1
|
Dutt S, Bachman SL, Dahl MJ, Li Y, Yew B, Jang JY, Ho JK, Nashiro K, Min J, Yoo HJ, Gaubert A, Nguyen A, Blanken AE, Sible IJ, Marshall AJ, Kapoor A, Alitin JPM, Hoang K, Rouanet J, Sordo L, Head E, Shao X, Wang DJJ, Mather M, Nation DA. Locus coeruleus MRI contrast, cerebral perfusion, and plasma Alzheimer's disease biomarkers in older adults. Neurobiol Aging 2025; 147:12-21. [PMID: 39637519 PMCID: PMC11781958 DOI: 10.1016/j.neurobiolaging.2024.11.008] [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: 06/05/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 12/07/2024]
Abstract
The locus coeruleus (LC) is among the first brain structures impacted by Alzheimer's disease (AD), and noradrenergic denervation may contribute to early neurovascular dysfunction in AD. Mechanistic links between the LC and cerebral perfusion have been demonstrated in rodents, but there have been no similar studies in aging humans. Community-dwelling older adults with no history of stroke or dementia (N=66) underwent structural (T1-MPRAGE; T1-FSE) and perfusion (resting pCASL) MRI. Plasma AD biomarkers levels were evaluated for Aβ42/40 ratio (n=56) and pTau181 (n=60). Higher rostral LC structural MRI contrast was associated with lower perfusion in entorhinal and limbic regions but higher perfusion in lateral and medial orbitofrontal cortices. Relationships between LC structure and regional cerebral perfusion were attenuated in older adults with higher plasma pTau levels and lower plasma Aβ42/40 ratios. Previously unstudied links between LC structure and cerebral perfusion are detectible in older adults using MRI and are attenuated in those showing greater AD pathophysiologic change, suggesting an uncoupling of LC-cerebral perfusion relationships in older adults with aggregating AD-related pathophysiology.
Collapse
Affiliation(s)
- Shubir Dutt
- Department of Psychology, University of Southern California, Los Angeles, CA, USA; Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA; Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, CA, USA; Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA
| | - Shelby L Bachman
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Martin J Dahl
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA; Center for Lifespan Psychology, Max Planck Institute for Human Development, Berlin, Germany
| | - Yanrong Li
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Belinda Yew
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jung Yun Jang
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Jean K Ho
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Kaoru Nashiro
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Jungwon Min
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Hyun Joo Yoo
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Aimée Gaubert
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Amy Nguyen
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Anna E Blanken
- Department of Psychiatry and Behavioral Sciences, University of California, San Francisco, CA, USA; San Francisco Veterans Affairs Health Care System, CA, USA
| | - Isabel J Sible
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Anisa J Marshall
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Arunima Kapoor
- Department of Psychological Science, University of California, Irvine, CA, USA
| | - John Paul M Alitin
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Kim Hoang
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Jeremy Rouanet
- Department of Pathology & Laboratory Medicine, University of California, Irvine, CA, USA
| | - Lorena Sordo
- Department of Pathology & Laboratory Medicine, University of California, Irvine, CA, USA
| | - Elizabeth Head
- Department of Pathology & Laboratory Medicine, University of California, Irvine, CA, USA
| | - Xingfeng Shao
- Laboratory of FMRI Technology, Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, CA, Los Angeles, USA
| | - Danny J J Wang
- Laboratory of FMRI Technology, Mark & Mary Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, CA, Los Angeles, USA
| | - Mara Mather
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA
| | - Daniel A Nation
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, USA; Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, CA, Los Angeles, USA.
| |
Collapse
|
2
|
Bennett SK, Zeng J, Dounavi ME, Majid A, Baig SS, De Marco M, Ritchie C, O’Brien JT, Su L. Cerebral perfusion alterations in healthy young adults due to two genetic risk factors of Alzheimer's disease: APOE and MAPT. J Cereb Blood Flow Metab 2025:271678X241310731. [PMID: 39763384 PMCID: PMC11705314 DOI: 10.1177/0271678x241310731] [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] [Received: 05/26/2024] [Revised: 11/05/2024] [Accepted: 12/07/2024] [Indexed: 01/11/2025]
Abstract
Functional brain changes such as altered cerebral blood flow occur long before the onset of clinical symptoms in Alzheimer's disease (AD) and other neurodegenerative disorders. While cerebral hypoperfusion occurs in established AD, middle-aged carriers of genetic risk factors for AD, including APOE ε4, display regional hyperperfusion due to hypothesised pleiotropic or compensatory effects, representing a possible early biomarker of AD and facilitating earlier AD diagnosis. However, it is not clear whether hyperperfusion already exists even earlier in life. Here, 160 young and cognitively healthy participants from the Chinese PREVENT cohort underwent 3 T arterial spin labelling and T1 MRI and genetic testing for APOE and MAPT rs242557 status. Using FSL, we performed a whole brain voxel-wise analysis and a global mean grey matter analysis comparing for the effects of both risk genes on cerebral perfusion. No significant alterations were seen for APOE genotype, but in MAPT rs242557 A carriers, we observed a significantly hyperperfusion in the left anterior cingulate cortex and left insular cortex. There were no effects of APOE or MAPT status on the global perfusion. These results are novel and may suggest that MAPT genotypes demonstrated a distinct hemodynamic profile in a very young age.
Collapse
Affiliation(s)
- Samuel K Bennett
- Sheffield Institute for Translational Neuroscience, Division of Neuroscience, School of Medicine and Population Heath, University of Sheffield, Sheffield, UK
| | - Jianmin Zeng
- Sino-Britain Centre for Cognition and Ageing Research, Faculty of Psychology, Southwest University, Chongqing, China
| | - Maria-Eleni Dounavi
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Arshad Majid
- Sheffield Institute for Translational Neuroscience, Division of Neuroscience, School of Medicine and Population Heath, University of Sheffield, Sheffield, UK
| | - Sheharyar S Baig
- Sheffield Institute for Translational Neuroscience, Division of Neuroscience, School of Medicine and Population Heath, University of Sheffield, Sheffield, UK
| | - Matteo De Marco
- Department of Life Sciences, Brunel University London, Uxbridge, UK
| | - Craig Ritchie
- School of Medicine, University of St Andrews, St Andrews, UK
- Scottish Brain Sciences, Edinburgh, UK
| | - John T O’Brien
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Li Su
- Sheffield Institute for Translational Neuroscience, Division of Neuroscience, School of Medicine and Population Heath, University of Sheffield, Sheffield, UK
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
3
|
Mares J, Kumar G, Sharma A, Emrani S, McIntire LB, Guo J, Menon V, Nuriel T. APOE ε4-associated heterogeneity of neuroimaging biomarkers across the Alzheimer's disease continuum. Alzheimers Dement 2025; 21:e14392. [PMID: 39575672 PMCID: PMC11775459 DOI: 10.1002/alz.14392] [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: 05/06/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 01/30/2025]
Abstract
INTRODUCTION While the role of apolipoprotein E (APOE) ε4 in Alzheimer's disease (AD) susceptibility has been studied extensively, much less is known about the differences in disease presentation in APOE ε4 carriers versus non-carriers. METHODS To help elucidate these differences, we performed a broad analysis comparing the regional levels of six different neuroimaging biomarkers in the brains of APOE ε4 carriers versus non-carriers who participated in the Alzheimer's Disease Neuroimaging Initiative (ADNI). RESULTS We observed significant APOE ε4-associated heterogeneity in regional amyloid beta deposition, tau accumulation, glucose uptake, brain volume, cerebral blood flow, and white matter hyperintensities within each AD diagnostic group. We also observed important APOE ε4-associated differences in cognitively unimpaired individuals who converted to mild cognitive impairment/AD versus those who did not convert. DISCUSSION This observed heterogeneity in neuroimaging biomarkers between APOE ε4 carriers versus non-carriers may have important implications regarding the prevention, diagnosis, and treatment of AD in different subpopulations. HIGHLIGHTS An extensive study was performed on the apolipoprotein E (APOE) ε4-associated heterogeneity in neuroimaging biomarkers from the Alzheimer's Disease Neuroimaging Initiative. Robust APOE ε4-associated increases in amyloid beta (Aβ) deposition throughout the brain, in every diagnostic group, were observed. APOE ε4-associated increases in tau pathology, decreases in glucose uptake, and increases in brain atrophy, which expand in regional scope and magnitude with disease progression, were observed. Significant sex- and age-related differences in APOE ε4-associated neuroimaging biomarker heterogeneity, with overall increases in pathological presentation in female APOE ε4 carriers, were observed. Regional differences in Aβ deposition, tau accumulation, glucose uptake, ventricle size, and white matter hyperintensities were observed in cognitively normal participants who converted to mild cognitive impairment/Alzheimer's disease, which may hold potential predictive value.
Collapse
Affiliation(s)
- Jason Mares
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
- Department of NeurologyColumbia UniversityNew YorkNew YorkUSA
| | - Gautam Kumar
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
- Department of Pathology and Cell BiologyColumbia UniversityNew YorkNew YorkUSA
- Department of NeurobiologyUniversity of MarylandBaltimoreMarylandUSA
| | - Anurag Sharma
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
- Department of Pathology and Cell BiologyColumbia UniversityNew YorkNew YorkUSA
| | - Sheina Emrani
- Department of NeurologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Laura Beth McIntire
- Lipidomics and Biomarker Discovery LabBrain Health Imaging InstituteDepartment of RadiologyWeill Cornell MedicineNew YorkNew YorkUSA
| | - Jia Guo
- Department of PsychiatryColumbia UniversityNew YorkNew YorkUSA
- Zuckerman InstituteColumbia UniversityNew YorkNew YorkUSA
| | - Vilas Menon
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
- Department of NeurologyColumbia UniversityNew YorkNew YorkUSA
| | - Tal Nuriel
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainColumbia UniversityNew YorkNew YorkUSA
- Department of Pathology and Cell BiologyColumbia UniversityNew YorkNew YorkUSA
| | | |
Collapse
|
4
|
Callewaert B, Gsell W, Lox M, Himmelreich U, Jones EAV. A timeline study on vascular co-morbidity induced cerebral endothelial dysfunction assessed by perfusion MRI. Neurobiol Dis 2024; 202:106709. [PMID: 39433136 DOI: 10.1016/j.nbd.2024.106709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 10/18/2024] [Accepted: 10/18/2024] [Indexed: 10/23/2024] Open
Abstract
Endothelial dysfunction is considered a key element in the early pathogenesis of neurodegenerative disorders. Dysfunction of the cerebral endothelial cells can result in dysregulation of cerebral perfusion and disruption of the Blood Brain Barrier (BBB), leading to brain damage, neurodegeneration and cognitive decline. It has been shown that the presence of modifiable risk factors exacerbates endothelial dysfunction. This study primarily aimed to identify which among various perfusion MRI methodologies could be effectively utilized to non-invasively identify early pathological alterations as a result of endothelial dysfunction. We compared these perfusion MRI measurements to invasive immunohistochemistry to detect early pathological alterations in the cerebral vasculature of a rat model of multiple cardiovascular co-morbidities (the ZSF1 Obese rat) at several stages of the cerebrovascular pathology. We observed cerebral hyperperfusion, expressed by increased Cerebral Blood Flow (CBF) and increased BBB permeability in the ZSF1 Obese rats, at an early stage of disease development. The increase in CBF observed with Arterial Spin Labeling (ASL) was lost during later stages of disease progression. These findings are in line with recent clinical findings in early stages of Alzheimer's disease (AD), that also show early increases in CBF.
Collapse
Affiliation(s)
- Bram Callewaert
- Center for Molecular and Vascular Biology (CMVB), Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium; Biomedical MRI unit, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
| | - Willy Gsell
- Biomedical MRI unit, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
| | - Marleen Lox
- Center for Molecular and Vascular Biology (CMVB), Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium
| | - Uwe Himmelreich
- Biomedical MRI unit, Department of Imaging and Pathology, KU Leuven, 3000 Leuven, Belgium
| | - Elizabeth A V Jones
- Center for Molecular and Vascular Biology (CMVB), Department of Cardiovascular Sciences, KU Leuven, 3000 Leuven, Belgium; School for Cardiovascular Diseases (CARIM), Department of Cardiology, Maastricht University, 6200 Maastricht, the Netherlands.
| |
Collapse
|
5
|
Muer JD, Didier KD, Wannebo BM, Sanchez S, Khademi Motlagh H, Haley TL, Carter KJ, Banks NF, Eldridge MW, Serlin RC, Wieben O, Schrage WG. Sex differences in gray matter, white matter, and regional brain perfusion in young, healthy adults. Am J Physiol Heart Circ Physiol 2024; 327:H847-H858. [PMID: 39120466 PMCID: PMC11482274 DOI: 10.1152/ajpheart.00341.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/26/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
Cerebrovascular and neurological diseases exhibit sex-specific patterns in prevalence, severity, and regional specificity, some of which are associated with altered cerebral blood flow (CBF). Females often exhibit higher resting CBF, but understanding the impact of sex per se on CBF is hampered by study variability in age, comorbidities, medications, and control for menstrual cycle or hormone therapies. A majority of studies report whole brain CBF without differentiating between gray and white matter or without assessing regional CBF. Thus fundamental sex differences in regional or whole brain CBF remain unclarified. While controlling for the above confounders, we tested the hypothesis that females will exhibit higher total gray and white matter perfusion as well as regional gray matter perfusion. Adults 18-30 yr old (females = 22 and males = 26) were studied using arterial spin labeling (ASL) magnetic resonance imaging (MRI) scans followed by computational anatomy toolbox (CAT12) analysis in statistical parametric mapping (SPM12) to quantify CBF relative to brain volume. Females displayed 40% higher perfusion globally (females = 62 ± 9 and males = 45 ± 10 mL/100 g/min, P < 0.001), gray matter (females = 75 ± 11 and males = 54 ± 12 mL/100 g/min, P < 0.001), and white matter (females = 44 ± 6 and males = 32 ± 7 mL/100 g/min, P < 0.001). Females exhibited greater perfusion than males in 67 of the 68 regions tested, ranging from 14% to 66% higher. A second MRI approach (4-dimensional flow) focused on large arteries confirmed the sex difference in global CBF. These data indicate strikingly higher basal CBF in females at global, gray, and white matter levels and across dozens of brain regions and offer new clarity into fundamental sex differences in global and regional CBF regulation before aging or pathology.NEW & NOTEWORTHY MRI used to measure cerebral blood flow (CBF) in gray matter, white matter, and 68 regions in healthy men and women. This study demonstrated that CBF is 40% higher in women, the highest sex difference reported, when controlling for numerous important clinical confounders like age, smoking, menstrual cycle, comorbidities, and medications.
Collapse
Affiliation(s)
- Jessica D Muer
- Department of Kinesiology, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Kaylin D Didier
- Department of Kinesiology, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Brett M Wannebo
- Department of Kinesiology, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Sophie Sanchez
- Department of Kinesiology, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Hedyeh Khademi Motlagh
- Department of Kinesiology, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Travis L Haley
- Department of Kinesiology, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Katrina J Carter
- Department of Kinesiology, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Nile F Banks
- Department of Kinesiology, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Marlowe W Eldridge
- Department of Pediatrics, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Ronald C Serlin
- Department of Educational Physcology, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - Oliver Wieben
- Department of Radiology, University of Wisconsin-Madison, Madison, Wisconsin, United States
- Department of Medical Physics, University of Wisconsin-Madison, Madison, Wisconsin, United States
| | - William G Schrage
- Department of Kinesiology, University of Wisconsin-Madison, Madison, Wisconsin, United States
| |
Collapse
|
6
|
Aamand R, Rasmussen PM, Andersen KS, de Paoli S, Weitzberg E, Christiansen M, Lund TE, Østergaard L. Cerebral microvascular changes in healthy carriers of the APOE-ɛ4 Alzheimer's disease risk gene. PNAS NEXUS 2024; 3:pgae369. [PMID: 39253395 PMCID: PMC11382292 DOI: 10.1093/pnasnexus/pgae369] [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: 05/08/2024] [Accepted: 08/13/2024] [Indexed: 09/11/2024]
Abstract
APOE-ɛ4 is a genetic risk factor for Alzheimer's disease (AD). AD is associated with reduced cerebral blood flow (CBF) and with microvascular changes that limit the transport of oxygen from blood into brain tissue: reduced microvascular cerebral blood volume and high relative transit time heterogeneity (RTH). Healthy APOE-ɛ4 carriers reveal brain regions with elevated CBF compared with carriers of the common ɛ3 allele. Such asymptomatic hyperemia may reflect microvascular dysfunction: a vascular disease entity characterized by suboptimal tissue oxygen uptake, rather than limited blood flow per se. Here, we used perfusion MRI to show that elevated regional CBF is accompanied by reduced capillary blood volume in healthy APOE-ɛ4 carriers (carriers) aged 30-70 years compared with similarly aged APOE-ɛ3 carriers (noncarriers). Younger carriers have elevated hippocampal RTH and more extreme RTH values throughout both white matter (WM) and cortical gray matter (GM) compared with noncarriers. Older carriers have reduced WM CBF and more extreme GM RTH values than noncarriers. Across all groups, lower WM and hippocampal RTH correlate with higher educational attainment, which is associated with lower AD risk. Three days of dietary nitrate supplementation increased carriers' WM CBF but caused older carriers to score worse on two of six aggregate neuropsychological scores. The intervention improved late recall in younger carriers and in noncarriers. The APOE-ɛ4 gene is associated with microvascular changes that may impair tissue oxygen extraction. We speculate that vascular risk factor control is particularly important for APOE-ɛ4 carriers' healthy aging.
