1
|
Gallet Q, Bouteloup V, Locatelli M, Habert MO, Chupin M, Campion JY, Michels PE, Delrieu J, Lebouvier T, Balageas AC, Surget A, Belzung C, Arlicot N, Ribeiro MJS, Gissot V, El-Hage W, Camus V, Gohier B, Desmidt T. Cerebral Metabolic Signature of Chronic Benzodiazepine Use in Nondemented Older Adults: An FDG-PET Study in the MEMENTO Cohort. Am J Geriatr Psychiatry 2024; 32:665-677. [PMID: 37973486 DOI: 10.1016/j.jagp.2023.10.002] [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: 03/22/2023] [Revised: 10/09/2023] [Accepted: 10/09/2023] [Indexed: 11/19/2023]
Abstract
OBJECTIVE We sought to examine the association between chronic Benzodiazepine (BZD) use and brain metabolism obtained from 2-deoxy-2-fluoro-D-glucose (FDG) positron emission tomography (PET) in the MEMENTO clinical cohort of nondemented older adults with an isolated memory complaint or mild cognitive impairment at baseline. METHODS Our analysis focused on 3 levels: (1) the global mean brain standardized uptake value (SUVR), (2) the Alzheimer's disease (AD)-specific regions of interest (ROIs), and (3) the ratio of total SUVR on the brain and different anatomical ROIs. Cerebral metabolism was obtained from 2-deoxy-2-fluoro-D-glucose-FDG-PET and compared between chronic BZD users and nonusers using multiple linear regressions adjusted for age, sex, education, APOE ε 4 copy number, cognitive and neuropsychiatric assessments, history of major depressive episodes and antidepressant use. RESULTS We found that the SUVR was significantly higher in chronic BZD users (n = 192) than in nonusers (n = 1,122) in the whole brain (beta = 0.03; p = 0.038) and in the right amygdala (beta = 0.32; p = 0.012). Trends were observed for the half-lives of BZDs (short- and long-acting BZDs) (p = 0.051) and Z-drug hypnotic treatments (p = 0.060) on the SUVR of the right amygdala. We found no significant association in the other ROIs. CONCLUSION Our study is the first to find a greater global metabolism in chronic BZD users and a specific greater metabolism in the right amygdala. Because the acute administration of BZDs tends to reduce brain metabolism, these findings may correspond to a compensatory mechanism while the brain adapts with global metabolism upregulation, with a specific focus on the right amygdala.
Collapse
Affiliation(s)
- Quentin Gallet
- Department of Psychiatry, University Hospital, Angers, France
| | - Vincent Bouteloup
- Centre Inserm U1219 Bordeaux Population Health, CIC1401-EC, Institut de Santé Publique, d'Epidémiologie et de Développement, Université de Bordeaux, CHU de Bordeaux, Pôle Santé Publique, Bordeaux, France
| | - Maxime Locatelli
- CATI, US52-UAR2031, CEA, ICM, Sorbonne Université, CNRS, INSERM, APHP, Ile de France, France; Paris Brain Institute - Institut du Cerveau (ICM), CNRS UMR 7225, INSERM, U 1127, Sorbonne Université F-75013, Paris, France; Sorbonne Université, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, LIB, F-75006, Paris, France
| | - Marie-Odile Habert
- CATI, US52-UAR2031, CEA, ICM, Sorbonne Université, CNRS, INSERM, APHP, Ile de France, France; Sorbonne Université, CNRS, INSERM, Laboratoire d'Imagerie Biomédicale, LIB, F-75006, Paris, France; Service de médecine nucléaire, Hôpital Pitié-Salpêtrière, APHP, Paris 75013, France
| | - Marie Chupin
- CATI, US52-UAR2031, CEA, ICM, Sorbonne Université, CNRS, INSERM, APHP, Ile de France, France; Paris Brain Institute - Institut du Cerveau (ICM), CNRS UMR 7225, INSERM, U 1127, Sorbonne Université F-75013, Paris, France
| | | | | | - Julien Delrieu
- Gérontopôle, Department of Geriatrics, CHU Toulouse, Purpan University Hospital, Toulouse, France; UMR1027, Université de Toulouse, UPS, INSERM, Toulouse, France
| | | | | | | | | | - Nicolas Arlicot
- UMR 1253, iBrain, Université de Tours, INSERM, Tours, France; CIC 1415, Université de Tours, INSERM, Tours, France
| | - Maria-Joao Santiago Ribeiro
- CHU de Tours, Tours, France; UMR 1253, iBrain, Université de Tours, INSERM, Tours, France; CIC 1415, Université de Tours, INSERM, Tours, France
| | - Valérie Gissot
- CHU de Tours, Tours, France; UMR 1253, iBrain, Université de Tours, INSERM, Tours, France
| | - Wissam El-Hage
- CHU de Tours, Tours, France; UMR 1253, iBrain, Université de Tours, INSERM, Tours, France; CIC 1415, Université de Tours, INSERM, Tours, France
| | - Vincent Camus
- CHU de Tours, Tours, France; UMR 1253, iBrain, Université de Tours, INSERM, Tours, France
| | - Bénédicte Gohier
- Department of Psychiatry, University Hospital, Angers, France; Université d'Angers, Université de Nantes, LPPL, SFR CONFLUENCES, F-49000 Angers, France
| | - Thomas Desmidt
- CHU de Tours, Tours, France; UMR 1253, iBrain, Université de Tours, INSERM, Tours, France.
| |
Collapse
|
2
|
Chang Y, Xie X, Liu Y, Liu M, Zhang H. Exploring clinical applications and long-term effectiveness of benzodiazepines: An integrated perspective on mechanisms, imaging, and personalized medicine. Biomed Pharmacother 2024; 173:116329. [PMID: 38401518 DOI: 10.1016/j.biopha.2024.116329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/13/2024] [Accepted: 02/21/2024] [Indexed: 02/26/2024] Open
Abstract
Benzodiazepines have been long-established treatments for various conditions, including anxiety disorders and insomnia. Recent FDA warnings emphasize the risks of misuse and dependence associated with benzodiazepines. This article highlights their benefits and potential drawbacks from various perspectives. It achieves this by explaining how benzodiazepines work in terms of neuroendocrinology, immunomodulation, sleep, anxiety, cognition, and addiction, ultimately improving their clinical effectiveness. Benzodiazepines play a regulatory role in the HPA axis and impact various systems, including neuropeptide Y and cholecystokinin. Benzodiazepines can facilitate sleep-dependent memory consolidation by promoting spindle wave activity, but they can also lead to memory deficits in older individuals due to reduced slow-wave sleep. The cognitive effects of chronic benzodiazepines use remain uncertain; however, no adverse findings have been reported in clinical imaging studies. This article aims to comprehensively review the evidence on benzodiazepines therapy, emphasizing the need for more clinical studies, especially regarding long-term benzodiazepines use.
