201
|
Wu DP, Wei YS, Du YX, Liu LL, Yan QQ, Zhao YD, Yu C, Liu JY, Zhong ZG, Huang JL. Ameliorative Role of Mitochondrial Therapy in Cognitive Function of Vascular Dementia Mice. J Alzheimers Dis 2024; 97:1381-1392. [PMID: 38250768 DOI: 10.3233/jad-230293] [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: 01/23/2024]
Abstract
BACKGROUND Mitochondrial dysfunction plays a vital role in the progression of vascular dementia (VaD). We hypothesized that transfer of exogenous mitochondria might be a beneficial strategy for VaD treatment. OBJECTIVE The study was aimed to investigate the role of mitochondrial therapy in cognitive function of VaD. METHODS The activity and integrity of isolated mitochondria were detected using MitoTracker and Janus Green B staining assays. After VaD mice were intravenously injected with exogenous mitochondria, Morris water maze and passive avoidance tests were used to detect cognitive function of VaD mice. Haematoxylin and eosin, Nissl, TUNEL, and Golgi staining assays were utilized to measure neuronal and synaptic injury in the hippocampus of VaD mice. Detection kits were performed to detect mitochondrial membrane potential (ΔΨ), SOD activity and the levels of ATP, ROS, and MDA in the brains of VaD mice. RESULTS The results showed that isolated mitochondria were intact and active. Mitochondrial therapy could ameliorate cognitive performance of VaD mice. Additionally, mitochondrial administration could attenuate hippocampal neuronal and synaptic injury, improve mitochondrial ΔΨ, ATP level and SOD activity, and reduce ROS and MDA levels in the brains of VaD mice. CONCLUSIONS The study reports profitable effect of mitochondrial therapy against cognitive impairment of VaD, making mitochondrial treatment become a promising therapeutic strategy for VaD.
Collapse
Affiliation(s)
- Deng-Pan Wu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Xuzhou Ruihu Health Management Consulting Co., Ltd, Xuzhou, Jiangsu, China
| | - Yan-Su Wei
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yu-Xuan Du
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Ling-Ling Liu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Qiu-Qing Yan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yuan-Dan Zhao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Chao Yu
- School of Basic Medicine, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jin-Yuan Liu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhen-Guo Zhong
- Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Jin-Lan Huang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Pharmacy School, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Xuzhou Ruihu Health Management Consulting Co., Ltd, Xuzhou, Jiangsu, China
| |
Collapse
|
202
|
Danilov AB, Shindryaeva NN, Borodulina IV, Lunegov TD, Kristeleva DA. [Clinical efficacy and safety of Picamilon in patients with progressive chronic cerebral ischemia]. Zh Nevrol Psikhiatr Im S S Korsakova 2024; 124:71-80. [PMID: 39269299 DOI: 10.17116/jnevro202412408171] [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: 09/15/2024]
Abstract
OBJECTIVE To study the efficacy and safety of the drug Picamilon with various therapy regimens in patients with stage II chronic cerebral ischemia (CCI). MATERIAL AND METHODS An open cohort clinical study involved 50 patients diagnosed with stage II CCI aged 51 to 69 years (average age 62.2±8.98 years). Patients received Picamilon first parenterally 200 mg (100 mg/ml, 2 ml) intravenously for 10 days, then orally in 50 mg tablets 3 times a day for 60 days. The total duration of therapy in the group was 70 days. The study included 3 visits (before treatment, after completion of the course, 1.5 months after completion of treatment). The dynamics of cognitive status according to the Montreal Cognitive Assessment Scale, vegetative disorders according to the A.M. Vane scale, neurological disorders according to the A.I. Fedin scale, and sleep quality according to the Ya. I. Levin scale were compared. The state of cerebral blood flow and endothelium was studied before and after treatment: dopplerography of cranial vessels, assessment of the level of methylated forms of arginine (ADMA, MMA, SDMA) and their ratios. The registration of adverse events against the background of therapy and the tolerability of treatment by patients was also carried out. RESULTS Against the background of Picamilon treatment, a significant positive dynamics of the MoCA scale results was observed in the general sample of patients, increasing in the delayed period (20.9, 24.6 and 25.9, p<0.0001 and p=0.0006); sleep normalization was observed in 55% of patients by Visit 2 and in 81% of patients by Visit 3 (p<0.0001 and p=0.0025). Improvement of neurological functions is noted in 84% of patients, the score on the Fedin A.I. scale significantly decreases after treatment from 17.4±9.34 to 8.06±6.84 (p<0.0001) and to 5.31±5.71 in the delayed period (p=0.0002). Normalization of vegetative status was observed in 38% of patients with stage II CCI, and in 60% of cases there was a decrease in the severity of vegetative dystonia syndrome (p=0.0001). Picamilon therapy has high efficacy in assessing clinical outcomes (100%), good tolerability in 98% of patients and is characterized by a favorable safety profile (in 92% of patients). Picamilon significantly affects the parameters of cerebral hemodynamics: increases the linear velocity of blood flow, reduces the thickness of the intima-media complex and the resistance index. It affects markers of NO metabolism and endothelial function: significantly reduces elevated levels of ADMA and ADMA/MMA and (ADMA+SDMA)/MMA ratios. CONCLUSION The use of Picamilon is effective in patients with stage II CCI in the form of step therapy contributes to a significant regression of neurological deficit, cognitive impairment, improvement of sleep quality and autonomic function; improves vascular endothelial function, reduces the risk of cardiovascular complications. Picamilon is a pathogenetic therapy agent that prevents the progression of CCI.
Collapse
Affiliation(s)
- A B Danilov
- Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - N N Shindryaeva
- Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - I V Borodulina
- Russian Medical Academy of Continuing Postgraduate Education, Moscow, Russia
| | | | - D A Kristeleva
- Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| |
Collapse
|
203
|
Andriuta D, Wiener E, Perron A, Ouin E, Masmoudi I, Thibaut W, Martin J, Roussel M, Constans JM, Aarabi A, Godefroy O. Neuroimaging determinants of cognitive impairment in the memory clinic: how important is the vascular burden? J Neurol 2024; 271:504-518. [PMID: 37777991 DOI: 10.1007/s00415-023-12009-1] [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: 05/10/2023] [Revised: 09/14/2023] [Accepted: 09/15/2023] [Indexed: 10/03/2023]
Abstract
While neurodegenerative and vascular neurocognitive disorder (NCD) often co-occur, the contribution of vascular lesions, especially stroke lesions identified on MRI, to global cognition in a real-life memory clinic population remains unclear. The main objective of this retrospective study was to determine NCD neuroimaging correlates: the GM atrophy pattern and vascular lesions (especially stroke lesion localization by voxel-based lesion-symptom mapping, VLSM) in a memory clinic. We included 336 patients with mild or major NCD who underwent cerebral MRI and a neuropsychological assessment. The GM atrophy pattern (obtained by voxel-based morphometry, VBM) and the stroke lesion localization (obtained by VLSM) associated with G5 z-score (a global cognitive score), were included as independent variables with other neuroimaging and clinical indices in a stepwise linear regression model. The mean age was 70.3 years and the mean MMSE score 21.3. On MRI, 75 patients had at least one stroke lesion. The G 5 z-score was associated with GM density in the pattern selected by the VBM analysis (R2 variation = 0.166, p < 0.001) and the presence of a stroke lesion in the region selected by the VSLM analysis (mainly in the right frontal region; R2 variation = 0.018, p = 0.008). The interaction between the two factors was insignificant (p = 0.374). In conclusion, in this first study combining VBM and VLSM analysis in a memory clinic, global cognition was associated with a specific GM atrophy pattern and the presence of a stroke lesion mainly in the right frontal region.
Collapse
Affiliation(s)
- Daniela Andriuta
- Department of Neurology, Amiens University Medical Center, Amiens University Hospital, 80054, Amiens, France.
- Laboratoire de Neurosciences Fonctionnelles Et Pathologies (UR UPJV 4559), Jules Verne University of Picardy, Amiens, France.
| | - Emmanuel Wiener
- Department of Neurology, Versailles - Le Chesnay Medical Center, Le Chesnay-Rocquencourt, France
| | - Alexandre Perron
- Department of Neurology, Amiens University Medical Center, Amiens University Hospital, 80054, Amiens, France
- Laboratoire de Neurosciences Fonctionnelles Et Pathologies (UR UPJV 4559), Jules Verne University of Picardy, Amiens, France
| | - Elisa Ouin
- Department of Neurology, Amiens University Medical Center, Amiens University Hospital, 80054, Amiens, France
- Laboratoire de Neurosciences Fonctionnelles Et Pathologies (UR UPJV 4559), Jules Verne University of Picardy, Amiens, France
| | - Ines Masmoudi
- Department of Neurology, Amiens University Medical Center, Amiens University Hospital, 80054, Amiens, France
- Laboratoire de Neurosciences Fonctionnelles Et Pathologies (UR UPJV 4559), Jules Verne University of Picardy, Amiens, France
| | - William Thibaut
- Department of Neurology, La Reunion University Medical Center, Site South Saint-Pierre, Saint-Pierre, La Reunion, France
| | - Jeanne Martin
- Department of Neurology, Bretagne Atlantique Medical Center, Vannes, France
| | - Martine Roussel
- Department of Neurology, Amiens University Medical Center, Amiens University Hospital, 80054, Amiens, France
- Laboratoire de Neurosciences Fonctionnelles Et Pathologies (UR UPJV 4559), Jules Verne University of Picardy, Amiens, France
| | - Jean-Marc Constans
- Department of Radiology, Amiens University Medical Center, Amiens, France
| | - Ardalan Aarabi
- Laboratoire de Neurosciences Fonctionnelles Et Pathologies (UR UPJV 4559), Jules Verne University of Picardy, Amiens, France
| | - Olivier Godefroy
- Department of Neurology, Amiens University Medical Center, Amiens University Hospital, 80054, Amiens, France
- Laboratoire de Neurosciences Fonctionnelles Et Pathologies (UR UPJV 4559), Jules Verne University of Picardy, Amiens, France
| |
Collapse
|
204
|
Fang X, Fan F, Border JJ, Roman RJ. Cerebrovascular Dysfunction in Alzheimer's Disease and Transgenic Rodent Models. JOURNAL OF EXPERIMENTAL NEUROLOGY 2024; 5:42-64. [PMID: 38434588 PMCID: PMC10906803 DOI: 10.33696/neurol.5.087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/05/2024]
Abstract
Alzheimer's Disease (AD) and Alzheimer's Disease-Related Dementia (ADRD) are the primary causes of dementia that has a devastating effect on the quality of life and is a tremendous economic burden on the healthcare system. The accumulation of extracellular beta-amyloid (Aβ) plaques and intracellular hyperphosphorylated tau-containing neurofibrillary tangles (NFTs) in the brain are the hallmarks of AD. They are also thought to be the underlying cause of inflammation, neurodegeneration, brain atrophy, and cognitive impairments that accompany AD. The discovery of APP, PS1, and PS2 mutations that increase Aβ production in families with early onset familial AD led to the development of numerous transgenic rodent models of AD. These models have provided new insight into the role of Aβ in AD; however, they do not fully replicate AD pathology in patients. Familial AD patients with mutations that elevate the production of Aβ represent only a small fraction of dementia patients. In contrast, those with late-onset sporadic AD constitute the majority of cases. This observation, along with the failure of previous clinical trials targeting Aβ or Tau and the modest success of recent trials using Aβ monoclonal antibodies, has led to a reappraisal of the view that Aβ accumulation is the sole factor in the pathogenesis of AD. More recent studies have established that cerebral vascular dysfunction is one of the earliest changes seen in AD, and 67% of the candidate genes linked to AD are expressed in the cerebral vasculature. Thus, there is an increasing appreciation of the vascular contribution to AD, and the National Institute on Aging (NIA) and the Alzheimer's Disease Foundation recently prioritized it as a focused research area. This review summarizes the strengths and limitations of the most commonly used transgenic AD animal models and current views about the contribution of Aβ accumulation versus cerebrovascular dysfunction in the pathogenesis of AD.
Collapse
Affiliation(s)
- Xing Fang
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Fan Fan
- Department of Physiology, Augusta University, Augusta, GA 30912, USA
| | - Jane J. Border
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Richard J. Roman
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| |
Collapse
|
205
|
Han S, Jeong S, Choi S, Park SJ, Kim KH, Lee G, Cho Y, Son JS, Park SM. Association of Thyroid Hormone Medication Adherence With Risk of Dementia. J Clin Endocrinol Metab 2023; 109:e225-e233. [PMID: 37515589 DOI: 10.1210/clinem/dgad447] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 04/18/2023] [Accepted: 07/28/2023] [Indexed: 07/31/2023]
Abstract
CONTEXT Recent studies suggest that hypothyroidism is a risk factor for dementia. Based on existing literature, it is unclear if treatment or compliance to treatment is associated with decreased dementia risk in hypothyroidism patients. OBJECTIVE To determine the association between thyroid hormone medication adherence and risk of dementia. METHODS A population-based cohort study following patients with newly diagnosed hypothyroidism until incidence of dementia, death, or December 31, 2020, whichever came earliest. The study comprised 41 554 older adults age 50 and above with newly diagnosed hypothyroidism between 2004 and 2008 who underwent health screening. Risk of dementia was evaluated using multivariable-adjusted Cox proportional hazards regression. RESULTS Among 5188 men (12.5%) and 36 366 (87.5%) women, 2120 cases of incident dementia were identified. After stratification of the participants according to the interquartile range of the medication possession ratio (MPR), the fourth quartile (highest adherence) showed a 14% lower risk of overall dementia compared with the first quartile (lowest adherence) (adjusted hazard ratio 0.86; 95% CI 0.76-0.97). No consistent association was observed between thyroid hormone medication adherence and vascular dementia. After a dichotomous stratification of the MPR, higher MPR (≥0.8) showed a lower risk of Alzheimer disease (adjusted hazard ratio 0.91; 95% CI 0.84-0.99) than lower MPR (<0.8). CONCLUSION Better adherence to thyroid hormone medication may be beneficial in preventing incident dementia, especially Alzheimer disease, in older adults with newly diagnosed hypothyroidism. Along with the resolution of hypothyroid symptoms and the reduction of cardiovascular risk factors, the reduction of dementia risk may be another potential benefit of thyroid hormone medication.
Collapse
Affiliation(s)
- Saemi Han
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, South Korea
- Department of Medicine, Seoul National University College of Medicine, Seoul 03080, South Korea
| | - Seogsong Jeong
- Department of Biomedical Informatics, CHA University School of Medicine, Seongnam 13448, Korea
| | - Seulggie Choi
- Department of Internal Medicine, Seoul National University Hospital, Seoul 03080, Korea
| | - Sun Jae Park
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, South Korea
| | - Kyae Hyung Kim
- Department of Family Medicine, Seoul National University Hospital, Seoul 03080, Korea
| | - Gyeongsil Lee
- Department of Family Medicine, Seoul National University Hospital, Seoul 03080, Korea
| | - Yoosun Cho
- Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 04514, South Korea
| | - Joung Sik Son
- Department of Family Medicine, Korea University Guro Hospital, Seoul 08308, South Korea
| | - Sang Min Park
- Department of Biomedical Sciences, Seoul National University College of Medicine, Seoul 03080, South Korea
- Department of Family Medicine, Seoul National University Hospital, Seoul 03080, Korea
| |
Collapse
|
206
|
Xiang LS, Zhang Y, Xie F, Fei X, Wang Y, Shi Y. Reliability and validity of the Cognitive Assessment for Stroke Patients (Chinese version) for patients with nonaphasic stroke. APPLIED NEUROPSYCHOLOGY. ADULT 2023:1-7. [PMID: 38104350 DOI: 10.1080/23279095.2023.2292236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2023]
Abstract
This study aimed to preliminarily explore the reliability and validity of the Chinese version of the Cognitive Assessment for Stroke Patients (CASP) in patients with nonaphasic stroke and provide a reliable basis for its clinical application in China. The original French version of the CASP was translated into Mandarin Chinese. The study enrolled 58 patients in the neurological center. Mini-Mental State Examination (MMSE), Montreal Cognitive Assessment (MoCA), and CASP were used to evaluate cognitive function. Content validity, structural validity, and concurrent validity, internal consistency, interrater consistency, and retesting reliability were used to evaluate the results. The Spearman correlation coefficient of each item and the total CASP score was between 0.320 and 0.905 (p < .05), showing good content validity. Two initial factors were extracted using principal component analysis and the orthogonal rotation method, with a cumulative contribution rate of 70.100%. Except for the subitem reproducing a copy of a cube, the factor loading of all subitems was >0.5, indicating good construct validity. The total CASP score significantly correlated with the total MMSE (r = 0.796, p < .001) and total MoCA (r = 0.816, p < .001) scores, indicating good concurrent validity. Cronbach's α of the CASP was 0.812, showing good internal consistency. The interrater consistency (ICC > 0.85) and retesting reliability (ICC = 0.7-0.951) were good. The Chinese version of the CASP has good reliability and validity and can play a good auxiliary role in evaluating cognitive dysfunction in patients with stroke.
Collapse
Affiliation(s)
- Li-Sha Xiang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Department of Rehabilitation Medicine, School of Clinical Medicine, Soochow University, Soochow, China
| | - Yi Zhang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Fan Xie
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xiao Fei
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Ya Wang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Yue Shi
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
207
|
Saks DG, Smith EE, Sachdev PS. National and international collaborations to advance research into vascular contributions to cognitive decline. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2023; 6:100195. [PMID: 38226362 PMCID: PMC10788430 DOI: 10.1016/j.cccb.2023.100195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 12/13/2023] [Accepted: 12/13/2023] [Indexed: 01/17/2024]
Abstract
Cerebrovascular disease is the second most common cause of cognitive disorders, usually referred to as vascular contributions to cognitive impairment and dementia (VCID) and makes some contribution to about 70 % of all dementias. Despite its importance, research into VCID has lagged as compared to cognitive impairment due to Alzheimer's disease. There is an increasing appreciation that closing this gap requires large national and international collaborations. This paper highlights 24 notable large-scale national and international efforts to advance research into VCID (MarkVCID, DiverseVCID, DISCOVERY, COMPASS-ND, HBC, RHU SHIVA, UK DRI Vascular Theme, STROKOG, Meta VCI Map, ISGC, ENIGMA-Stroke Recovery, CHARGE, SVDs@target, BRIDGET, CADASIL Consortium, CADREA, AusCADASIL, DPUK, DPAU, STRIVE, HARNESS, FINESSE, VICCCS, VCD-CRE Delphi). These collaborations aim to investigate the effects on cognition from cerebrovascular disease or impaired cerebral blood flow, the mechanisms of action, means of prevention and avenues for treatment. Consensus groups have been developed to harmonise global approaches to VCID, standardise terminology and inform management and treatment, and data sharing is becoming the norm. VCID research is increasingly a global collaborative enterprise which bodes well for rapid advances in this field.
