1
|
Weekman EM, Johnson SN, Rogers CB, Sudduth TL, Xie K, Qiao Q, Fardo DW, Bottiglieri T, Wilcock DM. Atorvastatin rescues hyperhomocysteinemia-induced cognitive deficits and neuroinflammatory gene changes. J Neuroinflammation 2023; 20:199. [PMID: 37658433 PMCID: PMC10474691 DOI: 10.1186/s12974-023-02883-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 08/28/2023] [Indexed: 09/03/2023] Open
Abstract
BACKGROUND Epidemiological data suggests statins could reduce the risk of dementia, and more specifically, Alzheimer's disease (AD). Pre-clinical data suggests statins reduce the risk of dementia through their pleiotropic effects rather than their cholesterol lowering effects. While AD is a leading cause of dementia, it is frequently found co-morbidly with cerebral small vessel disease and other vascular contributions to cognitive impairment and dementia (VCID), which are another leading cause of dementia. In this study, we determined if atorvastatin ameliorated hyperhomocysteinemia (HHcy)-induced VCID. METHODS Wild-type (C57Bl6/J) mice were placed on a diet to induce HHcy or a control diet each with or without atorvastatin for 14 weeks. Mice underwent novel object recognition testing before tissue collection. Plasma total cholesterol and total homocysteine as well as related metabolites were measured. Using qPCR and NanoString technology, we profiled glial cell-associated gene expression changes. Finally, microglial morphology, astrocyte end feet, and microhemorrhages were analyzed using histological methods. RESULTS Atorvastatin treatment of HHcy in mice led to no changes in total cholesterol but decreases in total homocysteine in plasma. While HHcy decreased expression of many glial genes, atorvastatin rescued these gene changes, which mostly occurred in oligodendrocytes and microglia. Microglia in HHcy mice with atorvastatin were trending towards fewer processes compared to control with atorvastatin, but there were no atorvastatin effects on astrocyte end feet. While atorvastatin treatment was trending towards increasing the area of microhemorrhages in HHcy mice in the frontal cortex, it only slightly (non-significantly) reduced the number of microhemorrhages. Finally, atorvastatin treatment in HHcy mice led to improved cognition on the novel object recognition task. CONCLUSIONS These data suggest that atorvastatin rescued cognitive changes induced by HHcy most likely through lowering plasma total homocysteine and rescuing gene expression changes rather than impacts on vascular integrity or microglial changes.
Collapse
Affiliation(s)
- Erica M Weekman
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA.
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| | - Sherika N Johnson
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Colin B Rogers
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
| | - Tiffany L Sudduth
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
| | - Kevin Xie
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
| | - Qi Qiao
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
| | - David W Fardo
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
| | - Teodoro Bottiglieri
- Center of Metabolomics, Institute of Metabolic Disease, Baylor Scott and White Research Institute, Dallas, TX, 75204, USA
| | - Donna M Wilcock
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, 40536, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| |
Collapse
|
2
|
Butterfield DA, Boyd-Kimball D, Reed TT. Cellular Stress Response (Hormesis) in Response to Bioactive Nutraceuticals with Relevance to Alzheimer Disease. Antioxid Redox Signal 2023; 38:643-669. [PMID: 36656673 PMCID: PMC10025851 DOI: 10.1089/ars.2022.0214] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/08/2023] [Indexed: 01/20/2023]
Abstract
Significance: Alzheimer's disease (AD) is the most common form of dementia associated with aging. As the large Baby Boomer population ages, risk of developing AD increases significantly, and this portion of the population will increase significantly over the next several decades. Recent Advances: Research suggests that a delay in the age of onset by 5 years can dramatically decrease both the incidence and cost of AD. In this review, the role of nuclear factor erythroid 2-related factor 2 (Nrf2) in AD is examined in the context of heme oxygenase-1 (HO-1) and biliverdin reductase-A (BVR-A) and the beneficial potential of selected bioactive nutraceuticals. Critical Issues: Nrf2, a transcription factor that binds to enhancer sequences in antioxidant response elements (ARE) of DNA, is significantly decreased in AD brain. Downstream targets of Nrf2 include, among other proteins, HO-1. BVR-A is activated when biliverdin is produced. Both HO-1 and BVR-A also are oxidatively or nitrosatively modified in AD brain and in its earlier stage, amnestic mild cognitive impairment (MCI), contributing to the oxidative stress, altered insulin signaling, and cellular damage observed in the pathogenesis and progression of AD. Bioactive nutraceuticals exhibit anti-inflammatory, antioxidant, and neuroprotective properties and are potential topics of future clinical research. Specifically, ferulic acid ethyl ester, sulforaphane, epigallocatechin-3-gallate, and resveratrol target Nrf2 and have shown potential to delay the progression of AD in animal models and in some studies involving MCI patients. Future Directions: Understanding the regulation of Nrf2 and its downstream targets can potentially elucidate therapeutic options for delaying the progression of AD. Antioxid. Redox Signal. 38, 643-669.
Collapse
Affiliation(s)
- D. Allan Butterfield
- Department of Chemistry, University of Kentucky, Lexington, Kentucky, USA
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, USA
| | - Debra Boyd-Kimball
- Department of Biochemistry, Chemistry, and Physics, University of Mount Union, Alliance, Ohio, USA
| | - Tanea T. Reed
- Department of Chemistry, Eastern Kentucky University, Richmond, Kentucky, USA
| |
Collapse
|
3
|
Pre-treatment and continuous administration of simvastatin during sepsis improve metabolic parameters and prevent CNS injuries in survivor rats. Mol Cell Biochem 2022; 477:2657-2667. [DOI: 10.1007/s11010-022-04463-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Accepted: 04/28/2022] [Indexed: 10/18/2022]
|
4
|
Kukula O, Günaydın C. Atorvastatin reduces alloxan-induced impairment of aversive stimulus memory in mice. ASIAN BIOMED 2022; 16:71-78. [PMID: 37551286 PMCID: PMC10321169 DOI: 10.2478/abm-2022-0009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Background An association between dysregulated glucose levels in patients with diabetes mellitus and detrimental effects on the central nervous system, particularly in Alzheimer disease, has been recognized. Atorvastatin treatment has improved memory and cognition in some patients with diabetes mellitus and Alzheimer disease. Objectives To determine possible neuroprotective effects of atorvastatin on memory and cognition by measuring changes in an adverse stimulus avoidance learning deficit induced by alloxan in a murine model of diabetes mellitus and impaired memory and cognition. Methods We administered 150 mg/kg and 100 mg/kg alloxan in saline (intraperitoneally, i.p.) at a 48 h interval to produce a model of diabetes mellitus in male BALB/c mice. An oral glucose tolerance test (OGTT) was used to assess blood glucose regulation. After demonstrating hyperglycemia in mice (n = 7 per group) we administered vehicle (saline, i.p.), atorvastatin (10 mg/kg, i.p.), or liraglutide (200 μg/kg, i.p.) for 28 d except for those in a negative control group, which were given saline instead of alloxan, and a group administered atorvastatin alone, which were given saline instead of alloxan followed by atorvastatin (10 mg/kg, i.p.) for 28 d. Locomotor activity was measured 24 h after the final drug treatments, and subsequently their learned behavioral response to an adverse electrical stimulus to their plantar paw surface in a dark compartment was measured using a passive avoidance apparatus (Ugo Basile) in a model of impaired memory and cognition associated with Alzheimer disease. To determine any deficit in their learned avoidance of the adverse stimulus, we measured the initial latency or time mice spent in an illuminated white compartment before entering the dark compartment in the learning trial, and on the day after learning to avoid the adverse stimulus, the retention period latency in the light compartment and time spent in the dark compartment. Results Atorvastatin alone produced no significant change in blood glucose levels (F4,10 = 0.80, P = 0.55) within 2 h. Liraglutide decreased blood glucose levels after 0.5 h (F4,10 = 11.7, P < 0.001). We found no significant change in locomotor activity in any group. In mice with alloxan-induced diabetes, atorvastatin significantly attenuated the decreased avoidance associated with the diabetes (F4,30 = 38.0, P = 0.02) and liraglutide also significantly attenuated the decreased avoidance (F4,30 = 38.0, P < 0.001). Atorvastatin alone had no significant effect on the adversive learned response compared with vehicle treatment (F4,30 = 38.0, P > 0.05). Atorvastatin significantly decreased the time mice with alloxan-induced diabetes spent in the dark compartment compared with mice in the diabetes group without atorvastatin treatment (F4,30 = 53.9, P = 0.046). Liraglutide also significantly reduced the time mice with alloxan-induced diabetes spent in the dark compartment compared with vehicle-treated mice with alloxan-induced diabetes (F4,30 = 53.9, P < 0.001). Atorvastatin treatment alone had no significant effect on the time mice spent in dark compartment compared with the control group (F4,30 = 53.9, P > 0.05). Conclusion Atorvastatin significantly attenuated the adverse stimulus avoidance learning deficit in the alloxan-induced murine model of diabetes suggesting decreased impairment of memory and cognition.
Collapse
Affiliation(s)
- Osman Kukula
- Department of Pharmacology, Ondokuz Mayıs University, Faculty of Medicine, Atakum, Samsun55139, Turkey
| | - Caner Günaydın
- Department of Pharmacology, Ondokuz Mayıs University, Faculty of Medicine, Atakum, Samsun55139, Turkey
| |
Collapse
|
5
|
Markowicz-Piasecka M, Markiewicz A, Darłak P, Sikora J, Adla SK, Bagina S, Huttunen KM. Current Chemical, Biological, and Physiological Views in the Development of Successful Brain-Targeted Pharmaceutics. Neurotherapeutics 2022; 19:942-976. [PMID: 35391662 PMCID: PMC9294128 DOI: 10.1007/s13311-022-01228-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/24/2022] [Indexed: 12/13/2022] Open
Abstract
One of the greatest challenges with successful pharmaceutical treatments of central nervous system (CNS) diseases is the delivery of drugs into their target sites with appropriate concentrations. For example, the physically tight blood-brain barrier (BBB) effectively blocks compounds from penetrating into the brain, also by the action of metabolizing enzymes and efflux transport mechanisms. However, many endogenous compounds, including both smaller compounds and macromolecules, like amino acids, sugars, vitamins, nucleosides, hormones, steroids, and electrolytes, have their peculiar internalization routes across the BBB. These delivery mechanisms, namely carrier-mediated transport and receptor-mediated transcytosis have been utilized to some extent in brain-targeted drug development. The incomplete knowledge of the BBB and the smaller than a desirable number of chemical tools have hindered the development of successful brain-targeted pharmaceutics. This review discusses the recent advancements achieved in the field from the point of medicinal chemistry view and discusses how brain drug delivery can be improved in the future.
