1
|
Catalão CHR, da Costa LHA, Dos Santos JR, Alberici LC, Falconi-Sobrinho LL, Coimbra NC, Dominguini D, Dal-Pizzol F, Barichello T, Rocha MJA. Mitigating neuroinflammation in cognitive areas: exploring the impact of HMG-CoA reductase inhibitor. Biochem J 2024; 481:1585-1602. [PMID: 39466125 DOI: 10.1042/bcj20240217] [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: 05/08/2024] [Revised: 09/20/2024] [Accepted: 10/28/2024] [Indexed: 10/29/2024]
Abstract
Existing literature suggests that infection-specific mechanisms may play a significant role in the onset and progression of dementia, as opposed to the broader phenomenon of systemic inflammation. In addition, 3-hydroxy-3-methylglutaryl (HMG)-coenzyme A (CoA) reductase inhibitors have been proposed as a potential therapeutic approach for sepsis, given their anti-inflammatory and antioxidant properties. We investigated the neuroprotective effect of an HMG-CoA reductase inhibitor (simvastatin) by analyzing neurodegenerative markers, mitochondrial respiration, and neuronal tracing in the prefrontal cortex (PFC) and thalamic nucleus reuniens (RE) of sepsis survivor animals. Adult Wistar rats were subjected to sepsis by cecal ligation and puncture or left non-manipulated. The animals were treated with simvastatin or vehicle for 4 days before and 10 days after surgery. The treatment preserved the non-associative memory (P < 0.05), recovered expression of Smad-3 in the hippocampus (P < 0.05), and prevented increased expression of calpain-1 (hippocampus: P < 0.0001; PFC: P < 0.05) and GSKβ (hippocampus: P < 0.0001; PFC: P < 0.0001) in the brain structures of the sepsis survivor animals. These animals also showed mitochondrial dysfunction and decreased axon terminals in the RE. Simvastatin seems to restore energy metabolism by improving the electron transfer system (ETS) values in the hippocampus (P < 0.01) and the oxidative phosphorylation/ETS (P/E) ratio in the PFC (P < 0.05), in addition to preventing the reduction of axon terminals in survivor animals. These results suggest a potential neuroprotective effect and the importance of considering HMG-CoA reductase inhibitors as a possible adjuvant therapy in sepsis.
Collapse
Affiliation(s)
- Carlos Henrique Rocha Catalão
- Department of Neurosciences and Behavioral Sciences of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto-SP, Brazil
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, U.S.A
- Department of Psychology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto-SP, Brazil
| | - Luis Henrique Angenendt da Costa
- Department of Neurosciences and Behavioral Sciences of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto-SP, Brazil
| | - Jonathas Rodrigo Dos Santos
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto-SP, Brazil
| | - Luciane Carla Alberici
- Department of Physics and Chemistry, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto-SP, Brazil
| | | | - Norberto Cysne Coimbra
- Department of Pharmacology of Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto-SP, Brazil
| | - Diogo Dominguini
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Felipe Dal-Pizzol
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Tatiana Barichello
- Department of Psychiatry and Behavioral Sciences, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, U.S.A
- Laboratory of Experimental Pathophysiology, Graduate Program in Health Sciences, University of Southern Santa Catarina (UNESC), Criciúma, SC, Brazil
| | - Maria José Alves Rocha
- Department of Psychology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto-SP, Brazil
| |
Collapse
|
2
|
Pordel S, McCloskey AP, Almahmeed W, Sahebkar A. The protective effects of statins in traumatic brain injury. Pharmacol Rep 2024; 76:235-250. [PMID: 38448729 DOI: 10.1007/s43440-024-00582-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/28/2024] [Accepted: 02/29/2024] [Indexed: 03/08/2024]
Abstract
Traumatic brain injury (TBI), often referred to as the "silent epidemic", is the most common cause of mortality and morbidity worldwide among all trauma-related injuries. It is associated with considerable personal, medical, and economic consequences. Although remarkable advances in therapeutic approaches have been made, current treatments and clinical management for TBI recovery still remain to be improved. One of the factors that may contribute to this gap is that existing therapies target only a single event or pathology. However, brain injury after TBI involves various pathological mechanisms, including inflammation, oxidative stress, blood-brain barrier (BBB) disruption, ionic disturbance, excitotoxicity, mitochondrial dysfunction, neuronal necrosis, and apoptosis. Statins have several beneficial pleiotropic effects (anti-excitotoxicity, anti-inflammatory, anti-oxidant, anti-thrombotic, immunomodulatory activity, endothelial and vasoactive properties) in addition to promoting angiogenesis, neurogenesis, and synaptogenesis in TBI. Supposedly, using agents such as statins that target numerous and diverse pathological mechanisms, may be more effective than a single-target approach in TBI management. The current review was undertaken to investigate and summarize the protective mechanisms of statins against TBI. The limitations of conducted studies and directions for future research on this potential therapeutic application of statins are also discussed.
Collapse
Affiliation(s)
- Safoora Pordel
- Department of Immunology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alice P McCloskey
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, UK
| | - Wael Almahmeed
- Heart and Vascular Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Katlowitz K, Gopinath S, Cruz Navarro J, Robertson C. HMG-CoA Reductase Inhibitors for Traumatic Brain Injury. Neurotherapeutics 2023; 20:1538-1545. [PMID: 37351829 PMCID: PMC10684840 DOI: 10.1007/s13311-023-01399-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/09/2023] [Indexed: 06/24/2023] Open
Abstract
Traumatic brain injuries (TBIs) are associated with high morbidity and mortality due to both the original insult as well as the destructive biological response that follows. Medical management aims to slow or even halt secondary neurological injury while simultaneously laying the groundwork for recovery. Statins are one class of medications that is showing increased promise in the management of TBI. Used extensively in cardiovascular disease, these drugs were originally developed as competitive inhibitors within the cholesterol production pipeline. They are now used in diverse disease states due to their pleiotropic effects on other biological processes such as inflammation and angiogenesis. Preclinical studies, retrospective reviews, and randomized clinical trials have shown a variety of benefits in the management of TBI, but to date, no large-scale randomized clinical trial has been performed. Despite this limitation, statins' early promise and well-tolerated side effect profile make them a promising new tool in the management of TBIs. More bench and clinical studies are needed to delineate proper treatment regimens as well as understand their true potential.
Collapse
Affiliation(s)
- Kalman Katlowitz
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, 77030, USA.
| | - Shankar Gopinath
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Jovany Cruz Navarro
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Claudia Robertson
- Department of Neurosurgery, Baylor College of Medicine, Houston, TX, 77030, USA
| |
Collapse
|
4
|
Susanto M, Pangihutan Siahaan AM, Wirjomartani BA, Setiawan H, Aryanti C, Michael. The neuroprotective effect of statin in traumatic brain injury: A systematic review. World Neurosurg X 2023; 19:100211. [PMID: 37251243 PMCID: PMC10220252 DOI: 10.1016/j.wnsx.2023.100211] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 04/23/2023] [Accepted: 05/02/2023] [Indexed: 07/22/2023] Open
Abstract
Despite recent encouraging pharmaceutical and technical breakthroughs in neurosurgical critical care, traumatic brain injury (TBI)-related mortality and morbidity remain substantial clinical issues. Medication of statins was revealed to enhance outcomes following TBI in animal research. In addition to their main role of decreasing serum cholesterol, statins decrease inflammation and enhance cerebral blood flow. However, research on the efficacy of statins in TBI is still limited. This systematic review was conducted to determine the efficacy of statins in enhancing the clinical outcomes of TBI individuals, and specifically investigate the optimal dose and form of statins. The databases of PubMed, DOAJ, EBSCO, and Cochrane were extensively researched. The date of publication within the last fifteen years was the inclusion criterion. Meta-analyses, clinical trials, and randomized controlled trials were prioritized forms of research publications. Ambiguous remarks, irrelevant correlations to the main issue, or a focus on disorders other than TBI were the exclusion criteria. Thirteen research were included in this study. Simvastatin, atorvastatin, and rosuvastatin were the main form of statins discussed in this study. Enhancement of the Glasgow Coma Scale, survival rates, hospital length of stay, and cognitive outcomes were revealed in this study. This study suggests either simvastatin 40 mg, atorvastatin 20 mg, or rosuvastatin 20 mg for 10 days as the optimal therapeutic forms and doses to be applied in the management of TBI. Pre-TBI statin use was linked to lower risk of mortality in TBI individuals compared to nonusers, whereas statin discontinuation was linked to an increase in mortality.
Collapse
Affiliation(s)
- Martin Susanto
- Faculty of Medicine, University of Sumatera Utara, Medan, North Sumatra, Indonesia
| | | | | | - Hendy Setiawan
- Department of Neurosurgery, University of Sumatera Utara, Medan, North Sumatra, Indonesia
| | - Citra Aryanti
- Department of Surgery, University of Udayana, Denpasar, Bali, Indonesia
| | - Michael
- Department of Neurosurgery, University of Padjadjaran, Bandung, West Java, Indonesia
| |
Collapse
|
5
|
Stefanou MI, Palaiodimou L, Katsanos AH, Milionis H, Kosmidou M, Lambadiari V, Halvatsiotis P, Ferentinos P, Andreadou E, Marinos G, Theodorou A, Tzartos JS, Voumvourakis K, Tsivgoulis G, Giannopoulos S. The effects of HMG-CoA reductase inhibitors on disease activity in multiple sclerosis: A systematic review and meta-analysis. Mult Scler Relat Disord 2022; 58:103395. [PMID: 35216778 DOI: 10.1016/j.msard.2021.103395] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 11/08/2021] [Indexed: 12/27/2022]
Abstract
OBJECTIVE To assess whether statins (3‑hydroxy-3-methylglutaryl coenzyme A reductase inhibitors) exert disease-modifying effects in multiple sclerosis (MS). APPROACH A systematic review and meta-analysis was performed including randomized-controlled clinical trials (RCTs) on statin use in MS. A random-effects model was applied to calculate pooled estimates and odds ratios (ORs) with corresponding 95% confidence intervals (CIs), when comparing patients treated with statins alone or adjunct to disease modifying treatment (DMT) to non-statin-treated patients. RESULTS We identified 7 RCTs including 789 patients with relapsing-remitting MS (RRMS), all of whom received additional DMT with IFN-β. Single identified RCTs in secondary-progressive MS (SPMS), clinically isolated syndrome (CIS) and optic neuritis (ON) were not meta-analyzed. In RRMS, add-on statin use was not associated with the risk of clinical relapse (OR=1.30, 95%CI: 0.901.87) or EDSS-progression from baseline, neither appeared related to the risk of new contrast-enhancing or T2 lesions (OR=1.28, 95%CI: 0.364.58), and the risk of whole-brain volume reduction on MRI. Add-on statins to IFN-β were safe and well-tolerated. In SPMS, stand-alone simvastatin led to significantly reduced annualized rate of whole-brain volume reduction. In CIS and ON, statins were associated with reduced risk for new T2 lesions and improved visual recovery, respectively. CONCLUSIONS We detected no benefit from statin treatment as add-on to IFN-β in RRMS. However, a potential beneficial effect in SPMS, CIS and ON deserves independent confirmation and further evaluation within adequately powered RCTs.
