1
|
Xu X, Xiao C, Yi M, Yang J, Liao M, Zhou K, Hu L, Ouyang F, Lan L, Fan Y. Cerebral Perfusion Characteristics and Dynamic Brain Structural Changes in Stroke-Prone Renovascular Hypertensive Rats: A Preclinical Model for Cerebral Small Vessel Disease. Transl Stroke Res 2025; 16:612-624. [PMID: 38443727 DOI: 10.1007/s12975-024-01239-8] [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: 12/15/2023] [Revised: 02/08/2024] [Accepted: 02/29/2024] [Indexed: 03/07/2024]
Abstract
Hypertension is a leading cause of cerebral small vessel disease (CSVD) and vascular dementia in elderly individuals. We aimed to assess cerebral perfusion and dynamic changes in brain structure in stroke-prone renovascular hypertensive rats (RHRSPs) with different durations of hypertension and to investigate whether they have pathophysiological features similar to those of humans with CSVD. The RHRSP model was established using the two-kidney, two-clip (2k2c) method, and the Morris water maze (MWM) test, MRI, immunohistochemistry, and biochemical analysis were performed at multiple time points for up to six months following the 2k2c operation. Systolic blood pressure was significantly greater in the RHRSP group than in the sham-operated group at week 4 post-surgery and continued to increase over time, leading to cognitive decline by week 20. Arterial spin labeling revealed cerebral hypoperfusion in the RHRSP group at 8 weeks, accompanied by vascular remodeling and decreased vessel density. Diffusion tensor imaging and Luxol fast blue staining indicated that white matter disintegration and demyelination gradually progressed in the corpus callosum and that myelin basic protein levels decreased. Eight weeks after surgery, blood-brain barrier (BBB) leakage into the corpus callosum was observed. The albumin leakage area was negatively correlated with the myelin sheath area (r=-0.88, p<0.001). RNA-seq analysis revealed downregulation of most angiogenic genes and upregulation of antiangiogenic genes in the corpus callosum of RHRSPs 24 weeks after surgery. RHRSPs developed cerebral hypoperfusion, BBB disruption, spontaneous white matter damage, and cognitive impairment as the duration of hypertension increased. RHRSPs share behavioral and neuropathological characteristics with CSVD patients, making them suitable animal models for preclinical trials related to CSVD.
Collapse
Affiliation(s)
- Xiangming Xu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Chi Xiao
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Ming Yi
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Jing Yang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Mengshi Liao
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Kun Zhou
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Liuting Hu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Fubing Ouyang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Linfang Lan
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Yuhua Fan
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University; Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases; National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China.
| |
Collapse
|
2
|
Wu ZP, Wei W, Liu S, Hu MD, Zhao H, Li XF, Chen X. The effect of hypertension, obesity, and type 2 diabetes on lacunar stroke: A network Mendelian randomization study. Nutr Metab Cardiovasc Dis 2025; 35:103974. [PMID: 40189994 DOI: 10.1016/j.numecd.2025.103974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND AND AIMS Previous studies have reported an association of lacunar stroke with hypertension, obesity, and type 2 diabetes (T2D). The aim of this study was to investigate whether the association was causal and whether body mass index mediated the effect of hypertension on lacunar stroke. METHODS AND RESULTS The independence and causal association of hypertension, obesity, and T2D with lacunar stroke were assessed by multivariate Mendelian randomization (MVMR) and network Mendelian randomization (NMR) with inverse variance weighting (IVW). The reliability of the results was increased by sensitivity analyses including MR-Egger, Cochrane's Q test, Mendelian Randomization Pleiotropy RESidual Sum and Outlier (MR-PRESSO), and leave-one-out. MVMR analysis found that genetically predicted hypertension had a 42 % higher lacunar stroke risk (OR: 1.42, 95 % CI: 1.29-1.56, P < 0.001) when adjusted for obesity and T2D, genetically predicted T2D had a 9 % higher lacunar stroke risk (OR: 1.09, 95 % CI: 1.03-1.16, P < 0.004) when adjusted for hypertension and obesity, and genetically predicted obesity had a 15 % lower lacunar stroke risk (OR: 0.85, 95 % CI: 0.77-0.93, P < 0.001) when adjusted for hypertension and T2D. NMR found that 44 % of the association between hypertension and lacunar stroke risk was mediated by obesity. CONCLUSION This genetic association study found novel independent genetic associations between hypertension and T2D with high risk of lacunar stroke, whereas obesity attenuated the risk of lacunar stroke. The findings emphasize the importance of individualized lacunar stroke prevention strategies rather than uniform weight management optimize medical care in high-risk populations.
Collapse
Affiliation(s)
- Zhi-Ping Wu
- Department of Epidemiology, School of Public Health, Dalian Medical University, Dalian, China
| | - Wei Wei
- Department of Neurosurgery, Central Hospital of Dalian University of Technology, Dalian, China
| | - Shan Liu
- Department of Epidemiology, School of Public Health, Dalian Medical University, Dalian, China; Emergency Department, Dalian Center for Disease Control and Prevention, Dalian, China
| | - Meng-Die Hu
- Department of Epidemiology, School of Public Health, Dalian Medical University, Dalian, China
| | - Heng Zhao
- Department of Epidemiology, School of Public Health, Dalian Medical University, Dalian, China
| | - Xiao-Feng Li
- Department of Epidemiology, School of Public Health, Dalian Medical University, Dalian, China
| | - Xin Chen
- Department of Epidemiology, School of Public Health, Dalian Medical University, Dalian, China.
| |
Collapse
|
3
|
Yang G, Zhu L, Wang Y, Yu Y, Liu X, Xia J, Yang Y, Wang H, Feng B. Antihypertensive effect of sinapine extracted from rapeseed meal in 2K1C hypertensive rats. Sci Rep 2025; 15:4133. [PMID: 39900955 PMCID: PMC11790856 DOI: 10.1038/s41598-025-88926-0] [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: 04/21/2024] [Accepted: 01/31/2025] [Indexed: 02/05/2025] Open
Abstract
To extract sinapine from rapeseed meal and investigate its antihypertensive function and mechanism. Blood pressure was measured before and after sinapine administration to evaluate sinapine's immediate antihypertensive function. Twokidney, oneclip (2K1C) hypertensive rats were given sinapine for four weeks, with weekly blood pressure monitoring. The renin angiotensin aldosterone system (RAAS), including the levels of renin, angiotensin I (Ang I), angiotensin II (Ang II), aldosterone (ALD), angiotensin-converting enzyme (ACE) and other molecules related to blood pressure, such as NO, prostacyclin (PGI2), endothelin-1 (ET1), and thromboxane A2 (TXA2), were measured in rat blood. The impact of sinapine on vascular endothelial cell (A10) calcium and potassium channels was assessed using the patch-clamp technique. One-time or long-term administration of sinapine significantly reduced the rats' systolic blood pressure (SBP), diastolic pressure (DBP), and mean blood pressure (MBP). Sinapine also decreased the levels of Ang II and ALD. Furthermore, sinapine effectively inhibited ACE activation, increased NO levels, and blocked L-type calcium channels. Sinapine has an antihypertensive function and achieves this process through multiple targets.
Collapse
Affiliation(s)
- Gaoyuan Yang
- Guangzhou Sport University, Guangzhou, 510500, Guangdong, China
| | - Lin Zhu
- Guangzhou Sport University, Guangzhou, 510500, Guangdong, China
| | - Yu Wang
- Guangzhou Sport University, Guangzhou, 510500, Guangdong, China
| | - Yang Yu
- Guangzhou Sport University, Guangzhou, 510500, Guangdong, China
| | - Xiaoguang Liu
- Guangzhou Sport University, Guangzhou, 510500, Guangdong, China
| | - Jingbo Xia
- Guangzhou Sport University, Guangzhou, 510500, Guangdong, China
| | - Yunjie Yang
- Guangzhou Sport University, Guangzhou, 510500, Guangdong, China
| | - Huiguo Wang
- Guangzhou Sport University, Guangzhou, 510500, Guangdong, China.
| | - Baomin Feng
- College of Life and Health, Dalian University, Liaoning, Dalian, 116622, China.
| |
Collapse
|
4
|
Zhào H, Zhang H, Ding Y, Li H, Huang Y. Circadian rest-activity rhythm pattern in the elderly with cerebral small vessel disease: Using multiple estimated methods. J Alzheimers Dis 2025; 103:856-864. [PMID: 39784723 DOI: 10.1177/13872877241307254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Background: Disruption of circadian rest-activity rhythm (RAR) has been found in many neurological disorders. Objective: In this study, actigraphic data were collected and analyzed to identify the RAR pattern in the elderly with cerebral small vessel disease. Methods: 115 cerebral small vessel disease (CSVD) cases were recruited. The presence of lacune infarct, white matter hyperintensities, and cerebral microbleeds in magnetic resonance imaging (MRI) images were rated independently, as well as using a simple MRI score of 0-3 points. Each subject wore an Actigraph device in their nondominant hand for 4-7 days to collect raw data. RAR parameters were generated using both extended cosinor model (RAR α, RAR β, amplitude, acrophase, up-mesor, down-mesor, and pseudo-F statistic) and non-parametric methods (interdaily stability, intradaily variability, and relative amplitude). Results: Elder patients with a simple MRI score of 2-3 points showed a statistically lower amplitude compared with individuals with a simple MRI score of 0 points in the extended cosinor model. For the non-parametric method, elderly people with a simple MRI score of 1-3 points exhibited higher intradaily variability relative to those participants with a simple MRI score of 0 points. However, no differences were found regarding sleep quality among individuals with different simple MRI scores. White matter hyperintensities, lacune infarct, and cerebral microbleeds were independently associated with RAR β, RAR α, and intradaily variability, respectively. Conclusions: The RAR pattern was disturbed in elderly adults with CSVD. Abnormal RAR parameters were independently associated with CSVD MRI markers.
