1
|
Yasmin S, Ashique S, Taj T, Garg A, Das J, Shorog E, Bhui U, Pal R, Selim S, Panigrahy UP, Begum N, Islam A, Ansari MY. The role of ACE inhibitors and ARBs in preserving cognitive function via hypertension Management: A critical Update. Brain Res 2025; 1850:149400. [PMID: 39681155 DOI: 10.1016/j.brainres.2024.149400] [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: 07/15/2024] [Revised: 11/27/2024] [Accepted: 12/10/2024] [Indexed: 12/18/2024]
Abstract
Hypertension poses a significant risk to cognition-related disorders like dementia. As the global population ages, age-related neurological illnesses such as Alzheimer's disease are becoming increasingly prevalent. The primary hypertension treatments, angiotensin receptor blockers, and angiotensin-converting enzyme inhibitors, exhibit neuroprotective properties. However, observational studies suggest that they may independently contribute to cognitive decline and dementia. Some of these medications have shown promise in reducing cognitive impairment and amyloid buildup in Alzheimer's models. While direct comparisons between the two drug classes are limited, angiotensin receptor blockers have been associated with less brain shrinkage, lower dementia incidence, and slower cognitive decline compared to angiotensin-converting enzyme inhibitors. Both types of medications can influence cognition by passing the blood-brain barrier, with angiotensin receptor blockers potentially offering superior neuroprotective effects due to their selective blockade of the angiotensin type 1 receptor.
Collapse
Affiliation(s)
- Sabina Yasmin
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha 62529, Saudi Arabia
| | - Sumel Ashique
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India.
| | - Tahreen Taj
- Department of Pharmacology, Yenepoya Pharmacy college and research centre, Yenepoya (Deemed to be) university, Mangalore 575018 , India
| | - Ashish Garg
- Guru Ramdas Khalsa Institute of Science and Technology, Pharmacy, Jabalpur, M.P, 483001, India
| | - Joy Das
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab-144411, India
| | - Eman Shorog
- Clinical Pharmacy Department, College of Pharmacy, King Khalid University, Abha, 62529, Saudi Arabia
| | - Utpal Bhui
- Department of Pharmacology, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab-144411, India
| | - Radheshyam Pal
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 144411, India
| | - Samy Selim
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Saudi Arabia
| | - Uttam Prasad Panigrahy
- Department of Pharmaceutical Analysis, Faculty of Pharmaceutical Science, Assam down town University, SankarMadhab Path,Gandhi Nagar, Panikhaiti, Guwahati, Assam 781026, India
| | - Naseem Begum
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University Abha-62529, Saudi Arabia
| | - Anas Islam
- Faculty of Pharmacy, Integral University, Lucknow, 226026, Uttar Pradesh, India
| | - Mohammad Yousuf Ansari
- MM college of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana 133207, India.
| |
Collapse
|
2
|
Anderson ME, Wind EJ, Robison LS. Exploring the neuroprotective role of physical activity in cerebral small vessel disease. Brain Res 2024; 1833:148884. [PMID: 38527712 PMCID: PMC12046637 DOI: 10.1016/j.brainres.2024.148884] [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: 12/27/2023] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 03/27/2024]
Abstract
Cerebral small vessel disease (cSVD) is a common neurological finding characterized by abnormalities of the small blood vessels in the brain. Previous research has established a strong connection between cSVD and stroke, as well as neurodegenerative disorders, notably Alzheimer's disease (AD) and other dementias. As the search for effective interventions continues, physical activity (PA) has emerged as a potential preventative and therapeutic avenue. This review synthesizes the human and animal literature on the influence of PA on cSVD, highlighting the importance of determining optimal exercise protocols, considering aspects such as intensity, duration, timing, and exercise type. Furthermore, the necessity of widening the age bracket in research samples is discussed, ensuring a holistic understanding of the interventions across varying pathological stages of the disease. The review also suggests the potential of exploring diverse biomarkers and risk profiles associated with clinically significant outcomes. Moreover, we review findings demonstrating the beneficial effects of PA in various rodent models of cSVD, which have uncovered numerous mechanisms of neuroprotection, including increases in neuroplasticity and integrity of the vasculature and white matter; decreases in inflammation, oxidative stress, and mitochondrial dysfunction; and alterations in amyloid processing and neurotransmitter signaling. In conclusion, this review highlights the potential of physical activity as a preventive strategy for addressing cSVD, offering insights into the need for refining exercise parameters, diversifying research populations, and exploring novel biomarkers, while shedding light on the intricate mechanisms through which exercise confers neuroprotection in both humans and animal models.
Collapse
Affiliation(s)
- Maria E Anderson
- Department of Psychology, Family, and Justice Studies, University of Saint Joseph, 1678 Asylum Ave, West Hartford, CT 06117, USA
| | - Eleanor J Wind
- Department of Psychology and Neuroscience, Nova Southeastern University, 3300 S. University Drive, Fort Lauderdale, FL 33328, USA
| | - Lisa S Robison
- Department of Psychology and Neuroscience, Nova Southeastern University, 3300 S. University Drive, Fort Lauderdale, FL 33328, USA.
| |
Collapse
|
3
|
Pacholko A, Iadecola C. Hypertension, Neurodegeneration, and Cognitive Decline. Hypertension 2024; 81:991-1007. [PMID: 38426329 PMCID: PMC11023809 DOI: 10.1161/hypertensionaha.123.21356] [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] [Indexed: 03/02/2024]
Abstract
Elevated blood pressure is a well-established risk factor for age-related cognitive decline. Long linked to cognitive impairment on vascular bases, increasing evidence suggests a potential association of hypertension with the neurodegenerative pathology underlying Alzheimer disease. Hypertension is well known to disrupt the structural and functional integrity of the cerebral vasculature. However, the mechanisms by which these alterations lead to brain damage, enhance Alzheimer pathology, and promote cognitive impairment remain to be established. Furthermore, critical questions concerning whether lowering blood pressure by antihypertensive medications prevents cognitive impairment have not been answered. Recent developments in neurovascular biology, brain imaging, and epidemiology, as well as new clinical trials, have provided insights into these critical issues. In particular, clinical and basic findings on the link between neurovascular dysfunction and the pathobiology of neurodegeneration have shed new light on the overlap between vascular and Alzheimer pathology. In this review, we will examine the progress made in the relationship between hypertension and cognitive impairment and, after a critical evaluation of the evidence, attempt to identify remaining knowledge gaps and future research directions that may advance our understanding of one of the leading health challenges of our time.
Collapse
Affiliation(s)
- Anthony Pacholko
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
| |
Collapse
|
4
|
Yao Q, Jiang K, Lin F, Zhu T, Khan NH, Jiang E. Pathophysiological Association of Alzheimer's Disease and Hypertension: A Clinical Concern for Elderly Population. Clin Interv Aging 2023; 18:713-728. [PMID: 37181536 PMCID: PMC10167960 DOI: 10.2147/cia.s400527] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 04/22/2023] [Indexed: 05/16/2023] Open
Abstract
Alzheimer's disease (AD), the most common cause of dementia and the fifth leading cause of death in the adult population has a complex pathophysiological link with hypertension (HTN). A growing volume of published literature on a parallel elevation of blood pressure (BP), amyloid plaques, and neurofibrillary tangles formation in post-middle of human brain cells has developed new, widely accepting foundations on this association. In particular, HTN in elderly life mediates cerebral blood flow dysfunction, neuronal dysfunction, and significant decline in cognitive impairment, primarily in the late-life populace, governing the onset of AD. Thus, HTN is an established risk factor for AD. Considering the impact of AD, 1.89 million deaths annually, and the failure of palliative therapies to cure AD, the scientific research community is looking to adopt integrated approaches to target early modified risk factors like HTN to reduce AD burden. The current review highlights the significance and impact of HTN-based prevention in lowering the AD burden in the elderly by providing a comprehensive overview of the physiological relationship between AD and HTN with an in-detail explanation of the role and applications of pathological biomarkers in this clinical association. The review will gain worth in presenting new insights and providing inclusive discussion on the correlation between HTN and cognitive impairment. It will increase across a wider scientific audience to expand understanding of this pathophysiological association.