Collapse
Affiliation(s)
- Maria-Ioanna Stefanou
- Second Department of Neurology, "Attikon" University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Department of Neurology & Stroke, Hertie-Institute for Clinical Brain Research, Eberhard-Karls University of Tübingen, Tübingen, Germany
| | - Lina Palaiodimou
- Second Department of Neurology, "Attikon" University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Aristeidis H Katsanos
- Second Department of Neurology, "Attikon" University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Division of Neurology, McMaster University/ Population Health Research Institute, Hamilton, Canada
| | - Haralampos Milionis
- Department of Internal Medicine, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Maria Kosmidou
- Department of Internal Medicine, Faculty of Medicine, School of Health Sciences, University of Ioannina, Ioannina, Greece
| | - Vaia Lambadiari
- Second Department of Internal Medicine Research Unit and Diabetes Center, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, Rimini 1 Chaidari, Athens, Greece
| | - Panagiotis Halvatsiotis
- Second Department of Internal Medicine Research Unit and Diabetes Center, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, Rimini 1 Chaidari, Athens, Greece
| | - Panagiotis Ferentinos
- Second Department of Psychiatry, Eating Disorders Unit, National and Kapodistrian University of Athens, Medical School, "Attikon" University General Hospital, Athens, Greece
| | - Elizabeth Andreadou
- First Department of Neurology, National & Kapodistiran University of Athens, School of Medicine, "Eginition" University Hospital, Athens, Greece
| | - Georgios Marinos
- Department of Hygiene, Epidemiology and Medical Statistics, National and Kapodistrian University of Athens, Athens, Greece
| | - Aikaterini Theodorou
- Second Department of Neurology, "Attikon" University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - John S Tzartos
- Second Department of Neurology, "Attikon" University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Voumvourakis
- Second Department of Neurology, "Attikon" University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Georgios Tsivgoulis
- Second Department of Neurology, "Attikon" University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Department of Neurology, University of Tennessee Health Science Center, Memphis, TN, United States of America.
| | - Sotirios Giannopoulos
- Second Department of Neurology, "Attikon" University Hospital, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
6
|
Sultan W, Sapkota A, Khurshid H, Qureshi IA, Jahan N, Went TR, Dominic JL, Win M, Kannan A, Tara A, Ruo SW, Alfonso M. Statins' Effect on Cognitive Outcome After Traumatic Brain Injury: A Systematic Review. Cureus 2021; 13:e16953. [PMID: 34405076 PMCID: PMC8352842 DOI: 10.7759/cureus.16953] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 08/06/2021] [Indexed: 11/05/2022] Open
Abstract
Traumatic brain injury (TBI), also known as the "Silent Epidemic," is a growing devastating global health problem estimated to affect millions of individuals yearly worldwide with little public recognition, leading to many individuals living with a TBI-related disability. TBI has been associated with up to five times increase in the risk of dementia among multiple neurologic complications compared with the general population. Several therapies, including statins, have been tried and showed promising benefits for TBI patients. In this systematic review, we evaluated the recent literature that tested the role of statins on neurological and cognitive outcomes such as Alzheimer's Disease and non-Alzheimer's dementia in survivors of TBI with various severities. We conducted a systematic search on PubMed, PubMed Central, MEDLINE, and Google Scholar. MeSH terms and keywords were used to search for full-text randomized clinical trials (RCTs), cross-sectional, case-control, cohort studies, systematic reviews, and animal studies published in English. Inclusion and exclusion criteria were applied, and the articles were subjected to quality appraisal by two reviewers. Our data search retrieved 4948 nonduplicate records. A total of 18 studies were included - nine human studies, and nine animal laboratory trials - after meeting inclusion, eligibility, and quality assessment criteria. Simvastatin was the most tested statin, and the oral route of administration was the most used. Eight human studies showed a significant neuroprotective effect and improvement in the cognitive outcomes, including dementia. Four randomized clinical trials with 296 patients showed that statins play a neuroprotective role and improve cognitive outcomes through different mechanisms, especially their anti-inflammatory effect; they were shown to lower tumor necrosis factor (TNF)-α and C-reactive protein (CRP) levels. Also, they decreased axonal injury and cortical thickness changes. In addition, four cohort studies compared a total of 867.953 patients. One study showed a decrease in mortality in statin-treated patients (p=0.05). Another study showed a reduction in the incidence of Alzheimer's disease and related dementias (RR, 0.77; 95% CI, 0.73-0.81), while one study showed a decreased risk of dementia after concussions by 6.13% (p=0.001). On the other hand, one cohort study showed no significant difference with the use of statins. In eight animal trials, statins showed a significant neuroprotective effect, improved cognitive outcomes, and neurological functions. Different molecular and cellular mechanisms were suggested, including anti-inflammatory effects, promoting angiogenesis, neurogenesis, increasing cerebral blood flow, neurite outgrowth, promoting the proliferation and differentiation of neural stem cells, and reducing axonal injury. On the contrary, one study showed no benefit and actual adverse effect on the cognitive outcome. Most of the studies showed promising neuroprotective effects of statins in TBI patients. Cognitive outcomes, especially dementia, were improved. However, the optimal therapeutic protocol is still unknown. Thus, statins are candidates for more advanced studies to test their efficacy in preventing cognitive decline in patients with TBI.
Collapse
Affiliation(s)
- Waleed Sultan
- Medicine, California Institute of Behavioral Neurosciences & Psychology (CIBNP), Fairfield, USA
| | - Alisha Sapkota
- Psychiatry, California Institute of Behavioral Neurosciences & Psychology (CIBNP), Fairfield, USA
| | - Hajra Khurshid
- Psychiatry, California Institute of Behavioral Neurosciences & Psychology (CIBNP), Fairfield, USA
| | - Israa A Qureshi
- Medicine, California Institute of Behavioral Neurosciences & Psychology (CIBNP), Fairfield, USA
| | - Nasrin Jahan
- Psychiatry, California Institute of Behavioral Neurosciences & Psychology (CIBNP), Fairfield, USA
| | - Terry R Went
- Medicine, California Institute of Behavioral Neurosciences & Psychology (CIBNP), Fairfield, USA
| | - Jerry Lorren Dominic
- General Surgery, California Institute of Behavioral Neurosciences & Psychology (CIBNP), Fairfield, USA
| | - Myat Win
- General Surgery, California Institute of Behavioral Neurosciences & Psychology (CIBNP), Fairfield, USA
| | - Amudhan Kannan
- General Surgery, California Institute of Behavioral Neurosciences & Psychology (CIBNP), Fairfield, USA
| | - Anjli Tara
- General Surgery, California Institute of Behavioral Neurosciences & Psychology (CIBNP), Fairfield, USA
| | - Sheila W Ruo
- General Surgery, California Institute of Behavioral Neurosciences & Psychology (CIBNP), Fairfield, USA
| | - Michael Alfonso
- Medicine, California Institute of Behavioral Neurosciences & Psychology (CIBNP), Fairfield, USA
| |
Collapse
|
7
|
Du RW, Bu WG. Simvastatin Prevents Neurodegeneration in the MPTP Mouse Model of Parkinson's Disease via Inhibition of A1 Reactive Astrocytes. Neuroimmunomodulation 2021; 28:82-89. [PMID: 33735898 DOI: 10.1159/000513678] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 12/09/2020] [Indexed: 11/19/2022] Open
Abstract
Emerging evidence indicates that A1 reactive astrocytes play crucial roles in the pathogenesis of Parkinson's disease (PD). Thus, development of agents that could inhibit the formation of A1 reactive astrocytes could be used to treat PD. Simvastatin has been touted as a potential neuroprotective agent for neurologic disorders such as PD, but the specific underlying mechanism remains unclear. The 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD and primary astrocytes/neurons were prepared to investigate the effects of simvastatin on PD and its underlying mechanisms in vitro and in vivo. We show that simvastatin protects against the loss of dopamine neurons and behavioral deficits in the MPTP mouse model of PD. We also found that simvastatin suppressed the expression of A1 astrocytic specific markers in vivo and in vitro. In addition, simvastatin alleviated neuron death induced by A1 astrocytes. Our findings reveal that simvastatin is neuroprotective via the prevention of conversion of astrocytes to an A1 neurotoxic phenotype. In light of simvastatin favorable properties, it should be evaluated in the treatment of PD and related neurologic disorders characterized by A1 reactive astrocytes.
Collapse
Affiliation(s)
- Ren-Wei Du
- Department of Neurology, Chaoyang Hospital, Huainan, China,
| | - Wen-Guang Bu
- Department of Neurology, Chaoyang Hospital, Huainan, China
| |
Collapse
|
8
|
Neekhra A, Tran J, Esfahani PR, Schneider K, Pham K, Sharma A, Chwa M, Luthra S, Gramajo AL, Mansoor S, Kuppermann BD, Kenney MC. Memantine, Simvastatin, and Epicatechin Inhibit 7-Ketocholesterol-induced Apoptosis in Retinal Pigment Epithelial Cells But Not Neurosensory Retinal Cells In Vitro. J Ophthalmic Vis Res 2020; 15:470-480. [PMID: 33133437 PMCID: PMC7591846 DOI: 10.18502/jovr.v15i4.7781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Accepted: 06/21/2020] [Indexed: 11/27/2022] Open
Abstract
Purpose 7-ketocholesterol (7kCh), a natural byproduct of oxidation in lipoprotein deposits is implicated in the pathogenesis of diabetic retinopathy and age-related macular degeneration (AMD). This study was performed to investigate whether several clinical drugs can inhibit 7kCh-induced caspase activation and mitigate its apoptotic effects on retinal cells in vitro. Methods Two populations of retinal cells, human retinal pigment epithelial cells (ARPE-19) and rat neuroretinal cells (R28) were exposed to 7kCh in the presence of the following inhibitors: Z-VAD-FMK (pan-caspase inhibitor), simvastatin, memantine, epicatechin, and Z-IETD-FMK (caspase-8 inhibitor) or Z-ATAD-FMK (caspase-12 inhibitor). Caspase-3/7, -8, and -12 activity levels were measured by fluorochrome caspase assays to quantify cell death. IncuCyte live-cell microscopic images were obtained to quantify cell counts. Results Exposure to 7kCh for 24 hours significantly increased caspase activities for both ARPE-19 and R28 cells (P< 0.05). In ARPE cells, pretreatment with various drugs had significantly lower caspase-3/7, -8, and -12 activities, reported in % change in mean signal intensity (msi): Z-VAD-FMK (48% decrease, P< 0.01), memantine (decreased 47.8% at 1 µM, P = 0.0039 and 81.9% at 1 mM, P< 0.001), simvastatin (decreased 85.3% at 0.01 µM, P< 0.001 and 84.8% at 0.05 µM, P< 0.001) or epicatechin (83.6% decrease, P< 0.05), Z-IETD-FMK (68.1% decrease, P< 0.01), and Z-ATAD-FMK (47.7% decrease, P = 0.0017). In contrast, R28 cells exposed to 7kCh continued to have elevated caspase-3/7, -8, and -12 activities (between 25.7% decrease and 17.5% increase in msi, P> 0.05) regardless of the pretreatment. Conclusion Several current drugs protect ARPE-19 cells but not R28 cells from 7kCh-induced apoptosis, suggesting that a multiple-drug approach is needed to protect both cells types in various retinal diseases.
