1
|
Chen MD, Deng CF, Chen PF, Li A, Wu HZ, Ouyang F, Hu XG, Liu JX, Wang SM, Tang D. Non-invasive metabolic biomarkers in initial cognitive impairment in patients with diabetes: A systematic review and meta-analysis. Diabetes Obes Metab 2024; 26:5519-5536. [PMID: 39233493 DOI: 10.1111/dom.15916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/05/2024] [Accepted: 08/15/2024] [Indexed: 09/06/2024]
Abstract
AIM Diabetic cognitive impairment (DCI), considered one of the most severe and commonly overlooked complications of diabetes, has shown inconsistent findings regarding the metabolic profiles in DCI patients. This systematic review and meta-analysis aimed to identify dysregulated metabolites as potential biomarkers for early DCI, providing valuable insights into the underlying pathophysiological mechanisms. MATERIALS AND METHODS A systematic search of four databases, namely PubMed, Embase, Web of Science and Cochrane, was conducted up to March 2024. Subsequently, a qualitative review of clinical studies was performed followed by a meta-analysis of metabolite markers. Finally, the sources of heterogeneity were explored through subgroup and sensitivity analyses. RESULTS A total of 774 unique publications involving 4357 participants and the identification of multiple metabolites were retrieved. Of these, 13 clinical studies reported metabolite differences between the DCI and control groups. Meta-analysis was conducted for six brain metabolites and two metabolite ratios. The results revealed a significant increase in myo-inositol (MI) concentration and decreases in glutamate (Glu), Glx (glutamate and glutamine) and N-acetylaspartate/creatine (NAA/Cr) ratios in DCI, which have been identified as the most sensitive metabolic biomarkers for evaluating DCI progression. Notably, brain metabolic changes associated with cognitive impairment are more pronounced in type 2 diabetes mellitus than in type 1 diabetes mellitus, and the hippocampus emerged as the most sensitive brain region regarding metabolic changes associated with DCI. CONCLUSIONS Our results suggest that MI, Glu, and Glx concentrations and NAA/Cr ratios within the hippocampus may serve as metabolic biomarkers for patients with early-stage DCI.
Collapse
Affiliation(s)
- Meng-Di Chen
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM and Engineering and Technology Research Center for Chinese Materia Medica Quality of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, China
| | - Chao-Fan Deng
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM and Engineering and Technology Research Center for Chinese Materia Medica Quality of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, China
| | - Peng-Fei Chen
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM and Engineering and Technology Research Center for Chinese Materia Medica Quality of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, China
| | - Ao Li
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM and Engineering and Technology Research Center for Chinese Materia Medica Quality of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, China
| | - Hua-Ze Wu
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM and Engineering and Technology Research Center for Chinese Materia Medica Quality of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, China
| | - Fan Ouyang
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM and Engineering and Technology Research Center for Chinese Materia Medica Quality of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xu-Guang Hu
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM and Engineering and Technology Research Center for Chinese Materia Medica Quality of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, China
| | - Jian-Xin Liu
- School of Pharmaceutical Sciences, China-Pakistan International Science and Technology Innovation Cooperation Base for Ethnic Medicine Development in Hunan Province, Hunan University of Medicine, Huaihua City, China
| | - Shu-Mei Wang
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM and Engineering and Technology Research Center for Chinese Materia Medica Quality of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, China
| | - Dan Tang
- Key Laboratory of Digital Quality Evaluation of Chinese Materia Medica of State Administration of TCM and Engineering and Technology Research Center for Chinese Materia Medica Quality of Guangdong Province, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
2
|
Kara F, Kantarci K. Understanding Proton Magnetic Resonance Spectroscopy Neurochemical Changes Using Alzheimer's Disease Biofluid, PET, Postmortem Pathology Biomarkers, and APOE Genotype. Int J Mol Sci 2024; 25:10064. [PMID: 39337551 PMCID: PMC11432594 DOI: 10.3390/ijms251810064] [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: 08/23/2024] [Revised: 09/15/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
In vivo proton (1H) magnetic resonance spectroscopy (MRS) is a powerful non-invasive method that can measure Alzheimer's disease (AD)-related neuropathological alterations at the molecular level. AD biomarkers include amyloid-beta (Aβ) plaques and hyperphosphorylated tau neurofibrillary tangles. These biomarkers can be detected via postmortem analysis but also in living individuals through positron emission tomography (PET) or biofluid biomarkers of Aβ and tau. This review offers an overview of biochemical abnormalities detected by 1H MRS within the biologically defined AD spectrum. It includes a summary of earlier studies that explored the association of 1H MRS metabolites with biofluid, PET, and postmortem AD biomarkers and examined how apolipoprotein e4 allele carrier status influences brain biochemistry. Studying these associations is crucial for understanding how AD pathology affects brain homeostasis throughout the AD continuum and may eventually facilitate the development of potential novel therapeutic approaches.
Collapse
Affiliation(s)
- Firat Kara
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA
| |
Collapse
|
3
|
Azriel O, Arad G, Tik N, Weiser M, Bloch M, Garber E, Lazarov A, Pine DS, Tavor I, Bar-Haim Y. Neural activation changes following attention bias modification treatment or a selective serotonin reuptake inhibitor for social anxiety disorder. Psychol Med 2024; 54:1-13. [PMID: 39252484 PMCID: PMC11496228 DOI: 10.1017/s0033291724001521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 02/01/2024] [Accepted: 04/11/2024] [Indexed: 09/11/2024]
Abstract
BACKGROUND Delineation of changes in neural function associated with novel and established treatments for social anxiety disorder (SAD) can advance treatment development. We examined such changes following selective serotonin reuptake inhibitor (SSRI) and attention bias modification (ABM) variant - gaze-contingent music reward therapy (GC-MRT), a first-line and an emerging treatments for SAD. METHODS Eighty-one patients with SAD were allocated to 12-week treatments of either SSRI or GC-MRT, or waitlist (ns = 22, 29, and 30, respectively). Baseline and post-treatment functional magnetic resonance imaging (fMRI) data were collected during a social-threat processing task, in which attention was directed toward and away from threat/neutral faces. RESULTS Patients who received GC-MRT or SSRI showed greater clinical improvement relative to patients in waitlist. Compared to waitlist patients, treated patients showed greater activation increase in the right inferior frontal gyrus and anterior cingulate cortex when instructed to attend toward social threats and away from neutral stimuli. An additional anterior cingulate cortex cluster differentiated between the two active groups. Activation in this region increased in ABM and decreased in SSRI. In the ABM group, symptom change was positively correlated with neural activation change in the dorsolateral prefrontal cortex. CONCLUSIONS Brain function measures show both shared and treatment-specific changes following ABM and SSRI treatments for SAD, highlighting the multiple pathways through which the two treatments might work. Treatment-specific neural responses suggest that patients with SAD who do not fully benefit from SSRI or ABM may potentially benefit from the alternative treatment, or from a combination of the two. TRIAL REGISTRATION ClinicalTrials.gov, Identifier: NCT03346239. https://clinicaltrials.gov/ct2/show/NCT03346239.