Collapse
Affiliation(s)
- Rasmus Aamand
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience (CFIN), Aarhus University, 8000 Aarhus, Denmark
| | - Peter M Rasmussen
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience (CFIN), Aarhus University, 8000 Aarhus, Denmark
| | - Katrine Schilling Andersen
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience (CFIN), Aarhus University, 8000 Aarhus, Denmark
| | - Stine de Paoli
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience (CFIN), Aarhus University, 8000 Aarhus, Denmark
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden
- Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, 17177 Stockholm, Sweden
| | - Michael Christiansen
- Department for Congenital Disorders, Statens Serum Institut, 2300 Copenhagen, Denmark
- Department of Biomedical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Torben E Lund
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience (CFIN), Aarhus University, 8000 Aarhus, Denmark
| | - Leif Østergaard
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience (CFIN), Aarhus University, 8000 Aarhus, Denmark
- Department of Neuroradiology, Aarhus University Hospital, 8200 Aarhus, Denmark
| |
Collapse
|
7
|
Marshall AG, Neikirk K, Shao B, Crabtree A, Vue Z, Beasley HK, Garza-Lopez E, Scudese E, Wanjalla CN, Kirabo A, Albritton CF, Jamison S, Demirci M, Murray SA, Cooper AT, Taffet GE, Hinton AO, Reddy AK. Methods to Utilize Pulse Wave Velocity to Measure Alterations in Cerebral and Cardiovascular Parameters. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.22.546154. [PMID: 38798364 PMCID: PMC11118486 DOI: 10.1101/2023.06.22.546154] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Alzheimer's Disease (AD) is a global health issue, affecting over 6 million in the United States, with that number expected to increase as the aging population grows. As a neurodegenerative disorder that affects memory and cognitive functions, it is well established that AD is associated with cardiovascular risk factors beyond only cerebral decline. However, the study of cerebrovascular techniques for AD is still evolving. Here, we provide reproducible methods to measure impedance-based pulse wave velocity (PWV), a marker of arterial stiffness, in the systemic vascular (aortic PWV) and in the cerebral vascular (cerebral PWV) systems. Using aortic impedance and this relatively novel technique of cerebral impedance to comprehensively describe the systemic vascular and the cerebral vascular systems, we examined the sex-dependent differences in 5x transgenic mice (5XFAD) with AD under normal and high-fat diet, and in wild-type mice under a normal diet. Additionally, we validated our method for measuring cerebrovascular impedance in a model of induced stress in 5XFAD. Together, our results show that sex and diet differences in wildtype and 5XFAD mice account for very minimal differences in cerebral impedance. Interestingly, 5XFAD, and not wildtype, male mice on a chow diet show higher cerebral impedance, suggesting pathological differences. Opposingly, when we subjected 5XFAD mice to stress, we found that females showed elevated cerebral impedance. Using this validated method of measuring impedance-based aortic and cerebral PWV, future research may explore the effects of modifying factors including age, chronic diet, and acute stress, which may mediate cardiovascular risk in AD.
Collapse
Affiliation(s)
- Andrea G. Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Bryanna Shao
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Amber Crabtree
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Heather K. Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Edgar Garza-Lopez
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Estevão Scudese
- Laboratory of Biosciences of Human Motricity (LABIMH) of the Federal University of State of Rio de Janeiro (UNIRIO), Rio de Janeiro, Brazil; Sport Sciences and Exercise Laboratory (LaCEE), Catholic University of Petrópolis (UCP), Brazil
| | - Celestine N. Wanjalla
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Claude F Albritton
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- School of Graduate Studies, Meharry Medical College, Nashville, TN
| | - Sydney Jamison
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- School of Graduate Studies, Meharry Medical College, Nashville, TN
| | - Mert Demirci
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Sandra A. Murray
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Anthonya T. Cooper
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - George E Taffet
- Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| | - Antentor O. Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Anilkumar K. Reddy
- Department of Medicine, Baylor College of Medicine, One Baylor Plaza, Houston, TX 77030, USA
| |
Collapse
|
8
|
Musaeus CS, Kjaer TW, Lindberg U, Vestergaard MB, Bo H, Larsson W, Press DZ, Andersen BB, Høgh P, Kidmose P, Hemmsen MC, Rank ML, Hasselbalch SG, Waldemar G, Frederiksen KS. Subclinical epileptiform discharges in Alzheimer's disease are associated with increased hippocampal blood flow. Alzheimers Res Ther 2024; 16:80. [PMID: 38610005 PMCID: PMC11010418 DOI: 10.1186/s13195-024-01432-9] [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: 10/26/2023] [Accepted: 03/19/2024] [Indexed: 04/14/2024]
Abstract
BACKGROUND In epilepsy, the ictal phase leads to cerebral hyperperfusion while hypoperfusion is present in the interictal phases. Patients with Alzheimer's disease (AD) have an increased prevalence of epileptiform discharges and a study using intracranial electrodes have shown that these are very frequent in the hippocampus. However, it is not known whether there is an association between hippocampal hyperexcitability and regional cerebral blood flow (rCBF). The objective of the study was to investigate the association between rCBF in hippocampus and epileptiform discharges as measured with ear-EEG in patients with Alzheimer's disease. Our hypothesis was that increased spike frequency may be associated with increased rCBF in hippocampus. METHODS A total of 24 patients with AD, and 15 HC were included in the analysis. Using linear regression, we investigated the association between rCBF as measured with arterial spin-labelling MRI (ASL-MRI) in the hippocampus and the number of spikes/sharp waves per 24 h as assessed by ear-EEG. RESULTS No significant difference in hippocampal rCBF was found between AD and HC (p-value = 0.367). A significant linear association between spike frequency and normalized rCBF in the hippocampus was found for patients with AD (estimate: 0.109, t-value = 4.03, p-value < 0.001). Changes in areas that typically show group differences (temporal-parietal cortex) were found in patients with AD, compared to HC. CONCLUSIONS Increased spike frequency was accompanied by a hemodynamic response of increased blood flow in the hippocampus in patients with AD. This phenomenon has also been shown in patients with epilepsy and supports the hypothesis of hyperexcitability in patients with AD. The lack of a significant difference in hippocampal rCBF may be due to an increased frequency of epileptiform discharges in patients with AD. TRIAL REGISTRATION The study is registered at clinicaltrials.gov (NCT04436341).
Collapse
Affiliation(s)
- Christian Sandøe Musaeus
- Danish Dementia Research Centre (DDRC), Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Inge Lehmanns vej 8, Copenhagen, 2100, Denmark.
| | - Troels Wesenberg Kjaer
- Danish Dementia Research Centre (DDRC), Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Inge Lehmanns vej 8, Copenhagen, 2100, Denmark
| | - Ulrich Lindberg
- Functional Imaging Unit, Department of Clinical Physiology and Nuclear Medicine, University of Copenhagen, Valdemar Hansens Vej 13, Glostrup, 2600, Denmark
| | - Mark B Vestergaard
- Functional Imaging Unit, Department of Clinical Physiology and Nuclear Medicine, University of Copenhagen, Valdemar Hansens Vej 13, Glostrup, 2600, Denmark
| | - Henrik Bo
- Danish Dementia Research Centre (DDRC), Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Inge Lehmanns vej 8, Copenhagen, 2100, Denmark
| | - Wiberg Larsson
- Functional Imaging Unit, Department of Clinical Physiology and Nuclear Medicine, University of Copenhagen, Valdemar Hansens Vej 13, Glostrup, 2600, Denmark
| | - Daniel Zvi Press
- Berenson-Allen Center for Non-invasive Brain Stimulation, Beth Israel Deaconess Medical Center, Harvard Medical School, 330 Brookline Ave, Boston, MA, 02215, USA
| | - Birgitte Bo Andersen
- Danish Dementia Research Centre (DDRC), Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Inge Lehmanns vej 8, Copenhagen, 2100, Denmark
| | - Peter Høgh
- Regional Dementia Research Centre, Department of Neurology, Zealand University Hospital, Vestermarksvej 11, Roskilde, 4000, Denmark
- Department of Clinical Medicine, University of Copenhagen, Blegdamsvej 3B, Copenhagen, 2200, Denmark
| | - Preben Kidmose
- Department of Electrical and Computer Engineering, Aarhus University, Finlandsgade 22, Aarhus N, 8200, Denmark
| | | | | | - Steen Gregers Hasselbalch
- Danish Dementia Research Centre (DDRC), Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Inge Lehmanns vej 8, Copenhagen, 2100, Denmark
- Department of Clinical Medicine, University of Copenhagen, Blegdamsvej 3B, Copenhagen, 2200, Denmark
| | - Gunhild Waldemar
- Danish Dementia Research Centre (DDRC), Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Inge Lehmanns vej 8, Copenhagen, 2100, Denmark
- Department of Clinical Medicine, University of Copenhagen, Blegdamsvej 3B, Copenhagen, 2200, Denmark
| | - Kristian Steen Frederiksen
- Danish Dementia Research Centre (DDRC), Department of Neurology, Copenhagen University Hospital - Rigshospitalet, Inge Lehmanns vej 8, Copenhagen, 2100, Denmark
- Department of Clinical Medicine, University of Copenhagen, Blegdamsvej 3B, Copenhagen, 2200, Denmark
| |
Collapse
|
9
|
Motger-Albertí A, de la Calle E, Giménez M, Blasco G, Biarnés C, Arnoriaga-Rodríguez M, Puig J, Coll-Martínez C, Contreras-Rodríguez O, Fernández-Real JM. Increased brain fractional perfusion in obesity using intravoxel incoherent motion (IVIM) MRI metrics. Obesity (Silver Spring) 2024; 32:756-767. [PMID: 38383843 DOI: 10.1002/oby.24001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/24/2023] [Accepted: 12/22/2023] [Indexed: 02/23/2024]
Abstract
OBJECTIVE This research seeks to shed light on the associations between brain perfusion, cognitive function, and mental health in individuals with and without obesity. METHODS In this study, we employed the noninvasive intravoxel incoherent motion (IVIM) magnetic resonance imaging (MRI) technique to examine brain fractional perfusion (FP) in two groups: individuals with obesity (N = 72) and healthy controls (N = 66). Additionally, we investigated potential associations between FP, cognitive function, and depressive symptoms in the participants with and without obesity. Finally, artificial intelligence algorithms (Boruta analysis) were also used. RESULTS Participants with obesity exhibited increased FP within dopaminergic brain circuits, particularly involving prefrontal cortex areas, anterior and posterior sections of the cingulate cortex, the right striatum, and the midbrain. Additionally, these individuals demonstrated lower working memory and higher depressive symptoms compared to the control group. Notably, higher FP in the inferior temporal and occipital cortices correlated with greater depressive symptoms, whereas increased FP in the right ventral caudate and the midbrain was associated with better working memory performance. A link between inflammatory and metabolic variables, with a particular emphasis on monocytes, and FP in obesity was also evidenced by Boruta analysis. CONCLUSIONS Increased brain perfusion in individuals with obesity is associated with cognitive function and mental health through interaction with metabolic and inflammatory factors.
Collapse
Affiliation(s)
- Anna Motger-Albertí
- Department of Diabetes, Endocrinology, and Nutrition (UDEN), Girona Biomedical Research Institute, Josep Trueta University Hospital, Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Girona, Spain
- Department of Medical Sciences, School of Medicine, University of Girona, Girona, Spain
| | - Elena de la Calle
- Department of Radiology-Medical Imaging, Girona Biomedical Research Institute, Josep Trueta University Hospital, Girona, Spain
| | - Mònica Giménez
- Department of Radiology-Medical Imaging, Girona Biomedical Research Institute, Josep Trueta University Hospital, Girona, Spain
| | - Gerard Blasco
- Department of Radiology-Medical Imaging, Girona Biomedical Research Institute, Josep Trueta University Hospital, Girona, Spain
| | - Carles Biarnés
- Department of Radiology-Medical Imaging, Girona Biomedical Research Institute, Josep Trueta University Hospital, Girona, Spain
| | - María Arnoriaga-Rodríguez
- Department of Diabetes, Endocrinology, and Nutrition (UDEN), Girona Biomedical Research Institute, Josep Trueta University Hospital, Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Girona, Spain
| | - Josep Puig
- Department of Medical Sciences, School of Medicine, University of Girona, Girona, Spain
- Department of Radiology-Medical Imaging, Girona Biomedical Research Institute, Josep Trueta University Hospital, Girona, Spain
| | - Clàudia Coll-Martínez
- Neuroimmunology and Multiple Sclerosis Unit, Department of Neurology, Josep Trueta University Hospital, Girona, Spain
- Neurodegeneration and Neuroinflammation Research Group, Girona Biomedical Research Institute, Department of Medical Sciences, University of Girona, Girona, Spain
| | - Oren Contreras-Rodríguez
- Department of Psychiatry and Legal Medicine, Faculty of Medicine, Universitat Autònoma de Barcelona, Bellaterra, Spain
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain
| | - José Manuel Fernández-Real
- Department of Diabetes, Endocrinology, and Nutrition (UDEN), Girona Biomedical Research Institute, Josep Trueta University Hospital, Girona, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Girona, Spain
- Department of Medical Sciences, School of Medicine, University of Girona, Girona, Spain
| |
Collapse
|
10
|
Zhu W, Gao Z, Li H, Huang Z, Li X, Wang H, Wu X, Tian Y, Zhou S, Li X, Yu Y. Education reduces cognitive dysfunction in Alzheimer's disease by changing regional cerebral perfusion: An in-vivo arterial spin labeling study. Neurol Sci 2023; 44:2349-2361. [PMID: 36843146 DOI: 10.1007/s10072-023-06696-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 02/17/2023] [Indexed: 02/28/2023]
Abstract
OBJECTIVE Formal education and other cognitive challenges influence brain structure and improve function. It is believed that cognitive activities create a cognitive reserve (CR) that can slow the decline due to aging and neurodegenerative diseases. This study investigated alterations of regional cerebral blood flow (rCBF) associated with high and low CR in different stages of Alzheimer's disease (AD) and examined whether rCBF alteration mediates the relationship between education and cognitive performance. METHODS Patients with AD or amnestic mild cognitive impairment (aMCI) and healthy controls were divided into low cognitive reserve (LCR) and high cognitive reserve (HCR) subgroups according to median of education years (≤ 9 vs. > 9 years). The final study population included 89 AD patients (67 LCR, 22 HCR), 74 aMCI patients (44 LCR, 30 HCR), and 66 healthy controls (29 LCR, 37 HCR). All subjects were examined by arterial spin labeling magnetic resonance imaging and a neurocognitive test battery. rCBF was compared among groups by two-way analysis of variance. Mediation analyses were used to explore the relationships among education, rCBF, and cognitive test scores. RESULTS There were significant interaction effects of disease state (AD, aMCI, HC) and education level (LCR, HCR) on CBF in right hippocampus, posterior cingulate cortex, and right inferior parietal cortex (R_IPC). Education regulated episodic memory score by influencing right hippocampal CBF in HC_HCR and aMCI_HCR subgroups. CONCLUSION Our results indicate that the protective effect of education against cognitive dysfunction in early-stage AD is mediated at least partially by altered CBF in right hippocampus.