Collapse
Affiliation(s)
- Yiheng Chang
- Department of Neurology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Xueting Xie
- Department of Neurology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yudan Liu
- Department of Neurology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Meichen Liu
- Department of Neurology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China.
| | - Huimin Zhang
- Department of Neurology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|
3
|
Kim RT, Zhou L, Li Y, Krieger AC, Nordvig AS, Butler T, de Leon MJ, Chiang GC. Impaired sleep is associated with tau deposition on 18F-flortaucipir PET and accelerated cognitive decline, accounting for medications that affect sleep. J Neurol Sci 2024; 458:122927. [PMID: 38341949 PMCID: PMC10947806 DOI: 10.1016/j.jns.2024.122927] [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: 10/23/2023] [Revised: 01/06/2024] [Accepted: 02/06/2024] [Indexed: 02/13/2024]
Abstract
BACKGROUND Impaired sleep is commonly associated with Alzheimer's disease (AD), although the underlying mechanisms remain unclear. Furthermore, the moderating effects of sleep-affecting medications, which have been linked to AD pathology, are incompletely characterized. Using data from the Alzheimer's Disease Neuroimaging Initiative, we investigated whether a medical history of impaired sleep, informant-reported nighttime behaviors, and sleep-affecting medications are associated with beta-amyloid and tau deposition on PET and cognitive change, cross-sectionally and longitudinally. METHODS We included 964 subjects with 18F-florbetapir PET scans. Measures of sleep impairment and medication use were obtained from medical histories and the Neuropsychiatric Inventory Questionnaire. Multivariate models, adjusted for covariates, were used to assess associations among sleep-related features, beta-amyloid and tau, and cognition. Cortical tau deposition, categorized by Braak stage, was assessed using the standardized uptake value peak alignment (SUVP) method on 18F-flortaucipir PET. RESULTS Medical history of sleep impairment was associated with greater baseline tau in the meta-temporal, Braak 1, and Braak 4 regions (p = 0.04, p < 0.001, p = 0.025, respectively). Abnormal nighttime behaviors were also associated with greater baseline tau in the meta-temporal region (p = 0.024), and greater cognitive impairment, cross-sectionally (p = 0.007) and longitudinally (p < 0.001). Impaired sleep was not associated with baseline beta-amyloid (p > 0.05). Short-term use of selective serotonin reuptake inhibitors and benzodiazepines slightly weakened the sleep-tau relationship. CONCLUSIONS Sleep impairment was associated with tauopathy and cognitive decline, which could be linked to increased tau secretion from neuronal hyperactivity. Clinically, our results help identify high-risk individuals who could benefit from sleep-related interventions aimed to delay cognitive decline and AD.
Collapse
Affiliation(s)
- Ryan T Kim
- From the Department of Stem Cell and Regenerative Biology, Harvard University, Bauer-Sherman Fairchild Complex 7 Divinity Avenue, Cambridge, MA 02138, United States of America.
| | - Liangdong Zhou
- From the Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 407 E 61(st) Street, New York, NY 10065, United States of America.
| | - Yi Li
- From the Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 407 E 61(st) Street, New York, NY 10065, United States of America.
| | - Ana C Krieger
- From the Departments of Medicine and Neurology, Division of Sleep Neurology, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 425 E 61st St., 5th Floor, New York, NY 10065, United States of America.
| | - Anna S Nordvig
- From the Department of Neurology, Alzheimer's Disease and Memory Disorders Program, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 428 East 72(nd) Street Suite 500, New York, NY 10021, United States of America.
| | - Tracy Butler
- From the Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 407 E 61(st) Street, New York, NY 10065, United States of America.
| | - Mony J de Leon
- From the Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 407 E 61(st) Street, New York, NY 10065, United States of America.
| | - Gloria C Chiang
- From the Department of Radiology, Brain Health Imaging Institute, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 407 E 61(st) Street, New York, NY 10065, United States of America; From the Department of Radiology, Division of Neuroradiology, Weill Cornell Medicine, NewYork-Presbyterian Hospital, 525 East 68th Street, Starr Pavilion, Box 141, New York, NY 10065, United States of America.
| |
Collapse
|
4
|
Al‐kuraishy HM, Al‐Gareeb AI, Albuhadily AK, Elewa YHA, AL‐Farga A, Aqlan F, Zahran MH, Batiha GE. Sleep disorders cause Parkinson's disease or the reverse is true: Good GABA good night. CNS Neurosci Ther 2024; 30:e14521. [PMID: 38491789 PMCID: PMC10943276 DOI: 10.1111/cns.14521] [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: 07/13/2023] [Revised: 10/03/2023] [Accepted: 10/23/2023] [Indexed: 03/18/2024] Open
Abstract
BACKGROUND Parkinson's disease (PD) is a progressive neurodegenerative brain disease due to degeneration of dopaminergic neurons (DNs) presented with motor and non-motor symptoms. PD symptoms are developed in response to the disturbance of diverse neurotransmitters including γ-aminobutyric acid (GABA). GABA has a neuroprotective effect against PD neuropathology by protecting DNs in the substantia nigra pars compacta (SNpc). It has been shown that the degeneration of GABAergic neurons is linked with the degeneration of DNs and the progression of motor and non-motor PD symptoms. GABA neurotransmission is a necessary pathway for normal sleep patterns, thus deregulation of GABAergic neurotransmission in PD could be the potential cause of sleep disorders in PD. AIM Sleep disorders affect GABA neurotransmission leading to memory and cognitive dysfunction in PD. For example, insomnia and short sleep duration are associated with a reduction of brain GABA levels. Moreover, PD-related disorders including rigidity and nocturia influence sleep patterns leading to fragmented sleep which may also affect PD neuropathology. However, the mechanistic role of GABA in PD neuropathology regarding motor and non-motor symptoms is not fully elucidated. Therefore, this narrative review aims to clarify the mechanistic role of GABA in PD neuropathology mainly in sleep disorders, and how good GABA improves PD. In addition, this review of published articles tries to elucidate how sleep disorders such as insomnia and REM sleep behavior disorder (RBD) affect PD neuropathology and severity. The present review has many limitations including the paucity of prospective studies and most findings are taken from observational and preclinical studies. GABA involvement in the pathogenesis of PD has been recently discussed by recent studies. Therefore, future prospective studies regarding the use of GABA agonists in the management of PD are suggested to observe their distinct effects on motor and non-motor symptoms. CONCLUSION There is a bidirectional relationship between the pathogenesis of PD and sleep disorders which might be due to GABA deregulation.