Collapse
Affiliation(s)
- Qianqian Yao
- Institute of Nursing and Health, Henan University, Kaifeng, People’s Republic of China
| | - Kexin Jiang
- Institute of Nursing and Health, Henan University, Kaifeng, People’s Republic of China
| | - Fei Lin
- School of Medicine, Shangqiu Institute of Technology, Shangqiu, People’s Republic of China
| | - Tao Zhu
- Department of Geriatrics, Kaifeng Traditional Chinese Medicine Hospital, Kaifeng, People’s Republic of China
| | - Nazeer Hussain Khan
- Institute of Nursing and Health, Henan University, Kaifeng, People’s Republic of China
- Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, People’s Republic of China
| | - Enshe Jiang
- Institute of Nursing and Health, Henan University, Kaifeng, People’s Republic of China
- Henan International Joint Laboratory for Nuclear Protein Regulation, Henan University, Kaifeng, People’s Republic of China
| |
Collapse
|
5
|
Zhang P, Hou Y, Tu W, Campbell N, Pieper AA, Leverenz JB, Gao S, Cummings J, Cheng F. Population-based discovery and Mendelian randomization analysis identify telmisartan as a candidate medicine for Alzheimer's disease in African Americans. Alzheimers Dement 2023; 19:1876-1887. [PMID: 36331056 PMCID: PMC10156891 DOI: 10.1002/alz.12819] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 08/11/2022] [Accepted: 09/02/2022] [Indexed: 11/06/2022]
Abstract
INTRODUCTION African Americans (AAs) and European Americans (EAs) differ in Alzheimer's disease (AD) prevalence, risk factors, and symptomatic presentation and AAs are less likely to enroll in AD clinical trials. METHODS We conducted race-conscious pharmacoepidemiologic studies of 5.62 million older individuals (age ≥60) to investigate the association of telmisartan exposure and AD outcome using Cox analysis, Kaplan-Meier analysis, and log-rank test. We performed Mendelian randomization (MR) analysis of large ethnically diverse genetic data to test likely causal relationships between telmisartan's target and AD. RESULTS We identified that moderate/high telmisartan exposure was significantly associated with a reduced incidence of AD in the AAs compared to low/no telmisartan exposure (hazard ratio [HR] = 0.77, 95% CI: 0.65-0.91, p-value = 0.0022), but not in the non-Hispanic EAs (HR = 0.97, 95% CI: 0.89-1.05, p-value = 0.4110). Sensitivity and sex-/age-stratified patient subgroup analyses identified that telmisartan's medication possession ratio (MPR) and average hypertension daily dosage were significantly associated with a stronger reduction in the incidence of both AD and dementia in AAs. Using MR analysis from large genome-wide association studies (GWAS) (over 2 million individuals) across AD, hypertension, and diabetes, we further identified AA-specific beneficial effects of telmisartan for AD. DISCUSSION Randomized controlled trials with ethnically diverse patient cohorts are warranted to establish causality and therapeutic outcomes of telmisartan and AD. HIGHLIGHTS Telmisartan is associated with lower risk of Alzheimer's disease (AD) in African Americans (AAs). Telmisartan is the only angiotensin II receptor blockers having PPAR-γ agonistic properties with beneficial anti-diabetic and renal function effects, which mitigate AD risk in AAs. Mendelian randomization (MR) analysis demonstrates the specificity of telmisartan's protective mechanism to AAs.
Collapse
Affiliation(s)
- Pengyue Zhang
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis, Indiana, USA
| | - Yuan Hou
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Wanzhu Tu
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis, Indiana, USA
| | - Noll Campbell
- Department of Pharmacy Practice, Purdue University, West Lafayette, Indiana, USA
| | - Andrew A. Pieper
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, Ohio, USA
- Department of Psychiatry, Case Western Reserve University, Cleveland, Ohio, USA
- Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center, Cleveland, Ohio, USA
- Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Neuroscience, Case Western Reserve University, School of Medicine, Cleveland, Ohio, USA
| | - James B. Leverenz
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Lou Ruvo Center for Brain Health, Neurological Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Sujuan Gao
- Department of Biostatistics and Health Data Science, Indiana University, Indianapolis, Indiana, USA
| | - Jeffrey Cummings
- Chambers-Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health Sciences, University of Nevada Las Vegas, Las Vegas, Nevada, USA
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| |
Collapse
|
6
|
Naessens DMP, de Vos J, Richard E, Wilhelmus MMM, Jongenelen CAM, Scholl ER, van der Wel NN, Heijst JA, Teunissen CE, Strijkers GJ, Coolen BF, VanBavel E, Bakker ENTP. Effect of long-term antihypertensive treatment on cerebrovascular structure and function in hypertensive rats. Sci Rep 2023; 13:3481. [PMID: 36859481 PMCID: PMC9977931 DOI: 10.1038/s41598-023-30515-0] [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: 10/31/2022] [Accepted: 02/24/2023] [Indexed: 03/03/2023] Open
Abstract
Midlife hypertension is an important risk factor for cognitive impairment and dementia, including Alzheimer's disease. We investigated the effects of long-term treatment with two classes of antihypertensive drugs to determine whether diverging mechanisms of blood pressure lowering impact the brain differently. Spontaneously hypertensive rats (SHR) were either left untreated or treated with a calcium channel blocker (amlodipine) or beta blocker (atenolol) until one year of age. The normotensive Wistar Kyoto rat (WKY) was used as a reference group. Both drugs lowered blood pressure equally, while only atenolol decreased heart rate. Cerebrovascular resistance was increased in SHR, which was prevented by amlodipine but not atenolol. SHR showed a larger carotid artery diameter with impaired pulsatility, which was prevented by atenolol. Cerebral arteries demonstrated inward remodelling, stiffening and endothelial dysfunction in SHR. Both treatments similarly improved these parameters. MRI revealed that SHR have smaller brains with enlarged ventricles. In addition, neurofilament light levels were increased in cerebrospinal fluid of SHR. However, neither treatment affected these parameters. In conclusion, amlodipine and atenolol both lower blood pressure, but elicit a different hemodynamic profile. Both medications improve cerebral artery structure and function, but neither drug prevented indices of brain damage in this model of hypertension.
Collapse
Affiliation(s)
- Daphne M. P. Naessens
- grid.509540.d0000 0004 6880 3010Amsterdam UMC Location University of Amsterdam, Biomedical Engineering and Physics, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands ,Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands ,grid.484519.5Amsterdam Neuroscience, Neurovascular Disorders, Amsterdam, The Netherlands
| | - Judith de Vos
- grid.509540.d0000 0004 6880 3010Amsterdam UMC Location University of Amsterdam, Biomedical Engineering and Physics, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands ,Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands ,grid.484519.5Amsterdam Neuroscience, Neurovascular Disorders, Amsterdam, The Netherlands
| | - Edo Richard
- grid.509540.d0000 0004 6880 3010Amsterdam UMC Location University of Amsterdam, Public and Occupational Health, Amsterdam, The Netherlands ,grid.10417.330000 0004 0444 9382Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Micha M. M. Wilhelmus
- grid.509540.d0000 0004 6880 3010Amsterdam UMC Location Vrije Universiteit Amsterdam, Anatomy and Neurosciences, Amsterdam, The Netherlands ,grid.484519.5Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands
| | - Cornelis A. M. Jongenelen
- grid.509540.d0000 0004 6880 3010Amsterdam UMC Location Vrije Universiteit Amsterdam, Anatomy and Neurosciences, Amsterdam, The Netherlands
| | - Edwin R. Scholl
- grid.5650.60000000404654431Amsterdam UMC Location University of Amsterdam, Medical Biology, Electron Microscopy Center Amsterdam, Amsterdam, The Netherlands
| | - Nicole N. van der Wel
- grid.5650.60000000404654431Amsterdam UMC Location University of Amsterdam, Medical Biology, Electron Microscopy Center Amsterdam, Amsterdam, The Netherlands
| | - Johannes A. Heijst
- grid.509540.d0000 0004 6880 3010Amsterdam UMC Location Vrije Universiteit Amsterdam, Neurochemistry Laboratory, Clinical Chemistry, Amsterdam, The Netherlands
| | - Charlotte E. Teunissen
- grid.484519.5Amsterdam Neuroscience, Neurodegeneration, Amsterdam, The Netherlands ,grid.509540.d0000 0004 6880 3010Amsterdam UMC Location Vrije Universiteit Amsterdam, Neurochemistry Laboratory, Clinical Chemistry, Amsterdam, The Netherlands ,grid.484519.5Amsterdam Neuroscience, Neuroinfection and -Inflammation, Amsterdam, The Netherlands
| | - Gustav J. Strijkers
- grid.509540.d0000 0004 6880 3010Amsterdam UMC Location University of Amsterdam, Biomedical Engineering and Physics, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Bram F. Coolen
- grid.509540.d0000 0004 6880 3010Amsterdam UMC Location University of Amsterdam, Biomedical Engineering and Physics, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands ,Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
| | - Ed VanBavel
- grid.509540.d0000 0004 6880 3010Amsterdam UMC Location University of Amsterdam, Biomedical Engineering and Physics, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands ,Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands ,grid.484519.5Amsterdam Neuroscience, Neurovascular Disorders, Amsterdam, The Netherlands
| | - Erik N. T. P. Bakker
- grid.509540.d0000 0004 6880 3010Amsterdam UMC Location University of Amsterdam, Biomedical Engineering and Physics, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands ,Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands ,grid.484519.5Amsterdam Neuroscience, Neurovascular Disorders, Amsterdam, The Netherlands
| |
Collapse
|
7
|
Patil G, Kulsange S, Kazi R, Chirmade T, Kale V, Mote C, Aswar M, Koratkar S, Agawane S, Kulkarni M. Behavioral and Proteomic Studies Reveal Methylglyoxal Activate Pathways Associated with Alzheimer's Disease. ACS Pharmacol Transl Sci 2023; 6:65-75. [PMID: 36654748 PMCID: PMC9841776 DOI: 10.1021/acsptsci.2c00143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Indexed: 12/29/2022]
Abstract
Diabetes is one of the major risk factors for Alzheimer's disease (AD) development. The role of elevated levels of glucose, methylglyoxal (MGO), and advanced glycation end products (AGEs) in the pathogenesis of AD is not well understood. In this pursuit, we studied the role of methylglyoxal in the pathogenesis of AD in rat models. The elevated plus-maze (EPM) behavioral study indicated that MGO induces anxiety. Treatment of telmisartan (RAGE expression inhibitor) and aminoguanidine (MGO quencher) attenuated MGO induced anxiety. Further, hippocampal proteomics demonstrated that MGO treated rats differentially regulate proteins involved in calcium homeostasis, mitochondrial functioning, and apoptosis, which may affect neurotransmission and neuronal plasticity. The hippocampal tau phosphorylation level was increased in MGO treated rats, which was reduced in the presence of aminoguanidine and telmisartan. The plasma fructosamine level was increased upon MGO treatment. Hippocampal histochemistry showed vascular degeneration and neuronal loss upon MGO treatment. This study provides mechanistic insight into the role of MGO in the diabetes-associated development of AD.
