1
|
Dagher R, Arjmand P, Ozkara BB, Radmard M, Gad M, Sheikhy A, Wintermark M, Yedavalli V, Sair HI, Luna LP. Diagnostic Performance of ASL-MRI and FDG-PET in Frontotemporal Dementia: A Systematic Review and Meta-Analysis. AJNR Am J Neuroradiol 2025; 46:341-348. [PMID: 39134374 DOI: 10.3174/ajnr.a8440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 08/01/2024] [Indexed: 12/29/2024]
Abstract
BACKGROUND While the diagnosis of frontotemporal dementia (FTD) is based mostly on clinical features, [18F]-FDG-PET has been investigated as a potential imaging standard in ambiguous cases, with arterial spin-labeling (ASL) MRI gaining recent interest. PURPOSE The purpose of this study is to conduct a systematic review and meta-analysis on the diagnostic performance of ASL MRI in patients with FTD and compare it with that of [18F]-FDG-PET. DATA SOURCES A systematic search of PubMed, Scopus, and Embase was conducted until March 13, 2024. STUDY SELECTION Inclusion criteria were original articles, patients with FTD and/or its variants, use of ASL MR perfusion imaging with or without [18F]-FDG-PET, and presence of sufficient diagnostic performance data. Exclusion criteria were meeting abstracts, comments, summaries, protocols, letters and guidelines, longitudinal studies, and overlapping cohorts. DATA ANALYSIS The quality of eligible studies was assessed by using the Quality Assessment of Diagnostic Accuracy Studies-2. Pooled sensitivity, specificity, and diagnostic odds ratio (DOR) for [18F]-FDG-PET and ASL MRI were calculated, and a summary receiver operating characteristic curve was plotted. DATA SYNTHESIS Seven eligible studies were identified, which included a total of 102 patients with FTD. Aside from some of the studies showing, at worst, an unclear risk of bias in patient selection, index test, flow, and timing, all studies showed low risk of bias and applicability concerns in all categories. Data from 4 studies were included in our meta-analysis for ASL MRI and 3 studies for [18F]-FDG-PET. Pooled sensitivity, specificity, and DOR were 0.70 (95% CI: 0.59-0.79), 0.81 (95% CI: 0.71-0.88), and 8.00 (95% CI: 3.74-17.13) for ASL MRI and 0.88 (95% CI: 0.71-0.96), 0.89 (95% CI: 0.43-0.99), and 47.18 (95% CI: 10.77-206.75) for [18F]-FDG-PET. LIMITATIONS The number of studies was relatively small, with a small sample size. The studies used different scanning protocols as well as a mix of diagnostic metrics, all of which might have introduced heterogeneity in the data. CONCLUSIONS While ASL MRI performed worse than [18F]-FDG-PET in the diagnosis of FTD, it exhibited a decent diagnostic performance to justify its further investigation as a quicker and more convenient alternative.
Collapse
Affiliation(s)
- Richard Dagher
- From the Department of Neuroradiology (R.D., B.B.O., M.W.), MD Anderson Cancer Center, Houston, Texas
| | - Parisa Arjmand
- Mashhad University of Medical Sciences (P.A.), Mashhad, Iran
| | - Burak Berksu Ozkara
- From the Department of Neuroradiology (R.D., B.B.O., M.W.), MD Anderson Cancer Center, Houston, Texas
| | - Mahla Radmard
- Russell H. Morgan Department of Radiology and Radiological Science (M.R., V.Y., H.I.S., L.P.L.), Johns Hopkins Hospital, Baltimore, Maryland
| | - Mona Gad
- Diagnostic Radiology Department (M.G.), Faculty of Medicine, Mansoura University, Egypt
| | - Ali Sheikhy
- Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Max Wintermark
- From the Department of Neuroradiology (R.D., B.B.O., M.W.), MD Anderson Cancer Center, Houston, Texas
| | - Vivek Yedavalli
- Russell H. Morgan Department of Radiology and Radiological Science (M.R., V.Y., H.I.S., L.P.L.), Johns Hopkins Hospital, Baltimore, Maryland
| | - Haris I Sair
- Russell H. Morgan Department of Radiology and Radiological Science (M.R., V.Y., H.I.S., L.P.L.), Johns Hopkins Hospital, Baltimore, Maryland
| | - Licia P Luna
- Russell H. Morgan Department of Radiology and Radiological Science (M.R., V.Y., H.I.S., L.P.L.), Johns Hopkins Hospital, Baltimore, Maryland
| |
Collapse
|
2
|
Salminen A. The role of inhibitory immune checkpoint receptors in the pathogenesis of Alzheimer's disease. J Mol Med (Berl) 2025; 103:1-19. [PMID: 39601807 PMCID: PMC11739239 DOI: 10.1007/s00109-024-02504-x] [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: 06/24/2024] [Revised: 10/16/2024] [Accepted: 11/19/2024] [Indexed: 11/29/2024]
Abstract
There is mounting evidence that microglial cells have a key role in the pathogenesis of Alzheimer's disease (AD). In AD pathology, microglial cells not only are unable to remove β-amyloid (Aβ) plaques and invading pathogens but also are involved in synaptic pruning, chronic neuroinflammation, and neuronal degeneration. Microglial cells possess many different inhibitory immune checkpoint receptors, such as PD-1, LILRB2-4, Siglecs, and SIRPα receptors, which can be targeted by diverse cell membrane-bound and soluble ligand proteins to suppress the functions of microglia. Interestingly, in the brains of AD patients there are elevated levels of many of the inhibitory ligands acting via these inhibitory checkpoint receptors. For instance, Aβ oligomers, ApoE4, and fibronectin are able to stimulate the LILRB2-4 receptors. Increased deposition of sialoglycans, e.g., gangliosides, inhibits microglial function via Siglec receptors. AD pathology augments the accumulation of senescent cells, which are known to possess a high level of PD-L1 proteins, and thus, they can evade immune surveillance. A decrease in the expression of SIRPα receptor in microglia and its ligand CD47 in neurons enhances the phagocytic pruning of synapses in AD brains. Moreover, cerebral neurons contain inhibitory checkpoint receptors which can inhibit axonal growth, reduce synaptic plasticity, and impair learning and memory. It seems that inappropriate inhibitory immune checkpoint signaling impairs the functions of microglia and neurons thus promoting AD pathogenesis. KEY MESSAGES: Microglial cells have a major role in the pathogenesis of AD. A decline in immune activity of microglia promotes AD pathology. Microglial cells and neurons contain diverse inhibitory immune checkpoint receptors. The level of ligands for inhibitory checkpoint receptors is increased in AD pathology. Impaired signaling of inhibitory immune checkpoint receptors promotes AD pathology.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.
| |
Collapse
|
3
|
Evans AK, Park HH, Woods CE, Lam RK, Rijsketic DR, Xu C, Chu EK, Ciari P, Blumenfeld S, Vidano LM, Saw NL, Heifets BD, Shamloo M. Impact of noradrenergic inhibition on neuroinflammation and pathophysiology in mouse models of Alzheimer's disease. J Neuroinflammation 2024; 21:322. [PMID: 39696597 DOI: 10.1186/s12974-024-03306-1] [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: 10/24/2024] [Accepted: 11/19/2024] [Indexed: 12/20/2024] Open
Abstract
Norepinephrine (NE) modulates cognitive function, arousal, attention, and responses to novelty and stress, and it also regulates neuroinflammation. We previously demonstrated behavioral and immunomodulatory effects of beta-adrenergic pharmacology in mouse models of Alzheimer's disease (AD). The current studies were designed to block noradrenergic signaling in 5XFAD mice through (1) chemogenetic inhibition of the locus coeruleus (LC), (2) pharmacologic blocking of β-adrenergic receptors, and (3) conditional deletion of β1- or β2-adrenergic receptors (adrb1 or adrb2) in microglia.First, brain-wide AD pathology was mapped in 3D by imaging immunolabeled, cleared 5XFAD brains to assess the overlap between amyloid beta (Aβ) pathology, reactive microglia, and the loss of tyrosine hydroxylase (TH) expression in the catecholaminergic system. To examine the effects of inhibiting the LC NE system in the 5XFAD model, inhibitory (Gi) DREADD receptors were expressed specifically in LC NE neurons. LC NE neurons were chronically inhibited through the subcutaneous pump administration of the DREADD agonist clozapine-N-oxide (CNO). Plasma and brains were collected for assessment of neuroinflammation and pathology. A separate cohort of 5XFAD mice was chronically dosed with the beta-adrenergic antagonist propranolol or vehicle and evaluated for behavior, as well as post-mortem neuroinflammation and pathology. Finally, we used 5XFAD mice with conditional deletion of either adrb1 or adrb2 in microglia to assess neuroinflammation and pathology mediated by β-adrenergic signaling.Using iDISCO+, light sheet fluorescence microscopy, and novel analyses, we detected widespread microgliosis and Aβ pathology, along with modest TH downregulation in fibers across multiple brain regions, in contrast to the spatially limited TH downregulation observed in neurons. Both chemogenetic inhibition of LC adrenergic signaling and pharmacological inhibition of beta-adrenergic receptors potentiated neuroinflammation without altering Aβ pathology. Conditional deletion of adrb1 in microglia did not affect neuroinflammation. Conditional deletion of adrb2 in microglia attenuated inflammation and pathology in females but had no effect in males. Overall, these data support previous observations demonstrating the immunomodulatory effects of beta-adrenergic signaling in the pathophysiology of brain disorders and suggest that adrenergic receptors on cell types other than microglia, such as astrocytes, may mediate some of the disease-modifying effects of β-adrenergic agonists in the brain.
Collapse
Affiliation(s)
- Andrew K Evans
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, Stanford, CA, 94304, United States of America
| | - Heui Hye Park
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, Stanford, CA, 94304, United States of America
| | - Claire E Woods
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, Stanford, CA, 94304, United States of America
| | - Rachel K Lam
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, Stanford, CA, 94304, United States of America
| | - Daniel Ryskamp Rijsketic
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, Stanford, CA, 94304, United States of America
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, Stanford, CA, 94304, United States of America
| | - Christine Xu
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, Stanford, CA, 94304, United States of America
| | - Emily K Chu
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, Stanford, CA, 94304, United States of America
| | - Peter Ciari
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, Stanford, CA, 94304, United States of America
| | - Sarah Blumenfeld
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, Stanford, CA, 94304, United States of America
| | - Laura M Vidano
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, Stanford, CA, 94304, United States of America
| | - Nay Lui Saw
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, Stanford, CA, 94304, United States of America
| | - Boris D Heifets
- Department of Anesthesiology, Perioperative, and Pain Medicine, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, Stanford, CA, 94304, United States of America
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, 94305, United States of America
| | - Mehrdad Shamloo
- Department of Neurosurgery, Stanford University School of Medicine, 1050 Arastradero Road, Building A, Palo Alto, Stanford, CA, 94304, United States of America.
| |
Collapse
|
4
|
Evans AK, Park HH, Woods CE, Lam RK, Rijsketic DR, Xu C, Chu E, Ciari P, Blumenfeld S, Vidano LM, Saw NL, Heifets BD, Shamloo M. Impact of noradrenergic inhibition on neuroinflammation and pathophysiology in mouse models of Alzheimer's disease. RESEARCH SQUARE 2024:rs.3.rs-5328229. [PMID: 39574895 PMCID: PMC11581111 DOI: 10.21203/rs.3.rs-5328229/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2024]
Abstract
Norepinephrine (NE) modulates cognitive function, arousal, attention, and responses to novelty and stress, and also regulates neuroinflammation. We previously demonstrated behavioral and immunomodulatory effects of beta-adrenergic pharmacology in mouse models of Alzheimer's disease (AD). The current studies were designed to block noradrenergic signaling in 5XFAD mice through 1) chemogenetic inhibition of the locus coeruleus (LC), 2)pharmacologic blocking of β-adrenergic receptors, and 3) conditional deletion of β1- or β2-adrenergic receptors (adrb1 or adrb2) in microglia. First, brain-wide AD pathology was mapped in 3D by imaging immunolabeled, cleared 5XFAD brains to assess the overlap between Aβ pathology, reactive microglia, and the loss of tyrosine hydroxylase (TH) expression in the catecholaminergic system. To examine the effects of inhibiting the LC NE system in the 5XFAD model, inhibitory (Gi) DREADD receptors were expressed specifically in LC NE neurons. LC NE neurons were chronically inhibited through the subcutaneous pump administration of the DREADD agonist clozapine-N-oxide (CNO). Plasma and brains were collected for assessment of neuroinflammation and pathology. A separate cohort of 5XFAD mice was chronically dosed with the beta-adrenergic antagonist propranolol or vehicle and evaluated for behavior, as well as post-mortem neuroinflammation and pathology. Finally, we used 5XFAD mice with conditional deletion of either adrb1 or adrb2 in microglia to assess neuroinflammation and pathology mediated by β-adrenergic signaling. Using iDISCO, light sheet fluorescence microscopy, and novel analyses, we detected widespread microgliosis and amyloid pathology, along with modest TH downregulation in fibers across multiple brain regions, in contrast to the spatially limited TH downregulation observed in neurons. Both chemogenetic inhibition of LC adrenergic signaling and pharmacological inhibition of beta-adrenergic receptors potentiated neuroinflammation without altering amyloid beta pathology. Conditional deletion of adrb1 in microglia did not affect neuroinflammation. Conditional deletion of adrb2 in microglia attenuated inflammation and pathology in females but had no effect in males. Overall, these data support previous observations demonstrating the immunomodulatory effects of beta-adrenergic signaling in the pathophysiology of brain disorders and suggest that adrenergic receptors on cell types other than microglia, such as astrocytes, may predominantly mediate the disease-modifying effects of β-adrenergic agonists in the brain.
Collapse
|
5
|
Gao Y, Zhao P, Wang C, Fang K, Pan Y, Zhang Y, Miao Z, Wang M, Wei M, Zou W, Liu M, Peng K. Buqi Huoxue Tongnao prescription protects against chronic cerebral hypoperfusion via regulating PI3K/AKT and LXRα/CYP7A1 signaling pathways. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155844. [PMID: 38959552 DOI: 10.1016/j.phymed.2024.155844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/01/2024] [Accepted: 06/24/2024] [Indexed: 07/05/2024]
Abstract
BACKGROUND Chronic cerebral hypoperfusion (CCH) has been confirmed as one of the pathogenesis underlying vascular cognitive impairment. A series of pathological changes, including inflammation, oxidative stress, and apoptosis, are involved in this pathophysiology and contribute to cognitive impairment and neuropathological alterations. The traditional Chinese medicine (TCM) of Buqi Huoxue Tongnao (BQHXTN) prescription possesses a remarkable clinical efficacy for treating patients with CCH, but still lacks a scientific foundation for its pharmacological mechanisms. PURPOSE To investigate the role and underlying mechanism of the effects of BQHXTN on CCH both in vitro and in vivo. METHODS In this study, we established a two-vessel occlusion (2-VO) induced CCH model in Sprague-Dawley rats, an oxygen-glucose deprivation model in BV2 cells, and a steatosis cell model in L02 cells to reveal the underlying mechanisms of BQHXTN by behavioral test, histopathological analysis and the detection of pro-inflammatory cytokine, apoptotic factors and reactive oxide species. Donepezil hydrochloride and Buyang Huanwu decoction were used as positive drugs. RESULTS Compared with the 2-VO group, BQHXTN treatment at three doses significantly enhanced the memory and learning abilities in the Y-maze and novel object recognition tests. The hematoxylin-eosin staining indicated that BQHXTN protected against hippocampal injury induced by CCH. Of note, in both in vivo and in vitro experiments, BQHXTN prominently inhibited the production of IL-1β, TNF-α, cleaved-caspase 3, and iNOS by regulating the PI3K/AKT pathway, consequently exerting anti-inflammatory, anti-apoptotic, and antioxidant effects. Moreover, it provided the first initial evidence that BQHXTN treatment mitigated dyslipidemia by increasing the LXRα/CYP7A1 expression, thereby delaying the neuropathological process. CONCLUSION In summary, these findings firstly revealed the pharmacodynamics and mechanism of BQHXTN, that is, BQHXTN could alleviate cognitive impairment, neuropathological alterations and dyslipidemia in CCH rats by activating PI3K/AKT and LXRα/CYP7A1 signaling pathways, as well as providing a TCM treatment strategy for CCH.