Collapse
Affiliation(s)
- Danit G Saks
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Eric E Smith
- Department of Clinical Neurosciences and Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, University of New South Wales, Sydney, New South Wales, Australia
- Neuropsychiatric Institute, Prince of Wales Hospital, Sydney, New South Wales, Australia
| |
Collapse
|
208
|
Lohman T, Sible I, Kapoor A, Engstrom AC, Alitin JP, Gaubert A, Rodgers KE, Bradford D, Mather M, Han SD, Thayer JF, Nation DA. Blood pressure variability, central autonomic network dysfunction and cerebral small vessel disease in APOE4 carriers. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.12.13.23299556. [PMID: 38168394 PMCID: PMC10760290 DOI: 10.1101/2023.12.13.23299556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
Background Increased blood pressure variability (BPV) is a risk factor for cerebral small vessel disease (CSVD) and neurodegeneration, independent of age and average blood pressure, particularly in apolipoprotein E4 (APOE4) carriers. However, it remains uncertain whether BPV elevation is a cause or a consequence of vascular brain injury, or to what degree injury to the central autonomic network (CAN) may contribute to BPV-associated risk in APOE4 carriers. Methods Independently living older adults (n=70) with no history of stroke or dementia were recruited from the community and underwent 5 minutes of resting beat-to-beat blood pressure monitoring, genetic testing, and brain MRI. Resting BPV, APOE genotype, CSVD burden on brain MRI, and resting state CAN connectivity by fMRI were analyzed. Causal mediation and moderation analysis evaluated BPV and CAN effects on CSVD in APOE4 carriers (n=37) and non-carriers (n=33). Results Higher BPV was associated with the presence and extent of CSVD in APOE4 carriers, but not non-carriers, independent of CAN connectivity (B= 18.92, P= .02), and CAN connectivity did not mediate the relationship between BPV and CSVD. In APOE4 carriers, CAN connectivity moderated the relationship between BPV and CSVD, whereby BPV effects on CSVD were greater in those with lower CAN connectivity (B= 36.43, P= .02). Conclusions Older APOE4 carriers with higher beat-to-beat BPV exhibit more extensive CSVD, independent of average blood pressure, and the strength of CAN connectivity does not mediate these effects. Findings suggest increased BPV is more likely a cause, not a consequence, of CSVD. BPV is more strongly associated with CSVD in APOE4 carriers with lower rsCAN connectivity, suggesting CAN dysfunction and BPV elevation may have synergistic effects on CSVD. Further studies are warranted to understand the interplay between BPV and CAN function in APOE4 carriers.
Collapse
Affiliation(s)
- Trevor Lohman
- University of Southern California Leonard Davis School of Gerontology, Los Angeles, CA, USA
| | - Isabel Sible
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Arunima Kapoor
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| | - Allison C Engstrom
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| | - John Paul Alitin
- University of Southern California Leonard Davis School of Gerontology, Los Angeles, CA, USA
| | - Aimee Gaubert
- University of Southern California Leonard Davis School of Gerontology, Los Angeles, CA, USA
| | - Kathleen E Rodgers
- Center for Innovations in Brain Science, Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - David Bradford
- Center for Innovations in Brain Science, Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Mara Mather
- University of Southern California Leonard Davis School of Gerontology, Los Angeles, CA, USA
| | - S Duke Han
- University of Southern California Leonard Davis School of Gerontology, Los Angeles, CA, USA
| | - Julian F Thayer
- Department of Psychological Science, University of California, Irvine, Irvine, CA, USA
| | - Daniel A Nation
- University of Southern California Leonard Davis School of Gerontology, Los Angeles, CA, USA
- University of Southern California Keck School of Medicine, Los Angeles, CA, USA
| |
Collapse
|
209
|
Chua XY, Torta F, Chong JR, Venketasubramanian N, Hilal S, Wenk MR, Chen CP, Arumugam TV, Herr DR, Lai MKP. Lipidomics profiling reveals distinct patterns of plasma sphingolipid alterations in Alzheimer's disease and vascular dementia. Alzheimers Res Ther 2023; 15:214. [PMID: 38087395 PMCID: PMC10714620 DOI: 10.1186/s13195-023-01359-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) and vascular dementia (VaD) are two of the commonest causes of dementia in the elderly. Of the myriad biomolecules implicated in dementia pathogenesis, sphingolipids have attracted relatively scant research attention despite their known involvement in multiple pathophysiological processes. The potential utility of peripheral sphingolipids as biomarkers in dementia cohorts with high concomitance of cerebrovascular diseases is also unclear. METHODS Using a lipidomics platform, we performed a case-control study of plasma sphingolipids in a prospectively assessed cohort of 526 participants (non-cognitively impaired, NCI = 93, cognitively impaired = 217, AD = 166, VaD = 50) using a lipidomics platform. RESULTS Distinct patterns of sphingolipid alterations were found in AD and VaD, namely an upregulation of d18:1 species in AD compared to downregulation of d16:1 species in VaD. In particular, GM3 d18:1/16:0 and GM3 d18:1/24:1 showed the strongest positive associations with AD. Furthermore, evaluation of sphingolipids panels showed specific combinations with higher sensitivity and specificity for classification of AD (Cer d16:1/24:0. Cer d18:1/16:0, GM3 d16:1/22:0, GM3 d18:1/16:0, SM d16:1/22:0, HexCer d18:1/18:0) and VAD (Cer d16:1/24:0, Cer d18:1/16:0, Hex2Cer d16:1/16:0, HexCer d18:1/18:0, SM d16:1/16:0, SM d16:1/20:0, SM d18:2/22:0) compared to NCI. CONCLUSIONS AD and VaD are associated with distinct changes of plasma sphingolipids, warranting further studies into underlying pathophysiological mechanisms and assessments of their potential utility as dementia biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Xin Ying Chua
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Federico Torta
- Singapore Lipidomics Incubator (SLING), Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Joyce R Chong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Memory, Aging and Cognition Centre, National University Health System, Singapore, Singapore
| | | | - Saima Hilal
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Memory, Aging and Cognition Centre, National University Health System, Singapore, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, Singapore, Singapore
| | - Markus R Wenk
- Singapore Lipidomics Incubator (SLING), Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Christopher P Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Memory, Aging and Cognition Centre, National University Health System, Singapore, Singapore
| | - Thiruma V Arumugam
- Centre for Cardiovascular Biology and Disease Research, Department of Microbiology, Anatomy, Physiology and Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe University, Melbourne, VIC, Australia
| | - Deron R Herr
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Center for Genetic Disorders and Aging Research, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.
- Memory, Aging and Cognition Centre, National University Health System, Singapore, Singapore.
| |
Collapse
|
210
|
Al-Thani M, Goodwin-Trotman M, Bell S, Patel K, Fleming LK, Vilain C, Abramowicz M, Allan SM, Wang T, Cader MZ, Horsburgh K, Van Agtmael T, Sinha S, Markus HS, Granata A. A novel human iPSC model of COL4A1/A2 small vessel disease unveils a key pathogenic role of matrix metalloproteinases. Stem Cell Reports 2023; 18:2386-2399. [PMID: 37977146 PMCID: PMC10724071 DOI: 10.1016/j.stemcr.2023.10.014] [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: 02/17/2023] [Revised: 10/19/2023] [Accepted: 10/20/2023] [Indexed: 11/19/2023] Open
Abstract
Cerebral small vessel disease (SVD) affects the small vessels in the brain and is a leading cause of stroke and dementia. Emerging evidence supports a role of the extracellular matrix (ECM), at the interface between blood and brain, in the progression of SVD pathology, but this remains poorly characterized. To address ECM role in SVD, we developed a co-culture model of mural and endothelial cells using human induced pluripotent stem cells from patients with COL4A1/A2 SVD-related mutations. This model revealed that these mutations induce apoptosis, migration defects, ECM remodeling, and transcriptome changes in mural cells. Importantly, these mural cell defects exert a detrimental effect on endothelial cell tight junctions through paracrine actions. COL4A1/A2 models also express high levels of matrix metalloproteinases (MMPs), and inhibiting MMP activity partially rescues the ECM abnormalities and mural cell phenotypic changes. These data provide a basis for targeting MMP as a therapeutic opportunity in SVD.
Collapse
Affiliation(s)
- Maha Al-Thani
- Department of Clinical Neurosciences, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge and Royal Papworth Hospital, Cambridge, UK
| | - Mary Goodwin-Trotman
- Department of Clinical Neurosciences, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge and Royal Papworth Hospital, Cambridge, UK
| | - Steven Bell
- Department of Clinical Neurosciences, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge and Royal Papworth Hospital, Cambridge, UK
| | - Krushangi Patel
- Department of Clinical Neurosciences, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge and Royal Papworth Hospital, Cambridge, UK
| | - Lauren K Fleming
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Catheline Vilain
- Department of Genetics, Hôpital Erasme, ULB Center of Human Genetics, Universite Libre de Bruxelles, Bruxelles, Belgium
| | - Marc Abramowicz
- Department of Genetics, Hôpital Erasme, ULB Center of Human Genetics, Universite Libre de Bruxelles, Bruxelles, Belgium
| | - Stuart M Allan
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Tao Wang
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, The University of Manchester, Manchester, UK; Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - M Zameel Cader
- Nuffield Department of Clinical Neurosciences, Kavli Institute of Nanoscience Discovery, Dorothy Crowfoot Hodgkin Building, Sherrington Road, University of Oxford, Oxford, UK
| | - Karen Horsburgh
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Tom Van Agtmael
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Sanjay Sinha
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Hugh S Markus
- Department of Neurology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Alessandra Granata
- Department of Clinical Neurosciences, Victor Phillip Dahdaleh Heart and Lung Research Institute, University of Cambridge and Royal Papworth Hospital, Cambridge, UK.
| |
Collapse
|
211
|
Martemucci G, Fracchiolla G, Muraglia M, Tardugno R, Dibenedetto RS, D’Alessandro AG. Metabolic Syndrome: A Narrative Review from the Oxidative Stress to the Management of Related Diseases. Antioxidants (Basel) 2023; 12:2091. [PMID: 38136211 PMCID: PMC10740837 DOI: 10.3390/antiox12122091] [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: 10/10/2023] [Revised: 11/15/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
Metabolic syndrome (MS) is a growing disorder affecting thousands of people worldwide, especially in industrialised countries, increasing mortality. Oxidative stress, hyperglycaemia, insulin resistance, inflammation, dysbiosis, abdominal obesity, atherogenic dyslipidaemia and hypertension are important factors linked to MS clusters of different pathologies, such as diabesity, cardiovascular diseases and neurological disorders. All biochemical changes observed in MS, such as dysregulation in the glucose and lipid metabolism, immune response, endothelial cell function and intestinal microbiota, promote pathological bridges between metabolic syndrome, diabesity and cardiovascular and neurodegenerative disorders. This review aims to summarise metabolic syndrome's involvement in diabesity and highlight the link between MS and cardiovascular and neurological diseases. A better understanding of MS could promote a novel strategic approach to reduce MS comorbidities.
Collapse
Affiliation(s)
- Giovanni Martemucci
- Department of Agricultural and Environmental Sciences, University of Bari Aldo Moro, 70126 Bari, Italy;
| | - Giuseppe Fracchiolla
- Department of Pharmacy–Drug Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.M.); (R.T.); (R.S.D.)
| | - Marilena Muraglia
- Department of Pharmacy–Drug Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.M.); (R.T.); (R.S.D.)
| | - Roberta Tardugno
- Department of Pharmacy–Drug Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.M.); (R.T.); (R.S.D.)
| | - Roberta Savina Dibenedetto
- Department of Pharmacy–Drug Sciences, University of Bari Aldo Moro, 70126 Bari, Italy; (M.M.); (R.T.); (R.S.D.)
| | | |
Collapse
|
212
|
Wei C, Tan C, Zhang X, Shen X, Xu Z, Li J, Xu G. Chronic gastritis may predict risk of cerebral small vessel disease. BMC Gastroenterol 2023; 23:429. [PMID: 38062366 PMCID: PMC10702041 DOI: 10.1186/s12876-023-03009-6] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 10/20/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND AND PURPOSE Chronic gastritis, especially that caused by helicobacter pylori (HP) infection, has been associated with increased risk of ischemic stroke. But the relationship between chronic gastritis and cerebral small vessel disease (CSVD) remains largely undetermined. This study aimed to determine the potential predictors for CSVD, with chronic gastritis and its proxies as alternatives. METHOD Patients aged 18 years or older with indications for electronic gastroscopy were enrolled. Presence of CSVD was evaluated with brain magnetic resonance imaging (MRI) results. Degree of CSVD was scored according to established criteria. Logistic regression analysis was used for identifying possible risk factors for CSVD. RESULTS Of the 1191 enrolled patients, 757 (63.6%) were identified as with, and 434 (36.4%) as without CSVD. Multivariate analysis indicated that patients with chronic atrophic gastritis had an increased risk for CSVD than those without (adjusted odds ratio = 1.58; 95% CI, 1.08-2.32; P < 0.05). CONCLUSIONS Chronic atrophic gastritis is associated with the presence of CSVD. We should routinely screen the presence of CSVD for patients with chronic atrophic gastritis.
Collapse
Affiliation(s)
- Cunsheng Wei
- Department of Neurology, Affiliated Jiangning Hospital With Nanjing Medical University, 168 Gushan Road, Nanjing, 211100, Jiangsu, China
| | - Chang Tan
- Department of Neurology, Affiliated Jiangning Hospital With Nanjing Medical University, 168 Gushan Road, Nanjing, 211100, Jiangsu, China
| | - Xuemei Zhang
- Department of Neurology, Affiliated Jiangning Hospital With Nanjing Medical University, 168 Gushan Road, Nanjing, 211100, Jiangsu, China
| | - Xin Shen
- Department of Neurology, Affiliated Jiangning Hospital With Nanjing Medical University, 168 Gushan Road, Nanjing, 211100, Jiangsu, China
| | - Zongliang Xu
- Department of Neurology, Affiliated Jiangning Hospital With Nanjing Medical University, 168 Gushan Road, Nanjing, 211100, Jiangsu, China
| | - Junrong Li
- Department of Neurology, Affiliated Jiangning Hospital With Nanjing Medical University, 168 Gushan Road, Nanjing, 211100, Jiangsu, China.
| | - Gelin Xu
- Department of Neurology, Jinling Hospital, Medical School of Nanjing University, 305 East Zhongshan Road, Nanjing, 210002, Jiangsu, China.
| |
Collapse
|
213
|
Haqqani AS, Mianoor Z, Star AT, Detcheverry FE, Delaney CE, Stanimirovic DB, Hamel E, Badhwar A. Proteome Profiling of Brain Vessels in a Mouse Model of Cerebrovascular Pathology. BIOLOGY 2023; 12:1500. [PMID: 38132326 PMCID: PMC10740654 DOI: 10.3390/biology12121500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 11/21/2023] [Accepted: 11/23/2023] [Indexed: 12/23/2023]
Abstract
Cerebrovascular pathology that involves altered protein levels (or signaling) of the transforming growth factor beta (TGFβ) family has been associated with various forms of age-related dementias, including Alzheimer disease (AD) and vascular cognitive impairment and dementia (VCID). Transgenic mice overexpressing TGFβ1 in the brain (TGF mice) recapitulate VCID-associated cerebrovascular pathology and develop cognitive deficits in old age or when submitted to comorbid cardiovascular risk factors for dementia. We characterized the cerebrovascular proteome of TGF mice using mass spectrometry (MS)-based quantitative proteomics. Cerebral arteries were surgically removed from 6-month-old-TGF and wild-type mice, and proteins were extracted and analyzed by gel-free nanoLC-MS/MS. We identified 3602 proteins in brain vessels, with 20 demonstrating significantly altered levels in TGF mice. For total and/or differentially expressed proteins (p ≤ 0.01, ≥ 2-fold change), using multiple databases, we (a) performed protein characterization, (b) demonstrated the presence of their RNA transcripts in both mouse and human cerebrovascular cells, and (c) demonstrated that several of these proteins were present in human extracellular vesicles (EVs) circulating in blood. Finally, using human plasma, we demonstrated the presence of several of these proteins in plasma and plasma EVs. Dysregulated proteins point to perturbed brain vessel vasomotricity, remodeling, and inflammation. Given that blood-isolated EVs are novel, attractive, and a minimally invasive biomarker discovery platform for age-related dementias, several proteins identified in this study can potentially serve as VCID markers in humans.
Collapse
Affiliation(s)
- Arsalan S. Haqqani
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON K1A 0R6, Canada; (A.S.H.); (A.T.S.); (C.E.D.); (D.B.S.)
| | - Zainab Mianoor
- Multiomics Investigation of Neurodegenerative Diseases (MIND) Laboratory, 4545 Chemin Queen Mary, Montreal, QC H3W 1W4, Canada; (Z.M.); (F.E.D.)
- Département de Pharmacologie et Physiologie, Institut de Génie Biomédical, Université de Montréal, 2900 Boulevard Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
- Centre de Recherche de l’Institut Universitaire de Gériatrie (CRIUGM), 4545 Chemin Queen Mary, Montreal, QC H3W 1W4, Canada
| | - Alexandra T. Star
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON K1A 0R6, Canada; (A.S.H.); (A.T.S.); (C.E.D.); (D.B.S.)
| | - Flavie E. Detcheverry
- Multiomics Investigation of Neurodegenerative Diseases (MIND) Laboratory, 4545 Chemin Queen Mary, Montreal, QC H3W 1W4, Canada; (Z.M.); (F.E.D.)
- Département de Pharmacologie et Physiologie, Institut de Génie Biomédical, Université de Montréal, 2900 Boulevard Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
- Centre de Recherche de l’Institut Universitaire de Gériatrie (CRIUGM), 4545 Chemin Queen Mary, Montreal, QC H3W 1W4, Canada
| | - Christie E. Delaney
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON K1A 0R6, Canada; (A.S.H.); (A.T.S.); (C.E.D.); (D.B.S.)
| | - Danica B. Stanimirovic
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON K1A 0R6, Canada; (A.S.H.); (A.T.S.); (C.E.D.); (D.B.S.)
| | - Edith Hamel
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, 3801 Rue University, Montreal, QC H3A 2B4, Canada;
| | - AmanPreet Badhwar
- Human Health Therapeutics Research Centre, National Research Council Canada, 1200 Montreal Road, Ottawa, ON K1A 0R6, Canada; (A.S.H.); (A.T.S.); (C.E.D.); (D.B.S.)
