1
|
Hameed MQ, D'Ambrosio R, Eastman C, Hui B, Lin R, Vermudez SAD, Liebhardt A, Choe Y, Klein P, Rundfeldt C, Löscher W, Rotenberg A. A comparison of the antiepileptogenic efficacy of two rationally chosen multitargeted drug combinations in a rat model of posttraumatic epilepsy. Exp Neurol 2024; 382:114962. [PMID: 39288831 DOI: 10.1016/j.expneurol.2024.114962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/08/2024] [Accepted: 09/13/2024] [Indexed: 09/19/2024]
Abstract
Post-traumatic epilepsy (PTE) is a recurrent and often drug-refractory seizure disorder caused by traumatic brain injury (TBI). No single drug treatment prevents PTE, but preventive drug combinations that may prophylax against PTE have not been studied. Based on a systematic evaluation of rationally chosen drug combinations in the intrahippocampal kainate (IHK) mouse model of acquired epilepsy, we identified two multi-targeted drug cocktails that exert strong antiepileptogenic effects. The first, a combination of levetiracetam (LEV) and topiramate, only partially prevented spontaneous recurrent seizures in the model. We therefore added atorvastatin (ATV) to the therapeutic cocktail (TC) to increase efficacy, forming "TC-001". The second cocktail - a combination of LEV, ATV, and ceftriaxone, termed "TC-002" - completely prevented epilepsy in the mouse IHK model. In the present proof-of-concept study, we tested whether the two drug cocktails prevent epilepsy in a rat PTE model in which recurrent electrographic seizures develop after severe rostral parasagittal fluid percussion injury (FPI). Following FPI, rats were either treated over 3-4 weeks with vehicle or drug cocktails, starting either 1 or 4-6 h after the injury. Using mouse doses of TC-001 and TC-002, no significant antiepileptogenic effect was obtained in the rat PTE model. However, when using allometric scaling of drug doses to consider the differences in body surface area between mice and rats, PTE was prevented by TC-002. Furthermore, the latter drug cocktail partially prevented the loss of perilesional cortical parvalbumin-positive GABAergic interneurons. Plasma and brain drug analysis showed that these effects of TC-002 occurred at clinically relevant levels of the individual TC-002 drug components. In silico analysis of drug-drug brain protein interactions by the STITCH database indicated that TC-002 impacts a larger functional network of epilepsy-relevant brain proteins than each drug alone, providing a potential network pharmacology explanation for the observed antiepileptogenic and neuroprotective effects observed with this combination.
Collapse
Affiliation(s)
- Mustafa Q Hameed
- Department of Neurology and FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Raimondo D'Ambrosio
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Cliff Eastman
- Department of Neurological Surgery, University of Washington, Seattle, WA, USA
| | - Benjamin Hui
- Department of Neurology and FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rui Lin
- Department of Neurology and FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Sheryl Anne D Vermudez
- Department of Neurology and FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Amanda Liebhardt
- Department of Neurology and FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Yongho Choe
- Department of Neurology and FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Pavel Klein
- PrevEp, Inc., Bethesda, MD, USA; Mid-Atlantic Epilepsy and Sleep Center, Bethesda, MD, USA
| | | | - Wolfgang Löscher
- PrevEp, Inc., Bethesda, MD, USA; Translational Neuropharmacology Lab, NIFE, Department of Experimental Otology of the ENT Clinics, Hannover Medical School, Hannover, Germany.
| | - Alexander Rotenberg
- Department of Neurology and FM Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; PrevEp, Inc., Bethesda, MD, USA.
| |
Collapse
|
2
|
García-Zamora M, García-Lluch G, Moreno L, Pardo J, Cháfer-Pericás C. Influence of statin potency and liposolubility on Alzheimer's disease patients: A population-based study. Pharmacol Res 2024; 209:107446. [PMID: 39362508 DOI: 10.1016/j.phrs.2024.107446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 09/30/2024] [Accepted: 09/30/2024] [Indexed: 10/05/2024]
Abstract
Although Alzheimer's disease (AD) cause is still unknown, there are several known risk factors, such as dyslipidemia. Statins are the most prescribed lipid-modifying therapies. Recent research has suggested a relationship between statins and AD, nevertheless, their ability to prevent AD is still unclear. Therefore, this cross-sectional study aimed to examine the relationship between statin use and anti-AD drug prescription. For that purpose, a database containing information on medications prescribed to patients aged 50 years or older (n = 233183) between 2018 and 2020 was used. Defined daily doses (DDDs) were calculated according to the ATC/DDD index 2023. Statistical analyses, with logistic regression and cumulative incidence, were carried out to assess statins and anti-AD drug consumption. As a result, a total of 47852 patients aged more than 70 years who were prescribed at least one antihypertensive, antidiabetic or lipid-modifying agent were included in the study. Of these, 45345 patients were classified within the cardiovascular risk group and 2483 were classified as patients with only hyperlipidemia. Patients using low-potency or hydrophilic statins had lower odds of anti-AD usage when compared to high-potency or lipophilic statins, respectively. Similarly, rosuvastatin and pitavastatin had lower odds of anti-AD medication intake when compared to atorvastatin. Finally, pitavastatin DDDs were prone to lower the odds of anti-AD medication usage when compared to rosuvastatin. In conclusion, a potential association between statins and the intake of AD medication has been observed. Specifically, low-potency (pitavastatin) and hydrophilic (rosuvastatin) statins were associated with less use of anti-AD medication.
Collapse
Affiliation(s)
- Mar García-Zamora
- Research Group in Alzheimer Disease, Instituto de Investigación Sanitaria La Fe, Valencia, Spain; Cathedra DeCo MICOF-CEU UCH, University Cardenal Herrera-CEU, Valencia 46115, Spain
| | - Gemma García-Lluch
- Research Group in Alzheimer Disease, Instituto de Investigación Sanitaria La Fe, Valencia, Spain; Cathedra DeCo MICOF-CEU UCH, University Cardenal Herrera-CEU, Valencia 46115, Spain
| | - Lucrecia Moreno
- Cathedra DeCo MICOF-CEU UCH, University Cardenal Herrera-CEU, Valencia 46115, Spain; Department of Pharmacy, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia 46115, Spain
| | - Juan Pardo
- Embedded Systems and Artificial Intelligence Group, Universidad Cardenal Herrera-CEU, CEU Universities, Valencia 46115, Spain.
| | - Consuelo Cháfer-Pericás
- Research Group in Alzheimer Disease, Instituto de Investigación Sanitaria La Fe, Valencia, Spain.
| |
Collapse
|
3
|
Wojcicki K, Budzinska A, Jarmuszkiewicz W. Effects of Atorvastatin and Simvastatin on the Bioenergetic Function of Isolated Rat Brain Mitochondria. Int J Mol Sci 2024; 25:8494. [PMID: 39126062 PMCID: PMC11313418 DOI: 10.3390/ijms25158494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 07/27/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
Little is known about the effects of statins, which are cholesterol-lowering drugs, on the bioenergetic functions of mitochondria in the brain. This study aimed to elucidate the direct effects of atorvastatin and simvastatin on the bioenergetics of isolated rat brain mitochondria by measuring the statin-induced changes in respiratory chain activity, ATP synthesis efficiency, and the production of reactive oxygen species (ROS). Our results in isolated brain mitochondria are the first to demonstrate that atorvastatin and simvastatin dose-dependently significantly inhibit the activity of the mitochondrial respiratory chain, resulting in a decreased respiratory rate, a decreased membrane potential, and increased ROS formation. Moreover, the tested statins reduced mitochondrial coupling parameters, the ADP/O ratio, the respiratory control ratio, and thus, the oxidative phosphorylation efficiency in brain mitochondria. Among the oxidative phosphorylation complexes, statin-induced mitochondrial impairment concerned complex I, complex III, and ATP synthase activity. The calcium-containing atorvastatin had a significantly more substantial effect on isolated brain mitochondria than simvastatin. The higher inhibitory effect of atorvastatin was dependent on calcium ions, which may lead to the disruption of calcium homeostasis in mitochondria. These findings suggest that while statins are effective in their primary role as cholesterol-lowering agents, their use may impair mitochondrial function, which may have consequences for brain health, particularly when mitochondrial energy efficiency is critical.
Collapse
Affiliation(s)
| | | | - Wieslawa Jarmuszkiewicz
- Mitochondrial Biochemistry Research Group, Faculty of Biology, Adam Mickiewicz University in Poznan, 61-614 Poznan, Poland; (K.W.); (A.B.)
| |
Collapse
|
4
|
He L, Ioannidis A, Hoffman CJ, Arambula E, Joshi P, Whitelegge J, Liau LM, Kornblum HI, Pajonk F. Activation of the Mevalonate Pathway in Response to Anti-cancer Treatments Drives Glioblastoma Recurrences Through Activation of Rac-1. CANCER RESEARCH COMMUNICATIONS 2024; 4:1566-1580. [PMID: 38837899 PMCID: PMC11197925 DOI: 10.1158/2767-9764.crc-24-0049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/26/2024] [Accepted: 05/31/2024] [Indexed: 06/07/2024]
Abstract
Glioblastoma (GBM) is the deadliest adult brain cancer. Under the current standard of care, almost all patients succumb to the disease and novel treatments are urgently needed. Recognizing that GBMs are addicted to cholesterol, past clinical trials have repurposed statins against GBM but failed. The purpose of this study was to test whether treatments that upregulate the cholesterol biosynthesis pathway in GBM would generate a metabolic vulnerability that can be exploited using statins and to determine the underlying mechanisms.Effects of radiotherapy and temozolomide or dopamine receptor antagonists on the mevalonate pathway in GBM were assessed in vitro and in vivo. The impact of statins on self-renewal of glioma stem cells and median survival was studied. Branches of the mevalonate pathway were probed to identify relevant effector proteins.Cells surviving combination treatments that converge in activating the immediate early response, universally upregulated the mevalonate pathway and increased stemness of GBM cells through activation of the Rho-GTPase Rac-1. Activation of the mevalonate pathway and Rac-1 was inhibited by statins, which led to improved survival in mouse models of glioblastoma when combined with radiation and drugs that target the glioma stem cell pool and plasticity of glioma cells.We conclude that a combination of dopamine receptor antagonists and statins could potentially improve radiotherapy outcome and warrants further investigation. SIGNIFICANCE Combination therapies that activate the mevalonate pathway in GBM cells after sublethal treatment enhance self-renewal and migratory capacity through Rac-1 activation, which creates a metabolic vulnerability that can be further potentially exploited using statins.
Collapse
Affiliation(s)
- Ling He
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, Los Angeles, California
- Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, California
| | - Angeliki Ioannidis
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Carter J. Hoffman
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Evelyn Arambula
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Purva Joshi
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Julian Whitelegge
- Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, California
- Department of Psychiatry and Human Behavior, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Linda M. Liau
- Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, California
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Harley I. Kornblum
- Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, California
- Department of Psychiatry and Human Behavior, David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Frank Pajonk
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA, Los Angeles, California
- Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, California
- Department of Neurosurgery, David Geffen School of Medicine at UCLA, Los Angeles, California
| |
Collapse
|
5
|
Al-Shalchi RF, Mohammad FK. Oxidative Stress-Induced Adverse Effects of Three Statins Following Single or Repetitive Treatments in Mice. Cureus 2024; 16:e51433. [PMID: 38298275 PMCID: PMC10828976 DOI: 10.7759/cureus.51433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/01/2024] [Indexed: 02/02/2024] Open
Abstract
Background and objective The hypolipidemic statins have been associated with various side effects, and in some cases, adverse reactions in humans and experimental animals, such as myotoxicity, neurobehavioral toxicity, as well as liver and kidney injuries. The purpose of the present study was to examine the possibility of the induction of oxidative stress in the brain and plasma of mice dosed with single or repetitive doses of three statins (atorvastatin, simvastatin, and rosuvastatin). Methods Male Swiss-origin mice were dosed orally with single doses of each of the three statins at 500 or 1000 mg/kg of body weight. Other groups of mice were dosed orally with repeated daily doses of each of the statins at 200 mg/kg of body weight/day for 14 or 28 consecutive days. These doses of statins were chosen to not produce overt toxicity in mice within the time frame allocated for each experiment. Brain and plasma glutathione (GSH) and malondialdehyde (MDA) levels, as well as liver enzymes activities alanine transaminase (ALT) and aspartate transaminase (AST), were determined using commercial kits. Results Single-dose treatments of the mice with the statins at either 500 or 1000 mg/kg significantly and dose-dependently (p < 0.05) reduced the GSH level in the plasma and the whole brain when compared with respective control values. Atorvastatin was the least effective statin, as only the high dose achieved a significant reduction in brain GSH level in comparison with the respective control value. Repetitive administration of the three statins at 200 mg/kg of body weight/day for 14 or 28 consecutive days significantly and time-dependently reduced plasma and brain GSH levels in comparison with respective control values. The oxidative stress biomarker MDA level significantly increased in the plasma and brain of mice following single or repetitive treatments with the three statins, and the most effective one was rosuvastatin. In association with these changes, activities of the liver enzymes ALT and AST were also increased in the plasma with single and repetitive statin treatments, and the most effective one was rosuvastatin. Conclusion The data suggest an association of high doses of three statins (atorvastatin, simvastatin, and rosuvastatin) with the induction of oxidative stress manifested as GSH reduction and MDA elevation as adverse effects in the brain and plasma of mice, which suffered from the additional burden of liver injury. These effects could be the basis of an in-depth exploration of statin adverse effects in experimental animals and to find an animal model, probably the mice, for the induction of adverse effects of statins that target the brain, as well as to shed light on potential statin intolerance outcomes following single-dose treatments in this species.