Collapse
Affiliation(s)
- Magdalena Markowicz-Piasecka
- Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego1, 90-151 Lodz, Poland
| | - Agata Markiewicz
- Students Research Group, Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151 Lodz, Poland
| | - Patrycja Darłak
- Students Research Group, Laboratory of Bioanalysis, Department of Pharmaceutical Chemistry, Drug Analysis and Radiopharmacy, Medical University of Lodz, ul. Muszyńskiego 1, 90-151 Lodz, Poland
| | - Joanna Sikora
- Department of Bioinorganic Chemistry, Medical University of Lodz, Medical University of Lodz, ul. Muszyńskiego1, 90-151 Lodz, Poland
| | - Santosh Kumar Adla
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, POB 1627, 70211 Kuopio, Finland
- Institute of Organic Chemistry and Biochemistry (IOCB), Czech Academy of Sciences, Flemingovo Namesti 542/2, 160 00 Prague, Czech Republic
| | - Sreelatha Bagina
- Charles River Discovery Research Services Finland Oy, Neulaniementie 4, 70210 Kuopio, Finland
| | - Kristiina M. Huttunen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Yliopistonranta 1C, POB 1627, 70211 Kuopio, Finland
| |
Collapse
|
6
|
Zhao G, Li N, Yin M, Xu M. Atorvastatin (ATV)-Loaded Lipid Bilayer-Coated Mesoporous Silica Nanoparticles Enhance the Therapeutic Efficacy of Acute Kidney Injury. J Biomed Nanotechnol 2021; 17:1754-1764. [PMID: 34688320 DOI: 10.1166/jbn.2021.3153] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Background: Acute kidney injury (AKI) increases the risk of chronic kidney disease. Atorvastatin (ATV)-loaded lipid bilayer-coated mesoporous silica nanoparticles (L-AMSNs) were synthesized, and their physicochemical parameters were characterized. L-AMSNs exhibited excellent stability; it did not increase in size over time, indicating that the lipid membrane coating prohibited mesoporous silica nanoparticles (MSNs) coalescence. Results: The rate of drug release differed significantly between AMSNs and L-AMSNs at all tested time points. A remarkable improvement in hydrogen peroxide (H₂O₂)-treated human umbilical vein endothelial cell (HUVEC) viability was observed after treatment with L-AMSNs; the malondialdehyde (MDA) level was significantly reduced compared to control cells. The extent of apoptosis was only 15% that of control H₂O₂-treated cells. L-AMSNs induced a remarkable decrease in the levels of pro-inflammatory cytokines (tumor necrosis factor [TNF]-α and interleukin [IL]-6), showing the therapeutic potential of nanocarrier-based ATV. L-AMSNs significantly increased the superoxide dismutase level and decreased the MDA level, indicating superior anti-inflammatory activity under conditions of oxidative stress. The L-AMSN showed a remarkable improvement in the outer stripe of outer medulla (OSOM) region and maintained the tubular structure of the kidney tissue. Besides, kidney injury score of L-AMSN is significantly lower compared to that of LPS-AKI and ATV indicating the excellent therapeutic efficacy of nanoparticulate system based L-AMSN. Conclusions: Nanoparticles system-based L-AMSNs maintained the tubular structure of kidney tissue, indicating excellent therapeutic efficacy. After clinical translation, L-AMSNs could serve as a promising treatment for AKI.
Collapse
Affiliation(s)
- Guanjie Zhao
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130033, China
| | - Na Li
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130033, China
| | - Min Yin
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130033, China
| | - Mingzhu Xu
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130033, China
| |
Collapse
|
7
|
Chu Z, Liu Z, Li W, Xu D, Pang L. Simvastatin attenuates delayed encephalopathy induced by carbon monoxide poisoning in rats by regulating oxidative stress, inflammation and NF-κB pathway. Mol Cell Toxicol 2021. [DOI: 10.1007/s13273-021-00124-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
8
|
Gu M, Mei XL, Zhao YN. Sepsis and Cerebral Dysfunction: BBB Damage, Neuroinflammation, Oxidative Stress, Apoptosis and Autophagy as Key Mediators and the Potential Therapeutic Approaches. Neurotox Res 2020; 39:489-503. [PMID: 32876918 DOI: 10.1007/s12640-020-00270-5] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 08/04/2020] [Accepted: 08/06/2020] [Indexed: 02/07/2023]
Abstract
Sepsis-associated cerebral dysfunction is complex pathophysiology, generated from primary infections that are developed elsewhere in the body. The neonates, elderly population and chronically ill and long-term hospitalized patients are predominantly vulnerable to sepsis and related cerebral damage. Generally, electrophysiological recordings, severity and sedation scales, computerized imaging and spectroscopy techniques are used for its detection and diagnosis. About the underlying mechanisms, enhanced blood-brain barrier permeability and metalloprotease activity, tight junction protein loss and endothelial cell degeneration promote the influx of inflammatory and toxic mediators into the brain, triggering cerebrovascular damage. An altered neutrophil count and phenotype further dysregulate the normal neuroimmune responses and neuroendocrine stability via modulated activation of protein kinase C-delta, nuclear factor kappa-B and sphingolipid signaling. Glial activation, together with pro-inflammatory cytokines and chemokines and the Toll-like receptor, destabilize the immune system. Moreover, superoxides and hydroperoxides generate oxidative stress and perturb mitochondrial dynamics and ATP synthesis, propagating neuronal injury cycle. Activated mitochondrial apoptotic pathway, characterized by increased caspase-3 and caspase-9 cleavage and Bax/Bcl2 ratio in the hippocampal and cortical neurons, stimulate neurocognitive impairments. Additionally, altered LC3-II/I and P62/SQSTM1, p-mTOR, p-AMPK1 and p-ULK1 levels and dysregulated autophagosome-lysosome fusion decrease neuronal and glial energy homeostasis. The therapies and procedures for attenuating sepsis-induced brain damage include early resuscitation, cerebral blood flow autoregulation, implantable electric vagus nerve stimulation, antioxidants, statins, glucocorticoids, neuroimmune axis modulators and PKCδ inhibitors. The current review enumerates the pathophysiology of sepsis-induced brain damage, its diagnosis, the role of critical inducers and mediators and, ultimately, therapeutic measures attenuating cerebrovascular degeneration.
Collapse
Affiliation(s)
- Ming Gu
- Department of Emergency and Critical Care Medicine, The Second Hospital of Jilin University, Changchun, China
| | - Xiang-Lin Mei
- Department of Pathology, The Second Hospital of Jilin University, Changchun, China
| | - Ya-Nan Zhao
- Neurology Department, China-Japan Union Hospital of Jilin University, Changchun, 130000, People's Republic of China.
| |
Collapse
|
9
|
Oxidative Stress-Mediated Blood-Brain Barrier (BBB) Disruption in Neurological Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020. [DOI: 10.1155/2020/4356386] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The blood-brain barrier (BBB), as a crucial gate of brain-blood molecular exchange, is involved in the pathogenesis of multiple neurological diseases. Oxidative stress is caused by an imbalance between the production of reactive oxygen species (ROS) and the scavenger system. Since oxidative stress plays a significant role in the production and maintenance of the BBB, the cerebrovascular system is especially vulnerable to it. The pathways that initiate BBB dysfunction include, but are not limited to, mitochondrial dysfunction, excitotoxicity, iron metabolism, cytokines, pyroptosis, and necroptosis, all converging on the generation of ROS. Interestingly, ROS also provide common triggers that directly regulate BBB damage, parameters including tight junction (TJ) modifications, transporters, matrix metalloproteinase (MMP) activation, inflammatory responses, and autophagy. We will discuss the role of oxidative stress-mediated BBB disruption in neurological diseases, such as hemorrhagic stroke, ischemic stroke (IS), Alzheimer’s disease (AD), Parkinson’s disease (PD), traumatic brain injury (TBI), amyotrophic lateral sclerosis (ALS), and cerebral small vessel disease (CSVD). This review will also discuss the latest clinical evidence of potential biomarkers and antioxidant drugs towards oxidative stress in neurological diseases. A deeper understanding of how oxidative stress damages BBB may open up more therapeutic options for the treatment of neurological diseases.
Collapse
|
10
|
Bolandghamat S, Behnam-Rassouli M. Recent Findings on the Effects of Pharmacological Agents on the Nerve Regeneration after Peripheral Nerve Injury. Curr Neuropharmacol 2020; 18:1154-1163. [PMID: 32379588 PMCID: PMC7709152 DOI: 10.2174/1570159x18666200507084024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/27/2020] [Accepted: 04/24/2020] [Indexed: 12/22/2022] Open
Abstract
Peripheral nerve injuries (PNIs) are accompanied with neuropathic pain and functional disability. Despite improvements in surgical repair techniques in recent years, the functional recovery is yet unsatisfied. Indeed a successful nerve repair depends not only on the surgical strategy but also on the cellular and molecular mechanisms involved in traumatic nerve injury. In contrast to all strategies suggested for nerve repair, pharmacotherapy is a cheap, accessible and non-invasive treatment that can be used immediately after nerve injury. This study aimed to review the effects of some pharmacological agents on the nerve regeneration after traumatic PNI evaluated by functional, histological and electrophysiological assessments. In addition, some cellular and molecular mechanisms responsible for their therapeutic actions, restricted to neural tissue, are suggested. These findings can not only help to find better strategies for peripheral nerve repair, but also to identify the neuropathic effects of various medications and their mechanisms of action.
Collapse
Affiliation(s)
- Samira Bolandghamat
- Department of Biology, Faculty of Science, Ferdowsi University of Mashhad, Iran
| | | |
Collapse
|
11
|
Solmaz V, Atasoy Ö, Erbaş O. Atorvastatin has therapeutic potential for the fatty liver-induced memory dysfunction in rats, likely via its antioxidant and anti-inflammatory properties. Neurol Res 2020; 42:497-503. [PMID: 32252617 DOI: 10.1080/01616412.2020.1747718] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
This study aimed to evaluate the memory function in a rat model of fatty liver and to investigate the effects of statins on fatty liver, neuronal inflammation, oxidative stress and memory. In this study, 24 male rats were used and were divided into four groups consisting of 6 animals in each. Of them, 12 rats received liquid diet containing 35% fructose for 8 weeks in order to induce hepatosteatosis, while other animals had a normal nutrition. Group 1 served as controls and had a normal nutrition with no drug treatment. The animals in Group 2 had a normal nutrition and treated with atorvastatin. Group 3 received high-fructose diet with no drug treatment and Group 4 received high-fructose diet followed by atorvastatin treatment. After the two weeks of treatment period, passive avoidance tasks evaluating the memory were performed in both the study and control groups. The liver and brain were then removed for histologic, pathologic, and biochemical evaluation. In the non-treated rats with hepatosteatosis (Group 3), the lowest mean latency time and the highest mean histopathologic liver score, and brain TNF- α and MDA (Measurement of lipid peroxidation) were found (p < 0.00001). On the other hand, in the animals treated with atorvastatin, all these parameters were significantly higher than that of controls and significantly lower than that of Group 3 (p < 0.05). Fatty liver can increase inflammation and cause memory disorders, and atorvastatin may have a positive effect on cognitive disorders.
Collapse
Affiliation(s)
| | - Özüm Atasoy
- Radyasyon Onkolojisi, Kartal Dr. Lutfi Kırdar Eğitim Ve Araştırma Hastanesi, İstanbul, Turkey
| | - Oytun Erbaş
- Department of Physiology, Bilim University Medical Faculty, İstanbul, Turkey
| |
Collapse
|
12
|
Henriques D, Moreira R, Schwamborn J, Pereira de Almeida L, Mendonça LS. Successes and Hurdles in Stem Cells Application and Production for Brain Transplantation. Front Neurosci 2019; 13:1194. [PMID: 31802998 PMCID: PMC6877657 DOI: 10.3389/fnins.2019.01194] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 10/21/2019] [Indexed: 12/18/2022] Open
Abstract
Brain regenerative strategies through the transplantation of stem cells hold the potential to promote functional rescue of brain lesions caused either by trauma or neurodegenerative diseases. Most of the positive modulations fostered by stem cells are fueled by bystander effects, namely increase of neurotrophic factors levels and reduction of neuroinflammation. Nevertheless, the ultimate goal of cell therapies is to promote cell replacement. Therefore, the ability of stem cells to migrate and differentiate into neurons that later become integrated into the host neuronal network replacing the lost neurons has also been largely explored. However, as most of the preclinical studies demonstrate, there is a small functional integration of graft-derived neurons into host neuronal circuits. Thus, it is mandatory to better study the whole brain cell therapy approach in order to understand what should be better comprehended concerning graft-derived neuronal and glial cells migration and integration before we can expect these therapies to be ready as a viable solution for brain disorder treatment. Therefore, this review discusses the positive mechanisms triggered by cell transplantation into the brain, the limitations of adult brain plasticity that might interfere with the neuroregeneration process, as well as some strategies tested to overcome some of these limitations. It also considers the efforts that have been made by the regulatory authorities to lead to better standardization of preclinical and clinical studies in this field in order to reduce the heterogeneity of the obtained results.