Collapse
Affiliation(s)
- Yinhuang Gao
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Peng Kang National Famous Traditional Chinese Medicine Expert Inheritance Studio, Southern Medicine University, Guangzhou 510315, China; Key Laboratory of Drug Metabolism Research and Evaluation of the State Drug Administration, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Peng Zhao
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Peng Kang National Famous Traditional Chinese Medicine Expert Inheritance Studio, Southern Medicine University, Guangzhou 510315, China
| | - Chunyan Wang
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Peng Kang National Famous Traditional Chinese Medicine Expert Inheritance Studio, Southern Medicine University, Guangzhou 510315, China
| | - Keren Fang
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Peng Kang National Famous Traditional Chinese Medicine Expert Inheritance Studio, Southern Medicine University, Guangzhou 510315, China
| | - Yueqing Pan
- Key Laboratory of Drug Metabolism Research and Evaluation of the State Drug Administration, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Yan Zhang
- Key Laboratory of Drug Metabolism Research and Evaluation of the State Drug Administration, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Zhishuo Miao
- Key Laboratory of Drug Metabolism Research and Evaluation of the State Drug Administration, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Meirong Wang
- Key Laboratory of Drug Metabolism Research and Evaluation of the State Drug Administration, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Minlong Wei
- Key Laboratory of Drug Metabolism Research and Evaluation of the State Drug Administration, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Wei Zou
- Changsha Research and Development Center on Obstetric and Gynecologic Traditional Chinese Medicine Preparation, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan 410008, China
| | - Menghua Liu
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Peng Kang National Famous Traditional Chinese Medicine Expert Inheritance Studio, Southern Medicine University, Guangzhou 510315, China; Key Laboratory of Drug Metabolism Research and Evaluation of the State Drug Administration, Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China.
| | - Kang Peng
- Southern Medical University Hospital of Integrated Traditional Chinese and Western Medicine, Peng Kang National Famous Traditional Chinese Medicine Expert Inheritance Studio, Southern Medicine University, Guangzhou 510315, China.
| |
Collapse
|
6
|
Heholt J, Patel R, Vedaei F, Zabrecky G, Wintering N, Monti DA, Wang Z, Newberg AB, Mohamed FB. Simultaneous arterial spin labeling functional MRI and fluorodeoxyglucose PET in mild chronic traumatic brain injury. J Neuroradiol 2024; 51:101211. [PMID: 38908545 DOI: 10.1016/j.neurad.2024.101211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/12/2024] [Accepted: 06/18/2024] [Indexed: 06/24/2024]
Abstract
BACKGROUND AND PURPOSE To determine the effect of mild chronic traumatic brain injury (cTBI) on cerebral blood flow and metabolism. METHODS 62 cTBI and 40 healthy controls (HCs) with no prior history of cTBI underwent both pulsed arterial spin labeling functional magnetic resonance imaging (PASL-fMRI) and fluorodeoxyglucose positron emission tomography (FDG-PET) scanning via a Siemens mMR (simultaneous PET/MRI) scanner. 30 participants also took part in a series of neuropsychological clinical measures (NCMs). Images were processed using statistical parametric mapping software relevant to each modality to generate relative cerebral blood flow (rCBF) and glucose metabolic standardized uptake value ratio (gSUVR) grey matter maps. A voxel-wise two-sample T-test and two-tailed gaussian random field correction for multiple comparisons was performed. RESULTS cTBI patients showed a significant increase in rCBF and gSUVR in the right thalamus as well as a decrease in bilateral occipital lobes and calcarine sulci. An inverse relationship between rCBF and gSUVR was found in the left frontal lobe, the left precuneus and regions in the right temporal lobe. Within those regions rCBF values correlated with 9 distinct NCMs and gSUVR with 3. CONCLUSION Simultaneous PASL-fMRI and FDG-PET can identify functional changes in a mild cTBI population. Within this population FDG-PET identified more regions of functional disturbance than ASL fMRI and NCMs are shown to correlate with rCBF and glucose metabolism (gSUVR) in various brain regions. As a result, both imaging modalities contribute to understanding the underlying pathophysiology and clinical course of mild chronic traumatic brain injury.
Collapse
Affiliation(s)
- Justin Heholt
- Department of Radiology, State University of New York, Downstate Medical Center, Brooklyn, NY, USA
| | - Riya Patel
- Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA
| | - Faezeh Vedaei
- Jefferson Integrated Magnetic Resonance Imaging Center, Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| | - George Zabrecky
- Department of Integrative Medicine and Nutritional Sciences, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA, USA
| | - Nancy Wintering
- Department of Integrative Medicine and Nutritional Sciences, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA, USA
| | - Daniel A Monti
- Department of Integrative Medicine and Nutritional Sciences, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA, USA
| | - Ze Wang
- Department of Diagnostic Radiology & Nuclear Medicine, University of Maryland School of Medicine
| | - Andrew B Newberg
- Department of Integrative Medicine and Nutritional Sciences, Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA, USA.
| | - Feroze B Mohamed
- Jefferson Integrated Magnetic Resonance Imaging Center, Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
7
|
Li LY, Park E, He C, Abbasi AZ, Henderson JT, Fraser PE, Uetrecht JP, Rauth AM, Wu XY. Evaluation of the biodistribution and preliminary safety profile of a novel brain-targeted manganese dioxide-based nanotheranostic system for Alzheimer's disease. Nanotoxicology 2024; 18:315-334. [PMID: 38847611 DOI: 10.1080/17435390.2024.2361687] [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: 08/15/2023] [Revised: 05/07/2024] [Accepted: 05/27/2024] [Indexed: 08/03/2024]
Abstract
A novel brain-targeted and reactive oxygen species-activatable manganese dioxide containing nanoparticle system functionalized with anti-amyloid-β antibody (named aAβ-BTRA-NC) developed by our group has shown great promise as a highly selective magnetic resonance imaging (MRI) contrast agent for early detection and multitargeted disease-modifying treatment of Alzheimer's disease (AD). To further evaluate the suitability of the formulation for future clinical application, we investigated the safety, biodistribution, and pharmacokinetic profile of aAβ-BTRA-NC in a transgenic TgCRND8 mouse AD model, wild type (WT) littermate, and CD-1 mice. Dose-ascending studies demonstrated that aAβ-BTRA-NC was well-tolerated by the animals up to 300 μmol Mn/kg body weight [b.w.], 3 times the efficacious dose for early AD detection without apparent adverse effects; Histopathological, hematological, and biochemical analyses indicated that a single dose of aAβ-BTRA-NC did not cause any toxicity in major organs. Immunotoxicity data showed that aAβ-BTRA-NC was safer than commercially available gadolinium-based MRI contrast agents at an equivalent dose of 100 μmol/kg b.w. of metal ions. Intravenously administered aAβ-BTRA-NC was taken up by main organs with the order of liver, kidneys, intestines, spleen, followed by other organs, and cleared after one day to one week post injection. Pharmacokinetic analysis indicated that the plasma concentration profile of aAβ-BTRA-NC followed a 2-compartmental model with faster clearance in the AD mice than in the WT mice. The results suggest that aAβ-BTRA-NC exhibits a strong safety profile as a nanotheranostic agent which warrants more robust preclinical development for future clinical applications.
Collapse
Affiliation(s)
- Lily Yi Li
- Leslie L. Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Elliya Park
- Leslie L. Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Chunsheng He
- Leslie L. Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Azhar Z Abbasi
- Leslie L. Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Jeffrey T Henderson
- Leslie L. Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Paul E Fraser
- Tanz Centre for Research in Neurodegenerative Diseases, Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Jack P Uetrecht
- Leslie L. Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Andrew M Rauth
- Departments of Medical Biophysics and Radiation Oncology, University of Toronto, Toronto, Ontario, Canada
| | - Xiao Yu Wu
- Leslie L. Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
8
|
Rahimzadeh H, Kamkar H, Ghafarian P, Hoseini-Tabatabaei N, Mohammadi-Mobarakeh N, Mehvari-Habibabadi J, Hashemi-Fesharaki SS, Nazem-Zadeh MR. Exploring ASL perfusion MRI as a substitutive modality for 18F-FDG PET in determining the laterality of mesial temporal lobe epilepsy. Neurol Sci 2024; 45:2223-2243. [PMID: 37994963 DOI: 10.1007/s10072-023-07188-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Accepted: 11/03/2023] [Indexed: 11/24/2023]
Abstract
OBJECTIVE The aim of this investigation was to determine whether a correlation could be discerned between perfusion acquired through ASL MRI and metabolic data acquired via 18F-fluorodeoxyglucose (18F-FDG) PET in mesial temporal lobe epilepsy (mTLE). METHODS ASL MRI and 18F-FDG PET data were gathered from 22 mTLE patients. Relative cerebral blood flow (rCBF) asymmetry index (AIs) were measured using ASL MRI, and standardized uptake value ratio (SUVr) maps were obtained from 18F-FDG PET, focusing on bilateral vascular territories and key bitemporal lobe structures (amygdala, hippocampus, and parahippocampus). Intra-group comparisons were carried out to detect hypoperfusion and hypometabolism between the left and right brain hemispheres for both rCBF and SUVr in right and left mTLE. Correlations between the two AIs computed for each modality were examined. RESULTS Significant correlations were observed between rCBF and SUVr AIs in the middle temporal gyrus, superior temporal gyrus, and hippocampus. Significant correlations were also found in vascular territories of the distal posterior, intermediate anterior, intermediate middle, proximal anterior, and proximal middle cerebral arteries. Intra-group comparisons unveiled significant differences in rCBF and SUVr between the left and right brain hemispheres for right mTLE, while hypoperfusion and hypometabolism were infrequently observed in any intracranial region for left mTLE. CONCLUSION The study's findings suggest promising concordance between hypometabolism estimated by 18F-FDG PET and hypoperfusion determined by ASL perfusion MRI. This raises the possibility that, with prospective technical enhancements, ASL perfusion MRI could be considered an alternative modality to 18F-FDG PET in the future.
Collapse
Affiliation(s)
- Hossein Rahimzadeh
- Research Center for Molecular and Cellular Imaging, Advanced Medical Technologies and Equipment Institute (AMTEI), Tehran University of Medical Sciences, Tehran, Iran
- Department of Biomedical Engineering and Medical Physics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hadi Kamkar
- Research Center for Molecular and Cellular Imaging, Advanced Medical Technologies and Equipment Institute (AMTEI), Tehran University of Medical Sciences, Tehran, Iran
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Pardis Ghafarian
- Chronic Respiratory Diseases Research Center, National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Neda Mohammadi-Mobarakeh
- Research Center for Molecular and Cellular Imaging, Advanced Medical Technologies and Equipment Institute (AMTEI), Tehran University of Medical Sciences, Tehran, Iran
- Medical Physics and Biomedical Engineering Department, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Seyed-Sohrab Hashemi-Fesharaki
- Pars Advanced and Minimally Invasive Medical Manners Research Center, Pars Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad-Reza Nazem-Zadeh
- Research Center for Molecular and Cellular Imaging, Advanced Medical Technologies and Equipment Institute (AMTEI), Tehran University of Medical Sciences, Tehran, Iran.
- Medical Physics and Biomedical Engineering Department, Tehran University of Medical Sciences, Tehran, Iran.
- Department of Neuroscience, Monash University, Melbourne, Australia.
| |
Collapse
|
9
|
Bi S, Yan S, Chen Z, Cui B, Shan Y, Yang H, Qi Z, Zhao Z, Han Y, Lu J. Comparison of 18F-FDG PET and arterial spin labeling MRI in evaluating Alzheimer's disease and amnestic mild cognitive impairment using integrated PET/MR. EJNMMI Res 2024; 14:9. [PMID: 38270821 PMCID: PMC10811308 DOI: 10.1186/s13550-024-01068-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 01/14/2024] [Indexed: 01/26/2024] Open
Abstract
BACKGROUND Developing biomarkers for early stage AD patients is crucial. Glucose metabolism measured by 18F-FDG PET is the most common biomarker for evaluating cellular energy metabolism to diagnose AD. Arterial spin labeling (ASL) MRI can potentially provide comparable diagnostic information to 18F-FDG PET in patients with neurodegenerative disorders. However, the conclusions about the diagnostic performance of AD are still controversial between 18F-FDG PET and ASL. This study aims to compare quantitative cerebral blood flow (CBF) and glucose metabolism measured by 18F-FDG PET diagnostic values in patients with Alzheimer's disease (AD) and amnestic mild cognitive impairment (aMCI) using integrated PET/MR. RESULTS Analyses revealed overlapping between decreased regional rCBF and 18F-FDG PET SUVR in patients with AD compared with NC participants in the bilateral parietotemporal regions, frontal cortex, and cingulate cortex. Compared with NC participants, patients with aMCI exclusively demonstrated lower 18F-FDG PET SUVR in the bilateral temporal cortex, insula cortex, and inferior frontal cortex. Comparison of the rCBF in patients with aMCI and NC participants revealed no significant difference (P > 0.05). The ROC analysis of rCBF in the meta-ROI could diagnose patients with AD (AUC, 0.87) but not aMCI (AUC, 0.61). The specificity of diagnosing aMCI has been improved to 75.56% when combining rCBF and 18F-FDG PET SUVR. CONCLUSION ASL could detect similar aberrant patterns of abnormalities compared to 18F-FDG PET in patients with AD compared with NC participants but not in aMCI. The diagnostic efficiency of 18F-FDG-PET for AD and aMCI patients remained higher to ASL. Our findings support that applying 18F-FDG PET may be preferable for diagnosing AD and aMCI.
Collapse
Affiliation(s)
- Sheng Bi
- Department of Radiology & Nuclear Medicine, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Xicheng District, Beijing, 100053, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Shaozhen Yan
- Department of Radiology & Nuclear Medicine, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Xicheng District, Beijing, 100053, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Zhigeng Chen
- Department of Radiology & Nuclear Medicine, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Xicheng District, Beijing, 100053, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Bixiao Cui
- Department of Radiology & Nuclear Medicine, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Xicheng District, Beijing, 100053, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Yi Shan
- Department of Radiology & Nuclear Medicine, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Xicheng District, Beijing, 100053, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Hongwei Yang
- Department of Radiology & Nuclear Medicine, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Xicheng District, Beijing, 100053, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Zhigang Qi
- Department of Radiology & Nuclear Medicine, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Xicheng District, Beijing, 100053, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Zhilian Zhao
- Department of Radiology & Nuclear Medicine, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Xicheng District, Beijing, 100053, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China
| | - Ying Han
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jie Lu
- Department of Radiology & Nuclear Medicine, Xuanwu Hospital, Capital Medical University, 45 Changchun Street, Xicheng District, Beijing, 100053, China.
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China.
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, China.
| |
Collapse
|
10
|
Cerina V, Crivellaro C, Morzenti S, Pozzi FE, Bigiogera V, Jonghi-Lavarini L, Moresco RM, Basso G, De Bernardi E. A ROI-based quantitative pipeline for 18F-FDG PET metabolism and pCASL perfusion joint analysis: Validation of the 18F-FDG PET line. Heliyon 2024; 10:e23340. [PMID: 38163125 PMCID: PMC10755331 DOI: 10.1016/j.heliyon.2023.e23340] [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: 09/29/2023] [Accepted: 12/01/2023] [Indexed: 01/03/2024] Open
Abstract
In Mild Cognitive Impairment (MCI), the study of brain metabolism, provided by 18F-FluoroDeoxyGlucose Positron Emission Tomography (18F-FDG PET) can be integrated with brain perfusion through pseudo-Continuous Arterial Spin Labeling Magnetic Resonance sequences (MR pCASL). Cortical hypometabolism identification generally relies on wide control group datasets; pCASL control groups are instead not publicly available yet, due to lack of standardization in the acquisition parameters. This study presents a quantitative pipeline to be applied to PET and pCASL data to coherently analyze metabolism and perfusion inside 16 matching cortical regions of interest (ROIs) derived from the AAL3 atlas. The PET line is tuned on 36 MCI patients and 107 healthy control subjects, to agree in identifying hypometabolic regions with clinical reference methods (visual analysis supported by a vendor tool and Statistical Parametric Mapping, SPM, with two parametrizations here identified as SPM-A and SPM-B). The analysis was conducted for each ROI separately. The proposed PET analysis pipeline obtained accuracy 78 % and Cohen's к 60 % vs visual analysis, accuracy 79 % and Cohen's к 58 % vs SPM-A, accuracy 77 % and Cohen's к 54 % vs SPM-B. Cohen's к resulted not significantly different from SPM-A and SPM-B Cohen's к when assuming visual analysis as reference method (p-value 0.61 and 0.31 respectively). Considering SPM-A as reference method, Cohen's к is not significantly different from SPM-B Cohen's к as well (p-value = 1.00). The complete PET-pCASL pipeline was then preliminarily applied on 5 MCI patients and metabolism-perfusion regional correlations were assessed. The proposed approach can be considered as a promising tool for PET-pCASL joint analyses in MCI, even in the absence of a pCASL control group, to perform metabolism-perfusion regional correlation studies, and to assess and compare perfusion in hypometabolic or normo-metabolic areas.