Collapse
Affiliation(s)
- Wanqiu Zhu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230022, China
| | - Ziwen Gao
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230022, China
| | - Hui Li
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230022, China
| | - Ziang Huang
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230022, China
| | - Xiaohu Li
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230022, China
| | - Haibao Wang
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230022, China
| | - Xingqi Wu
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yanghua Tian
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Shanshan Zhou
- Department of Neurology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Xiaoshu Li
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230022, China.
| | - Yongqiang Yu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230022, China.
| |
Collapse
|
11
|
Press DZ, Musaeus CS, Zhao L, Breton J, Shafi MM, Dai W, Alsop DC. Levetiracetam Increases Hippocampal Blood Flow in Alzheimer's Disease as Measured by Arterial Spin Labelling MRI. J Alzheimers Dis 2023:JAD220614. [PMID: 37125545 DOI: 10.3233/jad-220614] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
BACKGROUND Patients with Alzheimer's disease (AD) have an increased risk of developing epileptiform discharges, which is associated with a more rapid rate of progression. This suggests that suppression of epileptiform activity could have clinical benefit in patients with AD. OBJECTIVE In the current study, we tested whether acute, intravenous administration of levetiracetam led to changes in brain perfusion as measured with arterial spin labeling MRI (ASL-MRI) in AD. METHODS We conducted a double-blind, within-subject crossover design study in which participants with mild AD (n = 9) received placebo, 2.5 mg/kg, and 7.5 mg/kg of LEV intravenously in a random order in three sessions. Afterwards, the participants underwent ASL-MRI. RESULTS Analysis of relative cerebral blood flow (rCBF) between 2.5 mg of levetiracetam and placebo showed significant decreases in a cluster that included the posterior cingulate cortex, the precuneus, the posterior part of the cingulate gyrus, while increased cerebral blood flow was found in both temporal lobes involving the hippocampus. CONCLUSION Administration of 2.5 mg/kg of LEV in patients without any history of epilepsy leads to changes in rCBF in areas known to be affected in the early stages of AD. These areas may be the focus of the epileptiform activity. Larger studies are needed to confirm the current findings.
Collapse
Affiliation(s)
- Daniel Zvi Press
- Berenson-Allen Center for Non-invasive Brain Stimulation, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Christian Sandøe Musaeus
- Berenson-Allen Center for Non-invasive Brain Stimulation, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Neurology, Danish Dementia Research Centre, Copenhagen University Hospital, Rigshospitalet, Denmark
| | - Li Zhao
- Key Laboratory for Biomedical Engineering of Ministry of Education, College of Biomedical Engineering & Instrument Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jocelyn Breton
- Berenson-Allen Center for Non-invasive Brain Stimulation, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Mouhsin M Shafi
- Berenson-Allen Center for Non-invasive Brain Stimulation, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Weiying Dai
- Department of Computer Science, State University of New York at Binghamton, NY, USA
| | - David C Alsop
- Department of Radiology, Division of MRI Research, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
12
|
Hays Weeks CC, Zlatar ZZ, Meloy MJ, Shin DD, Thomas L, Wierenga CE. APOE Genotype Modifies the Association of Fusiform Gyrus Cerebral Metabolic Rate of Oxygen Consumption and Object Naming Performance. J Alzheimers Dis 2023; 91:1371-1383. [PMID: 36641668 DOI: 10.3233/jad-220749] [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: 01/12/2023]
Abstract
BACKGROUND The apolipoprotein E (APOE) ɛ4 allele confers risk for age and Alzheimer's disease related cognitive decline but the mechanistic link remains poorly understood. Blood oxygenation level dependent (BOLD) response in the fusiform gyrus (FG) during object naming appears greater among APOEɛ4 carriers even in the face of equivalent cognitive performance, suggesting neural compensation. However, BOLD is susceptible to known age and APOE-related vascular changes that could confound its interpretation. OBJECTIVE To address this limitation, we used calibrated fMRI during an object naming task and a hypercapnic challenge to obtain a more direct measure of neural function - percent change cerebral metabolic rate of oxygen consumption (%ΔCMRO2). METHODS Participants were 45 older adults without dementia (28 ɛ4-, 17 ɛ4+) between the ages of 65 and 85. We examined APOE-related differences in %ΔCMRO2 in the FG during object naming and the extent to which APOE modified associations between FG %ΔCMRO2 and object naming accuracy. Exploratory analyses also tested the hypothesis that %ΔCMRO2 is less susceptible to vascular compromise than are measures of %ΔCBF and %ΔBOLD. RESULTS We observed a modifying role of APOE on associations between FG %ΔCMRO2 and cognition, with ɛ4 carriers (but not non-carriers) demonstrating a positive association between right FG %ΔCMRO2 and object naming accuracy. CONCLUSION Results suggest that the relationship between neural function and cognition is altered among older adult APOEɛ4 carriers prior to the onset of dementia, implicating CMRO2 response as a potential mechanism to support cognition in APOE-related AD risk.
Collapse
Affiliation(s)
- Chelsea C Hays Weeks
- VA San Diego Healthcare System, San Diego, CA, USA.,Department of Psychiatry, UC San Diego, La Jolla, CA, USA
| | | | - M J Meloy
- VA San Diego Healthcare System, San Diego, CA, USA
| | | | - Liu Thomas
- Department of Radiology, UC San Diego, La Jolla, CA, USA
| | - Christina E Wierenga
- VA San Diego Healthcare System, San Diego, CA, USA.,Department of Psychiatry, UC San Diego, La Jolla, CA, USA
| |
Collapse
|
13
|
The Utility of Arterial Spin Labeling MRI in Medial Temporal Lobe as a Vascular Biomarker in Alzheimer's Disease Spectrum: A Systematic Review and Meta-Analysis. Diagnostics (Basel) 2022; 12:diagnostics12122967. [PMID: 36552974 PMCID: PMC9776573 DOI: 10.3390/diagnostics12122967] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/14/2022] [Accepted: 11/18/2022] [Indexed: 11/29/2022] Open
Abstract
We sought to systematically review and meta-analy the role of cerebral blood flow (CBF) in the medial temporal lobe (MTL) using arterial spin labeling magnetic resonance imaging (ASL-MRI) and compare this in patients with Alzheimer's disease (AD), individuals with mild cognitive impairment (MCI), and cognitively normal adults (CN). The prevalence of AD is increasing and leading to high healthcare costs. A potential biomarker that can identify people at risk of developing AD, whilst cognition is normal or only mildly affected, will enable risk-stratification and potential therapeutic interventions in the future. All studies investigated the role of CBF in the MTL and compared this among AD, MCI, and CN participants. A total of 26 studies were included in the systematic review and 11 in the meta-analysis. Three separate meta-analyses were conducted. Four studies compared CBF in the hippocampus of AD compared with the CN group and showed that AD participants had 2.8 mL/min/100 g lower perfusion compared with the CN group. Eight studies compared perfusion in the hippocampus of MCI vs. CN group, which showed no difference. Three studies compared perfusion in the MTL of MCI vs. CN participants and showed no statistically significant differences. CBF measured via ASL-MRI showed impairment in AD compared with the CN group in subregions of the MTL. CBF difference was significant in hippocampus between the AD and CN groups. However, MCI and CN group showed no significant difference in subregions of MTL.
Collapse
|
14
|
Drake JA, Jakicic JM, Rogers RJ, Aghjayan SL, Stillman CM, Donofry SD, Roecklein KA, Lang W, Erickson KI. Reduced brain activity during a working memory task in middle-aged apolipoprotein E ε4 carriers with overweight/obesity. Front Hum Neurosci 2022; 16:1001229. [PMID: 36504632 PMCID: PMC9732810 DOI: 10.3389/fnhum.2022.1001229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 11/07/2022] [Indexed: 11/27/2022] Open
Abstract
Objective The apolipoprotein E ε4 (APOE ε4) allele and midlife obesity are independent risk factors for Alzheimer's disease (AD). Both of these risk factors are also associated with differences in brain activation, as measured by blood oxygenation level-dependent (BOLD) responses, in the absence of detectable cognitive deficits. Although the presence of these risk factors may influence brain activity during working memory tasks, no study to date has examined whether the presence of the ε4 allele explains variation in working memory brain activity while matching for levels of overweight/obesity. The primary aim of this study was to determine whether the presence of the ε4 allele is associated with differences in task-functional magnetic resonance imaging (fMRI) brain activation in adults with overweight/obesity. We predicted that ε4 carriers would have greater brain activation in regions that support working memory. Methods This ancillary study included 48 (n = 24 APOE ε4 carriers; n = 24 APOE ε4 non-carriers), sedentary middle-aged adults (Mean age = 44.63 ± 8.36 years) with overweight/obesity (Mean BMI = 32.43 ± 4.12 kg/m2) who were matched on demographic characteristics. Participants were a subsample enrolled in 12-month randomized clinical trial examining the impact of energy-restricted diet and exercise on cardiovascular health outcomes. Participants completed a n-back working memory task with fMRI, which were completed within one month of the start of the intervention. Participants also underwent pseudo-continuous arterial spin labeling scans, a MRI measure of cerebral blood flow (CBF). Results Compared to non-ε4 carriers with overweight/obesity, ε4 carriers with overweight/obesity had lower fMRI brain activity in the middle frontal gyrus, pre and post central gyrus, supramarginal gyrus, superior temporal gyrus, lateral occipital cortex, and angular gyrus (z range = 2.52-3.56) during the n-back working memory task. Differences persisted even when controlling for CBF in these brain regions. Conclusion These results indicate that presence of the APOE ε4 allele in middle-aged adults with overweight/obesity is related to altered brain activity during a working memory paradigm, which may confer risk for accelerated neurocognitive decline in late adulthood. Future research is needed to clarify the clinical implications of these findings in the context of risk for AD.
Collapse
Affiliation(s)
- Jermon A. Drake
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States,Center for Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, PA, United States,*Correspondence: Jermon A. Drake,
| | - John M. Jakicic
- Division of Physical Activity and Weight Management, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, United States
| | | | - Sarah L. Aghjayan
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States,Center for Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Chelsea M. Stillman
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Shannon D. Donofry
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States,Allegheny Health Network, Psychiatry and Behavioral Health Institute, Pittsburgh, PA, United States
| | - Kathryn A. Roecklein
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States
| | - Wei Lang
- Center on Aging and Mobility, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Kirk I. Erickson
- Department of Psychology, University of Pittsburgh, Pittsburgh, PA, United States,Center for Neural Basis of Cognition, Carnegie Mellon University, Pittsburgh, PA, United States,PROFITH “PROmoting FITness and Health Through Physical Activity” Research Group, Department of Physical and Sports Education, Faculty of Sport Sciences, Sport and Health University Research Institute (iMUDS), University of Granada, Granada, Spain,AdventHealth Research Institute, Neuroscience Institute, Orlando, FL, United States
| |
Collapse
|
15
|
Zhu WM, Neuhaus A, Beard DJ, Sutherland BA, DeLuca GC. Neurovascular coupling mechanisms in health and neurovascular uncoupling in Alzheimer's disease. Brain 2022; 145:2276-2292. [PMID: 35551356 PMCID: PMC9337814 DOI: 10.1093/brain/awac174] [Citation(s) in RCA: 55] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 05/05/2022] [Accepted: 05/07/2022] [Indexed: 11/25/2022] Open
Abstract
To match the metabolic demands of the brain, mechanisms have evolved to couple neuronal activity to vasodilation, thus increasing local cerebral blood flow and delivery of oxygen and glucose to active neurons. Rather than relying on metabolic feedback signals such as the consumption of oxygen or glucose, the main signalling pathways rely on the release of vasoactive molecules by neurons and astrocytes, which act on contractile cells. Vascular smooth muscle cells and pericytes are the contractile cells associated with arterioles and capillaries, respectively, which relax and induce vasodilation. Much progress has been made in understanding the complex signalling pathways of neurovascular coupling, but issues such as the contributions of capillary pericytes and astrocyte calcium signal remain contentious. Study of neurovascular coupling mechanisms is especially important as cerebral blood flow dysregulation is a prominent feature of Alzheimer’s disease. In this article we will discuss developments and controversies in the understanding of neurovascular coupling and finish by discussing current knowledge concerning neurovascular uncoupling in Alzheimer’s disease.
Collapse
Affiliation(s)
- Winston M Zhu
- Oxford Medical School, University of Oxford, Oxford, UK
| | - Ain Neuhaus
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Daniel J Beard
- Acute Stroke Programme, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.,School of Biomedical Sciences and Pharmacy, University of Newcastle, Newcastle, Australia
| | - Brad A Sutherland
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Australia
| | - Gabriele C DeLuca
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| |
Collapse
|
16
|
Thomas KR, Weigand AJ, Cota IH, Edmonds EC, Wierenga CE, Bondi MW, Bangen KJ. Intrusion errors moderate the relationship between blood glucose and regional cerebral blood flow in cognitively unimpaired older adults. Brain Imaging Behav 2022; 16:219-227. [PMID: 34415491 PMCID: PMC8825619 DOI: 10.1007/s11682-021-00495-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2021] [Indexed: 02/03/2023]
Abstract
Regional cerebral blood flow (CBF) has a complex relationship with cognitive functioning such that cognitively unimpaired individuals at risk for Alzheimer's disease (AD) may show regional hyperperfusion, while those with cognitive impairment typically show hypoperfusion. Diabetes and word-list intrusion errors are both linked to greater risk of cognitive decline and dementia. Our study examined associations between fasting blood glucose, word-list intrusion errors, and regional CBF. 113 cognitively unimpaired older adults had arterial spin labeling MRI to measure CBF in a priori AD vulnerable regions: medial temporal lobe (MTL), inferior parietal lobe (IPL), precuneus, medial orbitofrontal cortex (mOFC), and pericalcarine (control region). Hierarchical linear regressions, adjusting for demographics, vascular risk, and reference CBF region, examined the main effect of blood glucose on regional CBF as well as whether intrusions moderated this relationship. Higher glucose was associated with higher CBF in the precuneus (β = .134, 95% CI = .007 to .261, p = .039), IPL (β = .173, 95% CI = .072 to .276, p = .001), and mOFC (β = .182, 95% CI = .047 to .320, p = .009). There was no main effect of intrusions on CBF across regions. However, the glucose x intrusions interaction was significant such that having higher glucose levels and more intrusion errors was associated with reduced CBF in the MTL (β = -.186, 95% CI = -.334 to -.040, p = .013) and precuneus (β = -.146, 95% CI = -.273 to -.022, p = .022). These findings may reflect early neurovascular dysregulation, whereby higher CBF is needed to maintain unimpaired cognition in individuals with higher glucose levels. However, lower regional CBF in unimpaired participants with both higher glucose and more intrusions suggests a failure in this early compensatory mechanism that may signal a decrease in neural activity in AD vulnerable regions.
Collapse
Affiliation(s)
- Kelsey R Thomas
- Research Service, VA San Diego Healthcare System, 3350 La Jolla Village Drive (151), San Diego, CA, USA.
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA.
| | - Alexandra J Weigand
- San Diego State University/University of California, San Diego Joint Doctoral Program in Clinical Psychology, San Diego, CA, USA
| | - Isabel H Cota
- Research Service, VA San Diego Healthcare System, 3350 La Jolla Village Drive (151), San Diego, CA, USA
| | - Emily C Edmonds
- Research Service, VA San Diego Healthcare System, 3350 La Jolla Village Drive (151), San Diego, CA, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Christina E Wierenga
- Research Service, VA San Diego Healthcare System, 3350 La Jolla Village Drive (151), San Diego, CA, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Mark W Bondi
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
- Psychology Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - Katherine J Bangen
- Research Service, VA San Diego Healthcare System, 3350 La Jolla Village Drive (151), San Diego, CA, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
17
|
Pearce AM, Marr C, Dewar M, Gow AJ. Apolipoprotein E Genotype Moderation of the Association Between Physical Activity and Brain Health. A Systematic Review and Meta-Analysis. Front Aging Neurosci 2022; 13:815439. [PMID: 35153725 PMCID: PMC8833849 DOI: 10.3389/fnagi.2021.815439] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 12/17/2021] [Indexed: 12/12/2022] Open
Abstract
Introduction Possession of one or two e4 alleles of the apolipoprotein E (APOE) gene is associated with cognitive decline and dementia risk. Some evidence suggests that physical activity may benefit carriers of the e4 allele differently. Method We conducted a systematic review and meta-analysis of studies which assessed APOE differences in the association between physical activity and: lipid profile, Alzheimer's disease pathology, brain structure and brain function in healthy adults. Searches were carried out in PubMed, SCOPUS, Web of Science and PsycInfo. Results Thirty studies were included from 4,896 papers screened. Carriers of the e4 allele gained the same benefit from physical activity as non-carriers on most outcomes. For brain activation, e4 carriers appeared to gain a greater benefit from physical activity on task-related and resting-state activation and resting-state functional connectivity compared to non-carriers. Post-hoc analysis identified possible compensatory mechanisms allowing e4 carriers to maintain cognitive function. Discussion Though there is evidence suggesting physical activity may benefit e4 carriers differently compared to non-carriers, this may vary by the specific brain health outcome, perhaps limited to brain activation. Further research is required to confirm these findings and elucidate the mechanisms.