Collapse
Affiliation(s)
- Gouri Patil
- Biochemical
Sciences Division, CSIR-National Chemical
Laboratory, Pune 411008, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Shabda Kulsange
- Biochemical
Sciences Division, CSIR-National Chemical
Laboratory, Pune 411008, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Rubina Kazi
- Biochemical
Sciences Division, CSIR-National Chemical
Laboratory, Pune 411008, India
| | - Tejas Chirmade
- Biochemical
Sciences Division, CSIR-National Chemical
Laboratory, Pune 411008, India
| | - Vaikhari Kale
- Biochemical
Sciences Division, CSIR-National Chemical
Laboratory, Pune 411008, India
| | - Chandrashekhar Mote
- Department
of Veterinary Pathology, KNP College of Veterinary Science, Shirwal Satara (Maharashtra Animal and Fishery Sciences
University Nagpur), Satara 412801, Maharashtra, India
| | - Manoj Aswar
- Department
of Pharmacology, Sinhgad Institute of Pharmacy,
Narhe, Pune 411041, Maharashtra, India
| | - Santosh Koratkar
- Symbiosis
School of Biological Sciences, Symbiosis
International (Deemed University), Pune 412115, Maharashtra, India
| | - Sachin Agawane
- Biochemical
Sciences Division, CSIR-National Chemical
Laboratory, Pune 411008, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Mahesh Kulkarni
- Biochemical
Sciences Division, CSIR-National Chemical
Laboratory, Pune 411008, India
- Academy
of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| |
Collapse
|
8
|
Abstract
Hypertension affects a significant proportion of the adult and aging population and represents an important risk factor for vascular cognitive impairment and late-life dementia. Chronic high blood pressure continuously challenges the structural and functional integrity of the cerebral vasculature, leading to microvascular rarefaction and dysfunction, and neurovascular uncoupling that typically impairs cerebral blood supply. Hypertension disrupts blood-brain barrier integrity, promotes neuroinflammation, and may contribute to amyloid deposition and Alzheimer pathology. The mechanisms underlying these harmful effects are still a focus of investigation, but studies in animal models have provided significant molecular and cellular mechanistic insights. Remaining questions relate to whether adequate treatment of hypertension may prevent deterioration of cognitive function, the threshold for blood pressure treatment, and the most effective antihypertensive drugs. Recent advances in neurovascular biology, advanced brain imaging, and detection of subtle behavioral phenotypes have begun to provide insights into these critical issues. Importantly, a parallel analysis of these parameters in animal models and humans is feasible, making it possible to foster translational advancements. In this review, we provide a critical evaluation of the evidence available in experimental models and humans to examine the progress made and identify remaining gaps in knowledge.
Collapse
Affiliation(s)
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY
| | - Daniela Carnevale
- Department of Molecular Medicine, “Sapienza” University of Rome, Italy
- Research Unit of Neuro and Cardiovascular Pathophysiology, IRCCS Neuromed, Pozzilli, Italy
| |
Collapse
|
9
|
Chen F, Chen Y, Wang Y, Ke Q, Cui L. The COVID-19 pandemic and Alzheimer's disease: mutual risks and mechanisms. Transl Neurodegener 2022; 11:40. [PMID: 36089575 PMCID: PMC9464468 DOI: 10.1186/s40035-022-00316-y] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 09/02/2022] [Indexed: 11/10/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), which is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a life-threatening disease, especially in elderly individuals and those with comorbidities. The predominant clinical manifestation of COVID-19 is respiratory dysfunction, while neurological presentations are increasingly being recognized. SARS-CoV-2 invades host cells primarily via attachment of the spike protein to the angiotensin-converting enzyme 2 (ACE2) receptor expressed on cell membranes. Patients with Alzheimer's disease (AD) are more susceptible to SARS-CoV-2 infection and prone to severe clinical outcomes. Recent studies have revealed some common risk factors for AD and COVID-19. An understanding of the association between COVID-19 and AD and the potential related mechanisms may lead to the development of novel approaches to treating both diseases. In the present review, we first summarize the mechanisms by which SARS-CoV-2 invades the central nervous system (CNS) and then discuss the associations and potential shared key factors between COVID-19 and AD, with a focus on the ACE2 receptor, apolipoprotein E (APOE) genotype, age, and neuroinflammation.
Collapse
Affiliation(s)
- Feng Chen
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences & Yunnan Province Kunming Institute of Zoology Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Yanting Chen
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yongxiang Wang
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Qiongwei Ke
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Lili Cui
- Guangdong Key Laboratory of Age-Related Cardiac and Cerebral Diseases, Department of Neurology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.
| |
Collapse
|
10
|
Şen S, Hacıosmanoğlu E. Comparing the Neuroprotective Effects of Telmisartan, Perindopril, and Nebivolol Against Lipopolysaccharide-Induced Injury in Neuron-Like Cells. Cureus 2022; 14:e27429. [PMID: 36051740 PMCID: PMC9420193 DOI: 10.7759/cureus.27429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2022] [Indexed: 11/05/2022] Open
Abstract
The effect of antihypertensive drugs, especially drugs modulating the renin-angiotensin-aldosterone-system (RAAS), on neurodegenerative diseases still needs to be investigated. This study aimed to compare the effects of three different antihypertensive drugs (telmisartan, perindopril, and nebivolol) on neuroprotection and acetylcholine (ACh) levels against lipopolysaccharide (LPS)-induced injury in a differentiated SH-SY5Y cell line. Cells were treated with retinoic acid for differentiation to a neuronal phenotype. LPS 20 (μg/mL) was applied to the cells for one hour. Then, the cells were treated with 1, 5, and 10 µg/mL concentrations of telmisartan, perindopril, and nebivolol separately for 24 hours, except for the control and LPS alone groups. Cell viability was evaluated using a 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. ACh levels were analyzed using an enzyme immunosorbent assay in the culture medium. Tumor necrosis factor-alpha (TNF-α), interleukin 1 beta (IL-1β), and nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) expressions were evaluated using western blot analysis. Telmisartan demonstrated the highest cell viability against LPS-induced injury, whereas the protective effect of perindopril was moderate. Nebivolol showed no neuroprotective effect. The protective effect of 10-µg/mL telmisartan was superior to 10 µg/mL perindopril (p=0.006), 5 µg/mL perindopril (p=0.001), 1 µg/mL perindopril (p=0.001), and 1, 5, and 10 µg/mL nebivolol (p<0.001). Among all the study drugs, only telmisartan provided a statistically significant increase in ACh levels after LPS-induced injury. Additionally, the administration of telmisartan provided a concentration-dependent reduction in TNF-α, IL-1β, and NFκB expression against LPS-induced neuroinflammation. These findings suggest that telmisartan has a superior neuroprotective effect against LPS-induced injury in neuron-like cells compared with both perindopril and nebivolol.