Collapse
Affiliation(s)
- Valeria Cerina
- PhD program in Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Italy
| | - Cinzia Crivellaro
- Nuclear Medicine, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italia
| | - Sabrina Morzenti
- Medical Physics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italia
| | - Federico E. Pozzi
- PhD program in Neuroscience, School of Medicine and Surgery, University of Milano-Bicocca, Italy
- Neurology, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italia
- Milan center for Neuroscience (NeuroMI), University of Milano-Bicocca, Italy
| | | | | | - Rosa M. Moresco
- School of Medicine and Surgery, University of Milano-Bicocca, Italy
| | - Gianpaolo Basso
- Milan center for Neuroscience (NeuroMI), University of Milano-Bicocca, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, Italy
- Neuroradiology, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italia
| | | |
Collapse
|
11
|
Matt RA, Martin RS, Evans AK, Gever JR, Vargas GA, Shamloo M, Ford AP. Locus Coeruleus and Noradrenergic Pharmacology in Neurodegenerative Disease. Handb Exp Pharmacol 2024; 285:555-616. [PMID: 37495851 DOI: 10.1007/164_2023_677] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/28/2023]
Abstract
Adrenoceptors (ARs) throughout the brain are stimulated by noradrenaline originating mostly from neurons of the locus coeruleus, a brainstem nucleus that is ostensibly the earliest to show detectable pathology in neurodegenerative diseases such as Alzheimer's and Parkinson's diseases. The α1-AR, α2-AR, and β-AR subtypes expressed in target brain regions and on a range of cell populations define the physiological responses to noradrenaline, which includes activation of cognitive function in addition to modulation of neurometabolism, cerebral blood flow, and neuroinflammation. As these heterocellular functions are critical for maintaining brain homeostasis and neuronal health, combating the loss of noradrenergic tone from locus coeruleus degeneration may therefore be an effective treatment for both cognitive symptoms and disease modification in neurodegenerative indications. Two pharmacologic approaches are receiving attention in recent clinical studies: preserving noradrenaline levels (e.g., via reuptake inhibition) and direct activation of target adrenoceptors. Here, we review the expression and role of adrenoceptors in the brain, the preclinical studies which demonstrate that adrenergic stimulation can support cognitive function and cerebral health by reversing the effects of noradrenaline depletion, and the human data provided by pharmacoepidemiologic analyses and clinical trials which together identify adrenoceptors as promising targets for the treatment of neurodegenerative disease.
Collapse
Affiliation(s)
| | | | - Andrew K Evans
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA, USA
| | | | | | - Mehrdad Shamloo
- Department of Neurosurgery, Stanford University School of Medicine, Palo Alto, CA, USA
| | | |
Collapse
|
12
|
Wang H, Zhang Z, Hongpaisan J. PKCε activator protects hippocampal microvascular disruption and memory defect in 3×Tg-Alzheimer's disease mice with cerebral microinfarcts. Front Aging Neurosci 2023; 15:1272361. [PMID: 38187357 PMCID: PMC10768563 DOI: 10.3389/fnagi.2023.1272361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 11/23/2023] [Indexed: 01/09/2024] Open
Abstract
Background Current evidence suggests that microvessel disease is involved in Alzheimer's disease (AD). Cerebrovascular disease correlates with cardiovascular disease and is complicated in ≈40% of AD patients. The protein kinase C (PKC) ε activator DCPLA can stimulate human antigen (Hu) R that prevents degradation and promotes the translation of mitochondrial Mn-superoxide dismutase (MnSOD) and vascular endothelial growth factor-A (VEGF) mRNAs. Methods To induce brain microinfarcts, we injected triple transgenic (3×Tg) and wild-type (WT) control mice with microbeads (20 μm caliber) into common carotid arteries, with or without the DCPLA-ME (methyl-ester) for 2 weeks. After water maze training, mice at 16 months old were examined for confocal immunohistochemistry at a single cell or microvessel level in the hippocampal CA1 area, important for spatial memory storage, and in the dorsal hippocampus by western blots. Results In 3×Tg mice without cerebral microinfarcts, an accelerating age-related increase in (mild) oxidative stress and hypoxia inducible factor (HIF)-1α, but a reduction in VEGF, mitochondrial transcription factor A (TFAM), and MnSOD were associated with capillary loss. The change was less pronounced in arterioles. However, in 3×Tg mice with cerebral microinfarcts, increasing arteriolar diameter and their wall cells were related with the strong oxidative DNA damage 8-hydroxy-2'-deoxyguanosine (8-OHdG), apoptosis (cleaved caspase 3), and sustained hypoxia (increased HIF-1α and VEGF/PKCε/extracellular signal regulated kinase or ERK pathway). Microocclusion enhanced the loss of the synaptic marker spinophilin, astrocytic number, and astrocyte-vascular coupling areas and demyelination of axons. DCPLA-ME prevented spatial memory defect; strong oxidative stress-related apoptosis; sustained hypoxia (by reducing HIF-1α and VEGF); and exaggerated cell repair in arteriolar walls, pericapillary space dilation, neuro-glial-vascular disruption, and demyelination. Conclusion In conclusion, in 3×Tg mice with cerebral microinfarcts, sustained hypoxia (increased HIF-1α and VEGF signals) is dominant with arteriolar wall thickening, and DCPLA has a protective effect on sustained hypoxia.
Collapse
Affiliation(s)
| | | | - Jarin Hongpaisan
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
13
|
Haidar H, Majzoub RE, Hajeer S, Abbas LA. Arterial spin labeling (ASL-MRI) versus fluorodeoxyglucose-PET (FDG-PET) in diagnosing dementia: a systematic review and meta-analysis. BMC Neurol 2023; 23:385. [PMID: 37875879 PMCID: PMC10594722 DOI: 10.1186/s12883-023-03432-y] [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: 11/14/2022] [Accepted: 10/10/2023] [Indexed: 10/26/2023] Open
Abstract
BACKGROUND Dementia is generally caused by neurodegenerative diseases affecting the brain, which leads to a progressive neurocognitive decline characterized by inability to perform major higher functioning tasks. Fluorodeoxyglucose-positron emission tomography (FDG-PET) scan is one of the main imaging tests performed for diagnostic purposes. However, with FDG-PET being quite expensive and not widely available, an attempt to find an alternative is set. Arterial-spin-labelling magnetic resonance imaging (ASL-MRI) is an increasingly investigated substitute to FDG-PET for the diagnosis of dementia. Thereby, the main purpose of this systematic review and meta-analysis is to compare the diagnostic ability of FDG-PET and ASL-MRI in detecting dementia. METHODS PRISMA checklist for diagnostic test accuracy was employed in outlining this paper. A literature search was done using several search engines including PubMed, Core, and Cochrane. Two researchers (HH and SH) extracted the essential information from all included articles. Risk of bias was evaluated by the Quality Assessment of Diagnostic Accuracy Studies tool, version 2 (QUADAS-2). A qualitative analysis and summary of studies' results were provided. In addition, a meta-analysis was executed based on the studies which involved sensitivity and specificity measures of diagnostic accuracy. RESULTS Fourteen total studies were included in the given review. Qualitative analysis of the articles showed that nine studies demonstrated an overlap between metabolic and perfused brain maps as derived by FDG-PET and ASL-MRI respectively, while the remaining five studies registered significant differences across both modalities, with superiority to FDG-PET. As for the meta-analysis implemented, summary ROC-curve analysis revealed that FDG-PET performed better than ASL-MRI, with pooled sensitivity being significantly higher for FDG-PET. CONCLUSIONS Comparing the diagnostic value of FDG-PET and ASL-MRI, the results of this systematic review and meta-analysis indicate that FDG-PET still has an advantage over ASL-MRI. Such implication could be related to the technical differences relating to both modalities, with ASL-MRI having lower temporal resolution. It's worth mentioning that specificity was rather quite similar among both modalities and some studies found an overridden metabolic and perfused images. These findings call for future research to focus their scope of investigation while exploring the diagnostic value of ASL-MRI.
Collapse
Affiliation(s)
- Hiba Haidar
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon.
| | - Rania El Majzoub
- Department of Biomedical Sciences, School of Pharmacy, Lebanese International University, Beirut, Lebanon
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Shorouk Hajeer
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| | - Linda Abou Abbas
- Neuroscience Research Center, Faculty of Medical Sciences, Lebanese University, Beirut, Lebanon
| |
Collapse
|
14
|
Lista S, González-Domínguez R, López-Ortiz S, González-Domínguez Á, Menéndez H, Martín-Hernández J, Lucia A, Emanuele E, Centonze D, Imbimbo BP, Triaca V, Lionetto L, Simmaco M, Cuperlovic-Culf M, Mill J, Li L, Mapstone M, Santos-Lozano A, Nisticò R. Integrative metabolomics science in Alzheimer's disease: Relevance and future perspectives. Ageing Res Rev 2023; 89:101987. [PMID: 37343679 DOI: 10.1016/j.arr.2023.101987] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 06/12/2023] [Accepted: 06/15/2023] [Indexed: 06/23/2023]
Abstract
Alzheimer's disease (AD) is determined by various pathophysiological mechanisms starting 10-25 years before the onset of clinical symptoms. As multiple functionally interconnected molecular/cellular pathways appear disrupted in AD, the exploitation of high-throughput unbiased omics sciences is critical to elucidating the precise pathogenesis of AD. Among different omics, metabolomics is a fast-growing discipline allowing for the simultaneous detection and quantification of hundreds/thousands of perturbed metabolites in tissues or biofluids, reproducing the fluctuations of multiple networks affected by a disease. Here, we seek to critically depict the main metabolomics methodologies with the aim of identifying new potential AD biomarkers and further elucidating AD pathophysiological mechanisms. From a systems biology perspective, as metabolic alterations can occur before the development of clinical signs, metabolomics - coupled with existing accessible biomarkers used for AD screening and diagnosis - can support early disease diagnosis and help develop individualized treatment plans. Presently, the majority of metabolomic analyses emphasized that lipid metabolism is the most consistently altered pathway in AD pathogenesis. The possibility that metabolomics may reveal crucial steps in AD pathogenesis is undermined by the difficulty in discriminating between the causal or epiphenomenal or compensatory nature of metabolic findings.
Collapse
Affiliation(s)
- Simone Lista
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid, Spain.
| | - Raúl González-Domínguez
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, Universidad de Cádiz, Cádiz, Spain
| | - Susana López-Ortiz
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid, Spain
| | - Álvaro González-Domínguez
- Instituto de Investigación e Innovación Biomédica de Cádiz (INiBICA), Hospital Universitario Puerta del Mar, Universidad de Cádiz, Cádiz, Spain
| | - Héctor Menéndez
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid, Spain
| | - Juan Martín-Hernández
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid, Spain
| | - Alejandro Lucia
- Research Institute of the Hospital 12 de Octubre ('imas12'), Madrid, Spain; Faculty of Sport Sciences, European University of Madrid, Villaviciosa de Odón, Madrid, Spain; CIBER of Frailty and Healthy Ageing (CIBERFES), Madrid, Spain
| | | | - Diego Centonze
- Department of Systems Medicine, Tor Vergata University, Rome, Italy; Unit of Neurology, IRCCS Neuromed, Pozzilli, IS, Italy
| | - Bruno P Imbimbo
- Department of Research and Development, Chiesi Farmaceutici, Parma, Italy
| | - Viviana Triaca
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), Rome, Italy
| | - Luana Lionetto
- Clinical Biochemistry, Mass Spectrometry Section, Sant'Andrea University Hospital, Rome, Italy; Department of Neuroscience, Mental Health and Sensory Organs, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Maurizio Simmaco
- Clinical Biochemistry, Mass Spectrometry Section, Sant'Andrea University Hospital, Rome, Italy; Department of Neuroscience, Mental Health and Sensory Organs, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Italy
| | - Miroslava Cuperlovic-Culf
- Digital Technologies Research Center, National Research Council, Ottawa, Canada; Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Jericha Mill
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Lingjun Li
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA; School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
| | - Mark Mapstone
- Department of Neurology, Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA, USA
| | - Alejandro Santos-Lozano
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid, Spain; Research Institute of the Hospital 12 de Octubre ('imas12'), Madrid, Spain
| | - Robert Nisticò
- School of Pharmacy, University of Rome "Tor Vergata", Rome, Italy; Laboratory of Pharmacology of Synaptic Plasticity, EBRI Rita Levi-Montalcini Foundation, Rome, Italy
| |
Collapse
|
15
|
Mao C, You H, Hou B, Chu S, Jin W, Huang X, Shang L, Feng F, Peng B, Gao J. Differentiation of Alzheimer’s Disease from Frontotemporal Dementia and Mild Cognitive Impairment Based on Arterial Spin Labeling Magnetic Resonance Imaging: A Pilot Cross-Sectional Study from PUMCH Dementia Cohort. J Alzheimers Dis 2023; 93:509-519. [PMID: 37038812 DOI: 10.3233/jad-221023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2023]
Abstract
Background: Arterial spin labeling (ASL) is helpful in early diagnosis and differential diagnosis of Alzheimer’s disease (AD), with advantages including no exposure to radioactivity, no injection of a contrast agent, more accessible, and relatively less expensive. Objective: To establish the perfusion pattern of different dementia in Chinese population and evaluate the effectiveness of ASL in differentiating AD from cognitive unimpaired (CU), mild cognitive impairment (MCI), and frontotemporal dementia (FTD). Methods: Four groups of participants were enrolled, including AD, FTD, MCI, and CU based on clinical diagnosis from PUMCH dementia cohort. ASL image was collected using 3D spiral fast spin echo–based pseudo-continuous ASL pulse sequence with background suppression and a high resolution T1-weighted scan covering the whole brain. Data processing was performed using Dr. Brain Platform to get cerebral blood flow (ml/100g/min) in every region of interest cortices. Results: Participants included 66 AD, 26 FTD, 21 MCI, and 21 CU. Statistically, widespread hypoperfusion neocortices, most significantly in temporal-parietal-occipital cortices, but not hippocampus and subcortical nucleus were found in AD. Hypoperfusion in parietal lobe was most significantly associated with cognitive decline in AD. Hypoperfusion in parietal lobe was found in MCI and extended to adjacent temporal, occipital and posterior cingulate cortices in AD. Significant reduced perfusion in frontal and temporal cortices, including subcortical nucleus and anterior cingulate cortex were found in FTD. Hypoperfusion regions were relatively symmetrical in AD and left predominant especially in FTD. Conclusion: Specific patterns of ASL hypoperfusion were helpful in differentiating AD from CU, MCI, and FTD.
Collapse
Affiliation(s)
- Chenhui Mao
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science/Peking Union Medical College, Beijing, China
| | - Hui You
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of MedicalScience/ Peking Union Medical College, Beijing, China
| | - Bo Hou
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of MedicalScience/ Peking Union Medical College, Beijing, China
| | - Shanshan Chu
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science/Peking Union Medical College, Beijing, China
| | - Wei Jin
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science/Peking Union Medical College, Beijing, China
| | - Xinying Huang
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science/Peking Union Medical College, Beijing, China
| | - Li Shang
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science/Peking Union Medical College, Beijing, China
| | - Feng Feng
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of MedicalScience/ Peking Union Medical College, Beijing, China
| | - Bin Peng
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science/Peking Union Medical College, Beijing, China
| | - Jing Gao
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science/Peking Union Medical College, Beijing, China
| |
Collapse
|
16
|
Ahmadi K, Pereira JB, Berron D, Vogel J, Ingala S, Strandberg OT, Janelidze S, Barkhof F, Pfeuffer J, Knutsson L, van Westen D, Palmqvist S, Mutsaerts HJ, Hansson O. Gray matter hypoperfusion is a late pathological event in the course of Alzheimer's disease. J Cereb Blood Flow Metab 2023; 43:565-580. [PMID: 36412244 PMCID: PMC10063832 DOI: 10.1177/0271678x221141139] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Several studies have shown decreased cerebral blood flow (CBF) in Alzheimer's disease (AD). However, the role of hypoperfusion in the disease pathogenesis remains unclear. Combining arterial spin labeling MRI, PET, and CSF biomarkers, we investigated the associations between gray matter (GM)-CBF and the key mechanisms in AD including amyloid-β (Aβ) and tau pathology, synaptic and axonal degeneration. Further, we applied a disease progression modeling to characterize the temporal sequence of different AD biomarkers. Lower perfusion was observed in temporo-occipito-parietal cortex in the Aβ-positive cognitively impaired compared to both Aβ-negative and Aβ-positive cognitively unimpaired individuals. In participants along the AD spectrum, GM-CBF was associated with tau, synaptic and axonal dysfunction, but not Aβ in similar cortical regions. Axonal degeneration was further associated with hypoperfusion in cognitively unimpaired individuals. Disease progression modeling revealed that GM-CBF disruption Followed the abnormality of biomarkers of Aβ, tau and brain atrophy. These findings indicate that tau tangles and neurodegeneration are more closely connected with GM-CBF changes than Aβ pathology. Although subjected to the sensitivity of the employed neuroimaging techniques and the modeling approach, these findings suggest that hypoperfusion might not be an early event associated with the build-up of Aβ in preclinical phase of AD.
