1
|
Reeve EH, Barnes JN, Moir ME, Walker AE. Impact of arterial stiffness on cerebrovascular function: a review of evidence from humans and preclincal models. Am J Physiol Heart Circ Physiol 2024; 326:H689-H704. [PMID: 38214904 PMCID: PMC11221809 DOI: 10.1152/ajpheart.00592.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/08/2023] [Accepted: 01/08/2024] [Indexed: 01/13/2024]
Abstract
With advancing age, the cerebral vasculature becomes dysfunctional, and this dysfunction is associated with cognitive decline. However, the initiating cause of these age-related cerebrovascular impairments remains incompletely understood. A characteristic feature of the aging vasculature is the increase in stiffness of the large elastic arteries. This increase in arterial stiffness is associated with elevated pulse pressure and blood flow pulsatility in the cerebral vasculature. Evidence from both humans and rodents supports that increases in large elastic artery stiffness are associated with cerebrovascular impairments. These impacts on cerebrovascular function are wide-ranging and include reductions in global and regional cerebral blood flow, cerebral small vessel disease, endothelial cell dysfunction, and impaired perivascular clearance. Furthermore, recent findings suggest that the relationship between arterial stiffness and cerebrovascular function may be influenced by genetics, specifically APOE and NOTCH genotypes. Given the strength of the evidence that age-related increases in arterial stiffness have deleterious impacts on the brain, interventions that target arterial stiffness are needed. The purpose of this review is to summarize the evidence from human and rodent studies, supporting the role of increased arterial stiffness in age-related cerebrovascular impairments.
Collapse
Affiliation(s)
- Emily H Reeve
- Department of Human Physiology, University of Oregon, Eugene, Oregon, United States
| | - Jill N Barnes
- Department of Kinesiology University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - M Erin Moir
- Department of Kinesiology University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Ashley E Walker
- Department of Human Physiology, University of Oregon, Eugene, Oregon, United States
| |
Collapse
|
2
|
Baggeroer CE, Cambronero FE, Savan NA, Jefferson AL, Santisteban MM. Basic Mechanisms of Brain Injury and Cognitive Decline in Hypertension. Hypertension 2024; 81:34-44. [PMID: 37732479 PMCID: PMC10840624 DOI: 10.1161/hypertensionaha.123.19939] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2023]
Abstract
Dementia affects almost 50 million adults worldwide, and remains a major cause of death and disability. Hypertension is a leading risk factor for dementia, including Alzheimer disease and Alzheimer disease-related dementias. Although this association is well-established, the mechanisms underlying hypertension-induced cognitive decline remain poorly understood. By exploring the mechanisms mediating the detrimental effects of hypertension on the brain, studies have aimed to provide therapeutic insights and strategies on how to protect the brain from the effects of blood pressure elevation. In this review, we focus on the basic mechanisms contributing to the cerebrovascular adaptions to elevated blood pressure and hypertension-induced microvascular injury. We also assess the cellular mechanisms of neurovascular unit dysfunction, focusing on the premise that cognitive impairment ensues when the dynamic metabolic demands of neurons are not met due to neurovascular uncoupling, and summarize cognitive deficits across various rodent models of hypertension as a resource for investigators. Despite significant advances in antihypertensive therapy, hypertension remains a critical risk factor for cognitive decline, and several questions remain about the development and progression of hypertension-induced cognitive impairment.
Collapse
Affiliation(s)
- Caroline E. Baggeroer
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN
| | - Francis E. Cambronero
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN
| | - N. Anna Savan
- Medical Scientist Training Program, Yale University, New Haven, CT
| | - Angela L. Jefferson
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Monica M. Santisteban
- Vanderbilt Memory and Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
3
|
Baskaran K, Johnson JT, Prem PN, Ravindran S, Kurian GA. Evaluation of prophylactic efficacy of sodium thiosulfate in combating I/R injury in rat brain: exploring its efficiency further in vascular calcified brain slice model. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:2587-2598. [PMID: 37058187 DOI: 10.1007/s00210-023-02481-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 03/30/2023] [Indexed: 04/15/2023]
Abstract
Cerebral ischemia reperfusion injury (CIR) is one of the clinical manifestations encountered during the management of stroke. High prevalence of intracranial arterial calcification is reported in stroke patients. However, the impact of vascular calcification (VC) in the outcome of CIR and the efficacy of mechanical preconditioning (IPC) and pharmacological conditioning with sodium thiosulphate (STS) in ameliorating IR remains unclear. Two experimental models namely carotid artery occlusion (n = 36) and brain slice models (n = 18) were used to evaluate the efficacy of STS in male Wistar rats. IR was inflicted in rat by occluding carotid artery for 30 min followed by 24-h reperfusion after STS (100 mg/kg) administration. Brain slice model was used to reconfirm the results to account blood brain barrier permeability. Further, brain slice tissue was utilised to evaluate the efficacy of STS in VC rat brain by measuring the histological alterations and biochemical parameters. Pre-treatment of STS prior to CIR in intact animal significantly reduced the IR-associated histopathological alterations in brain, declined oxidative stress and improved the mitochondrial function found to be similar to IPC. Brain slice model data also confirmed the neuroprotective effect of STS similar to IPC in IR challenged tissue slice. Higher tissue injury was noted in VC brain IR tissue than normal IR tissue. Therapeutic efficacy of STS was evident in VC rat brain tissues and normal tissues subjected to IR. On the other hand, IPC-mediated protection was noted only in IR normal and adenine-induced VC brain tissues not in high-fat diet (HFD) induced VC brain tissues. Based on the results, we concluded that similar to IPC, STS was effective in attenuating IR injury in CIR rat brain. Vascular calcification adversely affected the recovery protocol of brain tissues from ischemic insult. STS was found to be an effective agent in ameliorating the IR injury in both adenine and HFD induced vascular calcified rat brain, but IPC-mediated neuroprotection was absent in HFD-induced VC brain tissues.