Collapse
Affiliation(s)
- Hayder M. Al‐kuraishy
- Department of Clinical Pharmacology and Medicine, College of MedicineAl‐Mustansiriya UniversityBaghdadIraq
| | - Ali I. Al‐Gareeb
- Department of Clinical Pharmacology and Medicine, College of MedicineAl‐Mustansiriya UniversityBaghdadIraq
| | - Ali K. Albuhadily
- Department of Clinical Pharmacology and Medicine, College of MedicineAl‐Mustansiriya UniversityBaghdadIraq
| | - Yaser Hosny Ali Elewa
- Department of Histology and Cytology, Faculty of Veterinary MedicineZagazig UniversityZagazigEgypt
- Faculty of Veterinary MedicineHokkaido UniversitySapporoJapan
| | - Ammar AL‐Farga
- Biochemistry Department, College of SciencesUniversity of JeddahJeddahSaudia Arbia
| | - Faisal Aqlan
- Department of Chemistry, College of SciencesIbb UniversityIbb GovernorateYemen
| | | | - Gaber El‐Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary MedicineDamanhur UniversityDamanhurEgypt
| |
Collapse
|
5
|
Canet G, Rocaboy E, Laliberté F, Boscher E, Guisle I, Diego-Diaz S, Fereydouni-Forouzandeh P, Whittington RA, Hébert SS, Pernet V, Planel E. Temperature-induced Artifacts in Tau Phosphorylation: Implications for Reliable Alzheimer's Disease Research. Exp Neurobiol 2023; 32:423-440. [PMID: 38196137 PMCID: PMC10789175 DOI: 10.5607/en23025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 12/10/2023] [Accepted: 12/19/2023] [Indexed: 01/11/2024] Open
Abstract
In preclinical research on Alzheimer's disease and related tauopathies, tau phosphorylation analysis is routinely employed in both cellular and animal models. However, recognizing the sensitivity of tau phosphorylation to various extrinsic factors, notably temperature, is vital for experimental accuracy. Hypothermia can trigger tau hyperphosphorylation, while hyperthermia leads to its dephosphorylation. Nevertheless, the rapidity of tau phosphorylation in response to unintentional temperature variations remains unknown. In cell cultures, the most significant temperature change occurs when the cells are removed from the incubator before harvesting, and in animal models, during anesthesia prior to euthanasia. In this study, we investigate the kinetics of tau phosphorylation in N2a and SH-SY5Y neuronal cell lines, as well as in mice exposed to anesthesia. We observed changes in tau phosphorylation within the few seconds upon transferring cell cultures from their 37°C incubator to room temperature conditions. However, cells placed directly on ice post-incubation exhibited negligible phosphorylation changes. In vivo, isoflurane anesthesia rapidly resulted in tau hyperphosphorylation within the few seconds needed to lose the pedal withdrawal reflex in mice. These findings emphasize the critical importance of preventing temperature variation in researches focused on tau. To ensure accurate results, we recommend avoiding anesthesia before euthanasia and promptly placing cells on ice after removal from the incubator. By controlling temperature fluctuations, the reliability and validity of tau phosphorylation studies can be significantly enhanced.
Collapse
Affiliation(s)
- Geoffrey Canet
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Laval University, Québec G1V 4G2, Canada
- Neurosciences Axis, Research Center of the CHU de Québec - Laval University, Québec G1V 4G2, Canada
| | - Emma Rocaboy
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Laval University, Québec G1V 4G2, Canada
| | - Francis Laliberté
- Neurosciences Axis, Research Center of the CHU de Québec - Laval University, Québec G1V 4G2, Canada
| | - Emmanuelle Boscher
- Neurosciences Axis, Research Center of the CHU de Québec - Laval University, Québec G1V 4G2, Canada
| | - Isabelle Guisle
- Neurosciences Axis, Research Center of the CHU de Québec - Laval University, Québec G1V 4G2, Canada
| | - Sofia Diego-Diaz
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Laval University, Québec G1V 4G2, Canada
| | | | - Robert A. Whittington
- Department of Anesthesiology and Perioperative Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Sébastien S. Hébert
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Laval University, Québec G1V 4G2, Canada
- Neurosciences Axis, Research Center of the CHU de Québec - Laval University, Québec G1V 4G2, Canada
| | - Vincent Pernet
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Laval University, Québec G1V 4G2, Canada
- Department of Neurology, Bern University Hospital, Bern 3010, Switzerland
| | - Emmanuel Planel
- Department of Psychiatry and Neurosciences, Faculty of Medicine, Laval University, Québec G1V 4G2, Canada
- Neurosciences Axis, Research Center of the CHU de Québec - Laval University, Québec G1V 4G2, Canada
| |
Collapse
|
6
|
Kaplan K, Hunsberger HC. Benzodiazepine-induced anterograde amnesia: detrimental side effect to novel study tool. Front Pharmacol 2023; 14:1257030. [PMID: 37781704 PMCID: PMC10536168 DOI: 10.3389/fphar.2023.1257030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Accepted: 09/04/2023] [Indexed: 10/03/2023] Open
Abstract
Benzodiazepines (BZDs) are anxiolytic drugs that act on GABAa receptors and are used to treat anxiety disorders. However, these drugs come with the detrimental side effect of anterograde amnesia, or the inability to form new memories. In this review we discuss, behavioral paradigms, sex differences and hormonal influences affecting BZD-induced amnesia, molecular manipulations, including the knockout of GABAa receptor subunits, and regional studies utilizing lesion and microinjection techniques targeted to the hippocampus and amygdala. Additionally, the relationship between BZD use and cognitive decline related to Alzheimer's disease is addressed, as there is a lack of consensus on whether these drugs are involved in inducing or accelerating pathological cognitive deficits. This review aims to inspire new research directions, as there is a gap in knowledge in understanding the cellular and molecular mechanisms behind BZD-induced amnesia. Understanding these mechanisms will allow for the development of alternative treatments and potentially allow BZDs to be used as a novel tool to study Alzheimer's disease.