- Multiomics Investigation of Neurodegenerative Diseases (MIND) Laboratory, 4545 Chemin Queen Mary, Montreal, QC H3W 1W4, Canada; (Z.M.); (F.E.D.)
- Département de Pharmacologie et Physiologie, Institut de Génie Biomédical, Université de Montréal, 2900 Boulevard Édouard-Montpetit, Montreal, QC H3T 1J4, Canada
- Centre de Recherche de l’Institut Universitaire de Gériatrie (CRIUGM), 4545 Chemin Queen Mary, Montreal, QC H3W 1W4, Canada
- Laboratory of Cerebrovascular Research, Montreal Neurological Institute, McGill University, 3801 Rue University, Montreal, QC H3A 2B4, Canada;
| |
Collapse
|
214
|
Costa Novo J, Rieffel E, Velarde GC, Costa F, Barros P, Veloso M, Costa H, Paredes L, Gregório T, Rodrigues M, Calvão-Pires P, Campolargo A, Battistella V. Shorter Reperfusion Time in Stroke is Associated with Better Cognition. Can J Neurol Sci 2023:1-6. [PMID: 38052728 DOI: 10.1017/cjn.2023.321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
BACKGROUND Cognitive changes that result from cerebrovascular disease contribute to a poor functional outcome with reduced quality of life. Among patients undergoing endovascular therapy (EVT), we aim to assess cognitive function and evaluate the impact of reperfusion time in cognitive performance. METHODS Patients with acute right anterior circulation strokes that underwent EVT between January 2018 and August 2020 at Centro Hospitalar de Vila Nova de Gaia/Espinho, participated in the study. Modified treatment in cerebral infarction (mTICI) assessed the level of recanalization. Cognitive evaluation was assessed with Addenbrooke's Cognitive Examination revised (ACE-R). Multiple linear regression analyses were used to determine the association between time for recanalization and ACE-R. The level of significance adopted was 0.05. RESULTS The mean age of participants was 71.5 (interquartile range [IQR] 62.0-78.2) years, and 50% (22) were women. The median time after stroke was 28.6 months (IQR 18.94-31.55). All patients in our sample had a successful level of recanalization with EVT (mTICI ≥ 2b). Time for recanalization showed an inverse association with the ACE-R (b = -0.0207, P = 0.0203). Also the mRS at 3 months had an inverse association with cognition (b = -5.2803, p = 0.0095). Level of education had a strong and direct relationship with ACE-R results (b = 3.0869, p < 0.0001). CONCLUSIONS Longer time between stroke symptoms and recanalization with EVT in patients with right hemisphere ischemic stroke lead to lower ACE-R scores. Measures to improve door-to-recanalization time are also important for cognitive performance after ischemic stroke.
Collapse
Affiliation(s)
- Joana Costa Novo
- Neurology Department, Centro Hospitalar Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
| | - Evelyne Rieffel
- Psychology department, Pontifical Catholic University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Francisca Costa
- Imagiology Department - Neurorradiology, Centro Hospitalar Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
| | - Pedro Barros
- Neurology Department, Centro Hospitalar Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
- Stroke Unit, Neurology Department, Centro Hospitalar Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
| | - Miguel Veloso
- Neurology Department, Centro Hospitalar Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
- Stroke Unit, Neurology Department, Centro Hospitalar Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
| | - Henrique Costa
- Neurology Department, Centro Hospitalar Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
- Stroke Unit, Neurology Department, Centro Hospitalar Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
| | - Ludovina Paredes
- Stroke Unit, Neurology Department, Centro Hospitalar Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
- Internal Medicine Department, Centro Hospitalar Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
| | - Tiago Gregório
- Stroke Unit, Neurology Department, Centro Hospitalar Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
- Internal Medicine Department, Centro Hospitalar Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
| | - Marta Rodrigues
- Imagiology Department - Neurorradiology, Centro Hospitalar Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
| | - Pedro Calvão-Pires
- Imagiology Department - Neurorradiology, Centro Hospitalar Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
| | - Ana Campolargo
- Physical Medicine and Rehabilitation Department, Centro Hospitalar Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
| | - Valéria Battistella
- Neurology Department, Centro Hospitalar Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
- Stroke Unit, Neurology Department, Centro Hospitalar Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
| |
Collapse
|
215
|
Liang J, Li C, Gao D, Ma Q, Wang Y, Pan Y, Zhang W, Xie W, Zheng F. Association Between Onset Age of Coronary Heart Disease and Incident Dementia: A Prospective Cohort Study. J Am Heart Assoc 2023; 12:e031407. [PMID: 38018492 PMCID: PMC10727352 DOI: 10.1161/jaha.123.031407] [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: 06/19/2023] [Accepted: 09/20/2023] [Indexed: 11/30/2023]
Abstract
BACKGROUND The association of age at coronary heart disease (CHD) onset with incident dementia remains unexplored. This study aimed to examine whether younger onset age of CHD is associated with a higher risk of incident dementia. METHODS AND RESULTS Data were obtained from the UK Biobank. Information on the diagnosis of CHD and dementia was collected at baseline and follow-ups. Propensity score matching method and Cox proportional hazards models were used to evaluate the association between different ages at CHD onset and incident dementia. A total of 432 667 adults (mean±SD age, 56.9±8.1 years) were included, of whom 11.7% had CHD. Compared with participants without CHD, participants with CHD exhibited higher risks of developing all-cause dementia, Alzheimer's disease, and vascular dementia. More importantly, younger age at CHD onset (per 10-year decrease) was significantly associated with elevated risks of all-cause dementia (hazard ratio [HR], 1.25 [95% CI, 1.20-1.30]; P<0.001), Alzheimer's disease (HR, 1.29 [95% CI, 1.20-1.38]; P<0.001), and vascular dementia (HR, 1.22 [95% CI, 1.13-1.31]; P<0.001). After propensity score matching, patients with CHD had significantly higher risks of all-cause dementia, Alzheimer's disease, and vascular dementia than matched controls among all onset age groups, and the HRs gradually elevated with decreasing age at CHD onset. CONCLUSIONS Younger onset age of CHD is associated with higher risks of incident all-cause dementia, Alzheimer's disease, and vascular dementia, underscoring the necessity to pay attention to the neurocognitive status of individuals diagnosed with CHD at younger age to conduct timely interventions to attenuate subsequent risk of incident dementia.
Collapse
Affiliation(s)
- Jie Liang
- Department of Clinical Nursing, School of NursingChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Chenglong Li
- Heart and Vascular Health Research CenterPeking University Clinical Research Institute, Peking University First HospitalBeijingChina
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of EducationBeijingChina
| | - Darui Gao
- Heart and Vascular Health Research CenterPeking University Clinical Research Institute, Peking University First HospitalBeijingChina
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of EducationBeijingChina
| | - Qian Ma
- Department of Cardiology, Beijing Anzhen HospitalCapital Medical UniversityBeijingChina
| | - Yongqian Wang
- Heart and Vascular Health Research CenterPeking University Clinical Research Institute, Peking University First HospitalBeijingChina
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of EducationBeijingChina
| | - Yang Pan
- Department of Clinical Nursing, School of NursingChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Wenya Zhang
- Department of Clinical Nursing, School of NursingChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| | - Wuxiang Xie
- Heart and Vascular Health Research CenterPeking University Clinical Research Institute, Peking University First HospitalBeijingChina
- Key Laboratory of Epidemiology of Major Diseases (Peking University), Ministry of EducationBeijingChina
| | - Fanfan Zheng
- Department of Clinical Nursing, School of NursingChinese Academy of Medical Sciences & Peking Union Medical CollegeBeijingChina
| |
Collapse
|
216
|
Dilliott AA, Berberian SA, Sunderland KM, Binns MA, Zimmer J, Ozzoude M, Scott CJM, Gao F, Lang AE, Breen DP, Tartaglia MC, Tan B, Swartz RH, Rogaeva E, Borrie M, Finger E, Fischer CE, Frank A, Freedman M, Kumar S, Pasternak S, Pollock BG, Rajji TK, Tang-Wai DF, Abrahao A, Turnbull J, Zinman L, Casaubon L, Dowlatshahi D, Hassan A, Mandzia J, Sahlas D, Saposnik G, Grimes D, Marras C, Steeves T, Masellis M, Farhan SMK, Bartha R, Symons S, Hegele RA, Black SE, Ramirez J. Rare neurovascular genetic and imaging markers across neurodegenerative diseases. Alzheimers Dement 2023; 19:5583-5595. [PMID: 37272523 DOI: 10.1002/alz.13316] [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: 03/03/2023] [Revised: 05/09/2023] [Accepted: 05/10/2023] [Indexed: 06/06/2023]
Abstract
INTRODUCTION Cerebral small vessel disease (SVD) is common in patients with cognitive impairment and neurodegenerative diseases such as Alzheimer's and Parkinson's. This study investigated the burden of magnetic resonance imaging (MRI)-based markers of SVD in patients with neurodegenerative diseases as a function of rare genetic variant carrier status. METHODS The Ontario Neurodegenerative Disease Research Initiative study included 520 participants, recruited from 14 tertiary care centers, diagnosed with various neurodegenerative diseases and determined the carrier status of rare non-synonymous variants in five genes (ABCC6, COL4A1/COL4A2, NOTCH3/HTRA1). RESULTS NOTCH3/HTRA1 were found to significantly influence SVD neuroimaging outcomes; however, the mechanisms by which these variants contribute to disease progression or worsen clinical correlates are not yet understood. DISCUSSION Further studies are needed to develop genetic and imaging neurovascular markers to enhance our understanding of their potential contribution to neurodegenerative diseases.
Collapse
Affiliation(s)
- Allison A Dilliott
- Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, McGill University, Montréal, Quebec, Canada
| | - Stephanie A Berberian
- Dr. Sandra Black Centre for Brain Resilience and Recovery, LC Campbell Cognitive Neurology, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Kelly M Sunderland
- Rotman Research Institute, Baycrest Health Sciences, Toronto, Ontario, Canada
| | - Malcolm A Binns
- Rotman Research Institute, Baycrest Health Sciences, Toronto, Ontario, Canada
- Division of Biostatistics, Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
| | - Julia Zimmer
- Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, McGill University, Montréal, Quebec, Canada
| | - Miracle Ozzoude
- Dr. Sandra Black Centre for Brain Resilience and Recovery, LC Campbell Cognitive Neurology, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Christopher J M Scott
- Dr. Sandra Black Centre for Brain Resilience and Recovery, LC Campbell Cognitive Neurology, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Fuqiang Gao
- Dr. Sandra Black Centre for Brain Resilience and Recovery, LC Campbell Cognitive Neurology, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| | - Anthony E Lang
- Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, Ontario, Canada
| | - David P Breen
- Centre for Clinical Brain Sciences, University of Edinburgh; Anne Rowling Regenerative Neurology Clinic, University of Edinburgh; Usher Institute of Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, UK
| | - Maria C Tartaglia
- Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, Ontario, Canada
- Division of Neurology, Toronto Western Hospital, University Health Network, Toronto, Ontario, Canada
| | - Brian Tan
- Rotman Research Institute, Baycrest Health Sciences, Toronto, Ontario, Canada
| | - Richard H Swartz
- Dr. Sandra Black Centre for Brain Resilience and Recovery, LC Campbell Cognitive Neurology, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine (Neurology), Sunnybrook Health Sciences and University of Toronto, Ontario, Canada
- Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Ekaterina Rogaeva
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada
| | - Michael Borrie
- Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
- St. Joseph's Healthcare Centre, London, Ontario, Canada
| | - Elizabeth Finger
- Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Corinne E Fischer
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Andrew Frank
- Department of Medicine (Neurology), University of Ottawa Brain and Mind Research Institute and Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
- Bruyère Research Institute, Ottawa, Ontario, Canada
| | - Morris Freedman
- Rotman Research Institute, Baycrest Health Sciences, Toronto, Ontario, Canada
- Department of Medicine (Neurology), Baycrest Health Sciences, Mt. Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Sanjeev Kumar
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Adult Neurodevelopment and Geriatric Psychiatry, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Stephen Pasternak
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Bruce G Pollock
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Adult Neurodevelopment and Geriatric Psychiatry, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Tarek K Rajji
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Adult Neurodevelopment and Geriatric Psychiatry, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Toronto Dementia Research Alliance, University of Toronto, Toronto, Ontario, Canada
| | - David F Tang-Wai
- Department of Medicine (Neurology), Sunnybrook Health Sciences and University of Toronto, Ontario, Canada
| | - Agessandro Abrahao
- Department of Medicine (Neurology), Sunnybrook Health Sciences and University of Toronto, Ontario, Canada
| | - John Turnbull
- Division of Neurology, Department of Medicine, Hamilton Health Sciences, McMaster University, Hamilton, Canada
| | - Lorne Zinman
- Department of Medicine (Neurology), Sunnybrook Health Sciences and University of Toronto, Ontario, Canada
| | - Leanne Casaubon
- Department of Medicine (Neurology), Sunnybrook Health Sciences and University of Toronto, Ontario, Canada
| | - Dar Dowlatshahi
- Department of Medicine (Neurology), University of Ottawa Brain and Mind Research Institute and Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Ayman Hassan
- Thunder Bay Regional Health Research Institute, Thunder Bay, Ontario, Canada
| | - Jennifer Mandzia
- Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Demetrios Sahlas
- Division of Neurology, Department of Medicine, Hamilton Health Sciences, McMaster University, Hamilton, Canada
| | - Gustavo Saposnik
- Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
- Division of Neurology, Department of Medicine, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - David Grimes
- Department of Medicine (Neurology), University of Ottawa Brain and Mind Research Institute and Ottawa Hospital Research Institute, Ottawa, Ontario, Canada
| | - Connie Marras
- Edmond J. Safra Program in Parkinson's Disease and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, Ontario, Canada
| | - Thomas Steeves
- Division of Neurology, Department of Medicine, St. Michael's Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Mario Masellis
- Dr. Sandra Black Centre for Brain Resilience and Recovery, LC Campbell Cognitive Neurology, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine (Neurology), Sunnybrook Health Sciences and University of Toronto, Ontario, Canada
| | - Sali M K Farhan
- Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, McGill University, Montréal, Quebec, Canada
| | - Robert Bartha
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Sean Symons
- Department of Medical Imaging, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Robert A Hegele
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Sandra E Black
- Dr. Sandra Black Centre for Brain Resilience and Recovery, LC Campbell Cognitive Neurology, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
- Rotman Research Institute, Baycrest Health Sciences, Toronto, Ontario, Canada
- Department of Medicine (Neurology), Sunnybrook Health Sciences and University of Toronto, Ontario, Canada
- Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Joel Ramirez
- Dr. Sandra Black Centre for Brain Resilience and Recovery, LC Campbell Cognitive Neurology, Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
217
|
Hosoki S, Hansra GK, Jayasena T, Poljak A, Mather KA, Catts VS, Rust R, Sagare A, Kovacic JC, Brodtmann A, Wallin A, Zlokovic BV, Ihara M, Sachdev PS. Molecular biomarkers for vascular cognitive impairment and dementia. Nat Rev Neurol 2023; 19:737-753. [PMID: 37957261 DOI: 10.1038/s41582-023-00884-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2023] [Indexed: 11/15/2023]
Abstract
As disease-specific interventions for dementia are being developed, the ability to identify the underlying pathology and dementia subtypes is increasingly important. Vascular cognitive impairment and dementia (VCID) is the second most common cause of dementia after Alzheimer disease, but progress in identifying molecular biomarkers for accurate diagnosis of VCID has been relatively limited. In this Review, we examine the roles of large and small vessel disease in VCID, considering the underlying pathophysiological processes that lead to vascular brain injury, including atherosclerosis, arteriolosclerosis, ischaemic injury, haemorrhage, hypoperfusion, endothelial dysfunction, blood-brain barrier breakdown, inflammation, oxidative stress, hypoxia, and neuronal and glial degeneration. We consider the key molecules in these processes, including proteins and peptides, metabolites, lipids and circulating RNA, and consider their potential as molecular biomarkers alone and in combination. We also discuss the challenges in translating the promise of these biomarkers into clinical application.
Collapse
Affiliation(s)
- Satoshi Hosoki
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Gurpreet K Hansra
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Tharusha Jayasena
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Anne Poljak
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
- Bioanalytical Mass Spectrometry Facility, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW, Australia
| | - Karen A Mather
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Vibeke S Catts
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia
| | - Ruslan Rust
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Abhay Sagare
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jason C Kovacic
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, NY, USA
- Victor Chang Cardiac Research Institute, Darlinghurst, NSW, Australia
- St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Amy Brodtmann
- Department of Neurology, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Anders Wallin
- Department of Psychiatry and Neurochemistry, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, Australia.
| |
Collapse
|
218
|
Guglielmi V, Quaranta D, Masone Iacobucci G, Citro S, Scala I, Genovese D, Brunetti V, Marra C, Calabresi P, Della Marca G. Basal ganglia ischaemic infarction after thrombectomy: cognitive impairment at acute stage. Eur J Neurol 2023; 30:3772-3779. [PMID: 37332125 DOI: 10.1111/ene.15933] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 06/01/2023] [Accepted: 06/13/2023] [Indexed: 06/20/2023]
Abstract
BACKGROUND AND PURPOSE After successful mechanical thrombectomy for middle cerebral artery occlusion, basal ganglia infarction is commonly detectable. Whilst the functional outcome of these patients is often good, less knowledge is available about the cognitive outcome. The aim of our study was to assess the presence of cognitive impairment within 1 week after thrombectomy. METHODS In all, 43 subjects underwent a general cognitive assessment using the Montreal Cognitive Assessment and an extensive battery of tests. Patients were classified as cognitively impaired (CImp) or not (noCImp) according to a Montreal Cognitive Assessment score below 18. RESULTS Cognitively impaired and noCImp subjects did not differ either in their National Institutes of Health Stroke Scale (NIHSS) and modified Rankin Scale (mRS) at admittance, or in their Fazekas score and Alberta Stroke Program Early Computed Tomography Score. At discharge, CImp subjects showed higher scores than noCImp subjects on NIHSS (p = 0.002) and mRS (p < 0.001). The percentage of pathological performances on each neuropsychological test in the whole sample and in CImp and noCImp patients shows a similar cognitive profile between the groups. CONCLUSIONS Some patients who underwent thrombectomy experienced a detectable cognitive impairment that probably led to worse NIHSS and mRS. The neuropsychological profile of such cognitive impairment at the acute stage consists of wide deficits in numerous cognitive domains, suggesting that basal ganglia damage may lead to complex functional impairments.