Collapse
Affiliation(s)
- Khazar Ahmadi
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden.,Institute of Cognitive Neuroscience, Faculty of Psychology, Ruhr University Bochum, Bochum, Germany
| | - Joana B Pereira
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden.,Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institute, Stockholm, Sweden
| | - David Berron
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Jacob Vogel
- Department of Psychiatry, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Silvia Ingala
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Location VUmc, Amsterdam Neuroscience, Amsterdam, the Netherlands
| | - Olof T Strandberg
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Shorena Janelidze
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Location VUmc, Amsterdam Neuroscience, Amsterdam, the Netherlands.,Queen's Square Institute of Neurology and Centre for Medical Image Computing, University College London, London, UK
| | - Josef Pfeuffer
- Application Development, Siemens Healthcare, Erlangen, Germany
| | - Linda Knutsson
- Department of Medical Radiation Physics, Lund University, Lund, Sweden.,The Russell H. Morgan Department of Radiology and Radiological Science, Division of MR Research, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Danielle van Westen
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden.,Diagnostic Radiology, Lund University, Lund, Sweden
| | - Sebastian Palmqvist
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden.,Department of Neurology, Skåne University Hospital, Lund, Sweden
| | - Henk Jmm Mutsaerts
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Location VUmc, Amsterdam Neuroscience, Amsterdam, the Netherlands.,Queen's Square Institute of Neurology and Centre for Medical Image Computing, University College London, London, UK
| | - Oskar Hansson
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Lund, Sweden.,Memory Clinic, Skåne University Hospital, Malmö, Sweden
| |
Collapse
|
17
|
Salminen A. Activation of aryl hydrocarbon receptor (AhR) in Alzheimer's disease: role of tryptophan metabolites generated by gut host-microbiota. J Mol Med (Berl) 2023; 101:201-222. [PMID: 36757399 PMCID: PMC10036442 DOI: 10.1007/s00109-023-02289-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 12/19/2022] [Accepted: 01/17/2023] [Indexed: 02/10/2023]
Abstract
Gut microbiota in interaction with intestinal host tissues influences many brain functions and microbial dysbiosis has been linked with brain disorders, such as neuropsychiatric conditions and Alzheimer's disease (AD). L-tryptophan metabolites and short-chained fatty acids (SCFA) are major messengers in the microbiota-brain axis. Aryl hydrocarbon receptors (AhR) are main targets of tryptophan metabolites in brain microvessels which possess an enriched expression of AhR protein. The Ah receptor is an evolutionarily conserved, ligand-activated transcription factor which is not only a sensor of xenobiotic toxins but also a pleiotropic regulator of both developmental processes and age-related tissue degeneration. Major microbiota-produced tryptophan metabolites involve indole derivatives, e.g., indole 3-pyruvic acid, indole 3-acetaldehyde, and indoxyl sulfate, whereas indoleamine and tryptophan 2,3-dioxygenases (IDO/TDO) of intestine host cells activate the kynurenine (KYN) pathway generating KYN metabolites, many of which are activators of AhR signaling. Chronic kidney disease (CKD) increases the serum level of indoxyl sulfate which promotes AD pathogenesis, e.g., it disrupts integrity of blood-brain barrier (BBB) and impairs cognitive functions. Activation of AhR signaling disturbs vascular homeostasis in brain; (i) it controls blood flow via the renin-angiotensin system, (ii) it inactivates endothelial nitric oxide synthase (eNOS), thus impairing NO production and vasodilatation, and (iii) it induces oxidative stress, stimulates inflammation, promotes cellular senescence, and enhances calcification of vascular walls. All these alterations are evident in cerebral amyloid angiopathy (CAA) in AD pathology. Moreover, AhR signaling can disturb circadian regulation and probably affect glymphatic flow. It seems plausible that dysbiosis of gut microbiota impairs the integrity of BBB via the activation of AhR signaling and thus aggravates AD pathology. KEY MESSAGES: Dysbiosis of gut microbiota is associated with dementia and Alzheimer's disease. Tryptophan metabolites are major messengers from the gut host-microbiota to brain. Tryptophan metabolites activate aryl hydrocarbon receptor (AhR) signaling in brain. The expression of AhR protein is enriched in brain microvessels and blood-brain barrier. Tryptophan metabolites disturb brain vascular integrity via AhR signaling. Dysbiosis of gut microbiota promotes inflammation and AD pathology via AhR signaling.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, Kuopio, 70211, Finland.
| |
Collapse
|
18
|
Bolton CJ, Tam JW. Differential Involvement of the Locus Coeruleus in Early- and Late-Onset Alzheimer's Disease: A Potential Mechanism of Clinical Differences? J Geriatr Psychiatry Neurol 2022; 35:733-739. [PMID: 34496652 DOI: 10.1177/08919887211044755] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Sporadic early-onset Alzheimer's disease (sEOAD) is often associated with atypical clinical features, yet the cause of this heterogeneity remains unclear. This study investigated post-mortem atrophy of the locus coeruleus (LC) in sEOAD and late-onset Alzheimer's disease (LOAD). Levels of LC atrophy, as estimated by pathologist-rating of hypopigmentation, were compared between sEOAD (n = 115) and LOAD (n = 672) participants while controlling for other measures of pathological progression. Subsequent analyses compared low vs. high LC atrophy sEOAD subgroups on neuropsychological test performance. Results show nearly 4 times greater likelihood of higher LC atrophy in sEOAD as compared to LOAD (p < .005). sEOAD participants with greater LC atrophy displayed significantly worse performance on various baseline measures of attentional functioning (p < .05), despite similar global cognition (p = .25). These findings suggest the LC is an important potential driver of clinical and pathological heterogeneity in sEOAD.
Collapse
Affiliation(s)
- Corey J Bolton
- Vanderbilt Memory and Alzheimer's Center, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joyce W Tam
- Department of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
19
|
Sharma AL, Wang H, Zhang Z, Millien G, Tyagi M, Hongpaisan J. HIV Promotes Neurocognitive Impairment by Damaging the Hippocampal Microvessels. Mol Neurobiol 2022; 59:4966-4986. [PMID: 35665894 PMCID: PMC10071835 DOI: 10.1007/s12035-022-02890-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/18/2022] [Indexed: 10/18/2022]
Abstract
Current evidence suggests that mild cerebrovascular changes could induce neurodegeneration and contribute to HIV-associated neurocognitive disease (HAND) in HIV patients. We investigated both the quantitative and qualitative impact of HIV infection on brain microvessels, especially on hippocampal microvessels, which are crucial for optimal O2 supply, and thus for maintaining memory and cognitive abilities. The results obtained using cultured human brain microvascular endothelial cells (HBMEC) were reproduced using a suitable mouse model and autopsied human HIV hippocampus. In HBMEC, we found significantly higher oxidative stress-dependent apoptotic cell loss following 5 h of treatment of GST-Tat (1 µg/ml) compared to GST (1 µg/ml) control. We noticed complete recovery of HBMEC cells after 24 h of GST-Tat treatment, due to temporal degradation or inactivation of GST-Tat. Interestingly, we found a sustained increase in mitochondrial oxidative DNA damage marker 8-OHdG, as well as an increase in hypoxia-inducible factor hypoxia-inducible factor-1α (HIF-1α). In our mouse studies, upon short-term injection of GST-Tat, we found the loss of small microvessels (mostly capillaries) and vascular endothelial growth factor (VEGF), but not large microvessels (arterioles and venules) in the hippocampus. In addition to capillary loss, in the post-mortem HIV-infected human hippocampus, we observed large microvessels with increased wall cells and perivascular tissue degeneration. Together, our data show a crucial role of Tat in inducing HIF-1α-dependent inhibition of mitochondrial transcriptional factor A (TFAM) and dilated perivascular space. Thus, our results further define the underlying molecular mechanism promoting mild cerebrovascular disease, neuropathy, and HAND pathogenesis in HIV patients.
Collapse
Affiliation(s)
- Adhikarimayum Lakhikumar Sharma
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust Street, Jefferson Alumni Hall, PA, 19107, Philadelphia, USA
| | - Huaixing Wang
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust Street, Jefferson Alumni Hall, PA, 19107, Philadelphia, USA
| | - Zongxiu Zhang
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust Street, Jefferson Alumni Hall, PA, 19107, Philadelphia, USA
| | - Guetchyn Millien
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust Street, Jefferson Alumni Hall, PA, 19107, Philadelphia, USA
| | - Mudit Tyagi
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust Street, Jefferson Alumni Hall, PA, 19107, Philadelphia, USA.
| | - Jarin Hongpaisan
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, 1020 Locust Street, Jefferson Alumni Hall, PA, 19107, Philadelphia, USA.
| |
Collapse
|
20
|
Xiong Y, Chen RS, Wang XY, Li X, Dai LQ, Yu RQ. Cerebral blood flow in adolescents with drug-naive, first-episode major depressive disorder: An arterial spin labeling study based on voxel-level whole-brain analysis. Front Neurosci 2022; 16:966087. [PMID: 35968369 PMCID: PMC9363766 DOI: 10.3389/fnins.2022.966087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 07/07/2022] [Indexed: 11/13/2022] Open
Abstract
PurposeThe major depressive disorder (MDD) can be a threat to the health of people all over the world. Although governments have developed and implemented evidence-based interventions and prevention programs to prevent MDD and maintain mental health in adolescents, the number of adolescents with this condition has been on the rise for the past 10 years.MethodsA total of 60 adolescents were recruited, including 32 drug-naive adolescents with first-episode MDD and 28 healthy controls (HCs). Alterations in the intrinsic cerebral activity of the adolescents with MDD were explored using arterial spin labeling (ASL) while differences in the regional cerebral blood flow (rCBF) of the two groups were assessed based on voxel-based whole-brain analysis. Finally, correlations between the regional functional abnormalities and clinical variables were investigated for adolescents with MDD.ResultsCompared with HCs, MDD patients had a lower rCBF in the left triangular part of the inferior frontal gyrus (IFGtriang) but a higher one in the right Precental gyrus (PreCG). Negative correlations were also noted between the CBF in the left IFGtriang and the Hamilton depression scale (HAMD) scores of MDD patients.ConclusionElucidating the neurobiological features of adolescent patients with MDD is important to adequately develop methods that can assist in early diagnosis, precaution and intervention.
Collapse
Affiliation(s)
- Ying Xiong
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Hematology, Chongqing General Hospital, Chongqing, China
| | - Rong-Sheng Chen
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xing-Yu Wang
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiao Li
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lin-Qi Dai
- Department of Psychiatry, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ren-Qiang Yu
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- *Correspondence: Ren-Qiang Yu,
| |
Collapse
|
21
|
Barloese MCJ, Bauer C, Petersen ET, Hansen CS, Madsbad S, Siebner HR. Neurovascular Coupling in Type 2 Diabetes With Cognitive Decline. A Narrative Review of Neuroimaging Findings and Their Pathophysiological Implications. Front Endocrinol (Lausanne) 2022; 13:874007. [PMID: 35860697 PMCID: PMC9289474 DOI: 10.3389/fendo.2022.874007] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 05/17/2022] [Indexed: 01/21/2023] Open
Abstract
Type 2 diabetes causes substantial long-term damage in several organs including the brain. Cognitive decline is receiving increased attention as diabetes has been established as an independent risk factor along with the identification of several other pathophysiological mechanisms. Early detection of detrimental changes in cerebral blood flow regulation may represent a useful clinical marker for development of cognitive decline for at-risk persons. Technically, reliable evaluation of neurovascular coupling is possible with several caveats but needs further development before it is clinically convenient. Different modalities including ultrasound, positron emission tomography and magnetic resonance are used preclinically to shed light on the many influences on vascular supply to the brain. In this narrative review, we focus on the complex link between type 2 diabetes, cognition, and neurovascular coupling and discuss how the disease-related pathology changes neurovascular coupling in the brain from the organ to the cellular level. Different modalities and their respective pitfalls are covered, and future directions suggested.
Collapse
Affiliation(s)
- Mads C. J. Barloese
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
- Department of Clinical Physiology and Nuclear Imaging, Center for Functional and Diagnostic Imaging, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
| | - Christian Bauer
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
- Radiography, Department of Technology, University College Copenhagen, Copenhagen, Denmark
| | - Esben Thade Petersen
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
- Center for Magnetic Resonance, Department of Electrical Engineering, Technical University of Denmark, Lyngby, Denmark
| | | | - Sten Madsbad
- Department of Endocrinology, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hartwig Roman Siebner
- Danish Research Centre for Magnetic Resonance, Centre for Functional and Diagnostic Imaging and Research, Copenhagen University Hospital - Amager and Hvidovre, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Neurology, Copenhagen University Hospital - Bispebjerg and Fredriksberg, Copenhagen, Denmark
| |
Collapse
|
22
|
Xiao W, Su J, Gao X, Yang H, Weng R, Ni W, Gu Y. The microbiota-gut-brain axis participates in chronic cerebral hypoperfusion by disrupting the metabolism of short-chain fatty acids. MICROBIOME 2022; 10:62. [PMID: 35430804 PMCID: PMC9013454 DOI: 10.1186/s40168-022-01255-6] [Citation(s) in RCA: 95] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 02/28/2022] [Indexed: 05/02/2023]
Abstract
BACKGROUND Chronic cerebral hypoperfusion (CCH) underlies secondary brain injury following certain metabolic disorders and central nervous system (CNS) diseases. Dysregulation of the microbiota-gut-brain axis can exacerbate various CNS disorders through aberrantly expressed metabolites such as short-chain fatty acids (SCFAs). Yet, its relationship with CCH remains to be demonstrated. And if so, it is of interest to explore whether restoring gut microbiota to maintain SCFA metabolism could protect against CCH. RESULTS Rats subjected to bilateral common carotid artery occlusion (BCCAO) as a model of CCH exhibited cognitive impairment, depressive-like behaviors, decreased gut motility, and compromised gut barrier functions. The 16S ribosomal RNA gene sequencing revealed an abnormal gut microbiota profile and decreased relative abundance of some representative SCFA producers, with the decreased hippocampal SCFAs as the further evidence. Using fecal microbiota transplantation (FMT), rats recolonized with a balanced gut microbiome acquired a higher level of hippocampal SCFAs, as well as decreased neuroinflammation when exposed to lipopolysaccharide. Healthy FMT promoted gut motility and gut barrier functions, and improved cognitive decline and depressive-like behaviors by inhibiting hippocampal neuronal apoptosis in BCCAO rats. Long-term SCFA supplementation further confirmed its neuroprotective effect in terms of relieving inflammatory response and hippocampal neuronal apoptosis following BCCAO. CONCLUSION Our results demonstrate that modulating the gut microbiome via FMT can ameliorate BCCAO-induced gut dysbiosis, cognitive decline, and depressive-like behaviors, possibly by enhancing the relative abundance of SCFA-producing floras and subsequently increasing SCFA levels. Video abstract.
Collapse
Affiliation(s)
- Weiping Xiao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040 China
- Institute of Neurosurgery, Fudan University, Shanghai, 200052 China
| | - Jiabin Su
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040 China
- Institute of Neurosurgery, Fudan University, Shanghai, 200052 China
| | - Xinjie Gao
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040 China
- Institute of Neurosurgery, Fudan University, Shanghai, 200052 China
| | - Heng Yang
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040 China
- Institute of Neurosurgery, Fudan University, Shanghai, 200052 China
| | - Ruiyuan Weng
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040 China
- Institute of Neurosurgery, Fudan University, Shanghai, 200052 China
| | - Wei Ni
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040 China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, 200052 China
| | - Yuxiang Gu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, 200040 China
- Institute of Neurosurgery, Fudan University, Shanghai, 200052 China
| |
Collapse
|
23
|
Recent Advances in Understanding of Alzheimer's Disease Progression through Mass Spectrometry-Based Metabolomics. PHENOMICS (CHAM, SWITZERLAND) 2022; 2:1-17. [PMID: 35656096 PMCID: PMC9159642 DOI: 10.1007/s43657-021-00036-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia in the aging population, but despite extensive research, there is no consensus on the biological cause of AD. While AD research is dominated by protein/peptide-centric research based on the amyloid hypothesis, a theory that designates dysfunction in beta-amyloid production, accumulation, or disposal as the primary cause of AD, many studies focus on metabolomics as a means of understanding the biological processes behind AD progression. In this review, we discuss mass spectrometry (MS)-based AD metabolomics studies, including sample type and preparation, mass spectrometry specifications, and data analysis, as well as biological insights gleaned from these studies, with the hope of informing future AD metabolomic studies.
Collapse
|
24
|
Imaging Clinical Subtypes and Associated Brain Networks in Alzheimer’s Disease. Brain Sci 2022; 12:brainsci12020146. [PMID: 35203910 PMCID: PMC8869882 DOI: 10.3390/brainsci12020146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 01/17/2022] [Accepted: 01/18/2022] [Indexed: 11/17/2022] Open
Abstract
Alzheimer’s disease (AD) does not present uniform symptoms or a uniform rate of progression in all cases. The classification of subtypes can be based on clinical symptoms or patterns of pathological brain alterations. Imaging techniques may allow for the identification of AD subtypes and their differentiation from other neurodegenerative diseases already at an early stage. In this review, the strengths and weaknesses of current clinical imaging methods are described. These include positron emission tomography (PET) to image cerebral glucose metabolism and pathological amyloid or tau deposits. Magnetic resonance imaging (MRI) is more widely available than PET. It provides information on structural or functional changes in brain networks and their relation to AD subtypes. Amyloid PET provides a very early marker of AD but does not distinguish between AD subtypes. Regional patterns of pathology related to AD subtypes are observed with tau and glucose PET, and eventually as atrophy patterns on MRI. Structural and functional network changes occur early in AD but have not yet provided diagnostic specificity.