Collapse
Affiliation(s)
- Keerthana Baskaran
- Vascular Biology Lab, SASTRA Deemed University, 117, Anusandhan Kendra, TirumalaisamudramThanjavur, 613401, Tamil Nadu, India
| | - Jefri Thimoathi Johnson
- Vascular Biology Lab, SASTRA Deemed University, 117, Anusandhan Kendra, TirumalaisamudramThanjavur, 613401, Tamil Nadu, India
| | - Priyanka N Prem
- Vascular Biology Lab, SASTRA Deemed University, 117, Anusandhan Kendra, TirumalaisamudramThanjavur, 613401, Tamil Nadu, India
- School of Chemical and Biotechnology, SASTRA Deemed University, TirumalaisamudramThanjavur, 613401, Tamil Nadu, India
| | - Sriram Ravindran
- Vascular Biology Lab, SASTRA Deemed University, 117, Anusandhan Kendra, TirumalaisamudramThanjavur, 613401, Tamil Nadu, India
| | - Gino A Kurian
- Vascular Biology Lab, SASTRA Deemed University, 117, Anusandhan Kendra, TirumalaisamudramThanjavur, 613401, Tamil Nadu, India.
- School of Chemical and Biotechnology, SASTRA Deemed University, TirumalaisamudramThanjavur, 613401, Tamil Nadu, India.
| |
Collapse
|
4
|
Reeve EH, Kronquist EK, Wolf JR, Lee B, Khurana A, Pham H, Cullen AE, Peterson JA, Meza A, Colton Bramwell R, Villasana L, Machin DR, Henson GD, Walker AE. Pyridoxamine treatment ameliorates large artery stiffening and cerebral artery endothelial dysfunction in old mice. J Cereb Blood Flow Metab 2023; 43:281-295. [PMID: 36189840 PMCID: PMC9903220 DOI: 10.1177/0271678x221130124] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Age-related increases in large artery stiffness are associated with cerebrovascular dysfunction and cognitive impairment. Pyridoxamine treatment prevents large artery stiffening with advancing age, but the effects of pyridoxamine treatment on the cerebral vasculature or cognition is unknown. The purpose of this study was to investigate the effects of pyridoxamine on blood pressure, large artery stiffness, cerebral artery function, and cognitive function in old mice. Old male C57BL/6 mice consumed either pyridoxamine (2 g/L) or vehicle control in drinking water for ∼7.5 months and were compared with young male C57BL/6 mice. From pre- to post-treatment, systolic blood pressure increased in old control mice, but was maintained in pyridoxamine treated mice. Large artery stiffness decreased in pyridoxamine-treated mice but was unaffected in control mice. Pyridoxamine-treated mice had greater cerebral artery endothelium-dependent dilation compared with old control mice, and not different from young mice. Old control mice had impaired cognitive function; however, pyridoxamine only partially preserved cognitive function in old mice. In summary, pyridoxamine treatment in old mice prevented age-related increases in blood pressure, reduced large artery stiffness, preserved cerebral artery endothelial function, and partially preserved cognitive function. Taken together, these results suggest that pyridoxamine treatment may limit vascular aging.
Collapse
Affiliation(s)
- Emily H Reeve
- Department of Human Physiology, 3265, University of Oregon, Eugene, OR, USA
| | - Elise K Kronquist
- Department of Human Physiology, 3265, University of Oregon, Eugene, OR, USA
| | - Julia R Wolf
- Department of Human Physiology, 3265, University of Oregon, Eugene, OR, USA
| | - Byron Lee
- Department of Human Physiology, 3265, University of Oregon, Eugene, OR, USA
| | - Aleena Khurana
- Department of Human Physiology, 3265, University of Oregon, Eugene, OR, USA
| | - Hanson Pham
- Department of Human Physiology, 3265, University of Oregon, Eugene, OR, USA
| | - Abigail E Cullen
- Department of Human Physiology, 3265, University of Oregon, Eugene, OR, USA
| | - Jessica A Peterson
- Department of Human Physiology, 3265, University of Oregon, Eugene, OR, USA
| | - Antonio Meza
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - R Colton Bramwell
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | | | - Daniel R Machin
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
- Department of Nutrition and Integrative Physiology, 7823, Florida State University, Tallahassee, FL, USA
| | - Grant D Henson
- Department of Human Physiology, 3265, University of Oregon, Eugene, OR, USA
| | - Ashley E Walker
- Department of Human Physiology, 3265, University of Oregon, Eugene, OR, USA
- Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
5
|
Malone JE, Elkasaby MI, Lerner AJ. Effects of Hypertension on Alzheimer's Disease and Related Disorders. Curr Hypertens Rep 2022; 24:615-625. [PMID: 36125695 DOI: 10.1007/s11906-022-01221-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2022] [Indexed: 01/31/2023]
Abstract
PURPOSE OF REVIEW To review the pathophysiology of hypertension in Alzheimer's disease and related dementias and explore the current landscape of clinical trials involving treatment of hypertension to improve cognition. RECENT FINDINGS Hypertension is increasingly recognized as a contributor to cognitive impairment. Clinical trials that explore blood pressure reductions with cognitive outcomes have been promising. Various antihypertensives have been evaluated in clinical trials, with growing interest in those agents that impact the renin-angiotensin-aldosterone system due to its own association with cognitive impairment. No antihypertensive agent has been found to be superior to others in reducing cognitive impairment risk or conferring neuroprotective benefits. In this review, the pathophysiology of and clinical trial data involving hypertension and dementia will be explored. Hypertension is a significant risk factor for the development of neurodegenerative dementias, and clinical trials have been overall favorable in improving cognition by reductions in blood pressure using antihypertensive agents.
Collapse
Affiliation(s)
- Joseph E Malone
- Department of Neurology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Mohamed I Elkasaby
- Department of Neurology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Alan J Lerner
- Department of Neurology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA.
- Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
6
|
Huo R, Liu Y, Xu H, Li J, Xin R, Xing Z, Deng S, Wang T, Yuan H, Zhao X. Associations between carotid atherosclerotic plaque characteristics determined by magnetic resonance imaging and improvement of cognition in patients undergoing carotid endarterectomy. Quant Imaging Med Surg 2022; 12:2891-2903. [PMID: 35502372 PMCID: PMC9014142 DOI: 10.21037/qims-21-981] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 02/05/2022] [Indexed: 08/29/2023]
Abstract
BACKGROUND To determine the predictive value of carotid plaque characteristics for the improvement of cognition in patients with moderate-to-severe carotid stenosis after carotid endarterectomy (CEA), using vessel wall magnetic resonance imaging (MRI). METHODS This was a prospective cohort study. Patients with unilateral, moderate-to-severe carotid stenosis referred to the Peking University Third Hospital for CEA were prospectively recruited and underwent carotid vessel wall MRI within 1 week before CEA. We performed Montreal Cognitive Assessment (MoCA) within 1 week before and 3-4 days after CEA. The morphological and compositional characteristics of carotid plaques on MRI were evaluated. Improvement of cognition was defined as >10% increase of the total MoCA score after CEA compared with baseline. Carotid plaque characteristics were compared between patients with and without cognitive improvement. RESULTS In total, 105 patients (91 males; mean age, 65.5±8.4 years) were included. The volume {48.0 [interquartile range (IQR), 21.0 to 91.6] vs. 16.3 (IQR, 8.1 to 53.1) mm3; P=0.005} and cumulative slice [4.0 (IQR, 3.0 to 7.0) vs. 3.0 (IQR, 2.0 to 5.0); P=0.019] of carotid calcification, and maximum percentage of calcification area [13.1% (IQR, 6.0% to 19.8%) vs. 6.2% (IQR, 3.7% to 10.8%); P=0.004] were significantly smaller in participants with cognitive improvement compared to those without. Univariate logistic regression analysis showed that volume [odds ratio (OR) =0.994; 95% confidence interval (CI): 0.989 to 1.000; P=0.043] and cumulative slice (OR =0.823; 95% CI: 0.698 to 0.970; P=0.020) of carotid calcification, and maximum percentage of calcification area (OR =0.949; 95% CI: 0.909 to 0.991; P=0.018) were significantly correlated with cognitive improvement. After adjusting for confounding factors, these associations remained statistically or marginally significant (volume: OR =0.994; 95% CI: 0.988 to 1.000; P=0.057; maximum percentage of calcification area: OR =0.937; 95% CI: 0.890 to 0.987; P=0.014; and cumulative slice: OR =0.791; 95% CI: 0.646 to 0.967; P=0.022). No significant associations were found between other plaque characteristics and cognitive improvement (all P>0.05). CONCLUSIONS More than half of the participants with unilateral, moderate-to-severe carotid atherosclerotic stenosis had cognitive improvement. The size of calcification might be an effective indicator of cognitive improvement after CEA.
Collapse
Affiliation(s)
- Ran Huo
- Department of Radiology, Peking University Third Hospital, Beijing, China
| | - Ying Liu
- Department of Radiology, Peking University Third Hospital, Beijing, China
| | - Huimin Xu
- Department of Radiology, Peking University Third Hospital, Beijing, China
| | - Jin Li
- Department of Radiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Ruijing Xin
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Zhangli Xing
- Department of Radiology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Shasha Deng
- School of Medical Imaging, Changsha Medical University, Changsha, China
| | - Tao Wang
- Department of Neurosurgery, Peking University Third Hospital, Beijing, China
| | - Huishu Yuan
- Department of Radiology, Peking University Third Hospital, Beijing, China
| | - Xihai Zhao
- Center for Biomedical Imaging Research, Department of Biomedical Engineering, School of Medicine, Tsinghua University, Beijing, China
| |
Collapse
|
7
|
Kehmeier MN, Walker AE. Sex Differences in Large Artery Stiffness: Implications for Cerebrovascular Dysfunction and Alzheimer’s Disease. FRONTIERS IN AGING 2021; 2. [PMID: 35072153 PMCID: PMC8782423 DOI: 10.3389/fragi.2021.791208] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
Two in every three Alzheimer’s disease diagnoses are females, calling attention to the need to understand sexual dimorphisms with aging and neurodegenerative disease progression. Dysfunction and damage to the vasculature with aging are strongly linked to Alzheimer’s disease. With aging there is an increase in stiffness of the large elastic arteries, and this stiffening is associated with cerebrovascular dysfunction and cognitive impairment. However, it is unclear how the deleterious effects of arterial stiffness may differ between females and males. While environmental, chromosomal, and sex hormone factors influence aging, there is evidence that the deficiency of estrogen post-menopause in females is a contributor to vascular aging and Alzheimer’s disease progression. The purpose of this mini review is to describe the recent developments in our understanding of sex differences in large artery stiffness, cerebrovascular dysfunction, and cognitive impairment, and their intricate relations. Furthermore, we will focus on the impact of the loss of estrogen post-menopause as a potential driving factor for these outcomes. Overall, a better understanding of how sex differences influence aging physiology is crucial to the prevention and treatment of neurodegenerative diseases.
Collapse
|
8
|
Youwakim J, Girouard H. Inflammation: A Mediator Between Hypertension and Neurodegenerative Diseases. Am J Hypertens 2021; 34:1014-1030. [PMID: 34136907 DOI: 10.1093/ajh/hpab094] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 05/03/2021] [Accepted: 06/15/2021] [Indexed: 12/14/2022] Open
Abstract
Hypertension is the most prevalent and modifiable risk factor for stroke, vascular cognitive impairment, and Alzheimer's disease. However, the mechanistic link between hypertension and neurodegenerative diseases remains to be understood. Recent evidence indicates that inflammation is a common pathophysiological trait for both hypertension and neurodegenerative diseases. Low-grade chronic inflammation at the systemic and central nervous system levels is now recognized to contribute to the physiopathology of hypertension. This review speculates that inflammation represents a mediator between hypertension and neurodegenerative diseases, either by a decrease in cerebral blood flow or a disruption of the blood-brain barrier which will, in turn, let inflammatory cells and neurotoxic molecules enter the brain parenchyma. This may impact brain functions including cognition and contribute to neurodegenerative diseases. This review will thus discuss the relationship between hypertension, systemic inflammation, cerebrovascular functions, neuroinflammation, and brain dysfunctions. The potential clinical future of immunotherapies against hypertension and associated cerebrovascular risks will also be presented.