Collapse
Affiliation(s)
- Daniel Henriques
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Ricardo Moreira
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | - Jens Schwamborn
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Luís Pereira de Almeida
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,Faculty of Pharmacy, University of Coimbra, Coimbra, Portugal
| | - Liliana S Mendonça
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
13
|
Qi W, Yan L, Liu Y, Zhou X, Li R, Wang Y, Bai L, Chen J, Nie X. Simvastatin aggravates impaired autophagic flux in NSC34-hSOD1G93A cells through inhibition of geranylgeranyl pyrophosphate synthesis. Neuroscience 2019; 409:130-141. [DOI: 10.1016/j.neuroscience.2019.04.034] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/05/2019] [Accepted: 04/13/2019] [Indexed: 11/28/2022]
|
14
|
Paseban M, Mohebbati R, Niazmand S, Sathyapalan T, Sahebkar A. Comparison of the Neuroprotective Effects of Aspirin, Atorvastatin, Captopril and Metformin in Diabetes Mellitus. Biomolecules 2019; 9:biom9040118. [PMID: 30934759 PMCID: PMC6523359 DOI: 10.3390/biom9040118] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 03/13/2019] [Accepted: 03/21/2019] [Indexed: 12/23/2022] Open
Abstract
Objective: The aim of this study was to investigate the effect of combined intake of a high dose of aspirin, atorvastatin, captopril and metformin on oxidative stress in the brain cortex and hippocampus of streptozotocin (STZ)-induced diabetic rats. Material and methods: Rats were randomly divided into the following 11 groups: control and diabetic (D), as well as 9 groups that were treated with metformin (M, 300 mg/kg) or aspirin (ASA, 120 mg/kg) alone or in different combinations with captopril (C, 50 mg/kg) and/or atorvastatin (AT, 40 mg/kg) as follows: (D + M), (D + ASA), (D + M + ASA), (D + M + C), (D + M + AT), (D + M + C + ASA), (D + M + C + AT), (D + M + AT + ASA) and (D + M + C + AT + ASA). The rats in treatment groups received drugs by gavage daily for six weeks. Serum lipid profile and levels of oxidative markers in the brain cortex and hippocampus tissues were evaluated. Results: The levels of malondialdehyde in the brain cortex and hippocampus in all the treated groups decreased significantly (p < 0.05). There was a significant increase in the total thiol concentration as well as catalase activity in treated rats in (M + AT), (M + C + ASA), (M + C + AT), (M + AT + ASA) and (M + C + AT + ASA) groups in cortex and hippocampus in comparison with the diabetic rats (p < 0.05). Also, the superoxide dismutase activity in all treated rats with medications was significantly increased compared to the diabetic rats (p < 0.05–0.01). Conclusion: Our findings showed that the combined use of high-dose aspirin, metformin, captopril and atorvastatin potentiated their antioxidant effects on the brain, and hence could potentially improve cognitive function with their neuroprotective effects on hippocampus.
Collapse
Affiliation(s)
- Maryam Paseban
- Department of Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran.
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran.
| | - Reza Mohebbati
- Department of Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran.
| | - Saeed Niazmand
- Department of Physiology, School of Medicine, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran.
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran.
| | - Thozhukat Sathyapalan
- Department of Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, HU3 2RW, UK.
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of MedicalSciences; Mashhad 9177948564, Iran.
- School of Pharmacy, Mashhad University of Medical Sciences; Mashhad 9177948564, Iran.
| |
Collapse
|
15
|
Fracassi A, Marangoni M, Rosso P, Pallottini V, Fioramonti M, Siteni S, Segatto M. Statins and the Brain: More than Lipid Lowering Agents? Curr Neuropharmacol 2019; 17:59-83. [PMID: 28676012 PMCID: PMC6341496 DOI: 10.2174/1570159x15666170703101816] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2017] [Revised: 05/24/2017] [Accepted: 06/26/2017] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Statins represent a class of medications widely prescribed to efficiently treat dyslipidemia. These drugs inhibit 3-βhydroxy 3β-methylglutaryl Coenzyme A reductase (HMGR), the rate-limiting enzyme of mevalonate (MVA) pathway. Besides cholesterol, MVA pathway leads to the production of several other compounds, which are essential in the regulation of a plethora of biological activities, including in the central nervous system. For these reasons, statins are able to induce pleiotropic actions, and acquire increased interest as potential and novel modulators in brain processes, especially during pathological conditions. OBJECTIVE The purpose of this review is to summarize and examine the current knowledge about pharmacokinetic and pharmacodynamic properties of statins in the brain. In addition, effects of statin on brain diseases are discussed providing the most up-to-date information. METHODS Relevant scientific information was identified from PubMed database using the following keywords: statins and brain, central nervous system, neurological diseases, neurodegeneration, brain tumors, mood, stroke. RESULTS 315 scientific articles were selected and analyzed for the writing of this review article. Several papers highlighted that statin treatment is effective in preventing or ameliorating the symptomatology of a number of brain pathologies. However, other studies failed to demonstrate a neuroprotective effect. CONCLUSION Even though considerable research studies suggest pivotal functional outcomes induced by statin therapy, additional investigation is required to better determine the pharmacological effectiveness of statins in the brain, and support their clinical use in the management of different neuropathologies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Marco Segatto
- Address correspondence to this author at the Department of Sense Organs, Sapienza University, viale del Policlinico 155, 00186 Rome, Italy; E-mail:
| |
Collapse
|
16
|
Husain I, Akhtar M, Madaan T, Abdin MZ, Islamuddin M, Najmi AK. Rosuvastatin alleviates high-salt and cholesterol diet-induced cognitive impairment in rats via Nrf2-ARE pathway. Redox Rep 2018; 23:168-179. [PMID: 29961403 PMCID: PMC6748700 DOI: 10.1080/13510002.2018.1492774] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
OBJECTIVE The objectives of our study were to investigate the possible effect of rosuvastatin in ameliorating high salt and cholesterol diet (HSCD)-induced cognitive impairment and to also investigate its possible action via the Nrf2-ARE pathway. METHODS In silico studies were performed to check the theoretical binding of rosuvastatin to the Nrf2 target. HSCD was used to induce cognitive impairment in rats and neurobehavioral studies were performed to evaluate the efficacy of rosuvastatin in enhancing cognition. Biochemical analyses were used to estimate changes in oxidative markers. Western blot and immunohistochemical analyses were done to check Nrf2 translocation. TUNEL and caspase 3 tests were performed to evaluate reversal of apoptosis by rosuvastatin. RESULTS Rosuvastatin showed good theoretical affinity to Nrf2, significantly reversed changes in oxidative biomarkers which were induced by HSCD, and also improved the performance of rats in the neurobehavioral test. A rise in nuclear translocation of Nrf2 was revealed through immunohistochemical analysis and western blot. TUNEL staining and caspase 3 activity showed attenuation of apoptosis. DISCUSSION We have investigated a novel mechanism of action for rosuvastatin (via the Nrf2-ARE pathway) and demonstrated that it has the potential to be used in the treatment of cognitive impairment.
Collapse
Affiliation(s)
- Ibraheem Husain
- a Department of Pharmacology, School of Pharmaceutical Education and Research , Jamia Hamdard (Hamdard University) , New Delhi , India
| | - Mohd Akhtar
- a Department of Pharmacology, School of Pharmaceutical Education and Research , Jamia Hamdard (Hamdard University) , New Delhi , India
| | - Tushar Madaan
- a Department of Pharmacology, School of Pharmaceutical Education and Research , Jamia Hamdard (Hamdard University) , New Delhi , India
| | - Malik Zainul Abdin
- b Department of Biotechnology, School of Chemical and Life Sciences , Jamia Hamdard (Hamdard University) , New Delhi , India
| | - Mohammad Islamuddin
- b Department of Biotechnology, School of Chemical and Life Sciences , Jamia Hamdard (Hamdard University) , New Delhi , India
| | - Abul Kalam Najmi
- a Department of Pharmacology, School of Pharmaceutical Education and Research , Jamia Hamdard (Hamdard University) , New Delhi , India
| |
Collapse
|
17
|
Morris G, Fernandes BS, Puri BK, Walker AJ, Carvalho AF, Berk M. Leaky brain in neurological and psychiatric disorders: Drivers and consequences. Aust N Z J Psychiatry 2018; 52:924-948. [PMID: 30231628 DOI: 10.1177/0004867418796955] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND The blood-brain barrier acts as a highly regulated interface; its dysfunction may exacerbate, and perhaps initiate, neurological and neuropsychiatric disorders. METHODS In this narrative review, focussing on redox, inflammatory and mitochondrial pathways and their effects on the blood-brain barrier, a model is proposed detailing mechanisms which might explain how increases in blood-brain barrier permeability occur and can be maintained with increasing inflammatory and oxidative and nitrosative stress being the initial drivers. RESULTS Peripheral inflammation, which is causatively implicated in the pathogenesis of major psychiatric disorders, is associated with elevated peripheral pro-inflammatory cytokines, which in turn cause increased blood-brain barrier permeability. Reactive oxygen species, such as superoxide radicals and hydrogen peroxide, and reactive nitrogen species, such as nitric oxide and peroxynitrite, play essential roles in normal brain capillary endothelial cell functioning; however, chronically elevated oxidative and nitrosative stress can lead to mitochondrial dysfunction and damage to the blood-brain barrier. Activated microglia, redox control of which is mediated by nitric oxide synthases and nicotinamide adenine dinucleotide phosphate (NADPH) oxidases, secrete neurotoxic molecules such as reactive oxygen species, nitric oxide, prostaglandin, cyclooxygenase-2, quinolinic acid, several chemokines (including monocyte chemoattractant protein-1 [MCP-1], C-X-C motif chemokine ligand 1 [CXCL-1] and macrophage inflammatory protein 1α [MIP-1α]) and the pro-inflammatory cytokines interleukin-6, tumour necrosis factor-α and interleukin-1β, which can exert a detrimental effect on blood-brain barrier integrity and function. Similarly, reactive astrocytes produce neurotoxic molecules such as prostaglandin E2 and pro-inflammatory cytokines, which can cause a 'leaky brain'. CONCLUSION Chronic inflammatory and oxidative and nitrosative stress is associated with the development of a 'leaky gut'. The following evidence-based approaches, which address the leaky gut and blood-brain barrier dysfunction, are suggested as potential therapeutic interventions for neurological and neuropsychiatric disorders: melatonin, statins, probiotics containing Bifidobacteria and Lactobacilli, N-acetylcysteine, and prebiotics containing fructo-oligosaccharides and galacto-oligosaccharides.
Collapse
Affiliation(s)
- Gerwyn Morris
- 1 IMPACT Strategic Research Centre, Deakin University School of Medicine, and Barwon Health, Geelong, VIC, Australia
| | - Brisa S Fernandes
- 1 IMPACT Strategic Research Centre, Deakin University School of Medicine, and Barwon Health, Geelong, VIC, Australia.,2 Centre for Addiction and Mental Health (CAMH) and Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Basant K Puri
- 3 Department of Medicine, Hammersmith Hospital, Imperial College London, London, UK
| | - Adam J Walker
- 1 IMPACT Strategic Research Centre, Deakin University School of Medicine, and Barwon Health, Geelong, VIC, Australia
| | - Andre F Carvalho
- 2 Centre for Addiction and Mental Health (CAMH) and Department of Psychiatry, University of Toronto, Toronto, ON, Canada
| | - Michael Berk
- 1 IMPACT Strategic Research Centre, Deakin University School of Medicine, and Barwon Health, Geelong, VIC, Australia.,4 Orygen, The National Centre of Excellence in Youth Mental Health, The Department of Psychiatry and The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
18
|
Brzica H, Abdullahi W, Reilly BG, Ronaldson PT. Sex-specific differences in organic anion transporting polypeptide 1a4 (Oatp1a4) functional expression at the blood-brain barrier in Sprague-Dawley rats. Fluids Barriers CNS 2018; 15:25. [PMID: 30208928 PMCID: PMC6136214 DOI: 10.1186/s12987-018-0110-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 08/14/2018] [Indexed: 12/19/2022] Open
Abstract
Background Targeting endogenous blood–brain barrier (BBB) transporters such as organic anion transporting polypeptide 1a4 (Oatp1a4) can facilitate drug delivery for treatment of neurological diseases. Advancement of Oatp targeting for optimization of CNS drug delivery requires characterization of sex-specific differences in BBB expression and/or activity of this transporter. Methods In this study, we investigated sex differences in Oatp1a4 functional expression at the BBB in adult and prepubertal (i.e., 6-week-old) Sprague–Dawley rats. We also performed castration or ovariectomy surgeries to assess the role of gonadal hormones on Oatp1a4 protein expression and transport activity at the BBB. Slco1a4 (i.e., the gene encoding Oatp1a4) mRNA expression and Oatp1a4 protein expression in brain microvessels was determined using quantitative real-time PCR and western blot analysis, respectively. Oatp transport function at the BBB was determined via in situ brain perfusion using [3H]taurocholate and [3H]atorvastatin as probe substrates. Data were expressed as mean ± SD and analyzed via one-way ANOVA followed by the post hoc Bonferroni t-test. Results Our results showed increased brain microvascular Slco1a4 mRNA and Oatp1a4 protein expression as well as increased brain uptake of [3H]taurocholate and [3H]atorvastatin in female rats as compared to males. Oatp1a4 expression at the BBB was enhanced in castrated male animals but was not affected by ovariectomy in female animals. In prepubertal rats, no sex-specific differences in brain microvascular Oatp1a4 expression were observed. Brain accumulation of [3H]taurocholate in male rats was increased following castration as compared to controls. In contrast, there was no difference in [3H]taurocholate brain uptake between ovariectomized and control female rats. Conclusions These novel data confirm sex-specific differences in BBB Oatp1a4 functional expression, findings that have profound implications for treatment of CNS diseases. Studies are ongoing to fully characterize molecular pathways that regulate sex differences in Oatp1a4 expression and activity.