Collapse
Affiliation(s)
- Kameron Kaplan
- Center for Neurodegenerative Diseases and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science, The Chicago Medical School, North Chicago, IL, United States
| | - Holly Christian Hunsberger
- Center for Neurodegenerative Diseases and Therapeutics, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
- Department of Neuroscience, Rosalind Franklin University of Medicine and Science, The Chicago Medical School, North Chicago, IL, United States
| |
Collapse
|
7
|
Al-Kuraishy HM, Al-Gareeb AI, Alsayegh AA, Abusudah WF, Almohmadi NH, Eldahshan OA, Ahmed EA, Batiha GES. Insights on benzodiazepines' potential in Alzheimer's disease. Life Sci 2023; 320:121532. [PMID: 36858314 DOI: 10.1016/j.lfs.2023.121532] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 02/16/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023]
Abstract
Alzheimer's disease (AD) is the most frequent type of dementia characterized by the deposition of amyloid beta (Aβ) plaque and tau-neurofibrillary tangles (TNTs) in the brain. AD is associated with the disturbances of various neurotransmitters including gamma-aminobutyric acid (GABA). Of note, GABA is reduced in AD, and restoration of GABA effect by benzodiazepines (BDZs) may improve AD outcomes. However, BDZs may adversely affect cognitive functions chiefly in elderly AD patients with sleep disorders. Besides, there is a controversy regarding the use of BDZs in AD. Consequently, the objective of the present review was to disclose the possible role of BDZs on the pathogenesis of AD that might be beneficial, neutral, or detrimental effects on AD. Prolonged use of intermediate-acting BDZ lorazepam exerts amnesic effects due to attenuation of synaptic plasticity and impairment of recognition memory. However, BDZs may have a protective effect against the development of AD by reducing tau phosphorylation, neuroinflammation, and progression of AD neuropathology. On the other side, other findings highlighted that extended use of BDZs was not associated with the development of AD. In conclusion, there are controversial points concerning the use of BDZs and the risk for the progression of AD. Thus, preclinical, and clinical studies are essential in this regard.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Bagdad, Iraq
| | - Ali I Al-Gareeb
- Department of Clinical Pharmacology and Medicine, College of Medicine, Al-Mustansiriya University, Bagdad, Iraq
| | - Abdulrahman A Alsayegh
- Clinical Nutrition Department Applied Medical Sciences, College Jazan University, Jazan 82817, Saudi Arabia.
| | - Wafaa Fouzi Abusudah
- Clinical Nutrition Department, College of Applied Medical Sciences, Umm Al-Qura University, Makkah 24381, Saudi Arabia.
| | - Najlaa Hamed Almohmadi
- Clinical Nutrition Department, College of Applied Medical Sciences, Umm Al-Qura University, Makkah 24381, Saudi Arabia.
| | - Omayma A Eldahshan
- Department of Pharmacognosy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| | - Eman A Ahmed
- Department of Pharmacology, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Al Beheira, Egypt.
| |
Collapse
|
8
|
Al-Kuraishy HM, Al-Gareeb AI, Saad HM, Batiha GES. Benzodiazepines in Alzheimer's disease: beneficial or detrimental effects. Inflammopharmacology 2023; 31:221-230. [PMID: 36418599 DOI: 10.1007/s10787-022-01099-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 11/04/2022] [Indexed: 11/26/2022]
Abstract
Dementia is considered a clinical syndrome characterized by cognitive dysfunction and memory loss. Alzheimer's disease (AD) is the most common type of dementia. AD is linked with the turbulence of diverse neurotransmitters including gamma-aminobutyric acid (GABA). Notably, GABA in the brain and cerebrospinal fluid was reduced in AD. Thus, allosteric modulation of the GABA effect by benzodiazepines (BDZs) may improve the clinical outcomes of AD patients. Therefore, the objective of the present review was to reveal the possible role of BDZs on the pathogenesis and clinical outcomes in AD patients. Though BDZs may adversely affect cognitive functions mainly in elderly patients, herein it was postulated that BDZs may have beneficial, neutral, or detrimental effects in AD. Taken together, there is strong controversy regarding the use of BDZs and the risk for the development of AD. Therefore, experimental, preclinical and clinical studies are critical to determine the potential protective or detrimental effects of BDZs on AD neuropathology.
Collapse
Affiliation(s)
- Hayder M Al-Kuraishy
- Professor in Department of Pharmacology, Toxicology and Medicine, Medical Faculty, College of Medicine, Al-Mustansiriyah University, P.O. Box 14132, Baghdad, Iraq
| | - Ali I Al-Gareeb
- Professor in Department of Pharmacology, Toxicology and Medicine, Medical Faculty, College of Medicine, Al-Mustansiriyah University, P.O. Box 14132, Baghdad, Iraq
| | - Hebatallah M Saad
- Department of Pathology, Faculty of Veterinary Medicine, Matrouh University, Marsa Matruh, 51744, Egypt.
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, 22511, Egypt.
| |
Collapse
|
9
|
Chen Z, Wang S, Meng Z, Ye Y, Shan G, Wang X, Zhao X, Jin Y. Tau protein plays a role in the mechanism of cognitive disorders induced by anesthetic drugs. Front Neurosci 2023; 17:1145318. [PMID: 36937655 PMCID: PMC10015606 DOI: 10.3389/fnins.2023.1145318] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 02/13/2023] [Indexed: 03/08/2023] Open
Abstract
Cognitive disorders are mental health disorders that can affect cognitive ability. Surgery and anesthesia have been proposed to increase the incidence of cognitive dysfunction, including declines in memory, learning, attention and executive function. Tau protein is a microtubule-associated protein located in the axons of neurons and is important for microtubule assembly and stability; its biological function is mainly regulated by phosphorylation. Phosphorylated tau protein has been associated with cognitive dysfunction mediated by disrupting the stability of the microtubule structure. There is an increasing consensus that anesthetic drugs can cause cognitive impairment. Herein, we reviewed the latest literature and compared the relationship between tau protein and cognitive impairment caused by different anesthetics. Our results substantiated that tau protein phosphorylation is essential in cognitive dysfunction caused by anesthetic drugs, and the possible mechanism can be summarized as "anesthetic drugs-kinase/phosphatase-p-Tau-cognitive impairment".