Collapse
Affiliation(s)
- Omer Azriel
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Gal Arad
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Niv Tik
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Mark Weiser
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Psychiatry, Sheba Medical Center, Tel Aviv, Israel
| | - Miki Bloch
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Psychiatric Department, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Eddie Garber
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Psychiatric Department, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Amit Lazarov
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Daniel S. Pine
- Section on Developmental Affective Neuroscience, National Institute of Mental Health, Bethesda, MD, USA
| | - Ido Tavor
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Yair Bar-Haim
- School of Psychological Sciences, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
4
|
Hu J, Zhang M, Zhang Y, Zhuang H, Zhao Y, Li Y, Jin W, Qian X, Wang L, Ye G, Tang H, Liu J, Li B, Nachev P, Liang Z, Li Y. Neurometabolic topography and associations with cognition in Alzheimer's disease: A whole-brain high-resolution 3D MRSI study. Alzheimers Dement 2024; 20:6407-6422. [PMID: 39073196 PMCID: PMC11497670 DOI: 10.1002/alz.14137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/29/2024] [Accepted: 06/22/2024] [Indexed: 07/30/2024]
Abstract
INTRODUCTION Altered neurometabolism, detectable via proton magnetic resonance spectroscopic imaging (1H-MRSI), is spatially heterogeneous and underpins cognitive impairments in Alzheimer's disease (AD). However, the spatial relationships between neurometabolic topography and cognitive impairment in AD remain unexplored due to technical limitations. METHODS We used a novel whole-brain high-resolution 1H-MRSI technique, with simultaneously acquired 18F-florbetapir positron emission tomography (PET) imaging, to investigate the relationship between neurometabolic topography and cognitive functions in 117 participants, including 22 prodromal AD, 51 AD dementia, and 44 controls. RESULTS Prodromal AD and AD dementia patients exhibited spatially distinct reductions in N-acetylaspartate, and increases in myo-inositol. Reduced N-acetylaspartate and increased myo-inositol were associated with worse global cognitive performance, and N-acetylaspartate correlated with five specific cognitive scores. Neurometabolic topography provides biological insights into diverse cognitive dysfunctions. DISCUSSION Whole-brain high-resolution 1H-MRSI revealed spatially distinct neurometabolic topographies associated with cognitive decline in AD, suggesting potential for noninvasive brain metabolic imaging to track AD progression. HIGHLIGHTS Whole-brain high-resolution 1H-MRSI unveils neurometabolic topography in AD. Spatially distinct reductions in NAA, and increases in mI, are demonstrated. NAA and mI topography correlates with global cognitive performance. NAA topography correlates with specific cognitive performance.
Collapse
Affiliation(s)
- Jialin Hu
- National Engineering Research Center of Advanced Magnetic Resonance Technologies for Diagnosis and Therapy, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Miao Zhang
- Department of Nuclear MedicineRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yaoyu Zhang
- National Engineering Research Center of Advanced Magnetic Resonance Technologies for Diagnosis and Therapy, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Huixiang Zhuang
- National Engineering Research Center of Advanced Magnetic Resonance Technologies for Diagnosis and Therapy, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
| | - Yibo Zhao
- Beckman Institute for Advanced Science and TechnologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
- Department of Electrical and Computer EngineeringUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Yudu Li
- Beckman Institute for Advanced Science and TechnologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
- National Center for Supercomputing ApplicationsUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Wen Jin
- Beckman Institute for Advanced Science and TechnologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
- Department of Electrical and Computer EngineeringUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Xiao‐Hang Qian
- Department of GeriatricsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Medical Center on Aging of Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Department of Neurology and Institute of NeurologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Lijun Wang
- Department of Neurovascular CenterChanghai HospitalNaval Medical UniversityShanghaiChina
| | - Guanyu Ye
- Department of Neurology and Institute of NeurologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Huidong Tang
- Department of GeriatricsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Medical Center on Aging of Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Jun Liu
- Department of Neurology and Institute of NeurologyRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Biao Li
- Department of Nuclear MedicineRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Parashkev Nachev
- High‐Dimensional Neurology GroupInstitute of NeurologyUniversity College LondonLondonUK
| | - Zhi‐Pei Liang
- Beckman Institute for Advanced Science and TechnologyUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
- Department of Electrical and Computer EngineeringUniversity of Illinois at Urbana‐ChampaignUrbanaIllinoisUSA
| | - Yao Li
- National Engineering Research Center of Advanced Magnetic Resonance Technologies for Diagnosis and Therapy, School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghaiChina
- Institute of Medical RoboticsShanghai Jiao Tong UniversityShanghaiChina
| |
Collapse
|
5
|
Doppler CEJ, Seger A, Farrher E, Régio Brambilla C, Hensel L, Filss CP, Hellmich M, Gogishvili A, Shah NJ, Lerche CW, Neumaier B, Langen KJ, Fink GR, Sommerauer M. Glutamate Signaling in Patients With Parkinson Disease With REM Sleep Behavior Disorder. Neurology 2024; 102:e209271. [PMID: 38630966 DOI: 10.1212/wnl.0000000000209271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Clinical heterogeneity of patients with Parkinson disease (PD) is well recognized. PD with REM sleep behavior disorder (RBD) is a more malignant phenotype with faster motor progression and higher nonmotor symptom burden. However, the neural mechanisms underlying this clinical divergence concerning imbalances in neurotransmitter systems remain elusive. METHODS Combining magnetic resonance (MR) spectroscopy and [11C]ABP688 PET on a PET/MR hybrid system, we simultaneously investigated two different mechanisms of glutamate signaling in patients with PD. Patients were grouped according to their RBD status in overnight video-polysomnography and compared with age-matched and sex-matched healthy control (HC) participants. Total volumes of distribution (VT) of [11C]ABP688 were estimated with metabolite-corrected plasma concentrations during steady-state conditions between 45 and 60 minutes of the scan following a bolus-infusion protocol. Glutamate, glutamine, and glutathione levels were investigated with single-voxel stimulated echo acquisition mode MR spectroscopy of the left basal ganglia. RESULTS We measured globally elevated VT of [11C]ABP688 in 16 patients with PD and RBD compared with 17 patients without RBD and 15 HC participants (F(2,45) = 5.579, p = 0.007). Conversely, glutamatergic metabolites did not differ between groups and did not correlate with the regional VT of [11C]ABP688. VT of [11C]ABP688 correlated with the amount of REM sleep without atonia (F(1,42) = 5.600, p = 0.023) and with dopaminergic treatment response in patients with PD (F(1,30) = 5.823, p = 0.022). DISCUSSION Our results suggest that patients with PD and RBD exhibit altered glutamatergic signaling indicated by higher VT of [11C]ABP688 despite unaffected glutamate levels. The imbalance of glutamate receptors and MR spectroscopy glutamate metabolite levels indicates a novel mechanism contributing to the heterogeneity of PD and warrants further investigation of drugs targeting mGluR5.