Collapse
Affiliation(s)
- Hrvoje Brzica
- Department of Pharmacology, College of Medicine, University of Arizona, P.O. Box 245050, 1501 N. Campbell Avenue, Tucson, AZ, 85724-5050, USA
| | - Wazir Abdullahi
- Department of Pharmacology, College of Medicine, University of Arizona, P.O. Box 245050, 1501 N. Campbell Avenue, Tucson, AZ, 85724-5050, USA
| | - Bianca G Reilly
- Department of Pharmacology, College of Medicine, University of Arizona, P.O. Box 245050, 1501 N. Campbell Avenue, Tucson, AZ, 85724-5050, USA
| | - Patrick T Ronaldson
- Department of Pharmacology, College of Medicine, University of Arizona, P.O. Box 245050, 1501 N. Campbell Avenue, Tucson, AZ, 85724-5050, USA.
| |
Collapse
|
19
|
Lu D, Liu Y, Mai H, Zang J, Shen L, Zhang Y, Xu A. Rosuvastatin Reduces Neuroinflammation in the Hemorrhagic Transformation After rt-PA Treatment in a Mouse Model of Experimental Stroke. Front Cell Neurosci 2018; 12:225. [PMID: 30116175 PMCID: PMC6082938 DOI: 10.3389/fncel.2018.00225] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 07/11/2018] [Indexed: 12/31/2022] Open
Abstract
Hemorrhagic transformation (HT) is a serious complication that stimulates inflammation during reperfusion therapy after acute ischemic stroke. Rosuvastatin, a 3-hydroxymethyl-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitor, might improve the outcome of HT by inhibiting neuroinflammation. This study aimed to explore the protective effects of rosuvastatin against HT after recombinant tissue plasminogen activator (rt-PA) treatment in mice with experimental stroke via the attenuation of inflammation. A total of one hundred sixty-nine male BALB/c mice were used in the experiment. HT was successfully established in 70 mice that were subjected to 3 h of middle cerebral artery occlusion (MCAO) followed by a 10 mg/kg rt-PA injection over 10 min and reperfusion for 24 h. The mice were then administered rosuvastatin (1 mg/kg, 5 mg/kg) or saline (vehicle). The brain water content and neurological deficits (wire hang and adhesive removal somatosensory tests) were assessed at 24 h after rt-PA reperfusion following MCAO surgery. The morphology, blood-brain barrier (BBB) permeability and number of astrocytes and microglia were assessed by immunohistochemistry, electron microscopy and western blotting at 24 h after rt-PA reperfusion following MCAO surgery. Rosuvastatin protected against impaired neurological function and reversed the BBB leakage observed in the HT group. The increased activation of astrocytes and microglia and secretion of inflammatory factors caused by HT damage were significantly attenuated by high-dose rosuvastatin treatment vs. normal-dose rosuvastatin treatment. Related inflammatory pathways, such as the nuclear factor kappa B (NF-κB) and mitogen-activated protein kinase (MAPK) pathways, were downregulated in the rosuvastatin-treated groups compared with the HT group. In conclusion, our results indicate that rosuvastatin is a promising therapeutic agent for HT after rt-PA reperfusion following MCAO surgery in mice, as it attenuates neuroinflammation. Additionally, high-dose rosuvastatin treatment could have a greater anti-inflammatory effect on HT than normal-dose rosuvastatin treatment.
Collapse
Affiliation(s)
- Dan Lu
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute of Jinan University, Guangzhou, China
| | - Yanfang Liu
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute of Jinan University, Guangzhou, China
| | - Hongcheng Mai
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute of Jinan University, Guangzhou, China
| | - Jiankun Zang
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute of Jinan University, Guangzhou, China
| | - Lingling Shen
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute of Jinan University, Guangzhou, China
| | - Yusheng Zhang
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China
| | - Anding Xu
- Department of Neurology and Stroke Center, The First Affiliated Hospital, Jinan University, Guangzhou, China.,Clinical Neuroscience Institute of Jinan University, Guangzhou, China
| |
Collapse
|
20
|
Abdullahi W, Tripathi D, Ronaldson PT. Blood-brain barrier dysfunction in ischemic stroke: targeting tight junctions and transporters for vascular protection. Am J Physiol Cell Physiol 2018; 315:C343-C356. [PMID: 29949404 DOI: 10.1152/ajpcell.00095.2018] [Citation(s) in RCA: 346] [Impact Index Per Article: 57.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The blood-brain barrier (BBB) is a physical and biochemical barrier that precisely controls cerebral homeostasis. It also plays a central role in the regulation of blood-to-brain flux of endogenous and exogenous xenobiotics and associated metabolites. This is accomplished by molecular characteristics of brain microvessel endothelial cells such as tight junction protein complexes and functional expression of influx and efflux transporters. One of the pathophysiological features of ischemic stroke is disruption of the BBB, which significantly contributes to development of brain injury and subsequent neurological impairment. Biochemical characteristics of BBB damage include decreased expression and altered organization of tight junction constituent proteins as well as modulation of functional expression of endogenous BBB transporters. Therefore, there is a critical need for development of novel therapeutic strategies that can protect against BBB dysfunction (i.e., vascular protection) in the setting of ischemic stroke. Such strategies include targeting tight junctions to ensure that they maintain their correct structure or targeting transporters to control flux of physiological substrates for protection of endothelial homeostasis. In this review, we will describe the pathophysiological mechanisms in cerebral microvascular endothelial cells that lead to BBB dysfunction following onset of stroke. Additionally, we will utilize this state-of-the-art knowledge to provide insights on novel pharmacological strategies that can be developed to confer BBB protection in the setting of ischemic stroke.
Collapse
Affiliation(s)
- Wazir Abdullahi
- Department of Pharmacology, College of Medicine, University of Arizona , Tucson, Arizona
| | - Dinesh Tripathi
- Department of Pharmacology, College of Medicine, University of Arizona , Tucson, Arizona
| | - Patrick T Ronaldson
- Department of Pharmacology, College of Medicine, University of Arizona , Tucson, Arizona
| |
Collapse
|
21
|
Butterfield DA. Perspectives on Oxidative Stress in Alzheimer’s Disease and Predictions of Future Research Emphases. J Alzheimers Dis 2018; 64:S469-S479. [DOI: 10.3233/jad-179912] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- D. Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, USA
| |
Collapse
|
22
|
Heme Oxygenase-1 Activity as a Correlate to Exercise-Mediated Amelioration of Cognitive Decline and Neuropathological Alterations in an Aging Rat Model of Dementia. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7212861. [PMID: 29662895 PMCID: PMC5831053 DOI: 10.1155/2018/7212861] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 11/24/2017] [Accepted: 01/01/2018] [Indexed: 11/17/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder with cognitive impairment. Physical exercise has long been proven to be beneficial in the disorder. The present study was designed to examine the effect of voluntary exercise on spatial memory, imaging, and pathological abnormalities. Particular focus has been given to the role of heme oxygenase-1 (HO-1)—an important cellular cytoprotectant in preserving mental acuity—using an aging rat model of dementia. Male and female Wistar rats were segregated into six groups—namely, (i) aged sedentary (control) females (ASF, n = 8); (ii) aged sedentary (control) males (ASM, n = 8); (iii) aged running females (ARF, n = 8); (iv) aged running males (ARM, n = 8); (v) young control females (YCF, n = 8); and (vi) young control males (YCM, n = 8). Rats in the ARF and ARM groups had free access to a standardized inbuilt running wheel during the 3-month evaluation period. Spatial memory was investigated using the Morris Water Test, imaging and pathological alterations were assessed using positron emission tomography (PET) imaging and histopathological examinations (H&E, Congo red staining), respectively, and HO-1 enzyme activity assays were also conducted. The outcomes suggest that voluntary physical exercise mitigates impaired spatial memory and neuropathological changes exhibited by the aging sedentary group, via elevated HO-1 activity, contributing to the antioxidant capacity in the aging brain.
Collapse
|
23
|
Saeedi Saravi SS, Saeedi Saravi SS, Arefidoust A, Dehpour AR. The beneficial effects of HMG-CoA reductase inhibitors in the processes of neurodegeneration. Metab Brain Dis 2017; 32:949-965. [PMID: 28578514 DOI: 10.1007/s11011-017-0021-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 04/28/2017] [Indexed: 12/13/2022]
Abstract
Statins, cholesterol lowering drugs, have been demonstrated to exert beneficial effects in other conditions such as primary and progressing neurodegenerative diseases beyond their original role. Observation that statins ameliorate the neurodegenerative diseases such as Parkinson's disease (PD), Alzheimer's disease (AD), multiple sclerosis (MS) and cerebral ischemic stroke, the neuroprotective effects of these drugs are thought to be linked to their anti-inflammatory, anti-oxidative, and anti-excitotoxic properties. Despite the voluminous literature on the clinical advantages of 3-hydroxy-3-methylglutaryl Co-enzyme A reductase (HMGCR) inhibitors (statins) in cardiovascular system, the neuroprotective effects and the underlying mechanisms are little understood. Hence, the present review tries to provide a critical overview on the statin-induced neuroprotection, which are presumed to be associated with the ability to reduce cholesterol, Amyloid-β and apolipoprotein E (ApoE) levels, decrease reactive oxygen and nitrogen species (ROS and RNS) formation, inhibit excitotoxicity, modulate matrix metalloproteinases (MMPs), stimulate endothelial nitric oxide synthase (eNOS), and increase cerebral blood perfusion. This review is also aimed to illustrate that statins protect neurons against the neuro-inflammatory processes through balancing pro-inflammatory/anti-inflammatory cytokines. Ultimately, the beneficial role of statins in ameliorating the development of PD, AD, MS and cerebral ischemic stroke has been separately reviewed.
Collapse
Affiliation(s)
- Seyed Soheil Saeedi Saravi
- Department of Toxicology-Pharmacology, Faculty of Pharmacy, Guilan University of Medical Sciences, Rasht, Iran
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Experimental Medicine Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Sobhan Saeedi Saravi
- Department of Toxicology-Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Alireza Arefidoust
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Experimental Medicine Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
- Experimental Medicine Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
24
|
Lee SE, Hyun H, Park MR, Choi Y, Son YJ, Park YG, Jeong SG, Shin MY, Ha HJ, Hong HS, Choi MK, Im GS, Park EW, Kim YH, Park C, Kim EY, Park SP. Production of transgenic pig as an Alzheimer's disease model using a multi-cistronic vector system. PLoS One 2017; 12:e0177933. [PMID: 28586343 PMCID: PMC5460854 DOI: 10.1371/journal.pone.0177933] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 05/08/2017] [Indexed: 01/10/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease associated with memory loss and cognitive impairments. An AD transgenic (Tg) pig model would be useful for preclinical testing of therapeutic agents. We generated an AD Tg pig by somatic cell nuclear transfer (SCNT) using a multi-cistronic vector that harbored three AD-related genes with a total of six well-characterized mutations: hAPP (K670N/M671L, I716V, and V717I), hTau (P301L), and hPS1 (M146V and L286P). Four AD Tg cell lines were established from Jeju black pig ear fibroblasts (JB-PEFs); the resultant JB-PEFAD cells harbored transgene integration, expressed transgene mRNAs, and had normal karyotypes. Tg line #2-1, which expressed high levels of the transgenes, was used for SCNT; cleavage and blastocyst rates of embryos derived from this line were lower than those of Non-Tg. These embryos yielded three piglets (Jeju National University AD-Tg pigs, JNUPIGs) revealed by microsatellite testing to be genetically identical to JB-PEFAD. Transgenes were expressed in multiple tissues, and at especially high levels in brain, and Aβ-40/42, total Tau, and GFAP levels were high in brains of the Tg animals. Five or more copies of transgenes were inserted into chromosome X. This is the first report of an AD Tg pig derived from a multi-cistronic vector.