Collapse
|
11
|
Gouveia F, Camins A, Ettcheto M, Bicker J, Falcão A, Cruz MT, Fortuna A. Targeting brain Renin-Angiotensin System for the prevention and treatment of Alzheimer's disease: Past, present and future. Ageing Res Rev 2022; 77:101612. [PMID: 35346852 DOI: 10.1016/j.arr.2022.101612] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 02/09/2022] [Accepted: 03/16/2022] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a well-known neurodegenerative disease characterized by the presence of two main hallmarks - Tau hyperphosphorylation and Aβ deposits. Notwithstanding, in the last few years the scientific evidence about the drivers of AD have been changing and nowadays age-related vascular alterations and several cardiovascular risk factors have been shown to trigger the development of AD. In this context, drugs targeting the Renin Angiotensin System (RAS), commonly used for the treatment of hypertension, are evidencing a high potential to delay AD development due to their action on brain RAS. Indeed, the ACE 1/Ang II/AT1R axis is believed to be upregulated in AD and to be responsible for deleterious effects such as increased oxidative stress, neuroinflammation, blood-brain barrier (BBB) hyperpermeability, astrocytes dysfunction and a decrease in cerebral blood flow. In contrast, the alternative axis - ACE 1/Ang II/AT2R; ACE 2/Ang (1-7)/MasR; Ang IV/ AT4R(IRAP) - seems to counterbalance the deleterious effects of the principal axis and to exert beneficial effects on memory and cognition. Accordingly, retrospective studies demonstrate a reduced risk of developing AD among people taking RAS medication as well as several in vitro and in vivo pre-clinical studies as it is herein critically reviewed. In this review, we first revise, at a glance, the pathophysiology of AD focused on its classic hallmarks. Secondly, an overview about the impact of the RAS on the pathophysiology of AD is also provided, focused on their four essential axes ACE 1/Ang II/AT2R; ACE 2/Ang (1-7)/MasR; Ang IV/ AT4R(IRAP) and ACE 1/Ang II/AT1R. Finally, the therapeutic potential of available drugs targeting RAS on AD, namely angiotensin II receptor blockers (ARBs) and angiotensin converting enzyme inhibitors (ACEIs), is highlighted and data supporting this hope will be presented, from in vitro and in vivo pre-clinical to clinical studies.
Collapse
|
12
|
Potential role of Drug Repositioning Strategy (DRS) for management of tauopathy. Life Sci 2022; 291:120267. [PMID: 34974076 DOI: 10.1016/j.lfs.2021.120267] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/14/2021] [Accepted: 12/22/2021] [Indexed: 01/08/2023]
Abstract
Tauopathy is a term that has been used to represent a pathological condition in which hyperphosphorylated tau protein aggregates in neurons and glia which results in neurodegeneration, synapse loss and dysfunction and cognitive impairments. Recently, drug repositioning strategy (DRS) becomes a promising field and an alternative approach to advancing new treatments from actually developed and FDA approved drugs for an indication other than the indication it was originally intended for. This paradigm provides an advantage because the safety of the candidate compound has already been established, which abolishes the need for further preclinical safety testing and thus substantially reduces the time and cost involved in progressing of clinical trials. In the present review, we focused on correlation between tauopathy and common diseases as type 2 diabetes mellitus and the global virus COVID-19 and how tau pathology can aggravate development of these diseases in addition to how these diseases can be a risk factor for development of tauopathy. Moreover, correlation between COVID-19 and type 2 diabetes mellitus was also discussed. Therefore, repositioning of a drug in the daily clinical practice of patients to manage or prevent two or more diseases at the same time with lower side effects and drug-drug interactions is a promising idea. This review concluded the results of pre-clinical and clinical studies applied on antidiabetics, COVID-19 medications, antihypertensives, antidepressants and cholesterol lowering drugs for possible drug repositioning for management of tauopathy.
Collapse
|
13
|
Molecular Mechanisms of Neuroimmune Crosstalk in the Pathogenesis of Stroke. Int J Mol Sci 2021; 22:ijms22179486. [PMID: 34502395 PMCID: PMC8431165 DOI: 10.3390/ijms22179486] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 08/26/2021] [Accepted: 08/28/2021] [Indexed: 12/21/2022] Open
Abstract
Stroke disrupts the homeostatic balance within the brain and is associated with a significant accumulation of necrotic cellular debris, fluid, and peripheral immune cells in the central nervous system (CNS). Additionally, cells, antigens, and other factors exit the brain into the periphery via damaged blood–brain barrier cells, glymphatic transport mechanisms, and lymphatic vessels, which dramatically influence the systemic immune response and lead to complex neuroimmune communication. As a result, the immunological response after stroke is a highly dynamic event that involves communication between multiple organ systems and cell types, with significant consequences on not only the initial stroke tissue injury but long-term recovery in the CNS. In this review, we discuss the complex immunological and physiological interactions that occur after stroke with a focus on how the peripheral immune system and CNS communicate to regulate post-stroke brain homeostasis. First, we discuss the post-stroke immune cascade across different contexts as well as homeostatic regulation within the brain. Then, we focus on the lymphatic vessels surrounding the brain and their ability to coordinate both immune response and fluid homeostasis within the brain after stroke. Finally, we discuss how therapeutic manipulation of peripheral systems may provide new mechanisms to treat stroke injury.
Collapse
|
14
|
Law CSW, Yeong KY. Repurposing Antihypertensive Drugs for the Management of Alzheimer's Disease. Curr Med Chem 2021; 28:1716-1730. [PMID: 32164502 DOI: 10.2174/0929867327666200312114223] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 02/04/2020] [Accepted: 02/18/2020] [Indexed: 11/22/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that has affected millions of people worldwide. However, currently, there is no treatment to cure the disease. The AD drugs available in the market only manage the disease symptomatically and the effects are usually short-term. Thus, there is a need to look at alternatives AD therapies. This literature review aims to shed some light on the potential of repurposing antihypertensives to treat AD. Mid-life hypertension has not only been recognised as a risk factor for AD, but its relation with AD has also been well established. Hence, antihypertensives were postulated to be beneficial in managing AD. Four classes of antihypertensives, as well as their potential limitations and prospects in being utilised as AD therapeutics, were discussed in this review.
Collapse
Affiliation(s)
- Christine Shing Wei Law
- School of Science, Monash University Malaysia Campus, Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor, Malaysia
| | - Keng Yoon Yeong
- School of Science, Monash University Malaysia Campus, Jalan Lagoon Selatan, Bandar Sunway, 47500, Selangor, Malaysia
| |
Collapse
|
15
|
Zhang CY, He FF, Su H, Zhang C, Meng XF. Association between chronic kidney disease and Alzheimer's disease: an update. Metab Brain Dis 2020; 35:883-894. [PMID: 32246323 DOI: 10.1007/s11011-020-00561-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 03/05/2020] [Indexed: 12/11/2022]
Abstract
It has been accepted that kidney function is connected with brain activity. In clinical studies, chronic kidney disease (CKD) patients have been found to be prone to suffering cognitive decline and Alzheimer's disease (AD). The cognitive function of CKD patients may improve after kidney transplantation. All these indicators show a possible link between kidney function and dementia. However, little is known about the mechanism behind the relation of CKD and AD. This review discusses the associations between CKD and AD from the perspective of the pathophysiology of the kidney and complications and/or concomitants of CKD that may lead to cognitive decline in the progression of CKD and AD. Potential preventive and therapeutic strategies for AD are also presented. Further studies are warranted in order to confirm whether the setting of CKD is a possible new determinant for cognitive impairment in AD.
Collapse
Affiliation(s)
- Chun-Yun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Fang-Fang He
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hua Su
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chun Zhang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xian-Fang Meng
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
16
|
Plascencia-Villa G, Perry G. Status and future directions of clinical trials in Alzheimer's disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2020; 154:3-50. [PMID: 32739008 DOI: 10.1016/bs.irn.2020.03.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Amyloid-β (Aβ) senile plaques and neurofibrillary tangles of tau are generally recognized as the culprits of Alzheimer's disease (AD) and related dementia. About 25 years ago, the amyloid cascade hypotheses postulated a direct correlation of plaques with the development of AD, and it has been the dominant theory since then. In this period, more than 200 clinical trials focused mainly on targeting components of the Aβ cascade have dramatically failed, some of them in Phase III. With a greater than 99.6% failure rate at a cost of several billion from governments, industry, and private funders, therapeutic strategies targeting amyloid and tau are now under scrutiny. Therefore, it is time to reevaluate alternatives to targeting Aβ and tau as effective therapeutic strategies for AD. The diagnosis of AD is currently based on medical examination of symptoms including tests to assess memory impairment, attention, language, and other thinking skills. This is complemented with brain scans, such as computed tomography, magnetic resonance imaging, or positron emission tomography with the help of imaging probes targeting Aβ or tau deposits. This approach has contributed to the tunnel vision focus on Aβ and tau as the main culprits of AD. However, events upstream of these proteopathies (age-related impaired neuronal bioenergetics, lysosome function, neurotrophic signaling, and neuroinflammation, among others) are almost surely where the development of alternative therapeutic interventions should be targeted. Here, we present the current status of therapeutic candidates targeting diverse mechanisms and strategies including Aβ and tau, proteins involved in Aβ production and trafficking (ApoE, α/β/γ-secretases), neuroinflammation, neurotransmitters, neuroprotective agents antimicrobials, and gene and stem cell therapy. There are currently around 33 compounds in Phase III, 78 in Phase II, and 32 more in Phase I trials. With the current world health crisis of increased dementia in a rapidly aging population, effective AD therapies are desperately needed.
