistical Sciences, Wake Forest University, Winston-Salem, NC (C.D.L., M.E.C.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (C.J.M., P.S., J.L.O., D.W.); Center for Genomic Medicine, Massachusetts General Hospital, Boston (J.R.); Departments of Neurology and Medical Imaging, University of Arizona, Tucson (C.S.K.); Michael E. DeBakey VA Medical Center and Department of Neurology, Baylor College of Medicine, Houston, TX (S.M.); and Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas (S.S., C.H.).
| | - Carl D Langefeld
- From the Department of Neurology, Southern Illinois University School of Medicine, Springfield (F.M.S.); Center for Public Health Genomics and Department of Biostatistical Sciences, Wake Forest University, Winston-Salem, NC (C.D.L., M.E.C.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (C.J.M., P.S., J.L.O., D.W.); Center for Genomic Medicine, Massachusetts General Hospital, Boston (J.R.); Departments of Neurology and Medical Imaging, University of Arizona, Tucson (C.S.K.); Michael E. DeBakey VA Medical Center and Department of Neurology, Baylor College of Medicine, Houston, TX (S.M.); and Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas (S.S., C.H.)
| | - Charles J Moomaw
- From the Department of Neurology, Southern Illinois University School of Medicine, Springfield (F.M.S.); Center for Public Health Genomics and Department of Biostatistical Sciences, Wake Forest University, Winston-Salem, NC (C.D.L., M.E.C.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (C.J.M., P.S., J.L.O., D.W.); Center for Genomic Medicine, Massachusetts General Hospital, Boston (J.R.); Departments of Neurology and Medical Imaging, University of Arizona, Tucson (C.S.K.); Michael E. DeBakey VA Medical Center and Department of Neurology, Baylor College of Medicine, Houston, TX (S.M.); and Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas (S.S., C.H.)
| | - Mary E Comeau
- From the Department of Neurology, Southern Illinois University School of Medicine, Springfield (F.M.S.); Center for Public Health Genomics and Department of Biostatistical Sciences, Wake Forest University, Winston-Salem, NC (C.D.L., M.E.C.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (C.J.M., P.S., J.L.O., D.W.); Center for Genomic Medicine, Massachusetts General Hospital, Boston (J.R.); Departments of Neurology and Medical Imaging, University of Arizona, Tucson (C.S.K.); Michael E. DeBakey VA Medical Center and Department of Neurology, Baylor College of Medicine, Houston, TX (S.M.); and Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas (S.S., C.H.)
| | - Padmini Sekar
- From the Department of Neurology, Southern Illinois University School of Medicine, Springfield (F.M.S.); Center for Public Health Genomics and Department of Biostatistical Sciences, Wake Forest University, Winston-Salem, NC (C.D.L., M.E.C.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (C.J.M., P.S., J.L.O., D.W.); Center for Genomic Medicine, Massachusetts General Hospital, Boston (J.R.); Departments of Neurology and Medical Imaging, University of Arizona, Tucson (C.S.K.); Michael E. DeBakey VA Medical Center and Department of Neurology, Baylor College of Medicine, Houston, TX (S.M.); and Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas (S.S., C.H.)
| | - Jonathan Rosand
- From the Department of Neurology, Southern Illinois University School of Medicine, Springfield (F.M.S.); Center for Public Health Genomics and Department of Biostatistical Sciences, Wake Forest University, Winston-Salem, NC (C.D.L., M.E.C.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (C.J.M., P.S., J.L.O., D.W.); Center for Genomic Medicine, Massachusetts General Hospital, Boston (J.R.); Departments of Neurology and Medical Imaging, University of Arizona, Tucson (C.S.K.); Michael E. DeBakey VA Medical Center and Department of Neurology, Baylor College of Medicine, Houston, TX (S.M.); and Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas (S.S., C.H.)
| | - Chelsea S Kidwell
- From the Department of Neurology, Southern Illinois University School of Medicine, Springfield (F.M.S.); Center for Public Health Genomics and Department of Biostatistical Sciences, Wake Forest University, Winston-Salem, NC (C.D.L., M.E.C.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (C.J.M., P.S., J.L.O., D.W.); Center for Genomic Medicine, Massachusetts General Hospital, Boston (J.R.); Departments of Neurology and Medical Imaging, University of Arizona, Tucson (C.S.K.); Michael E. DeBakey VA Medical Center and Department of Neurology, Baylor College of Medicine, Houston, TX (S.M.); and Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas (S.S., C.H.)
| | - Sharyl Martini
- From the Department of Neurology, Southern Illinois University School of Medicine, Springfield (F.M.S.); Center for Public Health Genomics and Department of Biostatistical Sciences, Wake Forest University, Winston-Salem, NC (C.D.L., M.E.C.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (C.J.M., P.S., J.L.O., D.W.); Center for Genomic Medicine, Massachusetts General Hospital, Boston (J.R.); Departments of Neurology and Medical Imaging, University of Arizona, Tucson (C.S.K.); Michael E. DeBakey VA Medical Center and Department of Neurology, Baylor College of Medicine, Houston, TX (S.M.); and Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas (S.S., C.H.)
| | - Jennifer L Osborne
- From the Department of Neurology, Southern Illinois University School of Medicine, Springfield (F.M.S.); Center for Public Health Genomics and Department of Biostatistical Sciences, Wake Forest University, Winston-Salem, NC (C.D.L., M.E.C.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (C.J.M., P.S., J.L.O., D.W.); Center for Genomic Medicine, Massachusetts General Hospital, Boston (J.R.); Departments of Neurology and Medical Imaging, University of Arizona, Tucson (C.S.K.); Michael E. DeBakey VA Medical Center and Department of Neurology, Baylor College of Medicine, Houston, TX (S.M.); and Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas (S.S., C.H.)