Collapse
Affiliation(s)
- Hóngyi Zhào
- Department of Neurology, The Seventh Medical Center of PLA General Hospital, Beijing, China
- Department of Neurology, NO 984 Hospital of PLA, Beijing, China
| | - Haiyang Zhang
- Center for Disease Control and Prevention of Central Theater Command, Beijing, China
| | - Yu Ding
- Department of Neurology, The Seventh Medical Center of PLA General Hospital, Beijing, China
| | - Hong Li
- Center for Disease Control and Prevention of Central Theater Command, Beijing, China
| | - Yonghua Huang
- Department of Neurology, The Seventh Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
5
|
Bushnell C, Kernan WN, Sharrief AZ, Chaturvedi S, Cole JW, Cornwell WK, Cosby-Gaither C, Doyle S, Goldstein LB, Lennon O, Levine DA, Love M, Miller E, Nguyen-Huynh M, Rasmussen-Winkler J, Rexrode KM, Rosendale N, Sarma S, Shimbo D, Simpkins AN, Spatz ES, Sun LR, Tangpricha V, Turnage D, Velazquez G, Whelton PK. 2024 Guideline for the Primary Prevention of Stroke: A Guideline From the American Heart Association/American Stroke Association. Stroke 2024; 55:e344-e424. [PMID: 39429201 DOI: 10.1161/str.0000000000000475] [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: 10/22/2024]
Abstract
AIM The "2024 Guideline for the Primary Prevention of Stroke" replaces the 2014 "Guidelines for the Primary Prevention of Stroke." This updated guideline is intended to be a resource for clinicians to use to guide various prevention strategies for individuals with no history of stroke. METHODS A comprehensive search for literature published since the 2014 guideline; derived from research involving human participants published in English; and indexed in MEDLINE, PubMed, Cochrane Library, and other selected and relevant databases was conducted between May and November 2023. Other documents on related subject matter previously published by the American Heart Association were also reviewed. STRUCTURE Ischemic and hemorrhagic strokes lead to significant disability but, most important, are preventable. The 2024 primary prevention of stroke guideline provides recommendations based on current evidence for strategies to prevent stroke throughout the life span. These recommendations align with the American Heart Association's Life's Essential 8 for optimizing cardiovascular and brain health, in addition to preventing incident stroke. We also have added sex-specific recommendations for screening and prevention of stroke, which are new compared with the 2014 guideline. Many recommendations for similar risk factor prevention were updated, new topics were reviewed, and recommendations were created when supported by sufficient-quality published data.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Eliza Miller
- American College of Obstetricians and Gynecologists liaison
| | | | | | | | | | | | | | - Alexis N Simpkins
- American Heart Association Stroke Council Scientific Statement Oversight Committee on Clinical Practice Guideline liaison
| | | | | | | | | | | | | |
Collapse
|
6
|
Pacella J, Lembo G, Carnevale L. A Translational Perspective on the Interplay Between Hypertension, Inflammation and Cognitive Impairment. Can J Cardiol 2024; 40:2368-2377. [PMID: 39455022 DOI: 10.1016/j.cjca.2024.10.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/26/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
Hypertension represents the major risk factor in the onset of cardiovascular disease worldwide. Preclinically, several mouse models of hypertension have been developed to investigate the pathophysiological link between hypertension and vascular impairment. Specifically, angiotensin-II infusion, transverse aortic constriction, deoxycorticosterone acetate salt, and N(ω)-nitro-L-arginine methyl ester (L-NAME) administration as hypertensive stimuli at the preclinical level permit the unveiling of a proinflammatory response driven by the innate and adaptive immune system and leads to vascular injury in terms of structural and functional alterations. Vascular impairment seems to be particularly critical at the cerebral level wherein arterioles, venules, and capillaries finely tune blood supply across the whole brain leading to the onset of a well known clinical condition named cerebral small vessel disease (cSVD) characterized by extensive brain injury, which culminates in the decline of cognitive functions. Advances in magnetic resonance imaging permit identification and accurate diagnosis of specific cSVD biomarkers including white matter hyperintensities, lacunar strokes, cerebral microbleeds, and enlarged perivascular spaces, each of which proved to be associated with a specific cognitive domain impairment. Such an approach in combination with pharmacological interventions targeted to the lowering of blood pressure and the prevention of vascular thrombosis formation represents a solid strategy in the prevention and the management of cSVD cognitive decay.
Collapse
Affiliation(s)
- Jacopo Pacella
- Department of Angiocardioneurology and Translational Medicine, IRCCS INM Neuromed, Pozzilli, Italy
| | - Giuseppe Lembo
- Department of Angiocardioneurology and Translational Medicine, IRCCS INM Neuromed, Pozzilli, Italy; Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy.
| | - Lorenzo Carnevale
- Department of Angiocardioneurology and Translational Medicine, IRCCS INM Neuromed, Pozzilli, Italy. https://twitter.com/LorCarnevale
| |
Collapse
|
7
|
Tian JS, Wei YC, Wang P, Ling QS, Wang DX, Wang Z, Miao ZW, Miao CY. Pharmacological effects of MT-1207 in bilateral renal artery stenosis hypertension and its hypotensive targets validation. Biomed Pharmacother 2024; 178:117234. [PMID: 39106710 DOI: 10.1016/j.biopha.2024.117234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 08/09/2024] Open
Abstract
MT-1207 (MT) as a new antihypertensive drug is under clinical trial. However, its hypotensive mechanism has not been experimentally explored, and it is unknown whether MT can be used for bilateral renal artery stenosis hypertension. Using two-kidney two-clip (2K2C) to mimic bilateral renal artery stenosis in rats, a stroke-prone renovascular hypertension model, the present study further verified its antihypertensive effect, cardiovascular and renal protection, mortality reduction and lifespan prolongation, as well as demonstrated its two novel pharmacological effects for uric acid-lowering and cognition-improving. Notably, MT did not aggravate renal dysfunction; instead, it had beneficial effects on reducing serum uric acid level and maintaining serum K+ at a relatively stable level in 2K2C rats. In contrast, angiotensin receptor blocker losartan aggravated renal dysfunction in 2K2C rats. Mechanistically, MT hypotensive effect was dependent on its blockade of α1 and 5-HT2 receptors, since MT pretreatment abolished these receptor agonists-induced blood pressure elevations in vivo. Further evidence showed MT bound to and interacted with these receptor subtypes including α1A, α1B, α1D, 5-HT2A, 5-HT2B, and 5-HT2C receptors known for control of blood pressure. In conclusion, MT may be used for treatment of bilateral renal artery stenosis hypertension, different from losartan that is prohibited for treatment of bilateral renal artery stenosis hypertension. Targets validation of MT hypotensive mechanism and beneficial effects of MT on uric acid and cognitive function provide new insights for this novel multitarget drug, deserving clinical trial attention.
Collapse
Affiliation(s)
- Jia-Sheng Tian
- Department of Pharmacology, Second Military Medical University / Naval Medical University, Shanghai, China
| | - Yu-Chen Wei
- School of Medicine, Shanghai University, Shanghai, China
| | - Peng Wang
- ORxes Therapeutics Co., Ltd, Shenyang, China
| | - Qi-Sheng Ling
- Department of Pharmacology, Second Military Medical University / Naval Medical University, Shanghai, China
| | - Dao-Xin Wang
- Department of Pharmacology, Second Military Medical University / Naval Medical University, Shanghai, China
| | - Zhi Wang
- Department of Pharmacology, Second Military Medical University / Naval Medical University, Shanghai, China
| | - Zhu-Wei Miao
- Department of Pharmacology, Second Military Medical University / Naval Medical University, Shanghai, China
| | - Chao-Yu Miao
- Department of Pharmacology, Second Military Medical University / Naval Medical University, Shanghai, China; School of Medicine, Shanghai University, Shanghai, China.
| |
Collapse
|
8
|
Liu C, Li Q, Li Z, Wang L, Wang C, Du X, Song W, Sun X, Lu C. Association between the incident hypertension duration and cognitive performance in older adults: data from the NHANES 2011-2014. Aging Clin Exp Res 2024; 36:181. [PMID: 39212760 PMCID: PMC11364694 DOI: 10.1007/s40520-024-02836-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Established evidences have demonstrated that hypertension was associated with the cognitive impairment. But the associations between the duration of hypertension exposure and cognitive performance are still inconclusive. OBJECTIVES The objective of this study was to assess the association between the duration of hypertension diagnosis and cognitive performance in older adults by the National Health and Nutrition Examination Survey (2011-2014). METHODS To evaluate the relationship between the hypertension duration and cognitive performance, we conducted the logistic regression analysis. Furthermore, we also performed the Restricted cubic spline (RCS) analysis to assess the nonlinear relationship between the duration of exposure to hypertension and cognitive performance. RESULTS Initially, total 19,931 participants were included in this study, and 2928 individuals were enrolled. With the increase of hypertension duration, more risk of cognitive impairment was observed in the Digit Symbol Substitution test (DSST) (OR = 1.012, 1.006-1.019), and a similar trend was observed in Animal Fluency test (AFT) (OR = 1.009,1.003-1.016). The RCS results showed that the hypertension duration pattern was linear associated with the risk of cognitive impairment in DDST (P for non-linearity = 0.758). Meanwhile, subgroups analysis of midlife hypertension, we revealed that linear association with the risk of cognitive impairment in DSST (P for non-linearity = 0.391) and CERAD (P for non-linearity = 0.849) among hypertension diagnose < 55 years populations. CONCLUSION Collectively, our finding indicates that longer duration of exposure to hypertension worsens the cognition performance, especially for middle-aged hypertension.
Collapse
Affiliation(s)
- Chunlei Liu
- School of Medicine, Nankai University, Tianjin, 300071, China
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, 300192, China
| | - Qi Li
- School of Medicine, Nankai University, Tianjin, 300071, China
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, 300192, China
| | - Zhuqing Li
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, 300192, China
| | - Li Wang
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, 300192, China
| | - Che Wang
- School of Medicine, Nankai University, Tianjin, 300071, China
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, 300192, China
| | - Xiaoyu Du
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, 300192, China
- Department of Cardiology, The First Central Clinical School, Tianjin Medical University, Tianjin, 300192, China
| | - Wenjuan Song
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, 300192, China
- Department of Cardiology, The First Central Clinical School, Tianjin Medical University, Tianjin, 300192, China
| | - Xiaotong Sun
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, 300192, China
- Department of Cardiology, The First Central Clinical School, Tianjin Medical University, Tianjin, 300192, China
| | - Chengzhi Lu
- School of Medicine, Nankai University, Tianjin, 300071, China.
- Department of Cardiology, Tianjin First Center Hospital, Tianjin, 300192, China.
| |
Collapse
|
9
|
Hanscom M, Morales-Soto W, Watts SW, Jackson WF, Gulbransen BD. Innervation of adipocytes is limited in mouse perivascular adipose tissue. Am J Physiol Heart Circ Physiol 2024; 327:H155-H181. [PMID: 38787382 PMCID: PMC11380956 DOI: 10.1152/ajpheart.00041.2024] [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: 01/23/2024] [Revised: 05/10/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024]
Abstract
Perivascular adipose tissue (PVAT) regulates vascular tone by releasing anticontractile factors. These anticontractile factors are driven by processes downstream of adipocyte stimulation by norepinephrine; however, whether norepinephrine originates from neural innervation or other sources is unknown. The goal of this study was to test the hypothesis that neurons innervating PVAT provide the adrenergic drive to stimulate adipocytes in aortic and mesenteric perivascular adipose tissue (aPVAT and mPVAT), and white adipose tissue (WAT). Healthy male and female mice (8-13 wk) were used in all experiments. Expression of genes associated with synaptic transmission were quantified by qPCR and adipocyte activity in response to neurotransmitters and neuron depolarization was assessed in AdipoqCre+;GCaMP5g-tdTf/WT mice. Immunostaining, tissue clearing, and transgenic reporter lines were used to assess anatomical relationships between nerves and adipocytes. Although synaptic transmission component genes are expressed in adipose tissues (aPVAT, mPVAT, and WAT), strong nerve stimulation with electrical field stimulation does not significantly trigger calcium responses in adipocytes. However, norepinephrine consistently elicits strong calcium responses in adipocytes from all adipose tissues studied. Bethanechol induces minimal adipocyte responses. Imaging neural innervation using various techniques reveals that nerve fibers primarily run alongside blood vessels and rarely branch into the adipose tissue. Although nerve fibers are associated with blood vessels in adipose tissue, they demonstrate limited anatomical and functional interactions with adjacent adipocytes, challenging the concept of classical innervation. These findings dispute the significant involvement of neural input in regulating PVAT adipocyte function and emphasize alternative mechanisms governing adrenergic-driven anticontractile functions of PVAT.NEW & NOTEWORTHY This study challenges prevailing views on neural innervation in perivascular adipose tissue (PVAT) and its role in adrenergic-driven anticontractile effects on vasculature. Contrary to existing paradigms, limited anatomical and functional connections were found between PVAT nerve fibers and adipocytes, underscoring the importance of exploring alternative mechanistic pathways. Understanding the mechanisms involved in PVAT's anticontractile effects is critical for developing potential therapeutic interventions against dysregulated vascular tone, hypertension, and cardiovascular disease.