Collapse
Affiliation(s)
- Seung-Eun Lee
- Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju-si, Jeju Special Self-Governing Province, Korea
- Stem Cell Research Center, Jeju National University, Jeju-si, Jeju Special Self-Governing Province, Korea
| | - Hyuk Hyun
- Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju-si, Jeju Special Self-Governing Province, Korea
- Stem Cell Research Center, Jeju National University, Jeju-si, Jeju Special Self-Governing Province, Korea
| | - Mi-Ryung Park
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Jeollabuk-do, Korea
| | - Youngsok Choi
- Department of Biomedical Science, CHA University, Pocheon-si, Gyeonggi-do, Korea
| | - Yeo-Jin Son
- Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju-si, Jeju Special Self-Governing Province, Korea
- Stem Cell Research Center, Jeju National University, Jeju-si, Jeju Special Self-Governing Province, Korea
| | - Yun-Gwi Park
- Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju-si, Jeju Special Self-Governing Province, Korea
- Stem Cell Research Center, Jeju National University, Jeju-si, Jeju Special Self-Governing Province, Korea
| | - Sang-Gi Jeong
- Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju-si, Jeju Special Self-Governing Province, Korea
- Stem Cell Research Center, Jeju National University, Jeju-si, Jeju Special Self-Governing Province, Korea
| | - Min-Young Shin
- Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju-si, Jeju Special Self-Governing Province, Korea
- Stem Cell Research Center, Jeju National University, Jeju-si, Jeju Special Self-Governing Province, Korea
| | - Hee-Jin Ha
- Medifron DBT, Ansan-si, Gyeonggi-do, Korea
| | | | - Min-Keyung Choi
- Department of Animal Biotechnology, Konkuk University, Seoul, Korea
| | - Gi-Sun Im
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Jeollabuk-do, Korea
| | - Eung-Woo Park
- Animal Biotechnology Division, National Institute of Animal Science, Rural Development Administration, Wanju-gun, Jeollabuk-do, Korea
| | | | - Chankyu Park
- Department of Animal Biotechnology, Konkuk University, Seoul, Korea
| | - Eun-Young Kim
- Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju-si, Jeju Special Self-Governing Province, Korea
- Stem Cell Research Center, Jeju National University, Jeju-si, Jeju Special Self-Governing Province, Korea
- Mirae Cell Bio, Seoul, Korea
- * E-mail: (SPP); (EYK)
| | - Se-Pill Park
- Faculty of Biotechnology, College of Applied Life Sciences, Jeju National University, Jeju-si, Jeju Special Self-Governing Province, Korea
- Stem Cell Research Center, Jeju National University, Jeju-si, Jeju Special Self-Governing Province, Korea
- Mirae Cell Bio, Seoul, Korea
- * E-mail: (SPP); (EYK)
| |
Collapse
|
25
|
Statins Reduce Lipopolysaccharide-Induced Cytokine and Inflammatory Mediator Release in an In Vitro Model of Microglial-Like Cells. Mediators Inflamm 2017; 2017:2582745. [PMID: 28546657 PMCID: PMC5435995 DOI: 10.1155/2017/2582745] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Revised: 02/15/2017] [Accepted: 03/13/2017] [Indexed: 01/10/2023] Open
Abstract
The anti-inflammatory effects of statins (HMG-CoA reductase inhibitors) within the cardiovascular system are well-established; however, their neuroinflammatory potential is unclear. It is currently unknown whether statins' neurological effects are lipid-dependent or due to pleiotropic mechanisms. Therefore, the assumption that all statin compounds will have the same effect within the central nervous system is potentially inappropriate, with no studies to date having compared all statins in a single model. Thus, the aim of this study was to compare the effects of the six statins (atorvastatin, fluvastatin, pitavastatin, pravastatin, rosuvastatin, and simvastatin) within a single in vitro model of neuroinflammation. To achieve this, PMA-differentiated THP-1 cells were used as surrogate microglial cells, and LPS was used to induce inflammatory conditions. Here, we show that pretreatment with all statins was able to significantly reduce LPS-induced interleukin (IL)-1β and tumour necrosis factor (TNF)-α release, as well as decrease LPS-induced prostaglandin E2 (PGE2). Similarly, global reactive oxygen species (ROS) and nitric oxide (NO) production were decreased following pretreatment with all statins. Based on these findings, it is suggested that more complex cellular models should be considered to further compare individual statin compounds, including translation into in vivo models of acute and/or chronic neuroinflammation.
Collapse
|
26
|
Husain I, Akhtar M, Abdin MZ, Islamuddin M, Shaharyar M, Najmi AK. Rosuvastatin ameliorates cognitive impairment in rats fed with high-salt and cholesterol diet via inhibiting acetylcholinesterase activity and amyloid beta peptide aggregation. Hum Exp Toxicol 2017; 37:399-411. [PMID: 28441890 DOI: 10.1177/0960327117705431] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Amyloid beta (Aβ) peptide aggregation and cholinergic neurodegeneration are involved in the development of cognitive impairment. Therefore, in this article, we examined rosuvastatin (RSV), an oral hypolipidemic drug, to determine its potential as a dual inhibitor of acetylcholinesterase (AChE) and Aβ peptide aggregation for the treatment of cognitive impairment. Molecular docking study was done to examine the affinity of RSV with Aβ1-42 and AChE in silico. We also employed neurobehavioral activity tests, biochemical estimation, and histopathology to study the anti-Aβ1-42 aggregation capability of RSV in vivo. Molecular docking study provided evidence that RSV has the best binding conformer at its receptor site or active site of an enzyme. The cognitive impairment in female Wistar rats was induced by high-salt and cholesterol diet (HSCD) ad libitum for 8 weeks. RSV ameliorated serum cholesterol level, AChE activity, and Aβ1-42 peptide aggregations in HSCD induced cognitive impairment. In addition, RSV-treated rats showed greater scores in the open field (locomotor activity) test. Moreover, the histopathological studies in the hippocampus and cortex of rat brain also supported that RSV markedly reduced the cognitive impairment and preserved the normal histoarchitectural pattern of the hippocampus and cortex. Taken together, these data indicate that RSV may act as a dual inhibitor of AChE and Aβ1-42 peptide aggregation, therefore suggesting a therapeutic strategy for cognitive impairment treatment.
Collapse
Affiliation(s)
- I Husain
- 1 Department of Pharmacology, Faculty of Pharmacy, Jamia Hamdard (Hamdard University), New Delhi, India
| | - M Akhtar
- 1 Department of Pharmacology, Faculty of Pharmacy, Jamia Hamdard (Hamdard University), New Delhi, India
| | - M Zainul Abdin
- 2 Department of Biotechnology, Faculty of Science, Jamia Hamdard (Hamdard University), New Delhi, India
| | - M Islamuddin
- 2 Department of Biotechnology, Faculty of Science, Jamia Hamdard (Hamdard University), New Delhi, India
| | - M Shaharyar
- 3 Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Jamia Hamdard (Hamdard University), New Delhi, India
| | - A K Najmi
- 1 Department of Pharmacology, Faculty of Pharmacy, Jamia Hamdard (Hamdard University), New Delhi, India
| |
Collapse
|
27
|
Affiliation(s)
- Eleni Kotsampasakou
- University of Vienna; Department of Pharmaceutical Chemistry; Althanstrasse 14 1090 Vienna Austria
| | - Gerhard F. Ecker
- University of Vienna; Department of Pharmaceutical Chemistry; Althanstrasse 14 1090 Vienna Austria
| |
Collapse
|
28
|
Simvastatin enhances the hippocampal klotho in a rat model of streptozotocin-induced cognitive decline. Prog Neuropsychopharmacol Biol Psychiatry 2017; 72:87-94. [PMID: 27687042 DOI: 10.1016/j.pnpbp.2016.09.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Revised: 09/13/2016] [Accepted: 09/25/2016] [Indexed: 12/13/2022]
Abstract
Brain oxidative status is a crucial factor in the development of sporadic Alzheimer's disease (AD). Klotho, an anti-aging protein, diminishes oxidative stress by the induction of manganese superoxide dismutase (MnSOD). Thus, the substances that increase klotho expression could be considered as a potential treatment for Alzheimer's disease when the oxidative imbalance is present. Statins are suggested to up-regulate klotho expression. We examined the effect of simvastatin (5mg/kg, daily for 3weeks) on hippocampal klotho and MnSOD expression in the cognitive declined animal model induced by intracerebroventricular (ICV)-streptozotocin (STZ) administration. Cognitive assessment was performed by the Morris Water Maze (MWM) test. The results indicated that mean escape latency and distance were prolonged in the ICV-STZ group compared with the control group. The expression of klotho and MnSOD were also down regulated in the hippocampus. Furthermore, improved spatial performance was observed in simvastatin-treated animals. This effect could be related to increase in oxidative stress tolerance as evidenced by klotho and MnSOD up-regulation. Our current study indicates that klotho upregulation may be a neuroprotective mechanism of simvastatin against cognitive decline in AD.
Collapse
|
29
|
Villa RF, Ferrari F, Moretti A. Effects of Neuroprotectants Before and After Stroke: Statins and Anti-hypertensives. SPRINGER SERIES IN TRANSLATIONAL STROKE RESEARCH 2017. [DOI: 10.1007/978-3-319-45345-3_14] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
30
|
Zhang H, Chen MK, Li K, Hu C, Lu MH, Situ J. Eupafolin nanoparticle improves acute renal injury induced by LPS through inhibiting ROS and inflammation. Biomed Pharmacother 2017; 85:704-711. [DOI: 10.1016/j.biopha.2016.11.083] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 11/18/2016] [Accepted: 11/21/2016] [Indexed: 12/13/2022] Open
|
31
|
Alternative Interventions to Prevent Oxidative Damage following Ischemia/Reperfusion. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:7190943. [PMID: 28116037 PMCID: PMC5225393 DOI: 10.1155/2016/7190943] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 09/23/2016] [Accepted: 10/12/2016] [Indexed: 12/25/2022]
Abstract
Ischemia/reperfusion (I/R) lesions are a phenomenon that occurs in multiple pathological states and results in a series of events that end in irreparable damage that severely affects the recovery and health of patients. The principal therapeutic approaches include preconditioning, postconditioning, and remote ischemic preconditioning, which when used separately do not have a great impact on patient mortality or prognosis. Oxidative stress is known to contribute to the damage caused by I/R; however, there are no pharmacological approaches to limit or prevent this. Here, we explain the relationship between I/R and the oxidative stress process and describe some pharmacological options that may target oxidative stress-states.
Collapse
|
32
|
Lam V, Hackett M, Takechi R. Antioxidants and Dementia Risk: Consideration through a Cerebrovascular Perspective. Nutrients 2016; 8:nu8120828. [PMID: 27999412 PMCID: PMC5188481 DOI: 10.3390/nu8120828] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/16/2016] [Accepted: 12/16/2016] [Indexed: 12/16/2022] Open
Abstract
A number of natural and chemical compounds that exert anti-oxidative properties are demonstrated to be beneficial for brain and cognitive function, and some are reported to reduce the risk of dementia. However, the detailed mechanisms by which those anti-oxidative compounds show positive effects on cognition and dementia are still unclear. An emerging body of evidence suggests that the integrity of the cerebrovascular blood-brain barrier (BBB) is centrally involved in the onset and progression of cognitive impairment and dementia. While recent studies revealed that some anti-oxidative agents appear to be protective against the disruption of BBB integrity and structure, few studies considered the neuroprotective effects of antioxidants in the context of cerebrovascular integrity. Therefore, in this review, we examine the mechanistic insights of antioxidants as a pleiotropic agent for cognitive impairment and dementia through a cerebrovascular axis by primarily focusing on the current available data from physiological studies. Conclusively, there is a compelling body of evidence that suggest antioxidants may prevent cognitive decline and dementia by protecting the integrity and function of BBB and, indeed, further studies are needed to directly examine these effects in addition to underlying molecular mechanisms.