Collapse
Affiliation(s)
- Christopher E J Doppler
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Aline Seger
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Ezequiel Farrher
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Cláudia Régio Brambilla
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Lukas Hensel
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Christian P Filss
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Martin Hellmich
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Ana Gogishvili
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - N Jon Shah
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Christoph W Lerche
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Bernd Neumaier
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Karl-Josef Langen
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Gereon R Fink
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| | - Michael Sommerauer
- From the Cognitive Neuroscience (C.E.J.D., A.S., L.H., G.R.F., M.S.), Institute of Neuroscience and Medicine (INM-3), Forschungszentrum Jülich; Department of Neurology (C.E.J.D., A.S., L.H., G.R.F., M.S.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Köln; Institute of Neuroscience and Medicine (INM-4) (E.F., C.R.B., A.G., N.J.S., C.W.L., K.-J.L.), Forschungszentrum Jülich; Department of Nuclear Medicine (C.P.F., K.-J.L.), RWTH University Hospital, Aachen; Institute of Medical Statistics and Computational Biology (M.H.), Faculty of Medicine and University Hospital of Cologne, University of Cologne; Faculty of Medicine (A.G.), RWTH Aachen University, Germany; Engineering Physics Department (A.G.), Georgian Technical University, Tbilisi, Georgia; Institute of Neuroscience and Medicine (INM-11) (N.J.S.), Molecular Neuroscience and Neuroimaging, JARA, Forschungszentrum Jülich; JARA-BRAIN-Translational Medicine (N.J.S.), Aachen; Department of Neurology (N.J.S.), RWTH Aachen University; and Institute of Neuroscience and Medicine (INM-5) (B.N.), Forschungszentrum Jülich, Germany
| |
Collapse
|
6
|
Ambeskovic M, Hopkins G, Hoover T, Joseph JT, Montina T, Metz GAS. Metabolomic Signatures of Alzheimer's Disease Indicate Brain Region-Specific Neurodegenerative Progression. Int J Mol Sci 2023; 24:14769. [PMID: 37834217 PMCID: PMC10573054 DOI: 10.3390/ijms241914769] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/25/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023] Open
Abstract
Pathological mechanisms contributing to Alzheimer's disease (AD) are still elusive. Here, we identified the metabolic signatures of AD in human post-mortem brains. Using 1H NMR spectroscopy and an untargeted metabolomics approach, we identified (1) metabolomic profiles of AD and age-matched healthy subjects in post-mortem brain tissue, and (2) region-common and region-unique metabolome alterations and biochemical pathways across eight brain regions revealed that BA9 was the most affected. Phenylalanine and phosphorylcholine were mainly downregulated, suggesting altered neurotransmitter synthesis. N-acetylaspartate and GABA were upregulated in most regions, suggesting higher inhibitory activity in neural circuits. Other region-common metabolic pathways indicated impaired mitochondrial function and energy metabolism, while region-unique pathways indicated oxidative stress and altered immune responses. Importantly, AD caused metabolic changes in brain regions with less well-documented pathological alterations that suggest degenerative progression. The findings provide a new understanding of the biochemical mechanisms of AD and guide biomarker discovery for personalized risk prediction and diagnosis.
Collapse
Affiliation(s)
- Mirela Ambeskovic
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (M.A.); (G.H.); (T.H.)
| | - Giselle Hopkins
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (M.A.); (G.H.); (T.H.)
| | - Tanzi Hoover
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (M.A.); (G.H.); (T.H.)
| | - Jeffrey T. Joseph
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 1N4, Canada;
| | - Tony Montina
- Department of Chemistry and Biochemistry, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
- Southern Alberta Genome Sciences Centre, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| | - Gerlinde A. S. Metz
- Canadian Centre for Behavioural Neuroscience, Department of Neuroscience, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada; (M.A.); (G.H.); (T.H.)
- Southern Alberta Genome Sciences Centre, University of Lethbridge, Lethbridge, AB T1K 3M4, Canada
| |
Collapse
|
7
|
Kau YL, Lin IH, Juang CL, Chang CK, Ho WH, Wen HC. Metabolite Variations in the Hippocampus and Corpus Callosum of Patients with Mild Cognitive Impairment Using Magnetic Resonance Spectroscopy with Three-Dimensional Chemical Shift Images. Brain Sci 2023; 13:1244. [PMID: 37759845 PMCID: PMC10526271 DOI: 10.3390/brainsci13091244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 07/30/2023] [Accepted: 08/21/2023] [Indexed: 09/29/2023] Open
Abstract
This study compared the metabolites in the brain regions of hippocampus and corpus callosum between patients with mild cognitive impairment (MCI) and healthy controls using no-radiation and high-sensitivity magnetic resonance spectroscopy (MRS) with three-dimensional chemical shift images (3D-CSI). Twenty volunteers (seven patients with MCI and 13 healthy controls) aged 50-71 years were recruited for this prospective study. MRS with 3D-CSI images of a variety of metabolites was collected from the hippocampus and corpus callosum. Sex and weight showed no significant differences between the two groups. The metabolite levels in the hippocampus and corpus callosum of the MCI group were generally lower than in those of the healthy group, especially for creatine (p < 0.001 in the hippocampus and p = 0.020 in the corpus callosum) and N-acetyl aspartate/creatine (p < 0.001 in the hippocampus and p = 0.020 in the corpus callosum); however, choline/creatine showed a significant difference (p < 0.001) only in the hippocampus, and myo-inositol/creatine showed a significant difference (p < 0.001) only in the corpus callosum. Our study demonstrated that MRS with 3D-CSI can be used to measure these metabolite levels to determine the differences between patients with MCI and healthy individuals. This would aid early diagnosis of MCI in clinical practice, and patients could receive prompt intervention to improve their quality of life.
Collapse
Affiliation(s)
- Yen-Lon Kau
- Department of Medical Imaging, Camillian St. Mary’s Hospital, Luodong, Yilan 265502, Taiwan; (Y.-L.K.); (W.-H.H.)
- Department of Medical Imaging and Radiological Sciences, Yuanpei University, Hsinchu 30015, Taiwan;
| | - I-Hung Lin
- Nobel Eye Institute, Taipei 100008, Taiwan;
- Department of Ophthalmology, Taipei Medical University Hospital, Taipei 11031, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Chi-Long Juang
- Department of Medical Imaging and Radiological Sciences, Yuanpei University, Hsinchu 30015, Taiwan;
| | - Chao-Kai Chang
- Nobel Eye Institute, Taipei 100008, Taiwan;
- Department of Optometry, Yuanpei University, Hsinchu 30015, Taiwan;
| | - Wen-Hsiang Ho
- Department of Medical Imaging, Camillian St. Mary’s Hospital, Luodong, Yilan 265502, Taiwan; (Y.-L.K.); (W.-H.H.)
