1
|
Li H, Liu J, Liang Q, Yu Y, Sun G. Effect of Vascular Senescence on the Efficacy and Safety of Warfarin: Insights from Rat Models and a Prospective Cohort Study. J Pharmacol Exp Ther 2024; 391:39-50. [PMID: 39095206 DOI: 10.1124/jpet.124.002265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 07/03/2024] [Accepted: 07/12/2024] [Indexed: 08/04/2024] Open
Abstract
Warfarin, with its narrow therapeutic range, requires the understanding of various influencing factors for personalized medication. Vascular senescence, marked by vascular stiffening and endothelial dysfunction, has an unclear effect on the efficacy and safety of warfarin. Based on previous studies, we hypothesized that vascular senescence increases the risk of bleeding during warfarin therapy. This study aimed to explore these effects using animal models and clinical cohorts. We established rat models of vascular senescence and calcification using d-galactose, vitamin D, and nicotine. After validating the models, we examined changes in the international normalized ratio (INR) at fixed warfarin doses (0.20 and 0.35 mg/kg). We found that vascular senescence caused significantly elevated INR values and increased bleeding risk. In the prospective clinical cohort study (NCT06428110), hospitalized warfarin patients with standard dose adjustments were divided into vascular senescence and control groups based on ultrasound and computed tomography diagnosis. Using propensity score matching to exclude the influence of confounding factors, we found that the vascular senescence group had lower steady-state warfarin doses and larger dose adjustments, with a higher probability of INR exceeding the therapeutic range. The vascular senescence group tended to experience more bleeding or thromboembolic/ischemic events during 1 year of follow-up, while there was no statistical difference. In conclusion, vascular senescence leads to unstable INR values and increases higher bleeding risk during warfarin therapy, highlighting the importance of considering vascular senescence in future precision warfarin therapies. SIGNIFICANCE STATEMENT: Many factors influence warfarin efficacy; however, the effect of vascular senescence remains unclear. This study aimed to investigate the effects of vascular senescence on the efficacy and safety of warfarin. Through both rat models and clinical cohort studies, our findings indicated that vascular senescence may compromise the stability of warfarin, presenting challenges in maintaining its efficacy and safety.
Collapse
Affiliation(s)
- Haobin Li
- Department of Pharmacy, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Jing Liu
- Department of Pharmacy, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Qing Liang
- Department of Pharmacy, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Yan Yu
- Department of Pharmacy, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Guangchun Sun
- Department of Pharmacy, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| |
Collapse
|
2
|
Charisis S, Short MI, Bernal R, Kautz TF, Treviño HA, Mathews J, Dediós AGV, Muhammad JAS, Luckey AM, Aslam A, Himali JJ, Shipp EL, Habes M, Beiser AS, DeCarli C, Scarmeas N, Ramachandran VS, Seshadri S, Maillard P, Satizabal CL. Leptin bioavailability and markers of brain atrophy and vascular injury in the middle age. Alzheimers Dement 2024; 20:5849-5860. [PMID: 39132759 PMCID: PMC11497668 DOI: 10.1002/alz.13879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 03/01/2024] [Accepted: 03/24/2024] [Indexed: 08/13/2024]
Abstract
INTRODUCTION We investigated the associations of leptin markers with cognitive function and magnetic resonance imaging (MRI) measures of brain atrophy and vascular injury in healthy middle-aged adults. METHODS We included 2262 cognitively healthy participants from the Framingham Heart Study with neuropsychological evaluation; of these, 2028 also had available brain MRI. Concentrations of leptin, soluble leptin receptor (sOB-R), and their ratio (free leptin index [FLI]), indicating leptin bioavailability, were measured using enzyme-linked immunosorbent assays. Cognitive and MRI measures were derived using standardized protocols. RESULTS Higher sOB-R was associated with lower fractional anisotropy (FA, β = -0.114 ± 0.02, p < 0.001), and higher free water (FW, β = 0.091 ± 0.022, p < 0.001) and peak-width skeletonized mean diffusivity (PSMD, β = 0.078 ± 0.021, p < 0.001). Correspondingly, higher FLI was associated with higher FA (β = 0.115 ± 0.027, p < 0.001) and lower FW (β = -0.096 ± 0.029, p = 0.001) and PSMD (β = -0.085 ± 0.028, p = 0.002). DISCUSSION Higher leptin bioavailability was associated with better white matter (WM) integrity in healthy middle-aged adults, supporting the putative neuroprotective role of leptin in late-life dementia risk. HIGHLIGHTS Higher leptin bioavailability was related to better preservation of white matter microstructure. Higher leptin bioavailability during midlife might confer protection against dementia. Potential benefits might be even stronger for individuals with visceral obesity. DTI measures might be sensitive surrogate markers of subclinical neuropathology.
Collapse
Affiliation(s)
- Sokratis Charisis
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesUT Health San AntonioSan AntonioTexasUSA
| | - Meghan I. Short
- Institute for Clinical Research and Health Policy StudiesTufts Medical CenterBostonMassachusettsUSA
| | - Rebecca Bernal
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesUT Health San AntonioSan AntonioTexasUSA
| | - Tiffany F. Kautz
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesUT Health San AntonioSan AntonioTexasUSA
| | - Hector A. Treviño
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesUT Health San AntonioSan AntonioTexasUSA
| | - Julia Mathews
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesUT Health San AntonioSan AntonioTexasUSA
| | | | - Jazmyn A. S. Muhammad
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesUT Health San AntonioSan AntonioTexasUSA
| | - Alison M. Luckey
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesUT Health San AntonioSan AntonioTexasUSA
| | - Asra Aslam
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesUT Health San AntonioSan AntonioTexasUSA
| | - Jayandra J. Himali
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesUT Health San AntonioSan AntonioTexasUSA
- The Framingham Heart StudyFraminghamMassachusettsUSA
- Department of BiostatisticsBoston University School of Public HealthBostonMassachusettsUSA
| | - Eric L. Shipp
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesUT Health San AntonioSan AntonioTexasUSA
| | - Mohamad Habes
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesUT Health San AntonioSan AntonioTexasUSA
| | - Alexa S. Beiser
- The Framingham Heart StudyFraminghamMassachusettsUSA
- Department of BiostatisticsBoston University School of Public HealthBostonMassachusettsUSA
| | - Charles DeCarli
- Department of NeurologyUniversity of California, DavisSacramentoCaliforniaUSA
| | - Nikolaos Scarmeas
- 1st Department of NeurologyNational and Kapodistrian University of AthensAthensGreece
- Taub Institute for Research in Alzheimer's Disease and the Aging Brainthe Gertrude H. Sergievsky CenterColumbia UniversityNew YorkNew YorkUSA
| | - Vasan S. Ramachandran
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesUT Health San AntonioSan AntonioTexasUSA
- The Framingham Heart StudyFraminghamMassachusettsUSA
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesUT Health San AntonioSan AntonioTexasUSA
- The Framingham Heart StudyFraminghamMassachusettsUSA
- Department of BiostatisticsBoston University School of Public HealthBostonMassachusettsUSA
| | - Pauline Maillard
- Department of NeurologyUniversity of California, DavisSacramentoCaliforniaUSA
| | - Claudia L. Satizabal
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative DiseasesUT Health San AntonioSan AntonioTexasUSA
- The Framingham Heart StudyFraminghamMassachusettsUSA
- Department of BiostatisticsBoston University School of Public HealthBostonMassachusettsUSA
| |
Collapse
|
3
|
Wong Zhang DE, Gibson Hughes TA, Figueiredo Galvao HB, Lo C, Dinh QN, Zhang SR, Kim HA, Selvaraji S, Clarkson AN, Arumugam TV, Drummond G, Sobey CG, De Silva TM. Post-stroke cognitive impairment and brain hemorrhage are augmented in hypertensive mice. J Cereb Blood Flow Metab 2024:271678X241262127. [PMID: 38886874 DOI: 10.1177/0271678x241262127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/20/2024]
Abstract
Hypertension is a major risk factor for both stroke and cognitive impairment, but it is unclear whether it may specifically affect post-stroke cognitive impairment. We assessed the effect of hypertension and/or stroke on brain injury, cognitive outcome, and the brain transcriptomic profile. C57BL/6J mice (n = 117; 3-5 mo.) received s.c. infusion of either saline or angiotensin II followed by sham surgery or photothrombotic stroke targeting the prefrontal cortex seven days later. Cognitive function was assessed with the Barnes maze and RNA sequencing was used to quantify transcriptomic changes in the brain. Angiotensin II treatment produced spontaneous hemorrhaging after stroke. In the Barnes maze, hypertensive mice that received stroke surgery had an increased escape latency compared to other groups (day 3: hypertensive + stroke = 166.6 ± 6.0 s vs. hypertensive + sham = 122.8 ± 13.8 s vs. normotensive + stroke = 139.9 ± 10.1 s vs. normotensive + sham = 101.9 ± 16.7 s), consistent with impaired cognition. RNA sequencing revealed >1500 differentially expressed genes related to neuroinflammation in hypertensive + stroke vs. normotensive + stroke, which included genes associated with apoptosis, microRNAs, autophagy, anti-cognitive biomarkers and Wnt signaling. Overall, we show that the combination of hypertension and stroke resulted in greater learning impairment and brain injury.
Collapse
Affiliation(s)
- David E Wong Zhang
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Tayla A Gibson Hughes
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Hericka B Figueiredo Galvao
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Cecilia Lo
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Quynh Nhu Dinh
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Shenpeng R Zhang
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Hyun Ah Kim
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Sharmalee Selvaraji
- Department of Physiology, Yong Loo Lin School Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Singapore, Singapore
| | - Andrew N Clarkson
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Thiruma V Arumugam
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Grant Drummond
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - Christopher G Sobey
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| | - T Michael De Silva
- Centre for Cardiovascular Biology and Disease Research and La Trobe Institute for Molecular Sciences (LIMS), La Trobe University, Victoria, Australia
- Department of Microbiology, Anatomy, Physiology & Pharmacology, School of Agriculture, Biomedicine, Environment, La Trobe University, Victoria, Australia
| |
Collapse
|
4
|
Rudolph MD, Sutphen CL, Register TC, Whitlow CT, Solingapuram Sai KK, Hughes TM, Bateman JR, Dage JL, Russ KA, Mielke MM, Craft S, Lockhart SN. Associations among plasma, MRI, and amyloid PET biomarkers of Alzheimer's disease and related dementias and the impact of health-related comorbidities in a community-dwelling cohort. Alzheimers Dement 2024; 20:4159-4173. [PMID: 38747525 PMCID: PMC11180870 DOI: 10.1002/alz.13835] [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: 11/08/2023] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 06/05/2024]
Abstract
INTRODUCTION We evaluated associations between plasma and neuroimaging-derived biomarkers of Alzheimer's disease and related dementias and the impact of health-related comorbidities. METHODS We examined plasma biomarkers (neurofilament light chain, glial fibrillary acidic protein, amyloid beta [Aβ] 42/40, phosphorylated tau 181) and neuroimaging measures of amyloid deposition (Aβ-positron emission tomography [PET]), total brain volume, white matter hyperintensity volume, diffusion-weighted fractional anisotropy, and neurite orientation dispersion and density imaging free water. Participants were adjudicated as cognitively unimpaired (CU; N = 299), mild cognitive impairment (MCI; N = 192), or dementia (DEM; N = 65). Biomarkers were compared across groups stratified by diagnosis, sex, race, and APOE ε4 carrier status. General linear models examined plasma-imaging associations before and after adjusting for demographics (age, sex, race, education), APOE ε4 status, medications, diagnosis, and other factors (estimated glomerular filtration rate [eGFR], body mass index [BMI]). RESULTS Plasma biomarkers differed across diagnostic groups (DEM > MCI > CU), were altered in Aβ-PET-positive individuals, and were associated with poorer brain health and kidney function. DISCUSSION eGFR and BMI did not substantially impact associations between plasma and neuroimaging biomarkers. HIGHLIGHTS Plasma biomarkers differ across diagnostic groups (DEM > MCI > CU) and are altered in Aβ-PET-positive individuals. Altered plasma biomarker levels are associated with poorer brain health and kidney function. Plasma and neuroimaging biomarker associations are largely independent of comorbidities.
Collapse
Affiliation(s)
- Marc D. Rudolph
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Courtney L. Sutphen
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Thomas C. Register
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Christopher T. Whitlow
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Kiran K. Solingapuram Sai
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Timothy M. Hughes
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - James R. Bateman
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Jeffrey L. Dage
- Department Of NeurologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Kristen A. Russ
- Department Of NeurologyIndiana University School of MedicineIndianapolisIndianaUSA
| | - Michelle M. Mielke
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Suzanne Craft
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| | - Samuel N. Lockhart
- Department of Internal MedicineWake Forest University School of MedicineWinston‐SalemNorth CarolinaUSA
| |
Collapse
|
5
|
Xu F, Xin Q, Ren M, Shi P, Wang B. Inhibition of piezo1 prevents chronic cerebral hypoperfusion-induced cognitive impairment and blood brain barrier disruption. Neurochem Int 2024; 175:105702. [PMID: 38401846 DOI: 10.1016/j.neuint.2024.105702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 02/15/2024] [Accepted: 02/18/2024] [Indexed: 02/26/2024]
Abstract
Chronic cerebral hypoperfusion (CCH) plays a critical role in the onset and progression of vascular dementia (VD), which is now recognized as the second most common form of dementia after Alzheimer's disease (AD). The mechanosensitive piezo1 channel has been identified to play important roles in several neurological disorders. However, the roles and possible mechanisms of piezo1 in CCH-induced cognitive decline and blood brain barrier (BBB) disruption, as well as the underlying mechanisms remain elusive. In this study, the CCH model was established by bilateral common carotid artery occlusion in rats and by oxygen and glucose deprivation/reoxygenation (OGD/R) in bEnd.3 cells. The results demonstrated that the antagonist of piezo1 GsMTx4 ameliorated CCH-induced cognitive dysfunction and mitigated cerebral edema. Furthermore, this study indicated that GsMTx4 improved the permeability and integrity of BBB and protected cerebral microvasculature after CCH. In vitro, GsMTx4 improved cell viability, promoted the ability of cell motility and migration, and inhibited the degradation of BBB integrity-related proteins by inhibiting NLRP3 inflammasome activation. In addition, NLRP3 agonist abolished the beneficial effects of GsMTx4. Collectively, our results demonstrate that piezo1 might be involved in CCH-induced cognitive impairment and BBB damage, which may be at least partially mediated through regulation of NLRP3 inflammasome.
Collapse
Affiliation(s)
- Fei Xu
- Department of Vascular Surgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Department of Vascular Surgery, Jining NO.1 People's Hospital, Jining, 272000, China
| | - Qing Xin
- Department of Physiology, Jining Medical University, Jining, 272000, China
| | - Mengyao Ren
- Department of Physiology, Jining Medical University, Jining, 272000, China
| | - Peixin Shi
- Department of Physiology, Jining Medical University, Jining, 272000, China
| | - Bing Wang
- Department of Vascular Surgery, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| |
Collapse
|
6
|
Li B, Gu Z, Wang W, Du B, Wu C, Li B, Wang T, Yin G, Gao X, Chen J, Bi X, Zhang H, Sun X. The associations between peripheral inflammatory and lipid parameters, white matter hyperintensity, and cognitive function in patients with non-disabling ischemic cerebrovascular events. BMC Neurol 2024; 24:86. [PMID: 38438839 PMCID: PMC10910845 DOI: 10.1186/s12883-024-03591-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Accepted: 02/27/2024] [Indexed: 03/06/2024] Open
Abstract
BACKGROUND The global prevalence of VCI has increased steadily in recent years, but diagnostic biomarkers for VCI in patients with non-disabling ischemic cerebrovascular incidents (NICE) remain indefinite. The primary objective of this research was to investigate the relationship between peripheral serological markers, white matter damage, and cognitive function in individuals with NICE. METHODS We collected clinical data, demographic information, and medical history from 257 patients with NICE. Using the MoCA upon admission, patients were categorized into either normal cognitive function (NCF) or VCI groups. Furthermore, they were classified as having mild white matter hyperintensity (mWMH) or severe WMH based on Fazekas scores. We then compared the levels of serological markers between the cognitive function groups and the WMH groups. RESULTS Among 257 patients with NICE, 165 were male and 92 were female. Lymphocyte count (OR = 0.448, P < 0.001) and LDL-C/HDL-C (OR = 0.725, P = 0.028) were protective factors for cognitive function in patients with NICE. The sWMH group had a higher age and inflammation markers but a lower MoCA score, and lymphocyte count than the mWMH group. In the mWMH group, lymphocyte count (AUC = 0.765, P < 0.001) and LDL-C/HDL-C (AUC = 0.740, P < 0.001) had an acceptable diagnostic value for the diagnosis of VCI. In the sWMH group, no significant differences were found in serological markers between the NCF and VCI groups. CONCLUSION Lymphocyte count, LDL-C/HDL-C were independent protective factors for cognitive function in patients with NICE; they can be used as potential biological markers to distinguish VCI in patients with NICE and are applicable to subgroups of patients with mWMH.