Collapse
Affiliation(s)
- Rawnaq F Al-Shalchi
- Department of Physiology, Biochemistry and Pharmacology, College of Veterinary Medicine, University of Mosul, Mosul, IRQ
| | - Fouad K Mohammad
- Department of Physiology, Biochemistry and Pharmacology, College of Veterinary Medicine, University of Mosul, Mosul, IRQ
- College of Nursing, The American University of Kurdistan, Duhok, IRQ
| |
Collapse
|
6
|
Snow NJ, Landine J, Chaves AR, Ploughman M. Age and asymmetry of corticospinal excitability, but not cardiorespiratory fitness, predict cognitive impairments in multiple sclerosis. IBRO Neurosci Rep 2023; 15:131-142. [PMID: 37577407 PMCID: PMC10412844 DOI: 10.1016/j.ibneur.2023.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 07/11/2023] [Accepted: 07/15/2023] [Indexed: 08/15/2023] Open
Abstract
Background Cognitive impairment is a disabling and underestimated consequence of multiple sclerosis (MS), with multiple determinants that are poorly understood. Objectives We explored predictors of MS-related processing speed impairment (PSI) and age-related mild cognitive impairment (MCI) and hypothesized that cardiorespiratory fitness and corticospinal excitability would predict these impairments. Methods We screened 73 adults with MS (53 females; median [range]: Age 48 [21-70] years, EDSS 2.0 [0.0-6.5]) for PSI and MCI using the Symbol Digit Modalities Test and Montréal Cognitive Assessment, respectively. We identified six persons with PSI (No PSI, n = 67) and 13 with MCI (No MCI, n = 60). We obtained clinical data from medical records and self-reports; used transcranial magnetic stimulation to test corticospinal excitability; and assessed cardiorespiratory fitness using a graded maximal exercise test. We used receiver operator characteristic (ROC) curves to discern predictors of PSI and MCI. Results Interhemispheric asymmetry of corticospinal excitability was specific for PSI, while age was both sensitive and specific for MCI. MS-related PSI was also associated with statin prescriptions, while age-related MCI was related to progressive MS and GABA agonist prescriptions. Cardiorespiratory fitness was associated with neither PSI nor MCI. Discussion Corticospinal excitability is a potential marker of neurodegeneration in MS-related PSI, independent of age-related effects on global cognitive function. Age is a key predictor of mild global cognitive impairment. Cardiorespiratory fitness did not predict cognitive impairments in this clinic-based sample of persons with MS.
Collapse
Affiliation(s)
- Nicholas J. Snow
- Recovery and Performance Laboratory, Faculty of Medicine, Memorial University of Newfoundland and Labrador, St. John's, Newfoundland and Labrador, Canada
| | - Josef Landine
- Recovery and Performance Laboratory, Faculty of Medicine, Memorial University of Newfoundland and Labrador, St. John's, Newfoundland and Labrador, Canada
| | - Arthur R. Chaves
- Recovery and Performance Laboratory, Faculty of Medicine, Memorial University of Newfoundland and Labrador, St. John's, Newfoundland and Labrador, Canada
| | - Michelle Ploughman
- Recovery and Performance Laboratory, Faculty of Medicine, Memorial University of Newfoundland and Labrador, St. John's, Newfoundland and Labrador, Canada
| |
Collapse
|
7
|
Sarkar P, Chattopadhyay A. Interplay of Cholesterol and Actin in Neurotransmitter GPCR Signaling: Insights from Chronic Cholesterol Depletion Using Statin. ACS Chem Neurosci 2023; 14:3855-3868. [PMID: 37804226 DOI: 10.1021/acschemneuro.3c00472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/09/2023] Open
Abstract
Serotonin1A receptors are important neurotransmitter receptors in the G protein-coupled receptor (GPCR) family and modulate a variety of neurological, behavioral, and cognitive functions. We recently showed that chronic cholesterol depletion by statins, potent inhibitors of HMG-CoA reductase (the rate-limiting enzyme in cholesterol biosynthesis), leads to polymerization of the actin cytoskeleton that alters lateral diffusion of serotonin1A receptors. However, cellular signaling by the serotonin1A receptor under chronic cholesterol depletion remains unexplored. In this work, we explored signaling by the serotonin1A receptor under statin-treated condition. We show that cAMP signaling by the receptor is reduced upon lovastatin treatment due to reduction in cholesterol as well as polymerization of the actin cytoskeleton. To the best of our knowledge, these results constitute the first report describing the effect of chronic cholesterol depletion on the signaling of a G protein-coupled neuronal receptor. An important message arising from these results is that it is prudent to include the contribution of actin polymerization while analyzing changes in membrane protein function due to chronic cholesterol depletion by statins. Notably, our results show that whereas actin polymerization acts as a negative regulator of cAMP signaling, cholesterol could act as a positive modulator. These results assume significance in view of reports highlighting symptoms of anxiety and depression in humans upon statin administration and the role of serotonin1A receptors in anxiety and depression. Overall, these results reveal a novel role of actin polymerization induced by chronic cholesterol depletion in modulating GPCR signaling, which could act as a potential therapeutic target.
Collapse
Affiliation(s)
- Parijat Sarkar
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
| | - Amitabha Chattopadhyay
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad 500 007, India
- Academy of Scientific and Innovative Research, Ghaziabad 201 002, India
| |
Collapse
|
8
|
He L, Ioannidis A, Arambula E, Hoffman CJ, Joshi P, Kathiravan A, Whitelegge J, Liau LM, Kornblum HI, Pajonk F. Activation of the mevalonate pathway in response to anti-cancer treatments drives glioblastoma recurrences through activation of Rac-1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.23.550205. [PMID: 37546917 PMCID: PMC10402033 DOI: 10.1101/2023.07.23.550205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Glioblastoma is the deadliest adult brain cancer. Under the current standard of care almost all patients succumb to the disease and novel treatments are urgently needed. Dopamine receptor antagonists have been shown to target cancer cell plasticity in GBM and repurposing these FDA-approved drugs in combination with radiation improves the efficacy of radiotherapy in glioma models. In cells surviving this combination treatment the mevalonate pathway is upregulated at the transcriptional and functional level. Here we report that glioblastoma treatments that converge in the immediate early response to radiation through activation of the MAPK cascade universally upregulate the mevalonate pathway and increase stemness of GBM cells through activation of the Rho-GTPase Rac-1. Activation of the mevalonate pathway and Rac-1 is inhibited by statins, which leads to improved survival in mouse models of glioblastoma when combined with radiation and drugs that target the glioma stem cell pool and plasticity of glioma cells.
Collapse
Affiliation(s)
- Ling He
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA
| | - Angeliki Ioannidis
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA
| | - Evelyn Arambula
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA
| | - Carter J. Hoffman
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA
| | - Purva Joshi
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA
| | | | - Julian Whitelegge
- Jonsson Comprehensive Cancer Center at UCLA
- Department of Psychiatry and Human Behavior, David Geffen School of Medicine at UCLA
| | - Linda M. Liau
- Jonsson Comprehensive Cancer Center at UCLA
- Department of Neurosurgery, David Geffen School of Medicine at UCLA
| | - Harley I. Kornblum
- Jonsson Comprehensive Cancer Center at UCLA
- Department of Psychiatry and Human Behavior, David Geffen School of Medicine at UCLA
| | - Frank Pajonk
- Department of Radiation Oncology, David Geffen School of Medicine at UCLA
- Jonsson Comprehensive Cancer Center at UCLA
| |
Collapse
|
9
|
Susanto M, Pangihutan Siahaan AM, Wirjomartani BA, Setiawan H, Aryanti C, Michael. The neuroprotective effect of statin in traumatic brain injury: A systematic review. World Neurosurg X 2023; 19:100211. [PMID: 37251243 PMCID: PMC10220252 DOI: 10.1016/j.wnsx.2023.100211] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 04/23/2023] [Accepted: 05/02/2023] [Indexed: 07/22/2023] Open
Abstract
Despite recent encouraging pharmaceutical and technical breakthroughs in neurosurgical critical care, traumatic brain injury (TBI)-related mortality and morbidity remain substantial clinical issues. Medication of statins was revealed to enhance outcomes following TBI in animal research. In addition to their main role of decreasing serum cholesterol, statins decrease inflammation and enhance cerebral blood flow. However, research on the efficacy of statins in TBI is still limited. This systematic review was conducted to determine the efficacy of statins in enhancing the clinical outcomes of TBI individuals, and specifically investigate the optimal dose and form of statins. The databases of PubMed, DOAJ, EBSCO, and Cochrane were extensively researched. The date of publication within the last fifteen years was the inclusion criterion. Meta-analyses, clinical trials, and randomized controlled trials were prioritized forms of research publications. Ambiguous remarks, irrelevant correlations to the main issue, or a focus on disorders other than TBI were the exclusion criteria. Thirteen research were included in this study. Simvastatin, atorvastatin, and rosuvastatin were the main form of statins discussed in this study. Enhancement of the Glasgow Coma Scale, survival rates, hospital length of stay, and cognitive outcomes were revealed in this study. This study suggests either simvastatin 40 mg, atorvastatin 20 mg, or rosuvastatin 20 mg for 10 days as the optimal therapeutic forms and doses to be applied in the management of TBI. Pre-TBI statin use was linked to lower risk of mortality in TBI individuals compared to nonusers, whereas statin discontinuation was linked to an increase in mortality.
Collapse
Affiliation(s)
- Martin Susanto
- Faculty of Medicine, University of Sumatera Utara, Medan, North Sumatra, Indonesia
| | | | | | - Hendy Setiawan
- Department of Neurosurgery, University of Sumatera Utara, Medan, North Sumatra, Indonesia
| | - Citra Aryanti
- Department of Surgery, University of Udayana, Denpasar, Bali, Indonesia
| | - Michael
- Department of Neurosurgery, University of Padjadjaran, Bandung, West Java, Indonesia
| |
Collapse
|
10
|
Khan S, Hasan N, Khan S, Akhtar M, Akhtar M, Najmi AK. Exploring effects of Simvastatin on coagulation mediators to alleviate the advancement of high cholesterol diet triggered neurodegeneration. J Biochem Mol Toxicol 2023:e23342. [PMID: 36992618 DOI: 10.1002/jbt.23342] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 01/17/2023] [Accepted: 03/20/2023] [Indexed: 03/31/2023]
Abstract
The objectives of our study were to investigate the possible effect of Simvastatin in ameliorating high cholesterol diet (HCD)-induced neurodegeneration and to also investigate its possible action on coagulation mediators. In silico and in vitro studies were performed to evaluate the impact of Simvastatin on prime coagulation mediators. HCD was used to induce neuropathology in wistar rats and histopathological and immunohistochemical studies were performed to evaluate the efficacy of Simvastatin in preventing the advancement of neurodegeneration in obese rats. Biochemical analyses were used to estimate changes in lipid profile, oxidative stress, inflammatory and coagulation markers. Simvastatin showed good theoretical affinity to coagulation proteins, significantly reversed changes in inflammatory and coagulation biomarkers which were induced by HCD. Enhanced fibrinolytic activity of Simvastatin was revealed through in vitro analysis. Immunohistoanalysis showed raised level of Nrf2. Histopathological studies also supported neuroprotective potential of Simvastatin in HCD fed rats. Simvastatin demonstrated reduced hypercoagulation, enhanced fibrinolysis and reversed neurodegeneration in HCD exposed rats suggesting its potential role in preventing the progression of neurodegeneration in obesity.