Collapse
Affiliation(s)
- Marie Hanscom
- Department of Physiology, Michigan State University, East Lansing, Michigan, United States
| | - Wilmarie Morales-Soto
- Department of Physiology, Michigan State University, East Lansing, Michigan, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Stephanie W Watts
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - William F Jackson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - Brian D Gulbransen
- Department of Physiology, Michigan State University, East Lansing, Michigan, United States
| |
Collapse
|
10
|
Hayden MR. Cerebral Microbleeds Associate with Brain Endothelial Cell Activation-Dysfunction and Blood-Brain Barrier Dysfunction/Disruption with Increased Risk of Hemorrhagic and Ischemic Stroke. Biomedicines 2024; 12:1463. [PMID: 39062035 PMCID: PMC11274519 DOI: 10.3390/biomedicines12071463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/25/2024] [Accepted: 06/27/2024] [Indexed: 07/28/2024] Open
Abstract
Globally, cerebral microbleeds (CMBs) are increasingly being viewed not only as a marker for cerebral small vessel disease (SVD) but also as having an increased risk for the development of stroke (hemorrhagic/ischemic) and aging-related dementia. Recently, brain endothelial cell activation and dysfunction and blood-brain barrier dysfunction and/or disruption have been shown to be associated with SVD, enlarged perivascular spaces, and the development and evolution of CMBs. CMBs are a known disorder of cerebral microvessels that are visualized as 3-5 mm, smooth, round, or oval, and hypointense (black) lesions seen only on T2*-weighted gradient recall echo or susceptibility-weighted sequences MRI images. CMBs are known to occur with high prevalence in community-dwelling older individuals. Since our current global population is the oldest recorded in history and is only expected to continue to grow, we can expect the healthcare burdens associated with CMBs to also grow. Increased numbers (≥10) of CMBs should raise a red flag regarding the increased risk of large symptomatic neurologic intracerebral hemorrhages. Importantly, CMBs are also currently regarded as markers of diffuse vascular and neurodegenerative brain damage. Herein author highlights that it is essential to learn as much as we can about CMB development, evolution, and their relation to impaired cognition, dementia, and the exacerbation of neurodegeneration.
Collapse
Affiliation(s)
- Melvin R Hayden
- Department of Internal Medicine, Endocrinology Diabetes and Metabolism, Diabetes and Cardiovascular Disease Center, University of Missouri School of Medicine, One Hospital Drive, Columbia, MO 65211, USA
| |
Collapse
|
11
|
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
|
12
|
Jia R, Solé-Guardia G, Kiliaan AJ. Blood-brain barrier pathology in cerebral small vessel disease. Neural Regen Res 2024; 19:1233-1240. [PMID: 37905869 PMCID: PMC11467932 DOI: 10.4103/1673-5374.385864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/27/2023] [Accepted: 08/22/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT Cerebral small vessel disease is a neurological disease that affects the brain microvasculature and which is commonly observed among the elderly. Although at first it was considered innocuous, small vessel disease is nowadays regarded as one of the major vascular causes of dementia. Radiological signs of small vessel disease include small subcortical infarcts, white matter magnetic resonance imaging hyperintensities, lacunes, enlarged perivascular spaces, cerebral microbleeds, and brain atrophy; however, great heterogeneity in clinical symptoms is observed in small vessel disease patients. The pathophysiology of these lesions has been linked to multiple processes, such as hypoperfusion, defective cerebrovascular reactivity, and blood-brain barrier dysfunction. Notably, studies on small vessel disease suggest that blood-brain barrier dysfunction is among the earliest mechanisms in small vessel disease and might contribute to the development of the hallmarks of small vessel disease. Therefore, the purpose of this review is to provide a new foundation in the study of small vessel disease pathology. First, we discuss the main structural domains and functions of the blood-brain barrier. Secondly, we review the most recent evidence on blood-brain barrier dysfunction linked to small vessel disease. Finally, we conclude with a discussion on future perspectives and propose potential treatment targets and interventions.
Collapse
Affiliation(s)
- Ruxue Jia
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, the Netherlands
| | - Gemma Solé-Guardia
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, the Netherlands
| | - Amanda J. Kiliaan
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, the Netherlands
| |
Collapse
|
13
|
Khoshneviszadeh M, Henneicke S, Pirici D, Senthilnathan A, Morton L, Arndt P, Kaushik R, Norman O, Jukkola J, Dunay IR, Seidenbecher C, Heikkinen A, Schreiber S, Dityatev A. Microvascular damage, neuroinflammation and extracellular matrix remodeling in Col18a1 knockout mice as a model for early cerebral small vessel disease. Matrix Biol 2024; 128:39-64. [PMID: 38387749 DOI: 10.1016/j.matbio.2024.02.007] [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: 04/26/2023] [Revised: 02/17/2024] [Accepted: 02/19/2024] [Indexed: 02/24/2024]
Abstract
Collagen type XVIII (COL18) is an abundant heparan sulfate proteoglycan in vascular basement membranes. Here, we asked (i) if the loss of COL18 would result in blood-brain barrier (BBB) breakdown, pathological alterations of small arteries and capillaries and neuroinflammation as found in cerebral small vessel disease (CSVD) and (ii) if such changes may be associated with remodeling of synapses and neural extracellular matrix (ECM). We found that 5-month-old Col18a1-/- mice had elevated BBB permeability for mouse IgG in the deep gray matter, and intravascular erythrocyte accumulations were observed brain-wide in capillaries and arterioles. BBB permeability increased with age and affected cortical regions and the hippocampus in 12-month-old Col18a1-/- mice. None of the Col18a1-/- mice displayed hallmarks of advanced CSVD, such as hemorrhages, and did not show perivascular space enlargement. Col18a1 deficiency-induced BBB leakage was accompanied by activation of microglia and astrocytes, a loss of aggrecan in the ECM of perineuronal nets associated with fast-spiking inhibitory interneurons and accumulation of the perisynaptic ECM proteoglycan brevican and the microglial complement protein C1q at excitatory synapses. As the pathway underlying these regulations, we found increased signaling through the TGF-ß1/Smad3/TIMP-3 cascade. We verified the pivotal role of COL18 for small vessel wall structure in CSVD by demonstrating the protein's involvement in vascular remodeling in autopsy brains from patients with cerebral hypertensive arteriopathy. Our study highlights an association between the alterations of perivascular ECM, extracellular proteolysis, and perineuronal/perisynaptic ECM, as a possible substrate of synaptic and cognitive alterations in CSVD.
Collapse
Affiliation(s)
- Mahsima Khoshneviszadeh
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany; Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany
| | - Solveig Henneicke
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany; Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany
| | - Daniel Pirici
- Department of Histology, University of Medicine and Pharmacy of Craiova, Craiova, Romania
| | | | - Lorena Morton
- Institute of Inflammation and Neurodegeneration, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Philipp Arndt
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany; Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany
| | - Rahul Kaushik
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - Oula Norman
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Finland
| | - Jari Jukkola
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Finland
| | - Ildiko Rita Dunay
- Institute of Inflammation and Neurodegeneration, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany; Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany
| | - Constanze Seidenbecher
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany; Leibniz Institute for Neurobiology, Magdeburg, Germany; Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany
| | - Anne Heikkinen
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Finland
| | - Stefanie Schreiber
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany; Department of Neurology, Otto-von-Guericke University, Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany; Center for Intervention and Research on Adaptive and Maladaptive Brain Circuits Underlying Mental Health (C-I-R-C), Jena-Magdeburg-Halle, Germany.
| | - Alexander Dityatev
- German Center for Neurodegenerative Diseases (DZNE), Magdeburg, Germany; Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany; Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany.
| |
Collapse
|
14
|
Månsson T, Rosso A, Ellström K, Abul-Kasim K, Elmståhl S. Chronic kidney disease and its association with cerebral small vessel disease in the general older hypertensive population. BMC Nephrol 2024; 25:93. [PMID: 38481159 PMCID: PMC10936027 DOI: 10.1186/s12882-024-03528-8] [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: 04/27/2023] [Accepted: 02/28/2024] [Indexed: 03/17/2024] Open
Abstract
BACKGROUND Cerebral small vessel disease can be identified using magnetic resonance imaging, and includes white matter hyperintensities, lacunar infarcts, cerebral microbleeds, and brain atrophy. Cerebral small vessel disease and chronic kidney disease share many risk factors, including hypertension. This study aims to explore an association between chronic kidney disease and cerebral small vessel disease, and also to explore the role of hypertension in this relationship. METHODS With a cross sectional study design, data from 390 older adults was retrieved from the general population study Good Aging in Skåne. Chronic kidney disease was defined as glomerular filtration rate < 60 ml/min/1,73m2. Associations between chronic kidney disease and magnetic resonance imaging markers of cerebral small vessel disease were explored using logistic regression models adjusted for age and sex. In a secondary analysis, the same calculations were performed with the study sample stratified based on hypertension status. RESULTS In the whole group, adjusted for age and sex, chronic kidney disease was not associated with any markers of cerebral small vessel disease. After stratification by hypertension status and adjusted for age and sex, we observed that chronic kidney disease was associated with cerebral microbleeds (OR 1.93, CI 1.04-3.59, p-value 0.037), as well as with cortical atrophy (OR 2.45, CI 1.34-4.48, p-value 0.004) only in the hypertensive group. In the non-hypertensive group, no associations were observed. CONCLUSIONS In this exploratory cross-sectional study, we observed that chronic kidney disease was associated with markers of cerebral small vessel disease only in the hypertensive subgroup of a general population of older adults. This might indicate that hypertension is an important link between chronic kidney disease and cerebral small vessel disease. Further studies investigating the relationship between CKD, CSVD, and hypertension are warranted.