Collapse
Affiliation(s)
- Binghan Li
- Department of Neurology, Shanghai Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Zhengsheng Gu
- Department of Neurology, Shanghai Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Weisen Wang
- Department of Neurology, Shanghai Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Bingying Du
- Department of Neurology, Shanghai Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Chenghao Wu
- Department of Neurology, Shanghai Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Bin Li
- Department of Neurology, Shanghai Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Tianren Wang
- Department of Neurology, Shanghai Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Ge Yin
- Department of Neurology, Shanghai Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Xin Gao
- Department of Neurology, Shanghai Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Jingjing Chen
- Department of Neurology, Shanghai Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Xiaoying Bi
- Department of Neurology, Shanghai Changhai Hospital, The Second Military Medical University, Shanghai, China
| | - Hailing Zhang
- Department of Neurology, Shanghai Changhai Hospital, The Second Military Medical University, Shanghai, China.
| | - Xu Sun
- Department of Neurology, Shanghai Changhai Hospital, The Second Military Medical University, Shanghai, China.
| |
Collapse
|
7
|
Fu X, Sun P, Zhang X, Zhu D, Qin Q, Lu J, Wang J. GABA in the anterior cingulate cortex mediates the association of white matter hyperintensities with executive function: a magnetic resonance spectroscopy study. Aging (Albany NY) 2024; 16:4282-4298. [PMID: 38441529 PMCID: PMC10968699 DOI: 10.18632/aging.205585] [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: 10/13/2023] [Accepted: 01/24/2024] [Indexed: 03/22/2024]
Abstract
White matter hyperintensities (WMH) and gamma-aminobutyric acid (GABA) are associated with executive function. Multiple studies suggested cortical alterations mediate WMH-related cognitive decline. The aim of this study was to investigate the crucial role of cortical GABA in the WMH patients. In the 87 WMH patients (46 mild and 41 moderate to severe) examined in this study, GABA levels in the anterior cingulate cortex (ACC) and posterior cingulate cortex (PCC) assessed by the Meshcher-Garwood point resolved spectroscopy (MEGA-PRESS) sequence, WMH volume and executive function were compared between the two groups. Partial correlation and mediation analyses were carried out to examine the GABA levels in mediating the association between WMH volume and executive function. Patients with moderate to severe WMH had lower GABA+/Cr in the ACC (p = 0.034) and worse executive function (p = 0.004) than mild WMH patients. In all WMH cases, the GABA+/Cr levels in the ACC mediated the negative correlation between WMH and executive function (ab: effect = -0.020, BootSE = 0.010, 95% CI: -0.042 to -0.004). This finding suggested GABA+/Cr levels in the ACC might serve as a protective factor or potential target for preventing the occurrence and progression of executive function decline in WMH people.
Collapse
Affiliation(s)
- Xiaona Fu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430030, China
| | - Peng Sun
- Clinical and Technical Support, Philips Healthcare, Beijing 100600, China
| | - Xinli Zhang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430030, China
| | - Dongyong Zhu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430030, China
| | - Qian Qin
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430030, China
| | - Jue Lu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430030, China
| | - Jing Wang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430030, China
| |
Collapse
|
8
|
Pavuluri K, Huston J, Ehman RL, Manduca A, Jack CR, Senjem ML, Vemuri P, Murphy MC. Associations between vascular health, brain stiffness and global cognitive function. Brain Commun 2024; 6:fcae073. [PMID: 38505229 PMCID: PMC10950054 DOI: 10.1093/braincomms/fcae073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 12/19/2023] [Accepted: 02/27/2024] [Indexed: 03/21/2024] Open
Abstract
Vascular brain injury results in loss of structural and functional connectivity and leads to cognitive impairment. Its various manifestations, including microinfarcts, microhaemorrhages and white matter hyperintensities, result in microstructural tissue integrity loss and secondary neurodegeneration. Among these, tissue microstructural alteration is a relatively early event compared with atrophy along the aging and neurodegeneration continuum. Understanding its association with cognition may provide the opportunity to further elucidate the relationship between vascular health and clinical outcomes. Magnetic resonance elastography offers a non-invasive approach to evaluate tissue mechanical properties, providing a window into the microstructural integrity of the brain. This retrospective study evaluated brain stiffness as a potential biomarker for vascular brain injury and its role in mediating the impact of vascular dysfunction on cognitive impairment. Seventy-five participants from the Mayo Clinic Study of Aging underwent brain imaging using a 3T MR imager with a spin-echo echo-planar imaging sequence for magnetic resonance elastography and T1- and T2-weighted pulse sequences. This study evaluated the effects of vascular biomarkers (white matter hyperintensities and cardiometabolic condition score) on brain stiffness using voxelwise analysis. Partial correlation analysis explored associations between brain stiffness, white matter hyperintensities, cardiometabolic condition and global cognition. Mediation analysis determined the role of stiffness in mediating the relationship between vascular biomarkers and cognitive performance. Statistical significance was set at P-values < 0.05. Diagnostic accuracy of magnetic resonance elastography stiffness for white matter hyperintensities and cardiometabolic condition was evaluated using receiver operator characteristic curves. Voxelwise linear regression analysis indicated white matter hyperintensities negatively correlate with brain stiffness, specifically in periventricular regions with high white matter hyperintensity levels. A negative association between cardiovascular risk factors and stiffness was also observed across the brain. No significant patterns of stiffness changes were associated with amyloid load. Global stiffness (µ) negatively correlated with both white matter hyperintensities and cardiometabolic condition when all other covariables including amyloid load were controlled. The positive correlation between white matter hyperintensities and cardiometabolic condition weakened and became statistically insignificant when controlling for other covariables. Brain stiffness and global cognition were positively correlated, maintaining statistical significance after adjusting for all covariables. These findings suggest mechanical alterations are associated with cognitive dysfunction and vascular brain injury. Brain stiffness significantly mediated the indirect effects of white matter hyperintensities and cardiometabolic condition on global cognition. Local cerebrovascular diseases (assessed by white matter hyperintensities) and systemic vascular risk factors (assessed by cardiometabolic condition) impact brain stiffness with spatially and statistically distinct effects. Global brain stiffness is a significant mediator between vascular disease measures and cognitive function, highlighting the value of magnetic resonance elastography-based mechanical assessments in understanding this relationship.
Collapse
Affiliation(s)
| | - John Huston
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Richard L Ehman
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Armando Manduca
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
- Department of Physiology and Biomedical Engineering, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | - Clifford R Jack
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Matthew L Senjem
- Department of Information Technology, Mayo Clinic, Rochester, MN 55905, USA
| | | | | |
Collapse
|
9
|
Hansra GK, Jayasena T, Hosoki S, Poljak A, Lam BCP, Rust R, Sagare A, Zlokovic B, Thalamuthu A, Sachdev PS. Fluid biomarkers of the neurovascular unit in cerebrovascular disease and vascular cognitive disorders: A systematic review and meta-analysis. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2024; 6:100216. [PMID: 38510579 PMCID: PMC10951911 DOI: 10.1016/j.cccb.2024.100216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 01/30/2024] [Accepted: 02/16/2024] [Indexed: 03/22/2024]
Abstract
Background The disruption of the neurovascular unit (NVU), which maintains the integrity of the blood brain barrier (BBB), has been identified as a critical mechanism in the development of cerebrovascular and neurodegenerative disorders. However, the understanding of the pathophysiological mechanisms linking NVU dysfunction to the disorders is incomplete, and reliable blood biomarkers to measure NVU dysfunction are yet to be established. This systematic review and meta-analysis aimed to identify biomarkers associated with BBB dysfunction in large vessel disease, small vessel disease (SVD) and vascular cognitive disorders (VCD). Methods A literature search was conducted in PubMed, EMBASE, Scopus and PsychINFO to identify blood biomarkers related to dysfunction of the NVU in disorders with vascular pathologies published until 20 November 2023. Studies that assayed one or more specific markers in human serum or plasma were included. Quality of studies was assessed using the Newcastle-Ottawa Quality Assessment Scale. Effects were pooled and methodological heterogeneity examined using the random effects model. Results A total of 112 studies were included in this review. Where study numbers allowed, biomarkers were analysed using random effect meta-analysis for VCD (1 biomarker; 5 studies) and cerebrovascular disorders, including stroke and SVD (9 biomarkers; 29 studies) while all remaining biomarkers (n = 17 biomarkers; 78 studies) were examined through qualitative analysis. Results of the meta-analysis revealed that cerebrospinal fluid/serum albumin quotient (Q-Alb) reliably differentiates VCD patients from healthy controls (MD = 2.77; 95 % CI = 1.97-3.57; p < 0.0001) while commonly measured biomarkers of endothelial dysfunction (VEGF, VCAM-1, ICAM-1, vWF and E-selectin) and neuronal injury (NfL) were significantly elevated in vascular pathologies. A qualitative assessment of non-meta-analysed biomarkers revealed NSE, NfL, vWF, ICAM-1, VCAM-1, lipocalin-2, MMP-2 and MMP-9 levels to be upregulated in VCD, although these findings were not consistently replicated. Conclusions This review identifies several promising biomarkers of NVU dysfunction which require further validation. A panel of biomarkers representing multiple pathophysiological pathways may offer greater discriminative power in distinguishing possible disease mechanisms of VCD.
Collapse
Affiliation(s)
- Gurpreet Kaur Hansra
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, University of New South Wales, Sydney, Australia
| | - Tharusha Jayasena
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, University of New South Wales, Sydney, Australia
| | - Satoshi Hosoki
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, University of New South Wales, Sydney, Australia
- Department of Neurology, National Cerebral and Cardiovascular Centre, Suita, Japan
| | - Anne Poljak
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, University of New South Wales, Sydney, Australia
- Bioanalytical Mass Spectrometry Facility, Mark Wainwright Analytical Centre, University of New South Wales, NSW, Australia
| | - Ben Chun Pan Lam
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, University of New South Wales, Sydney, Australia
- School of Psychology and Public Health, La Trobe University, Melbourne, Australia
| | - Ruslan Rust
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Abhay Sagare
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Berislav Zlokovic
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Anbupalam Thalamuthu
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, University of New South Wales, Sydney, Australia
| | - Perminder S. Sachdev
- Centre for Healthy Brain Ageing, Discipline of Psychiatry and Mental Health, School of Clinical Medicine, University of New South Wales, Sydney, Australia
| |
Collapse
|
10
|
Claypoole SM, Frank JA, Messmer SJ, Pennypacker KR. CCR3 Expression in Relation to Delayed Microbleeds in a Rat Model of Large Vessel Occlusion. JOURNAL OF EXPERIMENTAL NEUROLOGY 2024; 5:1-8. [PMID: 38332938 PMCID: PMC10852049 DOI: 10.33696/neurol.5.082] [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: 02/10/2024]
Abstract
Thirty percent of ischemic stroke patients develop vascular cognitive impairment and dementia (VCID) within 1 year of stroke onset. The expression of C-C motif chemokine receptor 3 (CCR3) is associated with endothelial dysfunction and memory impairment. CCR3 has been reported to increase after experimental stroke and in human stroke patients. Using an in vivo model of stroke, our study aims to link CCR3 expression with endothelial dysfunction in this rodent stroke model. Methods 5-hour transient Middle Cerebral Artery Occlusion (5t-MCAO) or sham surgery was performed on rats and tissue collected at 3- and 30-days post-stroke. We measured the change in expression of CCR3 and its ligands in the venous blood before and after occlusion in the rat model.Immunohistochemistry was performed on consecutive coronal brain sections using Prussian blue to visualize microbleeds and DAB to visualize CCR3. Images were quantified using HALO. Results Using linear regression, we found that increased expression of CCR3 and its ligands after stroke were positively correlated with infarct volume. CCR3 expression was significantly increased in the ipsilateral hemisphere at 30 days post 5t-MCAO. Prussian blue staining was significantly increased in ipsilateral sections at 30 days post-stroke. Immunostaining for CCR3 was primarily detected in endothelium in areas of Prussian blue staining. Conclusions Our results demonstrate that CCR3 expression is associated with the presence of microbleeds at 30 days but not 3 days post-stroke in the ipsilateral hemisphere, and further supports the link between CCR3 and the endothelial dysfunction that is associated with VCID. CCR3 and its inflammatory pathway is a potential target for reducing endothelial dysfunction after ischemic stroke that may lead to VCID.
Collapse
Affiliation(s)
- Sydney M Claypoole
- Department of Neurology, University of Kentucky, Lexington, KY 40536, USA
| | - Jacqueline A Frank
- Department of Neurosurgery, University of Kentucky, Lexington, KY 40536, USA
| | - Sarah J Messmer
- Department of Neurosurgery, University of Kentucky, Lexington, KY 40536, USA
| | - Keith R Pennypacker
- Department of Neurology, University of Kentucky, Lexington, KY 40536, USA
- Department of Neuroscience, University of Kentucky, Lexington, KY 40536, USA
| |
Collapse
|
11
|
Guha D, Misra V, Yin J, Horiguchi M, Uno H, Gabuzda D. Vascular injury markers associated with cognitive impairment in people with HIV on suppressive antiretroviral therapy. AIDS 2023; 37:2137-2147. [PMID: 37503603 PMCID: PMC10615701 DOI: 10.1097/qad.0000000000003675] [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] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
OBJECTIVE Human immunodeficiency virus (HIV)-associated neurocognitive disorders (HAND) remain prevalent despite viral suppression on antiretroviral therapy (ART). Vascular disease contributes to HAND, but peripheral markers that distinguish vascular cognitive impairment (VCI) from HIV-related etiologies remain unclear. DESIGN Cross-sectional study of vascular injury, inflammation, and central nervous system (CNS) injury markers in relation to HAND. METHODS Vascular injury (VCAM-1, ICAM-1, CRP), inflammation (IFN-γ, IL-1β, IL-6, IL-8, IL-15, IP-10, MCP-1, VEGF-A), and CNS injury (NFL, total Tau, GFAP, YKL-40) markers were measured in plasma and CSF from 248 individuals (143 HIV+ on suppressive ART and 105 HIV- controls). RESULTS Median age was 53 years, median CD4 + cell count, and duration of HIV infection were 505 cells/μl and 16 years, respectively. Vascular injury, inflammation, and CNS injury markers were increased in HIV+ compared with HIV- individuals ( P < 0.05). HAND was associated with increased plasma VCAM-1, ICAM-1, and YKL-40 ( P < 0.01) and vascular disease ( P = 0.004). In contrast, inflammation markers had no significant association with HAND. Vascular injury markers were associated with lower neurocognitive T scores in age-adjusted models ( P < 0.01). Furthermore, plasma VCAM-1 correlated with NFL ( r = 0.29, P = 0.003). Biomarker clustering separated HAND into three clusters: two clusters with high prevalence of vascular disease, elevated VCAM-1 and NFL, and distinctive inflammation profiles (CRP/ICAM-1/YKL-40 or IL-6/IL-8/IL-15/MCP-1), and one cluster with no distinctive biomarker elevations. CONCLUSIONS Vascular injury markers are more closely related to HAND and CNS injury in PWH on suppressive ART than inflammation markers and may help to distinguish relative contributions of VCI to HAND.