Collapse
Affiliation(s)
- Jessica Youwakim
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
- Centre interdisciplinaire de recherche sur le cerveau et l’apprentissage (CIRCA); Montreal, QC, Canada
- Groupe de Recherche sur le Système Nerveux Central, Montreal, QC, Canada
| | - Hélène Girouard
- Département de Pharmacologie et Physiologie, Université de Montréal, Montreal, QC, Canada
- Centre interdisciplinaire de recherche sur le cerveau et l’apprentissage (CIRCA); Montreal, QC, Canada
- Groupe de Recherche sur le Système Nerveux Central, Montreal, QC, Canada
- Centre de recherche de l’Institut Universitaire de Gériaterie de Montréal, Montreal, QC, Canada
| |
Collapse
|
9
|
Ungvari Z, Toth P, Tarantini S, Prodan CI, Sorond F, Merkely B, Csiszar A. Hypertension-induced cognitive impairment: from pathophysiology to public health. Nat Rev Nephrol 2021; 17:639-654. [PMID: 34127835 PMCID: PMC8202227 DOI: 10.1038/s41581-021-00430-6] [Citation(s) in RCA: 273] [Impact Index Per Article: 68.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2021] [Indexed: 02/06/2023]
Abstract
Hypertension affects two-thirds of people aged >60 years and significantly increases the risk of both vascular cognitive impairment and Alzheimer's disease. Hypertension compromises the structural and functional integrity of the cerebral microcirculation, promoting microvascular rarefaction, cerebromicrovascular endothelial dysfunction and neurovascular uncoupling, which impair cerebral blood supply. In addition, hypertension disrupts the blood-brain barrier, promoting neuroinflammation and exacerbation of amyloid pathologies. Ageing is characterized by multifaceted homeostatic dysfunction and impaired cellular stress resilience, which exacerbate the deleterious cerebromicrovascular effects of hypertension. Neuroradiological markers of hypertension-induced cerebral small vessel disease include white matter hyperintensities, lacunar infarcts and microhaemorrhages, all of which are associated with cognitive decline. Use of pharmaceutical and lifestyle interventions that reduce blood pressure, in combination with treatments that promote microvascular health, have the potential to prevent or delay the pathogenesis of vascular cognitive impairment and Alzheimer's disease in patients with hypertension.
Collapse
Affiliation(s)
- Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Peter Toth
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Neurosurgery, Medical School, University of Pecs, Pecs, Hungary
| | - Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Calin I Prodan
- Department of Neurology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Farzaneh Sorond
- Department of Neurology, Division of Stroke and Neurocritical Care, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Bela Merkely
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Institute of Clinical Experimental Research, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
10
|
Baradaran H, Delic A, McNally JS, Alexander M, Majersik JJ, Parker DL, de Havenon A. Carotid Compliance and Parahippocampal and Hippocampal Volume over a 20-Year Period. Dement Geriatr Cogn Dis Extra 2021; 11:227-234. [PMID: 34721500 PMCID: PMC8543351 DOI: 10.1159/000518234] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 06/30/2021] [Indexed: 11/21/2022] Open
Abstract
INTRODUCTION We evaluated the association between carotid compliance, a measure of arterial stiffness, to parahippocampal volume (PHV) and hippocampal volume (HV) over 20 years later in the Atherosclerosis Risk in the Community study. METHODS We included participants with common carotid compliance measurements at visit 1 (1987-1989) and volumetric brain MRI at visit 5 (2011-2013). The primary outcomes are pooled bilateral PHV and HV. We performed linear regression models adjusting for age, sex, vascular risk factors, and total brain volume. RESULTS Of the 614 participants, higher compliance was correlated with higher PHV (R = 0.218[0.144-0.291], p < 0.001) and HV (R = 0.181 [0.105-0.255, p < 0.001]). The association was linear and significant after adjusting for confounders. At follow-up MRI, 30 patients with dementia had lower PHV and HV than patients without dementia (p < 0.001 and p < 0.001, respectively). CONCLUSION Carotid compliance is associated with higher PHV and HV when measured 20 years later, further supporting the link between arterial stiffness and cognitive decline.
Collapse
Affiliation(s)
- Hediyeh Baradaran
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, Utah, USA
| | - Alen Delic
- Department of Neurology, University of Utah, Salt Lake City, Utah, USA
| | - J. Scott McNally
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, Utah, USA
| | - Matthew Alexander
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, Utah, USA
| | | | - Dennis L. Parker
- Department of Radiology and Imaging Sciences, University of Utah, Salt Lake City, Utah, USA
| | - Adam de Havenon
- Department of Neurology, University of Utah, Salt Lake City, Utah, USA
| |
Collapse
|
11
|
Winder NR, Reeve EH, Walker AE. Large artery stiffness and brain health: insights from animal models. Am J Physiol Heart Circ Physiol 2020; 320:H424-H431. [PMID: 33164578 DOI: 10.1152/ajpheart.00696.2020] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
There are no effective treatments available to halt or reverse the progression of age-related cognitive decline and Alzheimer's disease. Thus, there is an urgent need to understand the underlying mechanisms of disease etiology and progression to identify novel therapeutic targets. Age-related changes to the vasculature, particularly increases in stiffness of the large elastic arteries, are now recognized as important contributors to brain aging. There is a growing body of evidence for an association between greater large artery stiffness and cognitive impairment among both healthy older adults and patients with Alzheimer's disease. However, studies in humans are limited to only correlative evidence, whereas animal models allow researchers to explore the causative mechanisms linking arterial stiffness to neurocognitive dysfunction and disease. Recently, several rodent models of direct modulation of large artery stiffness and the consequent effects on the brain have been reported. Common outcomes among these models have emerged, including evidence that greater large artery stiffness causes cerebrovascular dysfunction associated with increased oxidative stress and inflammatory signaling. The purpose of this mini-review is to highlight the recent findings associating large artery stiffness with deleterious brain outcomes, with a specific focus on causative evidence obtained from animal models. We will also discuss the gaps in knowledge that remain in our understanding of how large artery stiffness affects brain function and disease outcomes.
Collapse
Affiliation(s)
- Nick R Winder
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| | - Emily H Reeve
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| | - Ashley E Walker
- Department of Human Physiology, University of Oregon, Eugene, Oregon
| |
Collapse
|
12
|
Presa JL, Saravia F, Bagi Z, Filosa JA. Vasculo-Neuronal Coupling and Neurovascular Coupling at the Neurovascular Unit: Impact of Hypertension. Front Physiol 2020; 11:584135. [PMID: 33101063 PMCID: PMC7546852 DOI: 10.3389/fphys.2020.584135] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 09/04/2020] [Indexed: 12/18/2022] Open
Abstract
Components of the neurovascular unit (NVU) establish dynamic crosstalk that regulates cerebral blood flow and maintain brain homeostasis. Here, we describe accumulating evidence for cellular elements of the NVU contributing to critical physiological processes such as cerebral autoregulation, neurovascular coupling, and vasculo-neuronal coupling. We discuss how alterations in the cellular mechanisms governing NVU homeostasis can lead to pathological changes in which vascular endothelial and smooth muscle cell, pericyte and astrocyte function may play a key role. Because hypertension is a modifiable risk factor for stroke and accelerated cognitive decline in aging, we focus on hypertension-associated changes on cerebral arteriole function and structure, and the molecular mechanisms through which these may contribute to cognitive decline. We gather recent emerging evidence concerning cognitive loss in hypertension and the link with vascular dementia and Alzheimer’s disease. Collectively, we summarize how vascular dysfunction, chronic hypoperfusion, oxidative stress, and inflammatory processes can uncouple communication at the NVU impairing cerebral perfusion and contributing to neurodegeneration.