Collapse
Affiliation(s)
- Sana Khan
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, India
| | - Noorul Hasan
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, India
| | - Saba Khan
- Department of Pharmaceutics, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, India
| | - Mymoona Akhtar
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, India
| | - Mohammad Akhtar
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, India
| | - Abul Kalam Najmi
- Department of Pharmacology, School of Pharmaceutical Education & Research, Jamia Hamdard, New Delhi, India
| |
Collapse
|
11
|
Vukšić A, Rašić D, Žunec S, Božina T, Konjevoda P, Lovrić J, Bilušić M, Bradamante V. The effects of simvastatin and fenofibrate on malondialdehyde and reduced glutathione concentrations in the plasma, liver, and brain of normolipidaemic and hyperlipidaemic rats. Arh Hig Rada Toksikol 2023; 74:34-41. [PMID: 37014683 DOI: 10.2478/aiht-2023-74-3697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/01/2023] [Indexed: 04/05/2023] Open
Abstract
The objective of study was to investigate the effects of different doses of simvastatin and fenofibrate on malondialdehyde (MDA) and reduced glutathione (GSH) in the plasma, liver, and brain tissue of male normolipidaemic and hyperlipidaemic rats. Normolipidaemic (Wistar) rats were receiving 10 or 50 mg/kg a day of simvastatin or 30 or 50 mg/kg a day of fenofibrate. Hyperlipidaemic (Zucker) rats were receiving 50 mg/kg/day of simvastatin or 30 mg/kg/day of fenofibrate. Control normolipidaemic and hyperlipidaemic rats were receiving saline. Simvastatin, fenofibrate, and saline were administered by gavage for three weeks. In normolipidaemic rats simvastatin and fenofibrate showed similar and dose-independent effects on plasma and brain MDA and GSH concentrations. Generally, plasma and brain MDA decreased, while brain GSH concentration increased. In hyperlipidaemic rats simvastatin did not affect plasma and brain MDA and GSH concentrations but significantly decreased liver GSH. Fenofibrate decreased plasma and liver MDA but increased brain MDA. In both rat strains fenofibrate significantly decreased liver GSH concentrations, most likely because fenofibrate metabolites bind to GSH. Our findings suggest that simvastatin acts as an antioxidant only in normolipidaemic rats, whereas fenofibrate acts as an antioxidant in both rat strains.
Collapse
Affiliation(s)
- Antonija Vukšić
- 1Clinical Hospital Centre Zagreb, Department of Neurology, Zagreb, Croatia
| | - Dubravka Rašić
- 2Institute for Medical Research and Occupational Health, Unit of Toxicology, Zagreb, Croatia
| | - Suzana Žunec
- 2Institute for Medical Research and Occupational Health, Unit of Toxicology, Zagreb, Croatia
| | - Tamara Božina
- 3University of Zagreb School of Medicine, Department of Medical Chemistry, Biochemistry, and Clinical Chemistry, Zagreb, Croatia
| | | | - Jasna Lovrić
- 3University of Zagreb School of Medicine, Department of Medical Chemistry, Biochemistry, and Clinical Chemistry, Zagreb, Croatia
| | - Marinko Bilušić
- 5Polyclinic Bonifarm, Department of Clinical Pharmacology and Toxicology, Zagreb, Croatia
| | - Vlasta Bradamante
- 6University of Zagreb School of Medicine, Department of Pharmacology, Zagreb, Croatia
| |
Collapse
|
12
|
An JY, Park JY, Cho J, Kim HE, Park J, Oh J. The Relationship between Delirium and Statin Use According to Disease Severity in Patients in the Intensive Care Unit. CLINICAL PSYCHOPHARMACOLOGY AND NEUROSCIENCE : THE OFFICIAL SCIENTIFIC JOURNAL OF THE KOREAN COLLEGE OF NEUROPSYCHOPHARMACOLOGY 2023; 21:179-187. [PMID: 36700324 PMCID: PMC9889904 DOI: 10.9758/cpn.2023.21.1.179] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/19/2022] [Accepted: 05/02/2022] [Indexed: 01/27/2023]
Abstract
Objective The aim of this study was to investigate the association between the use of statins and the occurrence of delirium in a large cohort of patients in the intensive care unit (ICU), considering disease severity and statin properties. Methods We obtained clinical and demographical information from 3,604 patients admitted to the ICU from January 2013 to April 2020. This included information on daily statin use and delirium state, as assessed by the Confusion Assessment Method for ICU. We used inverse probability of treatment weighting and categorized the patients into four groups based on the Acute Physiology and Chronic Health Evaluation II score (group 1: 0-10 - mild; group 2: 11-20 - mild to moderate; group 3: 21-30 - moderate to severe; group 4: > 30 - severe). We analyzed the association between the use of statin and the occurrence of delirium in each group, while taking into account the properties of statins. Results Comparisons between statin and non-statin patient groups revealed that only in group 2, patients who were administered statin showed significantly higher occurrence of delirium (p = 0.004, odds ratio [OR] = 1.58) compared to the patients who did not receive statin. Regardless of whether statins were lipophilic (p = 0.036, OR = 1.47) or hydrophilic (p = 0.032, OR = 1.84), the occurrence of delirium was higher only in patients from group 2. Conclusion The use of statins may be associated with the increases in the risk of delirium occurrence in patients with mild to moderate disease severity, irrespective of statin properties.
Collapse
Affiliation(s)
- Jun Yong An
- Department of Psychiatry, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea,Institute of Behavioral Sciences in Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jin Young Park
- Institute of Behavioral Sciences in Medicine, Yonsei University College of Medicine, Seoul, Korea,Department of Psychiatry, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin, Korea
| | - Jaehwa Cho
- Department of Pulmonary and Critical Care Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea
| | - Hesun Erin Kim
- Institute of Behavioral Sciences in Medicine, Yonsei University College of Medicine, Seoul, Korea
| | - Jaesub Park
- Institute of Behavioral Sciences in Medicine, Yonsei University College of Medicine, Seoul, Korea,Department of Psychiatry, National Health Insurance Service Ilsan Hospital, Goyang, Korea
| | - Jooyoung Oh
- Department of Psychiatry, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Korea,Institute of Behavioral Sciences in Medicine, Yonsei University College of Medicine, Seoul, Korea,Address for correspondence: Jooyoung Oh Department of Psychiatry, Gangnam Severance Hospital, Yonsei University College of Medicine, 211 Eonju-ro, Gangnam-gu, Seoul 06273, Korea, E-mail: , ORCID: https://orcid.org/0000-0001-6721-399X
| |
Collapse
|
13
|
Chattopadhyay A, Sharma A. Smith-Lemli-Opitz syndrome: A pathophysiological manifestation of the Bloch hypothesis. Front Mol Biosci 2023; 10:1120373. [PMID: 36714259 PMCID: PMC9878332 DOI: 10.3389/fmolb.2023.1120373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 01/02/2023] [Indexed: 01/15/2023] Open
Abstract
The biosynthesis of cholesterol, an essential component of higher eukaryotic membranes, was worked out by Konrad Bloch (and Feodor Lynen) in the 1960s and they received the Nobel Prize around that time in recognition of their pioneering contributions. An elegant consequence of this was a hypothesis proposed by Konrad Bloch (the Bloch hypothesis) which suggests that each subsequent intermediate in the cholesterol biosynthesis pathway is superior in supporting membrane function in higher eukaryotes relative to its precursor. In this review, we discuss an autosomal recessive metabolic disorder, known as Smith-Lemli-Opitz syndrome (SLOS), associated with a defect in the Kandutsch-Russell pathway of cholesterol biosynthesis that results in accumulation of the immediate precursor of cholesterol in its biosynthetic pathway (7-dehydrocholesterol) and an altered cholesterol to total sterol ratio. Patients suffering from SLOS have several developmental, behavioral and cognitive abnormalities for which no drug is available yet. We characterize SLOS as a manifestation of the Bloch hypothesis and review its molecular etiology and current treatment. We further discuss defective Hedgehog signaling in SLOS and focus on the role of the serotonin1A receptor, a representative neurotransmitter receptor belonging to the GPCR family, in SLOS. Notably, ligand binding activity and cellular signaling of serotonin1A receptors are impaired in SLOS-like condition. Importantly, cellular localization and intracellular trafficking of the serotonin1A receptor (which constitute an important determinant of a GPCR cellular function) are compromised in SLOS. We highlight some of the recent developments and emerging concepts in SLOS pathobiology and suggest that novel therapies based on trafficking defects of target receptors could provide new insight into treatment of SLOS.
Collapse
Affiliation(s)
- Amitabha Chattopadhyay
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India,Academy of Scientific and Innovative Research, Ghaziabad, India,*Correspondence: Amitabha Chattopadhyay,
| | - Ashwani Sharma
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India,Academy of Scientific and Innovative Research, Ghaziabad, India
| |
Collapse
|
14
|
Abstract
Parkinson's disease (PD) is a neurodegenerative disease manifesting with motor and non-motor symptoms. Current treatment mainly relies on medication as a symptomatic therapy modulating neurotransmitters. Dopamine replacement therapy has been established, and levodopa is the gold standard for treatment of PD. However, the emergence of motor complications, such as a wearing-off phenomenon, is a clinical problem. Both primary symptoms and motor complications have been targets for the development of treatments for PD. Recent progression in the management of motor complications is supported by newly developed agents and advances in device and formulation technology to deliver drugs continuously. Elucidation of the pathophysiology of PD and the development of disease-modifying therapy that affects the underlying fundamental pathophysiology of the disease are also progressing. In this review, we introduce current knowledge on developments concerning medications for patients with PD.
Collapse
Affiliation(s)
- Hidetomo Murakami
- Department of Neurology, the Jikei University School of Medicine, Japan
| | | | - Tadashi Umehara
- Department of Neurology, the Jikei University School of Medicine, Japan
| | - Shusaku Omoto
- Department of Neurology, the Jikei University Katsushika Medical Center, Japan
| | - Yasuyuki Iguchi
- Department of Neurology, the Jikei University School of Medicine, Japan
| |
Collapse
|
15
|
Españo E, Kim JK. Effects of Statin Combinations on Zika Virus Infection in Vero Cells. Pharmaceutics 2022; 15:pharmaceutics15010050. [PMID: 36678679 PMCID: PMC9864436 DOI: 10.3390/pharmaceutics15010050] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 12/09/2022] [Accepted: 12/13/2022] [Indexed: 12/28/2022] Open
Abstract
The Zika virus (ZIKV) remains a global health concern. Thus far, no antiviral or vaccine has been approved to prevent or treat ZIKV infection. In a previous study, we found that lipophilic statins can inhibit ZIKV production in Vero cells. These statins appear to have different potencies against ZIKV infection. Here, we determined whether combinations of statins would have synergistic effects to maximize the efficacy of the statins and to reduce potential side effects. Specifically, we used a modified fixed-ratio assay for the combinations of atorvastatin (ATO) or fluvastatin (FLU) with mevastatin (MEV) or simvastatin (SIM). All combinations with MEV tended towards synergy, especially with higher fractions of MEV in the combinations. The ATO + SIM combination tended towards additivity. The FLU + SIM combination also tended towards additivity except for one combination which had the highest fraction of FLU over SIM among the tested combinations. Overall, certain combinations of ATO or FLU with SIM or MEV may be synergistic. More exhaustive combinatorial assays in vitro and in vivo could help define whether combining lipophilic statins would be beneficial and safe for treating ZIKV infections.
Collapse
|
16
|
Microglia and Cholesterol Handling: Implications for Alzheimer's Disease. Biomedicines 2022; 10:biomedicines10123105. [PMID: 36551857 PMCID: PMC9775660 DOI: 10.3390/biomedicines10123105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 11/24/2022] [Accepted: 11/29/2022] [Indexed: 12/04/2022] Open
Abstract
Cholesterol is essential for brain function and structure, however altered cholesterol metabolism and transport are hallmarks of multiple neurodegenerative conditions, including Alzheimer's disease (AD). The well-established link between apolipoprotein E (APOE) genotype and increased AD risk highlights the importance of cholesterol and lipid transport in AD etiology. Whereas more is known about the regulation and dysregulation of cholesterol metabolism and transport in neurons and astrocytes, less is known about how microglia, the immune cells of the brain, handle cholesterol, and the subsequent implications for the ability of microglia to perform their essential functions. Evidence is emerging that a high-cholesterol environment, particularly in the context of defects in the ability to transport cholesterol (e.g., expression of the high-risk APOE4 isoform), can lead to chronic activation, increased inflammatory signaling, and reduced phagocytic capacity, which have been associated with AD pathology. In this narrative review we describe how cholesterol regulates microglia phenotype and function, and discuss what is known about the effects of statins on microglia, as well as highlighting areas of future research to advance knowledge that can lead to the development of novel therapies for the prevention and treatment of AD.