Collapse
Affiliation(s)
- Debjani Guha
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Vikas Misra
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jun Yin
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Miki Horiguchi
- Department of Data Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Hajime Uno
- Department of Data Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Dana Gabuzda
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
12
|
Hinman JD, Elahi F, Chong D, Radabaugh H, Ferguson A, Maillard P, Thompson JF, Rosenberg GA, Sagare A, Moghekar A, Lu H, Lee T, Wilcock D, Satizabal CL, Tracy R, Seshadri S, Schwab K, Helmer K, Singh H, Kivisäkk P, Greenberg S, DeCarli C, Kramer J. Placental growth factor as a sensitive biomarker for vascular cognitive impairment. Alzheimers Dement 2023; 19:3519-3527. [PMID: 36815663 PMCID: PMC10440207 DOI: 10.1002/alz.12974] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 11/14/2022] [Accepted: 12/19/2022] [Indexed: 02/24/2023]
Abstract
INTRODUCTION High-performing biomarkers measuring the vascular contributions to cognitive impairment and dementia are lacking. METHODS Using a multi-site observational cohort study design, we examined the diagnostic accuracy of plasma placental growth factor (PlGF) within the MarkVCID Consortium (n = 335; CDR 0-1). Subjects underwent clinical evaluation, cognitive testing, MRI, and blood sampling as defined by Consortium protocols. RESULTS In the prospective population of 335 subjects (72.2 ± 7.8 years of age, 49.3% female), plasma PlGF (pg/mL) shows an ordinal odds ratio (OR) of 1.16 (1.07-1.25; P = .0003) for increasing Fazekas score and ordinal OR of 1.22 (1.14-1.32; P < .0001) for functional cognitive impairment measured by the Clinical Dementia Rating scale. We achieved the primary study outcome of a site-independent association of plasma PlGF (pg/mL) with white matter injury and cognitive impairment in two of three study cohorts. Secondary outcomes using the full MarkVCID cohort demonstrated that plasma PlGF can significantly discriminate individuals with Fazekas ≥ 2 and CDR = 0.5 (area under the curve [AUC] = 0.74) and CDR = 1 (AUC = 0.89) from individuals with CDR = 0. DISCUSSION Plasma PlGF measured by standardized immunoassay functions as a stable, reliable, diagnostic biomarker for cognitive impairment associated with substantial white matter burden.
Collapse
Affiliation(s)
- Jason D. Hinman
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles
- Department of Neurology, West Los Angeles Veterans Association Medical Center, Department of Veterans Affairs
| | - Fanny Elahi
- Memory and Aging Center, Weill Institute for Neuroscience, University of California San Francisco
- Department of Neurology, San Francisco Veterans Association Medical Center, Department of Veterans Affairs
| | - Davis Chong
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles
| | - Hannah Radabaugh
- Department of Neurological Surgery, Weill Institute for Neuroscience, University of California San Francisco
| | - Adam Ferguson
- Department of Neurology, San Francisco Veterans Association Medical Center, Department of Veterans Affairs
- Department of Neurological Surgery, Weill Institute for Neuroscience, University of California San Francisco
| | | | | | | | - Abhay Sagare
- Zilkha Neurogenetic Institute, University of Southern California
| | | | - Hanzhang Lu
- Department of Radiology, Johns Hopkins University
| | - Tiffany Lee
- Sanders-Brown Center on Aging, Department of Physiology, University of Kentucky
| | - Donna Wilcock
- Sanders-Brown Center on Aging, Department of Physiology, University of Kentucky
| | - Claudia L. Satizabal
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, UT Health San Antonio
| | - Russell Tracy
- Department of Pathology & Laboratory Medicine, Larner College of Medicine, University of Vermont
| | - Sudha Seshadri
- Glenn Biggs Institute for Alzheimer’s & Neurodegenerative Diseases, UT Health San Antonio
| | - Kristin Schwab
- Department of Neurology, Massachusetts General Hospital, Harvard University
| | - Karl Helmer
- Department of Neurology, Massachusetts General Hospital, Harvard University
| | - Herpreet Singh
- Department of Neurology, Massachusetts General Hospital, Harvard University
| | - Pia Kivisäkk
- Department of Neurology, Massachusetts General Hospital, Harvard University
| | - Steve Greenberg
- Department of Neurology, Massachusetts General Hospital, Harvard University
| | | | - Joel Kramer
- Memory and Aging Center, Weill Institute for Neuroscience, University of California San Francisco
| |
Collapse
|
13
|
Guha D, Misra V, Yin J, Horiguchi M, Uno H, Gabuzda D. Vascular injury markers associated with cognitive impairment in people with HIV on suppressive antiretroviral therapy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.07.23.23293053. [PMID: 37546734 PMCID: PMC10402231 DOI: 10.1101/2023.07.23.23293053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
Objective Human immunodeficiency virus (HIV)-associated neurocognitive disorders (HAND) remain prevalent despite viral suppression on antiretroviral therapy (ART). Vascular disease contributes to HAND, but peripheral markers that distinguish vascular cognitive impairment (VCI) from HIV-related etiologies remain unclear. Design Cross-sectional study of vascular injury, inflammation, and central nervous system (CNS) injury markers in relation to HAND. Methods Vascular injury (VCAM-1, ICAM-1, CRP), inflammation (IFN-γ, IL-1β, IL-6, IL-8, IL-15, IP-10, MCP-1, VEGF-A), and CNS injury (NFL, total Tau, GFAP, YKL-40) markers were measured in plasma and CSF from 248 individuals (143 HIV+ on suppressive ART and 105 HIV- controls). Results Median age was 53 years, median CD4 count, and duration of HIV infection were 505 cells/μl and 16 years, respectively. Vascular injury, inflammation, and CNS injury markers were increased in HIV+ compared with HIV- individuals (p<0.05). HAND was associated with increased plasma VCAM-1, ICAM-1, and YKL-40 (p<0.01) and vascular disease (p=0.004). In contrast, inflammation markers had no significant association with HAND. Vascular injury markers were associated with lower neurocognitive T scores in age-adjusted models (p<0.01). Furthermore, plasma VCAM-1 correlated with NFL (r=0.29, p=0.003). Biomarker clustering separated HAND into three clusters: two clusters with high prevalence of vascular disease, elevated VCAM-1 and NFL, and distinctive inflammation profiles (CRP/ICAM-1/YKL-40 or IL-6/IL-8/IL-15/MCP-1), and one cluster with no distinctive biomarker elevations. Conclusions Vascular injury markers are more closely related to HAND and CNS injury in PWH on suppressive ART than inflammation markers and may help to distinguish relative contributions of VCI to HAND.
Collapse
Affiliation(s)
- Debjani Guha
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Vikas Misra
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jun Yin
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Miki Horiguchi
- Department of Data Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Hajime Uno
- Department of Data Science, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Dana Gabuzda
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
14
|
Rajeev V, Chai YL, Poh L, Selvaraji S, Fann DY, Jo DG, De Silva TM, Drummond GR, Sobey CG, Arumugam TV, Chen CP, Lai MKP. Chronic cerebral hypoperfusion: a critical feature in unravelling the etiology of vascular cognitive impairment. Acta Neuropathol Commun 2023; 11:93. [PMID: 37309012 PMCID: PMC10259064 DOI: 10.1186/s40478-023-01590-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 05/25/2023] [Indexed: 06/14/2023] Open
Abstract
Vascular cognitive impairment (VCI) describes a wide spectrum of cognitive deficits related to cerebrovascular diseases. Although the loss of blood flow to cortical regions critically involved in cognitive processes must feature as the main driver of VCI, the underlying mechanisms and interactions with related disease processes remain to be fully elucidated. Recent clinical studies of cerebral blood flow measurements have supported the role of chronic cerebral hypoperfusion (CCH) as a major driver of the vascular pathology and clinical manifestations of VCI. Here we review the pathophysiological mechanisms as well as neuropathological changes of CCH. Potential interventional strategies for VCI are also reviewed. A deeper understanding of how CCH can lead to accumulation of VCI-associated pathology could potentially pave the way for early detection and development of disease-modifying therapies, thus allowing preventive interventions instead of symptomatic treatments.
Collapse
Affiliation(s)
- Vismitha Rajeev
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
| | - Yuek Ling Chai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
| | - Luting Poh
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
| | - Sharmelee Selvaraji
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
- Integrative Sciences and Engineering Programme, NUS Graduate School, National University of Singapore, Singapore, Singapore
| | - David Y Fann
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
| | - T Michael De Silva
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Grant R Drummond
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Christopher G Sobey
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Thiruma V Arumugam
- School of Pharmacy, Sungkyunkwan University, Suwon, Republic of Korea
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Christopher P Chen
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore
- NUS Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mitchell K P Lai
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Memory Aging and Cognition Centre, National University Health System, Singapore, Singapore.
- NUS Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
15
|
Gao H, Findeis EL, Culmone L, Powell B, Landschoot-Ward J, Zacharek A, Wu T, Lu M, Chopp M, Venkat P. Early therapeutic effects of an Angiopoietin-1 mimetic peptide in middle-aged rats with vascular dementia. Front Aging Neurosci 2023; 15:1180913. [PMID: 37304071 PMCID: PMC10248134 DOI: 10.3389/fnagi.2023.1180913] [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: 03/06/2023] [Accepted: 05/04/2023] [Indexed: 06/13/2023] Open
Abstract
Background Vascular Dementia (VaD) refers to dementia caused by cerebrovascular disease and/or reduced blood flow to the brain and is the second most common form of dementia after Alzheimer's disease. We previously found that in middle-aged rats subjected to a multiple microinfarction (MMI) model of VaD, treatment with AV-001, a Tie2 receptor agonist, significantly improves short-term memory, long-term memory, as well as improves preference for social novelty compared to control MMI rats. In this study, we tested the early therapeutic effects of AV-001 on inflammation and glymphatic function in rats subjected to VaD. Methods Male, middle-aged Wistar rats (10-12 m), subjected to MMI, were randomly assigned to MMI and MMI + AV-001 treatment groups. A sham group was included as reference group. MMI was induced by injecting 800 ± 200, 70-100 μm sized, cholesterol crystals into the internal carotid artery. Animals were treated with AV-001 (1 μg/Kg, i.p.) once daily starting at 24 h after MMI. At 14 days after MMI, inflammatory factor expression was evaluated in cerebrospinal fluid (CSF) and brain. Immunostaining was used to evaluate white matter integrity, perivascular space (PVS) and perivascular Aquaporin-4 (AQP4) expression in the brain. An additional set of rats were prepared to test glymphatic function. At 14 days after MMI, 50 μL of 1% Tetramethylrhodamine (3 kD) and FITC conjugated dextran (500 kD) at 1:1 ratio were injected into the CSF. Rats (4-6/group/time point) were sacrificed at 30 min, 3 h, and 6 h from the start of tracer infusion, and brain coronal sections were imaged using a Laser scanning confocal microscope to evaluate tracer intensities in the brain. Result Treatment of MMI with AV-001 significantly improves white matter integrity in the corpus callosum at 14 days after MMI. MMI induces significant dilation of the PVS, reduces AQP4 expression and impairs glymphatic function compared to Sham rats. AV-001 treatment significantly reduces PVS, increases perivascular AQP4 expression and improves glymphatic function compared to MMI rats. MMI significantly increases, while AV-001 significantly decreases the expression of inflammatory factors (tumor necrosis factor-α (TNF-α), chemokine ligand 9) and anti-angiogenic factors (endostatin, plasminogen activator inhibitor-1, P-selectin) in CSF. MMI significantly increases, while AV-001 significantly reduces brain tissue expression of endostatin, thrombin, TNF-α, PAI-1, CXCL9, and interleukin-6 (IL-6). Conclusion AV-001 treatment of MMI significantly reduces PVS dilation and increases perivascular AQP4 expression which may contribute to improved glymphatic function compared to MMI rats. AV-001 treatment significantly reduces inflammatory factor expression in the CSF and brain which may contribute to AV-001 treatment induced improvement in white matter integrity and cognitive function.
Collapse
Affiliation(s)
- Huanjia Gao
- Department of Neurology, Henry Ford Health, Detroit, MI, United States
| | | | - Lauren Culmone
- Department of Neurology, Henry Ford Health, Detroit, MI, United States
| | - Brianna Powell
- Department of Neurology, Henry Ford Health, Detroit, MI, United States
| | | | - Alex Zacharek
- Department of Neurology, Henry Ford Health, Detroit, MI, United States
| | - Trueman Wu
- Public Health Sciences, Henry Ford Health, Detroit, MI, United States
| | - Mei Lu
- Public Health Sciences, Henry Ford Health, Detroit, MI, United States
| | - Michael Chopp
- Department of Neurology, Henry Ford Health, Detroit, MI, United States
- Department of Physics, Oakland University, Rochester, MI, United States
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| | - Poornima Venkat
- Department of Neurology, Henry Ford Health, Detroit, MI, United States
- Department of Physiology, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
16
|
Chai YL, Rajeev V, Poh L, Selvaraji S, Hilal S, Chen CP, Jo DG, Koo EH, Arumugam TV, Lai MKP. Chronic cerebral hypoperfusion alters the CypA-EMMPRIN-gelatinase pathway: Implications for vascular dementia. J Cereb Blood Flow Metab 2023; 43:722-735. [PMID: 36537035 PMCID: PMC10108186 DOI: 10.1177/0271678x221146401] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 11/17/2022] [Accepted: 11/17/2022] [Indexed: 03/21/2023]
Abstract
Chronic cerebral hypoperfusion (CCH) is postulated to underlie multiple pathophysiological processes in vascular dementia (VaD), including extracellular matrix dysfunction. While several extracellular matrix proteins, namely cyclophilin A (CypA), extracellular matrix metalloproteinase inducer (EMMPRIN) and gelatinases (matrix metalloproteinases, MMP-2 and -9) have been investigated in acute stroke, their involvement in CCH and VaD remains unclear. In this study, CypA-EMMPRIN-gelatinase proteins were analysed in a clinical cohort of 36 aged, cognitively unimpaired subjects and 48 VaD patients, as well as in a bilateral carotid artery stenosis mouse model of CCH. Lower CypA and higher EMMPRIN levels were found in both VaD serum and CCH mouse brain. Furthermore, gelatinases were differentially altered in CCH mice and VaD patients, with significant MMP-2 increase in CCH brain and serum, whilst serum MMP-9 was elevated in VaD but reduced in CCH, suggesting complex CypA-EMMPRIN-gelatinase regulatory mechanisms. Interestingly, subjects with cortical infarcts had higher serum MMP-2, while white matter hyperintensities, cortical infarcts and lacunes were associated with higher serum MMP-9. Taken together, our data indicate that perturbations of CypA-EMMPRIN signalling may be associated with gelatinase-mediated vascular sequelae, highlighting the potential utility of the CypA-EMMPRIN-gelatinase pathway as clinical biomarkers and therapeutic targets in VaD.