Collapse
|
25
|
Li J, Zhang B, Jia W, Yang M, Zhang Y, Zhang J, Li L, Jin T, Wang Z, Tao J, Chen L, Liang S, Liu W. Activation of Adenosine Monophosphate-Activated Protein Kinase Drives the Aerobic Glycolysis in Hippocampus for Delaying Cognitive Decline Following Electroacupuncture Treatment in APP/PS1 Mice. Front Cell Neurosci 2021; 15:774569. [PMID: 34867206 PMCID: PMC8636716 DOI: 10.3389/fncel.2021.774569] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 10/12/2021] [Indexed: 12/03/2022] Open
Abstract
Aerobic glycolysis (AG), an important pathway of glucose metabolism, is dramatically declined in Alzheimer’s disease (AD). AMP-activated protein kinase (AMPK) is a key regulator to maintain the stability of energy metabolism by promoting the process of AG and regulating glucose metabolism. Interestingly, it has been previously reported that electroacupuncture (EA) treatment can improve cognitive function in AD through the enhancement of glucose metabolism. In this study, we generated AMPK-knockdown mice to confirm the EA effect on AMPK activation and further clarify the mechanism of EA in regulating energy metabolism and improving cognitive function in APP/PS1 mice. The behavioral results showed that EA treatment can improve the learning and memory abilities in APP/PS1 mice. At the same time, the glucose metabolism in the hippocampus was increased detected by MRI-chemical exchange saturation transfer (MRI-CEST). The expression of proteins associated with AG in the hippocampus was increased simultaneously, including hexokinase II (HK2), 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (PFKFB3), and pyruvate kinase M2 (PKM2). Moreover, the knockdown of AMPK attenuated AG activated by EA treatment. In conclusion, this study proves that EA can activate AMPK to enhance the process of AG in the early stage of AD.
Collapse
Affiliation(s)
- Jianhong Li
- TCM Rehabilitation Research Center of SATCM, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Bingxue Zhang
- TCM Rehabilitation Research Center of SATCM, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Weiwei Jia
- TCM Rehabilitation Research Center of SATCM, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Minguang Yang
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yuhao Zhang
- TCM Rehabilitation Research Center of SATCM, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jiayong Zhang
- TCM Rehabilitation Research Center of SATCM, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Le Li
- TCM Rehabilitation Research Center of SATCM, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Tingting Jin
- TCM Rehabilitation Research Center of SATCM, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Zhifu Wang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jing Tao
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Lidian Chen
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Shengxiang Liang
- TCM Rehabilitation Research Center of SATCM, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Weilin Liu
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| |
Collapse
|
26
|
Salminen A, Kaarniranta K, Kauppinen A. Hypoxia/ischemia impairs CD33 (Siglec-3)/TREM2 signaling: Potential role in Alzheimer's pathogenesis. Neurochem Int 2021; 150:105186. [PMID: 34530055 DOI: 10.1016/j.neuint.2021.105186] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 09/10/2021] [Accepted: 09/12/2021] [Indexed: 12/22/2022]
Abstract
Recent genetic and molecular studies have indicated that the innate immune system, especially microglia, have a crucial role in the accumulation of β-amyloid plaques in Alzheimer's disease (AD). In particular, the CD33 receptor, also called Siglec-3, inhibits the TREM2 receptor-induced phagocytic activity of microglia. CD33 receptors recognize the α2,3 and α2,6-linked sialic groups in tissue glycocalyx, especially sialylated gangliosides in human brain. The CD33 receptor triggers cell-type specific responses, e.g., in microglia, CD33 inhibits phagocytosis, whereas in natural killer cells, it inhibits the cytotoxic activity of the NKG2D receptor. Nonetheless, the regulation of the activity of CD33 receptor needs to be clarified. For example, it seems that hypoxia/ischemia, a potential cause of AD pathology, increases the expression of CD33 and its downstream target SHP-1, a tyrosine phosphatase which suppresses the phagocytosis driven by TREM2. Moreover, hypoxia/ischemia increases the deposition of sialylated gangliosides, e.g., GM1, GM2, GM3, and GD1, which are ligands for inhibitory CD33/Siglec-3 receptors. In addition, β-amyloid peptides bind to the sialylated gangliosides in raft-like clusters and subsequently these gangliosides act as seeds for the formation of β-amyloid plaques in AD pathology. It is known that senile plaques contain sialylated GM1, GM2, and GM3 gangliosides, i.e., the same species induced by hypoxia/ischemia treatment. Sialylated gangliosides in plaques might stimulate the CD33/Siglec-3 receptors of microglia and thus impede TREM2-driven phagocytosis. We propose that hypoxia/ischemia, e.g., via the accumulation of sialylated gangliosides, prevents the phagocytosis of β-amyloid deposits by inhibiting CD33/TREM2 signaling.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland; Department of Ophthalmology, Kuopio University Hospital, P.O. Box 100, FI-70029, KYS, Finland
| | - Anu Kauppinen
- School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| |
Collapse
|
27
|
Kanno S, Ogawa KI, Kikuchi H, Toyoshima M, Abe N, Sato K, Miyazawa K, Oshima R, Ohtomo S, Arai H, Shibuya S, Suzuki K. Reduced default mode network connectivity relative to white matter integrity is associated with poor cognitive outcomes in patients with idiopathic normal pressure hydrocephalus. BMC Neurol 2021; 21:353. [PMID: 34517828 PMCID: PMC8436532 DOI: 10.1186/s12883-021-02389-0] [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: 06/09/2021] [Accepted: 09/06/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The aim of this study was to investigate whether default mode network (DMN) connectivity and brain white matter integrity at baseline were associated with severe cognitive impairments at baseline and poor cognitive outcomes after shunt placement in patients with idiopathic normal pressure hydrocephalus (iNPH). METHODS Twenty consecutive patients with iNPH whose symptoms were followed for 6 months after shunt placement and 10 healthy controls (HCs) were enrolled. DMN connectivity and brain white matter integrity at baseline in the patients with iNPH and HCs were detected by using resting-state functional magnetic resonance imaging (MRI) with independent component analysis and diffusion tensor imaging, respectively, and these MRI indexes were compared between the patients with iNPH and HCs. Performance on neuropsychological tests for memory and executive function and on the gait test was assessed in the patients with iNPH at baseline and 6 months after shunt placement. We divided the patients with iNPH into the relatively preserved and reduced DMN connectivity groups using the MRI indexes for DMN connectivity and brain white matter integrity, and the clinical measures were compared between the relatively preserved and reduced DMN connectivity groups. RESULTS Mean DMN connectivity in the iNPH group was significantly lower than that in the HC group and was significantly positively correlated with Rey auditory verbal learning test (RAVLT) immediate recall scores and frontal assessment battery (FAB) scores. Mean fractional anisotropy of the whole-brain white matter skeleton in the iNPH group was significantly lower than that in the HC group. The reduced DMN connectivity group showed significantly worse performance on the RAVLT at baseline and significantly worse improvement in the RAVLT immediate recall and recognition scores and the FAB scores than the preserved DMN connectivity group. Moreover, the RAVLT recognition score highly discriminated patients with relatively preserved DMN connectivity from those with relatively reduced DMN connectivity. CONCLUSIONS Our findings indicated that iNPH patients with reduced DMN connectivity relative to the severity of brain white matter disruption have severe memory deficits at baseline and poorer cognitive outcomes after shunt placement. However, further larger-scale studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Shigenori Kanno
- Department of Behavioral Neurology and Cognitive Neuroscience, Tohoku University Graduate School of Medicine, 2-1, Seiryo-machi, Aoba-ku, Miyagi, 980-8575, Sendai, Japan. .,Department of Neurology, South Miyagi Medical Center, Shibata, Japan.
| | - Kun-Ichi Ogawa
- Department of Radiology, South Miyagi Medical Center, Shibata, Japan
| | - Hiroaki Kikuchi
- Healthcare Center, South Miyagi Medical Center, Shibata, Japan
| | - Masako Toyoshima
- Department of Rehabilitation, South Miyagi Medical Center, Shibata, Japan
| | - Nobuhito Abe
- Kokoro Research Center, Kyoto University, Kyoto, Japan
| | - Kazushi Sato
- Department of Radiology, South Miyagi Medical Center, Shibata, Japan
| | - Koichi Miyazawa
- Department of Neurology, South Miyagi Medical Center, Shibata, Japan.,Department of Neurology, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Ryuji Oshima
- Department of Neurology, South Miyagi Medical Center, Shibata, Japan
| | - Satoru Ohtomo
- Department of Neurosurgery, South Miyagi Medical Center, Shibata, Japan
| | - Hiroaki Arai
- Department of Neurosurgery, South Miyagi Medical Center, Shibata, Japan
| | - Satoshi Shibuya
- Department of Neurology, South Miyagi Medical Center, Shibata, Japan.,Department of Neurology, Moriyama Memorial Hospital, Edogawa, Japan
| | - Kyoko Suzuki
- Department of Behavioral Neurology and Cognitive Neuroscience, Tohoku University Graduate School of Medicine, 2-1, Seiryo-machi, Aoba-ku, Miyagi, 980-8575, Sendai, Japan
| |
Collapse
|
28
|
Slowed Temporal and Parietal Cerebrovascular Response in Patients with Alzheimer's Disease. Can J Neurol Sci 2021; 47:366-373. [PMID: 32051047 DOI: 10.1017/cjn.2020.30] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Recent investigations now suggest that cerebrovascular reactivity (CVR) is impaired in Alzheimer's disease (AD) and may underpin part of the disease's neurovascular component. However, our understanding of the relationship between the magnitude of CVR, the speed of cerebrovascular response, and the progression of AD is still limited. This is especially true in patients with mild cognitive impairment (MCI), which is recognized as an intermediate stage between normal aging and dementia. The purpose of this study was to investigate AD and MCI patients by mapping repeatable and accurate measures of cerebrovascular function, namely the magnitude and speed of cerebrovascular response (τ) to a vasoactive stimulus in key predilection sites for vascular dysfunction in AD. METHODS Thirty-three subjects (age range: 52-83 years, 20 males) were prospectively recruited. CVR and τ were assessed using blood oxygen level-dependent MRI during a standardized carbon dioxide stimulus. Temporal and parietal cortical regions of interest (ROIs) were generated from anatomical images using the FreeSurfer image analysis suite. RESULTS Of 33 subjects recruited, 3 individuals were excluded, leaving 30 subjects for analysis, consisting of 6 individuals with early AD, 11 individuals with MCI, and 13 older healthy controls (HCs). τ was found to be significantly higher in the AD group compared to the HC group in both the temporal (p = 0.03) and parietal cortex (p = 0.01) following a one-way ANCOVA correcting for age and microangiopathy scoring and a Bonferroni post-hoc correction. CONCLUSION The study findings suggest that AD is associated with a slowing of the cerebrovascular response in the temporal and parietal cortices.
Collapse
|
29
|
Kaliszewska A, Allison J, Martini M, Arias N. Improving Age-Related Cognitive Decline through Dietary Interventions Targeting Mitochondrial Dysfunction. Int J Mol Sci 2021; 22:ijms22073574. [PMID: 33808221 PMCID: PMC8036520 DOI: 10.3390/ijms22073574] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 12/13/2022] Open
Abstract
Aging is inevitable and it is one of the major contributors to cognitive decline. However, the mechanisms underlying age-related cognitive decline are still the object of extensive research. At the biological level, it is unknown how the aging brain is subjected to progressive oxidative stress and neuroinflammation which determine, among others, mitochondrial dysfunction. The link between mitochondrial dysfunction and cognitive impairment is becoming ever more clear by the presence of significant neurological disturbances in human mitochondrial diseases. Possibly, the most important lifestyle factor determining mitochondrial functioning is nutrition. Therefore, with the present work, we review the latest findings disclosing a link between nutrition, mitochondrial functioning and cognition, and pave new ways to counteract cognitive decline in late adulthood through diet.
Collapse
Affiliation(s)
- Aleksandra Kaliszewska
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Denmark Hill, London SE5 8AF, UK; (A.K.); (J.A.)
| | - Joseph Allison
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Denmark Hill, London SE5 8AF, UK; (A.K.); (J.A.)
| | - Matteo Martini
- Department of Psychology, University of East London, London E154LZ, UK;
| | - Natalia Arias
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King’s College London, Denmark Hill, London SE5 8AF, UK; (A.K.); (J.A.)
- Instituto de Neurociencias del Principado de Asturias (INEUROPA), 33005 Oviedo, Spain
- Correspondence:
| |
Collapse
|
30
|
Zammit MD, Laymon CM, Tudorascu DL, Hartley SL, Piro‐Gambetti B, Johnson SC, Stone CK, Mathis CA, Zaman SH, Klunk WE, Handen BL, Cohen AD, Christian BT. Patterns of glucose hypometabolism in Down syndrome resemble sporadic Alzheimer's disease except for the putamen. ALZHEIMER'S & DEMENTIA (AMSTERDAM, NETHERLANDS) 2021; 12:e12138. [PMID: 33490360 PMCID: PMC7804861 DOI: 10.1002/dad2.12138] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 11/09/2020] [Accepted: 11/09/2020] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Adults with Down syndrome (DS) are predisposed to Alzheimer's disease (AD) and the relationship between cognition and glucose metabolism in this population has yet to be evaluated. METHODS Adults with DS (N = 90; mean age [standard deviation] = 38.0 [8.30] years) underwent [C-11]Pittsburgh compound B (PiB) and [F-18]fluorodeoxyglucose (FDG) positron emission tomography scans. Associations among amyloid beta (Aβ), FDG, and measures of cognition were explored. Interregional FDG metabolic connectivity was assessed to compare cognitively stable DS and mild cognitive impairment/AD (MCI-DS/AD). RESULTS Negative associations between Aβ and FDG were evident in regions affected in sporadic AD. A positive association was observed in the putamen, which is the brain region showing the earliest increases in Aβ deposition. Both Aβ and FDG were associated with measures of cognition, and metabolic connectivity distinguished cases of MCI-DS/AD from cognitively stable DS. DISCUSSION Associations among Aβ, FDG, and cognition reveal that neurodegeneration in DS resembles sporadic AD with the exception of the putamen, highlighting the usefulness of FDG in monitoring neurodegeneration in DS.
Collapse
Affiliation(s)
| | - Charles M. Laymon
- Department of Radiology, University of PittsburghPittsburghPennsylvaniaUSA
- Department of Bioengineering, University of PittsburghPittsburghPennsylvaniaUSA
| | - Dana L. Tudorascu
- Department of Psychiatry, University of PittsburghPittsburghPennsylvaniaUSA
| | - Sigan L. Hartley
- University of Wisconsin‐Madison Waisman CenterMadisonWisconsinUSA
| | | | - Sterling C. Johnson
- University of Wisconsin‐Madison Alzheimer's Disease Research CenterMadisonWisconsinUSA
| | - Charles K. Stone
- Department of Medicine, University of Wisconsin‐MadisonMadisonWisconsinUSA
| | - Chester A. Mathis
- Department of Radiology, University of PittsburghPittsburghPennsylvaniaUSA
| | - Shahid H. Zaman
- University of Cambridge Intellectual Disability Research GroupCambridgeUK
| | - William E. Klunk
- Department of Psychiatry, University of PittsburghPittsburghPennsylvaniaUSA
| | - Benjamin L. Handen
- Department of Psychiatry, University of PittsburghPittsburghPennsylvaniaUSA
| | - Ann D. Cohen
- Department of Psychiatry, University of PittsburghPittsburghPennsylvaniaUSA
| | | |
Collapse
|
31
|
Salminen A. Hypoperfusion is a potential inducer of immunosuppressive network in Alzheimer's disease. Neurochem Int 2020; 142:104919. [PMID: 33242538 DOI: 10.1016/j.neuint.2020.104919] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 10/12/2020] [Accepted: 11/19/2020] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease which causes a non-reversible cognitive impairment and dementia. The primary cause of late-onset AD remains unknown although its pathology was discovered over a century ago. Recently, the vascular hypothesis of AD has received backing from evidence emerging from neuroimaging studies which have revealed the presence of a significant hypoperfusion in the brain regions vulnerable to AD pathology. In fact, hypoxia can explain many of the pathological changes evident in AD pathology, e.g. the deposition of β-amyloid plaques and chronic low-grade inflammation. Hypoxia-inducible factor-1α (HIF-1α) stimulates inflammatory responses and modulates both innate and adaptive immunity. It is known that hypoxia-induced inflammation evokes compensatory anti-inflammatory response involving tissue-resident microglia/macrophages and infiltrated immune cells. Hypoxia/HIF-1α induce immunosuppression by (i) increasing the expression of immunosuppressive genes, (ii) stimulating adenosinergic signaling, (iii) enhancing aerobic glycolysis, i.e. lactate production, and (iv) augmenting the secretion of immunosuppressive exosomes. Interestingly, it seems that these common mechanisms are also involved in the pathogenesis of AD. In AD pathology, an enhanced immunosuppression appears, e.g. as a shift in microglia/macrophage phenotypes towards the anti-inflammatory M2 phenotype and an increase in the numbers of regulatory T cells (Treg). The augmented anti-inflammatory capacity promotes the resolution of acute inflammation but persistent inflammation has crucial effects not only on immune cells but also harmful responses to the homeostasis of AD brain. I will examine in detail the mechanisms of the hypoperfusion/hypoxia-induced immunosuppressive state in general and especially, in its association with AD pathogenesis. These immunological observations support the vascular hypothesis of AD pathology.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, FI-70211 Kuopio, Finland.
| |
Collapse
|
32
|
Wang Z. Brain Entropy Mapping in Healthy Aging and Alzheimer's Disease. Front Aging Neurosci 2020; 12:596122. [PMID: 33240080 PMCID: PMC7683386 DOI: 10.3389/fnagi.2020.596122] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 10/06/2020] [Indexed: 12/18/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease, for which aging remains the major risk factor. Aging is under a consistent pressure of increasing brain entropy (BEN) due to the progressive brain deteriorations. Noticeably, the brain constantly consumes a large amount of energy to maintain its functional integrity, likely creating or maintaining a big "reserve" to counteract the high entropy. Malfunctions of this latent reserve may indicate a critical point of disease progression. The purpose of this study was to characterize BEN in aging and AD and to test an inverse-U-shape BEN model: BEN increases with age and AD pathology in normal aging but decreases in the AD continuum. BEN was measured with resting state fMRI and compared across aging and the AD continuum. Associations of BEN with age, education, clinical symptoms, and pathology were examined by multiple regression. The analysis results highlighted resting BEN in the default mode network, medial temporal lobe, and prefrontal cortex and showed that: (1) BEN increased with age and pathological deposition in normal aging but decreased with age and pathological deposition in the AD continuum; (2) AD showed catastrophic BEN reduction, which was related to more severe cognitive impairment and daily function disability; and (3) BEN decreased with education years in normal aging, but not in the AD continuum. BEN evolution follows an inverse-U trajectory when AD progresses from normal aging to AD dementia. Education is beneficial for suppressing the entropy increase potency in normal aging.