Collapse
Affiliation(s)
- Tomas Månsson
- Department of Clinical Sciences in Malmö, Division of Geriatric Medicine, Lund University and Skåne University Hospital, Jan Waldenströms gata 35, pl 13, 205 02, Malmö, Sweden.
| | - Aldana Rosso
- Department of Clinical Sciences in Malmö, Division of Geriatric Medicine, Lund University and Skåne University Hospital, Jan Waldenströms gata 35, pl 13, 205 02, Malmö, Sweden
| | - Katarina Ellström
- Department of Clinical Sciences in Malmö, Division of Geriatric Medicine, Lund University and Skåne University Hospital, Jan Waldenströms gata 35, pl 13, 205 02, Malmö, Sweden
| | - Kasim Abul-Kasim
- Department of Clinical Sciences in Lund, Division of Diagnostic Radiology, Lund University, 221 85, Lund, Sweden
| | - Sölve Elmståhl
- Department of Clinical Sciences in Malmö, Division of Geriatric Medicine, Lund University and Skåne University Hospital, Jan Waldenströms gata 35, pl 13, 205 02, Malmö, Sweden
| |
Collapse
|
15
|
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
|
16
|
Petrea RE, Pinheiro A, Demissie S, Ekenze O, Aparicio HJ, Satizabal C, Maillard P, DeCarli C, Beiser AS, Seshadri S, Lioutas VA, Romero JR. Hypertension Trends and White Matter Brain Injury in the Offspring Framingham Heart Study Cohort. Hypertension 2024; 81:87-95. [PMID: 37855140 PMCID: PMC10896002 DOI: 10.1161/hypertensionaha.123.21264] [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: 03/23/2023] [Accepted: 10/03/2023] [Indexed: 10/20/2023]
Abstract
BACKGROUND Hypertension is the most potent stroke risk factor and is also related to cerebral small vessel disease. We studied the relation between mid-to-late-life hypertension trends and cerebral white matter injury in community-dwelling individuals from the FHS (Framingham Heart Study). METHODS FHS Offspring cohort participants with available mid-life and late-life blood pressure measurements and brain magnetic resonance imaging were included. Multiple regression analyses were used to relate hypertension trends (normotension-normotension [reference], normotension-hypertension, and hypertension-hypertension) to white matter injury metrics on diffusion tensor imaging (free water, fractional anisotropy, and peak skeletonized mean diffusivity) and Fluid Attenuated Inversion Recovery (white matter hyperintensity volume) by different blood pressure cutoffs (130/80, 140/90, and 150/90 mm Hg). RESULTS We included 1018 participants (mean age 47.3±7.4 years at mid-life and 73.2±7.3 at late-life). At the 140/90 mm Hg cutoff, the hypertension-hypertension trend was associated with higher free water (β, 0.16 [95% CI, 0.03-0.30]; P=0.021) and peak skeletonized mean diffusivity (β, 0.15 [95% CI, 0.01-0.29]; P=0.033). At a 130/80 mm Hg cutoff, the hypertension-hypertension trend had significantly higher free water (β, 0.16 [95% CI, 0.01-0.30]; P=0.035); and the normotension-hypertension (β, 0.24 [95% CI, 0.03-0.44]; P=0.027) and hypertension-hypertension (β, 0.22 [95% CI, 0.04-0.41]; P=0.022) trends had significantly increased white matter hyperintensity volume. Exploratory stratified analysis showed effect modifications by APOE ɛ4 allele and age. CONCLUSIONS Mid-to-late-life hypertension exposure is significantly associated with microstructural and to a lesser extent, visible white matter injury; the effects are observed at both conventional and lower blood pressure cutoffs and are associated with longer duration of hypertension.
Collapse
Affiliation(s)
- Rodica Elena Petrea
- Department of Medicine, Section of Preventive Medicine and Epidemiology, Boston University School of Medicine, Boston, MA, USA
- NHLBI’s Framingham Heart Study, Framingham, MA, USA
| | - Adlin Pinheiro
- NHLBI’s Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Serkalem Demissie
- NHLBI’s Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Oluchi Ekenze
- NHLBI’s Framingham Heart Study, Framingham, MA, USA
- Graduate medical sciences, Boston University School of Medicine
| | - Hugo J. Aparicio
- NHLBI’s Framingham Heart Study, Framingham, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Claudia Satizabal
- NHLBI’s Framingham Heart Study, Framingham, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| | - Pauline Maillard
- Alzheimer’s Disease Center and Imaging of Dementia and Aging (IDeA) Laboratory, Department of Neurology and Center for Neuroscience, University of California at Davis School of Medicine, Sacramento, CA, USA
| | - Charles DeCarli
- Alzheimer’s Disease Center and Imaging of Dementia and Aging (IDeA) Laboratory, Department of Neurology and Center for Neuroscience, University of California at Davis School of Medicine, Sacramento, CA, USA
| | - Alexa S Beiser
- NHLBI’s Framingham Heart Study, Framingham, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | - Sudha Seshadri
- NHLBI’s Framingham Heart Study, Framingham, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health Sciences Center, San Antonio, TX, USA
| | - Vasileios-Arsenios Lioutas
- NHLBI’s Framingham Heart Study, Framingham, MA, USA
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, MA, USA
| | - Jose Rafael Romero
- NHLBI’s Framingham Heart Study, Framingham, MA, USA
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| |
Collapse
|
17
|
Scheuermann BC, Parr SK, Schulze KM, Kunkel ON, Turpin VG, Liang J, Ade CJ. Associations of Cerebrovascular Regulation and Arterial Stiffness With Cerebral Small Vessel Disease: A Systematic Review and Meta-Analysis. J Am Heart Assoc 2023; 12:e032616. [PMID: 37930079 PMCID: PMC10727345 DOI: 10.1161/jaha.123.032616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 10/24/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND Cerebral small vessel disease (cSVD) is a major contributing factor to ischemic stroke and dementia. However, the vascular pathologies of cSVD remain inconclusive. The aim of this systematic review and meta-analysis was to characterize the associations between cSVD and cerebrovascular reactivity (CVR), cerebral autoregulation, and arterial stiffness (AS). METHODS AND RESULTS MEDLINE, Web of Science, and Embase were searched from inception to September 2023 for studies reporting CVR, cerebral autoregulation, or AS in relation to radiological markers of cSVD. Data were extracted in predefined tables, reviewed, and meta-analyses performed using inverse-variance random effects models to determine pooled odds ratios (ORs). A total of 1611 studies were identified; 142 were included in the systematic review, of which 60 had data available for meta-analyses. Systematic review revealed that CVR, cerebral autoregulation, and AS were consistently associated with cSVD (80.4%, 78.6%, and 85.4% of studies, respectively). Meta-analysis in 7 studies (536 participants, 32.9% women) revealed a borderline association between impaired CVR and cSVD (OR, 2.26 [95% CI, 0.99-5.14]; P=0.05). In 37 studies (27 952 participants, 53.0% women) increased AS, per SD, was associated with cSVD (OR, 1.24 [95% CI, 1.15-1.33]; P<0.01). Meta-regression adjusted for comorbidities accounted for one-third of the AS model variance (R2=29.4%, Pmoderators=0.02). Subgroup analysis of AS studies demonstrated an association with white matter hyperintensities (OR, 1.42 [95% CI, 1.18-1.70]; P<0.01). CONCLUSIONS The collective findings of the present systematic review and meta-analyses suggest an association between cSVD and impaired CVR and elevated AS. However, longitudinal investigations into vascular stiffness and regulatory function as possible risk factors for cSVD remain warranted.
Collapse
Affiliation(s)
| | - Shannon K. Parr
- Department of KinesiologyKansas State UniversityManhattanKSUSA
| | | | | | | | - Jia Liang
- Department of Biostatistics, St. Jude Children’s Research HospitalMemphisTNUSA
| | - Carl J. Ade
- Department of KinesiologyKansas State UniversityManhattanKSUSA
- Department of Physician’s Assistant Studies, Kansas State UniversityManhattanKSUSA
- Johnson Cancer Research CenterKansas State UniversityManhattanKSUSA
| |
Collapse
|
18
|
Miyagi T, Ishida A, Shinzato T, Ohya Y. Arterial Stiffness Is Associated With Small Vessel Disease Irrespective of Blood Pressure in Stroke-Free Individuals. Stroke 2023; 54:2814-2821. [PMID: 37846566 DOI: 10.1161/strokeaha.123.042512] [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: 01/09/2023] [Accepted: 08/03/2023] [Indexed: 10/18/2023]
Abstract
BACKGROUND Arterial stiffness and hypertension are important risk factors for cerebral small vessel disease (CSVD). Clinically, there are hypertensive patients with low pulse wave velocity (PWV) and nonhypertensive individuals with high PWV. We aimed to determine the effects of arterial stiffness on CSVD in normotensive individuals. METHODS An observational cross-sectional study was conducted in 1894 stroke-free participants who underwent brain magnetic resonance imaging and brachial-ankle pulse wave velocity (baPWV) measurements at a health checkup between 2013 and 2020. CSVD was defined as any of following: white matter hyperintensities, cerebral microbleeds, silent lacunar infarcts, and enlarged perivascular spaces. baPWV was measured using an automatic oscillometric device. Participants were divided into 4 groups according to the following cutoff points: low blood pressure (BP, <120/80 mm Hg) with low baPWV (<14.63 m/s, a cutoff value that predicted CSVD); high BP (≥120/80 mm Hg) with low baPWV; low BP with high baPWV (≥14.63 m/s); and high BP with high baPWV. RESULTS The mean age of the participants was 57±13 years (41% women). The prevalence of CSVD was 718 (38%), which was higher in the low BP with high baPWV (56%) and high BP with high baPWV (55%) groups than in the high BP with low baPWV (24%) and low BP with low baPWV (22%) groups. Compared with the low BP with low baPWV group, the low BP with high baPWV group (odds ratio, 1.63 [95% CI, 1.09-2.43]) and the high BP with high baPWV group (odds ratio, 1.86 [95% CI, 1.39-2.49]) had a significantly higher multivariable-adjusted risk for CSVD. CONCLUSIONS Individuals with a high baPWV had a higher prevalence of CSVD, independent of BP status. Higher arterial stiffness is likely to be a more important risk factor for CSVD than BP status in stroke-free individuals.