Collapse
Affiliation(s)
- Valeria Guglielmi
- Dipartimento Neuroscienze, Organi di Senso e Torace, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Davide Quaranta
- Dipartimento Neuroscienze, Organi di Senso e Torace, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Dipartimento di Psicologia, Università Cattolica del Sacro Cuore, Milan, Italy
- Facoltà di Medicina e Chirurgia, Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giovanna Masone Iacobucci
- Unità di Psicologia Clinica, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Salvatore Citro
- Facoltà di Medicina e Chirurgia, Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Irene Scala
- Facoltà di Medicina e Chirurgia, Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Danilo Genovese
- Dipartimento Neuroscienze, Organi di Senso e Torace, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Marlene and Paolo Fresco Institute for Parkinson's and Movement Disorders, NYU Langone Health, New York, New York, USA
| | - Valerio Brunetti
- Dipartimento Neuroscienze, Organi di Senso e Torace, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Camillo Marra
- Dipartimento Neuroscienze, Organi di Senso e Torace, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Facoltà di Medicina e Chirurgia, Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Paolo Calabresi
- Dipartimento Neuroscienze, Organi di Senso e Torace, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Facoltà di Medicina e Chirurgia, Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giacomo Della Marca
- Dipartimento Neuroscienze, Organi di Senso e Torace, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Facoltà di Medicina e Chirurgia, Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
219
|
Hu JR, Abdullah A, Nanna MG, Soufer R. The Brain-Heart Axis: Neuroinflammatory Interactions in Cardiovascular Disease. Curr Cardiol Rep 2023; 25:1745-1758. [PMID: 37994952 PMCID: PMC10908342 DOI: 10.1007/s11886-023-01990-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/30/2023] [Indexed: 11/24/2023]
Abstract
PURPOSE OF REVIEW The role of neuroimmune modulation and inflammation in cardiovascular disease has been historically underappreciated. Physiological connections between the heart and brain, termed the heart-brain axis (HBA), are bidirectional, occur through a complex network of autonomic nerves/hormones and cytokines, and play important roles in common disorders. RECENT FINDINGS At the molecular level, advances in the past two decades reveal complex crosstalk mediated by the sympathetic and parasympathetic nervous systems, the renin-angiotensin aldosterone and hypothalamus-pituitary axes, microRNA, and cytokines. Afferent pathways amplify proinflammatory signals via the hypothalamus and brainstem to the periphery, promoting neurogenic inflammation. At the organ level, while stress-mediated cardiomyopathy is the prototypical disorder of the HBA, cardiac dysfunction can result from a myriad of neurologic insults including stroke and spinal injury. Atrial fibrillation is not necessarily a causative factor for cardioembolic stroke, but a manifestation of an abnormal atrial substrate, which can lead to the development of stroke independent of AF. Central and peripheral neurogenic proinflammatory factors have major roles in the HBA, manifesting as complex bi-directional relationships in common conditions such as stroke, arrhythmia, and cardiomyopathy.
Collapse
Affiliation(s)
- Jiun-Ruey Hu
- Section of Cardiovascular Medicine, Yale School of Medicine, 789 Howard Ave, New Haven, CT, 06519, USA
| | - Ahmed Abdullah
- Section of Cardiovascular Medicine, Yale School of Medicine, 789 Howard Ave, New Haven, CT, 06519, USA
| | - Michael G Nanna
- Section of Cardiovascular Medicine, Yale School of Medicine, 789 Howard Ave, New Haven, CT, 06519, USA
| | - Robert Soufer
- Section of Cardiovascular Medicine, Yale School of Medicine, 789 Howard Ave, New Haven, CT, 06519, USA.
- VA Connecticut Healthcare System, 950 Campbell Ave, -111B, West Haven, CT, 06516, USA.
| |
Collapse
|
220
|
Young HA, Geurts L, Scarmeas N, Benton D, Brennan L, Farrimond J, Kiliaan AJ, Pooler A, Trovò L, Sijben J, Vauzour D. Multi-nutrient interventions and cognitive ageing: are we barking up the right tree? Nutr Res Rev 2023; 36:471-483. [PMID: 36156184 DOI: 10.1017/s095442242200018x] [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: 11/07/2022]
Abstract
As we continue to elucidate the mechanisms underlying age-related brain diseases, the reductionist strategy in nutrition–brain function research has focused on establishing the impact of individual foods. However, the biological processes connecting diet and cognition are complex. Therefore, consideration of a combination of nutritional compounds may be most efficacious. One barrier to establishing the efficacy of multi-nutrient interventions is that the area lacks an established set of evidence-based guidelines for studying their effect on brain health. This review is an output of the International Life Sciences Institute (ILSI) Europe. A multi-disciplinary expert group was assembled with the aim of developing a set of considerations to guide research into the effects of multi-nutrient combinations on brain functions. Consensus recommendations converged on six key issues that should be considered to advance research in this area: (1) establish working mechanisms of the combination and contributions of each individual compound; (2) validate the relevance of the mechanisms for the targeted human condition; (3) include current nutrient status, intake or dietary pattern as inclusion/exclusion criteria in the study design; (4) select a participant population that is clinically and biologically appropriate for all nutritional components of the combination; (5) consider a range of cognitive outcomes; (6) consider the limits of reductionism and the ‘gold standard’ randomised controlled trial. These guiding principles will enhance our understanding of the interactive/complementary activities of dietary components, thereby strengthening the evidence base for recommendations aimed at delaying cognitive decline.
Collapse
Affiliation(s)
| | - Lucie Geurts
- International Life Sciences Institute Europe, Brussels, Belgium
| | - Nikolaos Scarmeas
- 1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
- Department of Neurology, Columbia University, New York, USA
| | - David Benton
- Department of Psychology, Swansea University, Wales, UK
| | - Lorraine Brennan
- UCD Conway Institute of Biomolecular and Biomedical Research, UCD Institute of Food and Health, UCD School of Agriculture and Food Science, Dublin, Republic of Ireland
| | | | - Amanda J Kiliaan
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition and Behaviour, Nijmegen, The Netherlands
| | - Amy Pooler
- Formerly at Nestlé Institute of Health Sciences, Lausanne, Switzerland. Currently at Sangamo Therapeutics, Inc, San Francisco, USA
| | - Laura Trovò
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Vers-chez-les-Blanc, 1000 Lausanne 26, Switzerland
| | - John Sijben
- Danone Nutricia Research, Utrecht, The Netherlands
| | - David Vauzour
- Norwich Medical School, University of East Anglia, Norwich, UK
| |
Collapse
|
221
|
Hedberg L, Kumar A, Skott P, Smedberg JI, Seiger Å, Sandborgh-Englund G, Nordin LE, Kåreholt I, Tzortzakakis A, Westman E, Trulsson M, Ekman U. White matter abnormalities mediate the association between masticatory dysfunction and cognition among older adults. J Oral Rehabil 2023; 50:1422-1431. [PMID: 37710915 DOI: 10.1111/joor.13584] [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: 09/24/2022] [Revised: 06/30/2023] [Accepted: 08/17/2023] [Indexed: 09/16/2023]
Abstract
BACKGROUND Masticatory parameters, such as reduced number of teeth and posterior contacts, have been shown to be associated with reduced cognitive status. The underlying mechanisms that affect these associations, are however, not well understood. OBJECTIVES The study aims to investigate the association between masticatory dysfunction and cognition and explore the mediating effect of brain structure. METHODS In this cross-sectional study, 45 older adults with subjective masticatory dysfunction (mean age 72.3 ± 4.0 years) were included. Mini-Mental State Examination score <25, brain trauma, neurological disease, neurodegenerative disorders, depression or poor Swedish language skills were criteria for exclusion. Cognitive functions (executive function and episodic memory) and masticatory dysfunction defined by functional occluding status (FOS; the number of occluding units and number of remaining teeth) were analysed with partial correlation models. Structural magnetic resonance imaging was performed on 28 feasible participants. Multiple regression analyses were performed to evaluate the predictive value of brain structure and white matter hypointensities (WM-hypo) on cognitive functions. A mediation analysis was applied to assess significant predictor/s of the association between FOS and cognition. RESULTS Both episodic memory and executive functions were positively correlated with FOS. WM-hypo predicted cognitive status (executive function, p ≤ .01). WM-hypo mediated 66.6% (p = 0.06) of the association between FOS and executive functions. CONCLUSION Associations between FOS and cognitive functions are reported, where FOS, a potential modifiable risk factor, was related to both episodic memory and executive functions. The mediating effect of WM-hypo on the association between FOS and executive functions highlights the impact of the vascularisation of the brain on the link between mastication and cognition. The present study provides increased knowledge that bridges the gap between masticatory dysfunction and cognition.
Collapse
Affiliation(s)
- Linn Hedberg
- Folktandvården Eastmaninstitutet, Stockholm, Sweden
- Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
- Academic Centre for Geriatric Dentistry, Stockholm, Sweden
| | - Abhishek Kumar
- Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
- Academic Centre for Geriatric Dentistry, Stockholm, Sweden
| | - Pia Skott
- Folktandvården Eastmaninstitutet, Stockholm, Sweden
- Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
- Academic Centre for Geriatric Dentistry, Stockholm, Sweden
| | - Jan-Ivan Smedberg
- Folktandvården Eastmaninstitutet, Stockholm, Sweden
- Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Åke Seiger
- Academic Centre for Geriatric Dentistry, Stockholm, Sweden
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden
| | - Gunilla Sandborgh-Englund
- Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
- Academic Centre for Geriatric Dentistry, Stockholm, Sweden
| | - Love Engström Nordin
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden
- Department of Diagnostic Medical Physics, Karolinska University Hospital, Stockholm, Sweden
| | - Ingemar Kåreholt
- Aging Research Center, Karolinska Institutet and Stockholm University, Stockholm, Sweden
- School of Health and Welfare, Jönköping University, Jönköping, Sweden
| | - Antonios Tzortzakakis
- Department of Medical Radiation Physics and Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Eric Westman
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden
- Department of Neuroimaging, Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Mats Trulsson
- Department of Dental Medicine, Karolinska Institutet, Stockholm, Sweden
- Academic Centre for Geriatric Dentistry, Stockholm, Sweden
| | - Urban Ekman
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society (NVS), Karolinska Institutet, Stockholm, Sweden
- Women's Health and Allied Health Professionals Theme, Medical Unit, Medical Psychology, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
222
|
Premachandran K, Wilson Alphonse CR, Soundharapandiyan N. Nourishing the Cognition with Millets: A Comprehensive Review of Their Nutritional Impact and Potential as Cognitive Enhancers. Mol Nutr Food Res 2023; 67:e2300450. [PMID: 37899300 DOI: 10.1002/mnfr.202300450] [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: 06/30/2023] [Revised: 08/28/2023] [Indexed: 10/31/2023]
Abstract
Cognition is the mental processes and abilities involved in acquiring, storing, retrieving and using it for decision making. Cognitive decline due to aging, lifestyle factor, chronic health conditions, genetic, and environmental factors are rising global concern and propose a potential threat to the cognitive health. The nutritional imbalance has led to increase in cognitive disorders around the world. Millets can be a nutritional intervention for promoting cognitive health and preventing cognitive decline. Millets has abundant phenolic compounds, flavonoids, and antioxidants to protect against oxidative stress-induced cognitive impairment. Millets exert neuroprotective effects by modulating pathways involved in neuronal-survival, synaptic-plasticity, and release of brain-derived neurotrophic factor. Millets demonstrates anti-inflammatory properties by regulating inflammatory-pathways and suppressing cytokines associated with cognitive impairment. Millets maintain healthy gut microbiota by producing metabolites such as short-chain fatty acids, which influence brain function and cognition. However, further research is needed to elucidate the underlying mechanisms and on optimizing the proportion do exploit its potential. Implementing millet-based dietary strategies through public health initiatives and educational programs can be a practical approach to support cognitive health across populations. Harnessing the potential of millets as a nutritional intervention offers a promising avenue for promoting cognitive health and improving the quality of life.
Collapse
Affiliation(s)
| | - Carlton Ranjith Wilson Alphonse
- Neuroscience Lab, Molecular and Nanomedicine Research Unit, Centre for Nanoscience and Nanotechnology, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, 600119, India
| | - Nandhagopal Soundharapandiyan
- Neuroscience Lab, Molecular and Nanomedicine Research Unit, Centre for Nanoscience and Nanotechnology, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, 600119, India
| |
Collapse
|
223
|
Waziry R, Williams OA. Alzheimer Disease: A Life-Limiting Disease With a Burden of a Terminal Illness. Neurol Clin Pract 2023; 13:e200208. [PMID: 37854176 PMCID: PMC10581074 DOI: 10.1212/cpj.0000000000200208] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 09/15/2023] [Indexed: 10/20/2023]
Abstract
Purpose of Review An estimated 6.5 million Americans live with Alzheimer dementia. Better understanding of advanced stages of Alzheimer disease (AD) and timely monitoring of its preventable complications would translate to improved survival and quality of life in this population. Recent Findings In this perspective review, we proposed shifting the narrative to recognizing AD as a chronic life-limiting illness instead of a terminal disease. In support of this view, we discussed the following: (1) the biochemical, cellular (preclinical), and clinical phases of AD; (2) survival after AD; (3) AD therapeutics and potential implications for the population with AD in the advanced stages. Summary On the bases of the prolonged preclinical phase in AD, promising advances in AD therapeutics and the varying survival after AD, we proposed a new classification for AD and more broadly neurodegenerative disorders to be recognized as chronic life-limiting illnesses rather than terminal diseases with important implications for patients with AD in the advanced stages given the challenges that are specific to this population.
Collapse
Affiliation(s)
- Reem Waziry
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, Columbia University, NY
| | - Olajide A Williams
- Department of Neurology, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, Columbia University, NY
| |
Collapse
|
224
|
Hassen CB, Machado‐Fragua MD, Landré B, Fayosse A, Dumurgier J, Kivimaki M, Sabia S, Singh‐Manoux A. Change in lipids before onset of dementia, coronary heart disease, and mortality: A 28-year follow-up Whitehall II prospective cohort study. Alzheimers Dement 2023; 19:5518-5530. [PMID: 37243914 PMCID: PMC10679471 DOI: 10.1002/alz.13140] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 04/07/2023] [Indexed: 05/29/2023]
Abstract
INTRODUCTION The association of lipids with dementia remains a subject of debate. Using data from 7,672 participants of the Whitehall II prospective cohort study, we examined whether timing of exposure, length of follow-up, or sex modifies this association. METHODS Twelve markers of lipid levels were measured from fasting blood and eight among them a further five times. We performed time-to-event as well as trajectory analyses. RESULTS No associations were observed in men; in women most lipids were associated with the risk of dementia, but only for events occurring after the first 20 years of follow-up. Differences in lipid trajectories in men emerged only in the years immediately before diagnosis whereas in women total cholesterol (TC), LDL-cholesterol (LDL-C), non-HDL-cholesterol (non-HDL-C), TC/HDL-C, and LDL-C/HDL-C were higher in midlife among dementia cases before declining progressively. DISCUSSION Abnormal lipid levels in midlife seem to be associated with a higher risk of dementia in women.
Collapse
Affiliation(s)
- Céline Ben Hassen
- Université Paris Cité, Inserm U1153, Epidemiology of Ageing and Neurodegenerative diseasesParisFrance
| | - Marcos D Machado‐Fragua
- Université Paris Cité, Inserm U1153, Epidemiology of Ageing and Neurodegenerative diseasesParisFrance
| | - Benjamin Landré
- Université Paris Cité, Inserm U1153, Epidemiology of Ageing and Neurodegenerative diseasesParisFrance
| | - Aurore Fayosse
- Université Paris Cité, Inserm U1153, Epidemiology of Ageing and Neurodegenerative diseasesParisFrance
| | - Julien Dumurgier
- Université Paris Cité, Inserm U1153, Epidemiology of Ageing and Neurodegenerative diseasesParisFrance
- Cognitive Neurology Center, Lariboisière – Fernand Widal Hospital, AP‐HPUniversité Paris CitéParisFrance
| | - Mika Kivimaki
- Department of Mental Health of Older People, Faculty of Brain SciencesUniversity College LondonLondonUK
| | - Séverine Sabia
- Université Paris Cité, Inserm U1153, Epidemiology of Ageing and Neurodegenerative diseasesParisFrance
- Department of Mental Health of Older People, Faculty of Brain SciencesUniversity College LondonLondonUK
| | - Archana Singh‐Manoux
- Université Paris Cité, Inserm U1153, Epidemiology of Ageing and Neurodegenerative diseasesParisFrance
- Department of Mental Health of Older People, Faculty of Brain SciencesUniversity College LondonLondonUK
| |
Collapse
|
225
|
Sabayan B, Goudarzi R, Ji Y, Borhani‐Haghighi A, Olson‐Bullis BA, Murray AM, Sedaghat S. Intracranial Atherosclerosis Disease Associated With Cognitive Impairment and Dementia: Systematic Review and Meta-Analysis. J Am Heart Assoc 2023; 12:e032506. [PMID: 37955546 PMCID: PMC10727275 DOI: 10.1161/jaha.123.032506] [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: 09/03/2023] [Accepted: 11/03/2023] [Indexed: 11/14/2023]
Abstract
BACKGROUND Intracranial atherosclerosis disease (ICAD) alters cerebrovascular hemodynamics and brain structural integrity. Multiple studies have evaluated the link between ICAD and cognitive impairment, with mixed results. This study aims to systematically review and summarize the current evidence on this link. METHODS AND RESULTS PubMed, EMBASE, PsycInfo, and Web of Science were searched from 2000 to 2023 without language restriction. Cross-sectional and prospective cohort studies as well as postmortem studies were included. Studies containing data on the link between ICAD, defined as at least 50% stenosis in 1 intracranial vessel, and cognitive impairment and dementia were screened by 2 independent reviewers. A total of 22 (17 observational and 5 postmortem) unique studies, comprising 11 184 individuals (average age range, 59.8-87.6 years; 45.7% women; 36.5% Asian race), were included in the systematic review. Seven of 10 cross-sectional studies and 5 of 7 prospective studies showed a significant association between ICAD and cognitive impairment. In the pooled analysis, ICAD was associated with greater cognitive impairment (measure of association, 1.87 [95% CI, 1.49-2.35]). Meta-regression analyses did not show a significant impact of age, sex, and race. All postmortem studies showed that patients with Alzheimer disease and vascular dementia had a higher burden of ICAD compared with controls. CONCLUSIONS This study shows that ICAD is associated with cognitive impairment and dementia across age, sex, and race groups. Our findings may underscore the need to develop individualized dementia preventive care plans in patients with ICAD.