Collapse
Affiliation(s)
- Yuek Ling Chai
- Department of Pharmacology, Yong
Loo Lin School of Medicine, National University of Singapore, Kent Ridge,
Singapore
- Memory, Aging and Cognition Centre,
National University Health System, Kent Ridge, Singapore
| | - Vismitha Rajeev
- Department of Pharmacology, Yong
Loo Lin School of Medicine, National University of Singapore, Kent Ridge,
Singapore
| | - Luting Poh
- Department of Pharmacology, Yong
Loo Lin School of Medicine, National University of Singapore, Kent Ridge,
Singapore
| | - Sharmelee Selvaraji
- Department of Pharmacology, Yong
Loo Lin School of Medicine, National University of Singapore, Kent Ridge,
Singapore
| | - Saima Hilal
- Department of Pharmacology, Yong
Loo Lin School of Medicine, National University of Singapore, Kent Ridge,
Singapore
- Saw Swee Hock School of Public
Health, National University of Singapore, Kent Ridge, Singapore
| | - Christopher P Chen
- Department of Pharmacology, Yong
Loo Lin School of Medicine, National University of Singapore, Kent Ridge,
Singapore
- Memory, Aging and Cognition Centre,
National University Health System, Kent Ridge, Singapore
| | - Dong-Gyu Jo
- School of Pharmacy, Sungkyunkwan
University, Suwon, Republic of Korea
| | - Edward H Koo
- Department of Medicine, National
University of Singapore, Kent Ridge, Singapore
- Graduate School for Integrative
Sciences and Engineering, National University of Singapore, Kent Ridge,
Singapore
- Department of Neurosciences,
University of California San Diego, San Diego, CA, USA
| | - Thiruma V Arumugam
- School of Pharmacy, Sungkyunkwan
University, Suwon, Republic of Korea
- Centre for Cardiovascular Biology
and Disease Research, Department of Microbiology, Anatomy, Physiology and
Pharmacology, School of Agriculture, Biomedicine and Environment, La Trobe
University, Bundoora, VIC, Australia
| | - Mitchell KP Lai
- Department of Pharmacology, Yong
Loo Lin School of Medicine, National University of Singapore, Kent Ridge,
Singapore
- Memory, Aging and Cognition Centre,
National University Health System, Kent Ridge, Singapore
| |
Collapse
|
17
|
Prajjwal P, Marsool MDM, Inban P, Sharma B, Asharaf S, Aleti S, Gadam S, Al Sakini AS, Hadi DD. Vascular dementia subtypes, pathophysiology, genetics, neuroimaging, biomarkers, and treatment updates along with its association with Alzheimer's dementia and diabetes mellitus. Dis Mon 2023; 69:101557. [PMID: 37031059 DOI: 10.1016/j.disamonth.2023.101557] [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: 04/10/2023]
Abstract
Dementia is a chronic progressive cognitive decline illness that results in functional impairment. Vascular dementia (VaD), second only to Alzheimer's disease (AD), is one of the most prevalent forms of dementia in the elderly (aged over 65 years), with a varied presentation and unpredictable disease development caused by cerebrovascular or cardiovascular illness. To get a better understanding of the changes occurring in the brain and to drive therapy efforts, new biomarkers for early and precise diagnosis of AD and VaD are required. In this review, Firstly, we describe the subtypes of vascular dementia, their clinical features, pathogenesis, genetics implemented, and their associated neuroimaging and biomarkers, while describing extensively the recent biomarkers discovered in the literature. Secondly, we describe some of the well-documented and other less-defined risk factors and their association and pathophysiology in relation to vascular dementia. Finally, we follow recent updates in the management of vascular dementia along with its association and differentiation from Alzheimer's disease. The aim of this review is to gather the scattered updates and the most recent changes in blood, CSF, and neuroimaging biomarkers related to the multiple subtypes of vascular dementia along with its association with Alzheimer's dementia and diabetes mellitus.
Collapse
Affiliation(s)
| | | | - Pugazhendi Inban
- Internal Medicine, Government Medical College, Omandurar, Chennai, India
| | | | - Shahnaz Asharaf
- Internal Medicine, Travancore Medical College, Kollam, Kerala, India
| | - Soumya Aleti
- PGY-2, Internal Medicine, Berkshire Medical Center, Pittsfield, MA, USA
| | - Srikanth Gadam
- Internal Medicine, Postdoctoral Research Fellow, Mayo Clinic, USA
| | | | - Dalia Dhia Hadi
- University of Baghdad, Al-Kindy College of Medicine, Baghdad, Iraq
| |
Collapse
|
18
|
Ying C, Kang P, Binkley MM, Ford AL, Chen Y, Hassenstab J, Wang Q, Strain J, Morris JC, Lee JM, Benzinger TLS, An H. Neuroinflammation and amyloid deposition in the progression of mixed Alzheimer and vascular dementia. Neuroimage Clin 2023; 38:103373. [PMID: 36933348 PMCID: PMC10036862 DOI: 10.1016/j.nicl.2023.103373] [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: 10/29/2022] [Revised: 01/18/2023] [Accepted: 03/08/2023] [Indexed: 03/17/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) and vascular contributions to cognitive impairment and dementia (VCID) pathologies coexist in patients with cognitive impairment. Abnormal amyloid beta (Aβ) deposition is the hallmark pathologic biomarker for AD. Neuroinflammation may be a pathophysiological mechanism in both AD and VCID. In this study, we aimed to understand the role of neuroinflammation and Aβ deposition in white matter hyperintensities (WMH) progression and cognitive decline over a decade in patients with mixed AD and VCID pathologies. METHODS Twenty-four elderly participants (median [interquartile range] age 78 [64.8, 83] years old, 14 female) were recruited from the Knight Alzheimer Disease Research Center. 11C-PK11195 standard uptake value ratio (SUVR) and 11C-PiB mean cortical binding potential (MCBP) were used to evaluate neuroinflammation and Aβ deposition in-vivo, respectively. Fluid-attenuated inversion recovery MR images were acquired to obtain baseline WMH volume and its progression over 11.5 years. Composite cognitive scores (global, processing speed and memory) were computed at baseline and follow-up over 7.5 years. Multiple linear regression models evaluated the association between PET biomarkers (11C-PK11195 SUVR and 11C-PiB MCBP) and baseline WMH volume and cognitive function. Moreover, linear mixed-effects models evaluated whether PET biomarkers predicted greater WMH progression or cognitive decline over a decade. RESULTS Fifteen participants (62.5%) had mixed AD (positive PiB) and VCID (at least one vascular risk factor) pathologies. Elevated 11C-PK11195 SUVR, but not 11C-PiB MCBP, was associated with greater baseline WMH volume and predicted greater WMH progression. Elevated 11C-PiB MCBP was associated with baseline memory and global cognition. Elevated 11C-PK11195 SUVR and elevated 11C-PiB MCBP independently predicted greater global cognition and processing speed declines. No association was found between 11C-PK11195 SUVR and 11C-PiB MCBP. CONCLUSIONS Neuroinflammation and Aβ deposition may represent two distinct pathophysiological pathways, and both independently contributed to the progression of cognitive impairment in mixed AD and VCID pathologies. Neuroinflammation, but not Aβ deposition, contributed to WMH volume and progression.
Collapse
Affiliation(s)
- Chunwei Ying
- Department of Biomedical Engineering, Washington University in St. Louis, USA; Mallinckrodt Institute of Radiology, Washington University School of Medicine, USA
| | - Peter Kang
- Department of Neurology, Washington University School of Medicine, USA
| | - Michael M Binkley
- Department of Neurology, Washington University School of Medicine, USA
| | - Andria L Ford
- Department of Neurology, Washington University School of Medicine, USA; Mallinckrodt Institute of Radiology, Washington University School of Medicine, USA
| | - Yasheng Chen
- Department of Neurology, Washington University School of Medicine, USA
| | - Jason Hassenstab
- Department of Neurology, Washington University School of Medicine, USA; Knight Alzheimer Disease Research Center, Washington University School of Medicine, USA
| | - Qing Wang
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, USA; Knight Alzheimer Disease Research Center, Washington University School of Medicine, USA
| | - Jeremy Strain
- Department of Neurology, Washington University School of Medicine, USA
| | - John C Morris
- Knight Alzheimer Disease Research Center, Washington University School of Medicine, USA
| | - Jin-Moo Lee
- Department of Biomedical Engineering, Washington University in St. Louis, USA; Department of Neurology, Washington University School of Medicine, USA; Mallinckrodt Institute of Radiology, Washington University School of Medicine, USA
| | - Tammie L S Benzinger
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, USA; Knight Alzheimer Disease Research Center, Washington University School of Medicine, USA; Department of Neurosurgery, Washington University School of Medicine, USA
| | - Hongyu An
- Department of Biomedical Engineering, Washington University in St. Louis, USA; Department of Neurology, Washington University School of Medicine, USA; Mallinckrodt Institute of Radiology, Washington University School of Medicine, USA.
| |
Collapse
|
19
|
Jamali Q, Akinfala A, Upendram A. Identification of biomarkers for vascular dementia: a literature review. BJPSYCH ADVANCES 2023. [DOI: 10.1192/bja.2022.90] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
Abstract
SUMMARY
Alzheimer's disease and vascular dementia are the two most common types of dementia. It becomes difficult to distinguish between the two, especially when there are no specific genetic causes or vascular changes apparent. The aim of this review was to identify specific biomarkers supporting the diagnosis of vascular dementia by conducting a literature search for systematic reviews and observational studies. We found seven studies meeting our inclusion/exclusion criteria, and from these we identified four specific biomarkers supporting the diagnosis of vascular dementia: high levels of thyroid-stimulating hormone, lipoprotein(a), homocysteine and N-terminal prosomatostatin. However, the studies were small and a well-conducted study with larger populations is recommended to strengthen the evidence base.
Collapse
|
20
|
Gorelick PB. Blood and Cerebrospinal Fluid Biomarkers in Vascular Dementia and Alzheimer's Disease: A Brief Review. Clin Geriatr Med 2023; 39:67-76. [PMID: 36404033 DOI: 10.1016/j.cger.2022.08.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The Maintenance of brain health is a lifelong process whereby potentially deleterious exposures such as cardiovascular risks, amyloid beta, and phosphorylated tau may adversely affect the brain decades before there are clinical manifestations. Thus, the early structural and neuropathological foundation for the development of cognitive impairment and its allied features later in life may provide precursor targets such that interventions may be applied to prevent or slow cognitively impairing processes if the underlying mechanism(s) can be addressed in time.
Collapse
Affiliation(s)
- Philip B Gorelick
- Section of Stroke and Neurocritical Care, Davee Department of Neurology, Northwestern University Feinberg School of Medicine, 625 North Michigan Avenue Suite 1150, Chicago, IL 60611, USA.
| |
Collapse
|
21
|
Initial and ongoing tobacco smoking elicits vascular damage and distinct inflammatory response linked to neurodegeneration. Brain Behav Immun Health 2023; 28:100597. [PMID: 36817509 PMCID: PMC9931921 DOI: 10.1016/j.bbih.2023.100597] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 12/07/2022] [Accepted: 01/21/2023] [Indexed: 01/30/2023] Open
Abstract
Tobacco smoking is strongly linked to vascular damage contributing to the development of hypertension, atherosclerosis, as well as increasing the risk for neurodegeneration. Still, the involvement of the innate immune system in the development of vascular damage upon chronic tobacco use before the onset of clinical symptoms is not fully characterized. Our data provide evidence that a single acute exposure to tobacco elicits the secretion of extracellular vesicles expressing CD105 and CD49e from endothelial cells, granting further recognition of early preclinical biomarkers of vascular damage. Furthermore, we investigated the effects of smoking on the immune system of healthy asymptomatic chronic smokers compared to never-smokers, focusing on the innate immune system. Our data reveal a distinct immune landscape representative for early stages of vascular damage in clinically asymptomatic chronic smokers, before tobacco smoking related diseases develop. These results indicate a dysregulated immuno-vascular axis in chronic tobacco smokers that are otherwise considered as healthy individuals. The distinct alterations are characterized by increased CD36 expression by the blood monocyte subsets, neutrophilia and increased plasma IL-18 and reduced levels of IL-33, IL-10 and IL-8. Additionally, reduced levels of circulating BDNF and elevated sTREM2, which are associated with neurodegeneration, suggest a considerable impact of tobacco smoking on CNS function in clinically healthy individuals. These findings provide profound insight into the initial and ongoing effects of tobacco smoking and the potential vascular damage contributing to neurodegenerative disorders, specifically cerebrovascular dysfunction and dementia.
Collapse
|
22
|
Liu J, Chang D, Cordato D, Lee K, Dixson H, Bensoussan A, Chan DKY. A pilot randomized controlled trial of WeiNaoKang (SaiLuoTong) in treating vascular dementia. Aging Med (Milton) 2022; 5:246-256. [PMID: 36606270 PMCID: PMC9805291 DOI: 10.1002/agm2.12230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/26/2022] [Accepted: 10/31/2022] [Indexed: 11/24/2022] Open
Abstract
Objective WeiNaoKang (or SaiLuoTong) is an herbal formula consisting of ginkgo, ginseng, and saffron. Our objective was to investigate if WeiNaoKang could improve cognitive function and cerebral perfusion in patients suffering from vascular dementia. Methods A 16-week randomized double-blind, placebo-controlled trial was carried out in the setting of a memory disorder clinic at a single center. Patients with vascular dementia diagnosed clinically but supported by imaging and other investigations were invited to participate. The diagnoses were based on the National Institute of Neurological Disorders and Stroke/Association Internationale pour la Recherche et l'Enseignement en Neurosciences (NINDS-AIREN) criteria. An independent blinded assessor evaluated the effects of the formula. Intervention group was compared to the control group. A subgroup of participants was randomly chosen for further evaluation of cerebral perfusion by single photon emission computed tomography scans post-treatment. Results Both groups were comparable in age (mean = 74 ± 7.2 years in the placebo group and 75 ± 7.4 in the intervention group) and in other demographics. Sixty-two participants were included in final analysis. Alzheimer's Disease Assessment Scale - Cognitive Portion (ADAS-cog) was the primary outcome. By week 16, the mean ADAS-cog reduced from 24.48 to 20.30 (mean reduction = 4.18) for those in the treatment group, and from 18.98 to 17.81 (mean reduction = 1.18) for those in the placebo group. The difference in mean reduction of ADAS-cog was -3.00 (95% confidence interval [CI] = -4.910 to -1.100) in favor of the treatment group. Secondary outcomes of activities of daily living and quality of life measures also showed significant difference. In the perfusion scan analysis, the difference in the change in cerebral blood flow (t-scores) pre- and post-treatment between the intervention group (n = 7) and the placebo group (n = 11) was statistically significant (P < 0.001). Conclusion In this randomized, double-blind placebo-controlled trial, we demonstrated significant differences in improvement in cognitive function and activities of daily living. The clinical improvement is corroborated with improvement in cerebral perfusion in a subset of participants.
Collapse
Affiliation(s)
- Junguang Liu
- NICM Health Research InstituteWestern Sydney UniversityWestmeadNew South WalesAustralia
| | - Dennis Chang
- NICM Health Research InstituteWestern Sydney UniversityWestmeadNew South WalesAustralia
| | - Dennis Cordato
- Department of NeurologyLiverpool HospitalLiverpoolNew South WalesAustralia
| | - Kien Lee
- Department of Nuclear Medicine and UltrasoundBankstown‐Lidcombe HospitalBankstownNew South WalesAustralia
| | - Hugh Dixson
- Department of Nuclear Medicine and UltrasoundBankstown‐Lidcombe HospitalBankstownNew South WalesAustralia
| | - Alan Bensoussan
- NICM Health Research InstituteWestern Sydney UniversityWestmeadNew South WalesAustralia
| | - Daniel Kam Yin Chan
- NICM Health Research InstituteWestern Sydney UniversityWestmeadNew South WalesAustralia
- University of New South WalesKensingtonNew South WalesAustralia
- Department of Aged Care & RehabilitationBankstown‐Lidcombe HospitalBankstownNew South WalesAustralia
| |
Collapse
|
23
|
Butler R, Bradford D, Rodgers KE. Analysis of shared underlying mechanism in neurodegenerative disease. Front Aging Neurosci 2022; 14:1006089. [PMID: 36523957 PMCID: PMC9745190 DOI: 10.3389/fnagi.2022.1006089] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 11/08/2022] [Indexed: 08/27/2023] Open
Abstract
In this review, the relationship between bioenergetics, mitochondrial dysfunction, and inflammation will be and how they contribute to neurodegeneration, specifically in Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), and multiple sclerosis (MS) will be reviewed. Long-term changes in mitochondrial function, autophagy dysfunction, and immune activation are commonalities shared across these age-related disorders. Genetic risk factors for these diseases support an autophagy-immune connection in the underlying pathophysiology. Critical areas of deeper evaluation in these bioenergetic processes may lead to potential therapeutics with efficacy across multiple neurodegenerative diseases.