Collapse
|
17
|
Mirzaie J, Nasiry D, Ayna Ö, Raoofi A, Delbari A, Rustamzadeh A, Nezhadi A, Jamalpoor Z. Neuroprotective effects of lovastatin against traumatic spinal cord injury in rats. J Chem Neuroanat 2022; 125:102148. [PMID: 36031087 DOI: 10.1016/j.jchemneu.2022.102148] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 10/31/2022]
Abstract
BACKGROUND Lovastatin, as a drug of statins subgroup, has been conceptualized to have anti-inflammatory, antioxidant, and anti-apoptotic properties. Accordingly, the present study aimed to investigate the neuroprotective ramification of lovastatin on spinal cord injury (SCI). MATERIAL AND METHODS Seventy-five female adult Wistar rats were divided into five groups (n = 15). In addition to non-treated (Control group) and laminectomy alone (Sham group), SCI animals were randomly assigned to non-treated spinal cord injury (SCI group), treated with 2 mg/kg of lovastatin (Lova 2 group), and treated with 5 mg/kg of lovastatin (Lova 5 group). At the end of the study, to evaluate the treatments, MDA, CAT, SOD, and GSH factors were evaluated biochemically, apoptosis and gliosis were assessed by immunohistochemical while measuring caspase-3 and GFAP antibodies, and inflammation was estimated by examining the expression of IL-10, TNF-α, and IL-1β genes. The stereological method was used to appraise the total volume of the spinal cord at the site of injury, the volume of the central cavity created, and the density of neurons and glial cells in the traumatic area. In addition, Basso-Beattie-Bresnehan (BBB) and narrow beam test (NBT) were utilized to rate neurological functions. RESULTS Our results exposed the fact that biochemical factors (except MDA), stereological parameters, and neurological functions were significantly ameliorated in both lovastatin-treated groups, especially in Lova 5 ones, compared to the SCI group. The expression of the IL-10 gene was significantly upregulated in both lovastatin-treated groups compared to the SCI group and was considerably heighten in Lova 5 group. Expression of TNF-α and IL-1β, as well as the rate of apoptosis and GFAP positive cells significantly decreased in both lovastatin treated groups compared to the SCI group, and it was more pronounced in the Lova 5 ones. CONCLUSION Overall, using lovastatin, especially at a dose of 5 mg/kg, has a dramatic neuroprotective impact on SCI treatment.
Collapse
Affiliation(s)
- Jafar Mirzaie
- Neuroscience Research Center, Aja University of Medical Sciences, Tehran, Iran
| | - Davood Nasiry
- Amol Faculty of Paramedicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Ömer Ayna
- Kiev Medical University, Dermatology Departments, Kiev, Ukraine
| | - Amir Raoofi
- Cellular and Molecular Research Center, Department of Anatomical Sciences, School of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Ali Delbari
- Cellular and Molecular Research Center, Department of Anatomical Sciences, School of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Auob Rustamzadeh
- Department of Anatomy, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Akram Nezhadi
- Neuroscience Research Center, Aja University of Medical Sciences, Tehran, Iran.
| | - Zahra Jamalpoor
- Trauma Research Center, Aja University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
18
|
Tajbakhsh A, Gheibihayat SM, Askari H, Savardashtaki A, Pirro M, Johnston TP, Sahebkar A. Statin-regulated phagocytosis and efferocytosis in physiological and pathological conditions. Pharmacol Ther 2022; 238:108282. [DOI: 10.1016/j.pharmthera.2022.108282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/10/2022] [Accepted: 09/13/2022] [Indexed: 10/14/2022]
|
19
|
Wang H, Chen Y, Li P, Chen Y, Yu D, Tan Q, Liu X, Guo Z. Biphasic effects of statins on neuron cell functions under oxygen-glucose deprivation and normal culturing conditions via different mechanisms. Pharmacol Res Perspect 2022; 10:e01001. [PMID: 36029136 PMCID: PMC9419152 DOI: 10.1002/prp2.1001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 07/07/2022] [Accepted: 07/26/2022] [Indexed: 11/11/2022] Open
Abstract
While there is a growing interest in the use of statins, HMG‐CoA reductase inhibitors, to treat neurodegenerative diseases, statins are associated with conflicting effects within the central nervous system (CNS) without clear evidence of the underlying mechanisms. This study systematically investigated effects of four statins (atorvastatin, pitavastatin, cerivastatin, and lovastatin) on neuronal cells under pathological condition using an in vitro model depicting ischemic injury, as well as tested under physiological condition. All four statins at micromolar concentrations display toxic effects on neuron cells under physiological condition. Atorvastatin and cerivastatin but not pitavastatin or lovastatin at nanomolar concentrations display protective effects on neuron cells under ischemic injury condition, via decreased ischemic injury‐induced oxidative stress, oxidative damage, and inflammation. Mechanistically, atorvastatin, pitavastatin, and lovastatin induces neuron cell apoptosis via prenylation‐independent manner. Other mechanisms are involved in the pro‐apoptotic effect of cerivastatin. Prenylation is not involved in the protective effects of statins under ischemic injury condition. Our work provides better understanding on the multiple differential effects of statins on neuron cells under physiological condition and ischemic injury, and elucidate their underlying mechanisms, which may be of relevance to the influence of statins in CNS.
Collapse
Affiliation(s)
- Hui Wang
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, China
| | - Yun Chen
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, China
| | - Ping Li
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, China
| | - Yan Chen
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, China
| | - Danfang Yu
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, China
| | - Qian Tan
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, China
| | - Xiaoli Liu
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, China
| | - Zhenli Guo
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, China
| |
Collapse
|
20
|
Sarkar P, Chattopadhyay A. Statin-induced Increase in Actin Polymerization Modulates GPCR Dynamics and Compartmentalization. Biophys J 2022:S0006-3495(22)00708-1. [DOI: 10.1016/j.bpj.2022.08.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 07/16/2022] [Accepted: 08/25/2022] [Indexed: 11/29/2022] Open
|
21
|
Prud’homme GJ, Kurt M, Wang Q. Pathobiology of the Klotho Antiaging Protein and Therapeutic Considerations. FRONTIERS IN AGING 2022; 3:931331. [PMID: 35903083 PMCID: PMC9314780 DOI: 10.3389/fragi.2022.931331] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 06/06/2022] [Indexed: 12/06/2022]
Abstract
The α-Klotho protein (henceforth denoted Klotho) has antiaging properties, as first observed in mice homozygous for a hypomorphic Klotho gene (kl/kl). These mice have a shortened lifespan, stunted growth, renal disease, hyperphosphatemia, hypercalcemia, vascular calcification, cardiac hypertrophy, hypertension, pulmonary disease, cognitive impairment, multi-organ atrophy and fibrosis. Overexpression of Klotho has opposite effects, extending lifespan. In humans, Klotho levels decline with age, chronic kidney disease, diabetes, Alzheimer’s disease and other conditions. Low Klotho levels correlate with an increase in the death rate from all causes. Klotho acts either as an obligate coreceptor for fibroblast growth factor 23 (FGF23), or as a soluble pleiotropic endocrine hormone (s-Klotho). It is mainly produced in the kidneys, but also in the brain, pancreas and other tissues. On renal tubular-cell membranes, it associates with FGF receptors to bind FGF23. Produced in bones, FGF23 regulates renal excretion of phosphate (phosphaturic effect) and vitamin D metabolism. Lack of Klotho or FGF23 results in hyperphosphatemia and hypervitaminosis D. With age, human renal function often deteriorates, lowering Klotho levels. This appears to promote age-related pathology. Remarkably, Klotho inhibits four pathways that have been linked to aging in various ways: Transforming growth factor β (TGF-β), insulin-like growth factor 1 (IGF-1), Wnt and NF-κB. These can induce cellular senescence, apoptosis, inflammation, immune dysfunction, fibrosis and neoplasia. Furthermore, Klotho increases cell-protective antioxidant enzymes through Nrf2 and FoxO. In accord, preclinical Klotho therapy ameliorated renal, cardiovascular, diabetes-related and neurodegenerative diseases, as well as cancer. s-Klotho protein injection was effective, but requires further investigation. Several drugs enhance circulating Klotho levels, and some cross the blood-brain barrier to potentially act in the brain. In clinical trials, increased Klotho was noted with renin-angiotensin system inhibitors (losartan, valsartan), a statin (fluvastatin), mTOR inhibitors (rapamycin, everolimus), vitamin D and pentoxifylline. In preclinical work, antidiabetic drugs (metformin, GLP-1-based, GABA, PPAR-γ agonists) also enhanced Klotho. Several traditional medicines and/or nutraceuticals increased Klotho in rodents, including astaxanthin, curcumin, ginseng, ligustilide and resveratrol. Notably, exercise and sport activity increased Klotho. This review addresses molecular, physiological and therapeutic aspects of Klotho.
Collapse
Affiliation(s)
- Gérald J. Prud’homme
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, ON, Canada
- *Correspondence: Gérald J. Prud’homme,
| | - Mervé Kurt
- Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science, Unity Health Toronto, Toronto, ON, Canada
| | - Qinghua Wang
- Department of Endocrinology and Metabolism, Huashan Hospital, Shanghai Medical School, Fudan University, Shanghai, China
- Shanghai Yinuo Pharmaceutical Co., Ltd., Shanghai, China
| |
Collapse
|
22
|
Qiao L, Deng F, Hu X, Huang T, An J, Pan D, Yan J, Liang G, He J. Dual sustained-release PTMC/PCL porous microspheres for lipid-soluble drugs. Colloids Surf A Physicochem Eng Asp 2022. [DOI: 10.1016/j.colsurfa.2022.129628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
23
|
Lewis MM, Albertson RM, Du G, Kong L, Foy A, Huang X. Parkinson’s Disease Progression and Statins: Hydrophobicity Matters. JOURNAL OF PARKINSON'S DISEASE 2022; 12:821-830. [PMID: 34958045 PMCID: PMC10141621 DOI: 10.3233/jpd-212819] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Recent randomized clinical trials using hydrophobic statins reported no influence on Parkinson’s disease (PD) clinical progression. Hydrophobicity is a key determinant for blood-brain barrier penetrance. Objective: Investigate a potential effect of statins on PD progression. Methods: Statin use was determined at baseline and subtyped according to hydrophobicity in 125 PD patients participating in the PD Biomarker Program (PDBP, 2012–2015) at our site. Clinical (N = 125) and susceptibility MRI (N = 86) data were obtained at baseline and 18-months. Movement Disorders Society-Unified PD Rating Scales were used to track progression of non-motor (MDS-UPDRS-I) and motor (MDS-UPDRS-II) symptoms, and rater-based scores (MDS-UPDRS-III) of patients in the “on” drug state. R2* values were used to capture pathological progression in the substantia nigra. Associations between statin use, its subtypes, and PD progression were evaluated with linear mixed effect regressions. Results: Compared to statin non-users, overall statin or lipophilic statin use did not significantly influence PD clinical or imaging progression. Hydrophilic statin users, however, demonstrated faster clinical progression of non-motor symptoms [MDS-UPDRS-I (β= 4.8, p = 0.010)] and nigral R2* (β= 3.7, p = 0.043). A similar trend was found for MDS-UPDRS-II (β= 3.9, p = 0.10), but an opposite trend was observed for rater-based MDS-UPDRS-III (β= –7.3, p = 0.10). Compared to lipophilic statin users, hydrophilic statin users also showed significantly faster clinical progression of non-motor symptoms [MDS-UPDRS-I (β= 5.0, p = 0.020)], but R2* did not reach statistical significance (β= 2.5, p = 0.24). Conclusion: This study suggests that hydrophilic, but not lipophilic, statins may be associated with faster PD progression. Future studies may have clinical and scientific implications.