Collapse
Affiliation(s)
- Behnam Sabayan
- Department of Neurology, Hennepin Healthcare Research InstituteHennepin County Medical CenterMinneapolisMN
- Division of Epidemiology and Community Health, School of Public HealthUniversity of MinnesotaMinneapolisMN
| | - Roham Goudarzi
- Faculty of ScienceUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - Yuekai Ji
- Division of Epidemiology and Community Health, School of Public HealthUniversity of MinnesotaMinneapolisMN
| | | | | | - Anne M. Murray
- Berman Center for Outcomes and Clinical Research and Geriatrics Division, Department of MedicineHennepin Healthcare Research InstituteMinneapolisMN
| | - Sanaz Sedaghat
- Division of Epidemiology and Community Health, School of Public HealthUniversity of MinnesotaMinneapolisMN
| |
Collapse
|
226
|
Bidzan L, Jurek P, Olech M, Bidzan-Wiącek M, Bidzan-Bluma I, Bidzan M. Somatic comorbidity and the progression of cognitive impairment. Front Aging Neurosci 2023; 15:1219449. [PMID: 38046465 PMCID: PMC10691469 DOI: 10.3389/fnagi.2023.1219449] [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: 05/09/2023] [Accepted: 11/02/2023] [Indexed: 12/05/2023] Open
Abstract
Background There are usually multiple factors underlying dementia in old age. Somatic comorbidity is one important element that influences the progression of cognitive impairment. Objective The goal of this study was to assess the relationship between the progression of cognitive impairment and the presence and severity of comorbidities based on a four-year observation. Material Out of 128 patients from the Clinic for Outpatients in Gdansk, who were recruited into the study based on the criteria of the Working Group on Mild Cognitive Impairment, a total of 93 participants completed the four-year observation. Only the data from participants who completed the full period of observations were analysed. The mean age of the group was M = 75.93 (SD = 9.43). The level of progression of cognitive impairment was measured using the Clinical Dementia Rating Scale - Sum of Boxes, the severity of comorbidities was measured using the modified Cumulative Illness Rating Scale, and, additionally, at the time of inclusion in the study, participants were assessed using the MMSE scale and the Activity Scale, and sociodemographic data were collected. The Generalized Estimating Equations method was employed to fit a marginal model for analyzing the data collected in a repeated measures design. The tested model elucidated the role of the overall severity of comorbidities in explaining the progression of cognitive impairment, while controlling for everyday activity and basic demographic variables. Results During the four-year observation, a significant decline in cognitive function (B = 1.86, p < 0.01) was observed in the examined sample. The statistical analysis revealed that individuals with higher overall severity of comorbidities exhibited significantly more pronounced progression of cognitive impairment over time. Regarding particular comorbidities, metabolic diseases were found to be associated with a poorer prognosis (rho = 0.41, p < 0.05). Furthermore, a time physical activity interaction was identified as predicting cognitive impairment, indicating that individuals who were more physically active at the beginning of the study exhibited significantly less pronounced progression of cognitive impairment over the course of the 4 years. Conclusion This study suggests the important roles of comorbidities and physical activity for the prognosis of mild cognitive impairment.
Collapse
Affiliation(s)
- Leszek Bidzan
- Faculty of Medicine, Medical University of Gdańsk, Gdańsk, Poland
- Department of Health Sciences, Pomeranian University in Słupsk, Słupsk, Poland
| | - Paweł Jurek
- Institute of Psychology, University of Gdańsk, Gdańsk, Poland
| | - Michał Olech
- Faculty of Health Sciences, Medical University of Gdańsk, Gdańsk, Poland
| | | | - Ilona Bidzan-Bluma
- Institute of Psychology, University of Gdańsk, Gdańsk, Poland
- Institute of Pedagogy and Languages, University of Applied Sciences in Elbląg, Elbląg, Poland
| | - Mariola Bidzan
- Institute of Psychology, University of Gdańsk, Gdańsk, Poland
- Institute of Pedagogy and Languages, University of Applied Sciences in Elbląg, Elbląg, Poland
| |
Collapse
|
227
|
Liu M, Zhang J, Wang Y, Zhou Y, Xie F, Guo Q, Shi F, Zhang H, Wang Q, Shen D. A common spectrum underlying brain disorders across lifespan revealed by deep learning on brain networks. iScience 2023; 26:108244. [PMID: 38026184 PMCID: PMC10651682 DOI: 10.1016/j.isci.2023.108244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/26/2023] [Accepted: 10/16/2023] [Indexed: 12/01/2023] Open
Abstract
Brain disorders in the early and late life of humans potentially share pathological alterations in brain functions. However, the key neuroimaging evidence remains unrevealed for elucidating such commonness and the relationships among these disorders. To explore this puzzle, we build a restricted single-branch deep learning model, using multi-site functional magnetic resonance imaging data (N = 4,410, 6 sites), for classifying 5 different early- and late-life brain disorders from healthy controls (cognitively unimpaired). Our model achieves 62.6 ± 1.9% overall classification accuracy and thus supports us in detecting a set of commonly affected functional subnetworks, including default mode, executive control, visual, and limbic networks. In the deep-layer representation of data, we observe young and aging patients with disorders are continuously distributed, which is in line with the clinical concept of the "spectrum of disorders." The relationships among brain disorders from the revealed spectrum promote the understanding of disorder comorbidities and time associations in the lifespan.
Collapse
Affiliation(s)
- Mianxin Liu
- School of Biomedical Engineering, State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
- Shanghai Artificial Intelligence Laboratory, Shanghai 200232, China
| | - Jingyang Zhang
- School of Biomedical Engineering, State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Yao Wang
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China
| | - Yan Zhou
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200001, China
| | - Fang Xie
- PET Center, Huashan Hospital, Fudan University, Shanghai 200040, China
| | - Qihao Guo
- Department of Gerontology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200233, China
| | - Feng Shi
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd, Shanghai 200232, China
| | - Han Zhang
- School of Biomedical Engineering, State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Qian Wang
- School of Biomedical Engineering, State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Dinggang Shen
- School of Biomedical Engineering, State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd, Shanghai 200232, China
- Shanghai Clinical Research and Trial Center, Shanghai 201210, China
| |
Collapse
|
228
|
Akyol O, Akyol S, Chou MC, Chen S, Liu CK, Selek S, Soares JC, Chen CH. Lipids and lipoproteins may play a role in the neuropathology of Alzheimer's disease. Front Neurosci 2023; 17:1275932. [PMID: 38033552 PMCID: PMC10687420 DOI: 10.3389/fnins.2023.1275932] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 10/30/2023] [Indexed: 12/02/2023] Open
Abstract
Alzheimer's disease (AD) and other classes of dementia are important public health problems with overwhelming social, physical, and financial effects for patients, society, and their families and caregivers. The pathophysiology of AD is poorly understood despite the extensive number of clinical and experimental studies. The brain's lipid-rich composition is linked to disturbances in lipid homeostasis, often associated with glucose and lipid abnormalities in various neurodegenerative diseases, including AD. Moreover, elevated low-density lipoprotein (LDL) cholesterol levels may be related to a higher probability of AD. Here, we hypothesize that lipids, and electronegative LDL (L5) in particular, may be involved in the pathophysiology of AD. Although changes in cholesterol, triglyceride, LDL, and glucose levels are seen in AD, the cause remains unknown. We believe that L5-the most electronegative subfraction of LDL-may be a crucial factor in understanding the involvement of lipids in AD pathology. LDL and L5 are internalized by cells through different receptors and mechanisms that trigger separate intracellular pathways. One of the receptors involved in L5 internalization, LOX-1, triggers apoptotic pathways. Aging is associated with dysregulation of lipid homeostasis, and it is believed that alterations in lipid metabolism contribute to the pathogenesis of AD. Proposed mechanisms of lipid dysregulation in AD include mitochondrial dysfunction, blood-brain barrier disease, neuronal signaling, inflammation, and oxidative stress, all of which lead ultimately to memory loss through deficiency of synaptic integration. Several lipid species and their receptors have essential functions in AD pathogenesis and may be potential biomarkers.
Collapse
Affiliation(s)
- Omer Akyol
- Molecular Cardiology, Vascular and Medicinal Research, The Texas Heart Institute, Houston, TX, United States
| | | | - Mei-Chuan Chou
- Department of Neurology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shioulan Chen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-Kuan Liu
- Institute of Precision Medicine, College of Medicine, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Salih Selek
- Department of Psychiatry and Behavioral Sciences, UTHealth Houston McGovern Medical School, Houston, TX, United States
| | - Jair C. Soares
- Department of Psychiatry and Behavioral Sciences, UTHealth Houston McGovern Medical School, Houston, TX, United States
| | - Chu-Huang Chen
- Molecular Cardiology, Vascular and Medicinal Research, The Texas Heart Institute, Houston, TX, United States
| |
Collapse
|
229
|
Zhang Q, Liu X, Gao S, Yan S, Li A, Wei Z, Han S, Hou Y, Li X, Cao D, Yue J. Multimodal magnetic resonance imaging on brain structure and function changes in vascular cognitive impairment without dementia. Front Aging Neurosci 2023; 15:1278390. [PMID: 38035274 PMCID: PMC10687453 DOI: 10.3389/fnagi.2023.1278390] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/23/2023] [Indexed: 12/02/2023] Open
Abstract
Vascular cognitive impairment not dementia (VCIND) is one of the three subtypes of vascular cognitive impairment (VCI), with cognitive dysfunction and symptoms ranging between normal cognitive function and vascular dementia. The specific mechanisms underlying VCIND are still not fully understood, and there is a lack of specific diagnostic markers in clinical practice. With the rapid development of magnetic resonance imaging (MRI) technology, structural MRI (sMRI) and functional MRI (fMRI) have become effective methods for exploring the neurobiological mechanisms of VCIND and have made continuous progress. This article provides a comprehensive overview of the research progress in VCIND using multimodal MRI, including sMRI, diffusion tensor imaging, resting-state fMRI, and magnetic resonance spectroscopy. By integrating findings from these multiple modalities, this study presents a novel perspective on the neuropathological mechanisms underlying VCIND. It not only highlights the importance of multimodal MRI in unraveling the complex nature of VCIND but also lays the foundation for future research examining the relationship between brain structure, function, and cognitive impairment in VCIND. These new perspectives and strategies ultimately hold the potential to contribute to the development of more effective diagnostic tools and therapeutic interventions for VCIND.
Collapse
Affiliation(s)
- Qinhong Zhang
- Shenzhen Frontiers in Chinese Medicine Research Co., Ltd., Shenzhen, China
- Department of Acupuncture and Moxibustion, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiao Liu
- Department of Pediatrics, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shenglan Gao
- Graduate School of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shiyan Yan
- School of Acupuncture-Moxibustion and Tuina, Beijing University of Chinese Medicine, Beijing, China
| | - Ang Li
- Servier (Beijing) Pharmaceutical Research and Development Co., Ltd., Beijing, China
| | - Zeyi Wei
- Graduate School of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Shengwang Han
- Third Ward of Rehabilitation Department, Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yu Hou
- Department of Gynecology, Harbin Traditional Chinese Medicine Hospital, Harbin, China
| | - Xiaoling Li
- Division of CT and MRI, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Danna Cao
- Division of CT and MRI, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jinhuan Yue
- Shenzhen Frontiers in Chinese Medicine Research Co., Ltd., Shenzhen, China
- Department of Acupuncture and Moxibustion, Vitality University, Hayward, CA, United States
| |
Collapse
|
230
|
Mauricio D, Gratacòs M, Franch-Nadal J. Diabetic microvascular disease in non-classical beds: the hidden impact beyond the retina, the kidney, and the peripheral nerves. Cardiovasc Diabetol 2023; 22:314. [PMID: 37968679 PMCID: PMC10652502 DOI: 10.1186/s12933-023-02056-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 11/07/2023] [Indexed: 11/17/2023] Open
Abstract
Diabetes microangiopathy, a hallmark complication of diabetes, is characterised by structural and functional abnormalities within the intricate network of microvessels beyond well-known and documented target organs, i.e., the retina, kidney, and peripheral nerves. Indeed, an intact microvascular bed is crucial for preserving each organ's specific functions and achieving physiological balance to meet their respective metabolic demands. Therefore, diabetes-related microvascular dysfunction leads to widespread multiorgan consequences in still-overlooked non-traditional target organs such as the brain, the lung, the bone tissue, the skin, the arterial wall, the heart, or the musculoskeletal system. All these organs are vulnerable to the physiopathological mechanisms that cause microvascular damage in diabetes (i.e., hyperglycaemia-induced oxidative stress, inflammation, and endothelial dysfunction) and collectively contribute to abnormalities in the microvessels' structure and function, compromising blood flow and tissue perfusion. However, the microcirculatory networks differ between organs due to variations in haemodynamic, vascular architecture, and affected cells, resulting in a spectrum of clinical presentations. The aim of this review is to focus on the multifaceted nature of microvascular impairment in diabetes through available evidence of specific consequences in often overlooked organs. A better understanding of diabetes microangiopathy in non-target organs provides a broader perspective on the systemic nature of the disease, underscoring the importance of recognising the comprehensive range of complications beyond the classic target sites.
Collapse
Affiliation(s)
- Dídac Mauricio
- DAP-Cat group, Unitat de Suport a la Recerca Barcelona, Fundació Institut Universitari per a la recerca a l'Atenció Primària de Salut Jordi Gol i Gurina (IDIAPJGol), Barcelona, Spain.
- CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Barcelona, Spain.
- Department of Endocrinology and Nutrition, Hospital de la Santa Creu i Sant Pau, IR Sant Pau, Barcelona, Spain.
- Department of Medicine, University of Vic - Central University of Catalonia, Vic, Spain.
| | - Mònica Gratacòs
- DAP-Cat group, Unitat de Suport a la Recerca Barcelona, Fundació Institut Universitari per a la recerca a l'Atenció Primària de Salut Jordi Gol i Gurina (IDIAPJGol), Barcelona, Spain
| | - Josep Franch-Nadal
- DAP-Cat group, Unitat de Suport a la Recerca Barcelona, Fundació Institut Universitari per a la recerca a l'Atenció Primària de Salut Jordi Gol i Gurina (IDIAPJGol), Barcelona, Spain
- CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Instituto de Salud Carlos III (ISCIII), Barcelona, Spain
| |
Collapse
|
231
|
Koblinsky ND, Carmichael PH, Belleville S, Fiocco AJ, Gaudreau P, Greenwood CE, Kergoat MJ, Morais JA, Presse N, Laurin D, Ferland G. Associations between circulating cardiovascular disease risk factors and cognitive performance in cognitively healthy older adults from the NuAge study. Front Aging Neurosci 2023; 15:1274794. [PMID: 38020779 PMCID: PMC10668121 DOI: 10.3389/fnagi.2023.1274794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 09/26/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Cardiovascular disease risk factors (CVRFs) contribute to the development of cognitive impairment and dementia. Methods This study examined the associations between circulating CVRF biomarkers and cognition in 386 cognitively healthy older adults (mean age = 78 ± 4 years, 53% females) selected from the Quebec Longitudinal Study on Nutrition and Successful Aging (NuAge). Memory, executive function, and processing speed were assessed at baseline and 2-year follow-up. CVRF biomarkers included total cholesterol, high-density lipoprotein cholesterol (HDL-C), low-density lipoprotein-cholesterol (LDL-C), triglycerides, glucose, insulin, high sensitivity C-reactive protein (hs-CRP), homocysteine, protein carbonyls, and cortisol. Linear mixed models were used to determine associations between individual CVRF biomarkers and cognition at both time points. Results HDL-C was most consistently associated with cognition with higher values related to better performance across several domains. Overall, stronger and more consistent relationships between CVRF biomarkers and cognition were observed in females relative to males. Discussion Findings suggest that increases in the majority of circulating CVRFs are not associated with worse cognition in cognitively healthy older adults.
Collapse
Affiliation(s)
- Noah D. Koblinsky
- Rotman Research Institute, Baycrest Health Sciences, Toronto, ON, Canada
| | - Pierre-Hugues Carmichael
- Center d’excellence sur le Vieillissement de Québec, Center de Recherche du Center Hospitalier Universitaire (CHU) de Québec-Université Laval and VITAM-Center de Recherche en Santé Durable, Center Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de la Capitale Nationale, Québec, QC, Canada
| | - Sylvie Belleville
- Center de Recherche de l’Institut Universitaire de Gériatrie de Montréal, CIUSSS du Center-Sud-de-l’Île-de-Montréal, Montreal, QC, Canada
| | - Alexandra J. Fiocco
- Department of Psychology, Toronto Metropolitan University, Toronto, ON, Canada
| | - Pierrette Gaudreau
- Center de Recherche du Center Hospitalier de l’Université de Montréal and Faculté de Médecine, Université de Montréal, Montreal, QC, Canada
| | - Carol E. Greenwood
- Rotman Research Institute, Baycrest Health Sciences, Toronto, ON, Canada
- Department of Nutritional Sciences, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | | | - José A. Morais
- Division of Geriatrics, McGill University, Montreal, QC, Canada
| | - Nancy Presse
- Center de Recherche du Center Hospitalier de l’Université de Montréal and Faculté de Médecine, Université de Montréal, Montreal, QC, Canada
- Faculté de Médecine et des Sciences de la Santé de l’Université de Sherbrooke, Sherbrooke, QC, Canada
- Center de Recherche sur le Vieillissement du CIUSSS de l’Estrie-Center Hospitalier Universitaire de Sherbrooke (CHUS), Sherbrooke, QC, Canada
| | - Danielle Laurin
- Center d’excellence sur le Vieillissement de Québec, Center de Recherche du Center Hospitalier Universitaire (CHU) de Québec-Université Laval and VITAM-Center de Recherche en Santé Durable, Center Intégré Universitaire de Santé et de Services Sociaux (CIUSSS) de la Capitale Nationale, Québec, QC, Canada
- Faculté de Pharmacie, Institut sur le Vieillissement et la Participation Sociale des Aînés and Institut sur la Nutrition et les Aliments Fonctionnels (INAF), Université Laval, Montreal, ON, Canada
| | - Guylaine Ferland
- Montreal Heart Institute Research Center, and Département de Nutrition, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
232
|
Qin R, Li T, Li C, Li L, Wang X, Wang L. Investigating altered brain functional hubs and causal connectivity in coronary artery disease with cognitive impairment. PeerJ 2023; 11:e16408. [PMID: 38025718 PMCID: PMC10640849 DOI: 10.7717/peerj.16408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 10/15/2023] [Indexed: 12/01/2023] Open
Abstract
Background Coronary artery disease (CAD) and cognitive impairment (CI) have become significant global disease and medical burdens. There have been several reports documenting the alterations in regional brain function and their correlation with CI in CAD patients. However, there is limited research on the changes in brain network connectivity in CAD patients. To investigate the resting-state connectivity and further understand the effective connectivity strength and directionality in patients with CAD, we utilized degree centrality (DC) and spectral dynamic causal modeling (spDCM) to detect functional hubs in the whole brain network, followed by an analysis of directional connections. Using the aforementioned approaches, it is possible to investigate the hub regions and aberrant connections underlying the altered brain function in CAD patients, providing neuroimaging evidence for the cognitive decline in patients with coronary artery disease. Materials and Methods This study was prospectively conducted involving 24 patients diagnosed with CAD and 24 healthy controls (HC) who were matched in terms of age, gender, and education. Functional MRI (fMRI) scans were utilized to investigate brain activity in these individuals. Neuropsychological examinations were performed on all participants. DC analysis and spDCM were employed to investigate abnormal brain networks in patients with CAD. Additionally, the association between effective connectivity strength and cognitive function in patients with CAD was examined based on the aforementioned results. Results By assessing cognitive functions, we discovered that patients with CAD exhibited notably lower cognitive function compared to the HC group. By utilizing DC analysis and spDCM, we observed significant reductions in DC values within the left parahippocampal cortex (PHC) and the left medial temporal gyrus (MTG) in CAD patients when compared to the control group. In terms of effective connectivity, we observed the absence of positive connectivity between the right superior frontal gyrus (SFG) and PHC in CAD patients. Moreover, there was an increase in negative connectivity from PHC and MTG to SFG, along with a decrease in the strength of positive connectivity between PHC and MTG. Furthermore, we identified a noteworthy positive correlation (r = 0.491, p = 0.015) between the strength of connectivity between the PHC and the MTG and cognitive function in CAD patients. Conclusions These research findings suggest that alterations in the connectivity of the brain networks involving SFG, PHC, and MTG in CAD patients may mediate changes in cognitive function.