Collapse
Affiliation(s)
- Germán Plascencia-Villa
- Department of Biology and Neurosciences Institute, The University of Texas at San Antonio (UTSA), 1 UTSA Circle, San Antonio, TX, United States
| | - George Perry
- Department of Biology and Neurosciences Institute, The University of Texas at San Antonio (UTSA), 1 UTSA Circle, San Antonio, TX, United States.
| |
Collapse
|
17
|
Telmisartan/17β-estradiol mitigated cognitive deficit in an ovariectomized rat model of Alzheimer's disease: Modulation of ACE1/ACE2 and AT1/AT2 ratio. Life Sci 2020; 245:117388. [PMID: 32007576 DOI: 10.1016/j.lfs.2020.117388] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 01/22/2020] [Accepted: 01/29/2020] [Indexed: 12/29/2022]
Abstract
AIMS The higher incidence rate of Alzheimer's disease (AD) among women has led to explorations on the association between estrogen deficiency and AD. Also, usage of antihypertensive drugs has been suggested to reduce the incidence of AD in elderly hypertensive patients. Thus, this study aimed to investigate the effects of telmisartan and/or 17β-estradiol on a cognitively impaired ovariectomized rat model of AD. MAIN METHODS 75 female Wistar rats were randomly allocated into five groups. One group was sham operated and the other four groups were subjected to ovariectomy, received D-galactose and either untreated or treated with telmisartan and/or 17β-estradiol for 6 weeks. KEY FINDINGS Ovariectomized rats showed cognitive impairment in Morris water maze and novel object recognition tests, increasing inflammatory biomarkers (tumor necrosis factor-α, and interleukin-1β), increasing AD biomarkers (amyloid beta1-42, and acetylcholine esterase), and over activation of classical arm of renin angiotensin system (RAS) (ACE1/Ang2/AT1) in hippocampi. Also, hippocampi histopathological examination revealed amyloid beta deposition. Whereas, administration of telmisartan and/or 17β-estradiol improved animals' behavior, alleviated histopathological alterations and reduced the level of inflammatory and AD biomarkers, modulated RAS activity favoring the novel neuroprotective arm (ACE2/Ang(1-7)/MasR). SIGNIFICANCE Our findings suggest that combined administration of both drugs has synergetic neuroprotective effects; supporting their potential application in AD treatment.
Collapse
|
18
|
Telmisartan Protects Against Aluminum-Induced Alzheimer-like Pathological Changes in Rats. Neurotox Res 2019; 37:275-285. [PMID: 31332715 DOI: 10.1007/s12640-019-00085-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 06/23/2019] [Accepted: 06/28/2019] [Indexed: 12/23/2022]
Abstract
Currently, there is no effective mean for treatment or prevention of Alzheimer's disease (AD). Commonly used AD drugs have a moderate effect and treat only the associated symptoms, therefore there is a strong need to search for more effective agents. Our goal is to examine telmisartan neuroprotective effect in aluminum-induced cognitive impairment in rats. Aluminum chloride (10 mg/kg, i.p) was administered for 2 months then behavioral tests (Y-maze and Morris water maze) were done. Hippocampal biochemical and histological analysis were then carried out. AD-like histological, biochemical, and behavioral alterations appeared in aluminum-treated rats. Telmisartan improved rats' condition on behavioral and histological levels. It reversed the increase in hippocampal amyloid beta protein, phosphorylated tau protein contents together with augmentation of neprilysin level, it also diminished levels of nuclear factor kappa-B, FAS ligand, tumor necrosis factor-alpha, malondialdehyde, and acetylcholinesterase content.These findings show the protective action of telmisartan against AD-like pathological alterations.
Collapse
|
19
|
Ahmed HA, Ishrat T, Pillai B, Fouda AY, Sayed MA, Eldahshan W, Waller JL, Ergul A, Fagan SC. RAS modulation prevents progressive cognitive impairment after experimental stroke: a randomized, blinded preclinical trial. J Neuroinflammation 2018; 15:229. [PMID: 30103772 PMCID: PMC6090822 DOI: 10.1186/s12974-018-1262-x] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 07/29/2018] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND With the aging population, the prevalence and incidence of cerebrovascular disease will continue to rise, as well as the number of individuals with vascular cognitive impairment/dementia (VCID). No specific FDA-approved treatments for VCID exist. Although clinical evidence supports that angiotensin receptor blockers (ARBs) prevent cognitive decline in older adults, whether ARBs have a similar effect on VCID after stroke is unknown. Moreover, these agents reduce BP, which is undesirable in the acute stroke period, so we believe that giving C21 in this acute phase or delaying ARB administration would enable us to achieve the neurovascular benefits without the risk of unintended and potentially dangerous, acute BP lowering. METHODS The aim of our study was to determine the impact of candesartan (ARB) or compound-21 (an angiotensin type 2 receptor--AT2R--agonist) on long-term cognitive function post-stroke, in spontaneously hypertensive rats (SHRs). We hypothesized that AT2R stimulation, either directly with C21, or indirectly by blocking the angiotensin type 1 receptor (AT1R) with candesartan, initiated after stroke, would reduce cognitive impairment. Animals were subjected to a 60-min transient middle cerebral artery occlusion and randomly assigned to either saline/C21 monotherapy, for the full study duration (30 days), or given sequential therapy starting with saline/C21 (7 days) followed by candesartan for the remainder of the study (21 days). Outcome measures included sensorimotor/cognitive-function, amyloid-β determination, and histopathologic analyses. RESULTS Treatment with RAS modulators effectively preserved cognitive function, reduced cytotoxicity, and prevented chronic-reactive microgliosis in SHRs, post-stroke. These protective effects were apparent even when treatment was delayed up to 7 days post-stroke and were independent of blood pressure and β-amyloid accumulation. CONCLUSION Collectively, our findings demonstrate that RAS modulators effectively prevent cognitive impairment after stroke, even when treatment is delayed.
Collapse
Affiliation(s)
- Heba A. Ahmed
- Program in Clinical and Experimental Therapeutics, Charlie Norwood VA Medical Center and University of Georgia College of Pharmacy, HM Bldg., 1120 15th St, Augusta, GA 30912 USA
| | - Tauheed Ishrat
- Department of Anatomy and Neurobiology, College of Medicine, Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN USA
| | - Bindu Pillai
- Program in Clinical and Experimental Therapeutics, Charlie Norwood VA Medical Center and University of Georgia College of Pharmacy, HM Bldg., 1120 15th St, Augusta, GA 30912 USA
| | - Abdelrahman Y. Fouda
- Program in Clinical and Experimental Therapeutics, Charlie Norwood VA Medical Center and University of Georgia College of Pharmacy, HM Bldg., 1120 15th St, Augusta, GA 30912 USA
| | - Mohammed A. Sayed
- Program in Clinical and Experimental Therapeutics, Charlie Norwood VA Medical Center and University of Georgia College of Pharmacy, HM Bldg., 1120 15th St, Augusta, GA 30912 USA
| | - Wael Eldahshan
- Program in Clinical and Experimental Therapeutics, Charlie Norwood VA Medical Center and University of Georgia College of Pharmacy, HM Bldg., 1120 15th St, Augusta, GA 30912 USA
| | - Jennifer L. Waller
- Department of Biostatistics and Epidemiology, Augusta University, Augusta, GA USA
| | - Adviye Ergul
- Program in Clinical and Experimental Therapeutics, Charlie Norwood VA Medical Center and University of Georgia College of Pharmacy, HM Bldg., 1120 15th St, Augusta, GA 30912 USA
- Department of Physiology, Augusta University, Augusta, GA USA
| | - Susan C. Fagan
- Program in Clinical and Experimental Therapeutics, Charlie Norwood VA Medical Center and University of Georgia College of Pharmacy, HM Bldg., 1120 15th St, Augusta, GA 30912 USA
- Department of Neurology, Augusta University, Augusta, GA USA
| |
Collapse
|
20
|
Csiszar A, Tarantini S, Fülöp GA, Kiss T, Valcarcel-Ares MN, Galvan V, Ungvari Z, Yabluchanskiy A. Hypertension impairs neurovascular coupling and promotes microvascular injury: role in exacerbation of Alzheimer's disease. GeroScience 2017; 39:359-372. [PMID: 28853030 PMCID: PMC5636770 DOI: 10.1007/s11357-017-9991-9] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 07/26/2017] [Indexed: 12/13/2022] Open
Abstract
Hypertension in the elderly substantially increases both the risk of vascular cognitive impairment (VCI) and Alzheimer's disease (AD); however, the underlying mechanisms are not completely understood. This review discusses the effects of hypertension on structural and functional integrity of cerebral microcirculation, including hypertension-induced alterations in neurovascular coupling responses, cellular and molecular mechanisms involved in microvascular damage (capillary rarefaction, blood-brain barrier disruption), and the genesis of cerebral microhemorrhages and their potential role in exacerbation of cognitive decline associated with AD. Understanding and targeting the hypertension-induced cerebromicrovascular alterations that are involved in the onset and progression of AD and contribute to cognitive impairment are expected to have a major role in preserving brain health in high-risk older individuals.