| | - Sonja Stutzman
- From the Department of Neurology, Southern Illinois University School of Medicine, Springfield (F.M.S.); Center for Public Health Genomics and Department of Biostatistical Sciences, Wake Forest University, Winston-Salem, NC (C.D.L., M.E.C.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (C.J.M., P.S., J.L.O., D.W.); Center for Genomic Medicine, Massachusetts General Hospital, Boston (J.R.); Departments of Neurology and Medical Imaging, University of Arizona, Tucson (C.S.K.); Michael E. DeBakey VA Medical Center and Department of Neurology, Baylor College of Medicine, Houston, TX (S.M.); and Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas (S.S., C.H.)
| | - Christiana Hall
- From the Department of Neurology, Southern Illinois University School of Medicine, Springfield (F.M.S.); Center for Public Health Genomics and Department of Biostatistical Sciences, Wake Forest University, Winston-Salem, NC (C.D.L., M.E.C.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (C.J.M., P.S., J.L.O., D.W.); Center for Genomic Medicine, Massachusetts General Hospital, Boston (J.R.); Departments of Neurology and Medical Imaging, University of Arizona, Tucson (C.S.K.); Michael E. DeBakey VA Medical Center and Department of Neurology, Baylor College of Medicine, Houston, TX (S.M.); and Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas (S.S., C.H.)
| | - Daniel Woo
- From the Department of Neurology, Southern Illinois University School of Medicine, Springfield (F.M.S.); Center for Public Health Genomics and Department of Biostatistical Sciences, Wake Forest University, Winston-Salem, NC (C.D.L., M.E.C.); Department of Neurology and Rehabilitation Medicine, University of Cincinnati, OH (C.J.M., P.S., J.L.O., D.W.); Center for Genomic Medicine, Massachusetts General Hospital, Boston (J.R.); Departments of Neurology and Medical Imaging, University of Arizona, Tucson (C.S.K.); Michael E. DeBakey VA Medical Center and Department of Neurology, Baylor College of Medicine, Houston, TX (S.M.); and Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas (S.S., C.H.)
| |
Collapse
|
21
|
Chen Q, Shi X, Tan Q, Feng Z, Wang Y, Yuan Q, Tao Y, Zhang J, Tan L, Zhu G, Feng H, Chen Z. Simvastatin Promotes Hematoma Absorption and Reduces Hydrocephalus Following Intraventricular Hemorrhage in Part by Upregulating CD36. Transl Stroke Res 2017; 8:362-373. [PMID: 28102508 DOI: 10.1007/s12975-017-0521-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 01/08/2017] [Accepted: 01/11/2017] [Indexed: 10/20/2022]
Abstract
We previously found that hematoma worsens hydrocephalus after intraventricular hemorrhage (IVH) via increasing iron deposition and aggravating ependymal cilia injury; therefore, promoting hematoma absorption may be a promising strategy for IVH. Recently, some investigations imply that simvastatin has the ability of accelerating hematoma absorption. Thus, this study was designed to examine the efficacy of simvastatin for IVH in rats. Intracerebral hemorrhage with ventricular extension was induced in adult male Sprague-Dawley rats after autologous blood injection. Simvastatin or vehicle was administered orally at 1 day after IVH and then daily for 1 week. MRI studies were performed to measure the volumes of intracranial hematoma and lateral ventricle at days 1, 3, 7, 14, and 28 after IVH. Motor and neurocognitive functions were assessed at days 1 to 7 and 23 to 28, respectively. Iron deposition, iron-related protein expression, ependymal damage, and histology were detected at day 28. Expression of CD36 scavenger receptor (facilitating phagocytosis) was examined at day 3 after IVH using western blotting and immunofluorescence. Simvastatin significantly increased hematoma absorption ratio, reduced ventricular volume, and attenuated neurological dysfunction post-IVH. In addition, less iron accumulation and more cilia survival was observed in the simvastatin group when compared with the control. What's more, higher expression of CD36 was detected around the hematoma after simvastatin administration. Simvastatin significantly enhanced brain hematoma absorption, alleviated hydrocephalus, and improved neurological recovery after experimental IVH, which may in part by upregulating CD36 expression. Our data suggest that early simvastatin use may be a novel therapy for IVH patients.
Collapse
Affiliation(s)
- Qianwei Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, No. 30, Gaotanyan Street, Chongqing, 400038, China
| | - Xia Shi
- Department of Nutrition, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Qiang Tan
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, No. 30, Gaotanyan Street, Chongqing, 400038, China
| | - Zhou Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, No. 30, Gaotanyan Street, Chongqing, 400038, China
| | - Yuelong Wang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, No. 30, Gaotanyan Street, Chongqing, 400038, China
| | - Qiaoying Yuan
- Department of Nutrition, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Yihao Tao
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, No. 30, Gaotanyan Street, Chongqing, 400038, China
| | - Jianbo Zhang
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, No. 30, Gaotanyan Street, Chongqing, 400038, China
| | - Liang Tan
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, No. 30, Gaotanyan Street, Chongqing, 400038, China
| | - Gang Zhu
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, No. 30, Gaotanyan Street, Chongqing, 400038, China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, No. 30, Gaotanyan Street, Chongqing, 400038, China.
| | - Zhi Chen
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University, No. 30, Gaotanyan Street, Chongqing, 400038, China.
| |
Collapse
|
22
|
Multimodality MRI assessment of grey and white matter injury and blood-brain barrier disruption after intracerebral haemorrhage in mice. Sci Rep 2017; 7:40358. [PMID: 28084426 PMCID: PMC5234017 DOI: 10.1038/srep40358] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 12/05/2016] [Indexed: 11/26/2022] Open
Abstract
In this study, we examined injury progression after intracerebral haemorrhage (ICH) induced by collagenase in mice using a preclinical 11.7 Tesla MRI system. On T2-weighted MRI, lesion and striatal volumes were increased on day 3 and then decreased from days 7 to 28. On day 3, with an increase in striatal water content, vasogenic oedema in the perihaematomal region presented as increased T2 and increased apparent diffusion coefficient (ADC) signal. With a synchronous change in T2 and ADC signals, microglial activation peaked on day 3 in the same region and decreased over time. Iron deposition appeared on day 3 around the haematoma border but did not change synchronously with ADC signals. Vascular permeability measured by Evans blue extravasation on days 1, 3, and 7 correlated with the T1-gadolinium results, both of which peaked on day 3. On diffusion tensor imaging, white matter injury was prominent in the corpus callosum and internal capsule on day 3 and then partially recovered over time. Our results indicate that the evolution of grey/white matter injury and blood-brain barrier disruption after ICH can be assessed with multimodal MRI, and that perihaematomal vasogenic oedema might be attributable to microglial activation, iron deposition, and blood-brain barrier breakdown.