Collapse
|
10
|
Liu X, Guo L, Duan B, Wu J, Wang E. Novel benzodiazepine remimazolam tosylate delays neurodegeneration of aged mice via decreasing tau phosphorylation. Neurotoxicology 2022; 92:156-165. [DOI: 10.1016/j.neuro.2022.08.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 08/03/2022] [Accepted: 08/04/2022] [Indexed: 10/15/2022]
|
11
|
Zarhin D, Atsmon R, Ruggiero A, Baeloha H, Shoob S, Scharf O, Heim LR, Buchbinder N, Shinikamin O, Shapira I, Styr B, Braun G, Harel M, Sheinin A, Geva N, Sela Y, Saito T, Saido T, Geiger T, Nir Y, Ziv Y, Slutsky I. Disrupted neural correlates of anesthesia and sleep reveal early circuit dysfunctions in Alzheimer models. Cell Rep 2022; 38:110268. [PMID: 35045289 PMCID: PMC8789564 DOI: 10.1016/j.celrep.2021.110268] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/06/2021] [Accepted: 12/22/2021] [Indexed: 11/30/2022] Open
Abstract
Dysregulated homeostasis of neural activity has been hypothesized to drive Alzheimer's disease (AD) pathogenesis. AD begins with a decades-long presymptomatic phase, but whether homeostatic mechanisms already begin failing during this silent phase is unknown. We show that before the onset of memory decline and sleep disturbances, familial AD (fAD) model mice display no deficits in CA1 mean firing rate (MFR) during active wakefulness. However, homeostatic down-regulation of CA1 MFR is disrupted during non-rapid eye movement (NREM) sleep and general anesthesia in fAD mouse models. The resultant hyperexcitability is attenuated by the mitochondrial dihydroorotate dehydrogenase (DHODH) enzyme inhibitor, which tunes MFR toward lower set-point values. Ex vivo fAD mutations impair downward MFR homeostasis, resulting in pathological MFR set points in response to anesthetic drug and inhibition blockade. Thus, firing rate dyshomeostasis of hippocampal circuits is masked during active wakefulness but surfaces during low-arousal brain states, representing an early failure of the silent disease stage.
Collapse
Affiliation(s)
- Daniel Zarhin
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Refaela Atsmon
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Antonella Ruggiero
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Halit Baeloha
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Shiri Shoob
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Oded Scharf
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Leore R Heim
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Nadav Buchbinder
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ortal Shinikamin
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Ilana Shapira
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Boaz Styr
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Gabriella Braun
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Michal Harel
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Anton Sheinin
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Nitzan Geva
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yaniv Sela
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama 351-0198, Japan; Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Takaomi Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Saitama 351-0198, Japan
| | - Tamar Geiger
- Department of Human Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yuval Nir
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Yaniv Ziv
- Department of Neurobiology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Inna Slutsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 69978, Israel; Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel.
| |
Collapse
|
12
|
Khan AF, Adewale Q, Baumeister TR, Carbonell F, Zilles K, Palomero-Gallagher N, Iturria-Medina Y. Personalized brain models identify neurotransmitter receptor changes in Alzheimer's disease. Brain 2021; 145:1785-1804. [PMID: 34605898 DOI: 10.1093/brain/awab375] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 11/12/2022] Open
Abstract
Alzheimer's disease (AD) involves many neurobiological alterations from molecular to macroscopic spatial scales, but we currently lack integrative, mechanistic brain models characterizing how factors across different biological scales interact to cause clinical deterioration in a way that is subject-specific or personalized. Neurotransmitter receptors, as important signaling molecules and potential drug targets, are key mediators of interactions between many neurobiological processes altered in AD. We present a neurotransmitter receptor-enriched multifactorial brain model, which integrates spatial distribution patterns of 15 neurotransmitter receptors from post-mortem autoradiography with multiple in-vivo neuroimaging modalities (tau, amyloid-β and glucose PET, and structural, functional and arterial spin labeling MRI) in a personalized, generative, whole-brain formulation. Applying this data-driven model to a heterogeneous aged population (N = 423, ADNI data), we observed that personalized receptor-neuroimaging interactions explained about 70% (± 20%) of the across-population variance in longitudinal changes to the six neuroimaging modalities, and up to 39.7% (P < 0.003, FWE-corrected) of inter-individual variability in AD cognitive deterioration via an axis primarily affecting executive function. Notably, based on their contribution to the clinical severity in AD, we found significant functional alterations to glutamatergic interactions affecting tau accumulation and neural activity dysfunction, and GABAergic interactions concurrently affecting neural activity dysfunction, amyloid and tau distributions, as well as significant cholinergic receptor effects on tau accumulation. Overall, GABAergic alterations had the largest effect on cognitive impairment (particularly executive function) in our AD cohort (N = 25). Furthermore, we demonstrate the clinical applicability of this approach by characterizing subjects based on individualized 'fingerprints' of receptor alterations. This study introduces the first robust, data-driven framework for integrating several neurotransmitter receptors, multi-modal neuroimaging and clinical data in a flexible and interpretable brain model. It enables further understanding of the mechanistic neuropathological basis of neurodegenerative progression and heterogeneity, and constitutes a promising step towards implementing personalized, neurotransmitter-based treatments.