Collapse
|
18
|
Liu XJ, Che P, Xing M, Tian XB, Gao C, Li X, Zhang N. Cerebral Hemodynamics and Carotid Atherosclerosis in Patients With Subcortical Ischemic Vascular Dementia. Front Aging Neurosci 2021; 13:741881. [PMID: 34880744 PMCID: PMC8645960 DOI: 10.3389/fnagi.2021.741881] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/21/2021] [Indexed: 11/13/2022] Open
Abstract
A growing body of evidence indicates that atherosclerosis is correlated with cerebral small vessel disease and contributes to cognitive decline. This study aimed to explore the characteristics and contributions of intracranial hemodynamics and carotid atherosclerosis to cognitive dysfunction in subjects with subcortical ischemic vascular dementia (SIVD). Notably, 44 patients with SIVD, 30 patients with Alzheimer's disease (AD), and 30 healthy controls (HCs) were recruited from our longitudinal MRI study for AD and SIVD (ChiCTR1900027943). The cerebral mean flow velocity (MFV) and pulsatility index (PI) of both anterior and posterior circulations, artery plaque, and lumen diameter in carotid arteries were investigated using transcranial Doppler and carotid ultrasound, respectively. Their correlations with cognitive function were analyzed in patients with dementia. Decreased MFV and increased PI were found in patients with SIVD and AD. Patients with SIVD showed lower MFV and higher PI in the bilateral posterior cerebral arteries compared to patients with AD. Increases in lumen diameter, number of arteries with plaque, and total carotid plaque score were found in patients with SIVD. The Mini-Mental State Examination score was positively correlated with the MFV and negatively correlated with the PI of most major cerebral arteries, while it was negatively correlated with the lumen diameter of the common carotid artery, number of arteries with plaque, and total carotid plaque score in patients with dementia. There were also correlations between these parameters of some arteries and memory and executive function. Our results provide additional evidence suggesting that the pathological changes in macrovascular structure and function are correlated with cognitive impairment in dementia patients with SIVD and to a lesser extent AD.
Collapse
Affiliation(s)
- Xiao-Jiao Liu
- Department of Neurology, Tianjin Medical University General Hospital Airport Site, Tianjin, China
| | - Ping Che
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Mengya Xing
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiao-Bing Tian
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Chunli Gao
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiuyan Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Nan Zhang
- Department of Neurology, Tianjin Medical University General Hospital Airport Site, Tianjin, China.,Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
19
|
Dounavi ME, Low A, McKiernan EF, Mak E, Muniz-Terrera G, Ritchie K, Ritchie CW, Su L, O’Brien JT. Evidence of cerebral hemodynamic dysregulation in middle-aged APOE ε4 carriers: The PREVENT-Dementia study. J Cereb Blood Flow Metab 2021; 41:2844-2855. [PMID: 34078163 PMCID: PMC8543665 DOI: 10.1177/0271678x211020863] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 04/14/2021] [Accepted: 04/19/2021] [Indexed: 11/22/2022]
Abstract
Accumulating evidence suggests vascular dysregulation in preclinical Alzheimer's disease. In this study, cerebral hemodynamics and their coupling with cognition in middle-aged apolipoprotein ε4 carriers (APOEε4+) were investigated. Longitudinal 3 T T1-weighted and arterial spin labelling MRI data from 158 participants (40-59 years old) in the PREVENT-Dementia study were analysed (125 two-year follow-up). Cognition was evaluated using the COGNITO battery. Cerebral blood flow (CBF) and cerebrovascular resistance index (CVRi) were quantified for the flow territories of the anterior, middle and posterior cerebral arteries. CBF was corrected for underlying atrophy and individual hematocrit. Hemodynamic measures were the dependent variables in linear regression models, with age, sex, years of education and APOEε4 carriership as predictors. Further analyses were conducted with cognitive outcomes as dependent variables, using the same model as before with additional APOEε4 × hemodynamics interactions. At baseline, APOEε4+ showed increased CBF and decreased CVRi compared to non-carriers in the anterior and middle cerebral arteries, suggestive of potential vasodilation. Hemodynamic changes were similar between groups. Interaction analysis revealed positive associations between CBF changes and performance changes in delayed recall (for APOEε4 non-carriers) and verbal fluency (for APOEε4 carriers) cognitive tests. These observations are consistent with neurovascular dysregulation in middle-aged APOEε4+.
Collapse
Affiliation(s)
- Maria-Eleni Dounavi
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Audrey Low
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Elizabeth F McKiernan
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - Elijah Mak
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | | | - Karen Ritchie
- Centre for Dementia Prevention, University of Edinburgh, Edinburgh, UK
- INSERM, Montpellier, France
| | - Craig W Ritchie
- Centre for Dementia Prevention, University of Edinburgh, Edinburgh, UK
| | - Li Su
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - John T. O’Brien
- Department of Psychiatry, School of Clinical Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
20
|
Wang R, Oh JM, Motovylyak A, Ma Y, Sager MA, Rowley HA, Johnson KM, Gallagher CL, Carlsson CM, Bendlin BB, Johnson SC, Asthana S, Eisenmenger L, Okonkwo OC. Impact of sex and APOE ε4 on age-related cerebral perfusion trajectories in cognitively asymptomatic middle-aged and older adults: A longitudinal study. J Cereb Blood Flow Metab 2021; 41:3016-3027. [PMID: 34102919 PMCID: PMC8545048 DOI: 10.1177/0271678x211021313] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 03/16/2021] [Accepted: 03/20/2021] [Indexed: 01/03/2023]
Abstract
Cerebral hypoperfusion is thought to contribute to cognitive decline in Alzheimer's disease, but the natural trajectory of cerebral perfusion in cognitively healthy adults has not been well-studied. This longitudinal study is consisted of 950 participants (40-89 years), who were cognitively unimpaired at their first visit. We investigated the age-related changes in cerebral perfusion, and their associations with APOE-genotype, biological sex, and cardiometabolic measurements. During the follow-up period (range 0.13-8.24 years), increasing age was significantly associated with decreasing cerebral perfusion, in total gray-matter (β=-1.43), hippocampus (-1.25), superior frontal gyrus (-1.70), middle frontal gyrus (-1.99), posterior cingulate (-2.46), and precuneus (-2.14), with all P-values < 0.01. Compared with male-ɛ4 carriers, female-ɛ4 carriers showed a faster decline in global and regional cerebral perfusion with increasing age, whereas the age-related decline in cerebral perfusion was similar between male- and female-ɛ4 non-carriers. Worse cardiometabolic profile (i.e., increased blood pressure, body mass index, total cholesterol, and blood glucose) was associated with lower cerebral perfusion at all the visits. When time-varying cardiometabolic measurements were adjusted in the model, the synergistic effect of sex and APOE-ɛ4 on age-related cerebral perfusion-trajectories became largely attenuated. Our findings demonstrate that APOE-genotype and sex interactively impact cerebral perfusion-trajectories in mid- to late-life. This effect may be partially explained by cardiometabolic alterations.
Collapse
Affiliation(s)
- Rui Wang
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- The Swedish School of Sport and Health Science, GIH, Stockholm, Sweden
- Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Jennifer M Oh
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Alice Motovylyak
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Yue Ma
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Mark A Sager
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Howard A Rowley
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Kevin M Johnson
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Catherine L Gallagher
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Cynthia M Carlsson
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Barbara B Bendlin
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sanjay Asthana
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Laura Eisenmenger
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Ozioma C Okonkwo
- Wisconsin Alzheimer’s Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
- Geriatric Research Education and Clinical Center, William S. Middleton Memorial Veterans Hospital, Madison, WI, USA
- Wisconsin Alzheimer’s Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
21
|
Cao R, Tran A, Li J, Xu Z, Sun N, Zuo Z, Hu S. Hemodynamic and oxygen-metabolic responses of the awake mouse brain to hypercapnia revealed by multi-parametric photoacoustic microscopy. J Cereb Blood Flow Metab 2021; 41:2628-2639. [PMID: 33899557 PMCID: PMC8504963 DOI: 10.1177/0271678x211010352] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 03/13/2021] [Accepted: 03/18/2021] [Indexed: 11/16/2022]
Abstract
A widely used cerebrovascular stimulus and common pathophysiologic condition, hypercapnia is of great interest in brain research. However, it remains controversial how hypercapnia affects brain hemodynamics and energy metabolism. By using multi-parametric photoacoustic microscopy, the multifaceted responses of the awake mouse brain to different levels of hypercapnia are investigated. Our results show significant and vessel type-dependent increases of the vessel diameter and blood flow in response to the hypercapnic challenges, along with a decrease in oxygen extraction fraction due to elevated venous blood oxygenation. Interestingly, the increased blood flow and decreased oxygen extraction are not commensurate with each other, which leads to reduced cerebral oxygen metabolism. Further, time-lapse imaging over 2-hour chronic hypercapnic challenges reveals that the structural, functional, and metabolic changes induced by severe hypercapnia (10% CO2) are not only more pronounced but more enduring than those induced by mild hypercapnia (5% CO2), indicating that the extent of brain's compensatory response to chronic hypercapnia is inversely related to the severity of the challenge. Offering quantitative, dynamic, and CO2 level-dependent insights into the hemodynamic and metabolic responses of the brain to hypercapnia, these findings might provide useful guidance to the application of hypercapnia in brain research.
Collapse
Affiliation(s)
- Rui Cao
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Angela Tran
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - Jun Li
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, USA
| | - Zhiqiang Xu
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Naidi Sun
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | - Zhiyi Zuo
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, USA
| | - Song Hu
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| |
Collapse
|
22
|
Correia SC, Moreira PI. Oxygen Sensing and Signaling in Alzheimer's Disease: A Breathtaking Story! Cell Mol Neurobiol 2021; 42:3-21. [PMID: 34510330 DOI: 10.1007/s10571-021-01148-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Accepted: 09/07/2021] [Indexed: 10/20/2022]
Abstract
Oxygen sensing and homeostasis is indispensable for the maintenance of brain structural and functional integrity. Under low-oxygen tension, the non-diseased brain has the ability to cope with hypoxia by triggering a homeostatic response governed by the highly conserved hypoxia-inducible family (HIF) of transcription factors. With the advent of advanced neuroimaging tools, it is now recognized that cerebral hypoperfusion, and consequently hypoxia, is a consistent feature along the Alzheimer's disease (AD) continuum. Of note, the reduction in cerebral blood flow and tissue oxygenation detected during the prodromal phases of AD, drastically aggravates as disease progresses. Within this scenario a fundamental question arises: How HIF-driven homeostatic brain response to hypoxia "behaves" during the AD continuum? In this sense, the present review is aimed to critically discuss and summarize the current knowledge regarding the involvement of hypoxia and HIF signaling in the onset and progression of AD pathology. Importantly, the promises and challenges of non-pharmacological and pharmacological strategies aimed to target hypoxia will be discussed as a new "hope" to prevent and/or postpone the neurodegenerative events that occur in the AD brain.
Collapse
Affiliation(s)
- Sónia C Correia
- CNC - Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, Rua Larga, Polo I, 1st Floor, 3004-504, Coimbra, Portugal. .,CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal. .,Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal.
| | - Paula I Moreira
- CNC - Center for Neuroscience and Cell Biology, Faculty of Medicine, University of Coimbra, Rua Larga, Polo I, 1st Floor, 3004-504, Coimbra, Portugal.,CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.,Laboratory of Physiology, Faculty of Medicine, University of Coimbra, 3000-548, Coimbra, Portugal
| |
Collapse
|
23
|
Lotan R, Ganmore I, Livny A, Itzhaki N, Waserman M, Shelly S, Zacharia M, Moshier E, Uribarri J, Beisswenger P, Cai W, Troen AM, Beeri MS. Effect of Advanced Glycation End Products on Cognition in Older Adults with Type 2 Diabetes: Results from a Pilot Clinical Trial. J Alzheimers Dis 2021; 82:1785-1795. [PMID: 34250935 DOI: 10.3233/jad-210131] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
BACKGROUND Dietary advanced glycation end-products (AGEs) are linked to cognitive decline. However, clinical trials have not tested the effect of AGEs on cognition in older adults. OBJECTIVE The aim of the current pilot trial was to examine the feasibility of an intervention to reduce dietary AGEs on cognition and on cerebral blood flow (CBF). METHODS The design is a pilot randomized controlled trial of dietary AGEs reduction in older adults with type 2 diabetes. Seventy-five participants were randomized to two arms. The control arm received standard of care (SOC) guidelines for good glycemic control; the intervention arm, in addition to SOC guidelines, were instructed to reduce their dietary AGEs intake. Global cognition and CBF were assessed at baseline and after 6 months of intervention. RESULTS At baseline, we found a reverse association between AGEs and cognitive functioning, possibly reflecting the long-term toxicity of AGEs on the brain. There was a significant improvement in global cognition at 6 months in both the intervention and SOC groups which was more prominent in participants with mild cognitive impairment. We also found that at baseline, higher AGEs were associated with increased CBF in the left inferior parietal cortex; however, 6 months of the AGEs lowering intervention did not affect CBF levels, despite lowering AGEs exposure in blood. CONCLUSION The current pilot trial focused on the feasibility and methodology of intervening through diet to reduce AGEs in older adults with type 2 diabetes. Our results suggest that participants with mild cognitive impairment may benefit from an intensive dietary intervention.
Collapse
Affiliation(s)
- Roni Lotan
- The Nutrition and Brain Health Laboratory, The Institute of Biochemistry, Food and Nutrition Science, The Robert H. Smith Faculty of Agriculture Food and the Environment, The Hebrew University of Jerusalem, Rehovot, Israel.,The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel-Hashomer, Israel
| | - Ithamar Ganmore
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel-Hashomer, Israel.,Memory Clinic, Sheba Medical Center, Tel Hashomer, Israel.,Neurology department, Sheba Medical Center, Tel Hashomer, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Abigail Livny
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel-Hashomer, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Division of Diagnostic Imaging, Sheba Medical Center, Tel-Hashomer, Israel
| | - Nofar Itzhaki
- Division of Diagnostic Imaging, Sheba Medical Center, Tel-Hashomer, Israel
| | - Mark Waserman
- Division of Diagnostic Imaging, Sheba Medical Center, Tel-Hashomer, Israel
| | - Shahar Shelly
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel-Hashomer, Israel.,Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Moran Zacharia
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel-Hashomer, Israel
| | - Erin Moshier
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jaime Uribarri
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Weijing Cai
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aron M Troen
- The Nutrition and Brain Health Laboratory, The Institute of Biochemistry, Food and Nutrition Science, The Robert H. Smith Faculty of Agriculture Food and the Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Michal Schnaider Beeri
- The Joseph Sagol Neuroscience Center, Sheba Medical Center, Tel-Hashomer, Israel.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
24
|
Zhang H, Chiu PW, Ip I, Liu T, Wong GHY, Song YQ, Wong SWH, Herrup K, Mak HKF. Asymmetric left-right hippocampal glutamatergic modulation of cognitive control in ApoE-isoform subjects is unrelated to neuroinflammation. Eur J Neurosci 2021; 54:5310-5326. [PMID: 34309092 PMCID: PMC9290961 DOI: 10.1111/ejn.15399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 06/03/2021] [Accepted: 07/21/2021] [Indexed: 11/28/2022]
Abstract
The glutamatergic cycle is essential in modulating memory processing by the hippocampal circuitry. Our combined proton magnetic resonance spectroscopy (1H‐MRS) and task‐based functional magnetic resonance imaging (fMRI) study (using face‐name paired‐associates encoding and retrieval task) of a cognitively normal cohort of 67 healthy adults (18 ApoE4 carriers and 49 non‐ApoE4 carriers) found altered patterns of relationships between glutamatergic‐modulated synaptic signalling and neuronal activity or functional hyperaemia in the ApoE4 isoforms. Our study highlighted the asymmetric left–right hippocampal glutamatergic system in modulating neuronal activities in ApoE4 carriers versus non‐carriers. Such brain differentiation might be developmental cognitive advantages or compensatory due to impaired synaptic integrity and plasticity in ApoE4 carriers. As there was no difference in myoinositol levels measured by MRS between the ApoE4 and non‐ApoE4 subgroups, the mechanism is unlikely to be a response to neuroinflammation.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Diagnostic Radiology, The University of Hong Kong, Hong Kong.,Alzheimer's Disease Research Network, The University of Hong Kong, Hong Kong
| | - Pui Wai Chiu
- Department of Diagnostic Radiology, The University of Hong Kong, Hong Kong.,State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong
| | - Isaac Ip
- Department of Educational Psychology, Chinese University of Hong Kong, Hong Kong
| | - Tianyin Liu
- Department of Social Work and Administration, The University of Hong Kong, Hong Kong
| | - Gloria Hoi Yan Wong
- Department of Social Work and Administration, The University of Hong Kong, Hong Kong
| | - You-Qiang Song
- School of Biomedical Sciences, The University of Hong Kong, Hong Kong
| | - Savio Wai Ho Wong
- Department of Educational Psychology, Chinese University of Hong Kong, Hong Kong
| | - Karl Herrup
- Alzheimer Disease Research Centre, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Henry Ka Fung Mak
- Department of Diagnostic Radiology, The University of Hong Kong, Hong Kong.,Alzheimer's Disease Research Network, The University of Hong Kong, Hong Kong.,State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong
| |
Collapse
|
25
|
Memel M, Staffaroni AM, Cobigo Y, Casaletto KB, Fonseca C, Bettcher BM, Yassa MA, Elahi FM, Wolf A, Rosen HJ, Kramer JH. APOE moderates the effect of hippocampal blood flow on memory pattern separation in clinically normal older adults. Hippocampus 2021; 31:845-857. [PMID: 33835624 PMCID: PMC8295213 DOI: 10.1002/hipo.23327] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Revised: 01/25/2021] [Accepted: 03/07/2021] [Indexed: 11/10/2022]
Abstract
Pattern separation, the ability to differentiate new information from previously experienced similar information, is highly sensitive to hippocampal structure and function and declines with age. Functional MRI studies have demonstrated hippocampal hyperactivation in older adults compared to young, with greater task-related activation associated with worse pattern separation performance. The current study was designed to determine whether pattern separation was sensitive to differences in task-free hippocampal cerebral blood flow (CBF) in 130 functionally intact older adults. Given prior evidence that apolipoprotein E e4 (APOE e4) status moderates the relationship between CBF and episodic memory, we predicted a stronger negative relationship between hippocampal CBF and pattern separation in APOE e4 carriers. An interaction between APOE group and right hippocampal CBF was present, such that greater right hippocampal CBF was related to better lure discrimination in noncarriers, whereas the effect reversed directionality in e4 carriers. These findings suggest that neurovascular changes in the medial temporal lobe may underlie memory deficits in cognitively normal older adults who are APOE e4 carriers.