Collapse
Affiliation(s)
- Mechelle M. Lewis
- Department of Neurology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Pharmacology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Richard M. Albertson
- Department of Neurology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Guangwei Du
- Department of Neurology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Lan Kong
- Department of Public Health Sciences, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Andrew Foy
- Department of Public Health Sciences, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Medicine, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Xuemei Huang
- Department of Neurology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Pharmacology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Radiology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Neurosurgery, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
- Department of Kinesiology, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA, USA
| |
Collapse
|
24
|
Sarkar P, Kumar GA, Shrivastava S, Chattopadhyay A. Chronic cholesterol depletion increases F-actin levels and induces cytoskeletal reorganization via a dual mechanism. J Lipid Res 2022; 63:100206. [PMID: 35390404 PMCID: PMC9096963 DOI: 10.1016/j.jlr.2022.100206] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/22/2022] [Accepted: 03/28/2022] [Indexed: 12/24/2022] Open
Abstract
Previous work from us and others has suggested that cholesterol is an important lipid in the context of the organization of the actin cytoskeleton. However, reorganization of the actin cytoskeleton upon modulation of membrane cholesterol is rarely addressed in the literature. In this work, we explored the signaling crosstalk between cholesterol and the actin cytoskeleton by using a high-resolution confocal microscopic approach to quantitatively measure changes in F-actin content upon cholesterol depletion. Our results show that F-actin content significantly increases upon chronic cholesterol depletion, but not during acute cholesterol depletion. In addition, utilizing inhibitors targeting the cholesterol biosynthetic pathway at different steps, we show that reorganization of the actin cytoskeleton could occur due to the synergistic effect of multiple pathways, including prenylated Rho GTPases and availability of membrane phosphatidylinositol 4,5-bisphosphate. These results constitute one of the first comprehensive dissections of the mechanistic basis underlying the interplay between cellular actin levels and cholesterol biosynthesis. We envision these results will be relevant for future understating of the remodeling of the actin cytoskeleton in pathological conditions with altered cholesterol.
Collapse
Affiliation(s)
- Parijat Sarkar
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | - G Aditya Kumar
- CSIR-Centre for Cellular and Molecular Biology, Hyderabad, India
| | | | | |
Collapse
|
25
|
Fadeyibi O, Rybalchenko N, Mabry S, Nguyen DH, Cunningham RL. The Role of Lipid Rafts and Membrane Androgen Receptors in Androgen’s Neurotoxic Effects. J Endocr Soc 2022; 6:bvac030. [PMID: 35308305 PMCID: PMC8926069 DOI: 10.1210/jendso/bvac030] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Indexed: 12/05/2022] Open
Abstract
Sex differences have been observed in multiple oxidative stress–associated neurodegenerative diseases. Androgens, such as testosterone, can exacerbate oxidative stress through a membrane androgen receptor (mAR), AR45, localized to lipid rafts in the plasma membrane. The goal of this study is to determine if interfering with mAR localization to cholesterol-rich lipid rafts decreases androgen induced neurotoxicity under oxidative stress environments. We hypothesize that cholesterol-rich caveolar lipid rafts are necessary for androgens to induce oxidative stress generation in neurons via the mAR localized within the plasma membrane. Nystatin was used to sequester cholesterol and thus decrease cholesterol-rich caveolar lipid rafts in a neuronal cell line (N27 cells). Nystatin was applied prior to testosterone exposure in oxidatively stressed N27 cells. Cell viability, endocytosis, and protein analysis of oxidative stress, apoptosis, and mAR localization were conducted. Our results show that the loss of lipid rafts via cholesterol sequestering blocked androgen-induced oxidative stress in cells by decreasing the localization of mAR to caveolar lipid rafts.
Collapse
Affiliation(s)
- Oluwadarasimi Fadeyibi
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Nataliya Rybalchenko
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Steve Mabry
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, Fort Worth, Texas
| | - Dianna H Nguyen
- Department of Physiology & Anatomy, Graduate School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, Texas
| | - Rebecca L Cunningham
- Department of Pharmaceutical Sciences, School of Pharmacy, University of North Texas Health Science Center, Fort Worth, Texas
| |
Collapse
|
26
|
Statins and Gliomas: A Systematic Review of the Preclinical Studies and Meta-Analysis of the Clinical Literature. Drugs 2022; 82:293-310. [PMID: 35122635 DOI: 10.1007/s40265-021-01668-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Gliomas represent most common primary brain tumors. Glioblastoma (GBM) is the most common subtype and carries a poor prognosis. There is growing interest in the anti-glioma properties of statins. The aim of this study was to conduct a systematic review of the preclinical literature and to meta-analyze existing clinical studies to determine what benefit, if any, statins may confer in the context of glioma. METHODS The PubMed, Embase, Cochrane, and Web of Science libraries were queried in May 2021. Preclinical studies were included if they investigated the anti-cancer effects of statins in glioma in vitro and in vivo. Clinical studies were included if they reported incidence rates of glioma by statin use, or mortality outcomes among GBM patients by statin use. Pooled point estimates were calculated using a random-effects model. RESULTS In total, 64 publications, 51 preclinical and 13 clinical, were included. Preclinical studies indicated that statins inhibited glioma cell proliferation, migration, and invasion. These effects were time- and concentration-dependent. Synergistic anti-glioma effects were observed when statins were combined with other anti-cancer therapies. Clinical observational studies showed an inverse, albeit non-statistically significant, association between statin use and incidence rate of glioma (HR = 0.84, 95% CI 0.62-1.13, I2 = 72%, p-heterogeneity = 0.003, 6 studies). Statin use was not associated with better overall survival following GBM surgery (HR = 1.05, 95% CI 0.85-1.30, I2 = 30%, p-heterogeneity = 0.23, 4 studies). CONCLUSION Statins were potent anti-cancer drugs that suppressed glioma growth through various mechanisms in vitro; these effects have translated into the clinical realm, clinically but not statistically, in terms of glioma incidence but not GBM survival.
Collapse
|
27
|
Luttman JH, Hoj JP, Lin KH, Lin J, Gu JJ, Rouse C, Nichols AG, MacIver NJ, Wood KC, Pendergast AM. ABL allosteric inhibitors synergize with statins to enhance apoptosis of metastatic lung cancer cells. Cell Rep 2021; 37:109880. [PMID: 34706244 PMCID: PMC8579324 DOI: 10.1016/j.celrep.2021.109880] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 08/29/2021] [Accepted: 10/04/2021] [Indexed: 12/25/2022] Open
Abstract
Targeting mitochondrial metabolism has emerged as a treatment option for cancer patients. The ABL tyrosine kinases promote metastasis, and enhanced ABL signaling is associated with a poor prognosis in lung adenocarcinoma patients. Here we show that ABL kinase allosteric inhibitors impair mitochondrial integrity and decrease oxidative phosphorylation. To identify metabolic vulnerabilities that enhance this phenotype, we utilized a CRISPR/Cas9 loss-of-function screen and identified HMG-CoA reductase, the rate-limiting enzyme of the mevalonate pathway and target of statin therapies, as a top-scoring sensitizer to ABL inhibition. Combination treatment with ABL allosteric inhibitors and statins decreases metastatic lung cancer cell survival in vitro in a synergistic manner. Notably, combination therapy in mouse models of lung cancer brain metastasis and therapy resistance impairs metastatic colonization with a concomitant increase in animal survival. Thus, metabolic combination therapy might be effective to decrease metastatic outgrowth, leading to increased survival for lung cancer patients with advanced disease. Metabolic reprogramming in tumors is an adaptation that generates vulnerabilities that can be exploited for developing new therapies. Here Luttman et al. identify synergism between ABL allosteric inhibitors and lipophilic statins to impair metastatic lung cancer cell outgrowth and colonization, leading to increased survival in mouse models of advanced disease.
Collapse
Affiliation(s)
- Jillian Hattaway Luttman
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Jacob P Hoj
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Kevin H Lin
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Jiaxing Lin
- Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA
| | - Jing Jin Gu
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA
| | - Clay Rouse
- Division of Laboratory Animal Resources, Duke University School of Medicine, Durham, NC, USA
| | - Amanda G Nichols
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Nancie J MacIver
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA; Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA; Department of Immunology, Duke University School of Medicine, Durham, NC, USA
| | - Kris C Wood
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA; Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA
| | - Ann Marie Pendergast
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, NC, USA; Duke Cancer Institute, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
28
|
Song Y, Liu J, Zhao K, Gao L, Zhao J. Cholesterol-induced toxicity: An integrated view of the role of cholesterol in multiple diseases. Cell Metab 2021; 33:1911-1925. [PMID: 34562355 DOI: 10.1016/j.cmet.2021.09.001] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/16/2021] [Accepted: 09/07/2021] [Indexed: 12/23/2022]
Abstract
High levels of cholesterol are generally considered to be associated with atherosclerosis. In the past two decades, however, a number of studies have shown that excess cholesterol accumulation in various tissues and organs plays a critical role in the pathogenesis of multiple diseases. Here, we summarize the effects of excess cholesterol on disease pathogenesis, including liver diseases, diabetes, chronic kidney disease, Alzheimer's disease, osteoporosis, osteoarthritis, pituitary-thyroid axis dysfunction, immune disorders, and COVID-19, while proposing that excess cholesterol-induced toxicity is ubiquitous. We believe this concept will help broaden the appreciation of the toxic effect of excess cholesterol, and thus potentially expand the therapeutic use of cholesterol-lowering medications.
Collapse
Affiliation(s)
- Yongfeng Song
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong 250021, China; Shandong Institute of Endocrine & Metabolic Disease, Jinan, Shandong 250062, China
| | - Junjun Liu
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong 250021, China; Shandong Institute of Endocrine & Metabolic Disease, Jinan, Shandong 250062, China
| | - Ke Zhao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong 250021, China; Shandong Institute of Endocrine & Metabolic Disease, Jinan, Shandong 250062, China
| | - Ling Gao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong 250021, China; Shandong Institute of Endocrine & Metabolic Disease, Jinan, Shandong 250062, China
| | - Jiajun Zhao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Jinan, Shandong 250021, China; Shandong Institute of Endocrine & Metabolic Disease, Jinan, Shandong 250062, China.
| |
Collapse
|
29
|
Vogt NM, Hunt JFV, Ma Y, Van Hulle CA, Adluru N, Chappell RJ, Lazar KK, Jacobson LE, Austin BP, Asthana S, Johnson SC, Bendlin BB, Carlsson CM. Effects of simvastatin on white matter integrity in healthy middle-aged adults. Ann Clin Transl Neurol 2021; 8:1656-1667. [PMID: 34275209 PMCID: PMC8351379 DOI: 10.1002/acn3.51421] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/22/2021] [Accepted: 06/23/2021] [Indexed: 11/29/2022] Open
Abstract
Background The brain is the most cholesterol‐rich organ and myelin contains 70% of total brain cholesterol. Statins are potent cholesterol‐lowing medications used by millions of adults for prevention of vascular disease, yet the effect of statins on cholesterol‐rich brain white matter (WM) is largely unknown. Methods We used longitudinal neuroimaging data acquired from 73 healthy, cognitively unimpaired, statin‐naïve, middle‐aged adults during an 18‐month randomized controlled trial of simvastatin 40 mg daily (n = 35) or matching placebo (n = 38). ANCOVA models (covariates: age, sex, APOE‐ɛ4) tested the effect of treatment group on percent change in WM, gray matter (GM), and WM hyperintensity (WMH) neuroimaging measures at each study visit. Mediation analysis tested the indirect effects of simvastatin on WM microstructure through change in serum total cholesterol levels. Results At 18 months, the simvastatin group showed a significant preservation in global WM fractional anisotropy (β = 0.88%, 95% CI 0.27 to 1.50, P = 0.005), radial diffusivity (β = −1.10%, 95% CI −2.13 to −0.06, P = 0.039), and WM volume (β = 0.72%, 95% CI 0.13 to 1.32, P = 0.018) relative to the placebo group. There was no significant effect of simvastatin on GM or WMH volume. Change in serum total cholesterol mediated approximately 30% of the effect of simvastatin on WM microstructure. Conclusions Simvastatin treatment in healthy, middle‐aged adults resulted in preserved WM microstructure and volume at 18 months. The partial mediation by serum cholesterol reduction suggests both peripheral and central mechanisms. Future studies are needed to determine whether these effects persist and translate to cognitive outcomes. Trial Registration NCT00939822 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Nicholas M Vogt
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Jack F V Hunt
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Yue Ma
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Carol A Van Hulle
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Nagesh Adluru
- Waisman Laboratory for Brain Imaging and Behavior, Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - Richard J Chappell
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin.,Department of Biostatistics and Medical Informatics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Karen K Lazar
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Laura E Jacobson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Benjamin P Austin
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Sanjay Asthana
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin.,Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin.,Geriatrics Division, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Sterling C Johnson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin.,Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin.,Geriatrics Division, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin.,Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Barbara B Bendlin
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin.,Geriatrics Division, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin.,Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Cynthia M Carlsson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin.,Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin.,Geriatrics Division, Department of Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin.,Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| |
Collapse
|
30
|
Menze ET, Ezzat H, Shawky S, Sami M, Selim EH, Ahmed S, Maged N, Nadeem N, Eldash S, Michel HE. Simvastatin mitigates depressive-like behavior in ovariectomized rats: Possible role of NLRP3 inflammasome and estrogen receptors' modulation. Int Immunopharmacol 2021; 95:107582. [PMID: 33774267 DOI: 10.1016/j.intimp.2021.107582] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 02/01/2021] [Accepted: 03/09/2021] [Indexed: 12/27/2022]
Abstract
It is well known that females are more vulnerable than males to stress-related psychiatric disorders, particularly during perimenopausal and postmenopausal periods. Hormone replacement therapy (HRT) has been widely used for the management of postmenopausal depression. However, HRT could be associated with severe adverse effects, including increased risk for coronary heart disease, breast cancer and endometrial cancer. Thus, there is a pressing demand for novel therapeutic options for postmenopausal depression without sacrificing uterine health. Simvastatin (SIM) was proven to have neuroprotective activities besides its hypocholesterolemic effect, the former can be attributed to its, antioxidant, anti-apoptotic and anti-inflammatory activities. Moreover, many reports highlighted that SIM has estrogenic activity and was able to induce the expression of estrogen receptors in rats. The present study showed that SIM (20 mg/kg, p.o.) markedly attenuated depressive-like behavior in ovariectomized (OVX) rats. Moreover, SIM prohibited hippocampal microglial activation, abrogated P2X7 receptor, TLR2 and TLR4 expression, inhibited NLRP3 inflammasome activation, with subsequent reduction in the levels of pro-inflammatory mediators; IL-1β and IL-18. Furthermore, a marked elevation in hippocampal expression of ERα and ERβ was noted in SIM-treated animals, without any significant effect on uterine relative weight or ERα expression. Taken together, SIM could provide a safer alternative for HRT for the management of postmenopausal depression, without any hyperplastic effect on the uterus.