Collapse
Affiliation(s)
- Aneesh Neekhra
- Gavin Herbert Eye Institute, University of California, Irvine, California
| | - Julia Tran
- Gavin Herbert Eye Institute, University of California, Irvine, California
| | - Parsa R Esfahani
- Gavin Herbert Eye Institute, University of California, Irvine, California
| | - Kevin Schneider
- Gavin Herbert Eye Institute, University of California, Irvine, California
| | - Khoa Pham
- Gavin Herbert Eye Institute, University of California, Irvine, California
| | - Ashish Sharma
- Gavin Herbert Eye Institute, University of California, Irvine, California
| | - Marilyn Chwa
- Gavin Herbert Eye Institute, University of California, Irvine, California
| | - Saurabh Luthra
- Gavin Herbert Eye Institute, University of California, Irvine, California
| | - Ana L Gramajo
- Gavin Herbert Eye Institute, University of California, Irvine, California
| | - Saffar Mansoor
- Gavin Herbert Eye Institute, University of California, Irvine, California
| | | | - M Cristina Kenney
- Gavin Herbert Eye Institute, University of California, Irvine, California.,Department of Pathology and Laboratory Medicine, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
9
|
Simvastatin Prevents Long-Term Cognitive Deficits in Sepsis Survivor Rats by Reducing Neuroinflammation and Neurodegeneration. Neurotox Res 2020; 38:871-886. [PMID: 32524380 DOI: 10.1007/s12640-020-00222-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/09/2020] [Accepted: 05/01/2020] [Indexed: 02/07/2023]
Abstract
Sepsis-associated encephalopathy causes brain dysfunction that can result in cognitive impairments in sepsis survivor patients. In previous work, we showed that simvastatin attenuated oxidative stress in brain structures related to memory in septic rats. However, there is still a need to evaluate the long-term impact of simvastatin administration on brain neurodegenerative processes and cognitive damage in sepsis survivors. Here, we investigated the possible neuroprotective role of simvastatin in neuroinflammation, and neurodegeneration conditions of brain structures related to memory in rats at 10 days after sepsis survival. Male Wistar rats (250-300 g) were submitted to cecal ligation and puncture (CLP, n = 42) or remained as non-manipulated (naïve, n = 30). Both groups were treated (before and after the surgery) by gavage with simvastatin (20 mg/kg) or an equivalent volume of saline and observed for 10 days. Simvastatin-treated rats that survived to sepsis showed a reduction in the levels of nitrate, IL1-β, and IL-6 and an increase in Bcl-2 protein expression in the prefrontal cortex and hippocampus, and synaptophysin only in the hippocampus. Immunofluorescence revealed a reduction of glial activation, neurodegeneration, apoptosis, and amyloid aggregates confirmed by quantification of GFAP, Iba-1, phospho Ser396-tau, total tau, cleaved caspase-3, and thioflavin-S in the prefrontal cortex and hippocampus. In addition, treated animals presented better performance in tasks involving habituation memory, discriminative, and aversive memory. These results suggest that statins exert a neuroprotective role by upregulation of the Bcl-2 and gliosis reduction, which may prevent the cognitive deficit observed in sepsis survivor animals.
Collapse
|
10
|
Lovastatin, not Simvastatin, Corrects Core Phenotypes in the Fragile X Mouse Model. eNeuro 2019; 6:ENEURO.0097-19.2019. [PMID: 31147392 PMCID: PMC6565377 DOI: 10.1523/eneuro.0097-19.2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 05/11/2019] [Accepted: 05/13/2019] [Indexed: 02/07/2023] Open
Abstract
The cholesterol-lowering drug lovastatin corrects neurological phenotypes in animal models of fragile X syndrome (FX), a commonly identified genetic cause of autism and intellectual disability (ID). The therapeutic efficacy of lovastatin is being tested in clinical trials for FX; however, the structurally similar drug simvastatin has been proposed as an alternative due to an increased potency and brain penetrance. Here, we perform a side-by-side comparison of the effects of lovastatin and simvastatin treatment on two core phenotypes in Fmr1-/y mice versus WT littermates: excessive hippocampal protein synthesis and susceptibility to audiogenic seizures (AGSs). We find that simvastatin does not correct excessive hippocampal protein synthesis in the Fmr1-/y hippocampus at any dose tested. In fact, simvastatin significantly increases protein synthesis in both Fmr1-/y and WT. Moreover, injection of simvastatin does not reduce AGS in the Fmr1-/y mouse, while lovastatin significantly reduces AGS incidence and severity versus vehicle-treated animals. These results show that unlike lovastatin, simvastatin does not correct core phenotypes in the Fmr1-/y mouse model.
Collapse
|
11
|
Differences in statin associated neuroprotection corresponds with either decreased production of IL-1β or TNF-α in an in vitro model of neuroinflammation-induced neurodegeneration. Toxicol Appl Pharmacol 2018. [DOI: 10.1016/j.taap.2018.03.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
12
|
Robertson CS, McCarthy JJ, Miller ER, Levin H, McCauley SR, Swank PR. Phase II Clinical Trial of Atorvastatin in Mild Traumatic Brain Injury. J Neurotrauma 2017; 34:1394-1401. [PMID: 28006970 DOI: 10.1089/neu.2016.4717] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Statins constitute a class of medications commonly used in the treatment of elevated cholesterol. However, in experimental studies, statins also have other non-cholesterol-mediated mechanisms of action, which may have neuroprotective effects. The aim of this study was to determine whether administration of atorvastatin for 7 days post-injury would improve neurological recovery in patients with mild traumatic brain injury (mTBI). The hypothesis was that atorvastatin administration would reduce post-concussion symptoms and also that atorvastatin administration for 1 week post-injury would be safe. One hundred forty patients with mTBI were planned to be enrolled and randomly assigned to receive atorvastatin 1 mg/kg (up to 80 mg/kg) per day or placebo for 7 days starting within 24 h of injury. Assessments of post-concussion syndrome, post-traumatic stress and depressive symptoms, cognition, memory, verbal fluency, functional, and work status were performed at baseline, 1 week, and 1 and 3 months. The result on the Rivermead Post-Concussion Symptoms Questionnaire at 3 months was the primary outcome. Enrollment in the trial was stopped early because of difficulty in recruiting sufficient numbers of subjects. Fifty-two patients with mTBI were enrolled; 28 patients received atorvastatin and 24 received placebo. The median Rivermead score was 2 for the atorvastatin group, compared to 3.5 for the placebo group, at 3 months post-injury (χ2(1) = 0.0976; p = 0.7547). The change in the Rivermead score between baseline and 3 months was also analyzed. The median decrease in score was 4 for the atorvastatin group and 10.5 for the placebo group (χ2(1) = 0.8750; p = 0.3496). No serious adverse events occurred, and there was no significant difference in the incidence of adverse events in the two treatment groups. Atorvastatin administration for 7 days post-injury was safe, but there were no significant differences in neurological recovery post-mTBI with atorvastatin.
Collapse
Affiliation(s)
| | - James J McCarthy
- 2 University of Texas Health Science Center at Houston , Houston, Texas
| | | | - Harvey Levin
- 3 Michael E. De Bakey Veterans Affairs Medical Center and Baylor College of Medicine , Houston, Texas
| | | | - Paul R Swank
- 4 University of Texas Health Science Center at Houston , School of Public Health, Houston, Texas
| |
Collapse
|
13
|
Sohn HM, Hwang JY, Ryu JH, Kim J, Park S, Park JW, Han SH. Simvastatin protects ischemic spinal cord injury from cell death and cytotoxicity through decreasing oxidative stress: in vitro primary cultured rat spinal cord model under oxygen and glucose deprivation-reoxygenation conditions. J Orthop Surg Res 2017; 12:36. [PMID: 28241838 PMCID: PMC5330028 DOI: 10.1186/s13018-017-0536-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 02/15/2017] [Indexed: 01/21/2023] Open
Abstract
Background Ischemia and the following reperfusion damage are critical mechanisms of spinal cord injury. Statins have been reported to decrease ischemia–reperfusion injury in many organs including the spinal cord. Anti-oxidative effect is one of the main protective mechanisms of statin against neuronal death and cytotoxicity. We hypothesized that statins’ anti-oxidative property would yield neuroprotective effects on spinal cord ischemia–reperfusion injury Methods Primary cultured spinal cord motor neurons were isolated from Sprague–Dawley rat fetuses. Ischemia–reperfusion injury model was induced by 60 min of oxygen and glucose deprivation (OGD) and 24 h of reoxygenation. Healthy and OGD cells were treated with simvastatin at concentrations of 0.1, 1, and 10 μM for 24 h. Cell viability was assessed using water-soluble tetrazolium salt (WST)-8, cytotoxicity with LDH, and production of free radicals with DCFDA (2′,7′-dichlorofluorescein diacetate). Results OGD reduced neuronal viability compared to normoxic control by 35.3%; however, 0.1–10 μM of simvastatin treatment following OGD improved cell survival. OGD increased LDH release up to 214%; however, simvastatin treatment attenuated its cytotoxicity at concentrations of 0.1–10 μM (p < 0.001 and p = 0.001). Simvastatin also reduced deteriorated morphological changes of motor neurons following OGD. Oxidative stress was reduced by simvastatin (0.1–10 μM) compared to untreated cells exposed to OGD (p < 0.001). Conclusions Simvastatin effectively reduced spinal cord neuronal death and cytotoxicity against ischemia–reperfusion injury, probably via modification of oxidative stress.
Collapse
Affiliation(s)
- Hye-Min Sohn
- Department of Anesthesiology and Pain Medicine, Seoul National University Bundang Hospital, 82 Gumi-ro, 173 Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea
| | - Jin-Young Hwang
- Department of Anesthesiology and Pain Medicine, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea
| | - Jung-Hee Ryu
- Department of Anesthesiology and Pain Medicine, Seoul National University Bundang Hospital, 82 Gumi-ro, 173 Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea
| | - Jinhee Kim
- Department of Anesthesiology and Pain Medicine, Seoul National University Bundang Hospital, 82 Gumi-ro, 173 Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea
| | - Seongjoo Park
- Department of Anesthesiology and Pain Medicine, Seoul National University Bundang Hospital, 82 Gumi-ro, 173 Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea
| | - Jin-Woo Park
- Department of Anesthesiology and Pain Medicine, Seoul National University Bundang Hospital, 82 Gumi-ro, 173 Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea
| | - Sung-Hee Han
- Department of Anesthesiology and Pain Medicine, Seoul National University Bundang Hospital, 82 Gumi-ro, 173 Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, Republic of Korea.
| |
Collapse
|
14
|
Ferraro S, Astort F, Yakisich J, Tasat D. Particulate matter cytotoxicity in cultured SH-SY5Y cells is modulated by simvastatin: Toxicological assessment for oxidative damage. Neurotoxicology 2016; 53:108-114. [DOI: 10.1016/j.neuro.2016.01.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 01/05/2016] [Accepted: 01/05/2016] [Indexed: 11/25/2022]
|
15
|
Jeong JH, Yum KS, Chang JY, Kim M, Ahn JY, Kim S, Lapchak PA, Han MK. Dose-specific effect of simvastatin on hypoxia-induced HIF-1α and BACE expression in Alzheimer's disease cybrid cells. BMC Neurol 2015; 15:127. [PMID: 26228060 PMCID: PMC4521481 DOI: 10.1186/s12883-015-0390-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 07/22/2015] [Indexed: 11/12/2022] Open
Abstract
Background Alzheimer’s disease (AD) is associated with vascular risk factors; brain ischemia facilitates the pathogenesis of AD. Recent studies have suggested that the reduction of AD risk with statin was achieved by decreased amyloidogenic amyloid precursor protein. Methods We used mitochondrial transgenic neuronal cell (cybrid) models to investigate changes in the levels of intracellular hypoxia inducible factor 1α (HIF-1α) and β-site amyloid precursor protein cleaving enzyme (BACE) in the presence of simvastatin. Sporadic AD (SAD) and age-matched control (CTL) cybrids were exposed to 2 % O2 and incubated with 1 μM or 10 μM simvastatin. Results There was no significant difference between cell survival by 1 or 10 μM simvastatin in both SAD and CTL cybrids. In the presence of 1 μM simvastatin, intracellular levels of HIF-1α and BACE decreased by 40–70 % in SAD, but not CTL cybrids. However, 10 μM simvastatin increased HIF-1α and BACE expression in both cybrid models. Conclusion Our results suggest demonstrate differential dose-dependent effects of simvastatin on HIF-1α and BACE in cultured Alzheimer’s disease cybrid cells.