Collapse
Affiliation(s)
| | | | - Kathleen E. Rodgers
- Department of Medical Pharmacology, Center for Innovation in Brain Science, University of Arizona College of Medicine, Tucson, AZ, United States
| |
Collapse
|
24
|
Palm Oil Derived Tocotrienol-Rich Fraction Attenuates Vascular Dementia in Type 2 Diabetic Rats. Int J Mol Sci 2022; 23:ijms232113531. [PMID: 36362316 PMCID: PMC9653761 DOI: 10.3390/ijms232113531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 10/24/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
Vascular dementia (VaD) is a serious global health issue and type 2 diabetes mellitus (T2DM) patients are at higher risk. Palm oil tocotrienol-rich fraction (TRF) exhibits neuroprotective properties; however, its effect on VaD is not reported. Hence, we evaluated TRF effectiveness in T2DM-induced VaD rats. Rats were given a single dose of streptozotocin (STZ) and nicotinamide (NA) to develop T2DM. Seven days later, diabetic rats were given TRF doses of 30, 60, and 120 mg/kg orally for 21 days. The Morris water maze (MWM) test was performed for memory assessment. Biochemical parameters such as blood glucose, plasma homocysteine (HCY) level, acetylcholinesterase (AChE) activity, reduced glutathione (GSH), superoxide dismutase (SOD) level, and histopathological changes in brain hippocampus and immunohistochemistry for platelet-derived growth factor-C (PDGF-C) expression were evaluated. VaD rats had significantly reduced memory, higher plasma HCY, increased AChE activity, and decreased GSH and SOD levels. However, treatment with TRF significantly attenuated the biochemical parameters and prevented memory loss. Moreover, histopathological changes were attenuated and there was increased PDGF-C expression in the hippocampus of VaD rats treated with TRF, indicating neuroprotective action. In conclusion, this research paves the way for future studies and benefits in understanding the potential effects of TRF in VaD rats.
Collapse
|
25
|
Zhou Q, Le M, Yang Y, Wang W, Huang Y, Wang Q, Tian Y, Jiang M, Rao Y, Luo HB, Wu Y. Discovery of novel phosphodiesterase-1 inhibitors for curing vascular dementia: suppression of neuroinflammation by blocking NF-κB transcription regulation and activating cAMP/CREB axis. Acta Pharm Sin B 2022; 13:1180-1191. [PMID: 36970192 PMCID: PMC10031254 DOI: 10.1016/j.apsb.2022.09.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 08/31/2022] [Accepted: 09/20/2022] [Indexed: 11/01/2022] Open
Abstract
Vascular dementia (VaD) is the second commonest type of dementia which lacks of efficient treatments currently. Neuroinflammation as a prominent pathological feature of VaD, is highly involved in the development of VaD. In order to verify the therapeutic potential of PDE1 inhibitors against VaD, the anti-neuroinflammation, memory and cognitive improvement were evaluated in vitro and in vivo by a potent and selective PDE1 inhibitor 4a. Also, the mechanism of 4a in ameliorating neuroinflammation and VaD was systematically explored. Furthermore, to optimize the drug-like properties of 4a, especially for metabolic stability, 15 derivatives were designed and synthesized. As a result, candidate 5f, with a potent IC50 value of 4.5 nmol/L against PDE1C, high selectivity over PDEs, and remarkable metabolic stability, efficiently ameliorated neuron degeneration, cognition and memory impairment in VaD mice model by suppressing NF-κB transcription regulation and activating cAMP/CREB axis. These results further identified PDE1 inhibition could serve as a new therapeutic strategy for treatment of VaD.
Collapse
|
26
|
Zhao K, Zeng L, Cai Z, Liu M, Sun T, Li Z, Liu R. RNA sequencing-based identification of the regulatory mechanism of microRNAs, transcription factors, and corresponding target genes involved in vascular dementia. Front Neurosci 2022; 16:917489. [PMID: 36203804 PMCID: PMC9531238 DOI: 10.3389/fnins.2022.917489] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Vascular dementia (VaD) is the second most common form of dementia with uncertain mechanisms and no effective treatments. microRNAs (miRNAs) and transcription factors (TFs) are considered regulatory factors of genes involved in many diseases. Therefore, this work investigated the aberrantly expressed miRNAs, TFs, corresponding target genes, and their co-regulatory networks in the cortex of rats with bilateral common carotid artery occlusion (2VO) to uncover the potential mechanism and biomarkers of VaD. Differentially expressed genes (DEGs), miRNAs (DEMs), and TFs (DETFs) were identified using RNA sequencing, and their interaction networks were constructed using Cytoscape. The results showed that rats with 2VO had declined cognitive abilities and neuronal loss in the cortex than sham rats. DEGs, DEMs, and DETFs were discriminated between rats with 2VO and sham rats in the cortex, as shown by the 13 aberrantly expressed miRNAs, 805 mRNAs, and 63 TFs. The miRNA-TF-target gene network was constructed, showing 523 nodes and 7237 edges. Five miRNAs (miR-5132-5p, miR-764-3p, miR-223-3p, miR-145-5p, and miR-122-5p), ten TFs (Mxi1, Nfatc4, Rxrg, Zfp523, Foxj2, Nkx6-1, Klf4, Klf5, Csrnp1, and Prdm6), and seven target genes (Serpine1, Nedd4l, Pxn, Col1a1, Plec, Trip12, and Tpm1) were chosen as the significant nodes to construct feed-forward loops (FFLs). Gene Ontology and pathway enrichment analysis revealed that these miRNA and TF-associated genes are mostly involved in the PI3K/Akt, neuroactive ligand–receptor interaction, calcium signaling, and Wnt signaling pathways, along with central locations around the cell membrane. They exert functions such as growth factor binding, integrin binding, and extracellular matrix structural constituent, with representative biological processes like vasculature development, cell–substrate adhesion, cellular response to growth factor stimulus, and synaptic transmission. Furthermore, the expression of three miRNAs (miR-145-5p, miR-122-5p, and miR-5132-5p), six TFs (Csrnp1, Klf4, Nfatc4, Rxrg, Foxj2, and Klf5), and five mRNAs (Serpine1, Plec, Nedd4l, Trip12, and Tpm1) were significantly changed in rats with VaD, in line with the outcome of RNA sequencing. In the potential FFL, miR-145-5p directly bound Csrnp1 and decreased its mRNA expression. These results might help the understanding of the underlying regulatory mechanisms of miRNA-TF-genes, providing potential therapeutic targets in VaD.
Collapse
|
27
|
Bioenergetic and vascular predictors of potential super-ager and cognitive decline trajectories-a UK Biobank Random Forest classification study. GeroScience 2022; 45:491-505. [PMID: 36104610 PMCID: PMC9886787 DOI: 10.1007/s11357-022-00657-6] [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: 12/20/2021] [Accepted: 09/01/2022] [Indexed: 02/03/2023] Open
Abstract
Aging has often been characterized by progressive cognitive decline in memory and especially executive function. Yet some adults, aged 80 years or older, are "super-agers" that exhibit cognitive performance like younger adults. It is unknown if there are adults in mid-life with similar superior cognitive performance ("positive-aging") versus cognitive decline over time and if there are blood biomarkers that can distinguish between these groups. Among 1303 participants in UK Biobank, latent growth curve models classified participants into different cognitive groups based on longitudinal fluid intelligence (FI) scores over 7-9 years. Random Forest (RF) classification was then used to predict cognitive trajectory types using longitudinal predictors including demographic, vascular, bioenergetic, and immune factors. Feature ranking importance and performance metrics of the model were reported. Despite model complexity, we achieved a precision of 77% when determining who would be in the "positive-aging" group (n = 563) vs. cognitive decline group (n = 380). Among the top fifteen features, an equal number were related to either vascular health or cellular bioenergetics but not demographics like age, sex, or socioeconomic status. Sensitivity analyses showed worse model results when combining a cognitive maintainer group (n = 360) with the positive-aging or cognitive decline group. Our results suggest that optimal cognitive aging may not be related to age per se but biological factors that may be amenable to lifestyle or pharmacological changes.
Collapse
|
28
|
Bailey J, Coucha M, Bolduc DR, Burnett FN, Barrett AC, Ghaly M, Abdelsaid M. GLP-1 receptor nitration contributes to loss of brain pericyte function in a mouse model of diabetes. Diabetologia 2022; 65:1541-1554. [PMID: 35687178 DOI: 10.1007/s00125-022-05730-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 03/17/2022] [Indexed: 12/21/2022]
Abstract
AIMS/HYPOTHESIS We have previously shown that diabetes causes pericyte dysfunction, leading to loss of vascular integrity and vascular cognitive impairment and dementia (VCID). Glucagon-like peptide-1 (GLP-1) receptor agonists (GLP-1 RAs), used in managing type 2 diabetes mellitus, improve the cognitive function of diabetic individuals beyond glycaemic control, yet the mechanism is not fully understood. In the present study, we hypothesise that GLP-1 RAs improve VCID by preventing diabetes-induced pericyte dysfunction. METHODS Mice with streptozotocin-induced diabetes and non-diabetic control mice received either saline (NaCl 154 mmol/l) or exendin-4, a GLP-1 RA, through an osmotic pump over 28 days. Vascular integrity was assessed by measuring cerebrovascular neovascularisation indices (vascular density, tortuosity and branching density). Cognitive function was evaluated with Barnes maze and Morris water maze. Human brain microvascular pericytes (HBMPCs), were grown in high glucose (25 mmol/l) and sodium palmitate (200 μmol/l) to mimic diabetic conditions. HBMPCs were treated with/without exendin-4 and assessed for nitrative and oxidative stress, and angiogenic and blood-brain barrier functions. RESULTS Diabetic mice treated with exendin-4 showed a significant reduction in all cerebral pathological neovascularisation indices and an improved blood-brain barrier (p<0.05). The vascular protective effects were accompanied by significant improvement in the learning and memory functions of diabetic mice compared with control mice (p<0.05). Our results showed that HBMPCs expressed the GLP-1 receptor. Diabetes increased GLP-1 receptor expression and receptor nitration in HBMPCs. Stimulation of HBMPCs with exendin-4 under diabetic conditions decreased diabetes-induced vascular inflammation and oxidative stress, and restored pericyte function (p<0.05). CONCLUSIONS/INTERPRETATION This study provides novel evidence that brain pericytes express the GLP-1 receptor, which is nitrated under diabetic conditions. GLP-1 receptor activation improves brain pericyte function resulting in restoration of vascular integrity and BBB functions in diabetes. Furthermore, the GLP-1 RA exendin-4 alleviates diabetes-induced cognitive impairment in mice. Restoration of pericyte function in diabetes represents a novel therapeutic target for diabetes-induced cerebrovascular microangiopathy and VCID.
Collapse
Affiliation(s)
- Joseph Bailey
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, GA, USA
| | - Maha Coucha
- Department of Pharmaceutical Sciences, School of Pharmacy, South University, Savannah, GA, USA
| | - Deanna R Bolduc
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, GA, USA
| | - Faith N Burnett
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, GA, USA
| | - Amy C Barrett
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, GA, USA
| | - Mark Ghaly
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, GA, USA
| | - Mohammed Abdelsaid
- Department of Biomedical Sciences, School of Medicine, Mercer University, Savannah, GA, USA.
| |
Collapse
|
29
|
Guo W, Shi J. White matter hyperintensities volume and cognition: A meta-analysis. Front Aging Neurosci 2022; 14:949763. [PMID: 36118701 PMCID: PMC9476945 DOI: 10.3389/fnagi.2022.949763] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Background Cerebral small vessel disease (CSVD) is prevalent in the elderly and leads to an increased risk of cognitive impairment and dementia. The volume of white matter hyperintensities (WMHs) increases with age, which affects cognition. Objective To explore the relationship between WMH volume and cognitive decline in patients with CSVD. Methods We performed a systematic search of PubMed, Embase, and the Web of Science databases from their respective creation dates to the 5 May 2022 to identify all the clinical studies on either mild cognitive impairment (MCI) or dementia in regards to WMH volume in CSVD. Results White matter hyperintensities was associated with the risk of both the MCI and dementia, with a 35% increased risk [relative risk (RR) = 1.35; (95% CI: 1.01-1.81)] of progression from cognitively unimpaired (CU) to MCI (six studies, n = 2,278) and a 49% increased risk [RR = 1.49; (95% CI: 1.21-1.84)] of progression to dementia (six studies, n = 6,330). In a subgroup analysis, a follow-up period of over 5 years increased the risk of MCI by 40% [RR = 1.40; (95% CI: 1.07-1.82)] and dementia by 48% [RR = 1.48; (95% CI: 1.15-1.92)]. Conclusion White matter hyperintensities was found to be substantially correlated with the risk of cognitive impairment. Furthermore, cognitive decline was found to be a chronic process, such that WMH predicted the rate of cognitive decline in CSVD beyond 5 years. The cognitive decline observed in patients with WMH may, therefore, be minimized by early intervention.
Collapse
Affiliation(s)
| | - Jing Shi
- The 3rd Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
30
|
Zhang DD, Ou YN, Fu Y, Wang ZB, Huang LY, Tan L, Yu JT. Risk of Dementia in Cancer Survivors: A Meta-Analysis of Population-Based Cohort Studies. J Alzheimers Dis 2022; 89:367-380. [PMID: 35871349 DOI: 10.3233/jad-220436] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: A negative association between cancer and Alzheimer’s disease (AD) was revealed. Objective: We aimed to further explore the dementia risk among cancer survivors and then among cancer survivors who received cancer treatment in subsequent subgroup analyses. Methods: Databases of PubMed, Embase, and Cochrane Library were systematically searched from inception to April 1, 2021, following PRISMA and MOOSE guidelines. Relative risks (RR) of dementia were pooled by a random-effects model stratifying the data by potential confounding factors to explore the heterogeneity. This study is registered with PROSPERO, number CRD42021250654. Results: A total of 36 studies were included in this meta-analysis, of which 16 studies were about the risk of dementia in cancer survivors, and 20 studies were about the risk of dementia in survivors who accepted cancer treatment. The pooled RR reached 0.89 ([95% CI = 0.82–0.97], I2 = 97.9%) for dementia and 0.89 ([0.83–0.95], I2 = 92.6%) for AD in cancer survivors compared with non-cancer controls. Notably, both dementia risk and AD risk significantly decreased in survivors of colon, leukemia, small intestine, and thyroid cancers (RR ranged from 0.64 to 0.92). Furthermore, prostate cancer patients treated with androgen deprivation therapy exhibited a significantly increased risk of dementia (RR:1.18 [1.09–1.27], I2 = 89.5%) and AD (RR:1.17 [1.08–1.25], I2 = 81.3%), with evidence of between-study heterogeneity. Conclusion: Currently, available evidence suggests that the risk of dementia among cancer survivors is decreased. However, large-scale prospective cohort studies are warranted to further prove the association.
Collapse
Affiliation(s)
- Dan-Dan Zhang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Ya-Nan Ou
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Yan Fu
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Zhi-Bo Wang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Liang-Yu Huang
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Lan Tan
- Department of Neurology, Qingdao Municipal Hospital, Qingdao University, Qingdao, China
| | - Jin-Tai Yu
- Department of Neurology and Institute of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
31
|
Chen RF, Braidy N, Xu YH, Tan S, Chan DKY. Macrophage- and Microglia-Related Chemokines Are Associated with Small Vessel (White Matter) Vascular Dementia: A Case-Control Study. Dement Geriatr Cogn Disord 2022; 50:454-459. [PMID: 34775383 DOI: 10.1159/000519885] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 09/25/2021] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Little is known about the role of inflammation in the process of small vessel vascular dementia (VaD). Recently, the notion that small vessel VaD is caused solely by vascular pathology has been challenged by new evidence of concomitant breakdown of the blood-brain barrier and dysregulation of neuroinflammation in the white matter. METHODS We examined selected inflammatory cytokines and chemokines in the plasma from patients with small vessel VaD (n = 41) and from age-matched controls (n = 131) using multiplex bead-based assays. Participants were recruited from a memory disorder clinic and from a hospital or community. RESULTS When compared to controls, patients with small vessel VaD had a highly significant increase in the plasma interferon-γ-inducible protein 10 (IP-10) level (p < 0.0001) and a highly significant decrease in plasma macrophage inflammatory protein 1-beta (MIP-1β) level (p < 0.0001). We also observed a significant increase in patients' levels of interleukin-10 (IL-10) (p = 0.022) as well as decreases in interleukin-8 (IL-8) (p = 0.004) and interleukin-7 (IL-7) (p = 0.011) when compared to age-matched controls. CONCLUSION Both IP-10 and MIP-1β are macrophage-related chemokines. The significant differences between cases and controls suggest a potential role for macrophages in small vessel VaD neuroinflammation. Although it remains unclear whether there is a causal effect of their alteration for small vessel VaD, a better understanding of these molecules in the pathogenesis of small vessel VaD may lead to improved diagnosis and future treatment outcomes against this disease.