| | - Hsiao-Chuan Wen
- Department of Pet Healthcare, Yuanpei University, Hsinchu 300, Taiwan
| |
Collapse
|
8
|
Dang C, Wang Y, Li Q, Lu Y. Neuroimaging modalities in the detection of Alzheimer's disease-associated biomarkers. PSYCHORADIOLOGY 2023; 3:kkad009. [PMID: 38666112 PMCID: PMC11003434 DOI: 10.1093/psyrad/kkad009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 06/04/2023] [Accepted: 06/20/2023] [Indexed: 04/28/2024]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. Neuropathological changes in AD patients occur up to 10-20 years before the emergence of clinical symptoms. Specific diagnosis and appropriate intervention strategies are crucial during the phase of mild cognitive impairment (MCI) and AD. The detection of biomarkers has emerged as a promising tool for tracking the efficacy of potential therapies, making an early disease diagnosis, and prejudging treatment prognosis. Specifically, multiple neuroimaging modalities, including magnetic resonance imaging (MRI), positron emission tomography, optical imaging, and single photon emission-computed tomography, have provided a few potential biomarkers for clinical application. The MRI modalities described in this review include structural MRI, functional MRI, diffusion tensor imaging, magnetic resonance spectroscopy, and arterial spin labelling. These techniques allow the detection of presymptomatic diagnostic biomarkers in the brains of cognitively normal elderly people and might also be used to monitor AD disease progression after the onset of clinical symptoms. This review highlights potential biomarkers, merits, and demerits of different neuroimaging modalities and their clinical value in MCI and AD patients. Further studies are necessary to explore more biomarkers and overcome the limitations of multiple neuroimaging modalities for inclusion in diagnostic criteria for AD.
Collapse
Affiliation(s)
- Chun Dang
- Department of Periodical Press, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Yanchao Wang
- Department of Neurology, Chifeng University of Affiliated Hospital, Chifeng 024000, China
| | - Qian Li
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150081, China
| | - Yaoheng Lu
- Department of General Surgery, Chengdu Integrated Traditional Chinese Medicine and Western Medicine Hospital, Chengdu 610000, China
| |
Collapse
|
9
|
Rosenblum Y, Shiner T, Bregman N, Giladi N, Maidan I, Fahoum F, Mirelman A. Decreased aperiodic neural activity in Parkinson's disease and dementia with Lewy bodies. J Neurol 2023:10.1007/s00415-023-11728-9. [PMID: 37138179 DOI: 10.1007/s00415-023-11728-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 05/05/2023]
Abstract
Neural oscillations and signal complexity have been widely studied in neurodegenerative diseases, whereas aperiodic activity has not been explored yet in those disorders. Here, we assessed whether the study of aperiodic activity brings new insights relating to disease as compared to the conventional spectral and complexity analyses. Eyes-closed resting-state electroencephalography (EEG) was recorded in 21 patients with dementia with Lewy bodies (DLB), 28 patients with Parkinson's disease (PD), 27 patients with mild cognitive impairment (MCI) and 22 age-matched healthy controls. Spectral power was differentiated into its oscillatory and aperiodic components using the Irregularly Resampled Auto-Spectral Analysis. Signal complexity was explored using the Lempel-Ziv algorithm (LZC). We found that DLB patients showed steeper slopes of the aperiodic power component with large effect sizes compared to the controls and MCI and with a moderate effect size compared to PD. PD patients showed steeper slopes with a moderate effect size compared to controls and MCI. Oscillatory power and LZC differentiated only between DLB and other study groups and were not sensitive enough to detect differences between PD, MCI, and controls. In conclusion, both DLB and PD are characterized by alterations in aperiodic dynamics, which are more sensitive in detecting disease-related neural changes than the traditional spectral and complexity analyses. Our findings suggest that steeper aperiodic slopes may serve as a marker of network dysfunction in DLB and PD features.
Collapse
Affiliation(s)
- Yevgenia Rosenblum
- Laboratory of Early Markers of Neurodegeneration, Center for the Study of Movement, Cognition and Mobility, Neurological Institute, Tel Aviv Medical Center, Tel Aviv, Israel
- Cognitive Neurology Unit, Neurological Institute, Tel Aviv Medical Center, Tel Aviv, Israel
- Department of Neurology and Neurosurgery, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tamara Shiner
- Cognitive Neurology Unit, Neurological Institute, Tel Aviv Medical Center, Tel Aviv, Israel
- Department of Neurology and Neurosurgery, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Noa Bregman
- Cognitive Neurology Unit, Neurological Institute, Tel Aviv Medical Center, Tel Aviv, Israel
- Department of Neurology and Neurosurgery, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Nir Giladi
- Laboratory of Early Markers of Neurodegeneration, Center for the Study of Movement, Cognition and Mobility, Neurological Institute, Tel Aviv Medical Center, Tel Aviv, Israel
- Cognitive Neurology Unit, Neurological Institute, Tel Aviv Medical Center, Tel Aviv, Israel
- Epilepsy Unit, Neurological Institute, Tel Aviv Medical Center, Tel Aviv, Israel
- Department of Neurology and Neurosurgery, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Inbal Maidan
- Laboratory of Early Markers of Neurodegeneration, Center for the Study of Movement, Cognition and Mobility, Neurological Institute, Tel Aviv Medical Center, Tel Aviv, Israel
- Department of Neurology and Neurosurgery, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Firas Fahoum
- Epilepsy Unit, Neurological Institute, Tel Aviv Medical Center, Tel Aviv, Israel
- Department of Neurology and Neurosurgery, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Anat Mirelman
- Laboratory of Early Markers of Neurodegeneration, Center for the Study of Movement, Cognition and Mobility, Neurological Institute, Tel Aviv Medical Center, Tel Aviv, Israel.
- Department of Neurology and Neurosurgery, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
10
|
Hnilicova P, Kantorova E, Sutovsky S, Grofik M, Zelenak K, Kurca E, Zilka N, Parvanovova P, Kolisek M. Imaging Methods Applicable in the Diagnostics of Alzheimer's Disease, Considering the Involvement of Insulin Resistance. Int J Mol Sci 2023; 24:3325. [PMID: 36834741 PMCID: PMC9958721 DOI: 10.3390/ijms24043325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/29/2023] [Accepted: 01/30/2023] [Indexed: 02/10/2023] Open
Abstract
Alzheimer's disease (AD) is an incurable neurodegenerative disease and the most frequently diagnosed type of dementia, characterized by (1) perturbed cerebral perfusion, vasculature, and cortical metabolism; (2) induced proinflammatory processes; and (3) the aggregation of amyloid beta and hyperphosphorylated Tau proteins. Subclinical AD changes are commonly detectable by using radiological and nuclear neuroimaging methods such as magnetic resonance imaging (MRI), computed tomography (CT), positron emission tomography (PET), and single-photon emission computed tomography (SPECT). Furthermore, other valuable modalities exist (in particular, structural volumetric, diffusion, perfusion, functional, and metabolic magnetic resonance methods) that can advance the diagnostic algorithm of AD and our understanding of its pathogenesis. Recently, new insights into AD pathoetiology revealed that deranged insulin homeostasis in the brain may play a role in the onset and progression of the disease. AD-related brain insulin resistance is closely linked to systemic insulin homeostasis disorders caused by pancreas and/or liver dysfunction. Indeed, in recent studies, linkages between the development and onset of AD and the liver and/or pancreas have been established. Aside from standard radiological and nuclear neuroimaging methods and clinically fewer common methods of magnetic resonance, this article also discusses the use of new suggestive non-neuronal imaging modalities to assess AD-associated structural changes in the liver and pancreas. Studying these changes might be of great clinical importance because of their possible involvement in AD pathogenesis during the prodromal phase of the disease.