Collapse
Affiliation(s)
- Anna Csiszar
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Translational Geroscience Laboratory, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Stefano Tarantini
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Translational Geroscience Laboratory, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Gábor A Fülöp
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Translational Geroscience Laboratory, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Tamas Kiss
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Translational Geroscience Laboratory, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - M Noa Valcarcel-Ares
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Translational Geroscience Laboratory, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Veronica Galvan
- Department of Cellular and Integrative Physiology, Barshop Institute for Longevity and Aging Studies University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Zoltan Ungvari
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Translational Geroscience Laboratory, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andriy Yabluchanskiy
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Translational Geroscience Laboratory, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
21
|
Edwards JD, Ramirez J, Callahan BL, Tobe SW, Oh P, Berezuk C, Lanctôt K, Swardfager W, Nestor S, Kiss A, Strother S, Black SE. Antihypertensive Treatment is associated with MRI-Derived Markers of Neurodegeneration and Impaired Cognition: A Propensity-Weighted Cohort Study. J Alzheimers Dis 2017; 59:1113-1122. [DOI: 10.3233/jad-170238] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Jodi D. Edwards
- LC Campbell Cognitive Neurology Research Unit, Sunnybrook Health Sciences Centre, Toronto, Canada
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute>, University of Toronto, Toronto, Canada
- Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Site, Toronto, Canada
| | - Joel Ramirez
- LC Campbell Cognitive Neurology Research Unit, Sunnybrook Health Sciences Centre, Toronto, Canada
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute>, University of Toronto, Toronto, Canada
- Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Site, Toronto, Canada
| | - Brandy L. Callahan
- LC Campbell Cognitive Neurology Research Unit, Sunnybrook Health Sciences Centre, Toronto, Canada
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute>, University of Toronto, Toronto, Canada
| | | | - Paul Oh
- Toronto Rehabilitation Institute, Toronto, Canada
| | - Courtney Berezuk
- LC Campbell Cognitive Neurology Research Unit, Sunnybrook Health Sciences Centre, Toronto, Canada
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute>, University of Toronto, Toronto, Canada
- Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Site, Toronto, Canada
| | - Krista Lanctôt
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute>, University of Toronto, Toronto, Canada
- Geriatric Psychiatry, University of Toronto, Toronto, Canada
- Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Walter Swardfager
- LC Campbell Cognitive Neurology Research Unit, Sunnybrook Health Sciences Centre, Toronto, Canada
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute>, University of Toronto, Toronto, Canada
- Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Site, Toronto, Canada
- Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Sean Nestor
- LC Campbell Cognitive Neurology Research Unit, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Alexander Kiss
- Institute for Clinical Evaluative Sciences, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Stephen Strother
- Medical Biophysics, University of Toronto, Toronto, Canada
- Rotman Research Institute, Toronto, Canada
| | - Sandra E. Black
- LC Campbell Cognitive Neurology Research Unit, Sunnybrook Health Sciences Centre, Toronto, Canada
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute>, University of Toronto, Toronto, Canada
- Heart & Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Site, Toronto, Canada
- Department of Medicine (Neurology), Sunnybrook Health Sciences and University of Toronto, Toronto, Canada
| | | |
Collapse
|
22
|
Michalski D, Hofmann S, Pitsch R, Grosche J, Härtig W. Neurovascular Specifications in the Alzheimer-Like Brain of Mice Affected by Focal Cerebral Ischemia: Implications for Future Therapies. J Alzheimers Dis 2017; 59:655-674. [DOI: 10.3233/jad-170185] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
| | - Sarah Hofmann
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Roman Pitsch
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | | | - Wolfgang Härtig
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| |
Collapse
|
23
|
Cai W, Zhang K, Li P, Zhu L, Xu J, Yang B, Hu X, Lu Z, Chen J. Dysfunction of the neurovascular unit in ischemic stroke and neurodegenerative diseases: An aging effect. Ageing Res Rev 2017; 34:77-87. [PMID: 27697546 PMCID: PMC5384332 DOI: 10.1016/j.arr.2016.09.006] [Citation(s) in RCA: 194] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 08/15/2016] [Accepted: 09/26/2016] [Indexed: 12/23/2022]
Abstract
Current understanding on the mechanisms of brain injury and neurodegeneration highlights an appreciation of multicellular interactions within the neurovascular unit (NVU), which include the evolution of blood-brain barrier (BBB) damage, neuronal cell death or degeneration, glial reaction, and immune cell infiltration. Aging is an important factor that influences the integrity of the NVU. The age-related physiological or pathological changes in the cellular components of the NVU have been shown to increase the vulnerability of the NVU to ischemia/reperfusion injury or neurodegeneration, and to result in deteriorated brain damage. This review describes the impacts of aging on each NVU component and discusses the mechanisms by which aging increases NVU sensitivity to stroke and neurodegenerative diseases. Prophylactic or therapeutic perspectives that may delay or diminish aging and thus prevent the incidence of these neurological disorders will also be reviewed.
Collapse
Affiliation(s)
- Wei Cai
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA; Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510630, China; Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Kai Zhang
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA; Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Peiying Li
- Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China; Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Ling Zhu
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA; Department of Anesthesiology, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200127, China; Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jing Xu
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA; Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Boyu Yang
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA; Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Xiaoming Hu
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA; Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Zhengqi Lu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510630, China.
| | - Jun Chen
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261, USA; Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA.
| |
Collapse
|
24
|
Zhai Y, Yamashita T, Nakano Y, Sun Z, Shang J, Feng T, Morihara R, Fukui Y, Ohta Y, Hishikawa N, Abe K. Chronic Cerebral Hypoperfusion Accelerates Alzheimer’s Disease Pathology with Cerebrovascular Remodeling in a Novel Mouse Model. J Alzheimers Dis 2016; 53:893-905. [DOI: 10.3233/jad-160345] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
25
|
Toda N, Okamura T. Cigarette smoking impairs nitric oxide-mediated cerebral blood flow increase: Implications for Alzheimer's disease. J Pharmacol Sci 2016; 131:223-32. [DOI: 10.1016/j.jphs.2016.07.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 07/05/2016] [Accepted: 07/06/2016] [Indexed: 02/08/2023] Open
|
26
|
Salacz ME, Kast RE, Saki N, Brüning A, Karpel-Massler G, Halatsch ME. Toward a noncytotoxic glioblastoma therapy: blocking MCP-1 with the MTZ Regimen. Onco Targets Ther 2016; 9:2535-45. [PMID: 27175087 PMCID: PMC4854261 DOI: 10.2147/ott.s100407] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
To improve the prognosis of glioblastoma, we developed an adjuvant treatment directed to a neglected aspect of glioblastoma growth, the contribution of nonmalignant monocyte lineage cells (MLCs) (monocyte, macrophage, microglia, dendritic cells) that infiltrated a main tumor mass. These nonmalignant cells contribute to glioblastoma growth and tumor homeostasis. MLCs comprise of approximately 10%-30% of glioblastoma by volume. After integration into the tumor mass, these become polarized toward an M2 immunosuppressive, pro-angiogenic phenotype that promotes continued tumor growth. Glioblastoma cells initiate and promote this process by synthesizing 13 kDa MCP-1 that attracts circulating monocytes to the tumor. Infiltrating monocytes, after polarizing toward an M2 phenotype, synthesize more MCP-1, forming an amplification loop. Three noncytotoxic drugs, an antibiotic - minocycline, an antihypertensive drug - telmisartan, and a bisphosphonate - zoledronic acid, have ancillary attributes of MCP-1 synthesis inhibition and could be re-purposed, singly or in combination, to inhibit or reverse MLC-mediated immunosuppression, angiogenesis, and other growth-enhancing aspects. Minocycline, telmisartan, and zoledronic acid - the MTZ Regimen - have low-toxicity profiles and could be added to standard radiotherapy and temozolomide. Re-purposing older drugs has advantages of established safety and low drug cost. Four core observations support this approach: 1) malignant glioblastoma cells require a reciprocal trophic relationship with nonmalignant macrophages or microglia to thrive; 2) glioblastoma cells secrete MCP-1 to start the cycle, attracting MLCs, which subsequently also secrete MCP-1 perpetuating the recruitment cycle; 3) increasing cytokine levels in the tumor environment generate further immunosuppression and tumor growth; and 4) MTZ regimen may impede MCP-1-driven processes, thereby interfering with glioblastoma growth.