Collapse
Affiliation(s)
- Virginie Lam
- Curtin Health Innovation Research Institute, Curtin University, Perth WA 6845, Australia.
- School of Public Health, Faculty of Health Sciences, Curtin University, Perth WA 6845, Australia.
| | - Mark Hackett
- Curtin Health Innovation Research Institute, Curtin University, Perth WA 6845, Australia.
- Department of Chemistry, Faculty of Science and Engineering, Curtin University, Perth WA 6845, Australia.
| | - Ryusuke Takechi
- Curtin Health Innovation Research Institute, Curtin University, Perth WA 6845, Australia.
- School of Public Health, Faculty of Health Sciences, Curtin University, Perth WA 6845, Australia.
| |
Collapse
|
33
|
Li CQ, Zheng Q, Wang Q, Zeng QP. Biotic/Abiotic Stress-Driven Alzheimer's Disease. Front Cell Neurosci 2016; 10:269. [PMID: 27932953 PMCID: PMC5120111 DOI: 10.3389/fncel.2016.00269] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 11/07/2016] [Indexed: 01/10/2023] Open
Affiliation(s)
- Chang-Qing Li
- Tropical Medicine Institute, Guangzhou University of Chinese Medicine Guangzhou, China
| | - Qing Zheng
- Department of Biopharmaceutics, College of Pharmacy, Jinan University Guangzhou, China
| | - Qi Wang
- Clinical Pharmacology Institute, Guangzhou University of Chinese Medicine Guangzhou, China
| | - Qing-Ping Zeng
- Tropical Medicine Institute, Guangzhou University of Chinese Medicine Guangzhou, China
| |
Collapse
|
34
|
Neuroprotective Effects of Methyl 3,4-Dihydroxybenzoate against TBHP-Induced Oxidative Damage in SH-SY5Y Cells. Molecules 2016; 21:molecules21081071. [PMID: 27556437 PMCID: PMC6274188 DOI: 10.3390/molecules21081071] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 08/08/2016] [Accepted: 08/10/2016] [Indexed: 11/17/2022] Open
Abstract
This study investigated the neuroprotective effects of methyl 3,4-dihydroxybenzoate (MDHB) against t-butyl hydroperoxide (TBHP) induced oxidative damage in SH-SY5Y (human neuroblastoma cells) and the underlying mechanisms. SH-SY5Y were cultured in DMEM + 10% FBS for 24 h and pretreated with different concentrations of MDHB or N-acetyl-l-cysteine (NAC) for 4 h prior to the addition of 40 μM TBHP for 24 h. Cell viability was analyzed using the methylthiazolyltetrazolium (MTT) and lactate dehydrogenase (LDH) assays. An annexin V-FITC assay was used to detect cell apoptosis rates. The 2',7'-dichlorofluorescin diacetate (DCFH-DA) assay was used to determine intracellular ROS levels. The activities of antioxidative enzymes (GSH-Px and SOD) were measured using commercially available kits. The oxidative DNA damage marker 8-OHdG was detected using ELISA. Western blotting was used to determine the expression of Bcl-2, Bax, caspase 3, p-Akt and Akt proteins in treated SH-SY5Y cells. Our results showed that MDHB is an effective neuroprotective compound that can mitigate oxidative stress and inhibit apoptosis in SH-SY5Y cells.
Collapse
|
35
|
Yamamoto N, Fujii Y, Kasahara R, Tanida M, Ohora K, Ono Y, Suzuki K, Sobue K. Simvastatin and atorvastatin facilitates amyloid β-protein degradation in extracellular spaces by increasing neprilysin secretion from astrocytes through activation of MAPK/Erk1/2 pathways. Glia 2016; 64:952-62. [PMID: 26875818 DOI: 10.1002/glia.22974] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 12/19/2015] [Accepted: 01/13/2016] [Indexed: 12/21/2022]
Abstract
One of the major neuropathological hallmarks of Alzheimer's disease (AD) is the deposition of amyloid β-protein (Aβ) in the brain. Aβ accumulation seems to arise from an imbalance between Aβ production and clearance. Neprilysin (NEP) and insulin-degrading enzyme (IDE) are the important Aβ-degrading enzymes in the brain, and deficits in their expression may promote Aβ deposition in patients with sporadic late-onset AD. Statins, which are used clinically for reducing cholesterol levels, can exert beneficial effects on AD. Therefore, we examined whether various statins are associated with Aβ degradation by inducing NEP and IDE expression, and then evaluating the relation between activation of intracellular signaling transduction, inhibition of cholesterol production, and morphological changes to astrocytes. Treating cultured rat astrocytes with simvastatin and atorvastatin significantly decreased the expression of NEP but not IDE in a concentration- and time-dependent manner. The decrease in NEP expression was a result of activation of extracellular signal-regulated kinase (ERK) but not the reduction of cholesterol synthesis pathway. This NEP reduction was achieved by the release to the extracellular space of cultured astrocytes. Furthermore, the cultured medium prepared from simvastatin- and atorvastatin-treated astrocytes significantly induced the degradation of exogenous Aβ. These results suggest that simvastatin and atorvastatin induce the increase of Aβ degradation of NEP on the extracellular of astrocytes by inducing ERK-mediated pathway activity and that these reagents regulate the differential mechanisms between the secretion of NEP, the induction of cholesterol reduction, and the morphological changes in the cultured astrocytes.
Collapse
Affiliation(s)
- Naoki Yamamoto
- Faculty of Pharmaceutical Sciences, Hokuriku University, Kanazawa, Ishikawa, 920-1181, Japan.,Laboratory of Neurochemistry, Department of Pharmacy, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, 525-8577, Japan
| | - Yoko Fujii
- Laboratory of Neurochemistry, Department of Pharmacy, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, 525-8577, Japan
| | - Rika Kasahara
- Laboratory of Neurochemistry, Department of Pharmacy, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, 525-8577, Japan
| | - Mamoru Tanida
- Department of Physiology II, Kanazawa Medical University, Uchinada, Ishikawa, 920-0293, Japan
| | - Kentaro Ohora
- Laboratory of Neurochemistry, Department of Pharmacy, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, 525-8577, Japan
| | - Yoko Ono
- Laboratory of Neurochemistry, Department of Pharmacy, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, 525-8577, Japan
| | - Kenji Suzuki
- Laboratory of Molecular Medicinal Science, Department of Pharmacy, College of Pharmaceutical Sciences, Ritsumeikan University, Kusatsu, Shiga, 525-8577, Japan
| | - Kazuya Sobue
- Department of Anesthesiology and Medical Crisis Management, Nagoya City University Graduate School of Medical Sciences, Nagoya City, Aichi, 467-8622, Japan
| |
Collapse
|
36
|
Chen X, Bakillah A, Zhou L, Pan X, Hoepfner F, Jacob M, Jiang XC, Lazar J, Schlitt A, Hussain MM. Nitrated apolipoprotein AI/apolipoprotein AI ratio is increased in diabetic patients with coronary artery disease. Atherosclerosis 2016; 245:12-21. [PMID: 26687998 PMCID: PMC4738057 DOI: 10.1016/j.atherosclerosis.2015.11.021] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2015] [Revised: 11/13/2015] [Accepted: 11/19/2015] [Indexed: 12/17/2022]
Abstract
AIMS/HYPOTHESIS Recent studies have suggested that determination of HDL function may be more informative than its concentration in predicting its protective role in coronary artery disease (CAD). Apolipoprotein AI (apoAI), the major protein of HDL, is nitrosylated in vivo to nitrated apoAI (NT-apoAI) that might cause dysfunction. We hypothesized that NT-apoAI/apoAI ratio might be associated with diabetes mellitus (DM) in CAD patients. METHODS We measured plasma NT-apoAI and apoAI levels in 777 patients with coronary artery disease (CAD) by ELISA. Further, we measured plasma cholesterol efflux potential in subjects with similar apoAI but different NT-apoAI levels. RESULTS We found that median NT-apoAI/apoAI ratio was significantly higher in diabetes mellitus (DM) (n = 327) versus non-diabetic patients (n = 450). Further analysis indicated that DM, thiobarbituric acid-reactive substances and C-reactive protein levels were independent predictors of higher NT-apoAI/apoAI ratio. There was negative correlation between NT-apoAI/apoAI and use of anti-platelet and lipid lowering drugs. The cholesterol efflux capacity of plasma from 67 individuals with differing NT-apoAI but similar apoAI levels from macrophages in vitro was negatively correlated with NT-apoAI/apoAI ratio. CONCLUSIONS Higher NT-apoAI/apoAI ratio is significantly associated with DM in this relatively large German cohort with CAD and may contribute to associated complications by reducing cholesterol efflux capacity.
Collapse
Affiliation(s)
- Xueying Chen
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY, USA; Institute of Cardiology, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | - Ahmed Bakillah
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Liye Zhou
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Xiaoyue Pan
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | | | - Marrit Jacob
- Department of Medicine III, University Clinic Halle, Germany
| | - Xian-Cheng Jiang
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY, USA; VA New York Harbor Healthcare System, Brooklyn, NY 11209, USA
| | - Jason Lazar
- Division of Cardiovascular Medicine, SUNY Downstate Medical Center, Brooklyn, NY, USA
| | - Axel Schlitt
- Department of Medicine III, University Clinic Halle, Germany; Paracelsus-Harz-Clinic Bad Suderode, Quedlinburg, Germany
| | - M Mahmood Hussain
- Department of Cell Biology, SUNY Downstate Medical Center, Brooklyn, NY, USA; VA New York Harbor Healthcare System, Brooklyn, NY 11209, USA; Department of Pediatrics, SUNY Downstate Medical Center, Brooklyn, NY, USA.
| |
Collapse
|
37
|
Dattilo S, Mancuso C, Koverech G, Di Mauro P, Ontario ML, Petralia CC, Petralia A, Maiolino L, Serra A, Calabrese EJ, Calabrese V. Heat shock proteins and hormesis in the diagnosis and treatment of neurodegenerative diseases. Immun Ageing 2015; 12:20. [PMID: 26543490 PMCID: PMC4634585 DOI: 10.1186/s12979-015-0046-8] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Accepted: 10/15/2015] [Indexed: 12/16/2022]
Abstract
Modulation of endogenous cellular defense mechanisms via the vitagene system represents an innovative approach to therapeutic intervention in diseases causing chronic tissue damage, such as in neurodegeneration. The possibility of high-throughoutput screening using proteomic techniques, particularly redox proteomics, provide more comprehensive overview of the interaction of proteins, as well as the interplay among processes involved in neuroprotection. Here by introducing the hormetic dose response concept, the mechanistic foundations and applications to the field of neuroprotection, we discuss the emerging role of heat shock protein as prominent member of vitagene network in neuroprotection and redox proteomics as a tool for investigating redox modulation of stress responsive vitagenes. Hormetic mechanisms are reviewed as possibility of targeted therapeutic manipulation in a cell-, tissue- and/or pathway-specific manner at appropriate points in the neurodegenerative disease process.