Collapse
Affiliation(s)
- Esther T Menze
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Hager Ezzat
- Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Salma Shawky
- Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Marwa Sami
- Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Eman H Selim
- Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Samar Ahmed
- Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Nouran Maged
- Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Nancy Nadeem
- Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | | | - Haidy E Michel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
31
|
Lin CH, Chang CH, Tai CH, Cheng MF, Chen YC, Chao YT, Huang TL, Yen RF, Wu RM. A Double-Blind, Randomized, Controlled Trial of Lovastatin in Early-Stage Parkinson's Disease. Mov Disord 2021; 36:1229-1237. [PMID: 33449392 DOI: 10.1002/mds.28474] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 12/05/2020] [Accepted: 12/08/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Recent evidence indicates that lipophilic statins have a neuroprotective benefit in animal models of Parkinson's disease (PD). The objective of this study was to evaluate whether lovastatin has the potential to slow motor symptom progression in patients with early-stage PD. METHODS This double-blind, randomized, placebo-controlled trial enrolled 77 patients with early-stage PD between May 23, 2017, and July 12, 2018, with follow-up ending September 1, 2019. Lovastatin 80 mg/day or placebo with 1:1 randomization was administered for 48 weeks. Mean change in the parts I-III scores of the Movement Disorder Society-Unified Parkinson's Disease Rating Scale (MDS-UPDRS), changes in the striatal dopamine uptake ratio measured by 18 F-dopa PET scan, and changes in PD medications between baseline and the week 48 visit were measured. RESULTS Of the 77 randomized patients, 70 (90.9%) completed the study. There was a slightly beneficial trend of the MDS-UPDRS motor score in the lovastatin group (-3.18 ± 5.50) compared with the placebo group (-0.50 ± 6.11); P = 0.14 adjusted for age, sex, disease duration, and baseline LEDD. Mean percentage change in the striatal 18 F-dopa uptake ratio deteriorated less in the lovastatin group than in the placebo group on the dominant side of caudate (1.2% ± 7.3% vs -7.1% ± 8.2%, P < 0.01) and putamen (2.3% ± 7.1% vs -6.4% ± 8.1%, P < 0.01). We found no between-group differences in the change in part I or part II MDS-UPDRS scores. Lovastatin was generally well tolerated. CONCLUSIONS Lovastatin treatment in patients with early-stage PD was associated with a trend of less motor symptom worsening and was well tolerated. A future larger long-term follow-up study is needed to confirm our findings. © 2021 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Chin-Hsien Lin
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan.,Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Chin-Hao Chang
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Chun-Hwei Tai
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Mei-Fang Cheng
- Department of Nuclear Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Yi-Chieh Chen
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Ying-Ting Chao
- Department of Nuclear Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Tse-Le Huang
- Department of Nuclear Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Ruoh-Fang Yen
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Ruey-Meei Wu
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
32
|
Lipid-core nanocapsules containing simvastatin improve the cognitive impairment induced by obesity and hypercholesterolemia in adult rats. Eur J Pharm Sci 2020; 151:105397. [DOI: 10.1016/j.ejps.2020.105397] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 05/06/2020] [Accepted: 05/26/2020] [Indexed: 01/07/2023]
|
33
|
Bagheri H, Ghasemi F, Barreto GE, Sathyapalan T, Jamialahmadi T, Sahebkar A. The effects of statins on microglial cells to protect against neurodegenerative disorders: A mechanistic review. Biofactors 2020; 46:309-325. [PMID: 31846136 DOI: 10.1002/biof.1597] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 11/22/2019] [Indexed: 12/28/2022]
Abstract
Microglia are the primary innate immune system cells in the central nervous system (CNS). They are crucial for the immunity, neurogenesis, synaptogenesis, neurotrophic support, phagocytosis of cellular debris, and maintaining the CNS integrity and homeostasis. Invasion by pathogens as well as in CNS injuries and damages results in activation of microglia known as microgliosis. The activated microglia have the capacity to release proinflammatory mediators leading to neuroinflammation. However, uncontrolled neuroinflammation can give rise to various neurological disorders (NDs), especially the neurodegenerative diseases including Parkinson's disease (PD) and related disorders, Alzheimer's disease (AD) and other dementias, multiple sclerosis (MS), Huntington's disease (HD), spinocerebellar ataxia (SCA), spinal muscular atrophy (SMA), amyotrophic lateral sclerosis (ALS), and stroke. Statins (HMG-CoA reductase inhibitors) are among the most widely prescribed medications for the management of hypercholesterolemia worldwide. It can be used for primary prevention in healthy individuals who are at higher risk of cardiovascular and coronary heart diseases as well as the secondary prevention in patients with cardiovascular and coronary heart diseases disease. A growing body of evidence has indicated that statins have the potential to attenuate the proinflammatory mediators and subsequent NDs by controlling the microglial activation and consequent reduction in neuroinflammatory mediators. In this review, we have discussed the recent studies on the effects of statins on microglia activation and neuroinflammation.
Collapse
Affiliation(s)
- Hossein Bagheri
- Department of Biotechnology, Faculty of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Faezeh Ghasemi
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Tehran, Iran
| | - George E Barreto
- Department of Biological Sciences, University of Limerick, Limerick, Ireland
- Health Research Institute, University of Limerick, Limerick, Ireland
| | - Thozhukat Sathyapalan
- Department of Academic Diabetes, Endocrinology and Metabolism, Hull York Medical School, University of Hull, Hull, UK
| | - Tannaz Jamialahmadi
- Halal Research Center of IRI, FDA, Tehran, Iran
- Department of Nutrition, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
34
|
Sun D, Zhang F, Ma T, Zhang Y, Liang Z. Atorvastatin alleviates left ventricular remodeling in isoproterenol-induced chronic heart failure in rats by regulating the RhoA/Rho kinase signaling pathway. Pharmacol Rep 2020; 72:903-911. [PMID: 32144744 DOI: 10.1007/s43440-020-00085-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 02/13/2020] [Accepted: 02/21/2020] [Indexed: 01/11/2023]
Abstract
BACKGROUND Chronic heart failure (CHF) is characterized by left ventricular dysfunction and altered autonomic control of cardiac function. This study aimed to investigate the effects of atorvastatin on left ventricular remodeling (LVR) and cardiac function in rats with isoproterenol-induced CHF and the possible mechanism. METHODS An isoproterenol-induced CHF model was established in rata, which were subsequently treated with atorvastatin. Echocardiography, hemodynamic, and left ventricular mass indexes were assessed. The mRNA expression of RhoA, Rho kinase, and endothelial nitric oxide synthase (eNOS) was determined by RT-qPCR. The protein expression of myosin-binding subunit (MBS), MBS-P, eNOS, phosphorylated-eNOS, RhoA, and Rho kinase was measured by Western blot analysis. The relative activity of NADPH oxidase, ROS, and NO was assessed by ELISA. RESULTS Isoproterenol-induced CHF rats treated with atorvastatin exhibited decreased left ventricular end-systolic dimension, left ventricular end-diastolic dimension, left ventricular end-diastolic pressure, left ventricular mass index, maximum fall rate of change in left ventricular pressure, heart rate (p < 0.001), expression of RhoA, Rho kinase, MBS and MBS-P (p < 0.01), and relative activity of NADPH oxidase, ROS and NO (p < 0.05) and increased left ventricular short axis fractional shortening, left ventricular end-systolic pressure, maximum rise rate of change in left ventricular pressure (p < 0.001) and expression of eNOS, and phosphorylated-eNOS ser1177 (all p < 0.05) compared with those of rats with isoproterenol-induced CHF. CONCLUSION We demonstrated that atorvastatin inhibits LVR and improves cardiac function in rats with isoproterenol-induced CHF through inhibition of the RhoA/Rho kinase signaling pathway.
Collapse
Affiliation(s)
- Dingjun Sun
- Department of Cardiology, Central South University Xiangya School of Medicine Affiliated Haikou Hospital (Haikou People's Hospital), Haikou, 570208, People's Republic of China
| | - Fuwei Zhang
- Department of Cardiology, Central South University Xiangya School of Medicine Affiliated Haikou Hospital (Haikou People's Hospital), Haikou, 570208, People's Republic of China
| | - Tianyi Ma
- Department of Cardiology, Central South University Xiangya School of Medicine Affiliated Haikou Hospital (Haikou People's Hospital), Haikou, 570208, People's Republic of China
| | - Yixue Zhang
- Department of Cardiology, Central South University Xiangya School of Medicine Affiliated Haikou Hospital (Haikou People's Hospital), Haikou, 570208, People's Republic of China
| | - Zhongshu Liang
- Department of Cardiology, The Third Xiangya Hospital of Central South University, No. 138, Tongzipo Road, Yuelu District, Changsha, 410013, Hunan, People's Republic of China.
| |
Collapse
|
35
|
Hsieh PF, Tung H, Lin CH. Statin effects on post-stroke epilepsy and mortality – Taiwan population-based study. Neurol Res 2020; 42:422-429. [DOI: 10.1080/01616412.2020.1735821] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Peiyuan F. Hsieh
- Taichung Veterans General Hospital, Neurological Institute, Taichung, Taiwan, ROC
| | - Hsin Tung
- Department of Medical Education, Taichung Veterans General Hospital, Taichung, Taiwan, ROC
| | - Ching-Heng Lin
- Health Service Research Center, Taichung Veterans General Hospital, Taichung, Taiwan, ROC
| |
Collapse
|
36
|
Williams EI, Betterton RD, Davis TP, Ronaldson PT. Transporter-Mediated Delivery of Small Molecule Drugs to the Brain: A Critical Mechanism That Can Advance Therapeutic Development for Ischemic Stroke. Pharmaceutics 2020; 12:pharmaceutics12020154. [PMID: 32075088 PMCID: PMC7076465 DOI: 10.3390/pharmaceutics12020154] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 02/10/2020] [Accepted: 02/11/2020] [Indexed: 12/28/2022] Open
Abstract
Ischemic stroke is the 5th leading cause of death in the United States. Despite significant improvements in reperfusion therapies, stroke patients still suffer from debilitating neurocognitive deficits. This indicates an essential need to develop novel stroke treatment paradigms. Endogenous uptake transporters expressed at the blood-brain barrier (BBB) provide an excellent opportunity to advance stroke therapy via optimization of small molecule neuroprotective drug delivery to the brain. Examples of such uptake transporters include organic anion transporting polypeptides (OATPs in humans; Oatps in rodents) and organic cation transporters (OCTs in humans; Octs in rodents). Of particular note, small molecule drugs that have neuroprotective properties are known substrates for these transporters and include 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors (i.e., statins) for OATPs/Oatps and 1-amino-3,5-dimethyladamantane (i.e., memantine) for OCTs/Octs. Here, we review current knowledge on specific BBB transporters that can be targeted for improvement of ischemic stroke treatment and provide state-of-the-art perspectives on the rationale for considering BBB transport properties during discovery/development of stroke therapeutics.