Collapse
Affiliation(s)
- Michael E Salacz
- Department of Internal Medicine, University of Kansas, Kansas City, KS, USA; Department of Neurosurgery, University of Kansas, Kansas City, KS, USA
| | | | - Najmaldin Saki
- Health Research Institute, Research Center of Thalassemia and Hemoglobinopathy, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ansgar Brüning
- Molecular Biology Laboratory, University Hospital Munich, Munich, Germany
| | | | | |
Collapse
|
27
|
Michel MC, Brunner HR, Foster C, Huo Y. Angiotensin II type 1 receptor antagonists in animal models of vascular, cardiac, metabolic and renal disease. Pharmacol Ther 2016; 164:1-81. [PMID: 27130806 DOI: 10.1016/j.pharmthera.2016.03.019] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 03/30/2016] [Indexed: 02/07/2023]
Abstract
We have reviewed the effects of angiotensin II type 1 receptor antagonists (ARBs) in various animal models of hypertension, atherosclerosis, cardiac function, hypertrophy and fibrosis, glucose and lipid metabolism, and renal function and morphology. Those of azilsartan and telmisartan have been included comprehensively whereas those of other ARBs have been included systematically but without intention of completeness. ARBs as a class lower blood pressure in established hypertension and prevent hypertension development in all applicable animal models except those with a markedly suppressed renin-angiotensin system; blood pressure lowering even persists for a considerable time after discontinuation of treatment. This translates into a reduced mortality, particularly in models exhibiting marked hypertension. The retrieved data on vascular, cardiac and renal function and morphology as well as on glucose and lipid metabolism are discussed to address three main questions: 1. Can ARB effects on blood vessels, heart, kidney and metabolic function be explained by blood pressure lowering alone or are they additionally directly related to blockade of the renin-angiotensin system? 2. Are they shared by other inhibitors of the renin-angiotensin system, e.g. angiotensin converting enzyme inhibitors? 3. Are some effects specific for one or more compounds within the ARB class? Taken together these data profile ARBs as a drug class with unique properties that have beneficial effects far beyond those on blood pressure reduction and, in some cases distinct from those of angiotensin converting enzyme inhibitors. The clinical relevance of angiotensin receptor-independent effects of some ARBs remains to be determined.
Collapse
Affiliation(s)
- Martin C Michel
- Dept. Pharmacology, Johannes Gutenberg University, Mainz, Germany; Dept. Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim, Ingelheim, Germany.
| | | | - Carolyn Foster
- Retiree from Dept. of Research Networking, Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, CT, USA
| | - Yong Huo
- Dept. Cardiology & Heart Center, Peking University First Hospital, Beijing, PR China
| |
Collapse
|
28
|
Glodzik L, Rusinek H, Kamer A, Pirraglia E, Tsui W, Mosconi L, Li Y, McHugh P, Murray J, Williams S, Osorio RS, Randall C, Butler T, Deshpande A, Vallabhajolusa S, de Leon M. Effects of vascular risk factors, statins, and antihypertensive drugs on PiB deposition in cognitively normal subjects. ALZHEIMER'S & DEMENTIA: DIAGNOSIS, ASSESSMENT & DISEASE MONITORING 2016; 2:95-104. [PMID: 27239540 PMCID: PMC4879519 DOI: 10.1016/j.dadm.2016.02.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Hypertension, hypercholesterolemia, and obesity increase the risk of dementia. Although their detection is commonly followed by an introduction of treatment, little is known about how medications frequently used to treat vascular risk affect amyloid deposition. METHODS A cross-sectional study of 156 subjects who underwent positron emission tomography with PiB. Using linear regression, we tested whether blood pressure, cholesterol, overweight/obese status, angiotensin receptor blockers (ARBs), beta-blockers, diuretics, angiotensin converting enzyme inhibitors, and statins predicted amyloid deposition. RESULTS The use of ARBs (β = -.15, P = .044) and diuretics (β = -.20, P = .006) predicted less amyloid accumulation; older age (β = .29, P < .001) and statins (β = .23, P = .004) were related to greater amyloid deposition. Overweight and/or obese women had more cortical amyloid than their peers. DISCUSSION Prospective studies should confirm effects of drugs and increased body weight on amyloid accumulation and establish whether they translate into measurable clinical outcomes. Women may be more susceptible to harmful effects of obesity.
Collapse
Affiliation(s)
- Lidia Glodzik
- Department of Psychiatry, Center for Brain Health, NYU School of Medicine, New York, USA
- Department of Radiology, NYU School of Medicine, New York, USA
- Corresponding author. Tel.: +1 212 263 5698; Fax: +1 212 263 3270.
| | - Henry Rusinek
- Department of Radiology, NYU School of Medicine, New York, USA
| | - Angela Kamer
- Department of Periodontology and Implant Dentistry, College of Dentistry, New York University, New York, USA
| | - Elizabeth Pirraglia
- Department of Psychiatry, Center for Brain Health, NYU School of Medicine, New York, USA
| | - Wai Tsui
- Department of Psychiatry, Center for Brain Health, NYU School of Medicine, New York, USA
| | - Lisa Mosconi
- Department of Psychiatry, Center for Brain Health, NYU School of Medicine, New York, USA
| | - Yi Li
- Department of Psychiatry, Center for Brain Health, NYU School of Medicine, New York, USA
| | - Pauline McHugh
- Department of Psychiatry, Center for Brain Health, NYU School of Medicine, New York, USA
| | - John Murray
- Department of Psychiatry, Center for Brain Health, NYU School of Medicine, New York, USA
| | | | - Ricardo S. Osorio
- Department of Psychiatry, Center for Brain Health, NYU School of Medicine, New York, USA
| | - Catherine Randall
- Department of Psychiatry, Center for Brain Health, NYU School of Medicine, New York, USA
| | - Tracy Butler
- Department of Psychiatry, Center for Brain Health, NYU School of Medicine, New York, USA
| | - Anup Deshpande
- Department of Psychiatry, Center for Brain Health, NYU School of Medicine, New York, USA
| | | | - Mony de Leon
- Department of Psychiatry, Center for Brain Health, NYU School of Medicine, New York, USA
| |
Collapse
|
29
|
Levy Nogueira M, Epelbaum S, Steyaert JM, Dubois B, Schwartz L. Mechanical stress models of Alzheimer's disease pathology. Alzheimers Dement 2015; 12:324-33. [PMID: 26718585 DOI: 10.1016/j.jalz.2015.10.005] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 09/11/2015] [Accepted: 10/15/2015] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Extracellular accumulation of amyloid-β protein and intracellular accumulation of tau in brain tissues have been described in animal models of Alzheimer's disease (AD) and mechanical stress-based diseases of different mechanisms, such as traumatic brain injury (TBI), arterial hypertension (HTN), and normal pressure hydrocephalus (NPH). METHODS We provide a brief overview of experimental models of TBI, HTN, and NPH showing features of tau-amyloid pathology, neuroinflammation, and neuronal loss. RESULTS "Alzheimer-like" hallmarks found in these mechanical stress-based models were compared with AD features found in transgenic models. DISCUSSION The goal of this review is, therefore, to build on current concepts of onset and progression of AD lesions. We point to the importance of accumulated mechanical stress in brain as an environmental and endogenous factor that pushes protein deposition and neuronal injury over the disease threshold. We further encourage the development of preventing strategies and drug screening based on mechanical stress models.
Collapse
Affiliation(s)
- Marcel Levy Nogueira
- Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Paris, France; Institut des Neurosciences Translationnelles de Paris (IHU-A-ICM), Institut du Cerveau et de la Moelle Epinière (ICM), Paris, France; Laboratoire d'informatique (LIX), UMR 7161, Ecole Polytechnique, Université Paris-Saclay, Palaiseau, France.
| | - Stéphane Epelbaum
- Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Paris, France; INSERM, CNRS, UMR-S975, Institut du Cerveau et de la Moelle Epinière (ICM), Paris, France; Sorbonne Universités, Université Pierre et Marie Curie, Hôpital de la Pitié-Salpêtrière, AP-HP, Paris, France
| | - Jean-Marc Steyaert
- Laboratoire d'informatique (LIX), UMR 7161, Ecole Polytechnique, Université Paris-Saclay, Palaiseau, France
| | - Bruno Dubois
- Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Département de Neurologie, Hôpital de la Pitié-Salpêtrière, AP-HP, Paris, France; Institut des Neurosciences Translationnelles de Paris (IHU-A-ICM), Institut du Cerveau et de la Moelle Epinière (ICM), Paris, France; INSERM, CNRS, UMR-S975, Institut du Cerveau et de la Moelle Epinière (ICM), Paris, France; Sorbonne Universités, Université Pierre et Marie Curie, Hôpital de la Pitié-Salpêtrière, AP-HP, Paris, France
| | - Laurent Schwartz
- Laboratoire d'informatique (LIX), UMR 7161, Ecole Polytechnique, Université Paris-Saclay, Palaiseau, France
| |
Collapse
|
30
|
Levy Nogueira M, da Veiga Moreira J, Baronzio GF, Dubois B, Steyaert JM, Schwartz L. Mechanical Stress as the Common Denominator between Chronic Inflammation, Cancer, and Alzheimer's Disease. Front Oncol 2015; 5:197. [PMID: 26442209 PMCID: PMC4585184 DOI: 10.3389/fonc.2015.00197] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 08/24/2015] [Indexed: 11/13/2022] Open
Abstract
The pathogenesis of common diseases, such as Alzheimer's disease (AD) and cancer, are currently poorly understood. Inflammation is a common risk factor for cancer and AD. Recent data, provided by our group and from others, demonstrate that increased pressure and inflammation are synonymous. There is a continuous increase in pressure from inflammation to fibrosis and then cancer. This is in line with the numerous papers reporting high interstitial pressure in cancer. But most authors focus on the role of pressure in the lack of delivery of chemotherapy in the center of the tumor. Pressure may also be a key factor in carcinogenesis. Increased pressure is responsible for oncogene activation and cytokine secretion. Accumulation of mechanical stress plays a key role in the development of diseases of old age, such as cardiomyopathy, atherosclerosis, and osteoarthritis. Growing evidence suggest also a possible link between mechanical stress in the pathogenesis of AD. The aim of this review is to describe environmental and endogenous mechanical factors possibly playing a pivotal role in the mechanism of chronic inflammation, AD, and cancer.