Collapse
Affiliation(s)
- Jin-Heon Jeong
- Department of Neurology, College of Medicine, Chungbuk National University, Chungbuk National University Hospital, Cheongju, Korea.
| | - Kyu Sun Yum
- Department of Neurology, College of Medicine, Konyang University, Konyang University Hospital, Daejeon, Korea.
| | - Jun Young Chang
- Department of Neurology, College of Medicine, Seoul National University, Seoul National University Bundang Hospital, Seongnam, Korea.
| | - Manho Kim
- Department of Neurology, College of Medicine, Seoul National University, Seoul National University Hospital, Seoul, Korea.
| | - Jin-young Ahn
- Department of Neurology, Seoul Medical Center, Seoul, Korea.
| | - SangYun Kim
- Department of Neurology, College of Medicine, Seoul National University, Seoul National University Bundang Hospital, Seongnam, Korea.
| | - Paul A Lapchak
- Departments of Neurology and Neurosurgery, Cedars-Sinai Medical Center, Los Angeles, USA.
| | - Moon-Ku Han
- Department of Neurology, College of Medicine, Seoul National University, Seoul National University Bundang Hospital, Seongnam, Korea.
| |
Collapse
|
16
|
Lin CM, Lin YT, Lin RD, Huang WJ, Lee MH. Neurocytoprotective Effects of Aliphatic Hydroxamates from Lovastatin, a Secondary Metabolite from Monascus-Fermented Red Mold Rice, in 6-Hydroxydopamine (6-OHDA)-Treated Nerve Growth Factor (NGF)-Differentiated PC12 Cells. ACS Chem Neurosci 2015; 6:716-24. [PMID: 25692332 DOI: 10.1021/cn500275k] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Lovastatin, a secondary metabolite isolated from Monascus-fermented red rice mold, has neuroprotective activity and permeates the blood-brain barrier. The aim of this study was to enhance the activity of lovastatin for potential use as a treatment for neuronal degeneration in Parkinson's disease. Six lovastatin-derived compounds were semisynthesized and screened for neurocytoprotective activity against 6-hydroxydopamine (6-OHDA)-induced toxicity in human neuroblastoma PC12 cells. Four compounds, designated as 3a, 3d, 3e, and 3f, significantly enhanced cell viability. In particular, compound 3f showed excellent neurocytoprotective activity (97.0 ± 2.7%). Annexin V-FITC and propidium iodide double staining and 4',6-diamidino-2-phenylindole staining indicated that compound 3f reduced 6-OHDA-induced apoptosis in PC12 cells. Compound 3f also reduced caspase-3, -8, and -9 activities, and intracellular calcium concentrations elevated by 6-OHDA in a concentration-dependent manner, without inhibiting reactive oxygen species generation. JC-1 staining indicated that compound 3f also stabilized mitochondrial membrane potential. Thus, compound 3f may be used as a neurocytoprotective agent. Future studies should investigate its potential application as a treatment for Parkinson's disease.
Collapse
Affiliation(s)
- Chien-Min Lin
- Graduate
Institute of Clinical Medicine, Taipei Medical University, Taipei 110, Taiwan
- Department
of Neurosurgery, Taipei Medical University—Wan Fang Hospital, Taipei 116, Taiwan
- Department
of Neurosurgery, Taipei Medical University—Shuang Ho Hospital, Taipei County 235, Taiwan
| | - Yi-Tzu Lin
- Graduate
Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
| | - Rong-Dih Lin
- Department
of Internal Medicine, Heping Branch, Taipei City Hospital, Taipei 100, Taiwan
| | - Wei-Jan Huang
- Graduate
Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
- Ph.D. Program for the
Clinical Drug Discovery from Botanical Herbs, Taipei 110, Taiwan
- School
of Pharmacy, National Defense Medical Center, Taipei 114, Taiwan
| | - Mei-Hsien Lee
- Graduate
Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan
- Ph.D. Program for the
Clinical Drug Discovery from Botanical Herbs, Taipei 110, Taiwan
- Center for Reproductive Medicine & Sciences, Taipei Medical University Hospital, Taipei 110, Taiwan
| |
Collapse
|
17
|
Ghasemi F, Moradi A, Izadpanah E, Moloudi MR, Hassanzadeh K, Rahimmi A, Hassanzadeh K. Simvastatin prevents morphine antinociceptive tolerance and withdrawal symptoms in rats. J Formos Med Assoc 2015; 114:399-406. [DOI: 10.1016/j.jfma.2014.07.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Revised: 07/28/2014] [Accepted: 07/29/2014] [Indexed: 01/06/2023] Open
|
18
|
Lee SH, Choi NY, Yu HJ, Park J, Choi H, Lee KY, Huh YM, Lee YJ, Koh SH. Atorvastatin Protects NSC-34 Motor Neurons Against Oxidative Stress by Activating PI3K, ERK and Free Radical Scavenging. Mol Neurobiol 2015; 53:695-705. [PMID: 25577170 DOI: 10.1007/s12035-014-9030-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Accepted: 11/30/2014] [Indexed: 12/13/2022]
Abstract
Although statins, or hydroxymethylglutaryl coenzyme A (HMG-Co A) reductase inhibitors, are generally used to decrease levels of circulating cholesterol, they have also been reported to have neuroprotective effects through various mechanisms. However, recent results have indicated that they may be harmful in patients with amyotrophic lateral sclerosis (ALS). In this study, we investigate whether atorvastatin protects motor neuron-like cells (NSC-34D) from oxidative stress. To evaluate the effects of atorvastatin or hydrogen peroxide or both on NSC-34D cells, the cells were treated with various combinations of these agents. To evaluate the viability of the cells, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays and trypan blue staining were performed. Levels of free radicals and intracellular signaling proteins were evaluated using the fluorescent probe 2',7'-dichlorodihydrofluorescein diacetate (DCFH-DA) and Western blotting, respectively. Atorvastatin protected NSC-34D cells against oxidative stress in a concentration-dependent manner. This neuroprotective effect of atorvastatin was blocked by LY294002, a phosphatidylinositol 3-kinase (PI3K) inhibitor and by FR180204, a selective extracellular signal-related kinase (ERK) inhibitor. Atorvastatin treatment increased the expression levels of p85αPI3K, phosphorylated Akt, phosphorylated glycogen synthase kinase-3β, phosphorylated ERK, and Bcl-2, which are proteins related to survival. Furthermore, atorvastatin decreased the levels of cytosolic cytochrome C, Bax, cleaved caspase-9, and cleaved caspase-3, which are associated with death in oxidative stress-injured NSC-34D cells. We conclude that atorvastatin has a protective effect against oxidative stress in motor neurons by activating the PI3K and ERK pathways as well as by scavenging free radicals. These findings indicate that statins could help protect motor neurons from oxidative stress.
Collapse
Affiliation(s)
- Seok-Ho Lee
- Department of Neurology, Hanyang University College of Medicine, 249-1 Gyomun-dong, Guri-si, Gyeonggi-do, South Korea
| | - Na-Young Choi
- Department of Translational Medicine, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea
| | - Hyun-Jeung Yu
- Department of Neurology, Bundang Jesaeng Hospital, Gwangju, Gyeonggi Province, South Korea
| | - Jinse Park
- Department of Neurology, Haeundae Paik Hospital, College of Medicine, Inje University, Busan, South Korea
| | - Hojin Choi
- Department of Neurology, Hanyang University College of Medicine, 249-1 Gyomun-dong, Guri-si, Gyeonggi-do, South Korea
| | - Kyu-Yong Lee
- Department of Neurology, Hanyang University College of Medicine, 249-1 Gyomun-dong, Guri-si, Gyeonggi-do, South Korea
| | - Yong-Min Huh
- Department of Radiology, College of Medicine, Yonsei University, Seoul, South Korea
| | - Young Joo Lee
- Department of Neurology, Hanyang University College of Medicine, 249-1 Gyomun-dong, Guri-si, Gyeonggi-do, South Korea.
| | - Seong-Ho Koh
- Department of Neurology, Hanyang University College of Medicine, 249-1 Gyomun-dong, Guri-si, Gyeonggi-do, South Korea. .,Department of Translational Medicine, Graduate School of Biomedical Science and Engineering, Hanyang University, Seoul, South Korea.
| |
Collapse
|
19
|
Afshordel S, Kern B, Clasohm J, König H, Priester M, Weissenberger J, Kögel D, Eckert GP. Lovastatin and perillyl alcohol inhibit glioma cell invasion, migration, and proliferation--impact of Ras-/Rho-prenylation. Pharmacol Res 2014; 91:69-77. [PMID: 25497898 DOI: 10.1016/j.phrs.2014.11.006] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 11/27/2014] [Accepted: 11/29/2014] [Indexed: 11/19/2022]
Abstract
Alterations in small GTPase mediated signal transduction pathways have emerged as a central step in the molecular pathogenesis of glioblastoma (GBM), the most common malignant brain tumor in adults. Farnesylpyrophosphate (FPP) and geranylgeranylpyrophosphate (GGPP) are derived from mevalonate, whose production is catalyzed by 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA) reductase. Prenylation by FPP and GGPP is required for membrane insertion and oncogenic function of Ras- and Rho-proteins, within the stimulation of the Ras-Raf-MEK-ERK pathway. A straightforward prediction from HMG-CoA reductase inhibitor studies is that statins decrease FPP and GGPP levels and diminish ERK signaling ensuring less proliferation and migration of cancer cells. Perillyl alcohol (POH), a naturally occurring monoterpene inhibits prenyltransferases and is able to inhibit cancer cell growth, but the underlying mechanism is still unclear. We here report that lovastatin (LOV) and POH impair the regulation of the mevalonate- and the Ras-Raf-MEK-ERK pathway in U87 and U343 glioblastoma cells. Both compounds affected the post-translational modification of H-Ras and Rac1. While LOV diminished the substrates of the transferase reaction that catalyze prenylation, POH inhibited the enzymes itself. Our data highlight the impact of isoprenoids for post-translational modification of small GTPases promoting proliferation, migration and invasion capabilities in glioma cells.