Collapse
Affiliation(s)
- Ren Fen Chen
- Central Sydney Immunology Laboratory at Royal Prince Alfred Hospital, NSW Health Pathology, Sydney, New South Wales, Australia
| | - Nady Braidy
- Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia
| | - Ying Hua Xu
- Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia.,Aged Care Department, Bankstown-Lidcombe Hospital, Bankstown, New South Wales, Australia
| | - Sarah Tan
- Central Sydney Immunology Laboratory at Royal Prince Alfred Hospital, NSW Health Pathology, Sydney, New South Wales, Australia
| | - Daniel Kam Yin Chan
- Faculty of Medicine, University of New South Wales, Sydney, New South Wales, Australia.,Faculty of Medicine, Western Sydney University, Penrith, New South Wales, Australia.,Aged Care Department, Bankstown-Lidcombe Hospital, Bankstown, New South Wales, Australia
| |
Collapse
|
32
|
Kaur M, Sharma S. Molecular mechanisms of cognitive impairment associated with stroke. Metab Brain Dis 2022; 37:279-287. [PMID: 35029798 DOI: 10.1007/s11011-022-00901-0] [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: 06/09/2021] [Accepted: 01/03/2022] [Indexed: 10/19/2022]
Abstract
Stroke is the second leading cause of death after coronary heart disease in developed countries and is the greatest cause of disability and cognitive impairment. Risk factors for cognitive impairment and dementia after stroke are multifactorial including older age, family history, hypertension, arterial fibrillation, diabetes, genetic variants, low educational status, vascular comorbidities, prior transient ischaemic attack or recurrent stroke, depressive illness duration of a stroke, location, volume, intensity, and degree of neuronal degeneration, location and size of infarction after stroke, time interval after stroke other cerebral dysfunctions. The pathophysiology of stroke associated cognitive impairment is complex and recent molecular, cellular, and animal models studies have revealed that multiple cellular changes have been implicated, including altered redox state, mitochondrial dysfunction, disruption of the blood-brain barrier, perivascular spacing, glymphatic system impairment, microglia activation and amyloid-β deposition in the parenchyma of the brain. These studies have also evidenced the involvement of various transcription factors, intracellular adhesion molecules, and endogenous growth factors in the pathogenesis of cognitive impairment associated with stroke and providing scope for developing therapeutic strategies for treatment. This review summarizes the latest research findings on molecular mechanisms involved in cognitive impairment associated with stroke.
Collapse
Affiliation(s)
- Mandeep Kaur
- Department of Pharmacology, School of Pharmaceutical Sciences, CT University, Ludhiana, Punjab, India
| | - Saurabh Sharma
- School of Pharmaceutical Sciences, CT University, Ludhiana, Punjab, India.
| |
Collapse
|
33
|
Morozova A, Zorkina Y, Abramova O, Pavlova O, Pavlov K, Soloveva K, Volkova M, Alekseeva P, Andryshchenko A, Kostyuk G, Gurina O, Chekhonin V. Neurobiological Highlights of Cognitive Impairment in Psychiatric Disorders. Int J Mol Sci 2022; 23:1217. [PMID: 35163141 PMCID: PMC8835608 DOI: 10.3390/ijms23031217] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/18/2022] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
This review is focused on several psychiatric disorders in which cognitive impairment is a major component of the disease, influencing life quality. There are plenty of data proving that cognitive impairment accompanies and even underlies some psychiatric disorders. In addition, sources provide information on the biological background of cognitive problems associated with mental illness. This scientific review aims to summarize the current knowledge about neurobiological mechanisms of cognitive impairment in people with schizophrenia, depression, mild cognitive impairment and dementia (including Alzheimer's disease).The review provides data about the prevalence of cognitive impairment in people with mental illness and associated biological markers.
Collapse
Affiliation(s)
- Anna Morozova
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, 117152 Moscow, Russia; (A.M.); (O.A.); (K.S.); (M.V.); (P.A.); (A.A.); (G.K.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia; (O.P.); (K.P.); (O.G.); (V.C.)
| | - Yana Zorkina
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, 117152 Moscow, Russia; (A.M.); (O.A.); (K.S.); (M.V.); (P.A.); (A.A.); (G.K.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia; (O.P.); (K.P.); (O.G.); (V.C.)
| | - Olga Abramova
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, 117152 Moscow, Russia; (A.M.); (O.A.); (K.S.); (M.V.); (P.A.); (A.A.); (G.K.)
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia; (O.P.); (K.P.); (O.G.); (V.C.)
| | - Olga Pavlova
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia; (O.P.); (K.P.); (O.G.); (V.C.)
| | - Konstantin Pavlov
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia; (O.P.); (K.P.); (O.G.); (V.C.)
| | - Kristina Soloveva
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, 117152 Moscow, Russia; (A.M.); (O.A.); (K.S.); (M.V.); (P.A.); (A.A.); (G.K.)
| | - Maria Volkova
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, 117152 Moscow, Russia; (A.M.); (O.A.); (K.S.); (M.V.); (P.A.); (A.A.); (G.K.)
| | - Polina Alekseeva
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, 117152 Moscow, Russia; (A.M.); (O.A.); (K.S.); (M.V.); (P.A.); (A.A.); (G.K.)
| | - Alisa Andryshchenko
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, 117152 Moscow, Russia; (A.M.); (O.A.); (K.S.); (M.V.); (P.A.); (A.A.); (G.K.)
| | - Georgiy Kostyuk
- Mental-Health Clinic No. 1 Named after N.A. Alekseev, 117152 Moscow, Russia; (A.M.); (O.A.); (K.S.); (M.V.); (P.A.); (A.A.); (G.K.)
| | - Olga Gurina
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia; (O.P.); (K.P.); (O.G.); (V.C.)
| | - Vladimir Chekhonin
- Department of Basic and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology, 119034 Moscow, Russia; (O.P.); (K.P.); (O.G.); (V.C.)
- Department of Medical Nanobiotechnology, Pirogov Russian National Research Medical University, 117997 Moscow, Russia
| |
Collapse
|
34
|
Panes JD, Wendt A, Ramirez-Molina O, Castro PA, Fuentealba J. Deciphering the role of PGC-1α in neurological disorders: from mitochondrial dysfunction to synaptic failure. Neural Regen Res 2022; 17:237-245. [PMID: 34269182 PMCID: PMC8463972 DOI: 10.4103/1673-5374.317957] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
The onset and mechanisms underlying neurodegenerative diseases remain uncertain. The main features of neurodegenerative diseases have been related with cellular and molecular events like neuronal loss, mitochondrial dysfunction and aberrant accumulation of misfolded proteins or peptides in specific areas of the brain. The most prevalent neurodegenerative diseases belonging to age-related pathologies are Alzheimer's disease, Huntington's disease, Parkinson's disease and amyotrophic lateral sclerosis. Interestingly, mitochondrial dysfunction has been observed to occur during the early onset of several neuropathological events associated to neurodegenerative diseases. The master regulator of mitochondrial quality control and energetic metabolism is the transcriptional coactivator peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α). Additionally, it has been observed that PGC-1α appears to be a key factor in maintaining neuronal survival and synaptic transmission. In fact, PGC-1α downregulation in different brain areas (hippocampus, substantia nigra, cortex, striatum and spinal cord) that occurs in function of neurological damage including oxidative stress, neuronal loss, and motor disorders has been seen in several animal and cellular models of neurodegenerative diseases. Current evidence indicates that PGC-1α upregulation may serve as a potent therapeutic approach against development and progression of neuronal damage. Remarkably, increasing evidence shows that PGC-1α deficient mice have neurodegenerative diseases-like features, as well as neurological abnormalities. Finally, we discuss recent studies showing novel specific PGC-1α isoforms in the central nervous system that appear to exert a key role in the age of onset of neurodegenerative diseases and have a neuroprotective function in the central nervous system, thus opening a new molecular strategy for treatment of neurodegenerative diseases. The purpose of this review is to provide an up-to-date overview of the PGC-1α role in the physiopathology of neurodegenerative diseases, as well as establish the importance of PGC-1α function in synaptic transmission and neuronal survival.
Collapse
Affiliation(s)
- Jessica D Panes
- Laboratorio de Screening de Compuestos Neuroactivos (LSCN), Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Aline Wendt
- Laboratorio de Screening de Compuestos Neuroactivos (LSCN), Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Oscar Ramirez-Molina
- Laboratorio de Screening de Compuestos Neuroactivos (LSCN), Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Patricio A Castro
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Jorge Fuentealba
- Laboratorio de Screening de Compuestos Neuroactivos (LSCN), Departamento de Fisiología; Centro de Investigaciones Avanzadas en Biomedicina (CIAB-UdeC), Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| |
Collapse
|
35
|
Hoyer-Kimura C, Konhilas JP, Mansour HM, Polt R, Doyle KP, Billheimer D, Hay M. Neurofilament light: a possible prognostic biomarker for treatment of vascular contributions to cognitive impairment and dementia. J Neuroinflammation 2021; 18:236. [PMID: 34654436 PMCID: PMC8520282 DOI: 10.1186/s12974-021-02281-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 09/20/2021] [Indexed: 12/29/2022] Open
Abstract
Background Decreased cerebral blood flow and systemic inflammation during heart failure (HF) increase the risk for vascular contributions to cognitive impairment and dementia (VCID) and Alzheimer disease-related dementias (ADRD). We previously demonstrated that PNA5, a novel glycosylated angiotensin 1–7 (Ang-(1–7)) Mas receptor (MasR) agonist peptide, is an effective therapy to rescue cognitive impairment in our preclinical model of VCID. Neurofilament light (NfL) protein concentration is correlated with cognitive impairment and elevated in neurodegenerative diseases, hypoxic brain injury, and cardiac disease. The goal of the present study was to determine (1) if treatment with Ang-(1–7)/MasR agonists can rescue cognitive impairment and decrease VCID-induced increases in NfL levels as compared to HF-saline treated mice and, (2) if NfL levels correlate with measures of cognitive function and brain cytokines in our VCID model. Methods VCID was induced in C57BL/6 male mice via myocardial infarction (MI). At 5 weeks post-MI, mice were treated with daily subcutaneous injections for 24 days, 5 weeks after MI, with PNA5 or angiotensin 1–7 (500 microg/kg/day or 50 microg/kg/day) or saline (n = 15/group). Following the 24-day treatment protocol, cognitive function was assessed using the Novel Object Recognition (NOR) test. Cardiac function was measured by echocardiography and plasma concentrations of NfL were quantified using a Quanterix Simoa assay. Brain and circulating cytokine levels were determined with a MILLIPLEX MAP Mouse High Sensitivity Multiplex Immunoassay. Treatment groups were compared via ANOVA, significance was set at p < 0.05. Results Treatment with Ang-(1–7)/MasR agonists reversed VCID-induced cognitive impairment and significantly decreased NfL levels in our mouse model of VCID as compared to HF-saline treated mice. Further, NfL levels were significantly negatively correlated with cognitive scores and the concentrations of multiple pleiotropic cytokines in the brain. Conclusions These data show that treatment with Ang-(1–7)/MasR agonists rescues cognitive impairment and decreases plasma NfL relative to HF-saline-treated animals in our VCID mouse model. Further, levels of NfL are significantly negatively correlated with cognitive function and with several brain cytokine concentrations. Based on these preclinical findings, we propose that circulating NfL might be a candidate for a prognostic biomarker for VCID and may also serve as a pharmacodynamic/response biomarker for therapeutic target engagement.
Collapse
Affiliation(s)
| | - John P Konhilas
- Department of Physiology, The University of Arizona, Tucson, AZ, USA.,Department of Nutritional Sciences, The University of Arizona, Tucson, AZ, USA.,Department of Biomedical Engineering, The University of Arizona, Tucson, AZ, USA.,Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, USA
| | - Heidi M Mansour
- Department of Pharmacy, Skaggs Pharmaceutical Sciences Center, The University of Arizona, Tucson, AZ, USA.,Department of Medicine, Division of Translational and Regenerative Medicine, The University of Arizona, Tucson, AZ, USA
| | - Robin Polt
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, AZ, USA
| | - Kristian P Doyle
- Department of Immunobiology, The University of Arizona, Tucson, AZ, USA
| | - Dean Billheimer
- Department of Epidemiology and Biostatistics, The University of Arizona, Tucson, AZ, USA
| | - Meredith Hay
- Department of Physiology, The University of Arizona, Tucson, AZ, USA.,Department of Neurology, The University of Arizona, Tucson, AZ, USA.,Evelyn F. McKnight Brain Institute, The University of Arizona, Tucson, AZ, USA.,ProNeurogen, Inc, The University of Arizona, Tucson, AZ, USA
| |
Collapse
|
36
|
Hambali A, Kumar J, Hashim NFM, Maniam S, Mehat MZ, Cheema MS, Mustapha M, Adenan MI, Stanslas J, Hamid HA. Hypoxia-Induced Neuroinflammation in Alzheimer's Disease: Potential Neuroprotective Effects of Centella asiatica. Front Physiol 2021; 12:712317. [PMID: 34721056 PMCID: PMC8551388 DOI: 10.3389/fphys.2021.712317] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 09/13/2021] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that is characterised by the presence of extracellular beta-amyloid fibrillary plaques and intraneuronal neurofibrillary tau tangles in the brain. Recurring failures of drug candidates targeting these pathways have prompted research in AD multifactorial pathogenesis, including the role of neuroinflammation. Triggered by various factors, such as hypoxia, neuroinflammation is strongly linked to AD susceptibility and/or progression to dementia. Chronic hypoxia induces neuroinflammation by activating microglia, the resident immune cells in the brain, along with an increased in reactive oxygen species and pro-inflammatory cytokines, features that are common to many degenerative central nervous system (CNS) disorders. Hence, interests are emerging on therapeutic agents and plant derivatives for AD that target the hypoxia-neuroinflammation pathway. Centella asiatica is one of the natural products reported to show neuroprotective effects in various models of CNS diseases. Here, we review the complex hypoxia-induced neuroinflammation in the pathogenesis of AD and the potential application of Centella asiatica as a therapeutic agent in AD or dementia.
Collapse
Affiliation(s)
- Aqilah Hambali
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Jaya Kumar
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Malaysia
| | - Nur Fariesha Md Hashim
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Sandra Maniam
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Muhammad Zulfadli Mehat
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Manraj Singh Cheema
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Muzaimi Mustapha
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Malaysia
| | | | - Johnson Stanslas
- Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| | - Hafizah Abdul Hamid
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Malaysia
| |
Collapse
|
37
|
Caruso G, Torrisi SA, Mogavero MP, Currenti W, Castellano S, Godos J, Ferri R, Galvano F, Leggio GM, Grosso G, Caraci F. Polyphenols and neuroprotection: Therapeutic implications for cognitive decline. Pharmacol Ther 2021; 232:108013. [PMID: 34624428 DOI: 10.1016/j.pharmthera.2021.108013] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/07/2021] [Accepted: 09/28/2021] [Indexed: 02/09/2023]
Abstract
Dietary polyphenols have been the focus of major interest for their potential benefits on human health. Several preclinical studies have been conducted to provide a rationale for their potential use as therapeutic agents in preventing or ameliorating cognitive decline. However, results from human studies are scarce and poorly documented. The aim of this review was to discuss the potential mechanisms involved in age-related cognitive decline or early stage cognitive impairment and current evidence from clinical human studies conducted on polyphenols and the aforementioned outcomes. The evidence published so far is encouraging but contrasting findings are to be taken into account. Most studies on anthocyanins showed a consistent positive effect on various cognitive aspects related to aging or early stages of cognitive impairment. Studies on cocoa flavanols, resveratrol, and isoflavones provided substantial contrasting results and further research is needed to clarify the therapeutic potential of these compounds. Results from other studies on quercetin, green tea flavanols, hydroxycinnamic acids (such as chlorogenic acid), curcumin, and olive oil tyrosol and derivatives are rather promising but still too few to provide any real conclusions. Future translational studies are needed to address issues related to dosage, optimal formulations to improve bioavailability, as well as better control for the overall diet, and correct target population.