Collapse
|
37
|
Sahu SS, Sarkar P, Shrivastava S, Chattopadhyay A. Differential effects of simvastatin on membrane organization and dynamics in varying phases. Chem Phys Lipids 2019; 225:104831. [DOI: 10.1016/j.chemphyslip.2019.104831] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 09/15/2019] [Accepted: 09/20/2019] [Indexed: 12/24/2022]
|
38
|
Abstract
OBJECTIVE Statins are a class of drugs that competitively bind to the active site of HMG-CoA reductase enzyme, thereby inhibiting the initial steps in cholesterol synthesis. Originally approved for use in lowering serum cholesterol, a risk factor for developing atherosclerosis and coronary heart disease, statins have subsequently been noted to have myriad extrahepatic effects, including potential effects on cognition, diabetes, breast cancer, bone, and muscle. This narrative review assesses the current state of the science regarding the risks and benefits of statin therapy in women to identify areas where additional research is needed. METHODS Basic and clinical studies were identified by searching PubMed with particular attention to inclusion of female animals, women, randomized controlled trials, and sex-specific analyses. RESULTS Statin therapy is generally recommended to reduce the risk of cardiovascular disease. None of the current clinical guidelines, however, offer sex-specific recommendations for women due to lack of understanding of sex differences and underlying mechanisms of disease processes. In addition, conclusions regarding efficacy of treatments do not consider lipid solubility for the drug, dosing, duration of treatment, interactions with estrogen, or comorbidities. Pleiotropic effects of statins are often derived from secondary analysis of studies with cardiovascular events as primary outcomes. CONCLUSIONS Many of the trials that have established the efficacy and safety of statins were conducted predominantly or entirely in men, with results extrapolated to women. Additional research is needed to guide clinical recommendations specific to women. : Video Summary:http://links.lww.com/MENO/A462.
Collapse
Affiliation(s)
- Stephanie S. Faubion
- Center for Women’s Health, Mayo Clinic, Rochester, MN
- Division of General Internal Medicine, Mayo Clinic, Rochester, MN
| | - Ekta Kapoor
- Center for Women’s Health, Mayo Clinic, Rochester, MN
- Division of General Internal Medicine, Mayo Clinic, Rochester, MN
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Mayo Clinic, Rochester, MN
| | - Ann M. Moyer
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Rochester, MN
| | - Howard N. Hodis
- Atherosclerosis Research Unit, Departments of Medicine and Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA
| | - Virginia M. Miller
- Departments of Surgery and Physiology & Biomedical Engineering, Women’s Health Research Center, Mayo Clinic, Rochester, MN
| |
Collapse
|
39
|
Combined Treatment with Hydrophilic and Lipophilic Statins Improves Neurological Outcomes Following Experimental Cardiac Arrest in Mice. Neurocrit Care 2019; 33:64-72. [DOI: 10.1007/s12028-019-00862-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
40
|
Tsang JKW, Liu J, Lo ACY. Vascular and Neuronal Protection in the Developing Retina: Potential Therapeutic Targets for Retinopathy of Prematurity. Int J Mol Sci 2019; 20:E4321. [PMID: 31484463 PMCID: PMC6747312 DOI: 10.3390/ijms20174321] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/21/2019] [Accepted: 08/29/2019] [Indexed: 12/13/2022] Open
Abstract
Retinopathy of prematurity (ROP) is a common retinal disease in preterm babies. To prolong the lives of preterm babies, high oxygen is provided to mimic the oxygen level in the intrauterine environment for postnatal organ development. However, hyperoxia-hypoxia induced pathological events occur when babies return to room air, leading to ROP with neuronal degeneration and vascular abnormality that affects retinal functions. With advances in neonatal intensive care, it is no longer uncommon for increased survival of very-low-birth-weight preterm infants, which, therefore, increased the incidence of ROP. ROP is now a major cause of preventable childhood blindness worldwide. Current proven treatment for ROP is limited to invasive retinal ablation, inherently destructive to the retina. The lack of pharmacological treatment for ROP creates a great need for effective and safe therapies in these developing infants. Therefore, it is essential to identify potential therapeutic agents that may have positive ROP outcomes, especially in preserving retinal functions. This review gives an overview of various agents in their efficacy in reducing retinal damages in cell culture tests, animal experiments and clinical studies. New perspectives along the neuroprotective pathways in the developing retina are also reviewed.
Collapse
Affiliation(s)
- Jessica K W Tsang
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jin Liu
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Amy C Y Lo
- Department of Ophthalmology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
41
|
Transmucosal Absorption Enhancers in the Drug Delivery Field. Pharmaceutics 2019; 11:pharmaceutics11070339. [PMID: 31311173 PMCID: PMC6680553 DOI: 10.3390/pharmaceutics11070339] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 07/11/2019] [Indexed: 01/11/2023] Open
Abstract
Drug delivery systems that safely and consistently improve transport of poorly absorbed compounds across epithelial barriers are highly sought within the drug delivery field. The use of chemical permeation enhancers is one of the simplest and widely tested approaches to improve transmucosal permeability via oral, nasal, buccal, ocular and pulmonary routes. To date, only a small number of permeation enhancers have progressed to clinical trials, and only one product that includes a permeation enhancer has reached the pharmaceutical market. This editorial is an introduction to the special issue entitled Transmucosal Absorption Enhancers in the Drug Delivery Field (https://www.mdpi.com/journal/pharmaceutics/special_issues/transmucosal_absorption_enhancers). The guest editors outline the scope of the issue, reflect on the results and the conclusions of the 19 articles published in the issue and provide an outlook on the use of permeation enhancers in the drug delivery field.
Collapse
|
42
|
Agrawal S, Vamadevan P, Mazibuko N, Bannister R, Swery R, Wilson S, Edwards S. A New Method for Ethical and Efficient Evidence Generation for Off-Label Medication Use in Oncology (A Case Study in Glioblastoma). Front Pharmacol 2019; 10:681. [PMID: 31316378 PMCID: PMC6610246 DOI: 10.3389/fphar.2019.00681] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 05/27/2019] [Indexed: 12/23/2022] Open
Abstract
In oncology, preclinical and early clinical data increasingly support the use of a number of candidate "non-cancer" drugs in an off-label setting against multiple tumor types. In particular, metabolically targeted drugs show promise as adjuvant chemo and radiosensitizers, improving or restoring sensitivity to standard therapies. The time has come for large scale clinical studies of off-label drugs in this context. However, it is well recognized that high-cost randomized controlled trials may not be an economically viable option for studying patent-expired off-label drugs. In some cases, randomized trials could also be considered as ethically controversial. This perspective article presents a novel approach to generating additional clinical data of sufficient quality to support changes in clinical practice and relabeling of such drugs for use in oncology. Here, we suggest that a pluralistic evidence base and triangulation of evidence can support clinical trial data for off-label drug use in oncology. An example of an off-label drug protocol brought to the clinic for glioblastoma patients is presented, along with preliminary retrospective data from the METRICS study (NCT02201381). METRICS is a novel participant-funded, open-label, non-randomized, single-arm real-world study designed to gather high-quality evidence on the safety, tolerability, and effectiveness of four off-label metabolically targeted medicines as an adjunctive cancer treatment for glioblastoma patients.
Collapse
Affiliation(s)
- Samir Agrawal
- Blizard Institute, Queen Mary University of London, London, United Kingdom
- St Bartholomew’s Hospital, Bart’s Health NHS Trust, London, United Kingdom
| | | | - Ndaba Mazibuko
- Department of Neuroimaging, Institute of Psychiatry, Psychology & Neuroscience, King’s College London, London, United Kingdom
| | | | | | | | - Sarah Edwards
- Department of Science and Technology Studies, University College London, London, United Kingdom
| |
Collapse
|
43
|
Hirunpattarasilp C, Attwell D, Freitas F. The role of pericytes in brain disorders: from the periphery to the brain. J Neurochem 2019; 150:648-665. [PMID: 31106417 DOI: 10.1111/jnc.14725] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 03/15/2019] [Accepted: 05/15/2019] [Indexed: 12/13/2022]
Abstract
It is becoming increasingly apparent that disorders of the brain microvasculature contribute to many neurological disorders. In recent years it has become clear that a major player in these events is the capillary pericyte which, in the brain, is now known to control the blood-brain barrier, regulate blood flow, influence immune cell entry and be crucial for angiogenesis. In this review we consider the under-explored possibility that peripheral diseases which affect the microvasculature, such as hypertension, kidney disease and diabetes, produce central nervous system (CNS) dysfunction by mechanisms affecting capillary pericytes within the CNS. We highlight how cellular messengers produced peripherally can act via signalling pathways within CNS pericytes to reshape blood vessels, restrict blood flow or compromise blood-brain barrier function, thus causing neuronal dysfunction. Increased understanding of how renin-angiotensin, Rho-kinase and PDGFRβ signalling affect CNS pericytes may suggest novel therapeutic approaches to reducing the CNS effects of peripheral disorders.
Collapse
Affiliation(s)
- Chanawee Hirunpattarasilp
- Department of Neuroscience, Andrew Huxley Building, University College London, Physiology & Pharmacology, Gower Street, London, UK
| | - David Attwell
- Department of Neuroscience, Andrew Huxley Building, University College London, Physiology & Pharmacology, Gower Street, London, UK
| | - Felipe Freitas
- Department of Neuroscience, Andrew Huxley Building, University College London, Physiology & Pharmacology, Gower Street, London, UK
| |
Collapse
|
44
|
Lorenzoni R, Cordenonsi LM, Davies S, Antonow MB, Medina Diedrich AS, Santos CG, Vitalis GS, Garrastazu G, Buttini F, Sonvico F, Gomes P, Raffin RP. Lipid-core nanocapsules are an alternative to the pulmonary delivery and to increase the stability of statins. J Microencapsul 2019; 36:317-326. [PMID: 31159613 DOI: 10.1080/02652048.2019.1624849] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Aims: Lipid-core nanocapsules (LNCs) loaded with simvastatin (SV, SV-LNC) or lovastatin (LV, LV-LNC) were formulated for pulmonary administration. Methods: The LNC suspensions were characterized physicochemically, their stability was evaluated, and drug delivery by the pulmonary route was tested in vitro. Results: The loaded LNCs had a particle size close to 200 nm, a low polydispersity index, and a zeta potential around -20 mV. The encapsulation efficiency was high for SV (99.21 ± 0.7%) but low for LV (20.34 ± 1.2%). SV release from nanocapsules was slower than it was from SV in solution, with a monoexponential release profile, and the drug emitted and aerosol output rate was higher for SV-LNCs (1.58 µg/s) than for SV in suspension (0.54 µg/s). Conclusions: SV-LNCs had a median aerodynamic diameter of 3.51 µm and a highly respirable fraction (61.9%), indicating that nanoparticles are a suitable system for efficient delivery of simvastatin to the lung.