Collapse
Affiliation(s)
- Rui Qin
- Department of Radiology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong Province, China
| | - Tong Li
- Department of Radiology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong Province, China
| | - Cuicui Li
- Department of Radiology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong Province, China
| | - Lin Li
- Department of Radiology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong Province, China
| | - Ximing Wang
- Department of Radiology, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong Province, China
| | - Li Wang
- Department of Health Management Center, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong Province, China
| |
Collapse
|
233
|
Vuic B, Milos T, Tudor L, Nikolac Perkovic M, Konjevod M, Nedic Erjavec G, Farkas V, Uzun S, Mimica N, Svob Strac D. Pharmacogenomics of Dementia: Personalizing the Treatment of Cognitive and Neuropsychiatric Symptoms. Genes (Basel) 2023; 14:2048. [PMID: 38002991 PMCID: PMC10671071 DOI: 10.3390/genes14112048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
Dementia is a syndrome of global and progressive deterioration of cognitive skills, especially memory, learning, abstract thinking, and orientation, usually affecting the elderly. The most common forms are Alzheimer's disease, vascular dementia, and other (frontotemporal, Lewy body disease) dementias. The etiology of these multifactorial disorders involves complex interactions of various environmental and (epi)genetic factors and requires multiple forms of pharmacological intervention, including anti-dementia drugs for cognitive impairment, antidepressants, antipsychotics, anxiolytics and sedatives for behavioral and psychological symptoms of dementia, and other drugs for comorbid disorders. The pharmacotherapy of dementia patients has been characterized by a significant interindividual variability in drug response and the development of adverse drug effects. The therapeutic response to currently available drugs is partially effective in only some individuals, with side effects, drug interactions, intolerance, and non-compliance occurring in the majority of dementia patients. Therefore, understanding the genetic basis of a patient's response to pharmacotherapy might help clinicians select the most effective treatment for dementia while minimizing the likelihood of adverse reactions and drug interactions. Recent advances in pharmacogenomics may contribute to the individualization and optimization of dementia pharmacotherapy by increasing its efficacy and safety via a prediction of clinical outcomes. Thus, it can significantly improve the quality of life in dementia patients.
Collapse
Affiliation(s)
- Barbara Vuic
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| | - Tina Milos
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| | - Lucija Tudor
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| | - Matea Nikolac Perkovic
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| | - Marcela Konjevod
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| | - Gordana Nedic Erjavec
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| | - Vladimir Farkas
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| | - Suzana Uzun
- Department for Biological Psychiatry and Psychogeriatry, University Hospital Vrapce, 10000 Zagreb, Croatia; (S.U.); (N.M.)
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Ninoslav Mimica
- Department for Biological Psychiatry and Psychogeriatry, University Hospital Vrapce, 10000 Zagreb, Croatia; (S.U.); (N.M.)
- School of Medicine, University of Zagreb, 10000 Zagreb, Croatia
| | - Dubravka Svob Strac
- Laboratory for Molecular Neuropsychiatry, Division of Molecular Medicine, Rudjer Boskovic Institute, 10000 Zagreb, Croatia; (B.V.); (T.M.); (L.T.); (M.N.P.); (M.K.); (G.N.E.); (V.F.)
| |
Collapse
|
234
|
Lee M, Lim JS, Kim Y, Park SH, Lee SH, Kim C, Lee BC, Yu KH, Lee JJ, Oh MS. High ApoB/ApoA-I Ratio Predicts Post-Stroke Cognitive Impairment in Acute Ischemic Stroke Patients with Large Artery Atherosclerosis. Nutrients 2023; 15:4670. [PMID: 37960323 PMCID: PMC10648714 DOI: 10.3390/nu15214670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 10/30/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023] Open
Abstract
BACKGROUND We aimed to investigate the association between the ApoB/ApoA-I ratio and post-stroke cognitive impairment (PSCI) in patients with acute stroke of large artery atherosclerosis etiology. METHODS Prospective stroke registry data were used to consecutively enroll patients with acute ischemic stroke due to large artery atherosclerosis. Cognitive function assessments were conducted 3 to 6 months after stroke. PSCI was defined as a z-score of less than -2 standard deviations from age, sex, and education-adjusted means in at least one cognitive domain. The ApoB/ApoA-I ratio was calculated, and patients were categorized into five groups according to quintiles of the ratio. Logistic regression analyses were performed to assess the association between quintiles of the ApoB/ApoA-I ratio and PSCI. RESULTS A total of 263 patients were included, with a mean age of 65.9 ± 11.6 years. The median NIHSS score and ApoB/ApoA-I ratio upon admission were 2 (IQR, 1-5) and 0.81 (IQR, 0.76-0.88), respectively. PSCI was observed in 91 (34.6%) patients. The highest quintile (Q5) of the ApoB/ApoA-I ratio was a significant predictor of PSCI compared to the lowest quintile (Q1) (adjusted OR, 3.16; 95% CI, 1.19-8.41; p-value = 0.021) after adjusting for relevant confounders. Patients in the Q5 group exhibited significantly worse performance in the frontal domain. CONCLUSIONS The ApoB/ApoA-I ratio in the acute stage of stroke independently predicted the development of PSCI at 3-6 months after stroke due to large artery atherosclerosis. Further, a high ApoB/ApoA-I ratio was specifically associated with frontal domain dysfunction.
Collapse
Affiliation(s)
- Minwoo Lee
- Department of Neurology, Hallym University Sacred Heart Hospital, Hallym Neurological Institute, Hallym University College of Medicine, Anyang 14068, Republic of Korea; (M.L.); (B.-C.L.); (K.-H.Y.)
- Institute of New Frontier Research Team, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea;
| | - Jae-Sung Lim
- Department of Neurology, Asan Medical Center, Ulsan University College of Medicine, Seoul 05505, Republic of Korea;
| | - Yerim Kim
- Department of Neurology, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul 24252, Republic of Korea; (Y.K.); (S.H.P.)
| | - Soo Hyun Park
- Department of Neurology, Kangdong Sacred Heart Hospital, Hallym University College of Medicine, Seoul 24252, Republic of Korea; (Y.K.); (S.H.P.)
| | - Sang-Hwa Lee
- Department of Neurology, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea; (S.-H.L.); (C.K.)
| | - Chulho Kim
- Department of Neurology, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea; (S.-H.L.); (C.K.)
| | - Byung-Chul Lee
- Department of Neurology, Hallym University Sacred Heart Hospital, Hallym Neurological Institute, Hallym University College of Medicine, Anyang 14068, Republic of Korea; (M.L.); (B.-C.L.); (K.-H.Y.)
| | - Kyung-Ho Yu
- Department of Neurology, Hallym University Sacred Heart Hospital, Hallym Neurological Institute, Hallym University College of Medicine, Anyang 14068, Republic of Korea; (M.L.); (B.-C.L.); (K.-H.Y.)
| | - Jae-Jun Lee
- Institute of New Frontier Research Team, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea;
| | - Mi Sun Oh
- Department of Neurology, Hallym University Sacred Heart Hospital, Hallym Neurological Institute, Hallym University College of Medicine, Anyang 14068, Republic of Korea; (M.L.); (B.-C.L.); (K.-H.Y.)
| |
Collapse
|
235
|
Bhat A, Goel D. Coronary Heart Disease, Heart Failure, and Risk of Alzheimer's Disease: How Strong is the Association? Ann Indian Acad Neurol 2023; 26:852-853. [PMID: 38229647 PMCID: PMC10789407 DOI: 10.4103/aian.aian_649_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 07/24/2023] [Indexed: 01/18/2024] Open
Affiliation(s)
- Ashwani Bhat
- Department of Neurology, Himalayan Institute of Medical Sciences, Swami Ram Himalayan University, Dehradun, Uttarakhand, India
| | - Deepak Goel
- Department of Neurology, Himalayan Institute of Medical Sciences, Swami Ram Himalayan University, Dehradun, Uttarakhand, India
| |
Collapse
|
236
|
Ishikawa H, Shindo A, Mizutani A, Tomimoto H, Lo EH, Arai K. A brief overview of a mouse model of cerebral hypoperfusion by bilateral carotid artery stenosis. J Cereb Blood Flow Metab 2023; 43:18-36. [PMID: 36883344 PMCID: PMC10638994 DOI: 10.1177/0271678x231154597] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 12/23/2022] [Accepted: 01/04/2023] [Indexed: 03/09/2023]
Abstract
Vascular cognitive impairment (VCI) refers to all forms of cognitive disorder related to cerebrovascular diseases, including vascular mild cognitive impairment, post-stroke dementia, multi-infarct dementia, subcortical ischemic vascular dementia (SIVD), and mixed dementia. Among the causes of VCI, more attention has been paid to SIVD because the causative cerebral small vessel pathologies are frequently observed in elderly people and because the gradual progression of cognitive decline often mimics Alzheimer's disease. In most cases, small vessel diseases are accompanied by cerebral hypoperfusion. In mice, prolonged cerebral hypoperfusion is induced by bilateral carotid artery stenosis (BCAS) with surgically implanted metal micro-coils. This cerebral hypoperfusion BCAS model was proposed as a SIVD mouse model in 2004, and the spreading use of this mouse SIVD model has provided novel data regarding cognitive dysfunction and histological/genetic changes by cerebral hypoperfusion. Oxidative stress, microvascular injury, excitotoxicity, blood-brain barrier dysfunction, and secondary inflammation may be the main mechanisms of brain damage due to prolonged cerebral hypoperfusion, and some potential therapeutic targets for SIVD have been proposed by using transgenic mice or clinically used drugs in BCAS studies. This review article overviews findings from the studies that used this hypoperfused-SIVD mouse model, which were published between 2004 and 2021.
Collapse
Affiliation(s)
- Hidehiro Ishikawa
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Akihiro Shindo
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Akane Mizutani
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Hidekazu Tomimoto
- Department of Neurology, Mie University Graduate School of Medicine, Tsu, Japan
| | - Eng H Lo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Ken Arai
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
237
|
Kapila Sharma A, Mohan L, Mittal S, Bahadur A, Mirza AA, Thapiyal M. Assessment of Visual Evoked Potential to Detect Visual Pathway Dysfunction in Gestational Diabetes Mellitus: A Longitudinal Case-Control Study With Postpartum Follow-up. Cureus 2023; 15:e49619. [PMID: 38161906 PMCID: PMC10755644 DOI: 10.7759/cureus.49619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND The increasing prevalence of gestational diabetes mellitus (GDM) during pregnancy has opened the opportunity to study its short- and long-term effects on maternal ophthalmic health. Visual evoked potential (VEP) is a non-invasive electrophysiological test that detects functional disturbances along the visual pathway before the physical signs of diabetic retinopathy (DR) can set in. This longitudinal study is aimed at the assessment of changes in VEP in GDM during different stages of pregnancy and 6-12 weeks after parturition by comparing it with normoglycemic controls. DESIGN AND METHOD Diagnosed cases of GDM were recruited along with normoglycemic controls at 24-28 weeks of gestation. Each participant was required to attend two follow-up appointments at 32-38 weeks of gestation and 6-12 weeks after parturition. A blood sample was taken in a fasting state to record biochemical parameters. VEP was recorded using Neuropack S1 MEB-9400 electrodiagnostic equipment (Nihon Kohden, Tokyo, Japan) in a dark room by providing pattern reversal stimuli to each eye. RESULTS A total of 29 participants (15 in the control group and 14 in the GDM group) completed the entire study procedure. The observed mean P100 latency of both eyes in the GDM group was recorded longer as compared to that in the control group at baseline and during late pregnancy. Although the mean P100 latency saw a significant decline in postpartum visits as compared to that in late pregnancy, the values were higher than in the control group. P100 latency at baseline correlated significantly to serum advanced glycated end products' (AGE'S) levels in the GDM group. CONCLUSION Our study findings reflect that the diagnosis of GDM is associated with significant changes in VEP during and after pregnancy as compared to that of healthy pregnant women.
Collapse
Affiliation(s)
| | - Latika Mohan
- Physiology, All India Institute of Medical Sciences, Rishikesh, Rishikesh, IND
| | - Sunita Mittal
- Physiology, All India Institute of Medical Sciences, Rishikesh, Rishikesh, IND
| | - Anupama Bahadur
- Obstetrics and Gynecology, All India Institute of Medical Sciences, Rishikesh, Rishikesh, IND
| | - Anissa A Mirza
- Biochemistry, All India Institute of Medical Sciences, Rishikesh, Rishikesh, IND
| | - Manish Thapiyal
- Physiology, All India Institute of Medical Sciences, Rishikesh, Rishikesh, IND
| |
Collapse
|
238
|
Sleight E, Stringer MS, Clancy U, Arteaga C, Jaime Garcia D, Hewins W, Jochems AC, Hamilton OK, Manning C, Morgan AG, Locherty R, Cheng Y, Liu X, Zhang J, Hamilton I, Jardine C, Brown R, Sakka E, Kampaite A, Wiseman S, Valdés-Hernández MC, Chappell FM, Doubal FN, Marshall I, Thrippleton MJ, Wardlaw JM. Cerebrovascular Reactivity in Patients With Small Vessel Disease: A Cross-Sectional Study. Stroke 2023; 54:2776-2784. [PMID: 37814956 PMCID: PMC10589433 DOI: 10.1161/strokeaha.123.042656] [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: 02/23/2023] [Revised: 09/06/2023] [Accepted: 09/19/2023] [Indexed: 10/11/2023]
Abstract
BACKGROUND Cerebrovascular reactivity (CVR) is inversely related to white matter hyperintensity severity, a marker of cerebral small vessel disease (SVD). Less is known about the relationship between CVR and other SVD imaging features or cognition. We aimed to investigate these cross-sectional relationships. METHODS Between 2018 and 2021 in Edinburgh, we recruited patients presenting with lacunar or cortical ischemic stroke, whom we characterized for SVD features. We measured CVR in subcortical gray matter, normal-appearing white matter, and white matter hyperintensity using 3T magnetic resonance imaging. We assessed cognition using Montreal Cognitive Assessment. Statistical analyses included linear regression models with CVR as outcome, adjusted for age, sex, and vascular risk factors. We reported regression coefficients with 95% CIs. RESULTS Of 208 patients, 182 had processable CVR data sets (median age, 68.2 years; 68% men). Although the strength of association depended on tissue type, lower CVR in normal-appearing tissues and white matter hyperintensity was associated with larger white matter hyperintensity volume (BNAWM=-0.0073 [95% CI, -0.0133 to -0.0014] %/mm Hg per 10-fold increase in percentage intracranial volume), more lacunes (BNAWM=-0.00129 [95% CI, -0.00215 to -0.00043] %/mm Hg per lacune), more microbleeds (BNAWM=-0.00083 [95% CI, -0.00130 to -0.00036] %/mm Hg per microbleed), higher deep atrophy score (BNAWM=-0.00218 [95% CI, -0.00417 to -0.00020] %/mm Hg per score point increase), higher perivascular space score (BNAWM=-0.0034 [95% CI, -0.0066 to -0.0002] %/mm Hg per score point increase in basal ganglia), and higher SVD score (BNAWM=-0.0048 [95% CI, -0.0075 to -0.0021] %/mm Hg per score point increase). Lower CVR in normal-appearing tissues was related to lower Montreal Cognitive Assessment without reaching convention statistical significance (BNAWM=0.00065 [95% CI, -0.00007 to 0.00137] %/mm Hg per score point increase). CONCLUSIONS Lower CVR in patients with SVD was related to more severe SVD burden and worse cognition in this cross-sectional analysis. Longitudinal analysis will help determine whether lower CVR predicts worsening SVD severity or vice versa. REGISTRATION URL: https://www.isrctn.com; Unique identifier: ISRCTN12113543.