Collapse
|
23
|
|
24
|
Mittal MK, LacKamp A. Intracerebral Hemorrhage: Perihemorrhagic Edema and Secondary Hematoma Expansion: From Bench Work to Ongoing Controversies. Front Neurol 2016; 7:210. [PMID: 27917153 PMCID: PMC5116572 DOI: 10.3389/fneur.2016.00210] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 11/08/2016] [Indexed: 12/30/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a medical emergency, which often leads to severe disability and death. ICH-related poor outcomes are due to primary injury causing structural damage and mass effect and secondary injury in the perihemorrhagic region over several days to weeks. Secondary injury after ICH can be due to hematoma expansion (HE) or a consequence of repair pathway along the continuum of neuroinflammation, neuronal death, and perihemorrhagic edema (PHE). This review article is focused on PHE and HE and will cover the animal studies, related human studies, and clinical trials relating to these mechanisms of secondary brain injury in ICH patients.
Collapse
Affiliation(s)
- Manoj K Mittal
- Department of Neurology, University of Kansas Medical Center , Kansas City, KS , USA
| | - Aaron LacKamp
- Department of Anesthesiology, University of Kansas Medical Center , Kansas City, KS , USA
| |
Collapse
|
25
|
Tai SY, Lin FC, Lee CY, Chang CJ, Wu MT, Chien CY. Statin use after intracerebral hemorrhage: a 10-year nationwide cohort study. Brain Behav 2016; 6:e00487. [PMID: 27247857 PMCID: PMC4867570 DOI: 10.1002/brb3.487] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2016] [Revised: 03/28/2016] [Accepted: 04/08/2016] [Indexed: 11/06/2022] Open
Abstract
INTRODUCTION Although statin therapy is beneficial to patients with ischemic stroke, statin use, and intracerebral hemorrhage (ICH) remain a concern. ICH survivors commonly have comorbid cardiovascular risk factors that would otherwise warrant cholesterol-lowering medication, thus emphasizing the importance of assessing the characteristics of statin therapy in this population. METHODS We performed a cohort study by using 10 years of data collected from the National Health Insurance Research Database in Taiwan. We enrolled 726 patients admitted for newly diagnosed ICH from January 1, 2001 to December 31, 2010. The patients were categorized into high- (92), moderate- (545), and low-intensity (89) statin groups, and into hydrophilic (295) and lipophilic (431) statin groups. The composite outcomes included all-cause mortality, recurrent ICH, ischemic stroke, transient ischemic attack, and acute coronary events. RESULTS The patients in the low-intensity group did not differ significantly from the patients in the high-intensity group in risk of all-cause mortality (adjusted hazard ratio [aHR] = 0.65, 95% confidence interval [CI] = 0.28-1.55) and recurrent ICH (aHR = 0.66, 95% CI = 0.30-1.44). In contrast, the patients in the hydrophilic group had a significantly lower risk of recurrent ICH than did those in the lipophilic group (aHR = 0.69, 95% CI = 0.48-0.99). We determined no significant differences in other composite endpoints between hydrophilic and lipophilic statin use. CONCLUSION Hydrophilic statin therapy is associated with a reduced risk of recurrent ICH in post-ICH patients. The intensity of statin use had no significant effect on recurrent ICH or other components of the composite outcome. Additional studies are required to clarify the biological mechanisms underlying these observations.
Collapse
Affiliation(s)
- Shu-Yu Tai
- Department of Family Medicine School of Medicine College of Medicine Kaohsiung Medical University Kaohsiung Taiwan; Department of Family Medicine Kaohsiung Medical University Hospital Kaohsiung Medical University Kaohsiung Taiwan; Department of Family Medicine Kaohsiung Municipal Ta-Tung Hospital Kaohsiung Medical University Hospital Kaohsiung Medical University Kaohsiung Taiwan
| | - Feng-Cheng Lin
- Department of Neurology Kaohsiung Medical University Hospital Kaohsiung Taiwan; Department of Neurology Pingtung Hospital Ministry of Health and Welfare Pingtung Taiwan
| | - Chung-Yin Lee
- Department of Family Medicine Kaohsiung Medical University Hospital Kaohsiung Medical University Kaohsiung Taiwan
| | - Chai-Jan Chang
- Department of Family Medicine School of Medicine College of Medicine Kaohsiung Medical University Kaohsiung Taiwan; Department of Family Medicine Kaohsiung Medical University Hospital Kaohsiung Medical University Kaohsiung Taiwan; Department of Family Medicine Kaohsiung Municipal Hsiao-Kang Hospital Kaohsiung Medical University Kaohsiung Taiwan
| | - Ming-Tsang Wu
- Department of Family Medicine School of Medicine College of Medicine Kaohsiung Medical University Kaohsiung Taiwan; Department of Public Health Kaohsiung Medical University Kaohsiung Taiwan; Center of Environmental and Occupational Medicine Kaohsiung Municipal Hsiao-Kang Hospital Kaohsiung Medical University Kaohsiung Taiwan
| | - Chen-Yu Chien