Collapse
Affiliation(s)
- Molly Memel
- San Francisco VA Medical Center, San Francisco, California
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California
| | - Adam M. Staffaroni
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California
| | - Yann Cobigo
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California
| | - Kaitlin B. Casaletto
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California
| | - Corrina Fonseca
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California
| | - Brianne M. Bettcher
- Department of Neurology, University of Colorado Anschutz Medical Campus, CU Alzheimer’s and Cognition Center, Aurora, Colorado
| | - Michael A. Yassa
- Department of Neurobiology and Behavior and Center for the Neurobiology of Learning and Memory, University of California, Irvine, California
| | - Fanny M. Elahi
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California
| | - Amy Wolf
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California
| | - Howard J. Rosen
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California
| | - Joel H. Kramer
- Department of Neurology, Memory and Aging Center, University of California, San Francisco (UCSF), San Francisco, California
| |
Collapse
|
26
|
Bracko O, Cruz Hernández JC, Park L, Nishimura N, Schaffer CB. Causes and consequences of baseline cerebral blood flow reductions in Alzheimer's disease. J Cereb Blood Flow Metab 2021; 41:1501-1516. [PMID: 33444096 PMCID: PMC8221770 DOI: 10.1177/0271678x20982383] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/27/2020] [Accepted: 11/16/2020] [Indexed: 12/23/2022]
Abstract
Reductions of baseline cerebral blood flow (CBF) of ∼10-20% are a common symptom of Alzheimer's disease (AD) that appear early in disease progression and correlate with the severity of cognitive impairment. These CBF deficits are replicated in mouse models of AD and recent work shows that increasing baseline CBF can rapidly improve the performance of AD mice on short term memory tasks. Despite the potential role these data suggest for CBF reductions in causing cognitive symptoms and contributing to brain pathology in AD, there remains a poor understanding of the molecular and cellular mechanisms causing them. This review compiles data on CBF reductions and on the correlation of AD-related CBF deficits with disease comorbidities (e.g. cardiovascular and genetic risk factors) and outcomes (e.g. cognitive performance and brain pathology) from studies in both patients and mouse models, and discusses several potential mechanisms proposed to contribute to CBF reductions, based primarily on work in AD mouse models. Future research aimed at improving our understanding of the importance of and interplay between different mechanisms for CBF reduction, as well as at determining the role these mechanisms play in AD patients could guide the development of future therapies that target CBF reductions in AD.
Collapse
Affiliation(s)
- Oliver Bracko
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Jean C Cruz Hernández
- Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Laibaik Park
- Feil Family Brain and Mind Research Institute, Weill Cornell Medical College, New York, NY, USA
| | - Nozomi Nishimura
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Chris B Schaffer
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| |
Collapse
|
27
|
Kaufman CS, Morris JK, Vidoni ED, Burns JM, Billinger SA. Apolipoprotein E4 Moderates the Association Between Vascular Risk Factors and Brain Pathology. Alzheimer Dis Assoc Disord 2021; 35:223-229. [PMID: 33734100 PMCID: PMC8387316 DOI: 10.1097/wad.0000000000000442] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 01/18/2021] [Indexed: 01/06/2023]
Abstract
BACKGROUND The strongest genetic risk factor for late-onset Alzheimer disease (AD), Apolipoprotein E4 (APOE4), increases cardiovascular disease risk and may also act synergistically with vascular risk factors to contribute to AD pathogenesis. Here, we assess the interaction between APOE4 and vascular risk on cerebrovascular dysfunction and brain pathology. METHODS This is an observational study of cognitively normal older adults, which included positron emission tomography imaging and vascular risk factors. We measured beat-to-beat blood pressure and middle cerebral artery velocity at rest and during moderate-intensity exercise. Cerebrovascular measures included cerebrovascular conductance index and the cerebrovascular response to exercise. RESULTS There was a significant interaction between resting cerebrovascular conductance index and APOE4 carrier status on β-amyloid deposition (P=0.026), with poor conductance in the cerebrovasculature associated with elevated β-amyloid for the APOE4 carriers only. There was a significant interaction between non-high-density lipoprotein cholesterol and APOE4 carrier status (P=0.014), with elevated non-high-density lipoprotein cholesterol predicting a blunted cerebrovascular response to exercise in APOE4 carriers and the opposite relationship in noncarriers. CONCLUSIONS Both cerebral and peripheral vascular risk factors are preferentially associated with brain pathology in APOE4 carriers. These findings provide insight into pathogenic vascular risk mechanisms and target strategies to potentially delay AD onset.
Collapse
Affiliation(s)
- Carolyn S. Kaufman
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Jill K. Morris
- University of Kansas Alzheimer’s Disease Center, Fairway, KS, USA
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS
| | - Eric D. Vidoni
- University of Kansas Alzheimer’s Disease Center, Fairway, KS, USA
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS
| | - Jeffrey M. Burns
- University of Kansas Alzheimer’s Disease Center, Fairway, KS, USA
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS
| | - Sandra A. Billinger
- Department of Molecular & Integrative Physiology, University of Kansas Medical Center, Kansas City, KS, USA
- Department of Neurology, University of Kansas Medical Center, Kansas City, KS
- Department of Physical Therapy & Rehabilitation Science, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
28
|
White D, John CS, Kucera A, Truver B, Lepping RJ, Kueck PJ, Lee P, Martin L, Billinger SA, Burns JM, Morris JK, Vidoni ED. A methodology for an acute exercise clinical trial called dementia risk and dynamic response to exercise. Sci Rep 2021; 11:12776. [PMID: 34140586 PMCID: PMC8211849 DOI: 10.1038/s41598-021-92177-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 06/04/2021] [Indexed: 01/21/2023] Open
Abstract
Exercise likely has numerous benefits for brain and cognition. However, those benefits and their causes remain imprecisely defined. If the brain does benefit from exercise it does so primarily through cumulative brief, "acute" exposures over a lifetime. The Dementia Risk and Dynamic Response to Exercise (DYNAMIC) clinical trial seeks to characterize the acute exercise response in cerebral perfusion, and circulating neurotrophic factors in older adults with and without the apolipoprotein e4 genotype (APOE4), the strongest genetic predictor of sporadic, late onset Alzheimer's disease. DYNAMIC will enroll 60 older adults into a single moderate intensity bout of exercise intervention, measuring pre- and post-exercise cerebral blood flow (CBF) using arterial spin labeling, and neurotrophic factors. We expect that APOE4 carriers will have poor CBF regulation, i.e. slower return to baseline perfusion after exercise, and will demonstrate blunted neurotrophic response to exercise, with concentrations of neurotrophic factors positively correlating with CBF regulation. Preliminary findings on 7 older adults and 9 younger adults demonstrate that the experimental method can capture CBF and neurotrophic response over a time course. This methodology will provide important insight into acute exercise response and potential directions for clinical trial outcomes.ClinicalTrials.gov NCT04009629, Registered 05/07/2019.
Collapse
Affiliation(s)
- Dreu White
- University of Kansas Medical Center, Kansas City, KS, USA
| | - Casey S John
- University of Kansas Medical Center, Kansas City, KS, USA
| | - Ashley Kucera
- American Academy of Family Physicians, Leawood, KS, USA
| | - Bryce Truver
- University of Kansas Medical Center, Kansas City, KS, USA
| | | | - Paul J Kueck
- University of Kansas Medical Center, Kansas City, KS, USA
| | - Phil Lee
- University of Kansas Medical Center, Kansas City, KS, USA
| | - Laura Martin
- University of Kansas Medical Center, Kansas City, KS, USA
| | | | | | - Jill K Morris
- University of Kansas Medical Center, Kansas City, KS, USA
| | - Eric D Vidoni
- University of Kansas Medical Center, Kansas City, KS, USA.
- University of Kansas Alzheimer's Disease Center, 4350 Shawnee Mission Parkway, Fairway, KS, 60205, MS6002, USA.
| |
Collapse
|
29
|
Duan W, Sehrawat P, Balachandrasekaran A, Bhumkar AB, Boraste PB, Becker JT, Kuller LH, Lopez OL, Gach HM, Dai W. Cerebral Blood Flow Is Associated with Diagnostic Class and Cognitive Decline in Alzheimer's Disease. J Alzheimers Dis 2021; 76:1103-1120. [PMID: 32597803 DOI: 10.3233/jad-200034] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Reliable cerebral blood flow (CBF) biomarkers using a noninvasive imaging technique are sought to facilitate early diagnosis and intervention in early Alzheimer's disease (AD). OBJECTIVE We aim to identify brain regions in which CBF values are affected and related to cognitive decline in early AD using a large cohort. METHODS Perfusion MRIs using continuous arterial spin labeling were acquired at 1.5 T in 58 normal controls (NC), 50 mild cognitive impairments (MCI), and 40 AD subjects from the Cardiovascular Health Study Cognition Study. Regional absolute CBF and normalized CBF (nCBF) values, without and with correction of partial volume effects, were compared across three groups. Association between regional CBF values and Modified Mini-Mental State Examination (3MSE) were investigated by multiple linear regression analyses adjusted for cardiovascular risk factors. RESULTS After correcting for partial volume effects and cardiovascular risk factors, ADs exhibited decreased nCBF with the strongest reduction in the bilateral posterior cingulate & precuneus region (p < 0.001) compared to NCs, and the strongest reduction in the bilateral superior medial frontal region (p < 0.001) compared to MCIs. MCIs exhibited the strongest nCBF decrease in the left hippocampus and nCBF increase in the right inferior frontal and insular region. The 3MSE scores within the symptomatic subjects were significantly associated with nCBF in the bilateral posterior and middle cingulate and parietal (p < 0.001), bilateral superior medial frontal (p < 0.001), bilateral temporoparietal (p < 0.02), and right hippocampus (p = 0.02) regions. CONCLUSION Noninvasive perfusion MRI can detect functional changes across diagnostic class and serve as a staging biomarker of cognitive status.
Collapse
Affiliation(s)
- Wenna Duan
- Department of Computer Science, State University of New York at Binghamton, Binghamton, NY, USA
| | - Parshant Sehrawat
- Department of Computer Science, State University of New York at Binghamton, Binghamton, NY, USA
| | | | - Ashish B Bhumkar
- Department of Computer Science, State University of New York at Binghamton, Binghamton, NY, USA
| | - Paresh B Boraste
- Department of Computer Science, State University of New York at Binghamton, Binghamton, NY, USA
| | - James T Becker
- Departments of Psychiatry, Psychology, and Neurology, University of Pittsburgh, PA, USA
| | - Lewis H Kuller
- Department of Epidemiology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Oscar L Lopez
- Departments of Neurology and Psychiatry, University of Pittsburgh, PA, USA
| | - H Michael Gach
- Departments of Radiation Oncology, Radiology, and Biomedical Engineering, Washington University, Saint Louis, MO, USA
| | - Weiying Dai
- Department of Computer Science, State University of New York at Binghamton, Binghamton, NY, USA
| |
Collapse
|
30
|
Clark LR, Zuelsdorff M, Norton D, Johnson SC, Wyman MF, Hancock LM, Carlsson CM, Asthana S, Flowers-Benton S, Gleason CE, Johnson HM. Association of Cardiovascular Risk Factors with Cerebral Perfusion in Whites and African Americans. J Alzheimers Dis 2021; 75:649-660. [PMID: 32310160 DOI: 10.3233/jad-190360] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
BACKGROUND Midlife cardiovascular risk factors increase risk for Alzheimer's disease (AD). Despite disproportionately high cardiovascular disease and dementia rates, African Americans are under-represented in studies of AD risk and research-based guidance on targeting vascular risk factors is lacking. OBJECTIVE This study investigated relationships between specific cardiovascular risk factors and cerebral perfusion in White and African American adults enriched for AD risk. METHODS Participants included 397 cognitively unimpaired White (n = 330) and African American (n = 67) adults enrolled in the Wisconsin Alzheimer's Disease Research Center who underwent pseudo-continuous arterial spin labeling MRI. Multiple linear regression models examined independent relationships between cardiovascular risk factors and mean cerebral perfusion. Subsequent interaction and stratified models assessed the role for APOE genotype and race. RESULTS When risk factor p-values were FDR-adjusted, diastolic blood pressure was significantly associated with mean perfusion. Tobacco use, triglycerides, waist-to-hip ratio, and a composite risk score were not associated with perfusion. Without FDR adjustment, a relationship was also observed between perfusion and obesity, cholesterol, and fasting glucose. Neither APOE genotype nor race moderated relationships between risk factors and perfusion. CONCLUSION Higher diastolic blood pressure predicted lower perfusion more strongly than other cardiovascular risk factors. This relationship did not vary by racial group or genetic risk for AD, although the African American sample had greater vascular risk burden and lower perfusion rates. Our findings highlight the need to prioritize inclusion of underrepresented groups in neuroimaging studies and to continue exploring the link between modifiable risk factors, cerebrovascular health, and AD risk in underrepresented populations.
Collapse
Affiliation(s)
- Lindsay R Clark
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Geriatric Research Education and Clinical Center, William S Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Megan Zuelsdorff
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,University of Wisconsin School of Nursing, Madison, WI, USA
| | - Derek Norton
- Department of Biostatistics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Sterling C Johnson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Geriatric Research Education and Clinical Center, William S Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Mary F Wyman
- Geriatric Research Education and Clinical Center, William S Middleton Memorial Veterans Hospital, Madison, WI, USA.,Department of Psychiatry, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Laura M Hancock
- Department of Neurology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Cynthia M Carlsson
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Wisconsin Alzheimer's Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Geriatric Research Education and Clinical Center, William S Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Sanjay Asthana
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Geriatric Research Education and Clinical Center, William S Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Susan Flowers-Benton
- Department of Family Medicine and Community Health, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Carey E Gleason
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.,Geriatric Research Education and Clinical Center, William S Middleton Memorial Veterans Hospital, Madison, WI, USA
| | - Heather M Johnson
- Division of Cardiovascular Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
31
|
Thomas KR, Osuna JR, Weigand AJ, Edmonds EC, Clark AL, Holmqvist S, Cota IH, Wierenga CE, Bondi MW, Bangen KJ. Regional hyperperfusion in older adults with objectively-defined subtle cognitive decline. J Cereb Blood Flow Metab 2021; 41:1001-1012. [PMID: 32615887 PMCID: PMC8054731 DOI: 10.1177/0271678x20935171] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 04/21/2020] [Accepted: 05/16/2020] [Indexed: 11/16/2022]
Abstract
Although cerebral blood flow (CBF) alterations are associated with Alzheimer's disease (AD), CBF patterns across prodromal stages of AD remain unclear. Therefore, we investigated patterns of regional CBF in 162 Alzheimer's Disease Neuroimaging Initiative participants characterized as cognitively unimpaired (CU; n = 80), objectively-defined subtle cognitive decline (Obj-SCD; n = 31), or mild cognitive impairment (MCI; n = 51). Arterial spin labeling MRI quantified regional CBF in a priori regions of interest: hippocampus, inferior temporal gyrus, inferior parietal lobe, medial orbitofrontal cortex, and rostral middle frontal gyrus. Obj-SCD participants had increased hippocampal and inferior parietal CBF relative to CU and MCI participants and increased inferior temporal CBF relative to MCI participants. CU and MCI groups did not differ in hippocampal or inferior parietal CBF, but CU participants had increased inferior temporal CBF relative to MCI participants. There were no CBF group differences in the two frontal regions. Thus, we found an inverted-U pattern of CBF signal across prodromal AD stages in regions susceptible to early AD pathology. Hippocampal and inferior parietal hyperperfusion in Obj-SCD may reflect early neurovascular dysregulation, whereby higher CBF is needed to maintain cognitive functioning relative to MCI participants, yet is also reflective of early cognitive inefficiencies that distinguish Obj-SCD from CU participants.