Collapse
Affiliation(s)
- Jessica L Presa
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States.,Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - Flavia Saravia
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires and Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - Zsolt Bagi
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Jessica A Filosa
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, United States
| |
Collapse
|
13
|
Fedintsev A, Moskalev A. Stochastic non-enzymatic modification of long-lived macromolecules - A missing hallmark of aging. Ageing Res Rev 2020; 62:101097. [PMID: 32540391 DOI: 10.1016/j.arr.2020.101097] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/05/2020] [Accepted: 06/04/2020] [Indexed: 12/12/2022]
Abstract
Damage accumulation in long-living macromolecules (especially extracellular matrix (ECM) proteins, nuclear pore complex (NPC) proteins, and histones) is a missing hallmark of aging. Stochastic non-enzymatic modifications of ECM trigger cellular senescence as well as many other hallmarks of aging affect organ barriers integrity and drive tissue fibrosis. The importance of it for aging makes it a key target for interventions. The most promising of them can be AGE inhibitors (chelators, O-acetyl group or transglycating activity compounds, amadorins and amadoriases), glucosepane breakers, stimulators of elastogenesis, and RAGE antagonists.
Collapse
Affiliation(s)
- Alexander Fedintsev
- Institute of Biology of FRC of Komi Scientific Center, Ural Branch of Russian Academy of Sciences, Syktyvkar, Russia
| | - Alexey Moskalev
- Institute of Biology of FRC of Komi Scientific Center, Ural Branch of Russian Academy of Sciences, Syktyvkar, Russia.
| |
Collapse
|
14
|
Muhire G, Iulita MF, Vallerand D, Youwakim J, Gratuze M, Petry FR, Planel E, Ferland G, Girouard H. Arterial Stiffness Due to Carotid Calcification Disrupts Cerebral Blood Flow Regulation and Leads to Cognitive Deficits. J Am Heart Assoc 2020; 8:e011630. [PMID: 31057061 PMCID: PMC6512142 DOI: 10.1161/jaha.118.011630] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Background Arterial stiffness is associated with cognitive decline and dementia; however, the precise mechanisms by which it affects the brain remain unclear. Methods and Results Using a mouse model based on carotid calcification this study characterized mechanisms that could contribute to brain degeneration due to arterial stiffness. At 2 weeks postcalcification, carotid stiffness attenuated resting cerebral blood flow in several brain regions including the perirhinal/entorhinal cortex, hippocampus, and thalamus, determined by autoradiography (P<0.05). Carotid calcification impaired cerebral autoregulation and diminished cerebral blood flow responses to neuronal activity and to acetylcholine, examined by laser Doppler flowmetry (P<0.05, P<0.01). Carotid stiffness significantly affected spatial memory at 3 weeks (P<0.05), but not at 2 weeks, suggesting that cerebrovascular impairments precede cognitive dysfunction. In line with the endothelial deficits, carotid stiffness led to increased blood‐brain barrier permeability in the hippocampus (P<0.01). This region also exhibited reductions in vessel number containing collagen IV (P<0.01), as did the somatosensory cortex (P<0.05). No evidence of cerebral microhemorrhages was present. Carotid stiffness did not affect the production of mouse amyloid‐β (Aβ) or tau phosphorylation, although it led to a modest increase in the Aβ40/Aβ42 ratio in frontal cortex (P<0.01). Conclusions These findings suggest that carotid stiffness alters brain microcirculation and increases blood‐brain barrier permeability associated with cognitive impairments. Therefore, arterial stiffness should be considered a relevant target to protect the brain and prevent cognitive dysfunctions.
Collapse
Affiliation(s)
- Gervais Muhire
- 1 Département de Pharmacologie et Physiologie Université de Montréal Québec Canada
| | - M Florencia Iulita
- 2 Groupe de Recherche sur le Système Nerveux Central Université de Montréal Québec Canada.,3 Département de Neurosciences Université de Montréal Québec Canada
| | - Diane Vallerand
- 1 Département de Pharmacologie et Physiologie Université de Montréal Québec Canada
| | - Jessica Youwakim
- 1 Département de Pharmacologie et Physiologie Université de Montréal Québec Canada
| | - Maud Gratuze
- 4 Département de Psychiatrie et Neurosciences Université Laval Québec Québec Canada
| | - Franck R Petry
- 4 Département de Psychiatrie et Neurosciences Université Laval Québec Québec Canada
| | - Emmanuel Planel
- 4 Département de Psychiatrie et Neurosciences Université Laval Québec Québec Canada.,5 Centre de Recherche du CHU de Québec Québec Canada
| | - Guylaine Ferland
- 6 Département de Nutrition Université de Montréal Québec Canada.,7 Centre de Recherche de l'Institut de Cardiologie de Montréal Montréal Québec Canada
| | - Hélène Girouard
- 1 Département de Pharmacologie et Physiologie Université de Montréal Québec Canada.,2 Groupe de Recherche sur le Système Nerveux Central Université de Montréal Québec Canada.,8 Centre de Recherche de l'Institut Universitaire de Gériatrie de Montréal Montréal Québec Canada
| |
Collapse
|
15
|
de Montgolfier O, Thorin-Trescases N, Thorin E. Pathological Continuum From the Rise in Pulse Pressure to Impaired Neurovascular Coupling and Cognitive Decline. Am J Hypertens 2020; 33:375-390. [PMID: 32202623 PMCID: PMC7188799 DOI: 10.1093/ajh/hpaa001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 12/11/2019] [Accepted: 03/03/2020] [Indexed: 12/12/2022] Open
Abstract
The "biomechanical hypothesis" stipulates that with aging, the cumulative mechanical damages to the cerebral microvasculature, magnified by risk factors for vascular diseases, contribute to a breach in cerebral homeostasis producing neuronal losses. In other words, vascular dysfunction affects brain structure and function, and leads to cognitive failure. This is gathered under the term Vascular Cognitive Impairment and Dementia (VCID). One of the main culprits in the occurrence of cognitive decline could be the inevitable rise in arterial pulse pressure due to the age-dependent stiffening of large conductance arteries like the carotids, which in turn, could accentuate the penetration of the pulse pressure wave deeper into the fragile microvasculature of the brain and damage it. In this review, we will discuss how and why the vascular and brain cells communicate and are interdependent, describe the deleterious impact of a vascular dysfunction on brain function in various neurodegenerative diseases and even of psychiatric disorders, and the potential chronic deleterious effects of the pulsatile blood pressure on the cerebral microcirculation. We will also briefly review data from antihypertensive clinical trial aiming at improving or delaying dementia. Finally, we will debate how the aging process, starting early in life, could determine our sensitivity to risk factors for vascular diseases, including cerebral diseases, and the trajectory to VCID.