- Department of Otorhinolaryngology School of Medicine College of Medicine Kaohsiung Medical University Kaohsiung Taiwan; Department of Otorhinolaryngology Kaohsiung Medical University Hospital Kaohsiung Medical University Kaohsiung Taiwan; Department of Otorhinolaryngology Kaohsiung Municipal Hsiao-Kang Hospital Kaohsiung Medical University Kaohsiung Taiwan
| |
Collapse
|
26
|
Knight RA, Nagaraja TN, Li L, Jiang Q, Tundo K, Chopp M, Seyfried DM. A Prospective Safety Trial of Atorvastatin Treatment to Assess Rebleeding after Spontaneous Intracerebral Hemorrhage: A Serial MRI Investigation. AUSTIN JOURNAL OF CEREBROVASCULAR DISEASE & STROKE 2016; 3:1043. [PMID: 28529979 PMCID: PMC5436718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
AIM This study was designed to determine any rebleeding after atorvastatin treatment following spontaneous intracerebral hemorrhage (ICH) in a prospective safety trial. PATIENTS Atorvastatin (80 mg/day) therapy was initiated in 6 patients with primary ICH with admission Glasgow Coma Score (GCS) >5 within 24 hours of ictus and continued for 7 days, with the dose tapered and treatment terminated over the next 5 days. Patients were studied longitudinally by multiparametric magnetic resonance imaging (MRI) at three time points: acute (3 to 5 days), subacute (4 to 6 weeks) and chronic (3 to 4 months). Imaging sequences included T1, T2-weighted imaging (T2WI), diffusion tensor imaging (DTI) and contrast-enhanced MRI measures of cerebral perfusion, blood volume and blood-brain barrier (BBB) permeability. Susceptibility weighted imaging (SWI) was used to identify primary ICH and to check for secondary rebleeding. Final outcome was assessed using Glasgow Outcome Score (GOS) at 3-4 months. RESULTS Mean admission GCS was 13.2±4.0 and mean GOS at 3 months was 4.5±0.6. Hemorrhagic lesions were segmented into core and rim areas. Mean lesion volumes decreased significantly between the acute and chronic study time points (p=0.008). Average ipsilateral hemispheric tissue loss at 3 to 4 months was 11.4±4.6 cm3. MRI showed acutely reduced CBF (p=0.004) and CBV (p=0.002) in the rim, followed by steady normalization. Apparent diffusion coefficient of water (ADC) in the rim demonstrated no alterations at any of the time points (p>0.2). The T2 values were significantly elevated in the rim acutely (p=0.02), but later returned to baseline. The ICH core showed sustained low CBF and CBV values concurrent with a small reduction in ADC acutely, but significant ADC elevation at the end suggestive of irreversible injury. CONCLUSION Despite the presence of a small, probably permanent, cerebral lesion in the ICH core, no patients exhibited post-treatment rebleeding. These data suggest that larger, Phase 2 trials are warranted to establish long term clinical safety of atorvastatin in spontaneous ICH.
Collapse
Affiliation(s)
- R A Knight
- Department of Neurology, Henry Ford Hospital, USA
- Department of Physics, Oakland University, Rochester, USA
| | - T N Nagaraja
- Department of Neurosurgery, Henry Ford Hospital, USA
| | - L Li
- Department of Neurology, Henry Ford Hospital, USA
| | - Q Jiang
- Department of Neurology, Henry Ford Hospital, USA
| | - K Tundo
- Department of Neurosurgery, Henry Ford Hospital, USA
| | - M Chopp
- Department of Neurology, Henry Ford Hospital, USA
| | - D M Seyfried
- Department of Neurosurgery, Henry Ford Hospital, USA
| |
Collapse
|
27
|
Griffin JM, Kho D, Graham ES, Nicholson LFB, O’Carroll SJ. Statins Inhibit Fibrillary β-Amyloid Induced Inflammation in a Model of the Human Blood Brain Barrier. PLoS One 2016; 11:e0157483. [PMID: 27309956 PMCID: PMC4911157 DOI: 10.1371/journal.pone.0157483] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Accepted: 05/31/2016] [Indexed: 02/03/2023] Open
Abstract
Background Astrocytes and cerebral endothelial cells are important components of the blood-brain barrier (BBB). Disruption to this barrier through inflammation is a major contributor to Alzheimer’s disease (AD) pathology. The amyloid beta (Aβ) protein is known to exist in several forms and is a key modulator of AD that is known to cause inflammation and changes to BBB function. While one of these forms, fibrillary Aβ (fAβ), is known to cause endothelial cell death at the BBB, no studies have looked specifically at its role on inflammation in a model of the human BBB. Aims To determine if fAβ is inflammatory to the human BBB. As statins have been shown to be anti-inflammatory and protective in AD, we also tested if these could inhibit the inflammatory effect of fAβ. Methods Using cultured cerebral endothelial cells and astrocytes we determined changes in cytokine release, cell toxicity and barrier function in response to fibrillary β-amyloid1–42 (fAβ1–42) alone and in combination with statins. Results fAβ1–42 induced inflammatory cytokine release from endothelial cells in the absence of cell toxicity. It also induced astrocyte cytokine release and cell death and caused a loss of barrier integrity. Statin treatment inhibited all of these effects. Conclusions We conclude that fAβ1–42 has both inflammatory and cytotoxic effects on the BBB and the protective effect of statins in AD may in part be through inhibiting these effects.