Collapse
Affiliation(s)
- Kelsey R Thomas
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Jessica R Osuna
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Alexandra J Weigand
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
- San Diego State University/University of California, San Diego Joint Doctoral Program in Clinical Psychology, San Diego, CA, USA
| | - Emily C Edmonds
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Alexandra L Clark
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Sophia Holmqvist
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - Isabel H Cota
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - Christina E Wierenga
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | - Mark W Bondi
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
- Psychology Service, VA San Diego Healthcare System, San Diego, CA, USA
| | - Katherine J Bangen
- Research Service, VA San Diego Healthcare System, San Diego, CA, USA
- Department of Psychiatry, University of California, San Diego, La Jolla, CA, USA
| | | |
Collapse
|
32
|
Singer MB, Ringman JM, Chu Z, Zhou X, Jiang X, Shahidzadeh A, Wang RK, Kashani AH. Abnormal retinal capillary blood flow in autosomal dominant Alzheimer's disease. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2021; 13:e12162. [PMID: 33728371 PMCID: PMC7931411 DOI: 10.1002/dad2.12162] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/07/2021] [Accepted: 01/12/2021] [Indexed: 02/03/2023]
Abstract
INTRODUCTION This study characterizes retinal capillary blood flow in subjects with autosomal dominant Alzheimer's disease (ADAD)-causing mutations. METHODS Carriers of PSEN1 or APP mutations were prospectively recruited and split into early-stage (ES) and late-stage (LS) groups. Controls were normal subjects and non-carriers from the at-risk group. Capillary blood flow was quantified using an optical coherence tomography angiography-based measure of erythrocyte flux through capillary segments. Statistical analyses were adjusted for correlation between two eyes of the same subject. RESULTS ES carriers had significantly greater capillary blood flow than controls and LS carriers. ES and LS carriers had significantly greater capillary blood flow heterogeneity than controls. There was no difference between capillary blood flow of LS carriers and controls. DISCUSSION ES ADAD carriers demonstrate increased retinal capillary blood flow and flow heterogeneity compared to controls. These findings support the hypothesis that increased perfusion is a pathophysiologic feature of presymptomatic stages of ADAD.
Collapse
Affiliation(s)
- Maxwell B. Singer
- Department of OphthalmologyUSC Roski Eye InstituteKeck School of Medicine of the University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - John M. Ringman
- Department of NeurologyKeck School of Medicine of the University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Zhongdi Chu
- Department of BioengineeringUniversity of WashingtonSeattleWashingtonUSA
| | - Xiao Zhou
- Department of BioengineeringUniversity of WashingtonSeattleWashingtonUSA
| | - Xuejuan Jiang
- Department of OphthalmologyUSC Roski Eye InstituteKeck School of Medicine of the University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Anoush Shahidzadeh
- Department of OphthalmologyUSC Roski Eye InstituteKeck School of Medicine of the University of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Ruikang K. Wang
- Department of BioengineeringUniversity of WashingtonSeattleWashingtonUSA
- Department of OphthalmologyUniversity of WashingtonSeattleWashingtonUSA
| | - Amir H. Kashani
- Department of OphthalmologyUSC Roski Eye InstituteKeck School of Medicine of the University of Southern CaliforniaLos AngelesCaliforniaUSA
- Department of NeurologyKeck School of Medicine of the University of Southern CaliforniaLos AngelesCaliforniaUSA
- USC Ginsburg Institute for Biomedical TherapeuticsKeck School of Medicine of the University of Southern CaliforniaLos AngelesCaliforniaUSA
- Department of Biomedical EngineeringUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
33
|
Steinman J, Sun HS, Feng ZP. Microvascular Alterations in Alzheimer's Disease. Front Cell Neurosci 2021; 14:618986. [PMID: 33536876 PMCID: PMC7849053 DOI: 10.3389/fncel.2020.618986] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/17/2020] [Indexed: 12/27/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder associated with continual decline in cognition and ability to perform routine functions such as remembering familiar places or understanding speech. For decades, amyloid beta (Aβ) was viewed as the driver of AD, triggering neurodegenerative processes such as inflammation and formation of neurofibrillary tangles (NFTs). This approach has not yielded therapeutics that cure the disease or significant improvements in long-term cognition through removal of plaques and Aβ oligomers. Some researchers propose alternate mechanisms that drive AD or act in conjunction with amyloid to promote neurodegeneration. This review summarizes the status of AD research and examines research directions including and beyond Aβ, such as tau, inflammation, and protein clearance mechanisms. The effect of aging on microvasculature is highlighted, including its contribution to reduced blood flow that impairs cognition. Microvascular alterations observed in AD are outlined, emphasizing imaging studies of capillary malfunction. The review concludes with a discussion of two therapies to protect tissue without directly targeting Aβ for removal: (1) administration of growth factors to promote vascular recovery in AD; (2) inhibiting activity of a calcium-permeable ion channels to reduce microglial activation and restore cerebral vascular function.
Collapse
Affiliation(s)
- Joe Steinman
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Hong-Shuo Sun
- Department of Physiology, University of Toronto, Toronto, ON, Canada.,Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Zhong-Ping Feng
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
34
|
Blanken AE, Dutt S, Li Y, Nation DA. Disentangling Heterogeneity in Alzheimer's Disease: Two Empirically-Derived Subtypes. J Alzheimers Dis 2020; 70:227-239. [PMID: 31177226 DOI: 10.3233/jad-190230] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Clinical-pathological Alzheimer's disease (AD) subtypes may help distill heterogeneity in patient presentation. To date, no studies have utilized neuropsychological and biological markers to identify preclinical subtypes with longitudinal stability. OBJECTIVE The objective of this study was to empirically derive AD endophenotypes using a combination of cognitive and biological markers. METHODS Hierarchical cluster analysis grouped dementia-free older adults using memory, executive and language abilities, and cerebrospinal fluid amyloid-β and phosphorylated tau. Brain volume differences, neuropsychological trajectory, and progression to dementia were compared, controlling for age, gender, education, and apolipoprotein E4 (ApoE4). RESULTS Subgroups included asymptomatic-normal (n = 653) with unimpaired cognition and subthreshold biomarkers, typical AD (TAD; n = 191) showing marked memory decline, high ApoE4 rates and abnormal biomarkers, and atypical AD (AAD; n = 132) with widespread cognitive decline, intermediate biomarker levels, older age, less education and more white matter lesions. Cognitive profiles showed longitudinal stability with corresponding patterns of cortical atrophy, despite nearly identical rates of progression to AD dementia. CONCLUSION Two clinical-pathological AD subtypes are identified with potential implications for preventative efforts.
Collapse
Affiliation(s)
- Anna E Blanken
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Shubir Dutt
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Yanrong Li
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | - Daniel A Nation
- Department of Psychology, University of Southern California, Los Angeles, CA, USA
| | | |
Collapse
|
35
|
Archila-Meléndez ME, Sorg C, Preibisch C. Modeling the impact of neurovascular coupling impairments on BOLD-based functional connectivity at rest. Neuroimage 2020; 218:116871. [DOI: 10.1016/j.neuroimage.2020.116871] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 04/17/2020] [Accepted: 04/20/2020] [Indexed: 12/12/2022] Open
|
36
|
McKiernan EF, Mak E, Dounavi ME, Wells K, Ritchie C, Williams G, Su L, O'Brien J. Regional hyperperfusion in cognitively normal APOE ε4 allele carriers in mid-life: analysis of ASL pilot data from the PREVENT-Dementia cohort. J Neurol Neurosurg Psychiatry 2020; 91:861-866. [PMID: 32586852 DOI: 10.1136/jnnp-2020-322924] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 05/01/2020] [Accepted: 05/27/2020] [Indexed: 01/28/2023]
Abstract
BACKGROUND Regional cerebral hypoperfusion is characteristic of Alzheimer's disease (AD). Previous studies report conflicting findings in cognitively normal individuals at high risk of AD. Understanding early preclinical perfusion alterations may improve understanding of AD pathogenesis and lead to new biomarkers and treatment targets. METHODS 3T arterial spin labelling MRI scans from 162 participants in the PREVENT-Dementia cohort were analysed (cognitively normal participants aged 40-59, stratified by future dementia risk). Cerebral perfusion was compared vertex-wise according to APOE ε4 status and family history (FH). Correlations between individual perfusion, age and cognitive scores (COGNITO battery) were explored. RESULTS Regional hyperperfusion was found in APOE ε4+group (left cingulate and lateral frontal and parietal regions p<0.01, threshold-free cluster enhancement, TFCE) and in FH +group (left temporal and parietal regions p<0.01, TFCE). Perfusion did not correlate with cognitive test scores. CONCLUSIONS Regional cerebral hyperperfusion in individuals at increased risk of AD in mid-life may be a very early marker of functional brain change related to AD. Increased perfusion may reflect a functional 'compensation' mechanism, offsetting the effects of early neural damage or may itself be risk factor for accelerating spread of degenerative pathology.
Collapse
Affiliation(s)
| | - Elijah Mak
- Psychiatry, University of Cambridge, Cambridge, UK
| | | | - Katie Wells
- The Centre for Mental Health, Imperial College, London, UK
| | - Craig Ritchie
- Centre for Dementia Prevention, University of Edinburgh Centre for Clinical Brain Sciences, Edinburgh, Edinburgh, UK
| | - Guy Williams
- Wolfson Brain Imaging Centre, Cambridge University, Cambridge, UK
| | - Li Su
- Psychiatry, University of Cambridge, Cambridge, UK
| | - John O'Brien
- Psychiatry, University of Cambridge, Cambridge, UK
| |
Collapse
|
37
|
Knolle F, Garofalo S, Viviani R, Justicia A, Ermakova AO, Blank H, Williams GB, Arrondo G, Ramachandra P, Tudor-Sfetea C, Bunzeck N, Duezel E, Robbins TW, Barker RA, Murray GK. Altered subcortical emotional salience processing differentiates Parkinson's patients with and without psychotic symptoms. NEUROIMAGE-CLINICAL 2020; 27:102277. [PMID: 32540629 PMCID: PMC7298672 DOI: 10.1016/j.nicl.2020.102277] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 03/30/2020] [Accepted: 05/05/2020] [Indexed: 01/03/2023]
Abstract
Emotional salience processing differentiates PD patients with and without psychosis. Enhanced striatal, hippocampal and midbrain responses in PD patients with psychosis. Indication for ‘jumping to conclusions’ bias in the same PD patients with psychosis. Aberrant top-down and salience processing associated with PD psychosis. Similar deficits as proposed in ‘aberrant salience hypothesis’ of schizophrenia.
Objective Current research does not provide a clear explanation for why some patients with Parkinson’s Disease (PD) develop psychotic symptoms. The ‘aberrant salience hypothesis’ of psychosis has been influential and proposes that dopaminergic dysregulation leads to inappropriate attribution of salience to irrelevant/non-informative stimuli, facilitating the formation of hallucinations and delusions. The aim of this study is to investigate whether non-motivational salience is altered in PD patients and possibly linked to the development of psychotic symptoms. Methods We investigated salience processing in 14 PD patients with psychotic symptoms, 23 PD patients without psychotic symptoms and 19 healthy controls. All patients were on dopaminergic medication for their PD. We examined emotional salience using a visual oddball fMRI paradigm that has been used to investigate early stages of schizophrenia spectrum psychosis, controlling for resting cerebral blood flow as assessed with arterial spin labelling fMRI. Results We found significant differences between patient groups in brain responses to emotional salience. PD patients with psychotic symptoms had enhanced brain responses in the striatum, dopaminergic midbrain, hippocampus and amygdala compared to patients without psychotic symptoms. PD patients with psychotic symptoms showed significant correlations between the levels of dopaminergic drugs they were taking and BOLD signalling, as well as psychotic symptom scores. Conclusion Our study suggests that enhanced signalling in the striatum, dopaminergic midbrain, the hippocampus and amygdala is associated with the development of psychotic symptoms in PD, in line with that proposed in the ‘aberrant salience hypothesis’ of psychosis in schizophrenia.
Collapse
Affiliation(s)
- F Knolle
- Department of Psychiatry, University of Cambridge, Cambridge, UK; Department of Neuroradiology, Technical University Munich, Munich, Germany.
| | - S Garofalo
- University of Bologna, Department of Psychology, Bologna, Italy
| | - R Viviani
- Institute of Psychology, University of Innsbruck, Innsbruck, Austria; Psychiatry and Psychotherapy Clinic III, University of Ulm, Ulm, Germany
| | - A Justicia
- Department of Psychiatry, University of Cambridge, Cambridge, UK; IMIM (Hospital del Mar Medical Research Institute), Centro de Investigación Biomédica en Red de Salud Mental (CIBERSAM), Barcelona, Spain
| | - A O Ermakova
- Faculty of Natural Sciences, Imperial College London, UK
| | - H Blank
- Institute of Systems Neuroscience, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - G B Williams
- Department of Clinical Neuroscience and WT-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - G Arrondo
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - P Ramachandra
- Cambridgeshire and Peterborough NHS Foundation Trust, Cambridge, UK
| | - C Tudor-Sfetea
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| | - N Bunzeck
- Institute of Psychology I, University of Lübeck, Lübeck, Germany
| | - E Duezel
- Otto-von-Guericke University Magdeburg, Institute of Cognitive Neurology and Dementia Research, Magdeburg, Germany; German Centre for Neurodegenerative Diseases (DZNE), Magdeburg, Germany
| | - T W Robbins
- Department of Psychology, University of Cambridge, Cambridge, UK
| | - R A Barker
- Department of Clinical Neuroscience and WT-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - G K Murray
- Department of Psychiatry, University of Cambridge, Cambridge, UK
| |
Collapse
|
38
|
de la Torre JC. Hemodynamic Instability in Heart Failure Intensifies Age-Dependent Cognitive Decline. J Alzheimers Dis 2020; 76:63-84. [PMID: 32444552 DOI: 10.3233/jad-200296] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
This review attempts to examine two key elements in the evolution of cognitive impairment in the elderly who develop heart failure. First, major left side heart parts can structurally and functionally deteriorate from aging wear and tear to provoke hemodynamic instability where heart failure worsens or is initiated; second, heart failure is a major inducer of cognitive impairment and Alzheimer's disease in the elderly. In heart failure, when the left ventricular myocardium of an elderly person does not properly contract, it cannot pump out adequate blood to the brain, raising the risk of cognitive impairment due to the intensification of chronic brain hypoperfusion. Chronic brain hypoperfusion originates from chronically reduced cardiac output which progresses as heart failure worsens. Other left ventricular heart parts, including atrium, valves, myocardium, and aorta can contribute to the physiological shortfall of cardiac output. It follows that hemodynamic instability and perfusion changes occurring from the aging heart's blood pumping deficiency will, in time, damage vulnerable brain cells linked to specific cognitive regulatory sites, diminishing neuronal energy metabolism to a level where progressive cognitive impairment is the outcome. Could cognitive impairment progress be reversed with a heart transplant? Evidence is presented detailing the errant hemodynamic pathways leading to cognitive impairment during aging as an offshoot of inefficient structural and functional heart parts and their contribution to heart failure.