Collapse
Affiliation(s)
- Petra Hnilicova
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Ema Kantorova
- Clinic of Neurology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Stanislav Sutovsky
- 1st Department of Neurology, Faculty of Medicine, Comenius University in Bratislava and University Hospital, 813 67 Bratislava, Slovakia
| | - Milan Grofik
- Clinic of Neurology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Kamil Zelenak
- Clinic of Radiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Egon Kurca
- Clinic of Neurology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Norbert Zilka
- Institute of Neuroimmunology, Slovak Academy of Sciences, 845 10 Bratislava, Slovakia
| | - Petra Parvanovova
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| | - Martin Kolisek
- Biomedical Center Martin, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 036 01 Martin, Slovakia
| |
Collapse
|
11
|
Huang P, Zhang M. Magnetic Resonance Imaging Studies of Neurodegenerative Disease: From Methods to Translational Research. Neurosci Bull 2023; 39:99-112. [PMID: 35771383 PMCID: PMC9849544 DOI: 10.1007/s12264-022-00905-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 05/07/2022] [Indexed: 01/22/2023] Open
Abstract
Neurodegenerative diseases (NDs) have become a significant threat to an aging human society. Numerous studies have been conducted in the past decades to clarify their pathologic mechanisms and search for reliable biomarkers. Magnetic resonance imaging (MRI) is a powerful tool for investigating structural and functional brain alterations in NDs. With the advantages of being non-invasive and non-radioactive, it has been frequently used in both animal research and large-scale clinical investigations. MRI may serve as a bridge connecting micro- and macro-level analysis and promoting bench-to-bed translational research. Nevertheless, due to the abundance and complexity of MRI techniques, exploiting their potential is not always straightforward. This review aims to briefly introduce research progress in clinical imaging studies and discuss possible strategies for applying MRI in translational ND research.
Collapse
Affiliation(s)
- Peiyu Huang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009 China
| | - Minming Zhang
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009 China
| |
Collapse
|
12
|
Valkenborghs SR, Hillman CH, Al‐Iedani O, Nilsson M, Smith JJ, Leahy AA, Harries SK, Ramadan S, Lubans DR. Effect of high-intensity interval training on hippocampal metabolism in older adolescents. Psychophysiology 2022; 59:e14090. [PMID: 35599295 PMCID: PMC9787522 DOI: 10.1111/psyp.14090] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 04/26/2022] [Accepted: 04/26/2022] [Indexed: 12/30/2022]
Abstract
Although well-evidenced in older adults, the effects of exercise on the hippocampus in youth are relatively unknown. This study examined the impact of a 6-month school-based physical activity intervention on hippocampal metabolism in adolescents using magnetic resonance spectroscopy. A subset of lower fit older adolescents [N = 56, 61% female, 16.1 ± 0.4 years] was included from four secondary schools (10 classes) in New South Wales, Australia, who were participating in a larger cluster randomized controlled trial. Participants were randomized to the Burn 2 Learn (B2L) intervention (five classes, 30 participants) or a control group (five classes, 26 participants). Changes in hippocampal metabolism were assessed using linear mixed models adjusted for clustering at the class level. We observed group-by-time effects for the B2L intervention on N-acetylaspartate (NAA) (+2.66 mmol/L, 95% CI 0.20 to 5.11, d = 0.66) and glutamate+glutamine (Glx) (+3.38 mmol/L, 95% CI 0.34 to 6.42, d = 0.67) in the left hippocampus. Increases in left hippocampal NAA and Glx concentrations were associated with improvements in cardiorespiratory fitness (NAA: rs = 0.52, p = .016; Glx: rs = 0.57, p = .007), lower body muscular fitness (NAA: rs = 0.49, p = .018; Glx: rs = 0.59, p = .003), and working memory (NAA: rs = 0.42, p = .032; Glx: rs = 0.43, p = .028) in the intervention group. Our findings suggest physical activity may improve hippocampal metabolism in lower fit older adolescents with implications for working memory. Further studies involving larger samples are needed to replicate our findings.
Collapse
Affiliation(s)
- Sarah Ruth Valkenborghs
- School of Biomedical Sciences and PharmacyThe University of NewcastleCallaghanNew South WalesAustralia,Centre for Active Living and LearningThe University of NewcastleCallaghanNew South WalesAustralia
| | - Charles H. Hillman
- Center for Cognitive & Brain Health, Department of Psychology, Department of Physical Therapy, Movement, and Rehabilitation SciencesNortheastern UniversityBostonMassachusettsUSA
| | - Oun Al‐Iedani
- School of Health SciencesThe University of NewcastleCallaghanNew South WalesAustralia
| | - Michael Nilsson
- Centre for Rehab InnovationsThe University of NewcastleCallaghanNew South WalesAustralia,Priority Research Centre for Stroke and Brain InjuryThe University of NewcastleCallaghanNew South WalesAustralia,School of Medicine and Public HealthThe University of NewcastleCallaghanNew South WalesAustralia
| | - Jordan J. Smith
- Centre for Active Living and LearningThe University of NewcastleCallaghanNew South WalesAustralia,School of EducationThe University of NewcastleCallaghanNew South WalesAustralia
| | - Angus Aaron Leahy
- Centre for Active Living and LearningThe University of NewcastleCallaghanNew South WalesAustralia,School of EducationThe University of NewcastleCallaghanNew South WalesAustralia
| | - Simon K. Harries
- Centre for Active Living and LearningThe University of NewcastleCallaghanNew South WalesAustralia,School of EducationThe University of NewcastleCallaghanNew South WalesAustralia
| | - Saadallah Ramadan
- School of Health SciencesThe University of NewcastleCallaghanNew South WalesAustralia
| | - David Revalds Lubans
- Centre for Active Living and LearningThe University of NewcastleCallaghanNew South WalesAustralia,School of EducationThe University of NewcastleCallaghanNew South WalesAustralia
| |
Collapse
|
13
|
Spatio-temporal metabolic rewiring in the brain of TgF344-AD rat model of Alzheimer's disease. Sci Rep 2022; 12:16958. [PMID: 36216838 PMCID: PMC9550832 DOI: 10.1038/s41598-022-20962-6] [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: 05/10/2022] [Accepted: 09/21/2022] [Indexed: 12/29/2022] Open
Abstract
Brain damage associated with Alzheimer's disease (AD) occurs even decades before the symptomatic onset, raising the need to investigate its progression from prodromal stages. In this context, animal models that progressively display AD pathological hallmarks (e.g. TgF344-AD) become crucial. Translational technologies, such as magnetic resonance spectroscopy (MRS), enable the longitudinal metabolic characterization of this disease. However, an integrative approach is required to unravel the complex metabolic changes underlying AD progression, from early to advanced stages. TgF344-AD and wild-type (WT) rats were studied in vivo on a 7 Tesla MRI scanner, for longitudinal quantitative assessment of brain metabolic profile changes using MRS. Disease progression was investigated at 4 time points, from 9 to 18 months of age, and in 4 regions: cortex, hippocampus, striatum, and thalamus. Compared to WT, TgF344-AD rats replicated common findings in AD patients, including decreased N-acetylaspartate in the cortex, hippocampus and thalamus, and decreased glutamate in the thalamus and striatum. Different longitudinal evolution of metabolic concentration was observed between TgF344-AD and WT groups. Namely, age-dependent trajectories differed between groups for creatine in the cortex and thalamus and for taurine in cortex, with significant decreases in Tg344-AD animals; whereas myo-inositol in the thalamus and striatum showed greater increase along time in the WT group. Additional analysis revealed divergent intra- and inter-regional metabolic coupling in each group. Thus, in cortex, strong couplings of N-acetylaspartate and creatine with myo-inositol in WT, but with taurine in TgF344-AD rats were observed; whereas in the hippocampus, myo-inositol, taurine and choline compounds levels were highly correlated in WT but not in TgF344-AD animals. Furthermore, specific cortex-hippocampus-striatum metabolic crosstalks were found for taurine levels in the WT group but for myo-inositol levels in the TgF344-AD rats. With a systems biology perspective of metabolic changes in AD pathology, our results shed light into the complex spatio-temporal metabolic rewiring in this disease, reported here for the first time. Age- and tissue-dependent imbalances between myo-inositol, taurine and other metabolites, such as creatine, unveil their role in disease progression, while pointing to the inadequacy of the latter as an internal reference for quantification.