Collapse
Affiliation(s)
- Jarred M. Griffin
- Centre for Brain Research, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Department of Anatomy and Medical Imaging, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Dan Kho
- Centre for Brain Research, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - E. Scott Graham
- Centre for Brain Research, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Department of Pharmacology and Clinical Pharmacology, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Louise F. B. Nicholson
- Centre for Brain Research, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Department of Anatomy and Medical Imaging, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Simon J. O’Carroll
- Centre for Brain Research, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- Department of Anatomy and Medical Imaging, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
- * E-mail:
| |
Collapse
|
28
|
Askenase MH, Sansing LH. Stages of the Inflammatory Response in Pathology and Tissue Repair after Intracerebral Hemorrhage. Semin Neurol 2016; 36:288-97. [PMID: 27214704 DOI: 10.1055/s-0036-1582132] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Intracerebral hemorrhage (ICH) is a major health concern, with high rates of mortality and morbidity and no highly effective clinical interventions. Basic research in animal models of ICH has provided insight into its complex pathology, in particular revealing the role of inflammation in driving neuronal death and neurologic deficits after hemorrhage. The response to ICH occurs in four distinct phases: (1) initial tissue damage and local activation of inflammatory factors, (2) inflammation-driven breakdown of the blood-brain barrier, (3) recruitment of circulating inflammatory cells and subsequent secondary immunopathology, and (4) engagement of tissue repair responses that promote tissue repair and restoration of neurologic function. The development of CNS inflammation occurs over many days after initial hemorrhage and thus may represent an ideal target for treatment of the disease, but further research is required to identify the mechanisms that promote engagement of inflammatory versus anti-inflammatory pathways. In this review, the authors examine how experimental models of ICH have uncovered critical mediators of pathology in each of the four stages of the inflammatory response, and focus on the role of the immune system in these processes.
Collapse
Affiliation(s)
- Michael H Askenase
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut
| | - Lauren H Sansing
- Department of Neurology, Yale School of Medicine, New Haven, Connecticut
| |
Collapse
|
29
|
Abstract
While statins play an indisputable role in primary and secondary prevention of ischemic cardiovascular and cerebrovascular disease, a concern exists regarding a possible association between low lipoprotein levels and statin use on the risk of intracerebral hemorrhage (ICH). While these data may incline physicians to discontinue statins after ICH, an increasing amount of preclinical and clinical evidence suggests that statins might have a beneficial effect on outcome and recovery in this context that goes beyond lipid lowering effects. Different etiologies of ICH and the related risk of recurrence should also be taken into account when deciding about statin use/avoidance in patients with high risk of ICH. The problem is compounded by paucity of data from randomized controlled trials and well-designed prospective observational studies. This review will discuss the existing evidence on potential interactions between statins and risk of ICH as well as outcomes in order to provide practical recommendations for clinical decision-making.
Collapse
|
30
|
Ma Y, Li Z, Chen L, Li X. Blood lipid levels, statin therapy and the risk of intracerebral hemorrhage. Lipids Health Dis 2016; 15:43. [PMID: 26932585 PMCID: PMC4774119 DOI: 10.1186/s12944-016-0213-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2015] [Accepted: 02/24/2016] [Indexed: 02/08/2023] Open
Abstract
Dyslipidemia has been proven to play an important role in the occurrence and development of the ischemic stroke and lipid-lowering therapy could significantly decrease the risk of the ischemic stroke. However, the association between lipid levels, lipid-lowering therapy and the risk of intracerebral hemorrhage (ICH) is not clear. Studies have shown that low serum levels of total cholesterol might be associated with increasing risk of ICH, whereas the SPARCL study, a large prospective, randomized, placebo-controlled trial, demonstrated an increased risk of hemorrhagic stroke during high-dose statin therapy among the patients with previous stroke. The relationship between lipid-lowering therapy and ICH has become a hot topic in the recent years. We searched PubMed for articles published in English to review the existing evidence on the association of lipid levels, statin therapy and risk of ICH as well as the underlying mechanisms in order to provide practical recommendations for clinical decision-making and a foundation for further researches.
Collapse
Affiliation(s)
- Yingxu Ma
- Department of Cardiology, The Second Xiangya Hospital, Central South University, #139 Middle Renmin Road, Changsha, Hunan, 410011, PR China.,The Eight-Year Clinical Medicine of Grade 2012, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, PR China
| | - Zhaokai Li
- Department of Cardiology, The Second Xiangya Hospital, Central South University, #139 Middle Renmin Road, Changsha, Hunan, 410011, PR China.,The Eight-Year Clinical Medicine of Grade 2012, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, PR China
| | - Liang Chen
- Department of Cardiology, The Second Xiangya Hospital, Central South University, #139 Middle Renmin Road, Changsha, Hunan, 410011, PR China.,The Eight-Year Clinical Medicine of Grade 2012, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410013, PR China
| | - Xiangping Li
- Department of Cardiology, The Second Xiangya Hospital, Central South University, #139 Middle Renmin Road, Changsha, Hunan, 410011, PR China.
| |
Collapse
|
31
|
Tapia Pérez JH, Yildiz OC, Schneider T, Nimsky C. Meta-analysis of Statin Use for the Acute Therapy of Spontaneous Intracerebral Hemorrhage. J Stroke Cerebrovasc Dis 2015; 24:2521-6. [PMID: 26387046 DOI: 10.1016/j.jstrokecerebrovasdis.2015.06.036] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2015] [Accepted: 06/27/2015] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Growing evidence demonstrates the neuroprotective effects of statins, and the risk to develop an intracerebral hemorrhage (ICH) using statins has been refuted. However, some controversy remains regarding their role in the acute phase after ICH onset. Therefore, we performed a systematic review to investigate this issue. METHODS We searched in MEDLINE, Web of Knowledge, and Scopus databases for studies examining the outcome in patients with spontaneous ICH and statin use. The analysis was performed for short-term (≤3 months) and long-term outcome (≥6 months) and a further subanalysis considered studies seeking for the effects of the discontinuation of statin after ICH onset. A random-effect model was applied, and country was used as a cofactor for meta-regression; odds ratios (ORs) with 95% confidence intervals (CIs) are offered. RESULTS A total of 17 studies were included, only 1 pseudo cohort trial assessed the new use of statin after ICH onset and 3 studies evaluated the suspension of statin after ICH onset, the rest of the studies focused on the effect of the regular use of statin before ICH onset. The number of patients with an ICH exposed and not exposed to statins were 3455 and 11,821, respectively. The absolute short-term mortality was 27.3% in statin users and 33% in nonusers that represented a significant risk reduction of mortality (OR, .73; 95% CI, .54-.97). For long-term mortality, the effect was less evident (OR, .71; 95% CI, .43-1.15). The analysis of the 3 studies assessing the discontinuation of statins suggested a reduction of mortality risk by continuing statin (OR, .14; 95% CI, .1-.20). CONCLUSIONS The current evidence suggests that continuing statin after ICH onset might be highly related to improvement of the outcome of patients with ICH. Despite this strong suggestion, randomized controlled trials should be performed to further investigate this association.