Collapse
Affiliation(s)
- Ze Wang
- Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, United States
| | | |
Collapse
|
33
|
Bolton CJ, Tam JW. Differential Involvement of the Locus Coeruleus in Early- and Late-Onset Alzheimer's Disease: A Potential Mechanism of Clinical Differences? MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.11.01.20224139. [PMID: 33173930 PMCID: PMC7654926 DOI: 10.1101/2020.11.01.20224139] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Early-onset Alzheimer's disease (EOAD) has been associated with an increased likelihood of atypical clinical manifestations such as attentional impairment, yet the cause of this heterogeneity remains unclear. The locus coeruleus (LC) is implicated early in Alzheimer's disease pathology and is associated with attentional functioning. This study investigated post-mortem atrophy of the LC in EOAD and late-onset Alzheimer's disease (LOAD) in a large, well-characterized sample. Results show nearly four times greater likelihood of higher LC atrophy in EOAD as compared to LOAD after controlling for other measures of pathological progression ( p < .005). Follow-up analyses within the EOAD group revealed that compared to those who displayed mild or no LC atrophy at autopsy, those with moderate-severe atrophy of the LC displayed significantly worse performance on various baseline measures of attentional functioning ( p < .05), despite similar overall cognition ( p = .25). These findings suggest the LC is an important potential driver of clinical and pathological heterogeneity in EOAD.
Collapse
Affiliation(s)
- Corey J. Bolton
- Vanderbilt Memory & Alzheimer’s Center, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, TN, USA
- Deparment of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, IL, USA
| | - Joyce W. Tam
- Deparment of Psychiatry and Behavioral Sciences, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
34
|
Du J, Fang M, Yu Y, Lu G. An adaptive two-scale biomedical image fusion method with statistical comparisons. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2020; 196:105603. [PMID: 32570007 DOI: 10.1016/j.cmpb.2020.105603] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 06/06/2020] [Indexed: 06/11/2023]
Abstract
Two-scale image representation of base and detail in the spatial-domain is a well-known decomposition scheme for its lower computational complexity than that performed in the transform-domain in the field of image fusion. Unfortunately, for a pseudo-colour input image, the base and detail images in the spatial-domain obtained via image decomposition scheme always display in greyscale. In this paper, a two-scale image fusion method with adaptive threshold obtained by Otsu's method is proposed for pseudo-colour image in the colour space domain. For greyscale image, detail and base image are obtained using structural information extracted from the difference image between a global and a local patch size. Consequently, local edge-preserving filter for preserving luminance information and local energy with the discussed window size are adopted to combine base and detail image. Experimental results show that structural and luminance information has been better preserved in terms of subjective and objective evaluations for medical image and protein image fusion. Specially, a two-step non-parametric statistical test (Friedman test and Nemenyi post-hoc test) with p-values is adopted to analysis the statistical significant of the relative difference between the proposed and compared methods in terms of values of objective metrics including 30 co-registered pairs of imaging data.
Collapse
Affiliation(s)
- Jiao Du
- School of Computer Science and Cyber Engineering, Guangzhou University, Guangzhou 510006, China.
| | - Meie Fang
- School of Computer Science and Cyber Engineering, Guangzhou University, Guangzhou 510006, China
| | - Yufeng Yu
- Department of Statistics, Guangzhou University, Guangzhou 510006, China
| | - Gang Lu
- Laboratory of Image Science and Technology, Key Laboratory of Computer Network and Information Integration, Southeast University, Ministry of Education, Nanjing 210096, China
| |
Collapse
|
35
|
Semantic Processing in Healthy Aging and Alzheimer's Disease: A Systematic Review of the N400 Differences. Brain Sci 2020; 10:brainsci10110770. [PMID: 33114051 PMCID: PMC7690742 DOI: 10.3390/brainsci10110770] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/21/2020] [Accepted: 10/21/2020] [Indexed: 01/18/2023] Open
Abstract
Semantic deficits are common in individuals with Alzheimer’s disease (AD). These deficits notably impact the ability to understand words. In healthy aging, semantic knowledge increases but semantic processing (i.e., the ability to use this knowledge) may be impaired. This systematic review aimed to investigate semantic processing in healthy aging and AD through behavioral responses and the N400 brain event-related potential. The results of the quantitative and qualitative analyses suggested an overall decrease in accuracy and increase in response times in healthy elderly as compared to young adults, as well as in individuals with AD as compared to age-matched controls. The influence of semantic association, as measured by N400 effect amplitudes, appears smaller in healthy aging and even more so in AD patients. Thus, semantic processing differences may occur in both healthy and pathological aging. The establishment of norms of healthy aging for these outcomes that vary between normal and pathological aging could eventually help early detection of AD.
Collapse
|
36
|
Zheng W, Li H, Cui B, Liang P, Wu Y, Han X, Li CR, Li K, Wang Z. Altered multimodal magnetic resonance parameters of basal nucleus of Meynert in Alzheimer's disease. Ann Clin Transl Neurol 2020; 7:1919-1929. [PMID: 32888399 PMCID: PMC7545587 DOI: 10.1002/acn3.51176] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 08/12/2020] [Accepted: 08/19/2020] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVES We aimed to examine how gray matter volume (GMV), regional blood flow (rCBF), and resting-state functional connectivity (FC) of the basal nucleus of Meynert (BNM) are altered in 40 patients with AD, relative to 30 healthy controls (HCs). METHODS We defined the BNM on the basis of a mask histochemically reconstructed from postmortem human brains. We examined GMV with voxel-based morphometry of high-resolution structural images, rCBF with arterial spin labeling imaging, and whole-brain FC with published routines. We performed partial correlations to explore how the imaging metrics related to cognitive and living status in patients with AD. Further, we employed receiver operating characteristic analysis to compute the "diagnostic" accuracy of these imaging markers. RESULTS AD relative to HC showed lower GMV and higher rCBF of the BNM as well as lower BNM connectivity with the right insula and cerebellum. In addition, the GMVs of BNM were correlated with cognitive and daily living status in AD. Finally, these imaging markers predicted AD (vs. HC) with an accuracy (area under the curve) of 0.70 to 0.86. Combination of BNM metrics provided the best prediction accuracy. CONCLUSIONS By combining multimode MR imaging, we demonstrated volumetric atrophy, hyperperfusion, and disconnection of the BNM in AD. These findings support cholinergic dysfunction as an etiological marker of AD and related dementia.
Collapse
Affiliation(s)
- Weimin Zheng
- Department of RadiologyAerospace Center HospitalBeijing100049China
| | - Hui Li
- Department of RadiologyChaoyang Hospital of Capital Medical UniversityBeijing100020China
| | - Bin Cui
- Department of RadiologyAerospace Center HospitalBeijing100049China
| | - Peipeng Liang
- School of PsychologyCapital Normal UniversityBeijing Key Laboratory of Learning and CognitionBeijing100037China
| | - Ye Wu
- Department of RadiologyAerospace Center HospitalBeijing100049China
| | - Xu Han
- Department of RadiologyAerospace Center HospitalBeijing100049China
| | - Chiang‐shan R. Li
- Department of PsychiatryYale University School of MedicineNew HavenConnecticutUSA
- Department of NeuroscienceYale University School of MedicineNew HavenConnecticutUSA
| | - Kuncheng Li
- Department of RadiologyXuanwu Hospital of Capital Medical UniversityBeijing100053China
| | - Zhiqun Wang
- Department of RadiologyAerospace Center HospitalBeijing100049China
| |
Collapse
|
37
|
Giorgi FS, Galgani A, Puglisi-Allegra S, Limanaqi F, Busceti CL, Fornai F. Locus Coeruleus and neurovascular unit: From its role in physiology to its potential role in Alzheimer's disease pathogenesis. J Neurosci Res 2020; 98:2406-2434. [PMID: 32875628 DOI: 10.1002/jnr.24718] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/26/2020] [Accepted: 08/08/2020] [Indexed: 12/15/2022]
Abstract
Locus coeruleus (LC) is the main noradrenergic (NA) nucleus of the central nervous system. LC degenerates early during Alzheimer's disease (AD) and NA loss might concur to AD pathogenesis. Aside from neurons, LC terminals provide dense innervation of brain intraparenchymal arterioles/capillaries, and NA modulates astrocyte functions. The term neurovascular unit (NVU) defines the strict anatomical/functional interaction occurring between neurons, glial cells, and brain vessels. NVU plays a fundamental role in coupling the energy demand of activated brain regions with regional cerebral blood flow, it includes the blood-brain barrier (BBB), plays an active role in neuroinflammation, and participates also to the glymphatic system. NVU alteration is involved in AD pathophysiology through several mechanisms, mainly related to a relative oligoemia in activated brain regions and impairment of structural and functional BBB integrity, which contributes also to the intracerebral accumulation of insoluble amyloid. We review the existing data on the morphological features of LC-NA innervation of the NVU, as well as its contribution to neurovascular coupling and BBB proper functioning. After introducing the main experimental data linking LC with AD, which have repeatedly shown a key role of neuroinflammation and increased amyloid plaque formation, we discuss the potential mechanisms by which the loss of NVU modulation by LC might contribute to AD pathogenesis. Surprisingly, thus far not so many studies have tested directly these mechanisms in models of AD in which LC has been lesioned experimentally. Clarifying the interaction of LC with NVU in AD pathogenesis may disclose potential therapeutic targets for AD.
Collapse
Affiliation(s)
- Filippo Sean Giorgi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy.,Neurology Unit, Pisa University Hospital, Pisa, Italy
| | | | | | - Fiona Limanaqi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | | | - Francesco Fornai
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy.,I.R.C.C.S. I.N.M. Neuromed, Pozzilli, Italy
| |
Collapse
|
38
|
Pelizzari L, Di Tella S, Rossetto F, Laganà MM, Bergsland N, Pirastru A, Meloni M, Nemni R, Baglio F. Parietal Perfusion Alterations in Parkinson's Disease Patients Without Dementia. Front Neurol 2020; 11:562. [PMID: 32655485 PMCID: PMC7324722 DOI: 10.3389/fneur.2020.00562] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 05/18/2020] [Indexed: 12/23/2022] Open
Abstract
Fronto-parietal regions are involved in cognitive processes that are commonly affected in Parkinson's disease (PD). The aims of this study were to investigate cerebral blood flow (CBF) and gray matter (GM) volume within the regions belonging to the fronto-parietal circuit in people with PD (pwPD) without dementia, and to assess their association with cognitive performance. Twenty-seven pwPD without dementia (mean [SD] age = 67.4 [8.1] years, 20 males, mean [SD] Montreal Cognitive Assessment, MoCA score = 24.2 [2.9], median [IQR] Hoehn and Yahr scale = 1.5 [1–2]) and twenty-six age- and sex-matched healthy controls (HC) were scanned with arterial spin labeling (ASL) and T1-weighted magnetic resonance imaging (MRI) sequences to investigate CBF and GM volume, respectively. The cognitive performance of the enrolled pwPD was assessed with MoCA, Trail Making Test (TMT, part A, B, B-A), phonemic fluency and semantic fluency tests. The scores were adjusted for age and education. After standard preprocessing, CBF differences between pwPD and HC were tested with a voxel-wise approach. Voxel-based morphometry was used to compare pwPD and HC in terms of GM volume. Both voxel-wise comparisons between pwPD and HC were restricted to regions of the fronto-parietal circuit. The following additional voxel-wise analyses were performed within regions showing either perfusion or GM volume alterations: (1) correlation with neuropsychological test scores; (2) subgroup comparison after median split on each neuropsychological test score. Family-wise error-corrected (FWE) p-values lower than 0.05 were considered significant. Significant hypoperfusion was identified in the left inferior parietal lobule (IPL, ppeak = 0.037) and in the bilateral superior parietal lobule (SPL, left hemisphere: ppeak = 0.037; right hemisphere: ppeak = 0.049) of pwPD when compared to HC. No significant GM atrophy was observed. Local hypoperfusion did not correlate with any neuropsychological test scores. However, significantly lower CBF was observed in the left SPL and IPL of the pwPD subgroup who performed poorer on TMT part A in comparison with the pwPD subgroup that performed better. Perfusion alterations may occur in parietal regions of pwPD without dementia, and may be associated with lower visuomotor skills. Parietal CBF may be considered as a suitable early biomarker for longitudinal studies investigating cognitive decline in PD.
Collapse
Affiliation(s)
| | | | | | | | - Niels Bergsland
- IRCCS, Fondazione Don Carlo Gnocchi, Milan, Italy.,Buffalo Neuroimaging Analysis Center, Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, United States
| | | | - Mario Meloni
- IRCCS, Fondazione Don Carlo Gnocchi, Milan, Italy
| | - Raffaello Nemni
- IRCCS, Fondazione Don Carlo Gnocchi, Milan, Italy.,Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Milan, Italy
| | | |
Collapse
|
39
|
Chau ACM, Cheung EYW, Chan KH, Chow WS, Shea YF, Chiu PKC, Mak HKF. Impaired cerebral blood flow in type 2 diabetes mellitus - A comparative study with subjective cognitive decline, vascular dementia and Alzheimer's disease subjects. NEUROIMAGE-CLINICAL 2020; 27:102302. [PMID: 32521474 PMCID: PMC7284123 DOI: 10.1016/j.nicl.2020.102302] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 04/27/2020] [Accepted: 05/25/2020] [Indexed: 12/21/2022]
Abstract
CBF impairment is found in T2DM and SCD individuals, which might suggest a preclinical stage of dementia. Comparing to HC, lower CBF in T2DM was due to higher rate of multiple cerebrovascular risk factors. Unlike T2DM, CBF reduction in AD and VD was due to amyloid deposition and microangiopathy respectively. Significant negative correlation between adjusted CBF and HbA1c in all cortical regions in healthy control and T2DM.
The link between non-demented type 2 diabetes mellitus (T2DM) and different types of cognitive impairment is controversial. By controlling for co-morbidities such as cerebral macrovascular and microvascular changes, cerebral atrophy, amyloid burden, hypertension or hyperlipidemia, the current study investigated the cerebral blood flow of T2DM individuals as compared to cognitively impaired subjects recruited from a memory clinic. 15 healthy control (71.8 ± 6.1 years), 18 T2DM (62.5 ± 3.7 years), as well as 8 Subjective Cognitive Decline (69.5 ± 7.5 years), 12 Vascular Dementia (79.3 ± 4.2 years) and 17 Alzheimer’s Disease (75.1 ± 8.2 years) underwent multi-parametric MRI brain scanning. Subjects with T2DM and from the memory clinic also had 18-F Flutametamol PET-CT scanning to look for any amyloid burden. Pseudocontinuous Arterial Spin Labeling (PCASL), MR Angiography Head, 3D FLAIR and 3D T1-weighted sequences were used to quantify cerebral blood flow, cerebrovascular changes, white matter hyperintensities and brain atrophy respectively. Vascular risk factors were retrieved from the medical records. The 37 subjects from memory clinic were classified into subjective cognitive decline (SCD), vascular dementia (VD) and Alzheimer’s disease (AD) subgroups by a multi-disciplinary panel consisting of a neuroradiologist, and 2 geriatricians. Absolute cortical CBF in our cohort of T2DM, SCD, VD and AD was significantly decreased (p < 0.01) as compared to healthy controls (HC) in both whole brain and eight paired brain regions, after age, normalized grey matter volume and gender adjustment and Bonferroni correction. Subgroup analysis between T2DM, SCD, VD, and AD revealed that CBF of T2DM was not significantly different from AD, VD or SCD. By controlling for co-morbidities, impaired cortical CBF in T2DM was not related to microangiopathy or amyloid deposition, but to the interaction of triple risk factors (such as diabetes mellitus, hypertension, and hyperlipidemia). There was statistically significant negative correlation (p ≤ 0.05) between adjusted CBF and HbA1c in all brain regions of T2DM and HC (with partial correlation ranging from −0.30 to −0.46). Taken together, altered cerebral blood flow in T2DM might be related to disruption of cerebrovascular autoregulation related to vascular risk factors, and such oligemia occurred before clinical manifestation due to altered glycemic control.