Collapse
Affiliation(s)
- Tomo Miyagi
- Department of Cardiovascular Medicine, Nephrology and Neurology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan (T.M., A.I., Y.O.)
| | - Akio Ishida
- Department of Cardiovascular Medicine, Nephrology and Neurology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan (T.M., A.I., Y.O.)
| | | | - Yusuke Ohya
- Department of Cardiovascular Medicine, Nephrology and Neurology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan (T.M., A.I., Y.O.)
| |
Collapse
|
19
|
Kopczak A, Stringer MS, van den Brink H, Kerkhofs D, Blair GW, van Dinther M, Reyes CA, Garcia DJ, Onkenhout L, Wartolowska KA, Thrippleton MJ, Kampaite A, Duering M, Staals J, Lesnik-Oberstein S, Muir KW, Middeke M, Norrving B, Bousser MG, Mansmann U, Rothwell PM, Doubal FN, van Oostenbrugge R, Biessels GJ, Webb AJS, Wardlaw JM, Dichgans M. Effect of blood pressure-lowering agents on microvascular function in people with small vessel diseases (TREAT-SVDs): a multicentre, open-label, randomised, crossover trial. Lancet Neurol 2023; 22:991-1004. [PMID: 37863608 DOI: 10.1016/s1474-4422(23)00293-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/15/2023] [Accepted: 08/01/2023] [Indexed: 10/22/2023]
Abstract
BACKGROUND Hypertension is the leading risk factor for cerebral small vessel disease. We aimed to determine whether antihypertensive drug classes differentially affect microvascular function in people with small vessel disease. METHODS We did a multicentre, open-label, randomised crossover trial with blinded endpoint assessment at five specialist centres in Europe. We included participants aged 18 years or older with symptomatic sporadic small vessel disease or cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL) and an indication for antihypertensive treatment. Participants were randomly assigned (1:1:1) to one of three sequences of antihypertensive treatment using a computer-generated multiblock randomisation, stratified by study site and patient group. A 2-week washout period was followed by three 4-week periods of oral monotherapy with amlodipine, losartan, or atenolol at approved doses. The primary endpoint was change in cerebrovascular reactivity (CVR) determined by blood oxygen level-dependent MRI response to hypercapnic challenge in normal-appearing white matter from the end of washout to the end of each treatment period. Efficacy analyses were done by intention-to-treat principles in all randomly assigned participants who had at least one valid assessment for the primary endpoint, and analyses were done separately for participants with sporadic small vessel disease and CADASIL. This trial is registered at ClinicalTrials.gov, NCT03082014, and EudraCT, 2016-002920-10, and is terminated. FINDINGS Between Feb 22, 2018, and April 28, 2022, 75 participants with sporadic small vessel disease (mean age 64·9 years [SD 9·9]) and 26 with CADASIL (53·1 years [7·0]) were enrolled and randomly assigned to treatment. 79 participants (62 with sporadic small vessel disease and 17 with CADASIL) entered the primary efficacy analysis. Change in CVR did not differ between study drugs in participants with sporadic small vessel disease (mean change in CVR 1·8 × 10-4%/mm Hg [SE 20·1; 95% CI -37·6 to 41·2] for amlodipine; 16·7 × 10-4%/mm Hg [20·0; -22·3 to 55·8] for losartan; -7·1 × 10-4%/mm Hg [19·6; -45·5 to 31·1] for atenolol; poverall=0·39) but did differ in patients with CADASIL (15·7 × 10-4%/mm Hg [SE 27·5; 95% CI -38·3 to 69·7] for amlodipine; 19·4 × 10-4%/mm Hg [27·9; -35·3 to 74·2] for losartan; -23·9 × 10-4%/mm Hg [27·5; -77·7 to 30·0] for atenolol; poverall=0·019). In patients with CADASIL, pairwise comparisons showed that CVR improved with amlodipine compared with atenolol (-39·6 × 10-4%/mm Hg [95% CI -72·5 to -6·6; p=0·019) and with losartan compared with atenolol (-43·3 × 10-4%/mm Hg [-74·3 to -12·3]; p=0·0061). No deaths occurred. Two serious adverse events were recorded, one while taking amlodipine (diarrhoea with dehydration) and one while taking atenolol (fall with fracture), neither of which was related to study drug intake. INTERPRETATION 4 weeks of treatment with amlodipine, losartan, or atenolol did not differ in their effects on cerebrovascular reactivity in people with sporadic small vessel disease but did result in differential treatment effects in patients with CADASIL. Whether antihypertensive drug classes differentially affect clinical outcomes in people with small vessel diseases requires further research. FUNDING EU Horizon 2020 programme.
Collapse
Affiliation(s)
- Anna Kopczak
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany
| | - Michael S Stringer
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Hilde van den Brink
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, Netherlands
| | - Danielle Kerkhofs
- Department of Neurology and School for Cardiovascular Diseases, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Gordon W Blair
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Maud van Dinther
- Department of Neurology and School for Cardiovascular Diseases, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Carmen Arteaga Reyes
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Daniela Jaime Garcia
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Laurien Onkenhout
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, Netherlands
| | - Karolina A Wartolowska
- Wolfson Centre for Prevention of Stroke and Dementia, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Michael J Thrippleton
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Agniete Kampaite
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Marco Duering
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany; Medical Image Analysis Center and Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Julie Staals
- Department of Neurology and School for Cardiovascular Diseases, Maastricht University Medical Center+, Maastricht, Netherlands
| | | | - Keith W Muir
- School of Psychology and Neuroscience, University of Glasgow, Queen Elizabeth University Hospital, Glasgow, UK
| | - Martin Middeke
- Hypertoniezentrum München, Excellence Centre of the European Society of Hypertension, Munich, Germany
| | - Bo Norrving
- Department of Clinical Sciences Lund, Neurology, Skåne University Hospital, Lund University, Lund, Sweden
| | | | - Ulrich Mansmann
- Institute for Medical Information Processing, Biometry, and Epidemiology, LMU Munich, Munich, Germany
| | - Peter M Rothwell
- Wolfson Centre for Prevention of Stroke and Dementia, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Fergus N Doubal
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Robert van Oostenbrugge
- Department of Neurology and School for Cardiovascular Diseases, Maastricht University Medical Center+, Maastricht, Netherlands
| | - Geert Jan Biessels
- Department of Neurology, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht, Netherlands
| | - Alastair J S Webb
- Wolfson Centre for Prevention of Stroke and Dementia, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Joanna M Wardlaw
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK; UK Dementia Research Institute, University of Edinburgh, Edinburgh, UK
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, University Hospital, LMU Munich, Munich, Germany; Munich Cluster for Systems Neurology, Munich, Germany; German Center for Neurodegenerative Diseases, Munich, Germany; German Centre for Cardiovascular Research, Munich, Germany.
| |
Collapse
|
20
|
Shi H, Cui L, Hui Y, Wu S, Li X, Shu R, Song H, Wang J, Yu P, Chen S, Li J, Yang L, Wang Z, Yang Q, Gao Y. Enlarged Perivascular Spaces in Relation to Cumulative Blood Pressure Exposure and Cognitive Impairment. Hypertension 2023; 80:2088-2098. [PMID: 37476978 DOI: 10.1161/hypertensionaha.123.21453] [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: 04/29/2023] [Accepted: 06/29/2023] [Indexed: 07/22/2023]
Abstract
BACKGROUND Hypertension and enlarged perivascular spaces (EPVS) are thought to be associated with cognitive impairment. However, the correlations among hypertension, EPVS, and cognitive impairment have not been studied yet. We aimed to investigate the relationships between cumulative blood pressure (cBP) exposure with EPVS and cognitive impairment and whether EPVS may mediate the relationship between cBP and cognitive impairment. METHODS A total of 1507 subjects from the Kailuan prospective cohort study were enrolled. cBP was calculated from 2006 to 2022. The effects of cBP, EPVS scores, and cognitive impairment were evaluated using a logistic regression model. The relationships among cBP, EPVS score, and cognitive impairment were analyzed using a mediation model. RESULTS An increase in cBP was positively correlated with an increase in EPVS score. For every SD increase in cBP, the odds ratios (95% CI) of increased EPVS score of the centrum semiovale were 1.67 (1.43-1.95), 1.63 (1.4-1.9), and 1.35 (1.17-1.56), respectively; the odds ratios (95% CI) of increased EPVS score of the basal ganglia were 1.83 (1.56-2.15), 2.01 (1.7-2.36), and 1.31 (1.13-1.52), respectively; and the odds ratios (95% CI) of developing cognitive impairment were 1.28 (1.06-1.53), 1.13 (0.95-1.34), and 1.28 (1.07-1.5), respectively. Basal ganglia-EPVS score accounted for 10.46% to 18.32% of the mediating effects on the relationships of cBP/SD with cognitive impairment. CONCLUSIONS High cBP exposure was an independent risk factor for EPVS, and basal ganglia-EPVS score mediated the effects of cBP on cognitive impairment. REGISTRATION URL: https://www. CLINICALTRIALS gov; Unique identifier: ChiCTR-TNRC-11001489.
Collapse
Affiliation(s)
- Huijing Shi
- Department of Graduate School, Tianjin Medical University, Heping District, China (H. Shi)
- Department of Rheumatology and Immunology, Kailuan General Hospital, Tangshan, Hebei Province, China (H. Shi, L.C., R.S., H. Song, J.W., P.Y.)
| | - Liufu Cui
- Department of Rheumatology and Immunology, Kailuan General Hospital, Tangshan, Hebei Province, China (H. Shi, L.C., R.S., H. Song, J.W., P.Y.)
| | - Ying Hui
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, China (Y.H., X.L., Z.W.)
| | - Shouling Wu
- Department of Cardiology, Kailuan General Hospital, Tangshan, Hebei Province, China (S.W., S.C.)
| | - Xiaoshuai Li
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, China (Y.H., X.L., Z.W.)
| | - Rong Shu
- Department of Rheumatology and Immunology, Kailuan General Hospital, Tangshan, Hebei Province, China (H. Shi, L.C., R.S., H. Song, J.W., P.Y.)
| | - Haicheng Song
- Department of Rheumatology and Immunology, Kailuan General Hospital, Tangshan, Hebei Province, China (H. Shi, L.C., R.S., H. Song, J.W., P.Y.)
| | - Jierui Wang
- Department of Rheumatology and Immunology, Kailuan General Hospital, Tangshan, Hebei Province, China (H. Shi, L.C., R.S., H. Song, J.W., P.Y.)
| | - Ping Yu
- Department of Rheumatology and Immunology, Kailuan General Hospital, Tangshan, Hebei Province, China (H. Shi, L.C., R.S., H. Song, J.W., P.Y.)
| | - Shuohua Chen
- Department of Cardiology, Kailuan General Hospital, Tangshan, Hebei Province, China (S.W., S.C.)
| | | | - Ling Yang
- School of Public Health, North China University of Science and Technology, Tangshan, Hebei (L.Y.)
| | - Zhenchang Wang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, China (Y.H., X.L., Z.W.)
| | - Qing Yang
- Department of Cardiology, Tianjin Medical University General Hospital, Heping District, China (Q.Y., Y.G.)
| | - Yuxia Gao
- Department of Cardiology, Tianjin Medical University General Hospital, Heping District, China (Q.Y., Y.G.)