Collapse
Affiliation(s)
- Sarah Afshordel
- Department of Pharmacology, Goethe-University, Frankfurt, Germany
| | - Beatrice Kern
- Department of Pharmacology, Goethe-University, Frankfurt, Germany
| | - Jasmin Clasohm
- Department of Pharmacology, Goethe-University, Frankfurt, Germany
| | - Hildegard König
- Division of Experimental Neurosurgery, University Clinics of Frankfurt, Germany
| | - Maike Priester
- Division of Experimental Neurosurgery, University Clinics of Frankfurt, Germany
| | - Jakob Weissenberger
- Division of Experimental Neurosurgery, University Clinics of Frankfurt, Germany
| | - Donat Kögel
- Division of Experimental Neurosurgery, University Clinics of Frankfurt, Germany
| | - Gunter P Eckert
- Department of Pharmacology, Goethe-University, Frankfurt, Germany.
| |
Collapse
|
20
|
Antioxidants Improve the Viability of Stored Adult Retinal Pigment Epithelial-19 Cultures. Ophthalmol Ther 2014; 3:49-61. [PMID: 25134496 PMCID: PMC4254857 DOI: 10.1007/s40123-014-0024-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Indexed: 12/03/2022] Open
Abstract
Introduction There is increasing evidence that retinal pigment epithelium (RPE) can be used to treat age-related macular degeneration, one of the leading causes of blindness worldwide. However, the best way to store RPE to enable worldwide distribution is unknown. We investigated the effects of supplementing our previously published storage method with seven additives, attempting to improve the number of viable adult retinal pigment epithelial (ARPE)-19 cells after storage. Materials and methods ARPE-19 cells were cultured on multiwell plates before being stored for 1 week at 16 °C. Unsupplemented Minimal Essential Medium (MEM) (control) and a total of seven individual additives (DADLE ([d-Ala2, d-Leu5]-encephalin), capsazepine, docosahexaenoic acid (DHA), resveratrol, quercetin, simvastatin and sulforaphane) at three to four concentrations in MEM were tested. The individual effect of each additive on cell viability was analyzed with a microplate fluorometer. Cell phenotype was investigated by both microplate fluorometer and epifluorescence microscopy, and morphology by scanning electron microscopy. Results Supplementation of the storage medium with DADLE, capsazepine, DHA or resveratrol significantly increased the number of viable cells by 86.1% ± 41.9%, 67.9% ± 24.7%, 36.5% ± 10.3% and 21.1% ± 6.4%, respectively, compared to cells stored in unsupplemented MEM. DHA and resveratrol significantly reduced caspase-3 expression, while expression of RPE65 was maintained across groups. Conclusion The number of viable ARPE-19 cells can be increased by the addition of DADLE, capsazepine, DHA or resveratrol to the storage medium without perturbing apoptosis or differentiation. Electronic supplementary material The online version of this article (doi:10.1007/s40123-014-0024-9) contains supplementary material, which is available to authorized users.
Collapse
|
21
|
Wood WG, Mΰller WE, Eckert GP. Statins and Neuroprotection: Basic Pharmacology Needed. Mol Neurobiol 2014; 50:214-20. [DOI: 10.1007/s12035-014-8647-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 01/15/2014] [Indexed: 11/29/2022]
|
22
|
Neuhaus O, Hartung HP. Evaluation of atorvastatin and simvastatin for treatment of multiple sclerosis. Expert Rev Neurother 2014; 7:547-56. [PMID: 17492904 DOI: 10.1586/14737175.7.5.547] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Atorvastatin and simvastatin (members of the 3-hydroxy-3-methylglutaryl coenzyme A reductase inhibitor family) are widely prescribed as cholesterol-lowering agents. As they have been shown to exhibit potent immunomodulatory effects, they may become a future treatment option for autoimmune disease in general and multiple sclerosis (MS) in particular. Several recent reports have demonstrated that statins prevent and reverse chronic and relapsing experimental autoimmune encephalomyelitis, an animal model of MS. An open-label clinical trial assessing simvastatin in MS revealed a significant decrease in the number and volume of new MRI lesions and a favorable safety profile. The results of a large multicenter, placebo-controlled clinical trial assessing atorvastatin in patients with clinically isolated syndrome (a disease that predisposes to development MS) are expected soon. However, prospective placebo-controlled trials of atorvastatin or simvastatin in definite MS are difficult to perform due to ethical and financial objections. In this review, we discuss the backgrounds, mechanisms of action and future perspectives of atorvastatin and simvastatin as putative future treatment options in MS.
Collapse
Affiliation(s)
- Oliver Neuhaus
- Heinrich Heine University, Department of Neurology, Düsseldorf, Germany.
| | | |
Collapse
|
23
|
Shafaroodi H, Moezi L, Fakhrzad A, Hassanipour M, Rezayat M, Dehpour AR. The involvement of nitric oxide in the anti-seizure effect of acute atorvastatin treatment in mice. Neurol Res 2013; 34:847-53. [DOI: 10.1179/1743132812y.0000000080] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Affiliation(s)
- Hamed Shafaroodi
- Department of Pharmacology and ToxicologyPharmaceutical Sciences Branch and Pharmaceutical Sciences Research Center, Islamic Azad University, Tehran, Iran
| | - Leila Moezi
- Department of PharmacologySchool of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Fakhrzad
- Department of PharmacologySchool of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahsa Hassanipour
- Department of Pharmacology and ToxicologyPharmaceutical Sciences Branch and Pharmaceutical Sciences Research Center, Islamic Azad University, Tehran, Iran
| | - Mehdi Rezayat
- Department of Pharmacology and ToxicologyPharmaceutical Sciences Branch and Pharmaceutical Sciences Research Center, Islamic Azad University, Tehran, Iran
| | - Ahmad Reza Dehpour
- Department of PharmacologySchool of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
Cloutier FC, Rouleau DM, Hébert-Davies J, Beaumont PH, Beaumont E. Atorvastatin is beneficial for muscle reinnervation after complete sciatic nerve section in rats. J Plast Surg Hand Surg 2013; 47:446-50. [PMID: 23848426 DOI: 10.3109/2000656x.2013.778205] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Nerve regeneration and functional recovery are often incomplete after peripheral neurotmetic lesion. Atorvastatin has been shown to be neuroprotective after transient ischaemia or traumatic injury. The aim of this study was to establish if systemic administration of Atorvastatin could improve functional muscle reinnervation after complete sciatic nerve section. Sixteen female Sprague-Dawley rats were used in this study. After a complete right sciatic nerve section, end-to-end microsuture repair was performed and fibrin glue was added. Three groups were studied: (1) sutures (S) + fibrin glue (F) only + saline administration for 14 days; (2) S+F+Atorvastatin administration for 14 days; and (3) uninjured nerve. Five months later, the sciatic nerve and the gastrocnemius muscle were isolated to perform in vivo electrophysiological measurements. Better kinematics was observed in atorvastatin-treated rats 5 months after its administration. Indeed, a larger excursion of the hip-ankle-toe angle during walking was observed. This effect was associated with the preservation of electromyographic activity (2.91 mV vs 0.77 mV) and maximal muscle force (85.1 g vs 28.6 g) on stimulation of the proximal nerve section. Five months after a neurotmetic lesion, the recovery is incomplete when using suture and fibrin glue only. Furthermore, the systemic administration of Atorvastatin for 14 days after lesion was beneficial in improving locomotion capability associated with the re-establishment of muscle strength and EMG activity.
Collapse
Affiliation(s)
- Frédéric-Charles Cloutier
- Centre de Recherche, Hôpital du Sacré-Coeur de Montréal and Département de Chirurgie, Université de Montréal , Montréal, Québec , Canada
| | | | | | | | | |
Collapse
|
25
|
Wood WG, Igbavboa U, Muller WE, Eckert GP. Statins, Bcl-2, and apoptosis: cell death or cell protection? Mol Neurobiol 2013; 48:308-14. [PMID: 23821030 DOI: 10.1007/s12035-013-8496-5] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Accepted: 06/16/2013] [Indexed: 01/12/2023]
Abstract
Statins have proven their effectiveness in the treatment of cardiovascular disease. This class of drugs has also attracted attention as a potential treatment for dissimilar diseases such as certain types of cancers and neurodegenerative diseases. What appears to be a contradiction is that, in the case of cancer, it has been suggested that statins increase apoptosis and alter levels of Bcl-2 family members (e.g., reduce Bcl-2 and increase Bax), whereas studies mainly using noncancerous cells report opposite effects. This review examined studies reporting on the effects of statins on Bcl-2 family members, apoptosis, cell death, and cell protection. Much, but not all, of the evidence supporting the pro-apoptotic effects of statins is based on data in cancer cell lines and the use of relatively high drug concentrations. Studies indicating an anti-apoptotic effect of statins are fewer in number and generally used much lower drug concentrations and normal cells. Those conclusions are not definitive, and certainly, there is a need for additional research to determine if statin repositioning is justified for noncardiovascular diseases.
Collapse
Affiliation(s)
- W Gibson Wood
- Department of Pharmacology, Geriatric Research, Education and Clinical Center, VA Medical Center, University of Minnesota School of Medicine, Minneapolis, MN, 55455, USA,
| | | | | | | |
Collapse
|
26
|
Giovannoni G. ECTRIMS 2012: Treatment highlights. Mult Scler Relat Disord 2013; 2:68-72. [PMID: 25877625 DOI: 10.1016/j.msard.2012.12.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Revised: 12/14/2012] [Accepted: 12/14/2012] [Indexed: 11/26/2022]
Affiliation(s)
- Gavin Giovannoni
- Queen Mary University of London, Barts and The London School of Medicine, Newark Street, London E1 2AT, United Kingdom.
| |
Collapse
|
27
|
Statins reduce amyloid β-peptide production by modulating amyloid precursor protein maturation and phosphorylation through a cholesterol-independent mechanism in cultured neurons. Neurochem Res 2012; 38:589-600. [PMID: 23269484 DOI: 10.1007/s11064-012-0956-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Revised: 12/17/2012] [Accepted: 12/19/2012] [Indexed: 01/19/2023]
Abstract
Statins, 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors, have been reported to attenuate amyloid-β peptide (Aβ) production in various cellular models. However, the mechanisms by which statins affect neuronal Aβ production have not yet been clarified. Here, we investigated this issue in rat primary cortical neurons using two statins, pitavastatin (PV) and atorvastatin (AV). Treatment of neurons with 0.2-2.5 μM PV or AV for 4 days induced a concentration- and time-dependent reduction in the secretion of both Aβ40 and Aβ42. Moreover, Western blot analyses of cell lysates showed that treatment with PV or AV significantly reduced expression levels of the mature form of amyloid precursor protein (APP) and Thr668-phosphorylated APP (P-APP), but not immature form of APP; the decreases in P-APP levels were more notable than those of mature APP levels. The statin treatment did not alter expression of BACE1 (β-site APP-cleaving enzyme 1) or γ-secretase complex proteins (presenilin 1, nicastrin, APH-1, and PEN-2). In neurons overexpressing APP via recombinant adenoviruses, PV or AV similarly reduced Aβ secretion and the levels of mature APP and P-APP. Statins also markedly reduced cellular cholesterol content in neurons in a concentration-dependent manner. Co-treatment with mevalonate reversed the statin-induced decreases in Aβ secretion and mature APP and P-APP levels, whereas co-treatment with cholesterol did not, despite recovery of cellular cholesterol levels. Finally, cell-surface biotinylation experiments revealed that both statins significantly reduced the levels of cell-surface P-APP without changing those of cell surface mature APP. These results suggest that statins reduce Aβ production by selectively modulating APP maturation and phosphorylation through a mechanism independent of cholesterol reduction in cultured neurons.
Collapse
|
28
|
Neuroprotective effects of statins: evidence from preclinical and clinical studies. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2012; 14:252-9. [PMID: 22362392 DOI: 10.1007/s11936-012-0174-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
OPINION STATEMENT The benefits of statins for both primary and secondary prevention of ischemic stroke are clearly established. Evidence is accumulating that statin withdrawal after ischemic stroke may lead to worse outcome and that initiation of statins after ischemic stroke may reduce mortality and improve outcome. Current treatment guidelines recommend starting statins before discharge in patients with stroke related to atherosclerosis or who have elevated cholesterol. The primary treatment question then is not if to start statins in most ischemic stroke patients, but when. Our recommendation would be start a statin as soon as the patient passes a dysphagia screen and can safely take oral medication. Based on the results of the Heart Protection Study and the SPARCL trial, either simvastatin 40 mg or atorvastatin 80 mg are appropriate alternatives. Clinical trials are needed to demonstrate unequivocal efficacy of improved outcome and to determine if lower doses may have this effect. Additionally, improved outcome needs to be established in cardioembolic stroke patients before routine use of statins in this stroke subtype can be recommended.