Collapse
Affiliation(s)
- Ahmed Faraz Khan
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Canada H3A 2B4.,McConnell Brain Imaging Center, Montreal Neurological Institute, Montreal, Canada H3A 2B4.,Ludmer Centre for Neuroinformatics & Mental Health, Montreal, Canada H3A 2B4
| | - Quadri Adewale
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Canada H3A 2B4.,McConnell Brain Imaging Center, Montreal Neurological Institute, Montreal, Canada H3A 2B4.,Ludmer Centre for Neuroinformatics & Mental Health, Montreal, Canada H3A 2B4
| | - Tobias R Baumeister
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Canada H3A 2B4.,McConnell Brain Imaging Center, Montreal Neurological Institute, Montreal, Canada H3A 2B4.,Ludmer Centre for Neuroinformatics & Mental Health, Montreal, Canada H3A 2B4
| | | | - Karl Zilles
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, 52425 Jülich, Germany
| | - Nicola Palomero-Gallagher
- Institute of Neuroscience and Medicine (INM-1), Research Centre Jülich, 52425 Jülich, Germany.,Cécile and Oskar Vogt Institute of Brain Research, Medical Faculty, Heinrich-Heine University, 40225 Düsseldorf, Germany.,Department of Psychiatry, Psychotherapy, and Psychosomatics, Medical Faculty, RWTH Aachen, 52074 Aachen, Germany.,JARA, Translational Brain Medicine, 52074 Aachen, Germany
| | - Yasser Iturria-Medina
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Canada H3A 2B4.,McConnell Brain Imaging Center, Montreal Neurological Institute, Montreal, Canada H3A 2B4.,Ludmer Centre for Neuroinformatics & Mental Health, Montreal, Canada H3A 2B4
| | | |
Collapse
|
13
|
Ferreira P, Ferreira AR, Barreto B, Fernandes L. Is there a link between the use of benzodiazepines and related drugs and dementia? A systematic review of reviews. Eur Geriatr Med 2021; 13:19-32. [PMID: 34403113 DOI: 10.1007/s41999-021-00553-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/06/2021] [Indexed: 01/08/2023]
Abstract
PURPOSE Benzodiazepines (BZDs) and related drugs (BZRDs) are commonly used to treat diverse psychiatric disorders due to their anxiolytic, hypnotic and sedative properties, despite their known associated side effects, including acute consequences on cognition. Recently, some studies have also suggested that long-term cognitive effects may coexist, as the increased risk of cognitive decline and dementia. This review aims to appraise and summarise published synthesis studies on the risk of dementia development due to BZDs/BZRDs use. METHODS A comprehensive systematic search was carried out in PubMed, Web of Science, Cochrane Library and Epistemonikos databases. Grey literature and hand search of the studies' reference lists were undertaken. Meta-analysis, systematic and non-systematic reviews were included. Neither language nor date restrictions were applied. Search results other than synthesis studies were excluded. The methodological quality of the included reviews was analysed with AMSTAR-2 and SANRA tools. RESULTS Overall, 877 records were initially retrieved and 15 complied with the inclusion criteria. From these, five were systematic reviews with meta-analysis, two were systematic reviews and eight were non-systematic reviews. Most of the primary studies included in the analysed reviews found an association between BZDs/BZRDs use and subsequent dementia, with meta-analysis studies reporting an increased risk for users (ORs ranging from 1.38 to 1.78). However, the considerable clinical and methodological heterogeneity of the primary studies makes it difficult to establish a causal relationship. CONCLUSION Although hampered by the heterogeneity between the studies, the present findings suggest an association between BZDs/BZRDs use and increased risk of dementia in older adults.
Collapse
Affiliation(s)
| | - Ana Rita Ferreira
- Faculty of Medicine, University of Porto, Porto, Portugal.,Faculty of Medicine, CINTESIS-Center for Health Technology and Services Research, University of Porto, Porto, Portugal
| | | | - Lia Fernandes
- Faculty of Medicine, University of Porto, Porto, Portugal.,Faculty of Medicine, CINTESIS-Center for Health Technology and Services Research, University of Porto, Porto, Portugal.,Psychiatry Service, Centro Hospitalar Universitário de São João, Porto, Portugal
| |
Collapse
|
14
|
HIV Infection and Related Mental Disorders. Brain Sci 2021; 11:brainsci11020248. [PMID: 33671125 PMCID: PMC7922767 DOI: 10.3390/brainsci11020248] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 01/13/2023] Open
Abstract
Over the more than thirty-year period of the human immunodeficiency virus type 1 (HIV-1) epidemic, many data have been accumulated indicating that HIV infection predisposes one to the development of mental pathologies. It has been proven that cognitive disorders in HIV-positive individuals are the result of the direct exposure of the virus to central nervous system (CNS) cells. The use of antiretroviral therapy has significantly reduced the number of cases of mental disorders among people infected with HIV. However, the incidence of moderate to mild cognitive impairment at all stages of HIV infection is still quite high. This review describes the most common forms of mental pathology that occur in people living with HIV and presents the current concepts on the possible pathogenetic mechanisms of the influence of human immunodeficiency virus (HIV-1) and its viral proteins on the cells of the CNS and the CNS’s functions. This review also provides the current state of knowledge on the impact of the antiretroviral therapy on the development of mental pathologies in people living with HIV, as well as current knowledge on the interactions between antiretroviral and psychotropic drugs that occur under their simultaneous administration.
Collapse
|
15
|
Comparison of intravenous sedation using midazolam during dental treatment in elderly patients with/without dementia: a prospective, controlled clinical trial. Sci Rep 2021; 11:3617. [PMID: 33574437 PMCID: PMC7878763 DOI: 10.1038/s41598-021-83122-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 01/19/2021] [Indexed: 12/18/2022] Open
Abstract
The effects of intravenous sedation with midazolam on the cerebral function of elderly patients with severe dementia are unclear. This study aimed to evaluate its effects on parameters such as brainwaves and cerebral blood flow (CBF) and compare them between elderly individuals with dementia and without cognitive impairment. Ten patients with severe dementia and 10 without cognitive impairment were registered. The bispectral index (BIS) and normalized tissue hemoglobin index (nTHI), which reflects CBF using near-infrared spectroscopy, were measured. Midazolam was administered until a Modified Observer’s Assessment of Alertness/Sedation score of 2 was reached. The chi-squared, Mann–Whitney U, Wilcoxon signed-rank, and Friedman tests and multiple regression analysis were used for comparisons. Whereas a similar decline in BIS values was observed in both groups after midazolam administration (P < 0.018), there was a significant decrease by 9% in the nTHI of the dementia-positive group (P < 0.013). However, there was no significant difference in the nTHI between the dementia-positive and dementia-negative group according to the multiple regression analysis (P = 0.058). In the dementia-negative group, none of the measured values differed from the baseline values. In the dementia-positive group, sedation with midazolam resulted in a 9% decrease in the CBF.