| |
Collapse
|
21
|
Wu B, Liu F, Sun G, Wang S. Correlation between obstructive sleep apnea and cerebral small vessel disease: a mendelian randomization study. Genes Genomics 2023; 45:1179-1186. [PMID: 37300787 DOI: 10.1007/s13258-023-01402-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Accepted: 05/12/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND Whether obstructive sleep apnea (OSA) is causally associated with an increased risk of cerebral small vessel disease (CSVD) remains controversial. We conducted a two-sample Mendelian randomization (MR) study to clarify the causal relationship between OSA and CSVD risk. METHODS Single-nucleotide polymorphisms associated with OSA at the genome-wide significance level (P < 5 × 10- 8) in the FinnGen consortium were selected as instrumental variables. Summary-level data for white matter hyperintensities (WMHs), lacunar infarctions (LIs), cerebral microbleeds (CMBs), fractional anisotropy (FA), and mean diffusivity (MD) were obtained from three meta-analyses of genome-wide association studies (GWASs). The random-effects inverse-variance weighted (IVW) method was selected for the major analysis. Weighted-median, MR-Egger, MR pleiotropy residual sum and outlier (MR-PRESSO), and leave-one-out analysis methods were implemented for the sensitivity analyses. RESULTS Genetically predicted OSA was not associated with LIs (odds ratio [OR] = 1.10, 95% confidence interval [CI] = 0.86-1.40), WMHs (OR = 0.94, 95% CI = 0.83-1.07), FA (OR = 1.33, 95% CI = 0.75-2.33), MD (OR = 0.93, 95% CI = 0.58-1.47), CMBs (OR = 1.29, 95% CI = 0.86-1.94), mixed CMBs (OR = 1.17, 95% CI = 0.63-2.17), and lobar CMBs (OR = 1.15, 95% CI = 0.75-1.76) in IVW method. The results of the sensitivity analyses were generally consistent with the major analyses. CONCLUSIONS This MR study does not support causal associations between OSA and the risk of CSVD in individuals of European ancestry. These findings need to be further validated in randomized controlled trials, larger cohort studies, and MR studies based on larger GWASs.
Collapse
Affiliation(s)
- Bing Wu
- Department of Neurology, Army 78th Military Group Hospital, 1 Tian Qing Street, Ai Min District, Mudanjiang, 157000, China
| | - Fang Liu
- Department of Neurology, Army 78th Military Group Hospital, 1 Tian Qing Street, Ai Min District, Mudanjiang, 157000, China
| | - Guiyan Sun
- Department of Neurology, Army 78th Military Group Hospital, 1 Tian Qing Street, Ai Min District, Mudanjiang, 157000, China
| | - Shuang Wang
- Department of Neurology, Army 78th Military Group Hospital, 1 Tian Qing Street, Ai Min District, Mudanjiang, 157000, China.
| |
Collapse
|
22
|
Gibson M, Yiallourou S, Pase MP. The Association Between 24-Hour Blood Pressure Profiles and Dementia. J Alzheimers Dis 2023; 94:1303-1322. [PMID: 37458039 DOI: 10.3233/jad-230400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023]
Abstract
Midlife hypertension increases risk for dementia. Around one third of adults have diagnosed hypertension; however, many adults are undiagnosed, or remain hypertensive despite diagnosis or treatment. Since blood pressure (BP) follows a circadian rhythm, ambulatory BP monitoring allows for the assessment of BP over a 24-hour period and provides an important tool for improving the diagnosis and management of hypertension. The measurement of 24-hour BP profiles, especially nocturnal BP, demonstrate better predictive ability for cardiovascular disease and mortality than office measurement. However, few studies have examined 24-hour BP profiles with respect to dementia risk. This is an important topic since improvements in BP management could facilitate the primary prevention of vascular cognitive impairment and dementia. Therefore, this review discusses the evidence linking BP to dementia, with a focus on whether the implementation of 24-hour BP measurements can improve risk prediction and prevention strategies. Pathways linking nocturnal BP to dementia are also discussed as are risk reduction strategies. Overall, limited research suggests an association between 24-hour BP elevation and poorer cognition, cerebral small vessel disease, and dementia. However, most studies were cross-sectional. Further evidence is needed to substantiate 24-hour BP profiles, over and above office BP, as predictors of vascular cognitive impairment and incident dementia.
Collapse
Affiliation(s)
- Madeline Gibson
- The Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| | - Stephanie Yiallourou
- The Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
| | - Matthew P Pase
- The Turner Institute for Brain and Mental Health, Monash University, Clayton, VIC, Australia
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
23
|
Tayler HM, MacLachlan R, Güzel Ö, Miners JS, Love S. Elevated late-life blood pressure may maintain brain oxygenation and slow amyloid-β accumulation at the expense of cerebral vascular damage. Brain Commun 2023; 5:fcad112. [PMID: 37113314 PMCID: PMC10128877 DOI: 10.1093/braincomms/fcad112] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 02/16/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023] Open
Abstract
Hypertension in midlife contributes to cognitive decline and is a modifiable risk factor for dementia. The relationship between late-life hypertension and dementia is less clear. We have investigated the relationship of blood pressure and hypertensive status during late life (after 65 years) to post-mortem markers of Alzheimer's disease (amyloid-β and tau loads); arteriolosclerosis and cerebral amyloid angiopathy; and to biochemical measures of ante-mortem cerebral oxygenation (the myelin-associated glycoprotein:proteolipid protein-1 ratio, which is reduced in chronically hypoperfused brain tissue, and the level of vascular endothelial growth factor-A, which is upregulated by tissue hypoxia); blood-brain barrier damage (indicated by an increase in parenchymal fibrinogen); and pericyte content (platelet-derived growth factor receptor β, which declines with pericyte loss), in Alzheimer's disease (n = 75), vascular (n = 20) and mixed dementia (n = 31) cohorts. Systolic and diastolic blood pressure measurements were obtained retrospectively from clinical records. Non-amyloid small vessel disease and cerebral amyloid angiopathy were scored semiquantitatively. Amyloid-β and tau loads were assessed by field fraction measurement in immunolabelled sections of frontal and parietal lobes. Homogenates of frozen tissue from the contralateral frontal and parietal lobes (cortex and white matter) were used to measure markers of vascular function by enzyme-linked immunosorbent assay. Diastolic (but not systolic) blood pressure was associated with the preservation of cerebral oxygenation, correlating positively with the ratio of myelin-associated glycoprotein to proteolipid protein-1 and negatively with vascular endothelial growth factor-A in both the frontal and parietal cortices. Diastolic blood pressure correlated negatively with parenchymal amyloid-β in the parietal cortex. In dementia cases, elevated late-life diastolic blood pressure was associated with more severe arteriolosclerosis and cerebral amyloid angiopathy, and diastolic blood pressure correlated positively with parenchymal fibrinogen, indicating blood-brain barrier breakdown in both regions of the cortex. Systolic blood pressure was related to lower platelet-derived growth factor receptor β in controls in the frontal cortex and in dementia cases in the superficial white matter. We found no association between blood pressure and tau. Our findings demonstrate a complex relationship between late-life blood pressure, disease pathology and vascular function in dementia. We suggest that hypertension helps to reduce cerebral ischaemia (and may slow amyloid-β accumulation) in the face of increasing cerebral vascular resistance, but exacerbates vascular pathology.
Collapse
Affiliation(s)
- Hannah M Tayler
- Dementia Research Group, Institute of Clinical Neurosciences, Bristol Medical School, University of Bristol, Bristol, BS10 5NB, UK
| | - Robert MacLachlan
- Dementia Research Group, Institute of Clinical Neurosciences, Bristol Medical School, University of Bristol, Bristol, BS10 5NB, UK
| | - Özge Güzel
- Dementia Research Group, Institute of Clinical Neurosciences, Bristol Medical School, University of Bristol, Bristol, BS10 5NB, UK
| | - J Scott Miners
- Dementia Research Group, Institute of Clinical Neurosciences, Bristol Medical School, University of Bristol, Bristol, BS10 5NB, UK
| | - Seth Love
- Correspondence to: Seth Love South West Dementia Brain Bank, University of Bristol Learning & Research Level 1, Southmead Hospital, Bristol, BS10 5NB, UK E-mail:
| |
Collapse
|
24
|
Choi J, Kim JY, Kwon HJ, Choi HJ, Kim SH, Kim S, Lee J, Park JE. Association of cerebral white matter hyperintensities with coronary artery calcium in a healthy population: a cross-sectional study. Sci Rep 2022; 12:21562. [PMID: 36513747 PMCID: PMC9747792 DOI: 10.1038/s41598-022-25654-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 12/02/2022] [Indexed: 12/15/2022] Open
Abstract
In brain magnetic resonance imaging (MRI), white matter hyperintensity (WMH) is a commonly encountered finding and is known to reflect cerebral small vessel disease. The aim of our study was to investigate the association of coronary artery calcium (CAC) with WMH and elucidate the relationship between WMH and atherosclerotic risk factors in a large-scale healthy population. This retrospective study included 1337 individuals who underwent brain MRI and CAC scoring computed tomography at healthcare centers affiliated with a tertiary hospital. Cerebral WMH was defined as Fazekas score greater than 2 on brain MRI. Intracranial artery stenosis (ICAS) was also assessed and determined to be present when stenosis was more than 50% on angiography. The associations of risk factors, CAC score, and ICAS with cerebral WMH were assessed by multivariable regression analysis. In multivariable analysis, categories of higher CAC scores showed increased associations with both periventricular and deep WMHs in a dose-dependent relationship. The presence of ICAS was also significantly related to cerebral WMH, and among the clinical variables, age and hypertension were independent risk factors. In conclusion, CAC showed a significant association with cerebral WMH in a healthy population, which might provide evidence for referring to the CAC score to identify individuals with risk of cerebral WMH.