Collapse
|
29
|
Eckert GP, Renner K, Eckert SH, Eckmann J, Hagl S, Abdel-Kader RM, Kurz C, Leuner K, Muller WE. Mitochondrial Dysfunction—A Pharmacological Target in Alzheimer's Disease. Mol Neurobiol 2012; 46:136-50. [DOI: 10.1007/s12035-012-8271-z] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2012] [Accepted: 04/16/2012] [Indexed: 12/12/2022]
|
30
|
Moezi L, Shafaroodi H, Hassanipour M, Fakhrzad A, Hassanpour S, Dehpour AR. Chronic administration of atorvastatin induced anti-convulsant effects in mice: the role of nitric oxide. Epilepsy Behav 2012; 23:399-404. [PMID: 22405864 DOI: 10.1016/j.yebeh.2012.02.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2011] [Revised: 01/25/2012] [Accepted: 02/01/2012] [Indexed: 10/28/2022]
Abstract
Atorvastatin has neuroprotective effects, and there is some evidence that nitric oxide is involved in atorvastatin effects. In this study, we evaluated whether the nitrergic system is involved in the anticonvulsant effects of chronic atorvastatin administration. Intravenous and intraperitoneal pentylenetetrazol were used to induce seizures in mice. Chronic atorvastatin treatment significantly increased the seizure threshold which is induced by both intravenous and intraperitoneal pentylenetetrazol. Intraperitoneal pentylenetetrazol also decreased the incidence of tonic seizure and death in atorvastatin-treated groups. Chronic co-administration of a non-selective nitric oxide synthase inhibitor, l-NAME, or a selective inducible nitric oxide synthase inhibitor, aminoguanidine, with atorvastatin inhibited atorvastatin-induced anticonvulsant effects in intravenous model of pentylenetetrazol. Acute injection of l-NAME or aminoguanidine inhibited the anticonvulsant effects of atorvastatin in both models of intravenous- and intraperitoneal-pentylenetetrazol-induced seizures. In conclusion, we demonstrated that nitric oxide signaling probably through inducible nitric oxide synthase could be involved in the anticonvulsant effects of atorvastatin.
Collapse
Affiliation(s)
- Leila Moezi
- Department of Pharmacology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | | | | | | |
Collapse
|
31
|
Yang D, Han Y, Zhang J, Chopp M, Seyfried DM. Statins Enhance Expression of Growth Factors and Activate the PI3K/Akt-mediated Signaling Pathway after Experimental Intracerebral Hemorrhage. ACTA ACUST UNITED AC 2012; 2:74-80. [PMID: 23482588 DOI: 10.4236/wjns.2012.22011] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Previous studies have demonstrates that statins improve neurological outcome and promote neurovascular recovery after ICH. This study is designed to examine whether simvastatin and atorvastatin affect levels of growth factors and activate the Akt signaling pathway during the recovery phase after intracerebral hemorrhage (ICH) in rats. Sixty (60) male Wistar rats were subjected to ICH by stereotactic injection of 100 μL of autologous blood into the striatum and were treated with or without simvastatin or atorvastatin. Neurological functional outcome was evaluated by behavioral tests (mNSS and corner turn test) at different time points after ICH. Brain extracts were utilized for Enzyme-linked immunosorbent assay (ELISA) analyses to measure vascular endothelial growth factor (VEGF); brain-derived neurotrophin factor (BDNF) expression, and nerve growth factor (NGF). Western blot was used to measure the changes in the Akt-mediated signaling pathway. Both the simvastatin- and atorvastatin-treated animals had significant neurological improvement at 2 weeks post-ICH. Simvastatin and atorvastatin treatment increased the expression of BDNF, VEGF and NGF in both low- and high-dose groups at 7 days after ICH (p < 0.05). Phosphorylation of Akt, glycogen synthase kinase-3β (GSK-3β), and cAMP response element-binding proteins (CREB) were also increased at 7 days after statin treatment. These results suggest that the therapeutic effects of statins after experimental ICH may be mediated by the transient induction of BDNF, VEGF and NGF expression and the activation of the Akt-mediated signaling pathway.
Collapse
Affiliation(s)
- Dongmei Yang
- Department of Neurosurgery, Henry Ford Health System, Detroit MI
| | | | | | | | | |
Collapse
|
32
|
Deepe GS, Buesing WR. Deciphering the pathways of death of Histoplasma capsulatum-infected macrophages: implications for the immunopathogenesis of early infection. THE JOURNAL OF IMMUNOLOGY 2011; 188:334-44. [PMID: 22102723 DOI: 10.4049/jimmunol.1102175] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Apoptosis of leukocytes is known to strongly influence the immunopathogenesis of infection. In this study, we dissected the death pathways of murine macrophages (MΦs) infected with the intracellular pathogen Histoplasma capsulatum. Yeast cells caused apoptosis of MΦs at a wide range of multiplicity of infection, but smaller inocula resulted in delayed detection of apoptosis. Upon infection, caspases 3 and 1 were activated, and both contributed to cell death; however, only the former was involved in apoptosis. The principal driving force for apoptosis involved the extrinsic pathway via engagement of TNFR1 by TNF-α. Infected MΦs produced IL-10 that dampened apoptosis. The chronology of TNF-α and IL-10 release differed in vitro. The former was detected by 2 h postinfection, and the latter was not detected until 8 h postinfection. In vivo, the lungs of TNFR1(-/-) mice infected for 1 d contained fewer apoptotic MΦs than wild-type mice, whereas the lungs of IL-10(-/-) mice exhibited more. Blockade of apoptosis by a pan-caspase inhibitor or by simvastatin sharply reduced the release of TNF-α but enhanced IL-10. However, these treatments did not modify the fungal burden in vitro over 72 h. Thus, suppressing cell death modulated cytokine release but did not alter the fungal burden. These findings provide a framework for the early pathogenesis of histoplasmosis in which yeast cell invasion of lung MΦs engenders apoptosis, triggered in part in an autocrine TNF-α-dependent manner, followed by release of IL-10 that likely prevents apoptosis of newly infected neighboring phagocytes.
Collapse
Affiliation(s)
- George S Deepe
- Medical Service, Veterans Affairs Hospital, Cincinnati, OH 45220, USA.
| | | |
Collapse
|
33
|
Ko ML, Chen CF, Peng PH, Peng YH. Simvastatin upregulates Bcl-2 expression and protects retinal neurons from early ischemia/reperfusion injury in the rat retina. Exp Eye Res 2011; 93:580-5. [DOI: 10.1016/j.exer.2011.07.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 04/23/2011] [Accepted: 07/06/2011] [Indexed: 01/13/2023]
|
34
|
Paz Soldán MM, Pittock SJ, Weigand SD, Yawn BP, Rodriguez M. Statin therapy and multiple sclerosis disability in a population-based cohort. Mult Scler 2011; 18:358-63. [PMID: 21908483 DOI: 10.1177/1352458511421920] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Axonal damage and inflammatory demyelination both occur in multiple sclerosis (MS). Some studies suggest that statins, through pleiotropic effects, reduce inflammatory episodes and protect neurons. However, other studies suggest statins have disparate impacts on these pathologic processes. OBJECTIVE The objective of this study was to assess disability progression in MS patients receiving statin therapy. METHODS We performed a retrospective medical record review of an established population-based MS prevalence cohort in Olmsted County, Minnesota, comparing disability progression between patients receiving statins and controls. RESULTS Duration of statin use ranged from 1.9 to 20.3 years with a mean and standard deviation of 6.8 ± 4 years. Years between assessments ranged from 0.6 to 8.2 (75% of patients having intervals >6.4 years). The median (interquartile range) absolute change of disability among the statin group was 0 (0 to +1), compared with +0.5 (0, +1) in the no-statin group. Distributions were not significantly different (p = 0.39). The mean (standard deviation) absolute change of disability scores among the statin group was +0.69 (+1.49), not significantly different from +0.61 (+1.31) in the no-statin group. Likewise, annualized disability scores did not differ significantly (p = 0.23). Eighteen (40%) patients worsened by 1.0 or more on Expanded Disability Status Scale (EDSS) in the statin group and 36 (40%) in the no-statin group (p = 0.85, chi-squared test). CONCLUSIONS In this cohort, disability progression did not differ between those receiving statin therapy and controls. These findings support the hypothesis that statins, in doses currently prescribed for hyperlipidemia, do not affect the long-term course of MS.
Collapse
|
35
|
Bull ND, Johnson TV, Welsapar G, DeKorver NW, Tomarev SI, Martin KR. Use of an adult rat retinal explant model for screening of potential retinal ganglion cell neuroprotective therapies. Invest Ophthalmol Vis Sci 2011; 52:3309-20. [PMID: 21345987 DOI: 10.1167/iovs.10-6873] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
PURPOSE. To validate an established adult organotypic retinal explant culture system for use as an efficient medium-throughput screening tool to investigate novel retinal ganglion cell (RGC) neuroprotective therapies. METHODS. Optimal culture conditions for detecting RGC neuroprotection in rat retinal explants were identified. Retinal explants were treated with various recognized, or purported, neuroprotective agents and cultured for either 4 or 7 days ex vivo. The number of cells surviving in the RGC layer (RGCL) was quantified using histologic and immunohistochemical techniques, and statistical analyses were applied to detect neuroprotective effects. RESULTS. The ability to replicate previously reported in vivo RGC neuroprotection in retinal explants was verified by demonstrating that caspase inhibition, brain-derived neurotrophic factor treatment, and stem cell transplantation all reduced RGCL cell loss in this model. Further screening of potential neuroprotective pharmacologic agents demonstrated that betaxolol, losartan, tafluprost, and simvastatin all alleviated RGCL cell loss in retinal explants, supporting previous reports. However, treatment with brimonidine did not protect RGCL neurons from death in retinal explant cultures. Explants cultured for 4 days ex vivo proved most sensitive for detecting neuroprotection. CONCLUSIONS. The current adult rat retinal explant culture model offers advantages over other models for screening potential neuroprotective drugs, including maintenance of neurons in situ, control of environmental conditions, and dissociation from other factors such as intraocular pressure. Verification that neuroprotection by previously identified RGC-protective therapies could be replicated in adult retinal explant cultures suggests that this model could be used for efficient medium-throughput screening of novel neuroprotective therapies for retinal neurodegenerative disease.
Collapse
Affiliation(s)
- Natalie D Bull
- Cambridge Centre for Brain Repair, University of Cambridge, United Kingdom
| | | | | | | | | | | |
Collapse
|
36
|
Ford AL, An H, D’Angelo G, Ponisio R, Bushard P, Vo KD, Powers WJ, Lin W, Lee JM. Preexisting statin use is associated with greater reperfusion in hyperacute ischemic stroke. Stroke 2011; 42:1307-13. [PMID: 21454815 PMCID: PMC3125694 DOI: 10.1161/strokeaha.110.600957] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2010] [Accepted: 11/30/2010] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Statin pretreatment has been associated with improved outcomes in patients with ischemic stroke. Although several mechanisms have been examined in animal models, few have been examined in patients. We hypothesized that patients using statins before stroke onset may have greater reperfusion than patients not using statins. METHODS Acute ischemic stroke patients underwent 2 MR scans: within 4.5 (tp1) and at 6 hours (tp2) after stroke onset. Regions of reperfusion were defined by prolonged mean transit time (MTT) at tp1, which normalized at tp2. Four MTT thresholds were assessed to ensure that results were not spuriously based on an arbitrary threshold. Baseline characteristics, relative reperfusion, and change in NIHSS between tp1 and 1-month follow-up (ΔNIHSS) were compared between patients who were using statins at stroke onset and those who were not. RESULTS Thirty-one stroke patients were prospectively enrolled; 12 were using statins and 19 were not. Baseline characteristics did not differ between the 2 groups except the statin group had greater coronary artery disease (P=0.03). Patients using statins showed significantly greater reperfusion compared to untreated patients across all MTT thresholds. For MTT of 4 seconds, median relative reperfusion was 50% (interquartile range, 30%-56%) in the preexisting statin group versus 13% (interquartile range, 5%-36%) in the untreated group (P=0.014). The statin group had greater ΔNIHSS (8.8±4.0 points) compared to the untreated group (4.4±5.7 points; P=0.028). CONCLUSIONS Statin use before ischemic stroke onset was associated with greater early reperfusion and NIHSS improvement. Further studies in larger populations are required to confirm our preliminary findings.