Collapse
Affiliation(s)
| | - O Can Yildiz
- Klinik für Neurochirurgie, Universitätsklinik Otto von Guericke Magdeburg, Magdeburg, Germany
| | - Thomas Schneider
- Klinik für Neurochirurgie, Universitätsklinik Otto von Guericke Magdeburg, Magdeburg, Germany
| | - Christopher Nimsky
- Klinik für Neurochirurgie, Philipps-University Marburg, Marburg, Germany
| |
Collapse
|
32
|
Ahmed LA, Darwish HA, Abdelsalam RM, Amin HA. Role of Rho Kinase Inhibition in the Protective Effect of Fasudil and Simvastatin Against 3-Nitropropionic Acid-Induced Striatal Neurodegeneration and Mitochondrial Dysfunction in Rats. Mol Neurobiol 2015; 53:3927-3938. [DOI: 10.1007/s12035-015-9303-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Accepted: 06/10/2015] [Indexed: 10/23/2022]
|
33
|
Gioia LC, Kate M, McCourt R, Gould B, Coutts SB, Dowlatshahi D, Asdaghi N, Jeerakathil T, Hill MD, Demchuk AM, Buck B, Emery D, Shuaib A, Butcher K. Perihematoma cerebral blood flow is unaffected by statin use in acute intracerebral hemorrhage patients. J Cereb Blood Flow Metab 2015; 35:1175-80. [PMID: 25757757 PMCID: PMC4640272 DOI: 10.1038/jcbfm.2015.36] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2014] [Revised: 01/19/2015] [Accepted: 01/30/2015] [Indexed: 11/09/2022]
Abstract
Statin therapy has been associated with improved cerebral blood flow (CBF) and decreased perihematoma edema in animal models of intracerebral hemorrhage (ICH). We aimed to assess the relationship between statin use and cerebral hemodynamics in ICH patients. A post hoc analysis of 73 ICH patients enrolled in the Intracerebral Hemorrhage Acutely Decreasing Arterial Pressure Trial (ICH ADAPT). Patients presenting <24 hours from ICH onset were randomized to a systolic blood pressure target <150 or <180 mm Hg with computed tomography perfusion imaging 2 hours after randomization. Cerebral blood flow maps were calculated. Hematoma and edema volumes were measured planimetrically. Regression models were used to assess the relationship between statin use, perihematoma edema and cerebral hemodynamics. Fourteen patients (19%) were taking statins at the time of ICH. Statin-treated patients had similar median (IQR Q25 to 75) hematoma volumes (21.1 (9.5 to 38.3) mL versus 14.5 (5.6 to 27.7) mL, P=0.25), but larger median (IQR Q25 to 75) perihematoma edema volumes (2.9 (1.7 to 9.0) mL versus 2.2 (0.8 to 3.5) mL, P=0.02) compared with nontreated patients. Perihematoma and ipsilateral hemispheric CBF were similar in both groups. A multivariate linear regression model revealed that statin use and hematoma volumes were independent predictors of acute edema volumes. Statin use does not affect CBF in ICH patients. Statin use, along with hematoma volume, are independently associated with increased perihematoma edema volume.
Collapse
Affiliation(s)
- Laura C Gioia
- Division of Neurology, 2E3 WMC Health Sciences Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Mahesh Kate
- Division of Neurology, 2E3 WMC Health Sciences Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Rebecca McCourt
- Division of Neurology, 2E3 WMC Health Sciences Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Bronwen Gould
- Division of Neurology, 2E3 WMC Health Sciences Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Shelagh B Coutts
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | | | - Negar Asdaghi
- Division of Neurology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Thomas Jeerakathil
- Division of Neurology, 2E3 WMC Health Sciences Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Michael D Hill
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Andrew M Demchuk
- Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada
| | - Brian Buck
- Division of Neurology, 2E3 WMC Health Sciences Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Derek Emery
- Department of Diagnostic Imaging, University of Alberta, Edmonton, Alberta, Canada
| | - Ashfaq Shuaib
- Division of Neurology, 2E3 WMC Health Sciences Centre, University of Alberta, Edmonton, Alberta, Canada
| | - Kenneth Butcher
- Division of Neurology, 2E3 WMC Health Sciences Centre, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
34
|
Kotze MJ, Lückhoff HK, Brand T, Pretorius J, van Rensburg SJ. Apolipoprotein E ε-4 as a genetic determinant of Alzheimer's disease heterogeneity. Degener Neurol Neuromuscul Dis 2015; 5:9-18. [PMID: 32669910 PMCID: PMC7337157 DOI: 10.2147/dnnd.s41721] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 04/23/2015] [Indexed: 12/30/2022] Open
Abstract
Alzheimer's disease (AD) displays a high degree of heterogeneity in terms of its etiology, presentation, prognosis, and treatment response. This can partly be explained by high-penetrance mutations in the amyloid precursor protein, presenilin 1 and presenilin 2 genes causing amyloid beta aggregation, which is a major pathogenic mechanism in the development of early-onset AD in a small subgroup of patients. Late-onset AD is considered a polygenic disorder in which cumulative risk resulting from interaction with modifiable environmental risk factors may be responsible for the majority of cases. The ε-4 allele of the apolipoprotein E (APOE) gene has emerged as the most significant genetic risk factor for late-onset AD, influencing nearly every pathogenic domain affected in AD. It is a major risk factor for cerebral amyloid angiopathy, recognized as a common pathological finding in an AD subtype associated with white matter dysfunction. The APOE ε-4 allele is also a known risk factor for ischemic stroke, which can result in vascular dementia or contribute to subcortical vascular dysfunction. In this review, we evaluate the clinical relevance of APOE genotyping in relation to cholesterol metabolism and available evidence on risk reduction strategies applicable to AD.