Collapse
Affiliation(s)
- Olivia de Montgolfier
- Faculty of Medicine, Department of Pharmacology and Physiology, Université de Montréal, Montreal, Quebec, Canada
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | | | - Eric Thorin
- Faculty of Medicine, Department of Pharmacology and Physiology, Université de Montréal, Montreal, Quebec, Canada
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
- Faculty of Medicine, Department of Surgery, Université de Montréal, Montreal, Quebec, Canada
- Correspondence: Eric Thorin ()
| |
Collapse
|
16
|
miR32-5p promoted vascular smooth muscle cell calcification by upregulating TNFα in the microenvironment. BMC Immunol 2020; 21:3. [PMID: 31952480 PMCID: PMC6967090 DOI: 10.1186/s12865-019-0324-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 10/22/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Vascular calcification is often associated with chronic inflammation and is a risk factor for brain arterial stiffness. Our previous results showed that miR32-5p was positively correlated with vascular smooth muscle cells (VSMC) calcification, but it is unclear whether miR32-5p promoted VSMC calcification by regulating inflammatory factor production. RESULTS In this study, bioinformatics analysis was used to select tumour necrosis factor α (TNFα) as a candidate inflammatory factor associated with calcification. Moreover, alizarin red staining and qRT-PCR analysis revealed that TNFα produced by BV2 cells was the key promoting factor of VSMC calcification. Interestingly, the expression of TNFα was significantly increased at the mRNA and protein levels after miR32-5p mimic treatment but significantly decreased after miR32-5p antagomir treatment. To explore the mechanism of the regulation of TNFα expression by miR32-5p, bioinformatics analysis indicated that PIKfyve was a candidate target gene of miR32-5p, and luciferase assays verified that the expression of PIKfyve was significantly repressed by miR32-5p mimics. Importantly, rescue experiments showed that the expression of TNFα in BV2 cells treated with miR32-5p antagomir and the PIKfyve inhibitor YM201636 was significantly increased. CONCLUSIONS The production of TNFα in microglia could be affected by miR32-5p targeting PIKfyve, and these results will be beneficial to reveal the mechanism of brain arterial calcification.
Collapse
|
17
|
Menezes ST, Giatti L, Colosimo EA, Ribeiro ALP, Brant LCC, Viana MC, Cunha RS, Mill JG, Barreto SM. Aortic Stiffness and Age With Cognitive Performance Decline in the ELSA-Brasil Cohort. J Am Heart Assoc 2019; 8:e013248. [PMID: 31826726 PMCID: PMC6951068 DOI: 10.1161/jaha.119.013248] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Background Increased aortic stiffness has been associated with cognitive decline and dementia, but the results are inconsistent. This study investigated the longitudinal association of aortic stiffness and age with decreased cognitive performance in 3 cognitive tests. Methods and Results This study included 6927 participants, with a mean age of 58.8 years at baseline (2008-2010), who participated in the second wave (2012-2014) of the ELSA-Brasil (Brazilian Longitudinal Study of Adult Health) (interval between visits ranging from 2-6 years). Cognitive performance was evaluated by Memory, Phonemic, and Semantic Verbal Fluency and Trail B Tests, applied at both cohort visits. Associations with the carotid-femoral pulse wave velocity and age at baseline were investigated using linear models with mixed effects after adjusting for confounders. After all the adjustments, including for systolic blood pressure, the interaction term carotid-femoral pulse wave velocity×time proved to be statistically significant for Memory and Verbal Fluency Tests, indicating that the higher carotid-femoral pulse wave velocity at baseline was associated with a faster decline in cognitive performance in these tests between waves. The interaction term age×time was statistically significant for all cognitive tests, suggesting that increasing age at baseline was also associated with a faster decline in cognitive performance between waves. Conclusions In this relatively young cohort, and after a relatively short interval, an increased aortic stiffness at baseline was associated with a sharper decline in cognitive performances in memory and verbal fluency, regardless of systolic blood pressure levels. This study also showed that the decline in cognitive performance was faster among older individuals than among younger ones at baseline.