Collapse
Affiliation(s)
- Jinyoung Choi
- grid.264381.a0000 0001 2181 989XDepartment of Radiology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181 Republic of Korea
| | - Jung Youn Kim
- grid.410886.30000 0004 0647 3511Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Gyeonggi-Do 13496 Republic of Korea
| | - Heon-Ju Kwon
- grid.264381.a0000 0001 2181 989XDepartment of Radiology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181 Republic of Korea
| | - Hye Jeong Choi
- grid.410886.30000 0004 0647 3511Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Gyeonggi-Do 13496 Republic of Korea
| | - Sang Heum Kim
- grid.410886.30000 0004 0647 3511Department of Radiology, CHA Bundang Medical Center, CHA University, Seongnam, Gyeonggi-Do 13496 Republic of Korea
| | - Sinae Kim
- grid.264381.a0000 0001 2181 989XDivision of Biostatistics, Department of R&D Management, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, 03181 Republic of Korea
| | - Jungbin Lee
- grid.412678.e0000 0004 0634 1623Department of Radiology, Soonchunhyang University Bucheon Hospital, Bucheon, Gyeonggi-Do 14584 Republic of Korea
| | - Ji Eun Park
- grid.413967.e0000 0001 0842 2126Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, Seoul, 05505 Republic of Korea
| |
Collapse
|
25
|
Huang XT, Chen CY, Zhang QF, Lu LH, She YL, Fang XY. Meta-analysis of the efficacy of acupuncture in the treatment of the vascular cognitive impairment associated with cerebral small vessel disease. Explore (NY) 2022:S1550-8307(22)00203-8. [DOI: 10.1016/j.explore.2022.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/18/2022] [Accepted: 10/25/2022] [Indexed: 11/08/2022]
|
26
|
Shen J, Yang L, Xu Z, Wei W. Association between Twenty-Four-Hour Ambulatory Blood Pressure Variability and Cerebral Small Vessel Disease Burden in Acute Ischemic Stroke. Behav Neurol 2022; 2022:3769577. [PMID: 36304138 PMCID: PMC9596265 DOI: 10.1155/2022/3769577] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 09/15/2022] [Indexed: 02/02/2025] Open
Abstract
OBJECTIVE This study is aimed at investigating the association between the twenty-four-hour ambulatory blood pressure variability monitoring (ABPM) and cerebral small vessel disease (cSVD) burden in acute ischemic stroke (AIS) patients. METHODS 115 AIS patients with demographics, vascular risk factors, 24 h ABPM, and brain magnetic resonance imaging (MRI) were retrospectively enrolled. 3.0 T MRI was used to assess cSVD burden by combining four MRI markers including white matter hyperintensities (WMHs), cerebral microbleeds (CMBs), perivascular spaces (PVS), and lacunes. Correlation analysis was conducted to detect whether ABPM was associated with cSVD burden in AIS patients. RESULTS 115 AIS patients with mean age 68.77 ± 10.26 years and 75.7% male were enrolled in this study. 112 AIS patients (97.4%) had at least one cSVD marker. Spearman correlation analysis indicated that hypertension was positively correlated with cSVD burden (ρ = 0.21, P = 0.07). High-density lipoprotein (HDL) was negatively correlated with cSVD burden (ρ = -0.21, P = 0.02). Blood pressure variability such as 24 h mean SBP (ρ = 0.23, P = 0.01), day mean SBP (ρ = 0.23, P = 0.01), and night mean SBP (ρ = 0.20, P = 0.04) was positively correlated with higher cSVD burden. Ordinal logistic regression analysis demonstrated that higher 24 h SBP SD and day mean SBP were independent risk factors for cSVD after controlling for other confounders. CONCLUSIONS Higher BPV was significantly related to total cSVD burden in AIS patients. 24 h SBP SD and day mean SBP were independent risk factors for cSVD burden in AIS patients but not DBP or DBP variability.
Collapse
Affiliation(s)
- Jun Shen
- Department of Neurology, Huadong Hospital of Fudan University, 221 West Yan An Road, Shanghai 200040, China
| | - Lu Yang
- Department of Neurology, Huadong Hospital of Fudan University, 221 West Yan An Road, Shanghai 200040, China
| | - Ziwei Xu
- Department of Neurology, Huadong Hospital of Fudan University, 221 West Yan An Road, Shanghai 200040, China
| | - Wenshi Wei
- Department of Neurology, Huadong Hospital of Fudan University, 221 West Yan An Road, Shanghai 200040, China
- Clinical Research Center for Dementias and Cognitive Impairments, Huadong Hospital of Fudan University, 221 West Yan An Road, Shanghai 200040, China
| |
Collapse
|
27
|
Ji X, Tian L, Niu S, Yao S, Qu C. Trimethylamine N-oxide promotes demyelination in spontaneous hypertension rats through enhancing pyroptosis of oligodendrocytes. Front Aging Neurosci 2022; 14:963876. [PMID: 36072486 PMCID: PMC9441869 DOI: 10.3389/fnagi.2022.963876] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/29/2022] [Indexed: 12/24/2022] Open
Abstract
Background Hypertension is a leading risk factor for cerebral small vessel disease (CSVD), a brain microvessels dysfunction accompanied by white matter lesions (WML). Trimethylamine N-oxide (TMAO), a metabolite of intestinal flora, is correlated with cardiovascular and aging diseases. Here, we explored the effect of TMAO on the demyelination of WML. Methods Spontaneous hypertension rats (SHRs) and primary oligodendrocytes were used to explore the effect of TMAO on demyelination in vivo and in vitro. T2-weighted magnetic resonance imaging (MRI) was applied to characterize the white matter hyperintensities (WMH) in rats. TMAO level was evaluated using LC-MS/MS assay. The histopathological changes of corpus callosum were measured by hematoxylin-eosin and luxol fast blue staining. And the related markers were detected by IHC, IF and western blot assay. Mito Tracker Red probe, DCFH-DA assay, flow cytometry based on JC-1 staining and Annexin V-FITC/PI double staining were conducted to evaluate the mitochondrial function, intracellular ROS levels and cell apoptosis. Results SHRs exhibited stronger WMH signals and a higher TMAO level than age-matched normotensive Wistar-kyoto rats (WKY). The corpus callosum region of SHR showed decreased volumes and enhanced demyelination when treated with TMAO. Furthermore, TMAO significantly elevated ROS production and induced NLRP3 inflammasome and impairment of mitochondrial function of oligodendrocytes. More importantly, TMAO enhanced the pyroptosis-related inflammatory death of oligodendrocytes. Conclusion TMAO could cross the blood-brain barrier (BBB) and promote oligodendrocytes pyroptosis via ROS/NLRP3 inflammasome signaling and mitochondrial dysfunction to promote demyelination, revealing a new diagnostic marker for WML under hypertension.
Collapse
Affiliation(s)
- Xiaotan Ji
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, China
- Department of Neurology, Jining No. 1 People’s Hospital, Jining, China
| | - Long Tian
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shenna Niu
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Shumei Yao
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Chuanqiang Qu
- Department of Neurology, Shandong Provincial Hospital, Shandong University, Jinan, China
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
- *Correspondence: Chuanqiang Qu,
| |
Collapse
|
28
|
Cimolai N. Epilepsy and clinically latent cerebrovascular disease. Epileptic Disord 2022; 24:628-629. [PMID: 35770760 DOI: 10.1684/epd.2022.1426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 02/04/2022] [Indexed: 11/17/2022]
|
29
|
Is it possible to prevent cognitive decline among middle-aged and older hypertensive individuals? Hypertens Res 2022; 45:1079-1081. [PMID: 35365800 DOI: 10.1038/s41440-022-00863-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 01/07/2022] [Indexed: 11/08/2022]
|
30
|
Abstract
Although a relationship between traditional cardiovascular risk factors and stroke has long been recognized, these risk factors likely play a role in other aspects of brain health. Clinical stroke is only the tip of the iceberg of vascular brain injury that includes covert infarcts, white matter hyperintensities, and microbleeds. Furthermore, an individual's risk for not only stroke but poor brain health includes not only these traditional vascular risk factors but also lifestyle and genetic factors. The purpose of this narrative review is to summarize the state of the evidence on traditional and nontraditional vascular risk factors and their contributions to brain health. Additionally, we will review important modifiers that interact with these risk factors to increase, or, in some cases, reduce risk of adverse brain health outcomes, with an emphasis on genes and biomarkers associated with Alzheimer disease. Finally, we will consider the importance of social determinants of health in brain health outcomes.
Collapse
Affiliation(s)
- Rebecca F Gottesman
- Stroke Branch, National Institute of Neurological Disorders and Stroke Intramural Research Program, Bethesda, MD (R.F.G.)
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, UTHSA, San Antonio, TX (S.S.).,Department of Neurology, Boston University School of Medicine, Boston, MA (S.S.)
| |
Collapse
|
31
|
van Tuijl RJ, Ruigrok YM, Geurts LJ, van der Schaaf IC, Biessels GJ, Rinkel GJE, Velthuis BK, Zwanenburg JJM. Does the Internal Carotid Artery Attenuate Blood-Flow Pulsatility in Small Vessel Disease? A 7 T 4D-Flow MRI Study. J Magn Reson Imaging 2022; 56:527-535. [PMID: 34997655 PMCID: PMC9546379 DOI: 10.1002/jmri.28062] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/23/2021] [Accepted: 12/23/2021] [Indexed: 11/18/2022] Open
Abstract
Background Increased cerebral blood‐flow pulsatility is associated with cerebral small vessel disease (cSVD). Reduced pulsatility attenuation over the internal carotid artery (ICA) could be a contributing factor to the development of cSVD and could be associated with intracranial ICA calcification (iICAC). Purpose To compare pulsatility, pulsatility attenuation, and distensibility along the ICA between patients with cSVD and controls and to assess the association between iICAC and pulsatility and distensibility. Study Type Retrospective, explorative cross‐sectional study. Subjects A total of 17 patients with cSVD, manifested as lacunar infarcts or deep intracerebral hemorrhage, and 17 age‐ and sex‐matched controls. Field Strength/Sequence Three‐dimensional (3D) T1‐weighted gradient echo imaging and 4D phase‐contrast (PC) MRI with a 3D time‐resolved velocity encoded gradient echo sequence at 7 T. Assessment Blood‐flow velocity pulsatility index (vPI) and arterial distensibility were calculated for seven ICA segments (C1–C7). iICAC presence and volume were determined from available brain CT scans (acquired as part of standard clinical care) in patients with cSVD. Statistical Tests Independent t‐tests and linear mixed models. The threshold for statistically significance was P < 0.05 (two tailed). Results The cSVD group showed significantly higher ICA vPI and significantly lower distensibility compared to controls. Controls showed significant attenuation of vPI over the carotid siphon (−4.9% ± 3.6%). In contrast, patients with cSVD showed no attenuation, but a significant increase of vPI (+6.5% ± 3.1%). iICAC presence and volume correlated positively with vPI (r = 0.578) in patients with cSVD and negatively with distensibility (r = −0.386). Conclusion Decreased distensibility and reduced pulsatility attenuation are associated with increased iICAC and may contribute to cSVD. Confirmation in a larger prospective study is required. Evidence Level 2 Technical Efficacy Stage 2
Collapse
Affiliation(s)
- Rick J van Tuijl
- Department of Radiology, Brain Center, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Ynte M Ruigrok
- Department of Neurology and Neurosurgery, Brain Center, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Lennart J Geurts
- Department of Radiology, Brain Center, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Irene C van der Schaaf
- Department of Radiology, Brain Center, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Geert Jan Biessels
- Department of Neurology and Neurosurgery, Brain Center, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Gabriël J E Rinkel
- Department of Neurology and Neurosurgery, Brain Center, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Birgitta K Velthuis
- Department of Radiology, Brain Center, University Medical Center Utrecht, Utrecht University, the Netherlands
| | - Jaco J M Zwanenburg
- Department of Radiology, Brain Center, University Medical Center Utrecht, Utrecht University, the Netherlands
| |
Collapse
|
32
|
Stefanidou M, Himali JJ, Devinsky O, Romero JR, Ikram MA, Beiser AS, Seshadri S, Friedman D. Vascular risk factors as predictors of epilepsy in older age: The Framingham Heart Study. Epilepsia 2021; 63:237-243. [PMID: 34786697 DOI: 10.1111/epi.17108] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Revised: 10/06/2021] [Accepted: 10/12/2021] [Indexed: 01/14/2023]
Abstract
OBJECTIVE Stroke is the most common cause of epilepsy in older age. Subclinical cerebrovascular disease is believed to underlie some of the 30%-50% of late-onset epilepsy without a known cause (Li et al. Epilepsia. 1997;38:1216; Cleary et al. Lancet. 2004;363:1184). We studied the role of modifiable vascular risk factors in predicting subsequent epilepsy among participants ages 45 or older in the Framingham Heart Study (FHS), a longitudinal, community-based study. METHODS Participants of the Offspring Cohort who attended FHS exam 5 (1991-1995) were included who were at least 45-years-old at that time, had available vascular risk factor data, and epilepsy follow-up (n = 2986, mean age 58, 48% male). Adjudication of epilepsy cases included review of medical charts to exclude seizure mimics and acute symptomatic seizures. The vascular risk factors studied included hypertension, diabetes mellitus, smoking, and hyperlipidemia. The role of the Framingham Stroke Risk Profile score was also investigated. Cox proportional hazards regression models were used for the analyses. RESULTS Fifty-five incident epilepsy cases were identified during a mean of 19 years of follow-up. Hypertension was associated with a near 2-fold risk (hazard ratio [HR]: 1.93, 95% confidence interval [CI]: 1.10-3.37, p = .022) of developing epilepsy, even after adjustment for prevalent and interim stroke. In secondary analysis, excluding patients with normal blood pressure who were receiving anti-HTN (anti-hypertensive) treatment (n = 2613, 50 incident epilepsy cases) the association was (HR: 2.44, 95% CI: 1.36-4.35, p = .003). SIGNIFICANCE Our results offer further evidence that hypertension, a potentially modifiable and highly prevalent vascular risk factor in the general population, increases 2- to 2.5-fold the risk of developing late-onset epilepsy.