Collapse
Affiliation(s)
- MJ Kotze
- Division of Anatomical Pathology, Department of Pathology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - HK Lückhoff
- Division of Anatomical Pathology, Department of Pathology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - T Brand
- Division of Anatomical Pathology, Department of Pathology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - J Pretorius
- Division of Anatomical Pathology, Department of Pathology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - SJ van Rensburg
- Division of Chemical Pathology, Department of Pathology, Faculty of Medicine and Health Sciences, Stellenbosch University and the National Health Laboratory Service, Tygerberg Hospital, Tygerberg, South Africa
| |
Collapse
|
35
|
Targeting transporters: promoting blood-brain barrier repair in response to oxidative stress injury. Brain Res 2015; 1623:39-52. [PMID: 25796436 DOI: 10.1016/j.brainres.2015.03.018] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Revised: 03/09/2015] [Accepted: 03/10/2015] [Indexed: 01/06/2023]
Abstract
The blood-brain barrier (BBB) is a physical and biochemical barrier that precisely regulates the ability of endogenous and exogenous substances to accumulate within brain tissue. It possesses structural and biochemical features (i.e., tight junction and adherens junction protein complexes, influx and efflux transporters) that work in concert to control solute permeation. Oxidative stress, a critical component of several diseases including cerebral hypoxia/ischemia and peripheral inflammatory pain, can cause considerable injury to the BBB and lead to significant CNS pathology. This suggests a critical need for novel therapeutic approaches that can protect the BBB in diseases with an oxidative stress component. Recent studies have identified molecular targets (i.e., putative membrane transporters, intracellular signaling systems) that can be exploited for optimization of endothelial drug delivery or for control of transport of endogenous substrates such as the antioxidant glutathione (GSH). In particular, targeting transporters offers a unique approach to protect BBB integrity by promoting repair of cell-cell interactions at the level of the brain microvascular endothelium. This review summarizes current knowledge in this area and emphasizes those targets that present considerable opportunity for providing BBB protection and/or promoting BBB repair in the setting of oxidative stress. This article is part of a Special Issue entitled SI: Cell Interactions In Stroke.
Collapse
|
36
|
Urday S, Kimberly WT, Beslow LA, Vortmeyer AO, Selim MH, Rosand J, Simard JM, Sheth KN. Targeting secondary injury in intracerebral haemorrhage--perihaematomal oedema. Nat Rev Neurol 2015; 11:111-22. [PMID: 25623787 DOI: 10.1038/nrneurol.2014.264] [Citation(s) in RCA: 206] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Perihaematomal oedema (PHO) is an important pathophysiological marker of secondary injury in intracerebral haemorrhage (ICH). In this Review, we describe a novel method to conceptualize PHO formation within the framework of Starling's principle of movement of fluid across a capillary wall. We consider progression of PHO through three stages, characterized by ionic oedema (stage 1) and progressive vasogenic oedema (stages 2 and 3). In this context, possible modifiers of PHO volume and their value in identifying patients who would benefit from therapies that target secondary injury are discussed; the practicalities of using neuroimaging to measure PHO volume are also considered. We examine whether PHO can be used as a predictor of neurological outcome following ICH, and we provide an overview of emerging therapies. Our discussion emphasizes that PHO has clinical relevance both as a therapeutic target, owing to its augmentation of the mass effect of a haemorrhage, and as a surrogate marker for novel interventions that target secondary injury.
Collapse
Affiliation(s)
- Sebastian Urday
- Department of Neurology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| | - W Taylor Kimberly
- Department of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
| | - Lauren A Beslow
- Department of Neurology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| | - Alexander O Vortmeyer
- Department of Pathology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| | - Magdy H Selim
- Department of Neurology, Beth Israel Deaconess Medical Centre, 330 Brookline Avenue, Boston, MA 02215, USA
| | - Jonathan Rosand
- Department of Neurology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, USA
| | - J Marc Simard
- Department of Neurosurgery, University of Maryland School of Medicine, 655 West Baltimore Street, Baltimore, MD 21201-1559, USA
| | - Kevin N Sheth
- Department of Neurology, Yale University School of Medicine, 333 Cedar Street, New Haven, CT 06510, USA
| |
Collapse
|
37
|
Jiao X, He P, Li Y, Fan Z, Si M, Xie Q, Chang X, Huang D. The Role of Circulating Tight Junction Proteins in Evaluating Blood Brain Barrier Disruption following Intracranial Hemorrhage. DISEASE MARKERS 2015; 2015:860120. [PMID: 26586924 PMCID: PMC4637473 DOI: 10.1155/2015/860120] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/14/2015] [Accepted: 09/16/2015] [Indexed: 02/05/2023]
Abstract
Brain injury after intracranial hemorrhage (ICH) results in significant morbidity and mortality. Blood brain barrier (BBB) disruption is a hallmark of ICH-induced brain injury; however, data mirroring BBB disruption in human ICH are scarce. The aim of this study was to assess the significance of circulating biomarkers in evaluating BBB disruption after ICH. Twenty-two patients with ICH were recruited in this study. Concentrations of the tight junction proteins (TJs) Claudin-5 (CLDN5), Occludin (OCLN), and zonula occludens 1 (ZO-1) and vascular endothelial growth factor (VEGF) and matrix metalloproteinase-9 (MMP-9) were measured by using enzyme-linked immunosorbent assay in serum and cerebrospinal fluid (CSF) samples obtained from patients with ICH. The white blood cell (WBC) count in blood and CSF, albumin (ALB) levels in the CSF (ALBCSF), and the BBB ratio were significantly higher in the ICH than in controls (p < 0.05). Significantly higher levels of CLDN5, OCLN, ZO-1, MMP-9, and VEGF in CSF were observed in the ICH group; these biomarkers were also positively associated with BBB ratio (p < 0.05). Our data revealed that circulating TJs could be considered the potential biomarkers reflecting the integrity of the BBB in ICH.