Collapse
Affiliation(s)
- Marcel Levy Nogueira
- Département de Neurologie, Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Hôpital de la Pitié-Salpêtrière, AP-HP , Paris , France ; Institut des Neurosciences Translationnelles de Paris (IHU-A-ICM), Institut du Cerveau et de la Moelle Epinière (ICM) , Paris , France ; UMR 7161, Laboratoire d'informatique (LIX), Ecole Polytechnique, Université Paris-Saclay , Palaiseau , France
| | - Jorgelindo da Veiga Moreira
- UMR 7161, Laboratoire d'informatique (LIX), Ecole Polytechnique, Université Paris-Saclay , Palaiseau , France
| | | | - Bruno Dubois
- Département de Neurologie, Institut de la Mémoire et de la Maladie d'Alzheimer (IM2A), Hôpital de la Pitié-Salpêtrière, AP-HP , Paris , France ; Institut des Neurosciences Translationnelles de Paris (IHU-A-ICM), Institut du Cerveau et de la Moelle Epinière (ICM) , Paris , France ; UMR-S975, CNRS, INSERM, Institut du Cerveau et de la Moelle Epinière (ICM) , Paris , France
| | - Jean-Marc Steyaert
- UMR 7161, Laboratoire d'informatique (LIX), Ecole Polytechnique, Université Paris-Saclay , Palaiseau , France
| | - Laurent Schwartz
- UMR 7161, Laboratoire d'informatique (LIX), Ecole Polytechnique, Université Paris-Saclay , Palaiseau , France
| |
Collapse
|
31
|
The Role of MAPT in Neurodegenerative Diseases: Genetics, Mechanisms and Therapy. Mol Neurobiol 2015; 53:4893-904. [PMID: 26363795 DOI: 10.1007/s12035-015-9415-8] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2015] [Accepted: 09/01/2015] [Indexed: 12/11/2022]
Abstract
Microtubule-associated protein tau (MAPT) is a gene responsible for encoding tau protein, which is tightly implicated in keeping the function of microtubules and axonal transport. Hyperphosphorylated tau protein participates in the formation of neurofibrillary tangles (NFTs), which characterize many neurodegenerative disorders termed tauopathies. Genome-wide association studies (GWAS) have demonstrated numerous single nucleotide polymorphisms (SNPs) located in MAPT associated with various neurodegenerative diseases. Thus, it has been presumed that MAPT plays a crucial role in pathogenesis of neurodegeneration via affecting the structure and function of tau. Here, we review the advanced studies to summarize the biochemical properties of MAPT and its encoded protein, as well as the genetics and epigenetics of MAPT in neurodegeneration. Finally, given the potential mechanisms of MAPT to neurodegeneration pathogenesis, targeting MAPT and tau might present significant treatments of MAPT mutation-related neurodegeneration. Affirmatively, the identification of MAPT is extremely beneficial for improving our understanding of the pathogenesis of various neurodegenerative diseases and developing the mechanism-based therapies.
Collapse
|
32
|
Smart D, Garcia-Glaessner A, Palmieri D, Wong-Goodrich SJ, Kramp T, Gril B, Shukla S, Lyle T, Hua E, Cameron HA, Camphausen K, Steeg PS. Analysis of radiation therapy in a model of triple-negative breast cancer brain metastasis. Clin Exp Metastasis 2015; 32:717-27. [PMID: 26319493 DOI: 10.1007/s10585-015-9739-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 08/11/2015] [Indexed: 11/25/2022]
Abstract
Most cancer patients with brain metastases are treated with radiation therapy, yet this modality has not yet been meaningfully incorporated into preclinical experimental brain metastasis models. We applied two forms of whole brain radiation therapy (WBRT) to the brain-tropic 231-BR experimental brain metastasis model of triple-negative breast cancer. When compared to sham controls, WBRT as 3 Gy × 10 fractions (3 × 10) reduced the number of micrometastases and large metastases by 87.7 and 54.5 %, respectively (both p < 0.01); whereas a single radiation dose of 15 Gy × 1 (15 × 1) was less effective, reducing metastases by 58.4 % (p < 0.01) and 47.1 % (p = 0.41), respectively. Neuroinflammation in the adjacent brain parenchyma was due solely to a reaction from metastases, and not radiotherapy, while adult neurogenesis in brains was adversely affected following both radiation regimens. The nature of radiation resistance was investigated by ex vivo culture of tumor cells that survived initial WBRT ("Surviving" cultures). The Surviving cultures surprisingly demonstrated increased radiosensitivity ex vivo. In contrast, re-injection of Surviving cultures and re-treatment with a 3 × 10 WBRT regimen significantly reduced the number of large and micrometastases that developed in vivo, suggesting a role for the microenvironment. Micrometastases derived from tumor cells surviving initial 3 × 10 WBRT demonstrated a trend toward radioresistance upon repeat treatment (p = 0.09). The data confirm the potency of a fractionated 3 × 10 WBRT regimen and identify the brain microenvironment as a potential determinant of radiation efficacy. The data also nominate the Surviving cultures as a potential new translational model for radiotherapy.
Collapse
Affiliation(s)
- DeeDee Smart
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Building 10, Room B3B69, 10 Center Dr., Bethesda, MD, 20892, USA.
| | - Alejandra Garcia-Glaessner
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Building 10, Room B3B69, 10 Center Dr., Bethesda, MD, 20892, USA
| | - Diane Palmieri
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bldg 37, Rm. 1126, Bethesda, MD, 20892, USA
- NHLBI, Bldg 10-CRC, 10 Center Dr., Bethesda, MD, 20892, USA
| | | | - Tamalee Kramp
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Building 10, Room B3B69, 10 Center Dr., Bethesda, MD, 20892, USA
| | - Brunilde Gril
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bldg 37, Rm. 1126, Bethesda, MD, 20892, USA
| | - Sudhanshu Shukla
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Building 10, Room B3B69, 10 Center Dr., Bethesda, MD, 20892, USA
| | - Tiffany Lyle
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bldg 37, Rm. 1126, Bethesda, MD, 20892, USA
| | - Emily Hua
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bldg 37, Rm. 1126, Bethesda, MD, 20892, USA
| | - Heather A Cameron
- Section on Neuroplasticity, NIMH, NIH, 35 Convent Dr., Bethesda, MD, 20892, USA
| | - Kevin Camphausen
- Radiation Oncology Branch, Center for Cancer Research, National Cancer Institute, NIH, Building 10, Room B3B69, 10 Center Dr., Bethesda, MD, 20892, USA
| | - Patricia S Steeg
- Women's Malignancies Branch, Center for Cancer Research, National Cancer Institute, NIH, Bldg 37, Rm. 1126, Bethesda, MD, 20892, USA.
| |
Collapse
|
33
|
Liu W, Yamashita T, Kurata T, Kono S, Hishikawa N, Deguchi K, Zhai Y, Abe K. Protective effect of telmisartan on neurovascular unit and inflammasome in stroke-resistant spontaneously hypertensive rats. Neurol Res 2015; 37:491-501. [PMID: 25591419 DOI: 10.1179/1743132815y.0000000002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVES Hypertension is a crucial risk factor for both stroke and dementia, including Alzheimer's disease (AD). We inspected the effect of telmisartan on the neurovascular unit (NVU) and related inflammatory responses in spontaneously hypertensive rat stroke resistant (SHR-SR) by observing the components of NVU such as N-acetyl glucosamine oligomer (NAGO), collagen IV, astrocytes, and matrix metalloproteinase-9 (MMP-9), as well as inflammasome NOD-like receptors family protein 3 (NLRP3). METHODS In the present study, we examined the effect of a highly selective angiotensin type 1 (AT-1) antagonist of angiotensin 2 receptor with high lipid solubility, telmisartan, on NVU and related inflammatory responses in SHR-SR with a low dose (0.3 mg/kg/day) only for improving metabolic syndrome, and a high dose (3 mg/kg/day) for improving both metabolic syndrome and SHR-SR hypertension. RESULTS Compared to normotensive Wistar rats, long-lasting hypertension in SHR-SR disrupted NVU by changing immunohistological components such as NAGO, collagen IV, astrocytes, and MMP-9. SHR-SR also strongly induced AD-related inflammasome NLRP3 in neuronal cells with age. However, such NVU disruption and inflammasome activation were greatly improved with dose-dependent telmisartan treatments. DISCUSSION These results suggest that telmisartan comprehensively protected the NVU components by reducing inflammatory reactions relative to AD in hypertensive rats, which could also preclude the risk of AD under hypertension.
Collapse
|