Collapse
Affiliation(s)
- Sara T Menezes
- Medical School and Clinical Hospital Universidade Federal de Minas Gerais Belo Horizonte Brazil
| | - Luana Giatti
- Medical School and Clinical Hospital Universidade Federal de Minas Gerais Belo Horizonte Brazil
| | - Enrico A Colosimo
- Department of Statistics Universidade Federal de Minas Gerais Belo Horizonte Brazil
| | - Antônio L P Ribeiro
- Medical School and Clinical Hospital Universidade Federal de Minas Gerais Belo Horizonte Brazil
| | - Luisa C C Brant
- Medical School and Clinical Hospital Universidade Federal de Minas Gerais Belo Horizonte Brazil
| | - Maria C Viana
- Department of Social Medicine Universidade Federal do Espírito Santo Vitória Brazil
| | - Roberto S Cunha
- Department of Physiological Sciences Universidade Federal do Espírito Santo Vitória Brazil
| | - José G Mill
- Department of Physiological Sciences Universidade Federal do Espírito Santo Vitória Brazil
| | - Sandhi Maria Barreto
- Medical School and Clinical Hospital Universidade Federal de Minas Gerais Belo Horizonte Brazil
| |
Collapse
|
18
|
de Montgolfier O, Pinçon A, Pouliot P, Gillis MA, Bishop J, Sled JG, Villeneuve L, Ferland G, Lévy BI, Lesage F, Thorin-Trescases N, Thorin É. High Systolic Blood Pressure Induces Cerebral Microvascular Endothelial Dysfunction, Neurovascular Unit Damage, and Cognitive Decline in Mice. Hypertension 2019; 73:217-228. [PMID: 30571552 DOI: 10.1161/hypertensionaha.118.12048] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A chronic and gradual increase in pulse pressure (PP) is associated with cognitive decline and dementia in older individuals, but the mechanisms remain ill-defined. We hypothesized that a chronic elevation of PP would cause brain microvascular endothelial mechanical stress, damage the neurovascular unit, and ultimately induce cognitive impairment in mice, potentially contributing to the progression of vascular dementia and Alzheimer disease. To test our hypothesis, male control wild-type mice and Alzheimer disease model APP/PS1 (amyloid precursor protein/presenilin 1) mice were exposed to a transverse aortic constriction for 6 weeks, creating a PP overload in the right carotid (ipsilateral). We show that the transverse aortic constriction procedure associated with high PP induces a cascade of vascular damages in the ipsilateral parenchymal microcirculation: in wild-type mice, it impairs endothelial dilatory and blood brain barrier functions and causes microbleeds, a reduction in microvascular density, microvascular cell death by apoptosis, leading to severe hypoperfusion and parenchymal cell senescence. These damages were associated with brain inflammation and a significant reduction in learning and spatial memories. In APP/PS1 mice, that endogenously display severe cerebral vascular dysfunctions, microbleeds, parenchymal inflammation and cognitive dysfunction, transverse aortic constriction-induced high PP further aggravates cerebrovascular damage, Aβ (beta-amyloid) accumulation, and prevents learning. Our study, therefore, demonstrates that brain microvessels are vulnerable to a high PP and mechanical stress associated with transverse aortic constriction, promoting severe vascular dysfunction, disruption of the neurovascular unit, and cognitive decline. Hence, chronic elevated amplitude of the PP could contribute to the development and progression of vascular dementia including Alzheimer disease.
Collapse
Affiliation(s)
- Olivia de Montgolfier
- From the Department of Pharmacology and Physiology (O.d.M., A.P.), Université de Montréal, Quebec, Canada.,Montreal Heart Institute, Research Center, Quebec, Canada (O.d.M., A.P., M.-A.G., L.V., G.F., F.L., N.T.-T., E.T.)
| | - Anthony Pinçon
- From the Department of Pharmacology and Physiology (O.d.M., A.P.), Université de Montréal, Quebec, Canada.,Montreal Heart Institute, Research Center, Quebec, Canada (O.d.M., A.P., M.-A.G., L.V., G.F., F.L., N.T.-T., E.T.)
| | | | - Marc-Antoine Gillis
- Montreal Heart Institute, Research Center, Quebec, Canada (O.d.M., A.P., M.-A.G., L.V., G.F., F.L., N.T.-T., E.T.)
| | - Jonathan Bishop
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada (J.B., J.G.S.)
| | - John G Sled
- Mouse Imaging Centre, The Hospital for Sick Children, Toronto, Ontario, Canada (J.B., J.G.S.).,Department of Medical Biophysics, University of Toronto, Ontario, Canada (J.G.S.)
| | - Louis Villeneuve
- Montreal Heart Institute, Research Center, Quebec, Canada (O.d.M., A.P., M.-A.G., L.V., G.F., F.L., N.T.-T., E.T.)
| | - Guylaine Ferland
- Department of Nutrition (G.F.), Université de Montréal, Quebec, Canada.,Montreal Heart Institute, Research Center, Quebec, Canada (O.d.M., A.P., M.-A.G., L.V., G.F., F.L., N.T.-T., E.T.)
| | - Bernard I Lévy
- Institut des Vaisseaux et du Sang, Hôpital Lariboisière, Paris, France (B.I.L.)
| | - Frédéric Lesage
- Montreal Heart Institute, Research Center, Quebec, Canada (O.d.M., A.P., M.-A.G., L.V., G.F., F.L., N.T.-T., E.T.).,Ecole Polytechnique de Montréal, Quebec, Canada (P.P., F.L.)
| | - Nathalie Thorin-Trescases
- Montreal Heart Institute, Research Center, Quebec, Canada (O.d.M., A.P., M.-A.G., L.V., G.F., F.L., N.T.-T., E.T.)
| | - Éric Thorin
- Department of Surgery (E.T.), Université de Montréal, Quebec, Canada.,Montreal Heart Institute, Research Center, Quebec, Canada (O.d.M., A.P., M.-A.G., L.V., G.F., F.L., N.T.-T., E.T.)
| |
Collapse
|
19
|
Di Daniele N, Celotto R, Alunni Fegatelli D, Gabriele M, Rovella V, Scuteri A. Common Carotid Artery Calcification Impacts on Cognitive Function in Older Patients. High Blood Press Cardiovasc Prev 2019; 26:127-134. [PMID: 30779026 DOI: 10.1007/s40292-019-00301-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Accepted: 01/14/2019] [Indexed: 01/13/2023] Open
Abstract
INTRODUCTION Cognitive impairment and dementia represent an emerging health problem. Cardiovascular (CV) risk factors contribute to cognitive impairment. AIM To investigate the effect of vascular calcification on cognitive impairment and dementia, independently of plaque and traditional CV risk factors. METHODS Four hundred and sixty-nine patients (age of 78.6 ± 6.1 years, 74.4% women) were studied. Traditional CV risk factors levels, cognitive function (MMSE), brain CT scan, and other vascular parameters were measured. Common Carotid Artery (CCA) plaque and calcification were evaluated by ultrasound. RESULTS CCA calcification was associated with a lower MMSE score than in subjects with no CCA calcification (23.7 ± 0.3 versus 25.5 ± 0.8; p = 0.015), after controlling for age, sex, education, blood pressure levels, diabetes, creatinine, lipid lowering therapy, neuroimaging alteration, and CCA plaque. Similarly, CCA calcification was associated with higher odds of dementia regardless of the presence of CCA plaque (OR 1.70, 95% CI 1.01-2.94, p < 0.05). This trend was not observed when stratifying patients according to the presence of CCA plaque. CONCLUSION CCA calcification is associated with cognitive impairment and dementia, independently of established CV risk factors and CCA plaque. The impact of arterial calcification on cognition seems largely independent of arterial stiffness.