Collapse
Affiliation(s)
- Maria Stefanidou
- Framingham Heart Study, Framingham, Massachusetts, USA.,Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Jayandra J Himali
- Framingham Heart Study, Framingham, Massachusetts, USA.,Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA.,Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA.,Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center, San Antonio, Texas, USA.,Department of Population Health Sciences, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Orrin Devinsky
- Department of Neurology, NYU Grossman School of Medicine, New York, New York, USA
| | - Jose R Romero
- Framingham Heart Study, Framingham, Massachusetts, USA.,Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Mohammad Arfan Ikram
- Department of Epidemiology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Alexa S Beiser
- Framingham Heart Study, Framingham, Massachusetts, USA.,Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA.,Department of Biostatistics, Boston University School of Public Health, Boston, Massachusetts, USA
| | - Sudha Seshadri
- Framingham Heart Study, Framingham, Massachusetts, USA.,Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA.,Glenn Biggs Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center, San Antonio, Texas, USA
| | - Daniel Friedman
- Department of Neurology, NYU Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
33
|
Kalaria RN, Sepulveda-Falla D. Cerebral Small Vessel Disease in Sporadic and Familial Alzheimer Disease. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1888-1905. [PMID: 34331941 PMCID: PMC8573679 DOI: 10.1016/j.ajpath.2021.07.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 06/15/2021] [Accepted: 07/02/2021] [Indexed: 01/26/2023]
Abstract
Alzheimer disease (AD) is the most common cause of dementia. Biological definitions of AD are limited to the cerebral burden of amyloid β plaques, neurofibrillary pathology, and neurodegeneration. However, current evidence suggests that various features of small vessel disease (SVD) are part of and covertly modify both sporadic and familial AD. Neuroimaging studies suggest that white matter hyperintensities explained by vascular mechanisms occurs frequently in the AD spectrum. Recent advances have further emphasized that frontal periventricular and posterior white matter hyperintensities are associated with cerebral amyloid angiopathy in familial AD. Although whether SVD markers precede the classically recognized biomarkers of disease is debatable, post-mortem studies show that SVD pathology incorporating small cortical and subcortical infarcts, microinfarcts, microbleeds, perivascular spacing, and white matter attenuation is commonly found in sporadic as well as in mutation carriers with confirmed familial AD. Age-related cerebral vessel pathologies such as arteriolosclerosis and cerebral amyloid angiopathy modify progression or worsen risk by shifting the threshold for cognitive impairment and AD dementia. The incorporation of SVD as a biomarker is warranted in the biological definition of AD. Therapeutic interventions directly reducing the burden of brain amyloid β have had no major impact on the disease or delaying cognitive deterioration, but lowering the risk of vascular disease seems the only rational approach to tackle both early- and late-onset AD dementia.
Collapse
Affiliation(s)
- Rajesh N Kalaria
- Neurovascular Research Group, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom; Department of Human Anatomy, College of Health Sciences, University of Nairobi, Nairobi, Kenya.
| | - Diego Sepulveda-Falla
- Institute of Neuropathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
34
|
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
|
35
|
Cacciottolo M, Morgan TE, Finch CE. Age, sex, and cerebral microbleeds in EFAD Alzheimer disease mice. Neurobiol Aging 2021; 103:42-51. [PMID: 33813349 PMCID: PMC8178216 DOI: 10.1016/j.neurobiolaging.2021.02.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 02/23/2021] [Accepted: 02/23/2021] [Indexed: 01/03/2023]
Abstract
Cerebral microbleeds (MBs) increase at later ages in association with increased cognitive decline and Alzheimer Disease (AD). MB prevalence is also increased by APOE4 and hypertension. In EFAD mice (5XFAD+/-/human APOE+/+), cerebral cortex MBs are most prevalent in E4 females at 6 months, paralleling plaque amyloid. We evaluated MBs at 2, 4, and 6 months in relation to amyloid in plaques and cerebral amyloid angiopathy (CAA) by age, sex, APOE allele, and blood pressure. At 2 mo, MBs were 50% more numerous than plaques, followed by decreased ratio of MBs:Aβ plaques with female excess to 6 mo. The stable size of MBs suggests MBs arise as single events of extravasation, which may "seed" plaque formation. Blood pressure was normal from 2 to 6 months, minimizing a role of hypertension. Memory, assessed by fear conditioning, decreased with age in correlation with MBs and amyloid. Cortical layer analysis showed prevalent MBs and plaque in layers 4 and 5. Contrarily, CAA was prevalent in layers 1 and 2, discounting its contribution to MBs.
Collapse
Affiliation(s)
- Mafalda Cacciottolo
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Todd E Morgan
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Caleb E Finch
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA; Departments of Neurobiology and Molecular Biology, The Dornsife College, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
36
|
Cerebral microbleeds in vascular dementia from clinical aspects to host-microbial interaction. Neurochem Int 2021; 148:105073. [PMID: 34048844 DOI: 10.1016/j.neuint.2021.105073] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 05/15/2021] [Accepted: 05/16/2021] [Indexed: 12/30/2022]
Abstract
Vascular dementia is the second leading cause of dementia after Alzheimer's disease in the elderly population worldwide. Cerebral microbleeds (CMBs) are frequently observed in MRI of elderly subjects and considered as a possible surrogate marker. The number and location of CMBs reflect the severity of diseases and the underlying pathologies may involve cerebral amyloid angiopathy or hypertensive vasculopathy. Accumulating evidence demonstrated the clinicopathological discrepancies of CMBs, the clinical significance of CMBs associated with other MRI markers of cerebral small vessel disease, cognitive impairments, serum, and cerebrospinal fluid biomarkers. Moreover, emerging evidence has shown that genetic factors and gene-environmental interactions might shed light on the underlying etiologies of CMBs, focusing on blood-brain-barrier and inflammation. In this review, we introduce recent genetic and microbiome studies as a cutting-edge approach to figure out the etiology of CMBs through the "microbe-brain-oral axis" and "microbiome-brain-gut axis." Finally, we propose novel concepts, "microvascular matrisome" and "imbalanced proteostasis," which may provide better perspectives for elucidating the pathophysiology of CMBs and future development of therapeutics for vascular dementia using CMBs as a surrogate marker.
Collapse
|
37
|
Newby D, Winchester L, Sproviero W, Fernandes M, Wang D, Kormilitzin A, Launer LJ, Nevado-Holgado AJ. Associations Between Brain Volumes and Cognitive Tests with Hypertensive Burden in UK Biobank. J Alzheimers Dis 2021; 84:1373-1389. [PMID: 34690138 PMCID: PMC8673518 DOI: 10.3233/jad-210512] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/13/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Mid-life hypertension is an established risk factor for cognitive impairment and dementia and related to greater brain atrophy and poorer cognitive performance. Previous studies often have small sample sizes from older populations that lack utilizing multiple measures to define hypertension such as blood pressure, self-report information, and medication use; furthermore, the impact of the duration of hypertension is less extensively studied. OBJECTIVE To investigate the relationship between hypertension defined using multiple measures and length of hypertension with brain measure and cognition. METHODS Using participants from the UK Biobank MRI visit with blood pressure measurements (n = 31,513), we examined the cross-sectional relationships between hypertension and duration of hypertension with brain volumes and cognitive tests using generalized linear models adjusted for confounding. RESULTS Compared with normotensives, hypertensive participants had smaller brain volumes, larger white matter hyperintensities (WMH), and poorer performance on cognitive tests. For total brain, total grey, and hippocampal volumes, those with greatest duration of hypertension had the smallest brain volumes and the largest WMH, ventricular cerebrospinal fluid volumes. For other subcortical and white matter microstructural regions, there was no clear relationship. There were no significant associations between duration of hypertension and cognitive tests. CONCLUSION Our results show hypertension is associated with poorer brain and cognitive health however, the impact of duration since diagnosis warrants further investigation. This work adds further insights by using multiple measures defining hypertension and analysis on duration of hypertension which is a substantial advance on prior analyses-particularly those in UK Biobank which present otherwise similar analyses on smaller subsets.
Collapse
Affiliation(s)
- Danielle Newby
- University of Oxford, Department of Psychiatry, Warneford Hospital, Oxford, UK
| | - Laura Winchester
- University of Oxford, Department of Psychiatry, Warneford Hospital, Oxford, UK
| | - William Sproviero
- University of Oxford, Department of Psychiatry, Warneford Hospital, Oxford, UK
| | - Marco Fernandes
- University of Oxford, Department of Psychiatry, Warneford Hospital, Oxford, UK
| | | | - Andrey Kormilitzin
- University of Oxford, Department of Psychiatry, Warneford Hospital, Oxford, UK
| | | | - Alejo J. Nevado-Holgado
- University of Oxford, Department of Psychiatry, Warneford Hospital, Oxford, UK
- Big Data Institute, University of Oxford, Oxford, UK
- Akrivia Health, Oxford, UK
| |
Collapse
|