Collapse
Affiliation(s)
- Andria L. Ford
- Department of Neurology, Washington University, School of Medicine
| | - Hongyu An
- Department of Radiology, University of North Carolina at Chapel Hill
| | - Gina D’Angelo
- Division of Biostatistics, Washington University, School of Medicine
| | - Rosana Ponisio
- Department of Radiology, Washington University, School of Medicine
| | - Patrick Bushard
- Department of Neurology, Washington University, School of Medicine
| | - Katie D. Vo
- Department of Radiology, Washington University, School of Medicine
| | - William J. Powers
- Department of Neurology, University of North Carolina at Chapel Hill
| | - Weili Lin
- Department of Radiology, University of North Carolina at Chapel Hill
| | - Jin-Moo Lee
- Department of Neurology, Washington University, School of Medicine
| |
Collapse
|
37
|
Awad AM, Sellner J, Hemmer B, Stüve O. Role of statins in the treatment of multiple sclerosis: an update. Neurodegener Dis Manag 2011. [DOI: 10.2217/nmt.11.13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
SUMMARY This article discusses the role of statins in the treatment of multiple sclerosis. Statins are promising agents as adjunctive therapies in immune-mediated disorders such as multiple sclerosis owing to their immunomodulatory, anti-inflammtory and neuroprotective characteristics. On the other hand, there are some conflicting data that warrant further investigation before sound conclusions can be made. Clinical trials are planned and being conducted to help shed more light on the potential benefit and the optimal dose of statins in treating multiple sclerosis.
Collapse
Affiliation(s)
- Amer M Awad
- Baton Rouge Neurology Associates, Baton Rouge General Medical Center, Baton Rouge, LA, USA
| | - Johann Sellner
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
- Department of Neurology, Christian-Doppler-Klinik, Paracelsus Medical University, Salzburg, Austria
| | - Bernhard Hemmer
- Department of Neurology, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Olaf Stüve
- Department of Neurology, University of Texas Southwestern Medical Center at Dallas, TX, USA
- Neurology Section, VA North Texas Health Care System, Medical Service, Dallas, TX, USA
| |
Collapse
|
38
|
Bartolomé F, Muñoz Ú, Esteras N, Alquezar C, Collado A, Bermejo-Pareja F, Martín-Requero Á. Simvastatin overcomes the resistance to serum withdrawal-induced apoptosis of lymphocytes from Alzheimer's disease patients. Cell Mol Life Sci 2010; 67:4257-68. [PMID: 20614159 PMCID: PMC11115769 DOI: 10.1007/s00018-010-0443-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2010] [Revised: 05/20/2010] [Accepted: 06/21/2010] [Indexed: 10/19/2022]
Abstract
Statins may exert beneficial effects on Alzheimer's disease (AD) patients. Based on the antineoplastic and apoptotic effects of statins in a number of cell types, we hypothesized that statins may be able to protect neurons by controlling the regulation of cell cycle and/or apoptosis. A growing body of evidence indicates that neurodegeneration involves the cell-cycle activation in postmitotic neurons. Failure of cell-cycle control is not restricted to neurons in AD patients, but occurs in peripheral cells as well. For these reasons, we studied the role of simvastatin (SIM) on cell survival/death in lymphoblasts from AD patients. We report here that SIM induces apoptosis in AD lymphoblasts deprived of serum. SIM interacts with PI3K/Akt and ERK1/2 signaling pathways thereby decreasing the serum withdrawal-enhanced levels of the CDK inhibitor p21(Cip1) (p21) and restoring the vulnerability of AD cells to trophic factor deprivation.
Collapse
Affiliation(s)
- Fernando Bartolomé
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Úrsula Muñoz
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
- Present Address: Mount Sinai School of Medicine, 1425 Madison Avenue, New York, NY 10029 USA
| | - Noemí Esteras
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Carolina Alquezar
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Ramiro de Maéztu 9, 28040 Madrid, Spain
| | - Andrea Collado
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Félix Bermejo-Pareja
- Hospital Doce de Octubre, Avda de Córdoba s/n, 28041 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Avda de Córdoba s/n, 28041 Madrid, Spain
| | - Ángeles Martín-Requero
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Ramiro de Maéztu 9, 28040 Madrid, Spain
| |
Collapse
|
39
|
Liposome-incorporated DHA increases neuronal survival by enhancing non-amyloidogenic APP processing. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2010; 1808:236-43. [PMID: 21036142 DOI: 10.1016/j.bbamem.2010.10.014] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2010] [Revised: 10/01/2010] [Accepted: 10/25/2010] [Indexed: 12/26/2022]
Abstract
The fluidity of neuronal membranes plays a pivotal role in brain aging and neurodegeneration. In this study, we investigated the role of the omega-3 fatty acid docosahexaenoic acid (DHA) in modulation of membrane fluidity, APP processing, and protection from cytotoxic stress. To this end, we applied unilamellar transfer liposomes, which provided protection from oxidation and effective incorporation of DHA into cell membranes. Liposomes transferring docosanoic acid (DA), the completely saturated form of DHA, to the cell cultures served as controls. In HEK-APP cells, DHA significantly increased membrane fluidity and non-amyloidogenic processing of APP, leading to enhanced secretion of sAPPα. This enhanced secretion of sAPPα was associated with substantial protection against apoptosis induced by ER Ca(2+) store depletion. sAPPα-containing supernatants obtained from HEK-APP cells exerted similar protective effects as DHA in neuronal PC12 cells and HEK293 control cells. Correlating to further increased sAPPα levels, supernatants obtained from DHA-treated HEK-APP cells enhanced protection, whereas supernatants obtained from DHA-treated HEK293 control cells did not inhibit apoptosis, likely due to the low expression of endogenous APP and negligible sAPPα secretion in these cells. Further experiments with the small molecule inhibitors LY294002 and SP600125 indicated that sAPPα-induced cytoprotection relied on activation of the anti-apoptotic PI3K/Akt pathway and inhibition of the stress-triggered JNK signaling pathway in PC12 cells. Our data suggest that liposomal DHA is able to restore or maintain physiological membrane properties, which are required for neuroprotective sAPPα secretion and autocrine modulation of neuronal survival.
Collapse
|
40
|
Kalonia H, Kumar P, Kumar A. Comparative neuroprotective profile of statins in quinolinic acid induced neurotoxicity in rats. Behav Brain Res 2010; 216:220-8. [PMID: 20696189 DOI: 10.1016/j.bbr.2010.07.040] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Revised: 07/28/2010] [Accepted: 07/31/2010] [Indexed: 01/09/2023]
Abstract
A possible neuroprotective role has been recently suggested for 3H3MGCoA reductase inhibitors (statins). Here, we sought to determine neuroprotective effect of statins in quinolinic acid induced neurotoxicity in rats. Rats were surgically administered quinolinic acid and treated with Atorvastatin (10, 20 mg/kg), simvastatin (15, 30 mg/kg) and fluvastatin (5, 10 mg/kg) once daily up to 3 weeks. Atorvastatin (10, 20 mg/kg), simvastatin (30 mg/kg) and fluvastatin (10 mg/kg) treatment significantly attenuated the quinolinic acid induced behavioral (locomotor activity, rotarod performance and beam walk test), biochemical (lipid peroxidation, nitrite concentration, SOD and catalase), mitochondrial enzyme complex alterations in rats suggesting their free radical scavenging potential. Additionally, atorvastatin (10, 20 mg/kg), simvastatin (30 mg/kg) and fluvastatin (10 mg/kg) significantly decrease the TNF-α level and striatal lesion volume in quinolinic acid treated animals indicating their anti-inflammatory effects. In comparing the protective effect of different statins, atorvastatin is effective at both the doses while simvastatin and fluvastatins at respective lower doses were not able to produce the protective effect in quinolinic acid treated animals. These modulations can account, at least partly, for the beneficial effect of statins in our rodent model of striatal degeneration. Our findings show that statins could be explored as possible neuroprotective agents for neurodegenerative disorders such as HD.
Collapse
Affiliation(s)
- Harikesh Kalonia
- Pharmacology Division, University Institute of Pharmaceutical Sciences, UGC Centre of Advanced Study, Panjab University, Chandigarh 160014, India
| | | | | |
Collapse
|
41
|
Kuffler DP. Combinatorial techniques for enhancing neuroprotection: hypothermia and alkalinization. Ann N Y Acad Sci 2010; 1199:164-74. [PMID: 20633122 DOI: 10.1111/j.1749-6632.2009.05353.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Brain and spinal cord (CNS) trauma typically kill a number of neurons, but even more neurons are killed by secondary causes triggered by the initial trauma. Thus, a minor insult may rapidly cause the death of a vastly larger number of neurons and complete paralysis. The best mechanism for reducing the extent of neurological deficits is to minimize the number of neurons killed by post-trauma sequelae. Neuroprotection techniques take many diverse forms with a breadth too great for a short review. Therefore, this review focuses on the neuroprotection provided by hypothermia and a number of other neuroprotective techniques, when administered singly or in combination, because it is generally found that combinations of applications lead to significantly better neuroprotection than is achieved by any one alone. The combinatorial approach to neuroprotection holds great promise for enhancing the degree of neuroprotection following trauma, leading to maximum maintenance of neurological function.
Collapse
Affiliation(s)
- Damien P Kuffler
- Institute of Neurobiology, Medical Sciences Campus, University of Puerto Rico, San Juan.
| |
Collapse
|
42
|
Wood WG, Eckert GP, Igbavboa U, Müller WE. Statins and neuroprotection: a prescription to move the field forward. Ann N Y Acad Sci 2010; 1199:69-76. [PMID: 20633110 DOI: 10.1111/j.1749-6632.2009.05359.x] [Citation(s) in RCA: 108] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
There is growing interest in the use of statins, HMG-CoA reductase inhibitors, for treating specific neurodegenerative diseases (e.g., cerebrovascular disease, Parkinson's disease, Alzheimer's disease, multiple sclerosis) and possibly traumatic brain injury. Neither is there a consensus on the efficacy of statins in treating the aforementioned diseases nor are the mechanisms of the purported statin-induced neuroprotection well-understood. Part of the support for statin-induced neuroprotection comes from studies using animal models and cell culture. Important information has resulted from that work but there continues to be a lack of progress on basic issues pertaining to statins and brain that impedes advancement in understanding how statins alter brain function. For example, there are scant data on the pharmacokinetics of lipophilic and hydrophilic statins in brain, statin-induced neuroprotection versus cell death, and statins and brain isoprenoids. The purpose of this mini-review will be to examine those aforementioned issues and to identify directions of future research.