Collapse
Affiliation(s)
- Ricardo Lorenzoni
- a Nanoscience Post-Graduation Program , Franciscan University , Santa Maria , Rio Grande do Sul , Brazil
| | - Leticia Malgarim Cordenonsi
- a Nanoscience Post-Graduation Program , Franciscan University , Santa Maria , Rio Grande do Sul , Brazil.,b Pharmaceutical Sciences Post-Graduation Program , Federal University of Rio Grande do Sul State , Porto Alegre , Brazil
| | - Samuel Davies
- a Nanoscience Post-Graduation Program , Franciscan University , Santa Maria , Rio Grande do Sul , Brazil.,b Pharmaceutical Sciences Post-Graduation Program , Federal University of Rio Grande do Sul State , Porto Alegre , Brazil
| | - Michelli Barcelos Antonow
- a Nanoscience Post-Graduation Program , Franciscan University , Santa Maria , Rio Grande do Sul , Brazil.,c Pharmaceutical Nanotechnology Post-Graduation Program , Federal University of Rio Grande do Sul State , Porto Alegre , Brazil
| | | | - Cayane Genro Santos
- a Nanoscience Post-Graduation Program , Franciscan University , Santa Maria , Rio Grande do Sul , Brazil
| | - Graciela Schneider Vitalis
- a Nanoscience Post-Graduation Program , Franciscan University , Santa Maria , Rio Grande do Sul , Brazil
| | | | - Francesca Buttini
- d Faculty of Pharmacy , Universitá degli Studi di Parma , Parma , Italy
| | - Fabio Sonvico
- d Faculty of Pharmacy , Universitá degli Studi di Parma , Parma , Italy
| | - Patrícia Gomes
- a Nanoscience Post-Graduation Program , Franciscan University , Santa Maria , Rio Grande do Sul , Brazil
| | - Renata Platcheck Raffin
- a Nanoscience Post-Graduation Program , Franciscan University , Santa Maria , Rio Grande do Sul , Brazil
| |
Collapse
|
45
|
Pająk B, Kania E, Gołaszewska A, Orzechowski A. Preliminary Study on Clusterin Protein (sCLU) Expression in PC-12 Cells Overexpressing Wild-Type and Mutated (Swedish) AβPP genes Affected by Non-Steroid Isoprenoids and Water-Soluble Cholesterol. Int J Mol Sci 2019; 20:E1481. [PMID: 30909654 PMCID: PMC6470582 DOI: 10.3390/ijms20061481] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 03/04/2019] [Accepted: 03/18/2019] [Indexed: 02/07/2023] Open
Abstract
In this study we attempted to verify the hypothesis that the mevalonate pathway affects amyloid beta precursor protein (AβPP) processing and regulates clusterin protein levels. AβPP expression was monitored by green fluorescence (FL) and Western blot (WB). WB showed soluble amyloid protein precursor alpha (sAβPPα) presence in AβPP-wt cells and Aβ expression in AβPP-sw cells. Nerve growth factor (NGF)-differentiated rat neuronal pheochromocytoma PC-12 cells were untreated/treated with statins alone or together with non-sterol isoprenoids. Co-treatment with mevalonate, dolichol, ubiquinol, farnesol, geranylgeraniol, or water-soluble cholesterol demonstrated statin-dependent neurotoxicity resulted from the attenuated activity of mevalonate pathway rather than lower cholesterol level. Atorvastatin (50 μM) or simvastatin (50 μM) as well as cholesterol chelator methyl-β-cyclodextrin (0.2 mM) diminished cell viability (p < 0.05) and clusterin levels. Interestingly, co-treatment with mevalonate, dolichol, ubiquinol, farnesol, geranylgeraniol, or water-soluble cholesterol stimulated (p < 0.05) clusterin expression. Effects of non-sterol isoprenoids, but not water soluble cholesterol (Chol-PEG), were the most significant in mock-transfected cells. Geranylgeraniol (GGOH) overcame atorvastatin (ATR)-dependent cytotoxicity. This effect does not seem to be dependent on clusterin, as its level became lower after GGOH. The novelty of these findings is that they show that the mevalonate (MEV) pathway rather than cholesterol itself plays an important role in clusterin expression levels. In mock-transfected, rather than in AβPP-overexpressing cells, GGOH/farnesol (FOH) exerted a protective effect. Thus, protein prenylation with GGOH/FOH might play substantial role in neuronal cell survival.
Collapse
Affiliation(s)
- Beata Pająk
- Independent Laboratory of Genetics and Molecular Biology, Kaczkowski Military Institute of Hygiene and Epidemiology, Kozielska 4, 01-163 Warsaw, Poland.
| | - Elżbieta Kania
- Tumor Cell Death Laboratory, Cancer Research UK, Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK.
| | - Anita Gołaszewska
- Department of Neuroendocrinology, Centre of Postgraduate Medical Education, Marymoncka 99/103, 01-813 Warsaw, Poland.
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences ⁻ SGGW, Nowoursynowska 159, 02-776 Warsaw, Poland.
| | - Arkadiusz Orzechowski
- Department of Physiological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences ⁻ SGGW, Nowoursynowska 159, 02-776 Warsaw, Poland.
| |
Collapse
|
46
|
Harshman SG, Shea MK, Fu X, Grusak MA, Smith D, Lamon-Fava S, Kuliopulos A, Greenberg A, Booth SL. Atorvastatin Decreases Renal Menaquinone-4 Formation in C57BL/6 Male Mice. J Nutr 2019; 149:416-421. [PMID: 30753659 PMCID: PMC6398385 DOI: 10.1093/jn/nxy290] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 07/30/2018] [Accepted: 10/25/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Menaquinone-4 (MK4), a vitamin K metabolite, is converted from phylloquinone through a process that requires intermediates of endogenous cholesterol production. Recent evidence suggests that MK4 is involved in kidney function. OBJECTIVE The purpose of this study was to determine the effect of atorvastatin treatment on MK4 formation in young and old male mice. METHODS C57BL/6 male mice (4-mo-old and 20-mo-old) were randomly assigned to either a diet containing 300 mg atorvastatin/kg with 3 mg phylloquinone/kg or a control diet containing 3 mg phylloquinone/kg for 8 wk. During week 8, all mice received deuterium-labeled phylloquinone in the diet. Labeled and unlabeled phylloquinone and MK4 in liver, kidney, brain, and intestine were measured by atmospheric pressure chemical ionization LC/MS. 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase gene expression was quantified by reverse transcriptase-PCR. Tissue MK4 and phylloquinone concentrations were compared between atorvastatin treatment groups with use of general linear models. RESULTS There was no age-treatment interaction on MK4 tissue concentrations. In atorvastatin-treated mice, total MK4 and percentage of deuterium-labeled MK4 in kidney were both approximately 45% lower compared to values in mice not given atorvastatin (all P < 0.05). MK4 concentrations did not differ between groups in any other tissue measured. CONCLUSION In male mice, atorvastatin reduced endogenous MK4 formation in the kidney, but not other organs. These observations are consistent with our hypothesis that cholesterol metabolism is involved in the generation of MK4. Further research is needed to understand potential regulatory mechanisms and the unique functions of MK4 in the kidney.
Collapse
Affiliation(s)
| | - M Kyla Shea
- Jean Mayer USDA Human Nutrition Research Center on Aging
| | - Xueyan Fu
- Jean Mayer USDA Human Nutrition Research Center on Aging
| | - Michael A Grusak
- USDA/ARS Red River Valley Agricultural Research Center, Fargo, ND
| | - Donald Smith
- Jean Mayer USDA Human Nutrition Research Center on Aging
| | | | - Athan Kuliopulos
- Sackler School of Biomedical Sciences, Tufts University, Boston, MA
| | | | - Sarah L Booth
- Jean Mayer USDA Human Nutrition Research Center on Aging,Address correspondence to SLB (e-mail: )
| |
Collapse
|
47
|
Statins affect human glioblastoma and other cancers through TGF-β inhibition. Oncotarget 2019; 10:1716-1728. [PMID: 30899443 PMCID: PMC6422202 DOI: 10.18632/oncotarget.26733] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 02/09/2019] [Indexed: 12/17/2022] Open
Abstract
The cholesterol-lowering statins have known anti-cancer effects, but the mechanisms and how to utilize statins in oncology have been unclear. We noted in the CellMiner database that statin activity against cancer lines correlated with higher expression of TGF-β target genes such as SERPINE1 and ZYX. This prompted us to assess whether statins affected TGF-β activity in glioblastoma (GBM), a cancer strongly influenced by TGF-β and in dire need of new therapeutic approaches. We noted that statins reduced TGF-β activity, cell viability and invasiveness, Rho/ROCK activity, phosphorylation and activity of the TGF-β mediator Smad3, and expression of TGF-β targets ZYX and SERPINE1 in GBM and GBM-initiating cell (GIC) lines. Statins were most potent against GBM, GIC, and other cancer cells with high TGF-β activity, and exogenous TGF-β further sensitized mesenchymal GICs to statins. Statin toxicity was rescued by addition of exogenous mevalonolactone or geranylgeranyl pyrophosphate, indicating that the observed effects reflected inhibition of HMG CoA-reductase by the statins. Simvastatin significantly inhibited the growth of subcutaneous GIC grafts and prolonged survival in GIC intracranially grafted mice. These results indicate where the statins might best be applied as adjunct therapies in oncology, against GBM and other cancers with high TGF-β activity, and have implications for other statin roles outside of oncology.
Collapse
|
48
|
Jovin DG, Katlaps KG, Ellis BK, Dharmaraj B. Neuroprotection against stroke and encephalopathy after cardiac surgery. Interv Med Appl Sci 2019; 11:27-37. [PMID: 32148901 PMCID: PMC7044570 DOI: 10.1556/1646.11.2019.01] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Cerebral ischemia in the perioperative period is a major risk factor for stroke, encephalopathy, and cognitive decline after cardiothoracic surgery. After coronary artery bypass grafting, both stroke and encephalopathy can result in poor patient outcomes and increased mortality. Neuroprotection aims to lessen the severity and occurrence of further injury mediated by stroke and encephalopathy and to aid the recovery of conditions already present. Several pharmacological and non-pharmacological methods of neuroprotection have been investigated in experimental studies and in animal models, and, although some have shown effectiveness in protection of the central nervous system, for most, clinical research is lacking or did not show the expected results. This review summarizes the value and need for neuroprotection in the context of cardiothoracic surgery and examines the use and effectiveness of several agents and methods with an emphasis on clinical trials and clinically relevant neuroprotectants.
Collapse
Affiliation(s)
- Daniel G Jovin
- Cardiothoracic Research, Department of Surgery, Hunter Holmes McGuire Veterans Affairs Medical Center, Richmond, VA, USA
| | - Karl G Katlaps
- Cardiothoracic Research, Department of Surgery, Hunter Holmes McGuire Veterans Affairs Medical Center, Richmond, VA, USA
| | - Ben K Ellis
- Cardiothoracic Research, Department of Surgery, Hunter Holmes McGuire Veterans Affairs Medical Center, Richmond, VA, USA
| | - Benita Dharmaraj
- Cardiothoracic Research, Department of Surgery, Hunter Holmes McGuire Veterans Affairs Medical Center, Richmond, VA, USA
| |
Collapse
|
49
|
Bailey DM, Catron MA, Kovtun O, Macdonald RL, Zhang Q, Rosenthal SJ. Single Quantum Dot Tracking Reveals Serotonin Transporter Diffusion Dynamics are Correlated with Cholesterol-Sensitive Threonine 276 Phosphorylation Status in Primary Midbrain Neurons. ACS Chem Neurosci 2018; 9:2534-2541. [PMID: 29787674 DOI: 10.1021/acschemneuro.8b00214] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Serotonin transporter (SERT) terminates serotonin signaling in the brain by enabling rapid clearance of the neurotransmitter. SERT dysfunction has been associated with a variety of psychiatric disorders, including depression, anxiety, and autism. Visualizing SERT behavior at the single molecule level in endogenous systems remains a challenge. In this study, we utilize quantum dot (QD) single particle tracking (SPT) to capture SERT dynamics in primary rat midbrain neurons. Membrane microenvironment, specifically membrane cholesterol, plays a key role in SERT regulation and has been found to affect SERT conformational state. We sought to determine how reduced cholesterol content affects both lateral mobility and phosphorylation of conformationally sensitive threonine 276 (Thr276) in endogenous SERT using two different methods of cholesterol manipulation, statins and methyl-β-cyclodextrin. Both chronic and acute cholesterol depletion increased SERT lateral diffusion, radial displacement along the membrane, mobile fraction, and Thr276 phosphorylation levels. Overall, this work has provided new insights about endogenous neuronal SERT mobility and its associations with membrane cholesterol and SERT phosphorylation status.
Collapse
|
50
|
Exosomes impact survival to radiation exposure in cell line models of nervous system cancer. Oncotarget 2018; 9:36083-36101. [PMID: 30546829 PMCID: PMC6281426 DOI: 10.18632/oncotarget.26300] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 10/21/2018] [Indexed: 12/24/2022] Open
Abstract
Radiation is utilized in the therapy of more than 50% of cancer patients. Unfortunately, many malignancies become resistant to radiation over time. We investigated the hypothesis that one method of a cancer cell's ability to survive radiation occurs through cellular communication via exosomes. Exosomes are cell-derived vesicles containing DNA, RNA, and protein. Three properties were analyzed: 1) exosome function, 2) exosome profile and 3) exosome uptake/blockade. To analyze exosome function, we show radiation-derived exosomes increased proliferation and enabled recipient cancer cells to survive radiation in vitro. Furthermore, radiation-derived exosomes increased tumor burden and decreased survival in an in vivo model. To address the mechanism underlying the alterations by exosomes in recipient cells, we obtained a profile of radiation-derived exosomes that showed expression changes favoring a resistant/proliferative profile. Radiation-derived exosomes contain elevated oncogenic miR-889, oncogenic mRNAs, and proteins of the proteasome pathway, Notch, Jak-STAT, and cell cycle pathways. Radiation-derived exosomes contain decreased levels of tumor-suppressive miR-516, miR-365, and multiple tumor-suppressive mRNAs. Ingenuity pathway analysis revealed the most represented networks included cell cycle, growth/survival. Upregulation of DNM2 correlated with increased exosome uptake. To analyze the property of exosome blockade, heparin and simvastatin were used to inhibit uptake of exosomes in recipient cells resulting in inhibited induction of proliferation and cellular survival. Because these agents have shown some success as cancer therapies, our data suggest their mechanism of action could be limiting exosome communication between cells. The results of our study identify a novel exosome-based mechanism that may underlie a cancer cell's ability to survive radiation.
Collapse
|