Collapse
|
14
|
Li Y, Lu T, Wei W, Lin Z, Ding L, Li Z, Xue X. Swimming Training Mitigates Neurological Impairment of Intracerebral Haemorrhage in Mice via the Serine-Threonine Kinase/Glycogen Synthase Kinase 3β Signalling Pathway. Neuroscience 2022; 501:72-84. [PMID: 35961525 DOI: 10.1016/j.neuroscience.2022.07.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 07/25/2022] [Accepted: 07/27/2022] [Indexed: 10/15/2022]
Abstract
Swimming training (ST) can mitigate functional disorders in neurological diseases, but the effect and mechanism of ST in improving the neurological function of intracerebral haemorrhage (ICH) have not been reported. Our study aimed to explore the protective effect of early ST on ICH mice and its relationship with the serine-threonine kinase (Akt)/glycogen synthase kinase 3β (GSK3β) pathway. Our findings showed that the ICH model mice had poor behavioural manifestations in the Y maze test and open field test compared to the ST group and sham group. The modified neurological severity score was increased in the ICH mice, and 7 days of ST intervention significantly attenuated the neurological deficits. The ratios of myo-inositol/creatine, lactate/creatine and glutamate/creatine were decreased, and the ratios of N-acetylaspartate/creatine and choline/creatine were increased in the ICH mice with ST intervention. ST intervention decreased the expression of Iba1 and GFAP. Seven days of ST significantly increased the expression of p-Akt/Akt compared to that in the ICH mice. Furthermore, the Akt kinase inhibitor GSK690693 exacerbated neurological impairment, increased the expression of Iba1, GFAP and Bax/Bcl-2, and reversed the anti-apoptotic effects and anti-glia activation of ST, which was associated with the inhibition of p-Akt/Akt and p-GSK3β/GSK3β expression. These results indicated that the protective role of ST in ICH was mediated via the Akt/GSK3β pathway. In conclusion, ST displayed neuroprotection by inhibiting apoptosis and glial activation in ICH mice by activating the Akt/GSK3β signalling pathway.
Collapse
Affiliation(s)
- Yongxu Li
- Affiliated Rehabilitation Hospital of Fujian University of Traditional Chinese Medicine, China; College of Rehabilitation Medicine, Fujian University of Chinese Medicine, China
| | - Taotao Lu
- Affiliated Rehabilitation Hospital of Fujian University of Traditional Chinese Medicine, China; College of Rehabilitation Medicine, Fujian University of Chinese Medicine, China
| | - Wei Wei
- Affiliated Rehabilitation Hospital of Fujian University of Traditional Chinese Medicine, China
| | - Zhicheng Lin
- Affiliated Rehabilitation Hospital of Fujian University of Traditional Chinese Medicine, China
| | - Linlin Ding
- Affiliated Rehabilitation Hospital of Fujian University of Traditional Chinese Medicine, China
| | - Zhaohui Li
- Anxi County Hospital of Traditional Chinese Medicine, Quanzhou, Fujian Province, China.
| | - Xiehua Xue
- Affiliated Rehabilitation Hospital of Fujian University of Traditional Chinese Medicine, China; Fujian Key Laboratory of Rehabilitation Technology and Cognition Rehabilitation, China.
| |
Collapse
|
15
|
He C, Rong S, Zhang P, Li R, Li X, Li Y, Wang L, Zhang Y. Metabolite changes in prefrontal lobes and the anterior cingulate cortex correlate with processing speed and executive function in Parkinson disease patients. Quant Imaging Med Surg 2022; 12:4226-4238. [PMID: 35919059 PMCID: PMC9338382 DOI: 10.21037/qims-21-1126] [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: 11/22/2021] [Accepted: 05/23/2022] [Indexed: 11/06/2022]
Abstract
Background Processing speed and executive function can be impaired in patients with Parkinson disease (PD). However, the neural factors related to the slowdown in processing speed and dysexecutive function in PD are not completely understood. The objective of this study is to investigate the metabolic changes of the frontal and anterior cingulate cortex (ACC) through the use of 1H magnetic resonance spectroscopy and to explore the association between cognitive function and metabolic ratios. Methods In this retrospective case-control study, we conducted neuropsychological assessments of executive function and information processing speed in healthy controls (HCs) and in patients with PD. Chemical information was obtained for the of N-acetyl-aspartate (NAA):creatine (Cr) ratio and the choline-containing compounds (Cho):Cr ratio within the bilateral prefrontal cortex and ACC. Using hierarchical multiple regression analysis, we analyzed the relationship between cognitive function and metabolic ratios in the bilateral prefrontal lobe and ACC in patients with PD. Results In all, 59 patients with PD and 30 HCs were recruited. Patients with PD showed worse performance in executive function and processing speed compared with HCs (P<0.001). In patients with PD, the Cho:Cr ratios in the ACC (Z=2.20, P=0.028) and the right prefrontal cortex (t=2.16, P=0.034) were significantly increased. The hierarchical multiple regressions in patients with PD showed that the NAA:Cr ratio in the ACC correlated with the Stroop A completion times (P<0.05) and that the NAA:Cr ratio of the right prefrontal cortex correlated with the scores of the Wechsler Adult Intelligence Scale (WAIS)-Digit symbol test (P<0.05). Conclusions Information processing speed and executive function are impaired in patients with PD. Neuronal integrity and membrane turnover in the ACC and the right prefrontal cortex may be important factors in the slowdown of the information processing speed in patients with PD.