Collapse
Affiliation(s)
- Anson C M Chau
- The University of Hong Kong (Shenzhen) Teaching Hospital Limited, The University of Hong Kong, Pokfulam Road, Hong Kong.
| | - Eva Y W Cheung
- Department of Diagnostic Radiology, LKS Faculty of Medicine, The University of Hong Kong, K406, Queen Mary Hospital, Pokfulam Road, Hong Kong
| | - K H Chan
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, 405B, Professorial Block, Queen Mary Hospital, Pokfulam Road, Hong Kong.
| | - W S Chow
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, 405B, Professorial Block, Queen Mary Hospital, Pokfulam Road, Hong Kong.
| | - Y F Shea
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, 405B, Professorial Block, Queen Mary Hospital, Pokfulam Road, Hong Kong
| | - Patrick K C Chiu
- Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, 405B, Professorial Block, Queen Mary Hospital, Pokfulam Road, Hong Kong
| | - Henry K F Mak
- Department of Diagnostic Radiology, LKS Faculty of Medicine, The University of Hong Kong, K406, Queen Mary Hospital, Pokfulam Road, Hong Kong; State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong; Alzheimer's Disease Research Network, The University of Hong Kong, Hong Kong.
| |
Collapse
|
40
|
Wang X, Ding D, Zhao Q, Liang X, Peng L, Zhao X, Xi Q, Min Z, Wang W, Xu X, Guo Q, Wang PJ. Brain hemodynamic changes in amnestic mild cognitive impairment measured by pulsed arterial spin labeling. Aging (Albany NY) 2020; 12:4348-4356. [PMID: 32167487 PMCID: PMC7093201 DOI: 10.18632/aging.102888] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 02/22/2020] [Indexed: 01/10/2023]
Abstract
We used pulsed arterial spin labeling (PASL) to investigate differences in cerebral blood flow (CBF) between 26 patients with amnestic mild cognitive impairment (aMCI) and 27 controls with normal cognition (NC). Hypoperfusion was observed in the right temporal pole of the middle temporal gyrus and the right inferior temporal gyrus in the aMCI compared with NC group. Interestingly, hyperperfusion was observed in the left temporal pole of the middle temporal gyrus, left superior temporal gyrus, bilateral precuneus, postcentral gyrus, right inferior parietal lobule, and right angular gyrus in the aMCI group, which likely resulted from a compensatory mechanism to maintain advanced neural activities. We found that mean CBF in the right inferior temporal gyrus, precuneus, and postcentral gyrus was positively correlated with cognitive ability in the aMCI but not NC group. Collectively, our data indicate that PASL is a useful noninvasive technique for monitoring changes in CBF and predicting cognitive decline in aMCI.
Collapse
Affiliation(s)
- Xiangbin Wang
- Department of Radiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, PR China
| | - Ding Ding
- Institute of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Qianhua Zhao
- Institute of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Xiaoniu Liang
- Institute of Neurology, Huashan Hospital, Fudan University, Shanghai 200040, PR China
| | - Ling Peng
- Department of Radiology, Shanghai Liqun Hospital, Shanghai 200333, PR China
| | - Xiaohu Zhao
- Department of Radiology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai 200240, PR China
| | - Qian Xi
- Department of Radiology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, PR China
| | - Zhang Min
- Department of Radiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, PR China
| | - Wei Wang
- Department of Radiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, PR China
| | - Xiaowen Xu
- Department of Radiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, PR China
| | - Qihao Guo
- Department of Geriatrics, The Sixth People's Hospital of Shanghai, Shanghai Jiao Tong University, Shanghai 200233, PR China
| | - Pei-Jun Wang
- Department of Radiology, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, PR China
| |
Collapse
|
41
|
Ceccarini J, Bourgeois S, Van Weehaeghe D, Goffin K, Vandenberghe R, Vandenbulcke M, Sunaert S, Van Laere K. Direct prospective comparison of 18F-FDG PET and arterial spin labelling MR using simultaneous PET/MR in patients referred for diagnosis of dementia. Eur J Nucl Med Mol Imaging 2020; 47:2142-2154. [PMID: 31960098 DOI: 10.1007/s00259-020-04694-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 01/12/2020] [Indexed: 01/09/2023]
Abstract
PURPOSE 18F-FDG PET is routinely used as an imaging marker in the early and differential diagnosis of dementing disorders and has incremental value over the clinical neurological and neuropsychological evaluation. Perfusion MR imaging by means of arterial spin labelling (ASL) is an alternative modality to indirectly measure neuronal functioning and could be used as complement measurement in a single MR session in the workup of dementia. Using simultaneous PET-MR, we performed a direct head-to-head comparison between enhanced multiplane tagging ASL (eASL) and 18F-FDG PET in a true clinical context of subjects referred for suspicion of neurodegenerative dementia. METHODS Twenty-seven patients underwent a 20-min 18F-FDG PET/MR and simultaneously acquired eASL on a GE Signa PET/MR. Data were compared with 30 screened age- and gender-matched healthy controls. Both integral eASL and 18F-FDG datasets were analysed visually by two readers unaware of the final clinical diagnosis, either in normal/abnormal classes, or full differential diagnosis (normal, Alzheimer type dementia [AD], dementia with Lewy Bodies [LBD], frontotemporal dementia [FTD] or other). Reader confidence was assessed with a rating scale (range 1-4). Data were also analysed semiquantitatively by VOI and voxel-based analyses. RESULTS The ground truth diagnosis for the patient group resulted in 14 patients with a neurodegenerative cognitive disorder (AD, FTD, LBD) and 13 patients with no arguments for an underlying neurodegenerative cause. Visual analysis resulted in equal specificity (0.70) for differentiating normal and abnormal cases between the two modalities, but in a higher sensitivity (0.93), confidence rating (0.64) and interobserver agreement for 18F-FDG PET compared with eASL. The same was true for assigning a specific differential diagnosis (sensitivity: and 0.39 for 18F-FDG PET and eASL, respectively). Semiquantitative analyses revealed prototypical patterns for AD and FTD, with both higher volumes of abnormality and intensity differences on 18F-FDG PET. CONCLUSION In a direct head-to-head comparison on a simultaneous GE Signa PET/MR, 18F-FDG PET performed better compared with ASL in terms of sensitivity and reader confidence, as well as volume and intensity of abnormalities. However, using pure semiquantitative analysis, similar diagnostic accuracy between the two modalities was obtained. Therefore, ASL may still serve as complement to neuroreceptor or protein deposition PET studies when a single simultaneous investigation is warranted.
Collapse
Affiliation(s)
- Jenny Ceccarini
- Department of Imaging and Pathology, University Hospitals Leuven and KU Leuven, Herestraat 49, 3000, Leuven, Belgium.
| | - Sophie Bourgeois
- Division of Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Donatienne Van Weehaeghe
- Department of Imaging and Pathology, University Hospitals Leuven and KU Leuven, Herestraat 49, 3000, Leuven, Belgium.,Division of Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Karolien Goffin
- Department of Imaging and Pathology, University Hospitals Leuven and KU Leuven, Herestraat 49, 3000, Leuven, Belgium.,Division of Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Rik Vandenberghe
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | | | - Stefan Sunaert
- Department of Imaging and Pathology, University Hospitals Leuven and KU Leuven, Herestraat 49, 3000, Leuven, Belgium.,Department of Radiology, University Hospitals Leuven, Leuven, Belgium
| | - Koen Van Laere
- Department of Imaging and Pathology, University Hospitals Leuven and KU Leuven, Herestraat 49, 3000, Leuven, Belgium.,Division of Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
42
|
Dolui S, Li Z, Nasrallah IM, Detre JA, Wolk DA. Arterial spin labeling versus 18F-FDG-PET to identify mild cognitive impairment. NEUROIMAGE-CLINICAL 2019; 25:102146. [PMID: 31931403 PMCID: PMC6957781 DOI: 10.1016/j.nicl.2019.102146] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 12/19/2019] [Accepted: 12/21/2019] [Indexed: 01/08/2023]
Abstract
18F-FDG-PET is a neurodegeneration biomarker, but is costly and requires a radioactive tracer. Arterial spin labeled perfusion MRI (ASL-MRI) provides similar information noninvasively. We compared ASL-MRI and FDG-PET in mild cognitive impairment (MCI) and older controls. At a group level, patterns and strength of ASL hypoperfusion and FDG-PET hypometabolism in MCI were comparable. ASL can provide a reasonable alternative for FDG-PET in clinical research.
Neurodegenerative biomarkers support diagnosis and measurement of disease progression in the Alzheimer's disease (AD) continuum. 18F-Fluorodeoxyglucose Positron Emission Tomography (18F-FDG-PET), which measures glucose metabolism, is one of the most commonly used biomarkers of neurodegeneration, but is expensive and requires exposure to ionizing radiation. Arterial Spin Labeled (ASL) perfusion Magnetic Resonance Imaging (MRI) provides non invasive quantification of cerebral blood flow (CBF), which is believed to be tightly coupled to glucose metabolism. Here we aimed to compare the performances of ASL derived CBF and 18F-FDG-PET derived standardized uptake value ratio (SUVR) in discriminating patients with mild cognitive impairment (MCI) from older Controls. 2D pseudo continuous ASL and 18F-FDG-PET data with adequate scan quality from 50 MCI study participants (age=73.0 ± 7.0 years, 16 female) and 35 older controls (age=70.2 ± 6.9 years, 20 female), acquired in close temporal proximity, usually on the same day, were considered for this study. We assessed Control-patient group differences both at voxel level and within a priori regions of interest (ROIs). We also compared their area under receiver operating characteristic curves (AUC) with mean CBF or SUVR in a priori selected posterior cingulate cortex (PCC). CBF and 18F-FDG-PET showed abnormalities in similar areas, particularly in medial temporoparietal regions, consistent with the typically observed pattern of prodromal AD. The hypoperfusion pattern with relative CBF (obtained by normalizing voxel CBF values with mean CBF in putamen) was more localized than with absolute CBF. Pearson's correlation coefficients between the T-scores corresponding to the group-differences obtained with 18F-FDG-PET SUVR and absolute and relative ASL CBF were 0.46 and 0.43 (p<0.001), respectively. ROI analyses were also consistent, with the strongest differences observed in PCC (p<0.01). 18F-FDG-PET SUVR, absolute and relative CBF in the PCC ROI demonstrated moderate and similar discriminatory power in predicting MCI status with AUC of 0.71 ± 0.12, 0.77 ± 0.12 and 0.74 ± 0.13, respectively. In conclusion, ASL CBF may be a reasonable, less expensive and safer substitute for 18F-FDG-PET in clinical research.
Collapse
Affiliation(s)
- Sudipto Dolui
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States
| | - Zhengjun Li
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States
| | - Ilya M Nasrallah
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States
| | - John A Detre
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, United States; Department of Neurology, University of Pennsylvania, Philadelphia, PA, United States
| | - David A Wolk
- Department of Neurology, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
43
|
Bergau N, Maul S, Rujescu D, Simm A, Navarrete Santos A. Reduction of Glycolysis Intermediate Concentrations in the Cerebrospinal Fluid of Alzheimer's Disease Patients. Front Neurosci 2019; 13:871. [PMID: 31496932 PMCID: PMC6713159 DOI: 10.3389/fnins.2019.00871] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 08/02/2019] [Indexed: 12/22/2022] Open
Abstract
The profile of 122 metabolites in the cerebrospinal fluid (CSF) of patients suffering from Alzheimer's disease (AD) and controls was studied. Among the 122 metabolites analyzed, 61 could be detected. Statistically significant differences between the AD and control group were only detected for metabolites of the glycolysis. Thus, accurate quantification of 11 glycolytic metabolites was done. We detected a significant reduction of five of them, namely phosphoenolpyruvate, 2-phosphoglycerate, 3-phosphoglycerate, pyruvate and dihydroxyacetone phosphate in the AD CSF compared to controls. These results correlate with the known reduction of glucose metabolism in the brain of patients with AD and indicate that metabolic analysis of the central carbon metabolism can be a potential tool in AD diagnostic. Although the Receiver operating characteristic (ROC) analyses of the metabolites do not reach the level of the diagnostic informativity of AD biomarkers, the combination of specific glycolysis metabolites with the established biomarkers may lead to an improvement in sensitivity and specificity.
Collapse
Affiliation(s)
- Nick Bergau
- Department of Cell and Metabolic Biology, Leibniz Institute of Plant Biochemistry, Halle (Saale), Germany
| | - Stephan Maul
- Department of Psychiatry, Psychotherapy and Psychosomatics, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Dan Rujescu
- Department of Psychiatry, Psychotherapy and Psychosomatics, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Andreas Simm
- Center for Basic Medical Research, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany.,Clinic for Cardiac Surgery, University Hospital, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Alexander Navarrete Santos
- Center for Basic Medical Research, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany.,Clinic for Cardiac Surgery, University Hospital, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
44
|
Zheng W, Cui B, Han Y, Song H, Li K, He Y, Wang Z. Disrupted Regional Cerebral Blood Flow, Functional Activity and Connectivity in Alzheimer's Disease: A Combined ASL Perfusion and Resting State fMRI Study. Front Neurosci 2019; 13:738. [PMID: 31396033 PMCID: PMC6668217 DOI: 10.3389/fnins.2019.00738] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Accepted: 07/02/2019] [Indexed: 11/13/2022] Open
Abstract
Recent studies have demonstrated a close relationship between regional cerebral blood flow (rCBF) and resting state functional connectivity changes in normal healthy people. However, little is known about the parameter changes in the most vulnerable regions in Alzheimer's disease (AD). Forty AD patients and 30 healthy controls participated in this study. The data of resting-state perfusion and functional magnetic resonance imaging (fMRI) was collected. By using voxel-wise arterial spin labeling (ASL) perfusion, we identified several regions of altered rCBF in AD patients. Then, by using resting state fMRI analysis, including amplitude low frequency fluctuation (ALFF) and seed-based functional connectivity, we investigated the changes of functional activity and connectivity among the identified rCBF regions. We extracted cognition-related parameters and searched for a sensitive biomarker to differentiate the AD patients from the normal controls (NC). Compared with controls, AD patients showed special disruptions in rCBF, which were mainly located in the left posterior cingulate cortex (PCC), the left and right dorsolateral prefrontal cortex (DLPFC), the left inferior parietal lobule (IPL), the right middle temporal gyrus (MTG), the left middle occipital gyrus (MOG), and the left precuneus (PCu). ALFF was performed based on the seven regions identified by the ASL method, and AD patients presented significantly decreased ALFF in the left PCC, left IPL, right MTG, left MOG, and left PCu and increased ALFF in the bilateral DLPFC. We constituted the network based on the seven regions and found that there was decreased connectivity among the identified regions in the AD patients, which predicted a disruption in the default mode network (DMN), executive control network (ECN) and visual network (VN). Furthermore, these abnormal parameters are closely associated with cognitive performances in AD patients. We combined the rCBF and ALFF value of PCC/PCu as a biomarker to differentiate the two groups and reached a sensitivity of 85.3% and a specificity of 88.5%. Our findings suggested that there was disrupted rCBF, functional activity and connectivity in specific cognition-related regions in Alzheimer's disease, which can be used as a valuable imaging biomarker for the diagnosis of AD.
Collapse
Affiliation(s)
- Weimin Zheng
- Department of Radiology, Aerospace Center Hospital, Beijing, China
| | - Bin Cui
- Department of Radiology, Aerospace Center Hospital, Beijing, China
| | - Ying Han
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Haiqing Song
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Kuncheng Li
- Department of Radiology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Magnetic Resonance Imaging and Brain Informatics, Beijing, China
| | - Yong He
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
- Beijing Key Laboratory of Brain Imaging and Connectomics, Beijing Normal University, Beijing, China
- IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing, China
| | - Zhiqun Wang
- Department of Radiology, Aerospace Center Hospital, Beijing, China
| |
Collapse
|
45
|
Croteau E, Castellano CA, Richard MA, Fortier M, Nugent S, Lepage M, Duchesne S, Whittingstall K, Turcotte ÉE, Bocti C, Fülöp T, Cunnane SC. Ketogenic Medium Chain Triglycerides Increase Brain Energy Metabolism in Alzheimer's Disease. J Alzheimers Dis 2019; 64:551-561. [PMID: 29914035 DOI: 10.3233/jad-180202] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND In Alzheimer's disease (AD), it is unknown whether the brain can utilize additional ketones as fuel when they are derived from a medium chain triglyceride (MCT) supplement. OBJECTIVE To assess whether brain ketone uptake in AD increases in response to MCT as it would in young healthy adults. METHODS Mild-moderate AD patients sequentially consumed 30 g/d of two different MCT supplements, both for one month: a mixture of caprylic (55%) and capric acids (35%) (n = 11), followed by a wash-out and then tricaprylin (95%; n = 6). Brain ketone (11C-acetoacetate) and glucose (FDG) uptake were quantified by PET before and after each MCT intervention. RESULTS Brain ketone consumption doubled on both types of MCT supplement. The slope of the relationship between plasma ketones and brain ketone uptake was the same as in healthy young adults. Both types of MCT increased total brain energy metabolism by increasing ketone supply without affecting brain glucose utilization. CONCLUSION Ketones from MCT compensate for the brain glucose deficit in AD in direct proportion to the level of plasma ketones achieved.