Collapse
Affiliation(s)
- Giuseppe Caruso
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| | - Sebastiano A Torrisi
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Maria Paola Mogavero
- Istituti Clinici Scientifici Maugeri, IRCCS, Scientific Institute of Pavia, Pavia, Italy
| | - Walter Currenti
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Sabrina Castellano
- Department of Educational Sciences, University of Catania, Catania, Italy
| | - Justyna Godos
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | | | - Fabio Galvano
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Gian Marco Leggio
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Giuseppe Grosso
- Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy.
| | - Filippo Caraci
- Department of Drug and Health Sciences, University of Catania, Catania, Italy; Oasi Research Institute - IRCCS, Troina, Italy
| |
Collapse
|
38
|
Minta K, Brinkmalm G, Portelius E, Johansson P, Svensson J, Kettunen P, Wallin A, Zetterberg H, Blennow K, Andreasson U. Brevican and Neurocan Peptides as Potential Cerebrospinal Fluid Biomarkers for Differentiation Between Vascular Dementia and Alzheimer's Disease. J Alzheimers Dis 2021; 79:729-741. [PMID: 33337373 DOI: 10.3233/jad-201039] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Brevican and neurocan are central nervous system-specific extracellular matrix proteoglycans. They are degraded by extracellular enzymes, such as metalloproteinases. However, their degradation profile is largely unexplored in cerebrospinal fluid (CSF). OBJECTIVE The study aim was to quantify proteolytic peptides derived from brevican and neurocan in human CSF of patients with Alzheimer's disease (AD) and vascular dementia (VaD) compared with controls. METHODS The first cohort consisted of 75 individuals including 25 patients with AD, 7 with mild cognitive impairment (MCI) diagnosed with AD upon follow-up, 10 patients with VaD or MCI diagnosed with VaD upon follow-up, and 33 healthy controls and cognitively stable MCI patients. In the second cohort, 31 individuals were included (5 AD patients, 14 VaD patients and 12 healthy controls). Twenty proteolytic peptides derived from brevican (n = 9) and neurocan (n = 11) were quantified using high-resolution parallel reaction monitoring mass spectrometry. RESULTS In the first cohort, the majority of CSF concentrations of brevican and neurocan peptides were significantly decreased inVaDas compared withADpatients (AUC = 0.83.0.93, p≤0.05) and as compared with the control group (AUC = 0.79.0.87, p ≤ 0.05). In the second cohort, CSF concentrations of two brevican peptides (B87, B156) were significantly decreased in VaD compared with AD (AUC = 0.86.0.91, p ≤ 0.05) and to controls (AUC = 0.80.0.82, p ≤ 0.05), while other brevican and neurocan peptides showed a clear trend to be decreased in VaD compared with AD (AUC = 0.64.80, p > 0.05). No peptides differed between AD and controls. CONCLUSION Brevican and neurocan peptides are potential diagnostic biomarkers for VaD, with ability to separate VaD from AD.
Collapse
Affiliation(s)
- Karolina Minta
- Department of Psychiatry and Neurochemistry,Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Gunnar Brinkmalm
- Department of Psychiatry and Neurochemistry,Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Erik Portelius
- Department of Psychiatry and Neurochemistry,Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Per Johansson
- Department of Clinical Sciences Helsingborg, Lund University, Sweden.,Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Johan Svensson
- Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, the Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Department of Endocrinology, Skaraborg Central Hospital, Skövde, Sweden
| | - Petronella Kettunen
- Department of Psychiatry and Neurochemistry,Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Anders Wallin
- Department of Psychiatry and Neurochemistry,Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry,Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.,UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry,Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Ulf Andreasson
- Department of Psychiatry and Neurochemistry,Institute of Neuroscience and Physiology, the Sahlgrenska Academy at the University of Gothenburg, Mölndal, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| |
Collapse
|
39
|
Winder Z, Wilcock D, Jicha GA. Diagnostic and Prognostic Laboratory Testing for Alzheimer Disease. Clin Lab Med 2021; 40:289-303. [PMID: 32718500 DOI: 10.1016/j.cll.2020.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
This article focuses on current clinical laboratory testing to diagnose Alzheimer disease and monitor its progression throughout its disease course. Several clinically available tests focus on analysis of amyloid and tau levels in cerebrospinal fluid as well as autosomal dominant and risk factor genes. Although the current armament of clinical laboratory testing is limited by invasiveness of cerebrospinal fluid collection, rarity of autosomal dominant genetic mutations, and uncertainties of risk inherent in nonpenetrant genes, the field is poised to advance the clinical repertoire of laboratory diagnostic testing.
Collapse
Affiliation(s)
- Zachary Winder
- Department of Physiology, Sanders-Brown Center on Aging, University of Kentucky College of Medicine, 800 South Limestone Street, Lexington, KY 40536-0230, USA
| | - Donna Wilcock
- Department of Physiology, Sanders-Brown Center on Aging, University of Kentucky College of Medicine, 800 South Limestone Street, Lexington, KY 40536-0230, USA
| | - Gregory A Jicha
- Department of Neurology, Sanders-Brown Center on Aging, University of Kentucky College of Medicine, 800 South Limestone Street, Lexington, KY 40536-0230, USA.
| |
Collapse
|
40
|
Nicoli CD, Howard VJ, Judd SE, Struck J, Manly JJ, Cushman M. Pro-Neurotensin/Neuromedin N and Risk of Cognitive Impairment in a Prospective Study. J Alzheimers Dis 2021; 76:1403-1412. [PMID: 32623400 DOI: 10.3233/jad-200456] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND The neuropeptide neurotensin (NT) has been linked to cardiometabolic disease. Cardiovascular risk factors are being recognized as risk factors for cognitive impairment. OBJECTIVE To examine the association of the stable precursor of NT, pro-neurotensin/neuromedin N (pro-NT/NMN), with incident cognitive impairment (ICI). METHODS We conducted a prospective nested case-control study in the REasons for Geographic And Racial Differences in Stroke (REGARDS) cohort. In 2003-2007, REGARDS enrolled 30,239 Black and White adults aged ≥45 years. ICI was identified using a 3-test cognitive battery administered biannually. Baseline pro-NT/NMN was measured by immunoassay in 393 cases of ICI and 490 controls after 3.4 years follow up. Multivariable logistic regression was used to calculate odds ratios (OR) of ICI by pro-NT/NMN quartiles. Race, age, and sex differences were studied with stratified models and interaction testing. RESULTS Pro-NT/NMN was higher in Black participants and those with hypertension and diabetes. Women with a 4th versus 1st-quartile pro-NT/NMN had 2.28-fold increased odds of ICI (95% CI 1.08-4.78) after adjusting for risk factors and incident stroke. There was no association of higher pro-NT/NMN quartiles with ICI in the overall group or men. There were no race or age differences in associations. CONCLUSION In this biracial population-based study, elevated systemic pro-NT/NMN was associated with more than doubled risk of ICI in women but not men. Others reported sex-specific associations in women for cardiovascular mortality and diabetes with higher pro-NT/NMN, supporting a role for future research on sex differences in the neurotensinergic system.
Collapse
Affiliation(s)
- Charles D Nicoli
- University of Vermont Larner College of Medicine, Burlington, VT, USA
| | - Virginia J Howard
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Suzanne E Judd
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | - Jennifer J Manly
- Department of Neurology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, USA
| | - Mary Cushman
- Departments of Medicine and Pathology and Laboratory Medicine, University of Vermont Larner College of Medicine, Burlington, VT, USA
| |
Collapse
|
41
|
Serra MC, Dondero KR, Larkins D, Burns A, Addison O. Healthy Lifestyle and Cognition: Interaction between Diet and Physical Activity. Curr Nutr Rep 2021; 9:64-74. [PMID: 32166628 DOI: 10.1007/s13668-020-00306-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW This review discusses current research on the impact of specific dietary patterns and exercise, both individually and combined, on cognitive function in older adults. RECENT FINDINGS Observational evidence generally supports a relationship between diet adherence and positive cognitive outcomes related to memory, executive function, and risk for cognitive impairment; however, randomized controlled trials (RCTs) are limited. Exercise research is more extensive, showing improvements in cognitive performance after exercise interventions regardless of baseline cognitive status and noting lower incidences of cognitive impairment in people who engage in regular physical activity. Evidence supports adherence to specific dietary patterns and a combination of aerobic and resistance exercise as an effective approach to mitigate age-associated cognitive decline. Further research on older adults at various stages of cognitive decline, as well as longer-term RCTs, will help determine the best clinical markers of early cognitive dysfunction, and the effectiveness of early lifestyle intervention on cognitive function.
Collapse
Affiliation(s)
- Monica C Serra
- Division of Geriatrics, Gerontology & Palliative Medicine and the Sam & Ann Barshop Institute for Longevity & Aging Studies, UT Health San Antonio, San Antonio, TX, USA. .,San Antonio GRECC, South Texas VA Health Care System, San Antonio, TX, USA.
| | | | - Derrik Larkins
- Department of Physical Therapy and Rehabilitative Science, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Aisling Burns
- Division of Geriatrics, Gerontology & Palliative Medicine and the Sam & Ann Barshop Institute for Longevity & Aging Studies, UT Health San Antonio, San Antonio, TX, USA
| | - Odessa Addison
- Department of Physical Therapy and Rehabilitative Science, University of Maryland School of Medicine, Baltimore, MD, USA.,Baltimore VA Medical Center GRECC, VA Maryland Health Care System, Baltimore, MD, USA
| |
Collapse
|
42
|
Frantellizzi V, Pani A, Ricci M, Locuratolo N, Fattapposta F, De Vincentis G. Neuroimaging in Vascular Cognitive Impairment and Dementia: A Systematic Review. J Alzheimers Dis 2021; 73:1279-1294. [PMID: 31929166 DOI: 10.3233/jad-191046] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cerebrovascular diseases are well established causes of cognitive impairment. Different etiologic entities, such as vascular dementia (VaD), vascular cognitive impairment, subcortical (ischemic) VaD, and vascular cognitive disorder, are included in the umbrella definition of vascular cognitive impairment and dementia (VCID). Because of the variability of VCID clinical presentation, there is no agreement on criteria defining the neuropathological threshold of this disorder. In fact, VCID is characterized by cerebral hemodynamic alteration which ranges from decreased cerebral blood flow to small vessels disease and involves a multifactorial process that leads to demyelination and gliosis, including blood-brain barrier disruption, hypoxia, and hypoperfusion, oxidative stress, neuroinflammation and alteration on neurovascular unit coupling, cerebral microbleeds, or superficial siderosis. Numerous criteria for the definition of VaD have been described: the National Institute of Neurological Disorders and Stroke Association Internationale pour Recherche'-et-l'Enseignement en Neurosciences criteria, the State of California Alzheimer's Disease Diagnostic and Treatment Centers criteria, DSM-V criteria, the Diagnostic Criteria for Vascular Cognitive Disorders (a VASCOG Statement), and Vascular Impairment of Cognition Classification Consensus Study. Neuroimaging is fundamental for definition and diagnosis of VCID and should be used to assess the extent, location, and type of vascular lesions. MRI is the most sensible technique, especially if used according to standardized protocols, even if CT plays an important role in several conditions. Functional neuroimaging, in particular functional MRI and PET, may facilitate differential diagnosis among different forms of dementia. This systematic review aims to explore the state of the art and future perspective of non-invasive diagnostics of VCID.
Collapse
Affiliation(s)
| | - Arianna Pani
- Clinical Pharmacology and Toxicology, University of Milan "Statale", Italy
| | - Maria Ricci
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, Rome, Italy
| | | | | | - Giuseppe De Vincentis
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
43
|
Caprihan A, Raja R, Hillmer LJ, Erhardt EB, Prestopnik J, Thompson J, Adair JC, Knoefel JE, Rosenberg GA. A double-dichotomy clustering of dual pathology dementia patients. CEREBRAL CIRCULATION - COGNITION AND BEHAVIOR 2021; 2:100011. [PMID: 34746872 PMCID: PMC8570532 DOI: 10.1016/j.cccb.2021.100011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 03/22/2021] [Accepted: 03/27/2021] [Indexed: 12/02/2022]
Abstract
INTRODUCTION Subcortical ischemic vascular disease (SIVD) and Alzheimer's disease (AD) related dementia can coexist in older subjects, leading to mixed dementia (MX). Identification of dementia sub-groups is important for designing proper treatment plans and clinical trials. METHOD An Alzheimer's disease severity (ADS) score and a vascular disease severity (VDS) score are calculated from CSF and MRI biomarkers, respectively. These scores, being sensitive to different Alzheimer's and vascular disease processes are combined orthogonally in a double-dichotomy plot. This formed an objective basis for clustering the subjects into four groups, consisting of AD, SIVD, MX and leukoaraiosis (LA). The relationship of these four groups is examined with respect to cognitive assessments and clinical diagnosis. RESULTS Cluster analysis had at least 83% agreement with the clinical diagnosis for groups based either on Alzheimer's or on vascular sensitive biomarkers, and a combined agreement of 68.8% for clustering the four groups. The VDS score was correlated to executive function (r = -0.28, p < 0.01) and the ADS score to memory function (r = -0.35, p < 0.002) after adjusting for age, sex, and education. In the subset of patients for which the cluster scores and clinical diagnoses agreed, the correlations were stronger (VDS score-executive function: r = -0.37, p < 0.006 and ADS score-memory function: r = -0.58, p < 0.0001). CONCLUSIONS The double-dichotomy clustering based on imaging and fluid biomarkers offers an unbiased method for identifying mixed dementia patients and selecting better defined sub-groups. Differential correlations with neuropsychological tests support the hypothesis that the categories of dementia represent different etiologies.
Collapse
Affiliation(s)
| | - Rajikha Raja
- The Mind Research Network, Albuquerque, NM, United States
- Department of Radiology, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Laura J. Hillmer
- Department of Neurology, University of New Mexico, Albuquerque, NM, United States
| | - Erik Barry Erhardt
- Departments of Mathematics and Statistics, University of New Mexico, Albuquerque, NM, United States
| | - Jill Prestopnik
- Department of Neurology, University of New Mexico, Albuquerque, NM, United States
| | - Jeffrey Thompson
- Department of Neurology, University of New Mexico, Albuquerque, NM, United States
| | - John C Adair
- Department of Neurology, University of New Mexico, Albuquerque, NM, United States
| | - Janice E. Knoefel
- Department of Neurology, University of New Mexico, Albuquerque, NM, United States
| | - Gary A. Rosenberg
- Department of Neurology, University of New Mexico, Albuquerque, NM, United States
| |
Collapse
|
44
|
Hosoki S, Tanaka T, Ihara M. Diagnostic and prognostic blood biomarkers in vascular dementia: From the viewpoint of ischemic stroke. Neurochem Int 2021; 146:105015. [PMID: 33781849 DOI: 10.1016/j.neuint.2021.105015] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/25/2021] [Accepted: 03/02/2021] [Indexed: 12/14/2022]
Abstract
Reliable quantitative blood biomarkers are important in vascular dementia (VaD) because early diagnosis and therapeutic intervention are effective in preventing progression of dementia. Although many blood biomarkers for acute ischemic stroke (AIS) or VaD have been reported, there are few reliable blood biomarkers. VaD and AIS have similar pathological conditions that are associated with small vessel disease (SVD) such as oxidative stress, inflammation, endothelial dysfunction, and neuronal injury. Therefore, it may be possible to find superior blood biomarkers of VaD among AIS blood biomarkers. Owing to recent developments, noncoding RNAs such as microRNA and long noncoding RNA, which can be analyzed using a single drop of blood, are also particularly reliable VaD markers because they stably reflect brain tissue damage. A multimarker combining several blood biomarkers or artificial intelligence technology may also be beneficial to compensate for insufficiencies of a single blood biomarker. This review describes the blood biomarkers of VaD and how they are related to blood biomarkers of AIS.