Collapse
Affiliation(s)
- Nicola Di Daniele
- Hypertension and Nephrology Unit, Department of Medicine, Policinico Tor Vergata, Universita'di Roma Tor Vergata, Rome, Italy
| | - Roberto Celotto
- Hypertension and Nephrology Unit, Department of Medicine, Policinico Tor Vergata, Universita'di Roma Tor Vergata, Rome, Italy
| | | | - Marco Gabriele
- Hypertension and Nephrology Unit, Department of Medicine, Policinico Tor Vergata, Universita'di Roma Tor Vergata, Rome, Italy
| | - Valentina Rovella
- Hypertension and Nephrology Unit, Department of Medicine, Policinico Tor Vergata, Universita'di Roma Tor Vergata, Rome, Italy
| | - Angelo Scuteri
- Department of Medical, Surgical, and Experimental Sciences, University of Sassari, Sassari, Italy.
| |
Collapse
|
20
|
Iulita MF, Vallerand D, Beauvillier M, Haupert N, A Ulysse C, Gagné A, Vernoux N, Duchemin S, Boily M, Tremblay MÈ, Girouard H. Differential effect of angiotensin II and blood pressure on hippocampal inflammation in mice. J Neuroinflammation 2018; 15:62. [PMID: 29490666 PMCID: PMC6389185 DOI: 10.1186/s12974-018-1090-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 02/05/2018] [Indexed: 12/11/2022] Open
Abstract
Background Angiotensin II (Ang II), a peptide hormone involved in the development of hypertension, causes systemic and cerebral inflammation, affecting brain regions important for blood pressure control. The cause-and-effect relationship between hypertension and inflammation is two-way, but the role of blood pressure in the induction of cerebral inflammation is less clear. The vulnerability of specific brain regions, particularly those important for memory, is also of interest. Methods We used molecular biology approaches, immunohistochemistry, and electron microscopy to examine the interdependence between the hypertensive and pro-inflammatory effects of Ang II. We examined the effect of blood pressure by administering a subpressive (200 ng/kg/min) or a pressive Ang II dose (1000 or 1900 ng/kg/min) with and without hydralazine (150 mg/L) for 1 week and used phenylephrine to increase blood pressure independently of the renin-angiotensin system. Results Ang II increased ionized calcium-binding adaptor molecule 1 (Iba-1) levels (marker of microgliosis) in the whole brain and in the hippocampus in a dose-dependent manner. Pressive Ang II induced specific changes in microglial morphology, indicating differences in functional phenotype. An increase in hippocampal glial fibrillary acidic protein (GFAP) was seen in mice receiving pressive Ang II, while no induction of cerebral gliosis was observed after 7 days of subpressive Ang II infusion. Although phenylephrine led to increased astrogliosis, it did not affect Iba-1 expression. Pressive Ang II stimulated TNF-α production in the hippocampus, and daily treatment with hydralazine prevented this increase. Hydralazine also reduced GFAP and Iba-1 levels. With longer perfusion (14 days), subpressive Ang II led to some but not all the inflammatory changes detected with the pressive doses, mainly an increase in CD68 and Iba-1 but not of GFAP or TNF-α. Conclusions Blood pressure and Ang II differentially contribute to hippocampal inflammation in mice. Control of blood pressure and Ang II levels should prevent or reduce brain inflammation and therefore brain dysfunctions associated with hypertension. Electronic supplementary material The online version of this article (10.1186/s12974-018-1090-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- M Florencia Iulita
- Department of Neurosciences, Université de Montréal, 2960 Chemin de la Tour, Montréal, Québec, H3T 1J4, Canada.,Groupe de recherche sur le système nerveux central (GRSNC), Université de Montréal, 2960 Chemin de la Tour, Montréal, Québec, H3T 1J4, Canada
| | - Diane Vallerand
- Department of Pharmacology and Physiology, Université de Montréal, Pavillon Roger-Gaudry, 2900 Boulevard Édouard-Montpetit, Montréal, Québec, H3T 1J4, Canada
| | - Mélissa Beauvillier
- Department of Pharmacology and Physiology, Université de Montréal, Pavillon Roger-Gaudry, 2900 Boulevard Édouard-Montpetit, Montréal, Québec, H3T 1J4, Canada
| | - Nathalie Haupert
- Department of Pharmacology and Physiology, Université de Montréal, Pavillon Roger-Gaudry, 2900 Boulevard Édouard-Montpetit, Montréal, Québec, H3T 1J4, Canada
| | - Corinne A Ulysse
- Department of Pharmacology and Physiology, Université de Montréal, Pavillon Roger-Gaudry, 2900 Boulevard Édouard-Montpetit, Montréal, Québec, H3T 1J4, Canada
| | - Audrey Gagné
- Axe Neurosciences, CRCHU de Québec-Université Laval, 2705 Boulevard Laurier, Québec, Québec, G1V 4G2, Canada
| | - Nathalie Vernoux
- Axe Neurosciences, CRCHU de Québec-Université Laval, 2705 Boulevard Laurier, Québec, Québec, G1V 4G2, Canada
| | - Sonia Duchemin
- Department of Pharmacology and Physiology, Université de Montréal, Pavillon Roger-Gaudry, 2900 Boulevard Édouard-Montpetit, Montréal, Québec, H3T 1J4, Canada
| | - Michaël Boily
- Department of Pharmacology and Physiology, Université de Montréal, Pavillon Roger-Gaudry, 2900 Boulevard Édouard-Montpetit, Montréal, Québec, H3T 1J4, Canada
| | - Marie-Ève Tremblay
- Axe Neurosciences, CRCHU de Québec-Université Laval, 2705 Boulevard Laurier, Québec, Québec, G1V 4G2, Canada.,Department of Molecular Medicine, Université Laval, 1050, Avenue de la Médecine, Québec, Québec, G1V 0A6, Canada
| | - Hélène Girouard
- Groupe de recherche sur le système nerveux central (GRSNC), Université de Montréal, 2960 Chemin de la Tour, Montréal, Québec, H3T 1J4, Canada. .,Department of Pharmacology and Physiology, Université de Montréal, Pavillon Roger-Gaudry, 2900 Boulevard Édouard-Montpetit, Montréal, Québec, H3T 1J4, Canada. .,Centre de recherche de l'Institut universitaire de gériatrie de Montréal, 545 Queen Mary Rd, Montréal, Québec, H3W 1W6, Canada.
| |
Collapse
|