Collapse
Affiliation(s)
- Jack C de la Torre
- Department of Psychology, University of Texas at Austin, Austin, TX, USA.,University of Valencia, Valencia, Spain
| |
Collapse
|
39
|
Abstract
It is a generally accepted observation that individuals act differently under stress. Recent task-based neuroimaging studies have shown that individuals under stress favor the intuitive and fast system over the deliberative and reflective system. In the present study, using a within-subjects design in thirty young adults, we examined whether and how acute social stress impacts regional neural activity in resting state. The results showed that stress induced lower coherence regional homogeneity (Cohe-ReHo) values in left hippocampus and right superior frontal gyrus, both of which are regions associated with deliberative decision making. Stress-induced cortisol change was significantly and positively correlated with the change in Cohe-ReHo value in the right midbrain, a region involved in habitual decision making. These results extend previous findings by demonstrating that stress modulates local synchrony in brain regions implicated in deliberative and intuitive decision making. Our findings might be useful in understanding the neural mechanisms underlying stress-related mental disorders.
Collapse
Affiliation(s)
- Jingjing Chang
- Guangdong Key Laboratory of Mental Health and Cognitive Science, Center for Studies of Psychological Application, School of Psychology, South China Normal University, Guangzhou, China
| | - Rongjun Yu
- Guangdong Key Laboratory of Mental Health and Cognitive Science, Center for Studies of Psychological Application, School of Psychology, South China Normal University, Guangzhou, China.
- Department of Psychology, National University of Singapore, 9 Arts Link, Singapore, 117570, Singapore.
| |
Collapse
|
40
|
Guo L, Li Z, Lyu J, Mei Y, Vardakis JC, Chen D, Han C, Lou X, Ventikos Y. On the Validation of a Multiple-Network Poroelastic Model Using Arterial Spin Labeling MRI Data. Front Comput Neurosci 2019; 13:60. [PMID: 31551742 PMCID: PMC6733888 DOI: 10.3389/fncom.2019.00060] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 08/19/2019] [Indexed: 02/05/2023] Open
Abstract
The Multiple-Network Poroelastic Theory (MPET) is a numerical model to characterize the transport of multiple fluid networks in the brain, which overcomes the problem of conducting separate analyses on individual fluid compartments and losing the interactions between tissue and fluids, in addition to the interaction between the different fluids themselves. In this paper, the blood perfusion results from MPET modeling are partially validated using cerebral blood flow (CBF) data obtained from arterial spin labeling (ASL) magnetic resonance imaging (MRI), which uses arterial blood water as an endogenous tracer to measure CBF. Two subjects—one healthy control and one patient with unilateral middle cerebral artery (MCA) stenosis are included in the validation test. The comparison shows several similarities between CBF data from ASL and blood perfusion results from MPET modeling, such as higher blood perfusion in the gray matter than in the white matter, higher perfusion in the periventricular region for both the healthy control and the patient, and asymmetric distribution of blood perfusion for the patient. Although the partial validation is mainly conducted in a qualitative way, it is one important step toward the full validation of the MPET model, which has the potential to be used as a testing bed for hypotheses and new theories in neuroscience research.
Collapse
Affiliation(s)
- Liwei Guo
- Department of Mechanical Engineering, University College London, London, United Kingdom
| | - Zeyan Li
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Jinhao Lyu
- Department of Radiology, Chinese PLA General Hospital, Beijing, China
| | - Yuqian Mei
- Department of Computer Science, INSIGNEO Institute, University of Sheffield, Sheffield, United Kingdom
| | - John C Vardakis
- Department of Mechanical Engineering, University College London, London, United Kingdom
| | - Duanduan Chen
- School of Life Science, Beijing Institute of Technology, Beijing, China
| | - Cong Han
- Department of Neurosurgery, The Fifth Medical Centre of PLA General Hospital, Beijing, China
| | - Xin Lou
- Department of Radiology, Chinese PLA General Hospital, Beijing, China
| | - Yiannis Ventikos
- Department of Mechanical Engineering, University College London, London, United Kingdom.,School of Life Science, Beijing Institute of Technology, Beijing, China
| |
Collapse
|
41
|
Rasmussen PM, Aamand R, Weitzberg E, Christiansen M, Østergaard L, Lund TE. APOE gene-dependent BOLD responses to a breath-hold across the adult lifespan. NEUROIMAGE-CLINICAL 2019; 24:101955. [PMID: 31408838 PMCID: PMC6699560 DOI: 10.1016/j.nicl.2019.101955] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 06/21/2019] [Accepted: 07/19/2019] [Indexed: 11/03/2022]
Abstract
Age and apolipoprotein E (APOE) e4 genotype are two of the strongest known risk factors for sporadic Alzheimer's disease (AD). Neuroimaging has shown hemodynamic response changes with age, in asymptomatic carriers of the APOE e4 allele, and in AD. In this study, we aimed to characterize and differentiate age- and APOE gene-specific hemodynamic changes to breath-hold and visual stimulation. A further aim was to study whether these responses were modulated by 3-day intake of nitrate, a nitric oxide (NO) source. The study was designed as a randomized, double-blinded, placebo-controlled crossover study, and the study cohort comprised 41 APOE e4 carriers (e3/e4 or e4/e4 genotype) and 40 non-carriers (e3/e3 genotype) aged 30-70 years at enrollment. The participants underwent two scanning sessions, each preceded by ingestion of sodium nitrate or sodium chloride (control). During functional magnetic resonance imaging (fMRI) sessions, participants performed two concurrent tasks; a breath-hold task to probe cerebrovascular reactivity and a visual stimulation task to evoke functional hyperemia, respectively. We found that the blood oxygenation level dependent (BOLD) hemodynamic response to breath-hold was altered in APOE e4 carriers relative to non-carriers. Mid-aged (50-60 years of age) e4 carriers exhibited a significantly increased peak time relative to mid-aged e3 carriers, and peak time for younger (30-40 years of age) e4 carriers was significantly shorter than that of mid-aged e4 carriers. The response width was significantly increased for e4 carriers. The response peak magnitude significantly decreased with age. For the visual stimulation task, we found age-related changes, with reduced response magnitude with age but no significant effect of APOE allele type. We found no effect of nitrate ingestion on BOLD responses evoked by the breath-hold and visual stimulation tasks. The APOE gene-dependent response to breath-hold may reflect NO-independent differences in vascular function.
Collapse
Affiliation(s)
- Peter M Rasmussen
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
| | - Rasmus Aamand
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Michael Christiansen
- Department for Congenital Disorders, Statens Serum Institut, Copenhagen, Denmark
| | - Leif Østergaard
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Torben E Lund
- Center of Functionally Integrative Neuroscience, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
42
|
Wang J, Peng G, Liu P, Tan X, Luo B. Regulating effect of CBF on memory in cognitively normal older adults with different ApoE genotype: the Alzheimer's Disease Neuroimaging Initiative (ADNI). Cogn Neurodyn 2019; 13:513-518. [PMID: 31741688 DOI: 10.1007/s11571-019-09536-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 04/07/2019] [Accepted: 04/25/2019] [Indexed: 12/16/2022] Open
Abstract
Apolipoprotein E (ApoE) ε4 allele and cerebral blood flow (CBF) changes are related to the increased risk of cognitive impairment independently. However, whether there are interactions between ApoE ε4 and CBF on memory performance in older adults with normal cognition remains unknown. This study determined whether the association between CBF and memory performance could be moderated by ApoE ε4 within a sample of cognitively normal older adults from the ADNI. 62 participants, including 23 with ApoE ε4 (ApoE ε4+) and 39 without ApoE ε4 (ApoE ε4-), underwent resting CBF measurement and memory testing. CBF was measured by arterial spin labeling MRI and memory performance was evaluated by the Rey Auditory Verbal Learning Test. By using linear regression models, CBF was negatively associated with memory function in ApoE ε4+ group, whereas positively in ApoE ε4- group by contrast. This study suggests that different CBF-memory relationships can be detected in cognitively normal ApoE ε4 carriers compared to ApoE ε4 non-carriers. Associations between hyperperfusion and worse memory performance in ApoE ε4 carriers may reflect vascular and/or cellular dysfunction.
Collapse
Affiliation(s)
- Junyang Wang
- 1Department of Neurology, The First Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, 310003 China
| | - Guoping Peng
- 1Department of Neurology, The First Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, 310003 China
| | - Ping Liu
- 1Department of Neurology, The First Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, 310003 China
| | - Xufei Tan
- 1Department of Neurology, The First Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, 310003 China
| | - Benyan Luo
- 1Department of Neurology, The First Affiliated Hospital of School of Medicine, Zhejiang University, Hangzhou, 310003 China.,Collaborative Innovation Center for Brain Science, Hangzhou, 310003 China
| | | |
Collapse
|
43
|
Govindpani K, McNamara LG, Smith NR, Vinnakota C, Waldvogel HJ, Faull RL, Kwakowsky A. Vascular Dysfunction in Alzheimer's Disease: A Prelude to the Pathological Process or a Consequence of It? J Clin Med 2019; 8:E651. [PMID: 31083442 PMCID: PMC6571853 DOI: 10.3390/jcm8050651] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 04/29/2019] [Accepted: 05/06/2019] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia. Despite decades of research following several theoretical and clinical lines, all existing treatments for the disorder are purely symptomatic. AD research has traditionally been focused on neuronal and glial dysfunction. Although there is a wealth of evidence pointing to a significant vascular component in the disease, this angle has been relatively poorly explored. In this review, we consider the various aspects of vascular dysfunction in AD, which has a significant impact on brain metabolism and homeostasis and the clearance of β-amyloid and other toxic metabolites. This may potentially precede the onset of the hallmark pathophysiological and cognitive symptoms of the disease. Pathological changes in vessel haemodynamics, angiogenesis, vascular cell function, vascular coverage, blood-brain barrier permeability and immune cell migration may be related to amyloid toxicity, oxidative stress and apolipoprotein E (APOE) genotype. These vascular deficits may in turn contribute to parenchymal amyloid deposition, neurotoxicity, glial activation and metabolic dysfunction in multiple cell types. A vicious feedback cycle ensues, with progressively worsening neuronal and vascular pathology through the course of the disease. Thus, a better appreciation for the importance of vascular dysfunction in AD may open new avenues for research and therapy.
Collapse
Affiliation(s)
- Karan Govindpani
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Laura G McNamara
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Nicholas R Smith
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Chitra Vinnakota
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Henry J Waldvogel
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Richard Lm Faull
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Andrea Kwakowsky
- Centre for Brain Research, Department of Anatomy and Medical Imaging, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
44
|
Sheikh-Bahaei N, Manavaki R, Sajjadi SA, Priest AN, O’Brien JT, Gillard JH. Correlation of Lobar Cerebral Microbleeds with Amyloid, Perfusion, and Metabolism in Alzheimer’s Disease. J Alzheimers Dis 2019; 68:1489-1497. [DOI: 10.3233/jad-180443] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Nasim Sheikh-Bahaei
- Department of Radiology, Keck School of Medicine of USC, University of Southern California, USA
| | - Roido Manavaki
- Department of Radiology, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - S. Ahmad Sajjadi
- Department of Neurology, University of California Irvine, CA, USA
| | - Andrew N. Priest
- Department of Radiology, Cambridge University Hospitals, Cambridge, UK
| | - John T. O’Brien
- Department of Psychiatry, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Jonathan H. Gillard
- Department of Radiology, University of Cambridge School of Clinical Medicine, Cambridge, UK
| |
Collapse
|
45
|
Lin Z, Sur S, Soldan A, Pettigrew C, Miller M, Oishi K, Bilgel M, Moghekar A, Pillai JJ, Albert M, Lu H. Brain Oxygen Extraction by Using MRI in Older Individuals: Relationship to Apolipoprotein E Genotype and Amyloid Burden. Radiology 2019; 292:140-148. [PMID: 31012816 DOI: 10.1148/radiol.2019182726] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Background Apolipoprotein E4 (APOE4) is a major genetic risk factor for late-onset Alzheimer disease. However, the mechanisms by which APOE4 affects the brain, underpinning this risk, have not been fully elucidated. Purpose To investigate the influence of APOE4 on global cerebral oxygen extraction fraction (OEF) and possible mediation through amyloid burden by using MRI-based brain oxygen extraction technique. Materials and Methods Participants were enrolled from a longitudinal prospective study, the Biomarkers for Older Controls at Risk for Dementia study (data collected from January 2015 to December 2017), of whom 35% (50 of 143 participants) were APOE4 carriers. OEF was measured by using a T2-relaxation-under-spin-tagging MRI technique with a 3.0-T MRI system. PET acquired with carbon 11-labeled Pittsburgh compound B tracer was available in 119 participants to measure amyloid burden. Cognitive performance was assessed by using domain-specific composite scores including executive function, episodic memory, visual-spatial processing, and language. Linear regression analysis was performed to investigate the relationship between APOE4, OEF, and amyloid burden. The association between OEF and cognitive function was studied for the entire study cohort and separately for the APOE4 carriers and noncarriers. Results A total of 143 cognitively healthy individuals (mean age 6 standard deviation, 69.1 years 6 8.2; 57 men and 86 women) were studied. APOE4 genetic status was associated with lower OEF (noncarriers, 41.1% 6 5.8; one E4 allele, 40.1% 6 4.9; two E4 alleles, 36.7% 6 4.5; P = .03). Furthermore, among APOE4 carriers, lower OEF correlated with lower executive function scores (b = 0.079 z score for each percent change in OEF; P = .03). Amyloid burden and OEF were independently associated with APOE4 but were not associated with one another, suggesting that the effect of APOE4 on OEF is not mediated by amyloid. Conclusion MRI-based brain oxygen extraction shows that cognitively healthy carriers of the apolipoprotein E4 gene manifest diminished brain oxygen extraction capacity independent of amyloid burden. ©RSNA, 2019 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Zixuan Lin
- From the Department of Biomedical Engineering (Z.L., M.M., H.L.), The Russell H. Morgan Department of Radiology and Radiological Science (Z.L., S.S., K.O., J.J.P., H.L.), and Department of Neurology (A.S., C.P., A.M., M.A.), Johns Hopkins University School of Medicine, 600 N Wolfe St, Park 322, Baltimore, Md; Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Md (M.B.); and F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, Md (H.L.)
| | - Sandeepa Sur
- From the Department of Biomedical Engineering (Z.L., M.M., H.L.), The Russell H. Morgan Department of Radiology and Radiological Science (Z.L., S.S., K.O., J.J.P., H.L.), and Department of Neurology (A.S., C.P., A.M., M.A.), Johns Hopkins University School of Medicine, 600 N Wolfe St, Park 322, Baltimore, Md; Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Md (M.B.); and F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, Md (H.L.)
| | - Anja Soldan
- From the Department of Biomedical Engineering (Z.L., M.M., H.L.), The Russell H. Morgan Department of Radiology and Radiological Science (Z.L., S.S., K.O., J.J.P., H.L.), and Department of Neurology (A.S., C.P., A.M., M.A.), Johns Hopkins University School of Medicine, 600 N Wolfe St, Park 322, Baltimore, Md; Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Md (M.B.); and F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, Md (H.L.)
| | - Corinne Pettigrew
- From the Department of Biomedical Engineering (Z.L., M.M., H.L.), The Russell H. Morgan Department of Radiology and Radiological Science (Z.L., S.S., K.O., J.J.P., H.L.), and Department of Neurology (A.S., C.P., A.M., M.A.), Johns Hopkins University School of Medicine, 600 N Wolfe St, Park 322, Baltimore, Md; Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Md (M.B.); and F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, Md (H.L.)
| | - Michael Miller
- From the Department of Biomedical Engineering (Z.L., M.M., H.L.), The Russell H. Morgan Department of Radiology and Radiological Science (Z.L., S.S., K.O., J.J.P., H.L.), and Department of Neurology (A.S., C.P., A.M., M.A.), Johns Hopkins University School of Medicine, 600 N Wolfe St, Park 322, Baltimore, Md; Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Md (M.B.); and F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, Md (H.L.)
| | - Kenichi Oishi
- From the Department of Biomedical Engineering (Z.L., M.M., H.L.), The Russell H. Morgan Department of Radiology and Radiological Science (Z.L., S.S., K.O., J.J.P., H.L.), and Department of Neurology (A.S., C.P., A.M., M.A.), Johns Hopkins University School of Medicine, 600 N Wolfe St, Park 322, Baltimore, Md; Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Md (M.B.); and F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, Md (H.L.)
| | - Murat Bilgel
- From the Department of Biomedical Engineering (Z.L., M.M., H.L.), The Russell H. Morgan Department of Radiology and Radiological Science (Z.L., S.S., K.O., J.J.P., H.L.), and Department of Neurology (A.S., C.P., A.M., M.A.), Johns Hopkins University School of Medicine, 600 N Wolfe St, Park 322, Baltimore, Md; Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Md (M.B.); and F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, Md (H.L.)