Collapse
Affiliation(s)
- Sandro Dattilo
- />Department of Biomedical and Biotechnological Sciences, University of Catania, Via Andrea Doria, 95100 Catania, Italy
| | - Cesare Mancuso
- />Institute of Pharmacology, Catholic University School of Medicine, Rome, Italy
| | - Guido Koverech
- />Department of Biomedical and Biotechnological Sciences, University of Catania, Via Andrea Doria, 95100 Catania, Italy
| | - Paola Di Mauro
- />Department of Medical and Surgery Specialties, University of Catania, Catania, Italy
| | - Maria Laura Ontario
- />Department of Biomedical and Biotechnological Sciences, University of Catania, Via Andrea Doria, 95100 Catania, Italy
| | | | - Antonino Petralia
- />Department of Clinical and Experimental Medicine, School of Medicine, University of Catania, Catania, Italy
| | - Luigi Maiolino
- />Department of Medical and Surgery Specialties, University of Catania, Catania, Italy
| | - Agostino Serra
- />Department of Medical and Surgery Specialties, University of Catania, Catania, Italy
| | - Edward J. Calabrese
- />Environmental Health Sciences Division, School of Public Health, University of Massachusetts, Amherst, MA USA
| | - Vittorio Calabrese
- />Department of Biomedical and Biotechnological Sciences, University of Catania, Via Andrea Doria, 95100 Catania, Italy
| |
Collapse
|
38
|
Filosa S, Pecorelli A, D'Esposito M, Valacchi G, Hajek J. Exploring the possible link between MeCP2 and oxidative stress in Rett syndrome. Free Radic Biol Med 2015; 88:81-90. [PMID: 25960047 DOI: 10.1016/j.freeradbiomed.2015.04.019] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Revised: 04/10/2015] [Accepted: 04/13/2015] [Indexed: 01/27/2023]
Abstract
Rett syndrome (RTT, MIM 312750) is a rare and orphan progressive neurodevelopmental disorder affecting girls almost exclusively, with a frequency of 1/15,000 live births of girls. The disease is characterized by a period of 6 to 18 months of apparently normal neurodevelopment, followed by early neurological regression, with a progressive loss of acquired cognitive, social, and motor skills. RTT is known to be caused in 95% of the cases by sporadic de novo loss-of-function mutations in the X-linked methyl-CpG-binding protein 2 (MECP2) gene encoding methyl-CpG binding protein 2 (MeCP2), a nuclear protein able to regulate gene expression. Despite almost two decades of research into the functions and role of MeCP2, little is known about the mechanisms leading from MECP2 mutation to the disease. Oxidative stress (OS) is involved in the pathogenic mechanisms of several neurodevelopmental and neurodegenerative disorders, although in many cases it is not clear whether OS is a cause or a consequence of the pathology. Fairly recently, the presence of a systemic OS has been demonstrated in RTT patients with a strong correlation with the patients' clinical status. The link between MECP2 mutation and the redox imbalance found in RTT is not clear. Animal studies have suggested a possible direct correlation between Mecp2 mutation and increased OS levels. In addition, the restoration of Mecp2 function in astrocytes significantly improves the developmental outcome of Mecp2-null mice and reexpression of Mecp2 gene in the brain of null mice restored oxidative damage, suggesting that Mecp2 loss of function can be involved in oxidative brain damage. Starting from the evidence that oxidative damage in the brain of Mecp2-null mice precedes the onset of symptoms, we evaluated whether, based on the current literature, the dysfunctions described in RTT could be a consequence or, in contrast, could be caused by OS. We also analyzed whether therapies that at least partially treated some RTT symptoms can play a role in defense against OS. At this stage we can propose that OS could be one of the main causes of the dysfunctions observed in RTT. In addition, the major part of the therapies recommended to alleviate RTT symptoms have been shown to interfere with oxidative homeostasis, suggesting that MeCP2 could somehow be involved in the protection of the brain from OS.
Collapse
Affiliation(s)
- Stefania Filosa
- Institute of Biosciences and BioResources-CNR, UOS Naples, Italy; IRCCS Neuromed, Pozzilli, Italy
| | - Alessandra Pecorelli
- Child Neuropsychiatry Unit, University General Hospital, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Maurizio D'Esposito
- Institute of Genetics and Biophysics "A. Buzzati-Traverso"-CNR, Naples, Italy; IRCCS Neuromed, Pozzilli, Italy
| | - Giuseppe Valacchi
- Department of Life Sciences and Biotechnology, University of Ferrara, Ferrara, Italy; Department of Food and Nutrition, Kyung Hee University, Seoul, South Korea.
| | - Joussef Hajek
- Child Neuropsychiatry Unit, University General Hospital, Azienda Ospedaliera Universitaria Senese, Siena, Italy
| |
Collapse
|
39
|
Mendoza-Oliva A, Ferrera P, Fragoso-Medina J, Arias C. Lovastatin Differentially Affects Neuronal Cholesterol and Amyloid-β Production in vivo and in vitro. CNS Neurosci Ther 2015; 21:631-41. [PMID: 26096465 DOI: 10.1111/cns.12420] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 05/08/2015] [Accepted: 05/22/2015] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND AIMS Epidemiological and experimental studies indicate that high cholesterol may increase susceptibility to age-associated neurodegenerative disorders, such as Alzheimer's disease (AD). Thus, it has been suggested that statins, which are inhibitors of the enzyme 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR), may be a useful therapeutic tool to diminish the risk of AD. However, several studies that analyzed the therapeutic benefits of statins have yielded conflicting results. Herein, we investigated the role of lovastatin on neuronal cholesterol homeostasis and its effects on amyloid β protein production in vivo and in vitro. METHODS AND RESULTS Lovastatin effects were analyzed in vitro using differentiated human neuroblastoma cells and in vivo in a lovastatin-fed rat model. We demonstrated that lovastatin can differentially affect the expression of APP and Aβ production in vivo and in vitro. Lovastatin-induced HMGCR inhibition was detrimental to neuronal survival in vitro via a mechanism unrelated to the reduction of cholesterol. We found that in vivo, dietary cholesterol was associated with increased Aβ production in the cerebral cortex, and lovastatin was not able to reduce cholesterol levels. However, lovastatin induced a remarkable increase in the mature form of the sterol regulatory element-binding protein-2 (SREBP-2) as well as its target gene HMGCR, in both neuronal cells and in the brain. CONCLUSIONS Lovastatin modifies the mevalonate pathway without affecting cholesterol levels in vivo and is able to reduce Aβ levels only in vitro.
Collapse
Affiliation(s)
- Aydé Mendoza-Oliva
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México D.F, México
| | - Patricia Ferrera
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México D.F, México
| | - Jorge Fragoso-Medina
- Departmento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, México D.F, México
| | - Clorinda Arias
- Departamento de Medicina Genómica y Toxicología Ambiental, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, México D.F, México
| |
Collapse
|
40
|
Li N, Yang YJ, Qian HY, Li Q, Zhang Q, Li XD, Dong QT, Xu H, Song L, Zhang H. Intravenous administration of atorvastatin-pretreated mesenchymal stem cells improves cardiac performance after acute myocardial infarction: role of CXCR4. Am J Transl Res 2015; 7:1058-1070. [PMID: 26279750 PMCID: PMC4532739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Accepted: 06/14/2015] [Indexed: 06/04/2023]
Abstract
BACKGROUND The interaction between stromal cell-derived factor 1 (SDF-1) and its receptor CXC chemokine receptor 4 (CXCR4) plays an important role in mesenchymal stem cells (MSCs) migration and engraftment. Statins can increase the survival of MSCs. However, whether statins could enhance MSCs migration and engraftment is still unknown. Therefore, we designed the study to investigate whether atorvastatin (ATV) could enhance CXCR4 expression of MSCs and promote them homing toward the injured myocardium. METHODS AND RESULTS Expression of CXCR4 was evaluated by flow cytometry and real time PCR. A transwell system was used to assess MSCs migration ability. Recruitment of systematically delivered MSCs to the infarcted heart was evaluated in Sprague-Dawley rats with acute myocardial infarction (AMI). ATV pretreatment enhanced the expression of CXCR4 and stimulated MSCs migration in vitro. However, the effect was largely abolished by CXCR4 neutralizing antibody. In AMI models, we found much more ATV-pretreated MSCs homing toward the infarcted myocardium than non-treated cells and this was accompanied by improved cardiac performance. CONCLUSIONS ATV increases the migration ability of MSCs and improves cardiac performance due to up-regulated expression of CXCR4. These results suggest that ATV pretreatment of donor MSCs is an effective way to promote cell therapeutic potential for AMI.
Collapse
Affiliation(s)
- Na Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100037, People's Republic of China
| | - Yue-Jin Yang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100037, People's Republic of China
| | - Hai-Yan Qian
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100037, People's Republic of China
| | - Qing Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100037, People's Republic of China
| | - Qian Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100037, People's Republic of China
| | - Xiang-Dong Li
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100037, People's Republic of China
| | - Qiu-Ting Dong
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100037, People's Republic of China
| | - Hui Xu
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100037, People's Republic of China
| | - Lei Song
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100037, People's Republic of China
| | - Hao Zhang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences and Peking Union Medical College Beijing 100037, People's Republic of China
| |
Collapse
|
41
|
Wanamaker BL, Swiger KJ, Blumenthal RS, Martin SS. Cholesterol, statins, and dementia: what the cardiologist should know. Clin Cardiol 2015; 38:243-50. [PMID: 25869997 PMCID: PMC4409455 DOI: 10.1002/clc.22361] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Revised: 10/22/2014] [Accepted: 10/26/2014] [Indexed: 01/22/2023] Open
Abstract
Alzheimer dementia (AD) is an important clinical problem that appears to be closely tied to comorbid cardiovascular disease, making it a relevant topic for the clinical cardiologist. Determinants of cardiovascular health, especially midlife dyslipidemia, are associated with an increased risk of dementia based on molecular and epidemiologic data. Given the potential role of dyslipidemia in the development of dementia, statins have been investigated as potential therapeutic options to slow or prevent disease. This review discusses the role of dyslipidemia and other cardiovascular risk factors in the pathogenesis of AD, with a focus on the existing evidence for the use of statin medications in the treatment and prevention of AD from observational studies and randomized clinical trials. Clinical questions for the practicing cardiologist are addressed.
Collapse
Affiliation(s)
- Brett L. Wanamaker
- Department of Medicine, Division of Cardiology, The Johns Hopkins Ciccarone Center for the Prevention of Heart DiseaseJohns Hopkins UniversityBaltimoreMaryland
| | - Kristopher J. Swiger
- Department of Medicine, Division of Cardiology, The Johns Hopkins Ciccarone Center for the Prevention of Heart DiseaseJohns Hopkins UniversityBaltimoreMaryland
| | - Roger S. Blumenthal
- Department of Medicine, Division of Cardiology, The Johns Hopkins Ciccarone Center for the Prevention of Heart DiseaseJohns Hopkins UniversityBaltimoreMaryland
| | - Seth S. Martin
- Department of Medicine, Division of Cardiology, The Johns Hopkins Ciccarone Center for the Prevention of Heart DiseaseJohns Hopkins UniversityBaltimoreMaryland
| |
Collapse
|
42
|
Atorvastatin Prevents Cognitive Deficits Induced by Intracerebroventricular Amyloid-β1–40 Administration in Mice: Involvement of Glutamatergic and Antioxidant Systems. Neurotox Res 2015; 28:32-42. [DOI: 10.1007/s12640-015-9527-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 03/12/2015] [Accepted: 03/19/2015] [Indexed: 12/12/2022]
|
43
|
Targeting transporters: promoting blood-brain barrier repair in response to oxidative stress injury. Brain Res 2015; 1623:39-52. [PMID: 25796436 DOI: 10.1016/j.brainres.2015.03.018] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2015] [Revised: 03/09/2015] [Accepted: 03/10/2015] [Indexed: 01/06/2023]
Abstract
The blood-brain barrier (BBB) is a physical and biochemical barrier that precisely regulates the ability of endogenous and exogenous substances to accumulate within brain tissue. It possesses structural and biochemical features (i.e., tight junction and adherens junction protein complexes, influx and efflux transporters) that work in concert to control solute permeation. Oxidative stress, a critical component of several diseases including cerebral hypoxia/ischemia and peripheral inflammatory pain, can cause considerable injury to the BBB and lead to significant CNS pathology. This suggests a critical need for novel therapeutic approaches that can protect the BBB in diseases with an oxidative stress component. Recent studies have identified molecular targets (i.e., putative membrane transporters, intracellular signaling systems) that can be exploited for optimization of endothelial drug delivery or for control of transport of endogenous substrates such as the antioxidant glutathione (GSH). In particular, targeting transporters offers a unique approach to protect BBB integrity by promoting repair of cell-cell interactions at the level of the brain microvascular endothelium. This review summarizes current knowledge in this area and emphasizes those targets that present considerable opportunity for providing BBB protection and/or promoting BBB repair in the setting of oxidative stress. This article is part of a Special Issue entitled SI: Cell Interactions In Stroke.