Collapse
Affiliation(s)
- W Gibson Wood
- Department of Pharmacology, University of Minnesota, Minneapolis, Minnesota 55455, USA.
| | | | | | | |
Collapse
|
43
|
Cholesterol and statins in Alzheimer's disease: Current controversies. Exp Neurol 2010; 223:282-93. [DOI: 10.1016/j.expneurol.2009.09.013] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2009] [Revised: 09/16/2009] [Accepted: 09/17/2009] [Indexed: 02/07/2023]
|
44
|
Butterick TA, Igbavboa U, Eckert GP, Sun GY, Weisman GA, Müller WE, Wood WG. Simvastatin stimulates production of the antiapoptotic protein Bcl-2 via endothelin-1 and NFATc3 in SH-SY5Y cells. Mol Neurobiol 2010; 41:384-91. [PMID: 20369390 PMCID: PMC3075856 DOI: 10.1007/s12035-010-8122-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2010] [Accepted: 03/17/2010] [Indexed: 01/11/2023]
Abstract
The use of statins for the prevention or treatment of different neurodegenerative diseases has generated considerable interest albeit with some controversy. Mechanisms of statin-induced neuroprotection are not well understood. Recently, we reported that simvastatin stimulated neuronal gene expression and protein levels of the major antiapoptotic protein Bcl-2 in vivo and in vitro; suppression of Bcl-2 in SH-SY5Y cells reduced simvastatin neuroprotection; effects were independent of cholesterol and other products of the 3-hydroxy-3-methylglutaryl-CoA reductase pathway. Endothelin-1 (ET-1) can increase Bcl-2 abundance via the transcription factor nuclear factor of activated thymocytes (NFATc), and simvastatin was reported to increase ET-1 gene expression. We tested the hypothesis that simvastatin stimulation of Bcl-2 involves up-regulation of ET-1 and binding of NFATc to Bcl-2 promoter sites in SH-SY5Y human neuroblastoma cells. Simvastatin increased both intracellular and secreted ET-1 protein levels. Exogenous ET-1 increased Bcl-2 protein abundance, which was inhibited by ET-1 receptor antagonists. Simvastatin increased translocation of NFATc3 to the nucleus while reducing nuclear NFATc1 and having no effect on NFATc4. Endothelin-1 also increased NFATc3 levels in the nucleus, and this increase was inhibited by ET-1 receptor antagonists. Treatment of cells with simvastatin stimulated binding of NFATc3 to the Bcl-2 promoter. We report novel findings showing that up-regulation of Bcl-2 by simvastatin involves ET-1 and the transcription factor NFATc3. Discovering how statins can selectively alter a specific NFATc isoform that leads to an increase in an antiapoptotic protein will provide a new approach to understanding statin-induced neuroprotection and conditions outside the brain in which apoptosis contributes to pathophysiology.
Collapse
Affiliation(s)
- Tammy A. Butterick
- Department of Pharmacology, Geriatric Research Education and Clinical Center, VA Medical Center, University of Minnesota, Minneapolis, MN, USA
| | - Urule Igbavboa
- Department of Pharmacology, Geriatric Research Education and Clinical Center, VA Medical Center, University of Minnesota, Minneapolis, MN, USA
| | - Gunter P. Eckert
- Department of Pharmacology, BiocenterNiederursel, Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt, Germany
| | - Grace Y. Sun
- Department of Biochemistry, Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, Canada
| | - Gary A. Weisman
- Department of Biochemistry, Bond Life Sciences Center, University of Missouri, Columbia, MO 65211, Canada
| | - Walter E. Müller
- Department of Pharmacology, BiocenterNiederursel, Goethe University, Max-von-Laue-Str. 9, 60438 Frankfurt, Germany
| | - W. Gibson Wood
- Department of Pharmacology, Geriatric Research Education and Clinical Center, VA Medical Center, University of Minnesota, Minneapolis, MN, USA. Department of Pharmacology, University of Minnesota, 6-120 Jackson Hall, 321 Church Street, SE, Minneapolis, MN 55455, USA
| |
Collapse
|
45
|
Müller WE, Eckert A, Kurz C, Eckert GP, Leuner K. Mitochondrial dysfunction: common final pathway in brain aging and Alzheimer's disease--therapeutic aspects. Mol Neurobiol 2010; 41:159-71. [PMID: 20461558 DOI: 10.1007/s12035-010-8141-5] [Citation(s) in RCA: 175] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2010] [Accepted: 04/15/2010] [Indexed: 12/21/2022]
Abstract
As a fully differentiated organ, our brain is very sensitive to cumulative oxidative damage of proteins, lipids, and DNA occurring during normal aging because of its high energy metabolism and the relative low activity of antioxidative defense mechanisms. As a major consequence, perturbations of energy metabolism including mitochondrial dysfunction, alterations of signaling mechanisms and of gene expression culminate in functional deficits. With the increasing average life span of humans, age-related cognitive disorders such as Alzheimer's disease (AD) are a major health concern in our society. Age-related mitochondrial dysfunction underlies most neurodegenerative diseases, where it is potentiated by disease-specific factors. AD is characterized by two major histopathological hallmarks, initially intracellular and with the progression of the disease extracellular accumulation of oligomeric and fibrillar beta-amyloid peptides and intracellular neurofibrillary tangles composed of hyperphosphorylated tau protein. In this review, we focus on findings in AD animal and cell models indicating that these histopathological alterations induce functional deficits of the respiratory chain complexes and therefore consecutively result in mitochondrial dysfunction and oxidative stress. These parameters lead synergistically with the alterations of the brain aging process to typical signs of neurodegeneration in the later state of the disease, including synaptic dysfunction, loss of synapses and neurites, and finally neuronal loss. We suggest that mitochondrial protection and subsequent reduction of oxidative stress are important targets for prevention and long-term treatment of early stages of AD.
Collapse
Affiliation(s)
- Walter E Müller
- Department of Pharmacology, Biocenter, University of Frankfurt, Max-von Laue-Strasse 9, 60438, Frankfurt, Germany.
| | | | | | | | | |
Collapse
|
46
|
Bil J, Zapala L, Nowis D, Jakobisiak M, Golab J. Statins potentiate cytostatic/cytotoxic activity of sorafenib but not sunitinib against tumor cell lines in vitro. Cancer Lett 2010; 288:57-67. [DOI: 10.1016/j.canlet.2009.06.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Revised: 06/20/2009] [Accepted: 06/23/2009] [Indexed: 12/11/2022]
|
47
|
Abstract
Cytoprotective or neuroprotective interventions would be of value if they could block the processes leading to delayed neuronal death or if they could delay the period between the onset of ischemia and irreversible necrotic injury, thereby lengthening the period for effective reperfusion therapy. Experimental studies in cell culture systems and laboratory animals show that statins have several potential cytoprotective actions, including promotion of angiogenesis, reduction of clot formation and facilitation of clot lysis, upregulation of endothelial nitric oxide synthase, downregulation of inducible nitric oxide synthase, reduction of excitotoxicity, and modulation of the inflammatory response. Clinically, statins appear to protect against vasospasm-related ischemic injury after subarachnoid hemorrhage. There have been no prospective randomized trials aimed at determining whether statins reduce acute stroke severity, and observational studies have had inconsistent results. Although a prospective, randomized trial assessing the effect of pre- or poststroke statin treatment on initial stroke severity would be the most appropriate study design to test for this type of effect, it is unlikely that such a trial will be conducted given the benefits of these drugs in reducing the risk of cardiovascular events and stroke in high-risk populations.
Collapse
Affiliation(s)
- Larry B Goldstein
- Department of Medicine (Neurology), Duke Stroke Center, Duke University Medical Center, Durham, NC 27710, USA.
| |
Collapse
|
48
|
Simvastatin improves cerebrovascular function and counters soluble amyloid-beta, inflammation and oxidative stress in aged APP mice. Neurobiol Dis 2009; 35:406-14. [DOI: 10.1016/j.nbd.2009.06.003] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2009] [Revised: 05/29/2009] [Accepted: 06/04/2009] [Indexed: 11/18/2022] Open
|
49
|
Igbavboa U, Sun GY, Weisman GA, He Y, Wood WG. Amyloid beta-protein stimulates trafficking of cholesterol and caveolin-1 from the plasma membrane to the Golgi complex in mouse primary astrocytes. Neuroscience 2009; 162:328-38. [PMID: 19401218 DOI: 10.1016/j.neuroscience.2009.04.049] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2009] [Revised: 04/03/2009] [Accepted: 04/20/2009] [Indexed: 11/19/2022]
Abstract
The Golgi complex plays a key role in cholesterol trafficking in cells. Our earlier study demonstrated amyloid beta-protein (Abeta) alters cholesterol distribution and abundance in the Golgi complex of astrocytes. We now test the hypothesis that the Abeta-induced increase in Golgi complex cholesterol is due to retrograde movement of the cholesterol carrier protein caveolin-1 from the cell plasma membrane to the Golgi complex in astrocytes. Results with mouse primary astrocytes indicated that Abeta(1-42)-induced increase in cholesterol and caveolin abundance in the Golgi complex was accompanied by a reduction in cholesterol and caveolin levels in the plasma membrane. Transfected rat astrocytes (DITNC1) with siRNA directed at caveolin-1 mRNA inhibited the Abeta(1-42)-induced redistribution of both cholesterol and caveolin from the plasma membrane to the Golgi complex. In astrocytes not treated with Abeta(1-42), suppression of caveolin-1 expression also significantly reduced cholesterol abundance in the Golgi complex, further demonstrating the role for caveolin in retrograde transport of cholesterol from the plasma membrane to the Golgi complex. Perturbation of this process by Abeta(1-42) could have consequences on membrane structure and cellular functions requiring optimal levels of cholesterol.
Collapse
Affiliation(s)
- U Igbavboa
- Department of Pharmacology, University of Minnesota School of Medicine and Geriatric Research, Education and Clinical Center, VA Medical Center, 6-120 Jackson Hall, 321 Church Street Southeast, Minneapolis, MN 55455, USA.
| | | | | | | | | |
Collapse
|
50
|
Paintlia AS, Paintlia MK, Singh I, Skoff RB, Singh AK. Combination therapy of lovastatin and rolipram provides neuroprotection and promotes neurorepair in inflammatory demyelination model of multiple sclerosis. Glia 2009; 57:182-93. [PMID: 18720408 DOI: 10.1002/glia.20745] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Drug combination therapies for central nervous system (CNS) demyelinating diseases including multiple sclerosis (MS) are gaining momentum over monotherapy. Over the past decade, both in vitro and in vivo studies established that statins (HMG-CoA reductase inhibitors) and rolipram (phosphodiesterase-4 inhibitor; blocks the degradation of intracellular cyclic AMP) can prevent the progression of MS in affected individuals via different mechanisms of action. In this study, we evaluated the effectiveness of lovastatin (LOV) and rolipram (RLP) in combination therapy to promote neurorepair in an inflammatory CNS demyelination model of MS, experimental autoimmune encephalomyelitis (EAE). Combination treatment with suboptimal doses of these drugs in an established case of EAE (clinical disease score > or = 2.0) significantly attenuated the infiltration of inflammatory cells and protected myelin sheath and axonal integrity in the CNS. It was accompanied with elevated level of cyclic AMP and activation of its associated protein kinase A. Interestingly, combination treatment with these drugs impeded neurodegeneration and promoted neurorepair in established EAE animals (clinical disease score > or = 3.5) as verified by quantitative real-time polymerase chain reaction, immunohistochemistry and electron microscopic analyses. These effects of combination therapy were minimal and/or absent with either drug alone in these settings. Together, these data suggest that combination therapy with LOV and RLP has the potential to provide neuroprotection and promote neurorepair in MS, and may have uses in other related CNS demyelinating diseases.
Collapse
Affiliation(s)
- Ajaib S Paintlia
- Department of Pediatrics, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | |
Collapse
|