Collapse
Affiliation(s)
- Chentao He
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Siming Rong
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Piao Zhang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ruitao Li
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xiaohong Li
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yan Li
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Lijuan Wang
- Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Yuhu Zhang
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.,Department of Neurology, Guangdong Neuroscience Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
16
|
Brown KM, Nair JK, Janas MM, Anglero-Rodriguez YI, Dang LTH, Peng H, Theile CS, Castellanos-Rizaldos E, Brown C, Foster D, Kurz J, Allen J, Maganti R, Li J, Matsuda S, Stricos M, Chickering T, Jung M, Wassarman K, Rollins J, Woods L, Kelin A, Guenther DC, Mobley MW, Petrulis J, McDougall R, Racie T, Bombardier J, Cha D, Agarwal S, Johnson L, Jiang Y, Lentini S, Gilbert J, Nguyen T, Chigas S, LeBlanc S, Poreci U, Kasper A, Rogers AB, Chong S, Davis W, Sutherland JE, Castoreno A, Milstein S, Schlegel MK, Zlatev I, Charisse K, Keating M, Manoharan M, Fitzgerald K, Wu JT, Maier MA, Jadhav V. Expanding RNAi therapeutics to extrahepatic tissues with lipophilic conjugates. Nat Biotechnol 2022; 40:1500-1508. [PMID: 35654979 DOI: 10.1038/s41587-022-01334-x] [Citation(s) in RCA: 98] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 04/22/2022] [Indexed: 01/03/2023]
Abstract
Therapeutics based on short interfering RNAs (siRNAs) delivered to hepatocytes have been approved, but new delivery solutions are needed to target additional organs. Here we show that conjugation of 2'-O-hexadecyl (C16) to siRNAs enables safe, potent and durable silencing in the central nervous system (CNS), eye and lung in rodents and non-human primates with broad cell type specificity. We show that intrathecally or intracerebroventricularly delivered C16-siRNAs were active across CNS regions and cell types, with sustained RNA interference (RNAi) activity for at least 3 months. Similarly, intravitreal administration to the eye or intranasal administration to the lung resulted in a potent and durable knockdown. The preclinical efficacy of an siRNA targeting the amyloid precursor protein was evaluated through intracerebroventricular dosing in a mouse model of Alzheimer's disease, resulting in amelioration of physiological and behavioral deficits. Altogether, C16 conjugation of siRNAs has the potential for safe therapeutic silencing of target genes outside the liver with infrequent dosing.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Jing Li
- Alnylam Pharmaceuticals, Cambridge, MA, USA
| | | | | | | | | | | | | | | | - Alex Kelin
- Alnylam Pharmaceuticals, Cambridge, MA, USA
| | | | | | | | | | | | | | - Diana Cha
- Alnylam Pharmaceuticals, Cambridge, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Mai W, Zhang A, Liu Q, Tang L, Wei Y, Su J, Duan G, Teng J, Nong X, Yu B, Li C, Shao L, Deng D, Chen S, Zhao L. Effects of Moxa Cone Moxibustion Therapy on Cognitive Function and Brain Metabolic Changes in MCI Patients: A Pilot 1H-MRS Study. Front Aging Neurosci 2022; 14:773687. [PMID: 35721029 PMCID: PMC9204283 DOI: 10.3389/fnagi.2022.773687] [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: 09/10/2021] [Accepted: 04/12/2022] [Indexed: 11/13/2022] Open
Abstract
Objective To explore the effect of moxa cone moxibustion on N-acetyl aspartate/total creatinine (NAA/tCr) and choline/total creatinine (Cho/tCr) in the bilateral hippocampus (HIP) and bilateral posterior cingulate gyrus (PCG) in patients with mild cognitive impairment (MCI) using hydrogen proton magnetic resonance spectroscopy (1H-MRS) and to provide imaging basis for moxa cone moxibustion treatment for MCI. Methods One hundred eight patients with MCI were served as the MCI group, and 67 age-matched subjects were enrolled as the normal control group. The MCI group was randomized and allocated into acupoint group, drug group, and sham acupoint group, with 36 cases in each group. Some patients in each group withdrew. Finally, 25 cases were included in the acupoint group, 24 cases in the drug group, and 20 cases in the sham acupoint group. The drug group was treated with oral donepezil hydrochloride. The acupoint group and sham acupoint group received moxa cone moxibustion treatment. Mini-mental state exam (MMSE) and Montreal cognitive assessment (MoCA) scores were recorded before intervention, at the end of the first and the second months of intervention, and in the 5th month of follow-up. The NAA/tCr and Cho/tCr ratios in the HIP and PCG were bilaterally measured by 1H-MRS before and after intervention. Results Before intervention, compared with the normal control group, the MMSE and MoCA scores, the Cho/tCr ratio in the right HIP, the NAA/tCr ratio in the bilateral HIP, and the NAA/tCr ratio in the left PCG in the three treatment groups decreased significantly (both p < 0.01), and the NAA/tCr ratio in the right PCG significantly reduced in the acupoint and drug groups (p < 0.05). After two months of treatment, compared with the normal control group, there were no differences in the MoCA scores, the NAA/tCr, and Cho/tCr ratios in the bilateral PCG and bilateral HIP in the three treatment groups (p > 0.05). However, the MMSE scores in the drug group decreased when compared with the acupoint group and normal control group (p < 0.05, p < 0.01). The scores of MMSE and MoCA in the acupoint group and sham acupoint group at all time points were better than those in the drug group, which were similar to those in the normal control group. Conclusion Our findings suggest that moxibustion could improve the cognitive function of patients with MCI. The mechanism may be related to the improvement of abnormal brain metabolism in HIP and PCG.