Collapse
|
44
|
Sehar N, Agarwal NB, Vohora D, Raisuddin S. Atorvastatin prevents development of kindling by modulating hippocampal levels of dopamine, glutamate, and GABA in mice. Epilepsy Behav 2015; 42:48-53. [PMID: 25499163 DOI: 10.1016/j.yebeh.2014.11.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Revised: 10/20/2014] [Accepted: 11/10/2014] [Indexed: 10/24/2022]
Abstract
PURPOSE Atorvastatin (ATV) is widely used for the treatment of dyslipidemias. Recent evidence has shown that ATV has protection effects against seizures. However, the effect of ATV on certain neurotransmitter and oxidative stress markers associated with seizures had not been reported. Therefore, the present study aimed to evaluate the effects of ATV on oxidative stress markers on whole brain and GABA, glutamate, and dopamine levels in the hippocampus of PTZ-kindled mice. Additionally, effects of ATV on animal models of seizures, anxiety, and depression were also assessed. MATERIALS AND METHODS Swiss albino mice were given ATV (20, 40, and 80mg/kg/p.o.) in an acute study. On the seventh day, animals were subjected to various neurological and neurobehavioral tests, viz, increasing current electroshock (ICES) test, pentylenetetrazole (PTZ)-induced seizures, Elevated Plus Maze (EPM), and Forced Swim Test (FST). For the development of kindling, a subconvulsant dose of PTZ, i.e., 25mg/kg, i.p., was administered every other day, and ATV in all the three doses was administered daily. Seizure score was continuously monitored until the development of kindling. Thiobarbituric acid reacting species (TBARS), glutathione, dopamine, GABA, and glutamate levels were also assessed in the brain tissues of mice. RESULTS The results showed that in the ICES test, ATV 80mg/kg increased the seizure threshold to hind limb extension (HLE), and a complete protection against HLE was observed when ATV 80mg/kg was combined with a subanticonvulsant dose of phenytoin. Atorvastatin in all the tested doses suppressed the development of kindling, reduced lipid peroxidation, and increased glutathione levels. All doses of ATV maintained the normal levels of glutamate, GABA, and dopamine in kindled mice. CONCLUSION Atorvastatin possesses anticonvulsant activity against electroconvulsions. It was found to suppress the development of PTZ kindling, presumably altering the redox status and hippocampal levels of dopamine, glutamate, and GABA.
Collapse
Affiliation(s)
- Nouroz Sehar
- Centre for Translational and Clinical Research, Jamia Hamdard, New Delhi 110062, India; Department of Medical Elementology and Toxicology, Jamia Hamdard, New Delhi 110062, India.
| | - Nidhi Bharal Agarwal
- Centre for Translational and Clinical Research, Jamia Hamdard, New Delhi 110062, India.
| | - Divya Vohora
- Department of Pharmacology, Faculty of Pharmacy, Jamia Hamdard, New Delhi 110062, India.
| | - S Raisuddin
- Centre for Translational and Clinical Research, Jamia Hamdard, New Delhi 110062, India; Department of Medical Elementology and Toxicology, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
45
|
McFarland AJ, Anoopkumar-Dukie S, Arora DS, Grant GD, McDermott CM, Perkins AV, Davey AK. Molecular mechanisms underlying the effects of statins in the central nervous system. Int J Mol Sci 2014; 15:20607-37. [PMID: 25391045 PMCID: PMC4264186 DOI: 10.3390/ijms151120607] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 10/23/2014] [Accepted: 10/30/2014] [Indexed: 02/06/2023] Open
Abstract
3-Hydroxy-3-methylglutaryl coenzyme A reductase inhibitors, commonly referred to as statins, are widely used in the treatment of dyslipidaemia, in addition to providing primary and secondary prevention against cardiovascular disease and stroke. Statins’ effects on the central nervous system (CNS), particularly on cognition and neurological disorders such as stroke and multiple sclerosis, have received increasing attention in recent years, both within the scientific community and in the media. Current understanding of statins’ effects is limited by a lack of mechanism-based studies, as well as the assumption that all statins have the same pharmacological effect in the central nervous system. This review aims to provide an updated discussion on the molecular mechanisms contributing to statins’ possible effects on cognitive function, neurodegenerative disease, and various neurological disorders such as stroke, epilepsy, depression and CNS cancers. Additionally, the pharmacokinetic differences between statins and how these may result in statin-specific neurological effects are also discussed.
Collapse
Affiliation(s)
| | | | - Devinder S Arora
- School of Pharmacy, Griffith University, Queensland 4222, Australia.
| | - Gary D Grant
- School of Pharmacy, Griffith University, Queensland 4222, Australia.
| | | | - Anthony V Perkins
- Griffith Health Institute, Griffith University, Queensland 4222, Australia.
| | - Andrew K Davey
- School of Pharmacy, Griffith University, Queensland 4222, Australia.
| |
Collapse
|
46
|
Abstract
Cerebral ischemia occurs when blood flow to the brain is insufficient to meet metabolic demand. This can result from cerebral artery occlusion that interrupts blood flow, limits CNS supply of oxygen and glucose, and causes an infarction/ischemic stroke. Ischemia initiates a cascade of molecular events in neurons and cerebrovascular endothelial cells including energy depletion, dissipation of ion gradients, calcium overload, excitotoxicity, oxidative stress, and accumulation of ions and fluid. Blood-brain barrier (BBB) disruption is associated with cerebral ischemia and leads to vasogenic edema, a primary cause of stroke-associated mortality. To date, only a single drug has received US Food and Drug Administration (FDA) approval for acute ischemic stroke treatment, recombinant tissue plasminogen activator (rt-PA). While rt-PA therapy restores perfusion to ischemic brain, considerable tissue damage occurs when cerebral blood flow is reestablished. Therefore, there is a critical need for novel therapeutic approaches that can "rescue" salvageable brain tissue and/or protect BBB integrity during ischemic stroke. One class of drugs that may enable neural cell rescue following cerebral ischemia/reperfusion injury is the HMG-CoA reductase inhibitors (i.e., statins). Understanding potential CNS drug delivery pathways for statins is critical to their utility in ischemic stroke. Here, we review molecular pathways associated with cerebral ischemia and novel approaches for delivering drugs to treat ischemic disease. Specifically, we discuss utility of endogenous BBB drug uptake transporters such as organic anion transporting polypeptides and nanotechnology-based carriers for optimization of CNS drug delivery. Overall, this chapter highlights state-of-the-art technologies that may improve pharmacotherapy of cerebral ischemia.
Collapse
|
47
|
The Therapeutic Potential of Atorvastatin in a Mouse Model of Postoperative Cognitive Decline. Ann Surg 2014; 259:1235-44. [DOI: 10.1097/sla.0000000000000257] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
48
|
Protective effects of a wheat germ peptide (RVF) against H2O 2-induced oxidative stress in human neuroblastoma cells. Biotechnol Lett 2014; 36:1615-22. [PMID: 24737076 DOI: 10.1007/s10529-014-1521-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 03/25/2014] [Indexed: 01/14/2023]
Abstract
RVF (Arg-Val-Phe), a peptide derived from wheat germ, shows antioxidant properties. Here, the neuroprotective efficacies of RVF were investigated in human neuroblastoma cells (SH-SY5Y) that were pretreated with RVF (150-250 μM, 4 h) and exposed to H2O2 (200 μM). RVF increased viable cell numbers by 37 % and reduced the release of lactate dehydrogenase. Pretreatment with RVF also inhibited H2O2-induced accumulation of reactive oxygen species and maintained the mitochondrial transmembrane potential as well as preventing intracellular Ca(2+) dysregulation during H2O2 exposure. Furthermore, pretreatment with RVF increased the Bcl-2/Bax ratio and blocked cleavage poly(ADP-ribose) polymerase by inhibiting caspase-3 activation, thus decreasing apoptosis.
Collapse
|
49
|
Honarmand AR, Pourtabatabaei N, Rahimi N, Dehpour AR, Javadi-Paydar M. Suppression of memory acquisition following co-administration of lithium and atorvastatin through nitric oxide pathway in mice. Pharmacol Biochem Behav 2014; 122:203-11. [PMID: 24708995 DOI: 10.1016/j.pbb.2014.03.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 02/28/2014] [Accepted: 03/27/2014] [Indexed: 01/09/2023]
Abstract
PURPOSE The aim of this study was to investigate the interactive effect of lithium and atorvastatin on cognitive performance and the role of NO as a potential mechanism involved in this interaction. MATERIALS AND METHODS Memory performance was evaluated in a two-trial recognition Y-maze test and a step-through passive avoidance task in mice. Lithium (5, 10, 20 or 40 mg/kg, i.p.) and atorvastatin (1 mg/kg, p.o.) were administered 1 h before each trial, L-NAME, a non-specific NO synthase inhibitor (3, 10 mg/kg, i.p.); aminoguanidine, a specific inducible NO synthase (iNOS) inhibitor (100 mg/kg); and L-arginine, a NO precursor (750 mg/kg) were administered 30 min before training sessions. The level of plasma NO end-products (NOx) was determined using Griess reagent protocol. RESULTS 1) Lithium (40 mg/kg) impaired the acquisition of spatial recognition memory; 2) lithium did not affect the retrieval phase of spatial memory; 3) atorvastatin (1 mg/kg) significantly impaired the memory performance, when co-administered with the sub-effective dose of lithium (10 mg/kg), but did not affect the status when administered with lithium (5 mg/kg); 4) L-NAME (10 mg/kg) and aminoguanidine (100 mg/kg) dramatically decreased memory performance in mice received sub-effective doses of both lithium (5 mg/kg) and atorvastatin (1 mg/kg); 5) L-arginine (750 mg/kg) improved the memory acquisition in mice administered lithium (10 mg/kg) and atorvastatin (1 mg/kg); 6) lithium did not affect the cognitive performance in the passive avoidance test. All results were compatible and confirmed with in vitro determination of plasma NOx levels. CONCLUSIONS Lithium, dose dependently, impaired acquisition phase of spatial recognition memory. Lithium and atorvastatin co-administration impaired spatial recognition memory mediating by nitrergic pathway. In addition to L-arginine, our data from L-NAME and aminoguanidine also support the involvement of NO pathway in this interaction.
Collapse
Affiliation(s)
- Amir Reza Honarmand
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Iran; Brain and Spinal Cord Injury Repair Research Center, Tehran University of Medical Sciences, Iran
| | - Nasim Pourtabatabaei
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Iran; Brain and Spinal Cord Injury Repair Research Center, Tehran University of Medical Sciences, Iran
| | - Nastaran Rahimi
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Iran; Experimental Medicine Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Iran; Experimental Medicine Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehrak Javadi-Paydar
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Iran; Brain and Spinal Cord Injury Repair Research Center, Tehran University of Medical Sciences, Iran.
| |
Collapse
|
50
|
Davis PR, Head E. Prevention approaches in a preclinical canine model of Alzheimer's disease: benefits and challenges. Front Pharmacol 2014; 5:47. [PMID: 24711794 PMCID: PMC3968758 DOI: 10.3389/fphar.2014.00047] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2014] [Accepted: 02/28/2014] [Indexed: 12/30/2022] Open
Abstract
Aged dogs spontaneously develop many features of human aging and Alzheimer's disease (AD) including cognitive decline and neuropathology. In this review, we discuss age-dependent learning tasks, memory tasks, and functional measures that can be used in aged dogs for sensitive treatment outcome measures. Neuropathology that is linked to cognitive decline is described along with examples of treatment studies that show reduced neuropathology in aging dogs (dietary manipulations, behavioral enrichment, immunotherapy, and statins). Studies in canine show that multi-targeted approaches may be more beneficial than single pathway manipulations (e.g., antioxidants combined with behavioral enrichment). Aging canine studies show good predictive validity for human clinical trials outcomes (e.g., immunotherapy) and several interventions tested in dogs strongly support a prevention approach (e.g., immunotherapy and statins). Further, dogs are ideally suited for prevention studies as they the age because onset of cognitive decline and neuropathology strongly support longitudinal interventions that can be completed within a 3-5 year period. Disadvantages to using the canine model are that they lengthy, use labor-intensive comprehensive cognitive testing, and involve costly housing (almost as high as that of non-human primates). However, overall, using the dog as a preclinical model for testing preventive approaches for AD may complement work in rodents and non-human primates.
Collapse
Affiliation(s)
- Paulina R Davis
- Sanders-Brown Center on Aging, University of Kentucky Lexington, KY, USA ; Department of Molecular and Biomedical Pharmacology, University of Kentucky Lexington, KY, USA
| | - Elizabeth Head
- Sanders-Brown Center on Aging, University of Kentucky Lexington, KY, USA ; Department of Molecular and Biomedical Pharmacology, University of Kentucky Lexington, KY, USA
| |
Collapse
|