Collapse
Affiliation(s)
- Emilie Sleight
- Centre for Clinical Brain Sciences (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
- UK Dementia Research Institute (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
| | - Michael S. Stringer
- Centre for Clinical Brain Sciences (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
- UK Dementia Research Institute (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
| | - Una Clancy
- Centre for Clinical Brain Sciences (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
- UK Dementia Research Institute (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
| | - Carmen Arteaga
- Centre for Clinical Brain Sciences (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
- UK Dementia Research Institute (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
| | - Daniela Jaime Garcia
- Centre for Clinical Brain Sciences (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
- UK Dementia Research Institute (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
| | - Will Hewins
- Centre for Clinical Brain Sciences (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
- UK Dementia Research Institute (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
| | - Angela C.C. Jochems
- Centre for Clinical Brain Sciences (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
- UK Dementia Research Institute (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
| | - Olivia K.L. Hamilton
- Centre for Clinical Brain Sciences (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
- UK Dementia Research Institute (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
| | - Cameron Manning
- Centre for Clinical Brain Sciences (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
- UK Dementia Research Institute (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
| | - Alasdair G. Morgan
- Centre for Clinical Brain Sciences (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
- UK Dementia Research Institute (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
| | - Rachel Locherty
- Centre for Clinical Brain Sciences (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
- UK Dementia Research Institute (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
| | - Yajun Cheng
- Centre for Clinical Brain Sciences (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
- UK Dementia Research Institute (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
- Department of Neurology, West China Hospital of Sichuan University, Chengdu (Y.C.)
| | - Xiaodi Liu
- Centre for Clinical Brain Sciences (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
- UK Dementia Research Institute (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
- Department of Medicine, University of Hong Kong (X.L.)
| | - Junfang Zhang
- Centre for Clinical Brain Sciences (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
- UK Dementia Research Institute (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, China (J.Z.)
| | - Iona Hamilton
- Edinburgh Imaging Facility RIE (I.H., C.J., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
| | - Charlotte Jardine
- Edinburgh Imaging Facility RIE (I.H., C.J., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
| | - Rosalind Brown
- Centre for Clinical Brain Sciences (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
- UK Dementia Research Institute (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
| | - Eleni Sakka
- Centre for Clinical Brain Sciences (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
- UK Dementia Research Institute (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
| | - Agniete Kampaite
- Centre for Clinical Brain Sciences (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
- UK Dementia Research Institute (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
| | - Stewart Wiseman
- Centre for Clinical Brain Sciences (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
- UK Dementia Research Institute (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
| | - Maria C. Valdés-Hernández
- Centre for Clinical Brain Sciences (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
- UK Dementia Research Institute (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
| | - Francesca M. Chappell
- Centre for Clinical Brain Sciences (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
- UK Dementia Research Institute (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
| | - Fergus N. Doubal
- Centre for Clinical Brain Sciences (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
- UK Dementia Research Institute (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
| | - Ian Marshall
- Centre for Clinical Brain Sciences (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
- UK Dementia Research Institute (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
| | - Michael J. Thrippleton
- Centre for Clinical Brain Sciences (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
- UK Dementia Research Institute (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
- Edinburgh Imaging Facility RIE (I.H., C.J., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
| | - Joanna M. Wardlaw
- Centre for Clinical Brain Sciences (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
- UK Dementia Research Institute (E. Sleight, M.S.S., U.C., C.A., D.J.G., W.H., A.C.C.J., O.K.L.H., C.M., A.G.M., R.L., Y.C., X.L., J.Z., R.B., E. Sakka, A.K., S.W., M.C.V.-H., F.M.C., F.N.D., I.M., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
- Edinburgh Imaging Facility RIE (I.H., C.J., M.J.T., J.M.W.), University of Edinburgh, United Kingdom
| |
Collapse
|
239
|
Katz BR, Khadanga S, Middleton WA, Mahoney K, Savage PD, DeSarno M, Ades PA, Gaalema DE. Self-Reported Executive Function in Hospitalized Cardiac Patients and Associations With Patient Characteristics and Cardiac Rehabilitation Attendance. J Cardiopulm Rehabil Prev 2023; 43:433-437. [PMID: 36857090 PMCID: PMC10474250 DOI: 10.1097/hcr.0000000000000785] [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] [Indexed: 03/02/2023]
Abstract
PURPOSE Executive function (ExF), the ability to do complex cognitive tasks like planning and refraining from impulsive behavior, is associated with compliance with medical recommendations. The present study identified associations between self-reported ExF and demographics of patients with cardiac disease as well as with cardiac rehabilitation (CR) attendance. METHODS Self-reported ExF impairment was measured using the Behavior Rating Inventory of Executive Function (BRIEF) on 316 individuals hospitalized for CR-qualifying cardiac events. Scores were calculated for a global measure (Global Executive Composite [GEC]) and the two BRIEF indices: Behavioral Regulation Index and Metacognition Index (MCI). Participants were followed up post-discharge to determine CR attendance. Univariate logistic regressions between ExF measures and demographic variables were conducted, as were multiple logistic regressions to identify significant, independent predictors. Analyses were conducted using clinical (T scores ≥ 65) and subclinical (T scores ≥ 60) criteria for significant ExF impairment as outcomes. One-way analyses of variance were performed between ExF impairment and CR attendance. RESULTS Self-reported ExF deficits were relatively rare; 8.9% had at least subclinical scores on the GEC. Using the subclinical criterion for the MCI, having diabetes mellitus (DM) and being male were significant, independent predictors of MCI impairment. No significant relationship was found between ExF and CR attendance. CONCLUSION Using the subclinical criterion only, individuals with DM and males were significantly more likely to have MCI impairment. No significant effect of ExF impairment on CR attendance was found, suggesting that self-reported ExF measured in the hospital may not be an appropriate measure for predicting behavioral outcomes.
Collapse
Affiliation(s)
- Brian R. Katz
- Vermont Center on Behavior and Health, Burlington VT
- University of Vermont Departments of Psychiatry and Psychology, Burlington VT
| | - Sherrie Khadanga
- Vermont Center on Behavior and Health, Burlington VT
- University of Vermont Medical Center Division of Cardiology, Burlington VT
| | - William A. Middleton
- Vermont Center on Behavior and Health, Burlington VT
- University of Vermont Departments of Psychiatry and Psychology, Burlington VT
| | - Katharine Mahoney
- Vermont Center on Behavior and Health, Burlington VT
- University of Vermont Departments of Psychiatry and Psychology, Burlington VT
- MedStar Health Research Institute, Hyattsville, MD
| | - Patrick D. Savage
- Vermont Center on Behavior and Health, Burlington VT
- University of Vermont Medical Center Division of Cardiology, Burlington VT
| | - Michael DeSarno
- Vermont Center on Behavior and Health, Burlington VT
- University of Vermont Department of Medical Biostatistics, Burlington VT
| | - Philip A. Ades
- Vermont Center on Behavior and Health, Burlington VT
- University of Vermont Medical Center Division of Cardiology, Burlington VT
| | - Diann E. Gaalema
- Vermont Center on Behavior and Health, Burlington VT
- University of Vermont Departments of Psychiatry and Psychology, Burlington VT
| |
Collapse
|
240
|
Canepa E, Parodi-Rullan R, Vazquez-Torres R, Gamallo-Lana B, Guzman-Hernandez R, Lemon NL, Angiulli F, Debure L, Ilies MA, Østergaard L, Wisniewski T, Gutiérrez-Jiménez E, Mar AC, Fossati S. FDA-approved carbonic anhydrase inhibitors reduce amyloid β pathology and improve cognition, by ameliorating cerebrovascular health and glial fitness. Alzheimers Dement 2023; 19:5048-5073. [PMID: 37186121 PMCID: PMC10600328 DOI: 10.1002/alz.13063] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/09/2023] [Accepted: 03/09/2023] [Indexed: 05/17/2023]
Abstract
INTRODUCTION Cerebrovascular pathology is an early and causal hallmark of Alzheimer's disease (AD), in need of effective therapies. METHODS Based on the success of our previous in vitro studies, we tested for the first time in a model of AD and cerebral amyloid angiopathy (CAA), the carbonic anhydrase inhibitors (CAIs) methazolamide and acetazolamide, Food and Drug Administration-approved against glaucoma and high-altitude sickness. RESULTS Both CAIs reduced cerebral, vascular, and glial amyloid beta (Aβ) accumulation and caspase activation, diminished gliosis, and ameliorated cognition in TgSwDI mice. The CAIs also improved microvascular fitness and induced protective glial pro-clearance pathways, resulting in the reduction of Aβ deposition. Notably, we unveiled that the mitochondrial carbonic anhydrase-VB (CA-VB) is upregulated in TgSwDI brains, CAA and AD+CAA human subjects, and in endothelial cells upon Aβ treatment. Strikingly, CA-VB silencing specifically reduces Aβ-mediated endothelial apoptosis. DISCUSSION This work substantiates the potential application of CAIs in clinical trials for AD and CAA.
Collapse
Affiliation(s)
- Elisa Canepa
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Rebecca Parodi-Rullan
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Rafael Vazquez-Torres
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Begona Gamallo-Lana
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Roberto Guzman-Hernandez
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Nicole L. Lemon
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Federica Angiulli
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Ludovic Debure
- Department on Neurology, Center for Cognitive Neurology, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Marc A. Ilies
- Department of Pharmaceutical Sciences and Moulder Center for Drug Discovery Research, Temple University School of Pharmacy, Temple University, Philadelphia, PA, 19140, USA
| | - Leif Østergaard
- Center of Functionally Integrative Neuroscience (CFIN), Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Thomas Wisniewski
- Department on Neurology, Center for Cognitive Neurology, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Eugenio Gutiérrez-Jiménez
- Center of Functionally Integrative Neuroscience (CFIN), Department of Clinical Medicine, Aarhus University, 8200 Aarhus, Denmark
| | - Adam C. Mar
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Grossman School of Medicine, New York, NY, 10016, USA
| | - Silvia Fossati
- Alzheimer’s Center at Temple, Department of Neural Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| |
Collapse
|
241
|
Lefferts WK, Reed KS, Rosonke RE, Augustine JA, Moreau KL. Age-associated increases in middle cerebral artery pulsatility differ between men and women. Am J Physiol Heart Circ Physiol 2023; 325:H1118-H1125. [PMID: 37682233 PMCID: PMC10908402 DOI: 10.1152/ajpheart.00453.2023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 08/21/2023] [Accepted: 09/05/2023] [Indexed: 09/09/2023]
Abstract
Mechanisms underlying sex differences in brain aging remain unclear but may relate to changes in cerebral pulsatile blood flow. Sex differences in the stiffening of the large arteries and expansion of pulse pressure with age may accelerate changes in pulsatile (i.e., discontinuous) blood flow in the brain that contribute to brain health. The purpose of this cross-sectional, secondary analysis was to examine sex differences in age-associated changes in large artery (aorta and carotid) stiffness, carotid pulse pressure, and cerebral pulsatility in 206 men and 217 women between 18 and 72 yr of age. Outcomes included aortic stiffness [carotid-femoral pulse wave velocity (cfPWV)] and carotid pulse pressure via tonometry, carotid β-stiffness via ultrasound, and middle cerebral artery (MCA) pulsatility index via transcranial Doppler. Regression analyses revealed a significant age-by-sex interaction, with women exhibiting a slower rate of change compared with men for cfPWV (β = -0.21, P = 0.04), and greater rate of change for carotid stiffness (β = 0.27, P = 0.02), carotid pulse pressure (β = 0.98, P < 0.001), and MCA pulsatility index (β = 0.49, P = 0.002) after adjustment for covariates. The significant age-by-sex interaction for MCA pulsatility was abolished after further adjustment for carotid pulse pressure. Women exhibit accelerated increases in cerebral pulsatility during midlife, likely driven by exaggerated increases in carotid stiffness and pulse pressure compared with men. These data suggest that there are disproportionate increases in cerebral pulsatility in women during midlife that could contribute to accelerated brain aging compared with men.NEW & NOTEWORTHY We identify sex-specific associations between increasing age and cerebral pulsatility and its vascular mechanisms. When compared with men, women in our cross-sectional analysis exhibited greater age-associated increases in carotid stiffness, carotid pulse pressure, and cerebral pulsatility particularly during midlife. These data suggest that the rapid expansion of pulse pressure during midlife contributes to an exaggerated increase in cerebral pulsatility among women and suggest a potential mechanism contributing to sex differences in brain aging.
Collapse
Affiliation(s)
- Wesley K Lefferts
- Department of Kinesiology, Iowa State University, Ames, Iowa, United States
| | - Krista S Reed
- Department of Kinesiology, Iowa State University, Ames, Iowa, United States
| | - Rachel E Rosonke
- Department of Kinesiology, Iowa State University, Ames, Iowa, United States
| | | | - Kerrie L Moreau
- Division of Geriatric Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| |
Collapse
|
242
|
Wu A, Sharrett AR, Folsom AR, Alonso A, Walker KA, Gottesman RF, Gross AL, Rawlings AM, Schneider ALC, Coresh J. Midlife Hemostasis Measures, 20-Year Cognitive Decline, and Incident Dementia. Neurology 2023; 101:e1697-e1707. [PMID: 37652701 PMCID: PMC10624500 DOI: 10.1212/wnl.0000000000207771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 06/27/2023] [Indexed: 09/02/2023] Open
Abstract
BACKGROUND AND OBJECTIVES Blood concentrations of hemostatic factors affect thrombosis and bleeding diathesis and may contribute to cognitive impairment through modifiable vascular pathologies. Whether hemostasis, assessed in middle age, is associated with late-life cognitive impairment remains largely unknown in a community-dwelling population. METHODS Using data from 14,128 participants with cognitive function measurements in 1990-1992 from the Atherosclerosis Risk in Communities study, we assessed the associations of hemostasis measures with 20-year changes in cognitive performance and incident dementia. Activated partial thromboplastin time (aPTT) and level of fibrinogen, von Willebrand factor (VWF), factor VIII, factor VII, factor XI, d-dimer, and soluble thrombomodulin were measured in 1987-1989 or 1993-1995. Hemostasis measures were categorized into quintiles, with the lowest quintile indicating low coagulability. Cognitive performance was characterized using a combined z-score from 3 tests (that is, delayed word recall test [DWRT], digit symbol substitution [DSST], and word fluency test [WFT]), assessed in 1990-1992, 1996-1998, and 2011-2013. Dementia was determined either from in-person evaluations or using dementia surveillance through 2017. Mixed-effects models and Cox proportional hazards models were used to assess cognitive trajectories and risk of dementia, respectively. RESULTS Among 12,765 participants with hemostasis measures in 1987-1989, who were aged 47-70 years at the first cognitive assessment, we observed significant trends of shorter aPTT (p for trend <0.001; difference in 20-year cognitive decline for fifth vs first quintile [Q5 vs Q1]: -0.104 [95% CI -0.160 to -0.048]) and higher levels of factor VII (p < 0.002; Q5 vs Q1: -0.085 [-0.142, -0.028]) and factor VIII (p = 0.033; Q4 vs Q1: -0.055 [-0.111, -0.000]) with greater 20-year cognitive declines. The associations with the decline in DSST were stronger than those with the decline in WFT or DWRT. Consistently, shorter aPTT and higher factor VIII levels were associated with higher dementia risk with HRs for Q5 vs Q1 of 1.23 (95% CI 1.07 to 1.42) and 1.17 (1.01-1.36), respectively, and p for trend of 0.008 and 0.024, respectively. DISCUSSION Overall, our study found consistent trend associations of aPTT and factor VIII measured in midlife with cognitive decline and incident dementia over 20 years, likely driven by vascular pathologies.
Collapse
Affiliation(s)
- Aozhou Wu
- From the Johns Hopkins University (A.W., A.R.S., A.L.G., J.C.), Bloomberg School of Public Health, Baltimore, MD; University of Minnesota (A.R.F.), School of Public Health, Minneapolis; Department of Epidemiology (A.A.), Rollins School of Public Health, Emory University, Atlanta, GA; Laboratory of Behavioral Neuroscience (K.A.W.), Intramural Research Program, National Institute on Aging, Baltimore; National Institute of Neurological Disorders and Stroke Intramural Program (R.F.G.), NIH, Bethesda, MD; Sanofi (A.M.R.), Cambridge, MA; and Division of Neurocritical Care (A.L.C.S.), Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - A Richey Sharrett
- From the Johns Hopkins University (A.W., A.R.S., A.L.G., J.C.), Bloomberg School of Public Health, Baltimore, MD; University of Minnesota (A.R.F.), School of Public Health, Minneapolis; Department of Epidemiology (A.A.), Rollins School of Public Health, Emory University, Atlanta, GA; Laboratory of Behavioral Neuroscience (K.A.W.), Intramural Research Program, National Institute on Aging, Baltimore; National Institute of Neurological Disorders and Stroke Intramural Program (R.F.G.), NIH, Bethesda, MD; Sanofi (A.M.R.), Cambridge, MA; and Division of Neurocritical Care (A.L.C.S.), Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Aaron R Folsom
- From the Johns Hopkins University (A.W., A.R.S., A.L.G., J.C.), Bloomberg School of Public Health, Baltimore, MD; University of Minnesota (A.R.F.), School of Public Health, Minneapolis; Department of Epidemiology (A.A.), Rollins School of Public Health, Emory University, Atlanta, GA; Laboratory of Behavioral Neuroscience (K.A.W.), Intramural Research Program, National Institute on Aging, Baltimore; National Institute of Neurological Disorders and Stroke Intramural Program (R.F.G.), NIH, Bethesda, MD; Sanofi (A.M.R.), Cambridge, MA; and Division of Neurocritical Care (A.L.C.S.), Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Alvaro Alonso
- From the Johns Hopkins University (A.W., A.R.S., A.L.G., J.C.), Bloomberg School of Public Health, Baltimore, MD; University of Minnesota (A.R.F.), School of Public Health, Minneapolis; Department of Epidemiology (A.A.), Rollins School of Public Health, Emory University, Atlanta, GA; Laboratory of Behavioral Neuroscience (K.A.W.), Intramural Research Program, National Institute on Aging, Baltimore; National Institute of Neurological Disorders and Stroke Intramural Program (R.F.G.), NIH, Bethesda, MD; Sanofi (A.M.R.), Cambridge, MA; and Division of Neurocritical Care (A.L.C.S.), Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Keenan A Walker
- From the Johns Hopkins University (A.W., A.R.S., A.L.G., J.C.), Bloomberg School of Public Health, Baltimore, MD; University of Minnesota (A.R.F.), School of Public Health, Minneapolis; Department of Epidemiology (A.A.), Rollins School of Public Health, Emory University, Atlanta, GA; Laboratory of Behavioral Neuroscience (K.A.W.), Intramural Research Program, National Institute on Aging, Baltimore; National Institute of Neurological Disorders and Stroke Intramural Program (R.F.G.), NIH, Bethesda, MD; Sanofi (A.M.R.), Cambridge, MA; and Division of Neurocritical Care (A.L.C.S.), Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Rebecca F Gottesman
- From the Johns Hopkins University (A.W., A.R.S., A.L.G., J.C.), Bloomberg School of Public Health, Baltimore, MD; University of Minnesota (A.R.F.), School of Public Health, Minneapolis; Department of Epidemiology (A.A.), Rollins School of Public Health, Emory University, Atlanta, GA; Laboratory of Behavioral Neuroscience (K.A.W.), Intramural Research Program, National Institute on Aging, Baltimore; National Institute of Neurological Disorders and Stroke Intramural Program (R.F.G.), NIH, Bethesda, MD; Sanofi (A.M.R.), Cambridge, MA; and Division of Neurocritical Care (A.L.C.S.), Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Alden L Gross
- From the Johns Hopkins University (A.W., A.R.S., A.L.G., J.C.), Bloomberg School of Public Health, Baltimore, MD; University of Minnesota (A.R.F.), School of Public Health, Minneapolis; Department of Epidemiology (A.A.), Rollins School of Public Health, Emory University, Atlanta, GA; Laboratory of Behavioral Neuroscience (K.A.W.), Intramural Research Program, National Institute on Aging, Baltimore; National Institute of Neurological Disorders and Stroke Intramural Program (R.F.G.), NIH, Bethesda, MD; Sanofi (A.M.R.), Cambridge, MA; and Division of Neurocritical Care (A.L.C.S.), Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Andreea M Rawlings
- From the Johns Hopkins University (A.W., A.R.S., A.L.G., J.C.), Bloomberg School of Public Health, Baltimore, MD; University of Minnesota (A.R.F.), School of Public Health, Minneapolis; Department of Epidemiology (A.A.), Rollins School of Public Health, Emory University, Atlanta, GA; Laboratory of Behavioral Neuroscience (K.A.W.), Intramural Research Program, National Institute on Aging, Baltimore; National Institute of Neurological Disorders and Stroke Intramural Program (R.F.G.), NIH, Bethesda, MD; Sanofi (A.M.R.), Cambridge, MA; and Division of Neurocritical Care (A.L.C.S.), Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Andrea Lauren Christman Schneider
- From the Johns Hopkins University (A.W., A.R.S., A.L.G., J.C.), Bloomberg School of Public Health, Baltimore, MD; University of Minnesota (A.R.F.), School of Public Health, Minneapolis; Department of Epidemiology (A.A.), Rollins School of Public Health, Emory University, Atlanta, GA; Laboratory of Behavioral Neuroscience (K.A.W.), Intramural Research Program, National Institute on Aging, Baltimore; National Institute of Neurological Disorders and Stroke Intramural Program (R.F.G.), NIH, Bethesda, MD; Sanofi (A.M.R.), Cambridge, MA; and Division of Neurocritical Care (A.L.C.S.), Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia
| | - Josef Coresh
- From the Johns Hopkins University (A.W., A.R.S., A.L.G., J.C.), Bloomberg School of Public Health, Baltimore, MD; University of Minnesota (A.R.F.), School of Public Health, Minneapolis; Department of Epidemiology (A.A.), Rollins School of Public Health, Emory University, Atlanta, GA; Laboratory of Behavioral Neuroscience (K.A.W.), Intramural Research Program, National Institute on Aging, Baltimore; National Institute of Neurological Disorders and Stroke Intramural Program (R.F.G.), NIH, Bethesda, MD; Sanofi (A.M.R.), Cambridge, MA; and Division of Neurocritical Care (A.L.C.S.), Department of Neurology, University of Pennsylvania Perelman School of Medicine, Philadelphia.