Collapse
Affiliation(s)
- Etienne Croteau
- Research Center on Aging, Université de Sherbrooke, Sherbrooke, QC, Canada
| | | | - Marie Anne Richard
- Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Mélanie Fortier
- Research Center on Aging, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Scott Nugent
- Centre de recherche CERVO de l'Institut universitaire en santé mentale de Québec, Québec, QC, Canada
| | - Martin Lepage
- Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Simon Duchesne
- Centre de recherche CERVO de l'Institut universitaire en santé mentale de Québec, Québec, QC, Canada.,Department of Radiology, Université Laval, Québec, QC, Canada
| | | | - Éric E Turcotte
- Department of Nuclear Medicine and Radiobiology, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Christian Bocti
- Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Tamàs Fülöp
- Research Center on Aging, Université de Sherbrooke, Sherbrooke, QC, Canada.,Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - Stephen C Cunnane
- Research Center on Aging, Université de Sherbrooke, Sherbrooke, QC, Canada.,Department of Medicine, Université de Sherbrooke, Sherbrooke, QC, Canada
| |
Collapse
|
46
|
Altomare D, Ferrari C, Caroli A, Galluzzi S, Prestia A, van der Flier WM, Ossenkoppele R, Van Berckel B, Barkhof F, Teunissen CE, Wall A, Carter SF, Schöll M, Choo ILH, Grimmer T, Redolfi A, Nordberg A, Scheltens P, Drzezga A, Frisoni GB. Prognostic value of Alzheimer's biomarkers in mild cognitive impairment: the effect of age at onset. J Neurol 2019; 266:2535-2545. [PMID: 31267207 DOI: 10.1007/s00415-019-09441-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 05/16/2019] [Accepted: 06/21/2019] [Indexed: 01/06/2023]
Abstract
OBJECTIVE The aim of this study is to assess the impact of age at onset on the prognostic value of Alzheimer's biomarkers in a large sample of patients with mild cognitive impairment (MCI). METHODS We measured Aβ42, t-tau, hippocampal volume on magnetic resonance imaging (MRI) and cortical metabolism on fluorodeoxyglucose-positron emission tomography (FDG-PET) in 188 MCI patients followed for at least 1 year. We categorised patients into earlier and later onset (EO/LO). Receiver operating characteristic curves and corresponding areas under the curve (AUCs) were performed to assess and compar the biomarker prognostic performances in EO and LO groups. Linear Model was adopted for estimating the time-to-progression in relation with earlier/later onset MCI groups and biomarkers. RESULTS In earlier onset patients, all the assessed biomarkers were able to predict cognitive decline (p < 0.05), with FDG-PET showing the best performance. In later onset patients, all biomarkers but t-tau predicted cognitive decline (p < 0.05). Moreover, FDG-PET alone in earlier onset patients showed a higher prognostic value than the one resulting from the combination of all the biomarkers in later onset patients (earlier onset AUC 0.935 vs later onset AUC 0.753, p < 0.001). Finally, FDG-PET showed a different prognostic value between earlier and later onset patients (p = 0.040) in time-to-progression allowing an estimate of the time free from disease. DISCUSSION FDG-PET may represent the most universal tool for the establishment of a prognosis in MCI patients and may be used for obtaining an onset-related estimate of the time free from disease.
Collapse
Affiliation(s)
- Daniele Altomare
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland.,Memory Clinic, University Hospital of Geneva, Geneva, Switzerland
| | - Clarissa Ferrari
- Service of Statistics, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, via Pilastroni 4, 25125, Brescia, Italy.
| | - Anna Caroli
- Medical Imaging Unit, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Samantha Galluzzi
- Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE), IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Annapaola Prestia
- Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE), IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Wiesje M van der Flier
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.,Department of Epidemiology and Biostatistics, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Rik Ossenkoppele
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.,Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.,Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden
| | - Bart Van Berckel
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.,Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.,Institute of Neurology, UCL, London, UK.,Institute of Healthcare Engineering, UCL, London, UK
| | - Charlotte E Teunissen
- Neurochemistry Laboratory, Department of Clinical Chemistry, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Anders Wall
- Section of Nuclear Medicine and PET, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Stephen F Carter
- Alzheimer Neurobiology Center, Karolinska Institutet, Stockholm, Sweden.,Wolfson Molecular Imaging Centre, University of Manchester, Manchester, UK
| | - Michael Schöll
- Clinical Memory Research Unit, Department of Clinical Sciences, Lund University, Malmö, Sweden.,Wallenberg Centre for Molecular and Translational Medicine, Department of Psychiatry and Neurochemistry, University of Gothenburg, Gothenburg, Sweden.,Dementia Research Centre, Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, UK
| | - I L Han Choo
- Alzheimer Neurobiology Center, Karolinska Institutet, Stockholm, Sweden.,Department of Neuropsychiatry, School of Medicine, Chosun University, Gwangju, Republic of Korea
| | - Timo Grimmer
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, Technische Universität München, Munich, Germany
| | - Alberto Redolfi
- Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE), IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Agneta Nordberg
- Center for Alzheimer Research, Division of Clinical Geriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Aging Theme, Karolinska University Hospital, Stockholm, Sweden
| | - Philip Scheltens
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Alexander Drzezga
- Department of Nuclear Medicine, University of Cologne, Cologne, Germany
| | - Giovanni B Frisoni
- Laboratory of Neuroimaging of Aging (LANVIE), University of Geneva, Geneva, Switzerland.,Laboratory of Alzheimer's Neuroimaging and Epidemiology (LANE), IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy.,Memory Clinic, University Hospital of Geneva, Geneva, Switzerland
| | | |
Collapse
|
47
|
Verclytte S, Lopes R, Viard R, Rollin A, Vanhoutte M, Pasquier F, Pruvo JP, Leclerc X. Differences in cortical perfusion detected by arterial spin labeling in nonamnestic and amnestic subtypes of early-onset Alzheimer's disease. J Neuroradiol 2019; 47:284-291. [PMID: 30981825 DOI: 10.1016/j.neurad.2019.03.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 03/26/2019] [Accepted: 03/30/2019] [Indexed: 11/24/2022]
Abstract
OBJECTIVES Early-onset Alzheimer's disease (EOAD) begins before the age of 65 and is characterized by a faster clinical course and the frequency of nonamnestic symptoms compared to late onset Alzheimer disease (LOAD). However, the pathophysiological process of EOAD remains unclear. We expected that ASL may show widespread cortical hypoperfusion in EOAD compared to LOAD and in nonamnestic EOAD compared to amnestic EOAD. METHODS In this study, 26 EOAD patients (16 amnestic and 10 nonamnestic patients), 29 LOAD patients and 12 healthy controls underwent pseudo-continuous ASL and 3D FFE T1 sequences. Statistical comparisons between EOAD, LOAD and control groups were made after surface-based analysis of CBF maps in regressing out the cortical thickness. RESULTS ASL showed a more severe hypoperfusion in nonamnestic EOAD patients compared to amnestic EOAD ones, with mean CBF values (± std) of 26.9 (± 3.8) and 46.6 (± 24.1) mL/100 g/min respectively (P = 0.014), located in the bilateral temporo-parietal neocortex, the precuneus, the posterior cingulate cortices (PCC) and frontal lobes. Comparison between EOAD and LOAD patients showed a trend to hypoperfusion in the left parietal lobe, PCC and precuneus in EOAD (P < 0.001 uncorrected). CONCLUSIONS Different patterns of hypoperfusion between nonamnestic and amnestic EOAD subtypes were identified, with a more severe and extensive hypoperfusion in nonamnestic patients. A trend towards more severe hypoperfusion was detected in EOAD compared to LOAD. Further studies are needed to validate ASL as a potential tool for the distinction of EOAD subtypes and the prediction of the time course of the disease.
Collapse
Affiliation(s)
- Sebastien Verclytte
- Imaging department, Lille catholic hospitals, Lille catholic university, 59000 Lille, France.
| | - Renaud Lopes
- U1171, In-vivo imaging platform, predictive medicine and therapeutic research institute, université Lille Nord de France, 59000 Lille, France
| | - Romain Viard
- U1171, In-vivo imaging platform, predictive medicine and therapeutic research institute, université Lille Nord de France, 59000 Lille, France
| | - Adeline Rollin
- Memory resources and research center, CHU de Lille, 59000 Lille, France
| | - Matthieu Vanhoutte
- U1171, In-vivo imaging platform, predictive medicine and therapeutic research institute, université Lille Nord de France, 59000 Lille, France
| | - Florence Pasquier
- Memory resources and research center, CHU de Lille, 59000 Lille, France
| | - Jean-Pierre Pruvo
- U1171, In-vivo imaging platform, predictive medicine and therapeutic research institute, université Lille Nord de France, 59000 Lille, France; Department of neuroradiology, CHU de Lille, 59000 Lille, France
| | - Xavier Leclerc
- U1171, In-vivo imaging platform, predictive medicine and therapeutic research institute, université Lille Nord de France, 59000 Lille, France; Department of neuroradiology, CHU de Lille, 59000 Lille, France
| |
Collapse
|
48
|
Guo Y, Li X, Zhang M, Chen N, Wu S, Lei J, Wang Z, Wang R, Wang J, Liu H. Age‑ and brain region‑associated alterations of cerebral blood flow in early Alzheimer's disease assessed in AβPPSWE/PS1ΔE9 transgenic mice using arterial spin labeling. Mol Med Rep 2019; 19:3045-3052. [PMID: 30816468 PMCID: PMC6423566 DOI: 10.3892/mmr.2019.9950] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 02/06/2019] [Indexed: 02/07/2023] Open
Abstract
It has been suggested that cerebral blood flow (CBF) alterations may be involved in the pathogenesis of Alzheimer's disease (AD). However, how CBF changes with age has not been detailed in AD, particularly in its early stages. The objective of the present study was to evaluate CBF in four brain regions (the hippocampus, entorhinal cortex, frontoparietal cortex and thalamus) of mice in four age groups, to mimic the respective stages of AD in humans [2 months (pre-clinical), 3.5 months (sub-clinical), 5 months (early-clinical) and 8 months (mid-clinical)], to understand the age-associated changes in selected brain regions and to elucidate the underlying vascular mechanisms. CBF was measured using magnetic resonance imaging-arterial spin labelling (ASL) under identical conditions across the age groups of AβPPSWE/PS1ΔE9 (APP/PS1) transgenic mice with AD. The results indicated age- and brain region-associated changes in CBF were associated with early AD. More precisely, an age-dependent increase in CBF (in the pre- and sub-clinical AD groups) was observed in the frontoparietal cortex and thalamus. Conversely, increased CBF demonstrated an age-dependent decline (in the early- and mid-clinical AD groups) in all examined brain regions. Among the regions, the thalamus had the greatest increase in CBF in the 2 and 3.5 months age groups, which was substantially different compared with the age-matched controls. An extension of vessel area was also noted to be age- and brain region-dependent. In particular, correlation analysis revealed significant associations of CBF with vessel area in the frontoparietal cortex and thalamus of APP/PS1 mice at ages 2 and 3.5 months, indicating that CBF increase may arise from vessel extension. The results of the present study suggested that ASL can detect age- and brain region-associated changes in CBF in mice with AD, and that ASL-measured CBF increase may be a potential diagnostic biomarker for early AD. The observation that CBF increase resulted from vessel extension may aid in the understanding of the vascular role in age-associated development of AD pathology, and provide preclinical evidence for AD patient management.
Collapse
Affiliation(s)
- Yapei Guo
- Department of Neurology, The Fifth Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Xueyuan Li
- Department of Neurosurgery, The First Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Min Zhang
- Department of Neurology, The Fifth Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Ningning Chen
- Department of Neurology, The Fifth Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Shitao Wu
- Department of Neurology, The Fifth Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Jianfeng Lei
- Center for Medical Experiments and Testing, Capital Medical University, Beijing 100069, P.R. China
| | - Zhanjing Wang
- Center for Medical Experiments and Testing, Capital Medical University, Beijing 100069, P.R. China
| | - Renzhi Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Beijing 100052, P.R. China
| | - Jianping Wang
- Department of Neurology, The Fifth Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Hengfang Liu
- Department of Neurology, The Fifth Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| |
Collapse
|
49
|
Possible Clues for Brain Energy Translation via Endolysosomal Trafficking of APP-CTFs in Alzheimer's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:2764831. [PMID: 30420907 PMCID: PMC6215552 DOI: 10.1155/2018/2764831] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 07/14/2018] [Accepted: 08/19/2018] [Indexed: 02/07/2023]
Abstract
Vascular dysfunctions, hypometabolism, and insulin resistance are high and early risk factors for Alzheimer's disease (AD), a leading neurological disease associated with memory decline and cognitive dysfunctions. Early defects in glucose transporters and glycolysis occur during the course of AD progression. Hypometabolism begins well before the onset of early AD symptoms; this timing implicates the vulnerability of hypometabolic brain regions to beta-secretase 1 (BACE-1) upregulation, oxidative stress, inflammation, synaptic failure, and cell death. Despite the fact that ketone bodies, astrocyte-neuron lactate shuttle, pentose phosphate pathway (PPP), and glycogenolysis compensate to provide energy to the starving AD brain, a considerable energy crisis still persists and increases during disease progression. Studies that track brain energy metabolism in humans, animal models of AD, and in vitro studies reveal striking upregulation of beta-amyloid precursor protein (β-APP) and carboxy-terminal fragments (CTFs). Currently, the precise role of CTFs is unclear, but evidence supports increased endosomal-lysosomal trafficking of β-APP and CTFs through autophagy through a vague mechanism. While intracellular accumulation of Aβ is attributed as both the cause and consequence of a defective endolysosomal-autophagic system, much remains to be explored about the other β-APP cleavage products. Many recent works report altered amino acid catabolism and expression of several urea cycle enzymes in AD brains, but the precise cause for this dysregulation is not fully explained. In this paper, we try to connect the role of CTFs in the energy translation process in AD brain based on recent findings.
Collapse
|
50
|
Li KC, Luo X, Zeng QZ, Xu XJ, Huang PY, Shen ZJ, Xu JJ, Zhou J, Zhang MM. Distinct Patterns of Interhemispheric Connectivity in Patients With Early- and Late-Onset Alzheimer's Disease. Front Aging Neurosci 2018; 10:261. [PMID: 30237764 PMCID: PMC6136638 DOI: 10.3389/fnagi.2018.00261] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 08/14/2018] [Indexed: 12/18/2022] Open
Abstract
Background: Early-onset Alzheimer’s disease (EOAD) presents a different clinical profile than late-onset Alzheimer’s disease (LOAD). Neuroimaging studies have demonstrated that patients with EOAD present more atrophy and functional disconnection than LOAD patients. However, it remains unknown whether the interhemispheric functional disconnection or its underlying structural impairment contributes to the different clinical profiles of EOAD and LOAD. Methods: According to the arbitrary cut-off age of 65, we included 22 EOAD patients, 27 LOAD patients and 38 healthy controls (further divided into 21 relatively young and 17 old controls). Participants underwent resting-state functional MRI, diffusion tensor imaging (DTI) and comprehensive neuropsychological assessments. We used voxel-mirrored homotopic connectivity (VMHC) to examine interhemispheric functional connectivity. Then, we calculated the diffusion index based on tract-based spatial statistics (TBSS). Two-sample t-tests were used to assess the interhemispheric connectivity differences between each patient group and its corresponding control group. Results: We found that the EOAD patients had lower VMHC in the hippocampus, parahippocampal gyrus (PHG), superior temporal gyrus (STG) and inferior parietal cortex (IPC) than did controls. Consistently, the EOAD patients exhibited white matter (WM) tract impairment in the posterior regions. On the other hand, the LOAD patients displayed increased VMHC and impaired WM tracts in the frontal region. Correlation analyses showed that VMHC in the IPC was related to executive function in the EOAD patients (r = −0.67, P < 0.05). Conclusion: In contrast to the LOAD patients, patients with EOAD exhibited more widely disrupted interhemispheric functional and structural connectivity, which overlapped well across brain regions. In addition, for the EOAD patients, decreased interhemispheric connectivity related to executive deficits. Our study suggested that different interhemispheric connectivity damage patterns may contribute to the distinct clinical profiles in EOAD and LOAD.
Collapse
Affiliation(s)
- Kai-Cheng Li
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao Luo
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Qing-Ze Zeng
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiao-Jun Xu
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Pei-Yu Huang
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Zhu-Jing Shen
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jing-Jing Xu
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Jiong Zhou
- Department of Neurology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Min-Ming Zhang
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|