Collapse
|
16
|
Gurunathan U, Rahman T, Williams Z, Vandeleur A, Sriram S, Harch J, Boggett S, Hill C, Bowyer A, Royse C. Effect of Midazolam in Addition to Propofol and Opiate Sedation on the Quality of Recovery After Colonoscopy: A Randomized Clinical Trial. Anesth Analg 2020; 131:741-750. [PMID: 31922999 DOI: 10.1213/ane.0000000000004620] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND There is a concern that midazolam, when used as a component of sedation for colonoscopy, may impair cognition and prolong recovery. We aimed to identify whether midazolam produced short- and longer-term effects on multiple dimensions of recovery including cognition. METHODS A 2-center double-blinded, placebo-controlled, parallel-group, randomized, phase IV study with a 1:1 allocation ratio was conducted in adults ≥18 years of age undergoing elective outpatient colonoscopy, with sufficient English language proficiency to complete the Postoperative Quality of Recovery Scale (PostopQRS). Participants were administered either midazolam (0.04 mg·kg) or an equivalent volume of 0.9% saline before sedation with propofol with or without an opiate. The primary outcome was incidence of recovery in the cognitive domain of the PostopQRS on day 3 after colonoscopy, which was analyzed using a χ test. Secondary outcomes included recovery in other domains of the PostopQRS over time, time to eye-opening, and hospital stay, and patient and endoscopist satisfaction. All hypotheses were defined before recruitment. RESULTS During September 2015 to June 2018, 406 patients were allocated to either midazolam (n = 201) or placebo (n = 205), with one withdrawn before allocation. There was no significant difference in recovery in the cognitive domain of the PostopQRS on day 3 after colonoscopy (midazolam 86.8% vs placebo 88.7%, odds ratio, 0.838; 95% confidence interval [CI], 0.42-1.683; P= .625). Furthermore, there was no difference in recovery over time in the cognitive domain of the PostopQRS (P = .534). Overall recovery of the PostopQRS increased over time but was not different between groups. Furthermore, there were no differences between groups for nociceptive, emotive, activities-of-daily-living domains of the PostopQRS. Patient and endoscopist satisfaction were high and not different. There were no differences in time to eye-opening (midazolam 9.4 ± 12.8 minutes vs placebo 7.3 ± 0.7 minutes; P = .055), or time to hospital discharge (midazolam 103.4 ± 1.4 minutes vs placebo 98.4 ± 37.0 minutes; P = .516). CONCLUSIONS The addition of midazolam 0.04 mg·kg as adjunct to propofol and opiate sedation for elective colonoscopy did not show evidence of any significant differences in recovery in the cognitive domain of the PostopQRS, overall quality of recovery as measured by the PostopQRS, or emergence and hospital discharge times. The use of midazolam should be determined by the anesthesiologist.
Collapse
Affiliation(s)
- Usha Gurunathan
- From the The Prince Charles Hospital, Brisbane, Queensland, Australia.,University of Queensland, Brisbane, Queensland, Australia
| | - Tony Rahman
- From the The Prince Charles Hospital, Brisbane, Queensland, Australia.,Queensland University of Technology and University of Sunshine Coast, Queensland, Australia
| | - Zelda Williams
- Department of Surgery, The University of Melbourne, Melbourne, Victoria, Australia
| | - Ann Vandeleur
- From the The Prince Charles Hospital, Brisbane, Queensland, Australia
| | - Sweta Sriram
- Department of Surgery, The University of Melbourne, Melbourne, Victoria, Australia
| | - Jennifer Harch
- From the The Prince Charles Hospital, Brisbane, Queensland, Australia
| | - Stuart Boggett
- Department of Surgery, The University of Melbourne, Melbourne, Victoria, Australia
| | - Cindy Hill
- From the The Prince Charles Hospital, Brisbane, Queensland, Australia
| | - Andrea Bowyer
- Department of Anesthesia and Pain Management, The Royal Melbourne Hospital, Melbourne, Victoria, Australia
| | - Colin Royse
- Department of Surgery, The University of Melbourne, Melbourne, Victoria, Australia.,Outcomes Research Consortium, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
17
|
Liu M, Dexheimer T, Sui D, Hovde S, Deng X, Kwok R, Bochar DA, Kuo MH. Hyperphosphorylated tau aggregation and cytotoxicity modulators screen identified prescription drugs linked to Alzheimer's disease and cognitive functions. Sci Rep 2020; 10:16551. [PMID: 33024171 PMCID: PMC7539012 DOI: 10.1038/s41598-020-73680-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 09/11/2020] [Indexed: 12/11/2022] Open
Abstract
The neurodegenerative Alzheimer's disease (AD) affects more than 30 million people worldwide. There is thus far no cure or prevention for AD. Aggregation of hyperphosphorylated tau in the brain correlates with the cognitive decline of patients of AD and other neurodegenerative tauopathies. Intracerebral injection of tau aggregates isolated from tauopathy brains causes similar pathology in the recipient mice, demonstrating the pathogenic role of abnormally phosphorylated tau. Compounds controlling the aggregation of hyperphosphorylated tau therefore are probable modulators for the disease. Here we report the use of recombinant hyperphosphorylated tau (p-tau) to identify potential tauopathy therapeutics and risk factors. Hyperphosphorylation renders tau prone to aggregate and to impair cell viability. Taking advantage of these two characters of p-tau, we performed a screen of a 1280-compound library, and tested a selective group of prescription drugs in p-tau aggregation and cytotoxicity assays. R-(-)-apomorphine and raloxifene were found to be p-tau aggregation inhibitors that protected p-tau-treated cells. In contrast, a subset of benzodiazepines exacerbated p-tau cytotoxicity apparently via enhancing p-tau aggregation. R-(-)apomorphine and raloxifene have been shown to improve cognition in animals or in humans, whereas benzodiazepines were linked to increased risks of dementia. Our results demonstrate the feasibility and potential of using hyperphosphorylated tau-based assays for AD drug discovery and risk factor identification.
Collapse
Affiliation(s)
- Mengyu Liu
- Department of Biochemistry and Molecular Biology, Michigan State University, 603 Wilson Road, Room 401, Biochemistry Building, East Lansing, MI, 48824, USA
| | - Thomas Dexheimer
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Dexin Sui
- Department of Biochemistry and Molecular Biology, Michigan State University, 603 Wilson Road, Room 401, Biochemistry Building, East Lansing, MI, 48824, USA
| | - Stacy Hovde
- Department of Biochemistry and Molecular Biology, Michigan State University, 603 Wilson Road, Room 401, Biochemistry Building, East Lansing, MI, 48824, USA
| | - Xiexiong Deng
- Department of Biochemistry and Molecular Biology, Michigan State University, 603 Wilson Road, Room 401, Biochemistry Building, East Lansing, MI, 48824, USA
- Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, 48109-1085, USA
| | - Roland Kwok
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI, USA
| | | | - Min-Hao Kuo
- Department of Biochemistry and Molecular Biology, Michigan State University, 603 Wilson Road, Room 401, Biochemistry Building, East Lansing, MI, 48824, USA.