Collapse
Affiliation(s)
- Xiaoyang Jiao
- Department of Cell Biology and Genetics, Shantou University Medical College, Guangdong 515041, China
| | - Ping He
- Shantou University Medical College, Guangdong 515041, China
| | - Yazhen Li
- Department of Cell Biology and Genetics, Shantou University Medical College, Guangdong 515041, China
| | - Zhicheng Fan
- Department of Cell Biology and Genetics, Shantou University Medical College, Guangdong 515041, China
| | - Mengya Si
- Department of Cell Biology and Genetics, Shantou University Medical College, Guangdong 515041, China
| | - Qingdong Xie
- Department of Cell Biology and Genetics, Shantou University Medical College, Guangdong 515041, China
| | - Xiaolan Chang
- Department of Cell Biology and Genetics, Shantou University Medical College, Guangdong 515041, China
| | - Dongyang Huang
- Department of Cell Biology and Genetics, Shantou University Medical College, Guangdong 515041, China
- *Dongyang Huang:
| |
Collapse
|
38
|
Lei B, Sheng H, Wang H, Lascola CD, Warner DS, Laskowitz DT, James ML. Intrastriatal injection of autologous blood or clostridial collagenase as murine models of intracerebral hemorrhage. J Vis Exp 2014. [PMID: 25046028 DOI: 10.3791/51439] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Intracerebral hemorrhage (ICH) is a common form of cerebrovascular disease and is associated with significant morbidity and mortality. Lack of effective treatment and failure of large clinical trials aimed at hemostasis and clot removal demonstrate the need for further mechanism-driven investigation of ICH. This research may be performed through the framework provided by preclinical models. Two murine models in popular use include intrastriatal (basal ganglia) injection of either autologous whole blood or clostridial collagenase. Since, each model represents distinctly different pathophysiological features related to ICH, use of a particular model may be selected based on what aspect of the disease is to be studied. For example, autologous blood injection most accurately represents the brain's response to the presence of intraparenchymal blood, and may most closely replicate lobar hemorrhage. Clostridial collagenase injection most accurately represents the small vessel rupture and hematoma evolution characteristic of deep hemorrhages. Thus, each model results in different hematoma formation, neuroinflammatory response, cerebral edema development, and neurobehavioral outcomes. Robustness of a purported therapeutic intervention can be best assessed using both models. In this protocol, induction of ICH using both models, immediate post-operative demonstration of injury, and early post-operative care techniques are demonstrated. Both models result in reproducible injuries, hematoma volumes, and neurobehavioral deficits. Because of the heterogeneity of human ICH, multiple preclinical models are needed to thoroughly explore pathophysiologic mechanisms and test potential therapeutic strategies.
Collapse
Affiliation(s)
- Beilei Lei
- Multidisciplinary Neuroprotection Laboratories, Department of Anesthesiology, Duke University
| | - Huaxin Sheng
- Multidisciplinary Neuroprotection Laboratories, Department of Anesthesiology, Duke University
| | | | | | - David S Warner
- Multidisciplinary Neuroprotection Laboratories, Department of Anesthesiology, Duke University; Department of Neurobiology, Duke University
| | - Daniel T Laskowitz
- Multidisciplinary Neuroprotection Laboratories, Department of Anesthesiology, Duke University; Department of Neurology, Duke University; Department of Neurobiology, Duke University
| | - Michael L James
- Multidisciplinary Neuroprotection Laboratories, Department of Anesthesiology, Duke University; Department of Neurology, Duke University;
| |
Collapse
|
39
|
Undas A, Brummel-Ziedins KE, Mann KG. Anticoagulant effects of statins and their clinical implications. Thromb Haemost 2013; 111:392-400. [PMID: 24285296 DOI: 10.1160/th13-08-0720] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 10/15/2013] [Indexed: 12/31/2022]
Abstract
There is evidence indicating that statins (3-hydroxy-methylglutaryl coenzyme A reductase inhibitors) may produce several cholesterol-independent antithrombotic effects. In this review, we provide an update on the current understanding of the interactions between statins and blood coagulation and their potential relevance to the prevention of venous thromboembolism (VTE). Anticoagulant properties of statins reported in experimental and clinical studies involve decreased tissue factor expression resulting in reduced thrombin generation and attenuation of pro-coagulant reactions catalysed by thrombin, such as fibrinogen cleavage, factor V and factor XIII activation, as well as enhanced endothelial thrombomodulin expression, resulting in increased protein C activation and factor Va inactivation. Observational studies and one randomized trial have shown reduced VTE risk in subjects receiving statins, although their findings still generate much controversy and suggest that the most potent statin rosuvastatin exerts the largest effect.
Collapse
Affiliation(s)
- A Undas
- Anetta Undas, MD, PhD, Institute of Cardiology, Jagiellonian University School of Medicine, 80 Pradnicka St., 31-202 Krakow, Poland, Tel.: +48 12 6143004, Fax: +48 12 4233900, E-mail:
| | | | | |
Collapse
|
40
|
Bustamante A, Montaner J. Statin Therapy Should Not be Discontinued in Patients With Intracerebral Hemorrhage. Stroke 2013; 44:2060-1. [DOI: 10.1161/strokeaha.113.000916] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Alejandro Bustamante
- From the Neurovascular Research Laboratory, Institut de Recerca, Hospital Universitari Vall d’Hebron, Barcelona, Spain
| | - Joan Montaner
- From the Neurovascular Research Laboratory, Institut de Recerca, Hospital Universitari Vall d’Hebron, Barcelona, Spain
| |
Collapse
|