| | - Abhay Moghekar
- From the Department of Biomedical Engineering (Z.L., M.M., H.L.), The Russell H. Morgan Department of Radiology and Radiological Science (Z.L., S.S., K.O., J.J.P., H.L.), and Department of Neurology (A.S., C.P., A.M., M.A.), Johns Hopkins University School of Medicine, 600 N Wolfe St, Park 322, Baltimore, Md; Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Md (M.B.); and F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, Md (H.L.)
| | - Jay J Pillai
- From the Department of Biomedical Engineering (Z.L., M.M., H.L.), The Russell H. Morgan Department of Radiology and Radiological Science (Z.L., S.S., K.O., J.J.P., H.L.), and Department of Neurology (A.S., C.P., A.M., M.A.), Johns Hopkins University School of Medicine, 600 N Wolfe St, Park 322, Baltimore, Md; Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Md (M.B.); and F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, Md (H.L.)
| | - Marilyn Albert
- From the Department of Biomedical Engineering (Z.L., M.M., H.L.), The Russell H. Morgan Department of Radiology and Radiological Science (Z.L., S.S., K.O., J.J.P., H.L.), and Department of Neurology (A.S., C.P., A.M., M.A.), Johns Hopkins University School of Medicine, 600 N Wolfe St, Park 322, Baltimore, Md; Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Md (M.B.); and F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, Md (H.L.)
| | - Hanzhang Lu
- From the Department of Biomedical Engineering (Z.L., M.M., H.L.), The Russell H. Morgan Department of Radiology and Radiological Science (Z.L., S.S., K.O., J.J.P., H.L.), and Department of Neurology (A.S., C.P., A.M., M.A.), Johns Hopkins University School of Medicine, 600 N Wolfe St, Park 322, Baltimore, Md; Laboratory of Behavioral Neuroscience, National Institute on Aging, National Institutes of Health, Baltimore, Md (M.B.); and F. M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Research Institute, Baltimore, Md (H.L.)
| |
Collapse
|
46
|
van der Thiel M, Rodriguez C, Van De Ville D, Giannakopoulos P, Haller S. Regional Cerebral Perfusion and Cerebrovascular Reactivity in Elderly Controls With Subtle Cognitive Deficits. Front Aging Neurosci 2019; 11:19. [PMID: 30837863 PMCID: PMC6390712 DOI: 10.3389/fnagi.2019.00019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 01/21/2019] [Indexed: 12/27/2022] Open
Abstract
Background: Recent studies suggested that arterial spin labeling (ASL)-based measures of cerebral blood flow (CBF) as well as cerebral vasoreactivity to CO2 (CVR CO2) show significant alterations mainly in posterior neocortical areas both in mild cognitive impairment (MCI) and Alzheimer disease. It remains, however, unknown whether similar changes occur in at risk healthy elders without clinically overt symptoms. This longitudinal study investigated patterns of ASL perfusion and CVR CO2 as a function of the cognitive trajectories in asymptomatic elderly individuals. Methods: Seventy-nine community-dwelling subjects (mean age: 78.7 years, 34 male) underwent three neuropsychological assessments during a subsequent 3-year period. Individuals were classified as stable-stable (SS), variable (V), or progressive-progressive (PP). Between-group comparisons were conducted for ASL CBF and transit-time delay maps and β-maps of CO2 response. Spearman's rho maps assessed the correlation between ASL (respectively, CVR CO2 measures) and Shapes test for working memory, as well as Verbal fluency test for executive functions. Three group-with-continuous-covariate-interaction designs were implemented to investigate group-based differences on the association between neuropsychological scores and ASL or CO2 measures. Results: Comparison of CBF maps demonstrates significantly lower perfusion in the V-group as to PP-cases predominantly in parietal regions, including the precuneus and, to a lesser degree, in temporal and frontal cortex. A stronger CVR CO2 response was found in the PP-group in left parietal areas compared to the V-group. V-cases showed a stronger ASL-Shape value relationship than V-group in right temporoparietal junction and superior parietal lobule. CO2-Shape value correlation was significantly higher in both SS and PP-groups compared to the V-group in right insular and superior perisylvian regions. Conclusion: Our data indicate the presence of decreased ASL and CVR CO2 values mainly in parietal and fronto-temporal areas in cases with the first signs of cognitive instability (V-group). Importantly, the PP-group, at high risk for MCI transition, displays an increase of both parameters in the same areas. Clinicoradiologic correlations also indicate a clear distinction between the V-group and both PP and SS-cases. These data imply the presence of an inverted U-shape pattern of regional blood flow and CVR in old age that might predict subsequent cognitive fate.
Collapse
Affiliation(s)
- Merel van der Thiel
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.,Faculté de Médecine, Université de Genève, Geneva, Switzerland
| | - Cristelle Rodriguez
- Division of Institutional Measures, Geneva University Hospitals, Geneva, Switzerland
| | - Dimitri Van De Ville
- Institute of Bioengineering, École Polytechnique Fédérale de Lausanne, Lausanne, Switzerland.,Faculté de Médecine, Université de Genève, Geneva, Switzerland
| | | | - Sven Haller
- Faculté de Médecine, Université de Genève, Geneva, Switzerland.,CIRD - Centre d'Imagerie Rive Droite, Geneva, Switzerland.,Department of Surgical Sciences, Division of Radiology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
47
|
Polygenic impact of common genetic risk loci for Alzheimer's disease on cerebral blood flow in young individuals. Sci Rep 2019; 9:467. [PMID: 30679549 PMCID: PMC6345995 DOI: 10.1038/s41598-018-36820-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 11/15/2018] [Indexed: 11/15/2022] Open
Abstract
Genome-wide association studies (GWAS) show that many common alleles confer risk for developing Alzheimer’s disease (AD). These risk loci may contribute to MRI alterations in young individuals, preceding the clinical manifestations of AD. Prior evidence identifies vascular dysregulation as the earliest marker of disease progression. However, it remains unclear whether cerebrovascular function (measured via grey-matter cerebral blood flow (gmCBF)) is altered in young individuals with increased AD genetic risk. We establish relationships between gmCBF with APOE and AD polygenic risk score in a young cohort (N = 75; aged: 19–32). Genetic risk was assessed via a) possessing at least one copy of the APOE ɛ4 allele and b) a polygenic risk score (AD-PRS) estimated from AD-GWAS. We observed a reduction in gmCBF in APOE ɛ4 carriers and a negative relationship between AD-PRS and gmCBF. We further found regional reductions in gmCBF in individuals with higher AD-PRS across the frontal cortex (PFWE < 0.05). Our findings suggest that a larger burden of AD common genetic risk alleles is associated with attenuated cerebrovascular function, during young adulthood. These results suggest that cerebral vasculature is a mechanism by which AD risk alleles confer susceptibility.
Collapse
|
48
|
Miyazawa T, Shibata S, Nagai K, Hirasawa A, Kobayashi Y, Koshiba H, Kozaki K. Relationship between cerebral blood flow estimated by transcranial Doppler ultrasound and single-photon emission computed tomography in elderly people with dementia. J Appl Physiol (1985) 2018; 125:1576-1584. [DOI: 10.1152/japplphysiol.00118.2018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Transcranial Doppler (TCD) ultrasonography is a noninvasive technique allowing continuous recording of cerebral blood flow (CBF) velocity. However, it is unclear whether the CBF estimated by TCD would be reliable for the comparison between individuals. The present study aimed to clarify the relationship between middle cerebral artery blood flow (MCA BF) measured by TCD and regional and total CBF measured by single-photon emission computed tomography (SPECT-CBF) with a quantification software program, a three-dimensional stereotaxic region of interest template. We recruited 91 elderly subjects with and without dementia. MCA blood flow velocity (MCA V) and middle cerebral artery cross-sectional area (AM) were measured by TCD and magnetic resonance angiography, respectively. MCA BF was calculated by the product of MCA V and AM. Diastolic or mean MCA V and MCA BF were significantly correlated with SPECT-CBF in several segments. Interestingly, the correlation coefficient in the temporal segment of SPECT-CBF was higher than those of the other segments. Moreover, correlations between MCA BF and SPECT-CBF were stronger as compared with those between MCA V and SPECT-CBF. These findings suggest that both mean MCA V and MCA BF with TCD ultrasonography would be useful for CBF comparison between individuals especially in the temporal region, although estimated blood flow with arterial area seems to be better than using simple flow velocity. NEW & NOTEWORTHY Correlations between middle cerebral artery blood flow (MCA BF) calculated by the product of MCA blood flow velocity (MCA V) and middle cerebral artery cross-sectional area and regional and total cerebral blood flow (CBF) measured by single-photon emission computed tomography (SPECT-CBF) were stronger as compared with those between MCA V and SPECT-CBF. These findings suggest that both mean MCA V and MCA BF would be useful for CBF comparison between individuals although estimated blood flow with arterial area seems to be better than using simple flow velocity.
Collapse
Affiliation(s)
- Taiki Miyazawa
- Department of Health and Sports Science, Faculty of Wellness, Shigakkan University, Obu-shi, Japan
- Department of Geriatric Medicine, School of Medicine, Kyorin University, Tokyo, Japan
| | - Shigeki Shibata
- Department of Geriatric Medicine, School of Medicine, Kyorin University, Tokyo, Japan
| | - Kumiko Nagai
- Department of Geriatric Medicine, School of Medicine, Kyorin University, Tokyo, Japan
| | - Ai Hirasawa
- Department of Geriatric Medicine, School of Medicine, Kyorin University, Tokyo, Japan
| | - Yoshio Kobayashi
- Department of Geriatric Medicine, School of Medicine, Kyorin University, Tokyo, Japan
| | - Hitomi Koshiba
- Department of Geriatric Medicine, School of Medicine, Kyorin University, Tokyo, Japan
| | - Koichi Kozaki
- Department of Geriatric Medicine, School of Medicine, Kyorin University, Tokyo, Japan
| |
Collapse
|
49
|
Bangen KJ, Werhane ML, Weigand AJ, Edmonds EC, Delano-Wood L, Thomas KR, Nation DA, Evangelista ND, Clark AL, Liu TT, Bondi MW. Reduced Regional Cerebral Blood Flow Relates to Poorer Cognition in Older Adults With Type 2 Diabetes. Front Aging Neurosci 2018; 10:270. [PMID: 30250430 PMCID: PMC6139361 DOI: 10.3389/fnagi.2018.00270] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 08/22/2018] [Indexed: 12/27/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) increases risk for dementia, including Alzheimer's disease (AD). Many previous studies of brain changes underlying cognitive impairment in T2DM have applied conventional structural magnetic resonance imaging (MRI) to detect macrostructural changes associated with cerebrovascular disease such as white matter hyperintensities or infarcts. However, such pathology likely reflects end-stage manifestations of chronic decrements in cerebral blood flow (CBF). MRI techniques that measure CBF may (1) elucidate mechanisms that precede irreversible parenchymal damage and (2) serve as a marker of risk for cognitive decline. CBF measured with arterial spin labeling (ASL) MRI may be a useful marker of perfusion deficits in T2DM and related conditions. We examined associations among T2DM, CBF, and cognition in a sample of 49 well-characterized nondemented older adults. Along with a standard T1-weighted scan, a pseudocontinuous ASL sequence optimized for older adults (by increasing post-labeling delays to allow more time for the blood to reach brain tissue) was obtained on a 3T GE scanner to measure regional CBF in FreeSurfer derived regions of interest. Participants also completed a neuropsychological assessment. Results showed no significant differences between individuals with and without T2DM in terms of cortical thickness or regional brain volume. However, adjusting for age, sex, comorbid vascular risk factors, and reference CBF (postcentral gyrus) older adults with T2DM demonstrated reduced CBF in the hippocampus, and inferior temporal, inferior parietal, and frontal cortices. Lower CBF was associated with poorer memory and executive function/processing speed. When adjusting for diabetes, the significant associations between lower regional CBF and poorer executive function/processing speed remained. Results demonstrate that CBF is reduced in older adults with T2DM, and suggest that CBF alterations likely precede volumetric changes. Notably, relative to nondiabetic control participants, those with T2DM showed lower CBF in predilection sites for AD pathology (medial temporal lobe and inferior parietal regions). Findings augment recent research suggesting that perfusion deficits may underlie cognitive decrements frequently observed among older adults with T2DM. Results also suggest that CBF measured with ASL MRI may reflect an early and important marker of risk of cognitive impairment in T2DM and related conditions.
Collapse
Affiliation(s)
- Katherine J Bangen
- Research Service, VA San Diego Healthcare System, San Diego, CA, United States.,Department of Psychiatry, University of California, San Diego, San Diego, CA, United States
| | - Madeleine L Werhane
- Department of Psychiatry, University of California, San Diego, San Diego, CA, United States.,Department of Psychology, San Diego State University, San Diego, CA, United States
| | - Alexandra J Weigand
- Research Service, VA San Diego Healthcare System, San Diego, CA, United States.,Department of Psychiatry, University of California, San Diego, San Diego, CA, United States
| | - Emily C Edmonds
- Research Service, VA San Diego Healthcare System, San Diego, CA, United States.,Department of Psychiatry, University of California, San Diego, San Diego, CA, United States
| | - Lisa Delano-Wood
- Psychology Service, VA San Diego Healthcare System, San Diego, CA, United States.,Department of Psychiatry, University of California, San Diego, San Diego, CA, United States
| | - Kelsey R Thomas
- Department of Psychiatry, University of California, San Diego, San Diego, CA, United States.,Psychology Service, VA San Diego Healthcare System, San Diego, CA, United States
| | - Daniel A Nation
- Department of Psychology, University of Southern California, Los Angeles, CA, United States
| | | | - Alexandra L Clark
- Department of Psychiatry, University of California, San Diego, San Diego, CA, United States.,Department of Psychology, San Diego State University, San Diego, CA, United States
| | - Thomas T Liu
- Department of Radiology and Bioengineering, University of California, San Diego, San Diego, CA, United States
| | - Mark W Bondi
- Department of Psychiatry, University of California, San Diego, San Diego, CA, United States.,Psychology Service, VA San Diego Healthcare System, San Diego, CA, United States
| |
Collapse
|
50
|
Zhang N, Gordon ML, Ma Y, Chi B, Gomar JJ, Peng S, Kingsley PB, Eidelberg D, Goldberg TE. The Age-Related Perfusion Pattern Measured With Arterial Spin Labeling MRI in Healthy Subjects. Front Aging Neurosci 2018; 10:214. [PMID: 30065646 PMCID: PMC6056623 DOI: 10.3389/fnagi.2018.00214] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 06/25/2018] [Indexed: 01/12/2023] Open
Abstract
Aim: To analyze age-related cerebral blood flow (CBF) using arterial spin labeling (ASL) MRI in healthy subjects with multivariate principal component analysis (PCA). Methods: 50 healthy subjects (mean age 45.8 ± 18.5 years, range 21-85) had 3D structural MRI and pseudo-continuous ASL MRI at resting state. The relationship between CBF and age was examined with voxel-based univariate analysis using multiple regression and two-sample t-test (median age 41.8 years as a cut-off). An age-related CBF pattern was identified using multivariate PCA. Results: Age correlated negatively with CBF especially anteriorly and in the cerebellum. After adjusting by global value, CBF was relatively decreased with aging in certain regions and relatively increased in others. The age-related CBF pattern showed relative reductions in frontal and parietal areas and cerebellum, and covarying increases in temporal and occipital areas. Subject scores of this pattern correlated negatively with age (R2 = 0.588; P < 0.001) and discriminated between the older and younger subgroups (P < 0.001). Conclusion: A distinct age-related CBF pattern can be identified with multivariate PCA using ASL MRI.
Collapse
Affiliation(s)
- Nan Zhang
- The Litwin-Zucker Research Center, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Marc L. Gordon
- The Litwin-Zucker Research Center, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
- Donald and Barbara Zucker School of Medicine at Hofstra-Northwell, Hofstra University, Hepstead, NY, United States
| | - Yilong Ma
- Center for Neurosciences, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Bradley Chi
- Donald and Barbara Zucker School of Medicine at Hofstra-Northwell, Hofstra University, Hepstead, NY, United States
| | - Jesus J. Gomar
- The Litwin-Zucker Research Center, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Shichun Peng
- Center for Neurosciences, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Peter B. Kingsley
- Donald and Barbara Zucker School of Medicine at Hofstra-Northwell, Hofstra University, Hepstead, NY, United States
- Department of Radiology, North Shore University Hospital, Northwell Health, Manhasset, NY, United States
| | - David Eidelberg
- Donald and Barbara Zucker School of Medicine at Hofstra-Northwell, Hofstra University, Hepstead, NY, United States
- Center for Neurosciences, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Terry E. Goldberg
- The Litwin-Zucker Research Center, The Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
- Donald and Barbara Zucker School of Medicine at Hofstra-Northwell, Hofstra University, Hepstead, NY, United States
| |
Collapse
|