| |
Collapse
|
243
|
Bradley CP, Berry C. Microvascular arterial disease of the brain and the heart: a shared pathogenesis. QJM 2023; 116:829-834. [PMID: 37467080 PMCID: PMC10593384 DOI: 10.1093/qjmed/hcad158] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 05/22/2023] [Indexed: 07/21/2023] Open
Abstract
Microvascular arterial disease in the heart manifest as coronary microvascular dysfunction. This condition causes microvascular angina and is associated increased morbidity and mortality. Microvascular arterial disease in the brain is referred to as cerebrovascular small vessel disease. This is responsible for 45% of dementias and 25% of ischaemic strokes. The heart and brain share similar vascular anatomy and common pathogenic risk factors are associated with the development of both coronary microvascular dysfunction and cerebrovascular small vessel disease. Microvascular disease in the heart and brain also appear to share common multisystem pathophysiological mechanisms. Further studies on diagnostic approaches, epidemiology and development of disease-modifying therapy seem warranted.
Collapse
Affiliation(s)
- C P Bradley
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
- NHS Golden Jubilee Hospital, Clydebank, UK
| | - C Berry
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, UK
- NHS Golden Jubilee Hospital, Clydebank, UK
| |
Collapse
|
244
|
Tarasova I, Trubnikova O, Kukhareva I, Syrova I, Sosnina A, Kupriyanova D, Barbarash O. A Comparison of Two Multi-Tasking Approaches to Cognitive Training in Cardiac Surgery Patients. Biomedicines 2023; 11:2823. [PMID: 37893196 PMCID: PMC10604887 DOI: 10.3390/biomedicines11102823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/13/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
BACKGROUND The multi-tasking approach may be promising for cognitive rehabilitation in cardiac surgery patients due to a significant effect on attentional and executive functions. This study aimed to compare the neuropsychological changes in patients who have undergone two variants of multi-tasking training and a control group in the early postoperative period of coronary artery bypass grafting (CABG). METHODS One hundred and ten CABG patients were divided into three groups: cognitive training (CT) I (a postural balance task with mental arithmetic, verbal fluency, and divergent tasks) (n = 30), CT II (a simple visual-motor reaction with mental arithmetic, verbal fluency, and divergent tasks) (n = 40), and control (n = 40). RESULTS Two or more cognitive indicators improved in 93.3% of CT I patients, in 72.5% of CT II patients, and in 62.5% of control patients; CT I patients differed from CT II and control (p = 0.04 and p = 0.008, respectively). The improving short-term memory and attention was found more frequently in the CT I group as compared to control (56.7% vs. 15%; p = 0.0005). The cognitive improvement of all domains (psychomotor and executive functions, attention, and short-term memory) was also revealed in CT I patients more frequently than CT II (46.7% vs. 20%; p = 0.02) and control (46.7% vs. 5%; p = 0.0005). CONCLUSIONS The CT I multi-tasking training was more effective at improving the cognitive performance in cardiac surgery patients as compared to CT II training and standard post-surgery management. The findings of this study will be helpful for future studies involving multi-tasking training.
Collapse
Affiliation(s)
- Irina Tarasova
- Department of Clinical Cardiology, Research Institute for Complex Issues of Cardiovascular Diseases, Sosnovy Blvd., 6, 650002 Kemerovo, Russia; (O.T.); (I.S.)
| | | | | | | | | | | | | |
Collapse
|
245
|
Mohamed-Mohamed H, García-Morales V, Sánchez Lara EM, González-Acedo A, Pardo-Moreno T, Tovar-Gálvez MI, Melguizo-Rodríguez L, Ramos-Rodríguez JJ. Physiological Mechanisms Inherent to Diabetes Involved in the Development of Dementia: Alzheimer's Disease. Neurol Int 2023; 15:1253-1272. [PMID: 37873836 PMCID: PMC10594452 DOI: 10.3390/neurolint15040079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/06/2023] [Accepted: 10/08/2023] [Indexed: 10/25/2023] Open
Abstract
Type 2 diabetes mellitus (T2D) is a metabolic disease reaching pandemic levels worldwide. In parallel, Alzheimer's disease (AD) and vascular dementia (VaD) are the two leading causes of dementia in an increasingly long-living Western society. Numerous epidemiological studies support the role of T2D as a risk factor for the development of dementia. However, few basic science studies have focused on the possible mechanisms involved in this relationship. On the other hand, this review of the literature also aims to explore the relationship between T2D, AD and VaD. The data found show that there are several alterations in the central nervous system that may be promoting the development of T2D. In addition, there are some mechanisms by which T2D may contribute to the development of neurodegenerative diseases such as AD or VaD.
Collapse
Affiliation(s)
- Himan Mohamed-Mohamed
- Department of Physiology, Faculty of Health Sciences of Ceuta, University of Granada, 51001 Ceuta, Spain
| | - Victoria García-Morales
- Physiology Area, Department of Biomedicine, Biotechnology and Public Health, Faculty of Medicine, University of Cádiz, Pl. Falla, 9, 11003 Cádiz, Spain
| | - Encarnación María Sánchez Lara
- Department of Personalidad, Evaluación y Tratamiento Psicológico, Faculty of Health Sciences (Ceuta), University of Granada, 51001 Ceuta, Spain;
| | - Anabel González-Acedo
- Department of Nursing, Faculty of Health Sciences of Ceuta, University of Granada, 51001 Ceuta, Spain
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, 18016 Granada, Spain
| | - Teresa Pardo-Moreno
- Department of Nursing, Faculty of Health Sciences of Ceuta, University of Granada, 51001 Ceuta, Spain
| | - María Isabel Tovar-Gálvez
- Department of Nursing, Faculty of Health Sciences of Ceuta, University of Granada, 51001 Ceuta, Spain
| | - Lucía Melguizo-Rodríguez
- Department of Nursing, Faculty of Health Sciences of Ceuta, University of Granada, 51001 Ceuta, Spain
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, 18016 Granada, Spain
| | - Juan José Ramos-Rodríguez
- Department of Physiology, Faculty of Health Sciences of Ceuta, University of Granada, 51001 Ceuta, Spain
| |
Collapse
|
246
|
Li T, Ye M, Yang G, Diao S, Zhou Y, Qin Y, Ding D, Zhu M, Fang Q. Regional white matter hyperintensity volume predicts persistent cognitive impairment in acute lacunar infarct patients. Front Neurol 2023; 14:1265743. [PMID: 37881309 PMCID: PMC10595143 DOI: 10.3389/fneur.2023.1265743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 09/15/2023] [Indexed: 10/27/2023] Open
Abstract
Background White matter hyperintensity (WMH) is often described in acute lacunar stroke (ALS) patients. However, the specific relationship between regional WMH volume and persistent cognitive impairment remains unclear. Methods We enrolled patients with ALS who were hospitalized at the First Affiliated Hospital of Soochow University between January 2020 and November 2022. All patients were assessed for global cognitive function using the Montreal Cognitive Assessment (MoCA) scale at 14 ± 2 days and 6 months after the onset of ALS. Manifestations of chronic cerebral small vessel disease (CSVD) were assessed via MRI scan. The distributions of regional WMH were segmented, and their relationship with cognitive impairment was evaluated. Results A total of 129 patients were enrolled. Baseline frontal WMH volume (OR = 1.18, P = 0.04) was an independent risk factor for long-term cognitive impairment after ALS. Furthermore, the presence of WMH at the genu of the corpus callosum (GCC) at baseline (OR = 3.1, P = 0.033) was strongly associated with persistent cognitive decline. Multivariable logistic regression analysis showed that depression (OR = 6.252, P = 0.029), NIHSS score (OR = 1.24, P = 0.011), and albumin at admission (OR = 0.841, P = 0.032) were also important determinants of long-term cognitive impairment after ALS. Conclusions Our study found that WMH, especially frontal WMH volume and the presence of WMH at the GCC at baseline, independently contributed to long-term cognitive decline in ALS patients. This study provides new evidence of the clinical relationship between regional WMH volume and cognitive impairment in ALS patients.
Collapse
Affiliation(s)
- Tan Li
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Mengfan Ye
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Guopeng Yang
- Suzhou Jiasheng Medical Instrument Co., Ltd., Suzhou, Jiangsu, China
| | - Shanshan Diao
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yun Zhou
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yiren Qin
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Dongxue Ding
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Mo Zhu
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qi Fang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
247
|
Filippenkov IB, Khrunin AV, Mozgovoy IV, Dergunova LV, Limborska SA. Are Ischemic Stroke and Alzheimer's Disease Genetically Consecutive Pathologies? Biomedicines 2023; 11:2727. [PMID: 37893101 PMCID: PMC10604604 DOI: 10.3390/biomedicines11102727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 10/01/2023] [Accepted: 10/03/2023] [Indexed: 10/29/2023] Open
Abstract
Complex diseases that affect the functioning of the central nervous system pose a major problem for modern society. Among these, ischemic stroke (IS) holds a special place as one of the most common causes of disability and mortality worldwide. Furthermore, Alzheimer's disease (AD) ranks first among neurodegenerative diseases, drastically reducing brain activity and overall life quality and duration. Recent studies have shown that AD and IS share several common risk and pathogenic factors, such as an overlapping genomic architecture and molecular signature. In this review, we will summarize the genomics and RNA biology studies of IS and AD, discussing the interconnected nature of these pathologies. Additionally, we highlight specific genomic points and RNA molecules that can serve as potential tools in predicting the risks of diseases and developing effective therapies in the future.
Collapse
Affiliation(s)
| | | | | | | | - Svetlana A. Limborska
- Laboratory of Human Molecular Genetics, National Research Center “Kurchatov Institute”, Kurchatov Sq. 2, 123182 Moscow, Russia (A.V.K.); (I.V.M.); (L.V.D.)
| |
Collapse
|
248
|
Zhu M, Pan G, Luo F, Sui S, Zhang Y. Modified Suanzaoren decoction in treating post-stroke cognitive impairment with comorbid insomnia symptoms: A clinical trial. Medicine (Baltimore) 2023; 102:e35239. [PMID: 37800827 PMCID: PMC10553162 DOI: 10.1097/md.0000000000035239] [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: 06/05/2023] [Accepted: 08/24/2023] [Indexed: 10/07/2023] Open
Abstract
BACKGROUND Cognitive impairment and insomnia are common complications for stroke patients, and often coexist without effective therapy. Modified Suanzaoren decoction (M-SZRD), derived from a famous classic prescription, has been used as an alternative treatment for these patients. The objective of this study is to investigate the effectiveness of M-SZRD in treating post-stroke cognitive impairment with comorbid insomnia symptoms. METHODS A total of 80 participants were randomly assigned into 2 groups to 40 cases in the treatment group (treated with modified Suanzaoren decoction) and 40 cases in the control group (treated with zolpidem). The intervention period was 4 weeks. Cognitive function, sleep quality, depression, and anxiety disorders were evaluated in both groups before and after treatment. Clinical assessment of patients with stroke included National Institutes of Health Stroke Scale and Barthel Index evaluations. Hormone levels of the hypothalamic-pituitary-adrenal and hypothalamus-pituitary-thyroid axis were also measured. RESULTS Out of the total 80 participants, 5 withdrew during the experiment and did not complete the study, leaving 75 patients for analysis to 38 in the treatment group and 37 in the control group. The findings showed that M-SZRD was more effective than the control group in improving cognitive function (P = .006). However, both groups were found to have a similar effect in improving insomnia (P = .323). There was no significant difference between the 2 groups in terms of activities of daily living and National Institutes of Health Stroke Scale improvement. M-SZRD was superior to the control group in improving depression state (P = .034), but when including dropouts in the intention-to-treat analysis, the difference was not statistically significant (P = .150). Furthermore, the M-SZRD group was better than the control group in reducing cortisol levels (P = .036), and the improvement in serum-free triiodothyronine (FT3) levels was also more significant in the M-SZRD group than in the control group (P = .0007). CONCLUSION M-SZRD is a more effective treatment for improving cognitive function in patients with post-stroke cognitive impairment and comorbid insomnia symptoms, possibly by regulating the cortisol levels of the hypothalamic-pituitary-adrenal axis and FT3 levels of the hypothalamus-pituitary-thyroid axis.
Collapse
Affiliation(s)
- Mingjin Zhu
- Department of Rehabilitation Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Guoyua Pan
- Department of Rehabilitation Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Fang Luo
- Department of Rehabilitation Medicine, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Shuyan Sui
- Department of Neurology, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Yonghua Zhang
- Department of Clinical Psychology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, China
| |
Collapse
|
249
|
Zhang T, Liu W, Bai Q, Gao S. Virtual reality technology in the rehabilitation of post-stroke cognitive impairment: an opinion article on recent findings. Front Psychol 2023; 14:1271458. [PMID: 37849482 PMCID: PMC10577207 DOI: 10.3389/fpsyg.2023.1271458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 09/20/2023] [Indexed: 10/19/2023] Open
Affiliation(s)
- Ting Zhang
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, China
- Department of Traditional Chinese Medicine, University Hospital, Zhejiang Normal University, Jinhua, China
| | - Wei Liu
- Physical Education College, Guangxi University of Science and Technology, Liuzhou, China
| | - Qingping Bai
- Physical Education College, Guangxi University of Science and Technology, Liuzhou, China
| | - Song Gao
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, China
| |
Collapse
|
250
|
Samuelsson J, Marseglia A, Lindberg O, Westman E, Pereira JB, Shams S, Kern S, Ahlner F, Rothenberg E, Skoog I, Zettergren A. Associations between dietary patterns and dementia-related neuroimaging markers. Alzheimers Dement 2023; 19:4629-4640. [PMID: 36960849 DOI: 10.1002/alz.13048] [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: 12/05/2022] [Revised: 02/03/2023] [Accepted: 02/21/2023] [Indexed: 03/25/2023]
Abstract
BACKGROUND The exploration of associations between dietary patterns and dementia-related neuroimaging markers can provide insights on food combinations that may impact brain integrity. METHODS Data were derived from the Swedish Gothenburg H70 Birth Cohort Study (n = 610). Three dietary patterns were obtained using principal component analysis. Magnetic resonance imaging markers included cortical thickness, an Alzheimer's disease (AD) signature score, small vessel disease, and white matter microstructural integrity. Adjusted linear/ordinal regression analyses were performed. RESULTS A high-protein and alcohol dietary pattern was negatively associated with cortical thickness in the whole brain (Beta: -0.011; 95% confidence interval [CI]: -0.018 to -0.003), and with an Alzheimer's disease cortical thickness signature score (Beta: -0.013; 95% CI: -0.024 to -0.001). A positive association was found between a Mediterranean-like dietary pattern and white matter microstructural integrity (Beta: 0.078; 95% CI: 0.002-0.154). No associations were found with a Western-like dietary pattern. DISCUSSION Dietary patterns may impact brain integrity through neurodegenerative and vascular pathways. HIGHLIGHTS Certain dietary patterns were associated with dementia-related neuroimaging markers. A Mediterranean dietary pattern was positively associated with white matter microstructure. A high-protein and alcohol pattern was negatively associated with cortical thickness.
Collapse
Affiliation(s)
- Jessica Samuelsson
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Neuropsychiatric Epidemiology Unit, Sahlgrenska Academy, Centre for Ageing and Health (AGECAP), University of Gothenburg, Mölndal, Sweden
| | - Anna Marseglia
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Olof Lindberg
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Eric Westman
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Joana B Pereira
- Department of Neurobiology, Care Sciences and Society, Division of Clinical Geriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
- Department of Clinical Sciences, Clinical Memory Research Unit, Lund University, Malmo, Sweden
| | - Sara Shams
- Department of Radiology, Karolinska University Hospital, The Institution for Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
- Department of Radiology, Stanford University Hospital, Stanford, California, USA
| | - Silke Kern
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Neuropsychiatric Epidemiology Unit, Sahlgrenska Academy, Centre for Ageing and Health (AGECAP), University of Gothenburg, Mölndal, Sweden
| | - Felicia Ahlner
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Neuropsychiatric Epidemiology Unit, Sahlgrenska Academy, Centre for Ageing and Health (AGECAP), University of Gothenburg, Mölndal, Sweden
| | | | - Ingmar Skoog
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Neuropsychiatric Epidemiology Unit, Sahlgrenska Academy, Centre for Ageing and Health (AGECAP), University of Gothenburg, Mölndal, Sweden
- Department of Psychiatry, Cognition and Old Age Psychiatry, Sahlgrenska University Hospital, Region Västra Götaland, Mölndal, Sweden
| | - Anna Zettergren
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, Neuropsychiatric Epidemiology Unit, Sahlgrenska Academy, Centre for Ageing and Health (AGECAP), University of Gothenburg, Mölndal, Sweden
| |
Collapse
|