| |
Collapse
|
18
|
Ettcheto M, Olloquequi J, Sánchez-López E, Busquets O, Cano A, Manzine PR, Beas-Zarate C, Castro-Torres RD, García ML, Bulló M, Auladell C, Folch J, Camins A. Benzodiazepines and Related Drugs as a Risk Factor in Alzheimer's Disease Dementia. Front Aging Neurosci 2020; 11:344. [PMID: 31969812 PMCID: PMC6960222 DOI: 10.3389/fnagi.2019.00344] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 11/26/2019] [Indexed: 12/21/2022] Open
Abstract
Benzodiazepines (BZDs) and Z-drugs are compounds widely prescribed in medical practice due to their anxiolytic, hypnotic, and muscle relaxant properties. Yet, their chronic use is associated with cases of abuse, dependence, and relapse in many patients. Furthermore, elderly people are susceptible to alterations in pharmacodynamics, pharmacokinetics as well as to drug interaction due to polypharmacy. These situations increase the risk for the appearance of cognitive affectations and the development of pathologies like Alzheimer's disease (AD). In the present work, there is a summary of some clinical studies that have evaluated the effect of BZDs and Z-drugs in the adult population with and without AD, focusing on the relationship between their use and the loss of cognitive function. Additionally, there is an assessment of preclinical studies focused on finding molecular proof on the pathways by which these drugs could be involved in AD pathogenesis. Moreover, available data (1990-2019) on BZD and Z-drug use among elderly patients, with and without AD, was compiled in this work. Finally, the relationship between the use of BZD and Z-drugs for the treatment of insomnia and the appearance of AD biomarkers was analyzed. Results pointed to a vicious circle that would worsen the condition of patients over time. Likewise, it put into relevance the need for close monitoring of those patients using BZDs that also suffer from AD. Consequently, future studies should focus on optimizing strategies for insomnia treatment in the elderly by using other substances like melatonin agonists, which is described to have a much more significant safety profile.
Collapse
Affiliation(s)
- Miren Ettcheto
- Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain
- Departament de Bioquímica i Biotecnologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Jordi Olloquequi
- Laboratory of Cellular and Molecular Pathology, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca, Chile
| | - Elena Sánchez-López
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Unitat de Farmàcia, Tecnologia Farmacèutica i Fisico-química, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), Universitat de Barcelona, Barcelona, Spain
| | - Oriol Busquets
- Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain
- Departament de Bioquímica i Biotecnologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Amanda Cano
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Unitat de Farmàcia, Tecnologia Farmacèutica i Fisico-química, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), Universitat de Barcelona, Barcelona, Spain
| | | | - Carlos Beas-Zarate
- Laboratorio de Regeneración y Desarrollo Neural, Departamento de Biología Celular y Molecular, Instituto de Neurobiología, CUCBA, Guadalajara, Mexico
| | - Rubén D. Castro-Torres
- Laboratorio de Regeneración y Desarrollo Neural, Departamento de Biología Celular y Molecular, Instituto de Neurobiología, CUCBA, Guadalajara, Mexico
| | - Maria Luisa García
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Unitat de Farmàcia, Tecnologia Farmacèutica i Fisico-química, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), Universitat de Barcelona, Barcelona, Spain
| | - Mónica Bulló
- Departament de Bioquímica i Biotecnologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
- Institut d'Investigació Sanitària Pere Virgili (IISPV), Reus, Spain
- Centro de Investigación Biomédica en Red Fisiopatologia de la Obesidad y la Nutrición (CIBEROBN), Institut de Salud Carlos III, Madrid, Spain
| | - Carme Auladell
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Departament de Biologia Cellular, Fisiologia i Immunologia, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain
| | - Jaume Folch
- Departament de Bioquímica i Biotecnologia, Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Reus, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Antonio Camins
- Departament de Farmacologia, Toxicologia i Química Terapèutica, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Barcelona, Spain
- Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
- Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
- Laboratory of Cellular and Molecular Pathology, Facultad de Ciencias de la Salud, Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Talca, Chile
| |
Collapse
|
19
|
Cil G, Park J, Bergen AW. Self-Reported Prescription Drug Use for Pain and for Sleep and Incident Frailty. J Am Geriatr Soc 2019; 67:2474-2481. [PMID: 31648384 DOI: 10.1111/jgs.16214] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 04/24/2019] [Accepted: 04/28/2019] [Indexed: 11/26/2022]
Abstract
OBJECTIVES We aimed to estimate incident frailty risks of prescription drugs for pain and for sleep in older US adults. DESIGN Longitudinal cohort. SETTING Health and Retirement Study. PARTICIPANTS Community-living respondents aged 65 years and older, excluding individuals who received recent treatment for cancer (N = 14 208). Our longitudinal analysis sample included respondents who were not frail at baseline and had at least one follow-up wave with complete information on both prescription drug use and frailty, or date of death (N = 7201). MEASUREMENTS Prescription drug use for pain and sleep, sociodemographics, other drug and substance use, and Burden frailty model components. Multivariable drug use stratified hazard models with death as a competing risk evaluated frailty risks associated with co-use and single use of prescription drugs for pain and for sleep. RESULTS Proportions endorsing prescription drug use were 22.1% for pain only, 6.8% for sleep only, and 7.7% for both indications. Burden frailty model prevalence was 41.0% and varied significantly by drug use. Among non-frail individuals at baseline, proportions endorsing prescription drug use were 14.9%, 5.6%, and 2.2% for the three indications. Prescription drug use was associated with increased risk of frailty (co-use adjusted subhazard ratio [sHR] = 1.95; 95% confidence interval [CI] = 1.6-2.4; pain only adjusted sHR = 1.58; CI = 1.4-1.8; sleep-only adjusted sHR = 1.35; CI = 1.1-1.6; no use = reference group). Cumulative incidence of frailty over 8 years for the four groups was 60.6%, 50.9%, 45.8%, and 34.1%. Sensitivity analyses controlling for chronic diseases associated with persistent pain resulted in minor risk reductions. CONCLUSION Prescription pain and sleep drug use is significantly associated with increased incidence of frailty. Research to estimate effects of pain and sleep indications and of drug class-specific dosage and duration on incident frailty is indicated before advocating deprescribing based on these findings. J Am Geriatr Soc 67:2474-2481, 2019.
Collapse
Affiliation(s)
| | - Juyoung Park
- Florida Atlantic University Phyllis and Harvey Sandler School of Social Work, Boca Raton, Florida
| | | |
Collapse
|