Collapse
Affiliation(s)
- Satoshi Hosoki
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Tomotaka Tanaka
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan
| | - Masafumi Ihara
- Department of Neurology, National Cerebral and Cardiovascular Center, Suita, Japan.
| |
Collapse
|
45
|
Plasma osteopontin as a biomarker of Alzheimer's disease and vascular cognitive impairment. Sci Rep 2021; 11:4010. [PMID: 33597603 PMCID: PMC7889621 DOI: 10.1038/s41598-021-83601-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 01/21/2021] [Indexed: 12/22/2022] Open
Abstract
Cerebrovascular disease (CeVD) and neurodegenerative dementia such as Alzheimer’s disease (AD) are frequently associated comorbidities in the elderly, sharing common risk factors and pathophysiological mechanisms including neuroinflammation. Osteopontin (OPN) is an inflammatory marker found upregulated in vascular diseases as well as in AD. However, its involvement in vascular dementia (VaD) and pre-dementia stages, namely cognitive impairment no dementia (CIND), both of which fall under the spectrum of vascular cognitive impairment (VCI), has yet to be examined. Its correlations with inflammatory cytokines in cognitive impairment also await investigation. 80 subjects with no cognitive impairment (NCI), 160 with CIND and 144 with dementia were included in a cross-sectional study on a Singapore-based memory clinic cohort. All subjects underwent comprehensive clinical, neuropsychological and brain neuroimaging assessments, together with clinical diagnoses based on established criteria. Blood samples were collected and OPN as well as inflammatory cytokines interleukin (IL)-6, IL-8 and tumor necrosis factor (TNF) were measured using immunoassays. Multivariate regression analyses showed significant associations between increased OPN and VCI groups, namely CIND with CeVD, AD with CeVD and VaD. Interestingly, higher OPN was also significantly associated with AD even in the absence of CeVD. We further showed that increased OPN significantly associated with neuroimaging markers of CeVD and neurodegeneration, including cortical infarcts, lacunes, white matter hyperintensities and brain atrophy. OPN also correlated with elevated levels of IL-6, IL-8 and TNF. Our findings suggest that OPN may play a role in both VCI and neurodegenerative dementias. Further longitudinal analyses are needed to assess the prognostic utility of OPN in disease prediction and monitoring.
Collapse
|
46
|
Leszek J, Mikhaylenko EV, Belousov DM, Koutsouraki E, Szczechowiak K, Kobusiak-Prokopowicz M, Mysiak A, Diniz BS, Somasundaram SG, Kirkland CE, Aliev G. The Links between Cardiovascular Diseases and Alzheimer's Disease. Curr Neuropharmacol 2021; 19:152-169. [PMID: 32727331 PMCID: PMC8033981 DOI: 10.2174/1570159x18666200729093724] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 06/02/2020] [Accepted: 07/16/2020] [Indexed: 12/17/2022] Open
Abstract
The root cause of non-inherited Alzheimer's disease (AD) remains unknown despite hundreds of research studies performed to attempt to solve this problem. Since proper prophylaxis remains the best strategy, many scientists have studied the risk factors that may affect AD development. There is robust evidence supporting the hypothesis that cardiovascular diseases (CVD) may contribute to AD progression, as the diseases often coexist. Therefore, a lack of well-defined diagnostic criteria makes studying the relationship between AD and CVD complicated. Additionally, inflammation accompanies the pathogenesis of AD and CVD, and is not only a consequence but also implicated as a significant contributor to the course of the diseases. Of note, АроЕε4 is found to be one of the major risk factors affecting both the cardiovascular and nervous systems. According to genome wide association and epidemiological studies, numerous common risk factors have been associated with the development of AD-related pathology. Furthermore, the risk of developing AD and CVDs appears to be increased by a wide range of conditions and lifestyle factors: hypertension, dyslipidemia, hypercholesterolemia, hyperhomocysteinemia, gut/oral microbiota, physical activity, and diet. This review summarizes the literature and provides possible mechanistic links between CVDs and AD.
Collapse
Affiliation(s)
- Jerzy Leszek
- Address correspondence to these authors at the Department of Psychiatry, Wrocław Medical University, Ul. Pasteura 10, 50-367, Wroclaw, Poland;, E-mail: and GALLY International Research Institute, 7733 Louis Pasteur Drive, #330, San Antonio, TX 78229, USA; Tel: +1-210-442-8625 or +1-440-263-7461; E-mails: ,
| | | | | | | | | | | | | | | | | | | | - Gjumrakch Aliev
- Address correspondence to these authors at the Department of Psychiatry, Wrocław Medical University, Ul. Pasteura 10, 50-367, Wroclaw, Poland;, E-mail: and GALLY International Research Institute, 7733 Louis Pasteur Drive, #330, San Antonio, TX 78229, USA; Tel: +1-210-442-8625 or +1-440-263-7461; E-mails: ,
| |
Collapse
|
47
|
Miao M, Yuan F, Ma X, Yang H, Gao X, Zhu Z, Bi J. Methylation of the RIN3 Promoter is Associated with Transient Ischemic Stroke/Mild Ischemic Stroke with Early Cognitive Impairment. Neuropsychiatr Dis Treat 2021; 17:2587-2598. [PMID: 34408420 PMCID: PMC8364373 DOI: 10.2147/ndt.s320167] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 07/23/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Early cognitive impairment after transient ischemic stroke (TIA)/mild ischemic stroke (MIS) is common but easily overlooked. It has been demonstrated that DNA methylation plays a significant role in cognitive impairment and ischemic stroke. Furthermore, it has been reported that the RIN3 gene influences transportation of the amyloid β-protein. However, to our knowledge, there has been no research related to correlations between RIN3 methylation and early-onset cognitive impairment after TIA/MIS. Therefore, this study aimed to investigate this relationship in TIA/MIS patients. METHODS This study include 28 control subjects and 84 patients with TIA/MIS who were evaluated within 7 days of TIA/MIS onset using four single-domain cognitive scales. In addition, DNA methylation of whole blood was tested. RIN3 methylation was compared between TIA/MIS and control groups and between TIA/MIS patients with early cognitive impairment and those without early cognitive impairment. Clinical variables and RIN3 methylation sites with statistical differences were then used to construct a predictive model. RESULTS Hypomethylation of the RIN3 gene was observed in the whole blood of TIA/MIS patients relative to healthy controls. Furthermore, patients with early cognitive impairment after TIA/MIS had hypomethylation of RIN3 relative to those without early cognitive impairment. CONCLUSION RIN3 methylation is strongly associated with TIA/MIS and TIA/MIS with early cognitive impairment. It is possible to influence the disease process by methylation via appropriate lifestyle and clinical interventions, and methylation of RIN3 gene sites may predict the occurrence of TIA/MIS with early cognitive impairment.
Collapse
Affiliation(s)
- Meng Miao
- Department of Neurology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, 266035, People's Republic of China
| | - Fang Yuan
- Department of Health Care, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, 266035, People's Republic of China
| | - Xiaotian Ma
- Department of Medicine Experimental Center, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, 266035, People's Republic of China
| | - Haiming Yang
- Department of Neurology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, 266035, People's Republic of China
| | - Xiang Gao
- Department of Neurology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, Shandong, 266035, People's Republic of China
| | - Zhengyu Zhu
- Department of Neurology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250033, People's Republic of China
| | - Jianzhong Bi
- Department of Neurology, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250033, People's Republic of China
| |
Collapse
|
48
|
Abstract
Cerebrovascular disease is a significant cause of cognitive impairment leading to a reduction or loss of functioning, including social and occupational. The connection cause-effect between cerebrovascular disease and cerebral infarction was originally theorized by the studies from Newcastle-Upon-Tyne, England, in the 1960s, where vascular dementia (VaD) was defined as a disease originated from several infarctions that overcome a determined threshold. It differs from Alzheimer's disease (AD), although there are various overlaps in risk factors, symptomatology, the similarity of vascular lesions, and treatment benefits. Nevertheless, AD is one-half of all cases of dementia. Cognitive impairment and dementia (VCID) has recently been proposed to include different entities such as VaD, Vascular cognitive impairment, subcortical (ischemic) VaD, and vascular cognitive disorders. VaD is the most common cause of dementia after AD. Neuroimaging is an essential part of the workup of patients with cognitive decline and in those with suspected VCID it should be used to assess the extent, location, and type of vascular lesions. Computed tomography (CT) or structural magnetic resonance imaging (MRI) are usually used for the diagnosis of vascular diseases of the brain. However, images obtained from new hybrid devices could help the neurologist in the differential diagnosis between various neuropathological entities related to VCID. Single-photon emission computed tomography (SPECT) combined with CT or MRI and positron emission tomography (PET) combined with CT or MRI represent the future of neuroimaging tools as morphological and functional data can be provided simultaneously. New prospects have been developed such as hybrid PET/SPECT/CT, a high-performance prototype able to produce high-quality images but for now suitable only for small animals. Nowadays, PET/CT and PET/MRI are good performance and high-quality instruments, even if the magnetic field of MRI represents a limitation that affects the PET electronics and positron detection ability. SPECT/MRI delineates as a potential and tempting device. It could give us both functional and anatomical details, with the advantage of lack of extra ionizing radiation and high soft-tissue contrast, important features, and considerable auxiliary for differential diagnosis in the variegate word of vascular cognitive impairment. The aim of this review is to summarize the newest viewpoints in hybrid imaging in the diagnosis of VaD and to highlight pros and cons of each methodic.
Collapse
Affiliation(s)
| | - Miriam Conte
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, Rome, Italy
| | - Giuseppe De Vincentis
- Department of Radiological Sciences, Oncology and Anatomical Pathology, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
49
|
Zhang J, Sun P, Zhou C, Zhang X, Ma F, Xu Y, Hamblin MH, Yin K. Regulatory microRNAs and vascular cognitive impairment and dementia. CNS Neurosci Ther 2020; 26:1207-1218. [PMID: 33459504 PMCID: PMC7702235 DOI: 10.1111/cns.13472] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 12/11/2022] Open
Abstract
Vascular cognitive impairment and dementia (VCID) is defined as a progressive dementia disease related to cerebrovascular injury and often occurs in aged populations. Despite decades of research, effective treatment for VCID is still absent. The pathological processes of VCID are mediated by the molecular mechanisms that are partly modulated at the post-transcriptional level. As small endogenous non-coding RNAs, microRNAs (miRs) can regulate target gene expression through post-transcriptional gene silencing. miRs have been reported to play an important role in the pathology of VCID and have recently been suggested as potential novel pharmacological targets for the development of new diagnosis and treatment strategies in VCID. In this review, we summarize the current understanding of VCID, the possible role of miRs in the regulation of VCID and attempt to envision future therapeutic strategies. Since manipulation of miR levels by either pharmacological or genetic approaches has shown therapeutic effects in experimental VCID models, we also emphasize the potential therapeutic value of miRs in clinical settings.
Collapse
Affiliation(s)
- Jing Zhang
- Department of NeurologyPittsburgh Institute of Brain Disorders & RecoveryUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Ping Sun
- Department of NeurologyPittsburgh Institute of Brain Disorders & RecoveryUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Chao Zhou
- Department of NeurologyPittsburgh Institute of Brain Disorders & RecoveryUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Xuejing Zhang
- Department of NeurologyPittsburgh Institute of Brain Disorders & RecoveryUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Feifei Ma
- Department of NeurologyPittsburgh Institute of Brain Disorders & RecoveryUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Yang Xu
- Department of NeurologyPittsburgh Institute of Brain Disorders & RecoveryUniversity of Pittsburgh School of MedicinePittsburghPAUSA
| | - Milton H. Hamblin
- Department of PharmacologyTulane University School of MedicineNew OrleansLAUSA
| | - Ke‐Jie Yin
- Department of NeurologyPittsburgh Institute of Brain Disorders & RecoveryUniversity of Pittsburgh School of MedicinePittsburghPAUSA
- Geriatric ResearchEducation and Clinical CenterVeterans Affairs Pittsburgh Healthcare SystemPittsburghPAUSA
| |
Collapse
|
50
|
Czakó C, Kovács T, Ungvari Z, Csiszar A, Yabluchanskiy A, Conley S, Csipo T, Lipecz A, Horváth H, Sándor GL, István L, Logan T, Nagy ZZ, Kovács I. Retinal biomarkers for Alzheimer's disease and vascular cognitive impairment and dementia (VCID): implication for early diagnosis and prognosis. GeroScience 2020; 42:1499-1525. [PMID: 33011937 PMCID: PMC7732888 DOI: 10.1007/s11357-020-00252-7] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 08/10/2020] [Indexed: 12/11/2022] Open
Abstract
Cognitive impairment and dementia are major medical, social, and economic public health issues worldwide with significant implications for life quality in older adults. The leading causes are Alzheimer's disease (AD) and vascular cognitive impairment/dementia (VCID). In both conditions, pathological alterations of the cerebral microcirculation play a critical pathogenic role. Currently, the main pathological biomarkers of AD-β-amyloid peptide and hyperphosphorylated tau proteins-are detected either through cerebrospinal fluid (CSF) or PET examination. Nevertheless, given that they are invasive and expensive procedures, their availability is limited. Being part of the central nervous system, the retina offers a unique and easy method to study both neurodegenerative disorders and cerebral small vessel diseases in vivo. Over the past few decades, a number of novel approaches in retinal imaging have been developed that may allow physicians and researchers to gain insights into the genesis and progression of cerebromicrovascular pathologies. Optical coherence tomography (OCT), OCT angiography, fundus photography, and dynamic vessel analyzer (DVA) are new imaging methods providing quantitative assessment of retinal structural and vascular indicators-such as thickness of the inner retinal layers, retinal vessel density, foveal avascular zone area, tortuosity and fractal dimension of retinal vessels, and microvascular dysfunction-for cognitive impairment and dementia. Should further studies need to be conducted, these retinal alterations may prove to be useful biomarkers for screening and monitoring dementia progression in clinical routine. In this review, we seek to highlight recent findings and current knowledge regarding the application of retinal biomarkers in dementia assessment.
Collapse
Affiliation(s)
- Cecilia Czakó
- Department of Ophthalmology, Semmelweis University, Budapest, Hungary
| | - Tibor Kovács
- Department of Neurology, Semmelweis University, Budapest, Hungary
| | - Zoltan Ungvari
- Translational Geroscience Laboratory, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- International Training Program in Geroscience, Theoretical Medicine Doctoral School/Departments of Medical Physics and Informatics & Cell Biology and Molecular Medicine, University of Szeged, Szeged, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Translational Geroscience Laboratory, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Theoretical Medicine Doctoral School/Departments of Medical Physics and Informatics & Cell Biology and Molecular Medicine, University of Szeged, Szeged, Hungary
| | - Andriy Yabluchanskiy
- Translational Geroscience Laboratory, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Shannon Conley
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Neuroscience, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Tamas Csipo
- Translational Geroscience Laboratory, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Agnes Lipecz
- Translational Geroscience Laboratory, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Vascular Cognitive Impairment and Neurodegeneration Program, Center for Geroscience and Healthy Brain Aging/Reynolds Oklahoma Center on Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Ophthalmology, Josa Andras Hospital, Nyiregyhaza, Hungary
| | - Hajnalka Horváth
- Department of Ophthalmology, Semmelweis University, Budapest, Hungary
| | | | - Lilla István
- Department of Ophthalmology, Semmelweis University, Budapest, Hungary
| | - Trevor Logan
- Department of Ophthalmology, Semmelweis University, Budapest, Hungary
| | - Zoltán Zsolt Nagy
- Department of Ophthalmology, Semmelweis University, Budapest, Hungary
| | - Illés Kovács
- Department of Ophthalmology, Semmelweis University, Budapest, Hungary.
- Department of Ophthalmology, Weill Cornell Medical College, New York City, NY, USA.
| |
Collapse
|