Collapse
Affiliation(s)
- Wei Mai
- Guangxi University of Chinese Medicine, Nanning, China
- Department of Traditional Chinese Medicine, Guangxi Tumour Hospital, Nanning, China
| | | | - Qiang Liu
- Xinghu Outpatient Department, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Liying Tang
- Xinghu Outpatient Department, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Yichen Wei
- Department of Radiology, The First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, China
| | - Jiahui Su
- Department of Acupuncture, The First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, China
| | - Gaoxiong Duan
- Department of Radiology, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
| | - Jinlong Teng
- Department of Acupuncture, The First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, China
| | - Xiucheng Nong
- Department of Acupuncture, The First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, China
| | - Bihan Yu
- Department of Acupuncture, The First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, China
| | - Chong Li
- Department of Acupuncture, The First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, China
| | - Lijuan Shao
- Department of Acupuncture, The First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, China
| | - Demao Deng
- Department of Radiology, The People’s Hospital of Guangxi Zhuang Autonomous Region, Nanning, China
- Demao Deng,
| | - Shangjie Chen
- Department of Rehabilitation, The Second Affiliated Hospital of Shenzhen University, Shenzhen, China
- Shangjie Chen,
| | - Lihua Zhao
- Department of Acupuncture, The First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, China
- *Correspondence: Lihua Zhao,
| |
Collapse
|
18
|
Liu W, Li J, Yang M, Ke X, Dai Y, Lin H, Wang S, Chen L, Tao J. Chemical genetic activation of the cholinergic basal forebrain hippocampal circuit rescues memory loss in Alzheimer's disease. Alzheimers Res Ther 2022; 14:53. [PMID: 35418161 PMCID: PMC9006585 DOI: 10.1186/s13195-022-00994-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 03/27/2022] [Indexed: 11/18/2022]
Abstract
Background The degeneration of the cholinergic circuit from the basal forebrain to the hippocampus contributes to memory loss in patients suffering from Alzheimer’s disease (AD). However, the internal relationships between the acetylcholine (Ach) cycle and memory decline during the early stages of AD currently remain unknown. Here, we investigate the mechanisms underlying the activation of the cholinergic circuit and its impact on learning and memory using APP/PS1 mice models. Methods Novel object recognition and Morris water maze tests were used to measure learning and memory function. Magnetic resonance spectrum (MRS) imaging was applied to longitudinally track changes in neurochemical metabolism in APP/PS1 mice aged 2, 4, 6, and 8 months. The number of neurons and the deposition of Aβ plaques were measured using Nissl, immunohistochemistry, and Thioflavin S staining. We then employed a chemogenetic strategy to selectively activate the cholinergic circuit from the medial septal nucleus (MS) and the vertical limb of the diagonal band nucleus (VDB) on the basal forebrain to the hippocampus. MRS and immunoblotting techniques were used to measure the neurochemical metabolism levels and cholinergic-related proteins, respectively. Results We found that the levels of choline (Cho) in the basal forebrain were markedly higher compared to other brain regions and that its decrease along with N-acetyl aspartate (NAA) levels in the hippocampus was accompanied by memory deficits in APP/PS1 mice aged 4, 6, and 8 months. In terms of pathology, we observed that the deposition of Aβ plaques gradually aggravated throughout the cerebral cortex and hippocampus in APP/PS1 mice aged 6 and 8 months, while no Aβ deposition was detected in the basal forebrain. In contrast, the activity of choline acetyltransferase (ChAT) enzyme in the basal forebrain was decreased at 6 months of age and the cholinergic neurons were lost in the basal forebrain at 8 months of age. In addition, the activation of the cholinergic circuit from the MS and VDB to the hippocampus using chemical genetics is able to improve learning and reduce memory impairment in APP/PS1 mice. Similarly, the levels of Cho in the basal forebrain; NAA in the hippocampus, as well as the expression of ChAT and vesicular acetylcholine transporter (vAchT) in the basal forebrain; and muscarinic acetylcholine receptor 2 (CHRM2) in the hippocampus all increased. Conclusions These findings demonstrate that the neurochemical Cho and NAA of the cholinergic circuit can be used as biomarkers to enable the early diagnosis of AD. In addition, memory impairment in APP/PS1 mice can be attenuated using chemical genetics-driven Ach cycle activity of the cholinergic circuit. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-022-00994-w.
Collapse
Affiliation(s)
- Weilin Liu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China.,The Academy of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Jianhong Li
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Minguang Yang
- The Academy of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Xiaohua Ke
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Yaling Dai
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Huawei Lin
- Rehabilitation Medical Technology Joint National Local Engineering Research Center, Fuzhou, 350122, Fujian, China
| | - Sinuo Wang
- Rehabilitation Medical Technology Joint National Local Engineering Research Center, Fuzhou, 350122, Fujian, China
| | - Lidian Chen
- The Academy of Rehabilitation Industry, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China
| | - Jing Tao
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, Fujian, China.
| |
Collapse
|
19
|
Kara F, Joers JM, Deelchand DK, Park YW, Przybelski SA, Lesnick TG, Senjem ML, Zeydan B, Knopman DS, Lowe VJ, Vemuri P, Mielke MM, Machulda MM, Jack CR, Petersen RC, Öz G, Kantarci K. 1H MR spectroscopy biomarkers of neuronal and synaptic function are associated with tau deposition in cognitively unimpaired older adults. Neurobiol Aging 2022; 112:16-26. [PMID: 35038671 PMCID: PMC8976711 DOI: 10.1016/j.neurobiolaging.2021.12.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 12/23/2021] [Accepted: 12/26/2021] [Indexed: 12/25/2022]
Abstract
Proton magnetic resonance spectroscopy (1H MRS) may provide information on pathophysiological changes associated with tau deposition in cognitively unimpaired older adults. In this study, the associations of posterior cingulate gyrus tau and amyloid beta (Aβ) deposition on PET with 1H MRS metabolite ratios acquired from bilateral posterior cingulate gyri were investigated in cognitively unimpaired older adults. Participants (n = 40) from the Mayo Clinic Study of Aging underwent single-voxel sLASER 1H MRS from the posterior cingulate gyrus at 3 Tesla, 18F-flortaucipir, and 11C- Pittsburgh Compound B (PiB) PET. An increase in posterior cingulate gyrus tau deposition, but not elevated Aβ, was associated with lower N-acetylaspartate/total creatine (tCr) and glutamate (Glu)/tCr ratios, and sex by tau interaction was observed in association with Glu/tCr. Higher tau levels in cognitively unimpaired older adults are associated with biomarkers of neural and synaptic injury even in the absence of cognitive impairment and these relationships appear to be stronger in women than in men.
Collapse
Affiliation(s)
- Firat Kara
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - James M Joers
- Department of Radiology, Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA
| | - Dinesh K Deelchand
- Department of Radiology, Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA
| | - Young Woo Park
- Department of Radiology, Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA
| | - Scott A Przybelski
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Timothy G Lesnick
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | - Matthew L Senjem
- Department of Radiology, Mayo Clinic, Rochester, MN, USA; Department of Information Technology, Mayo Clinic, Rochester, MN, USA
| | - Burcu Zeydan
- Department of Radiology, Mayo Clinic, Rochester, MN, USA; Department of Neurology, Mayo Clinic-Minnesota, Rochester, MN, USA
| | - David S Knopman
- Department of Neurology, Mayo Clinic-Minnesota, Rochester, MN, USA
| | - Val J Lowe
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | | | - Michelle M Mielke
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA; Department of Neurology, Mayo Clinic-Minnesota, Rochester, MN, USA
| | - Mary M Machulda
- Department of Psychiatry and Psychology, Mayo Clinic-Minnesota, Rochester, MN, USA
| | | | | | - Gülin Öz
- Department of Radiology, Center for Magnetic Resonance Research, University of Minnesota, Minneapolis, MN, USA
| | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
20
|
Liu W, Li J, Li L, Zhang Y, Yang M, Liang S, Li L, Dai Y, Chen L, Jia W, He X, Lin H, Tao J. Enhanced Medial Prefrontal Cortex and Hippocampal Activity Improves Memory Generalization in APP/PS1 Mice: A Multimodal Animal MRI Study. Front Cell Neurosci 2022; 16:848967. [PMID: 35386301 PMCID: PMC8977524 DOI: 10.3389/fncel.2022.848967] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 02/08/2022] [Indexed: 02/03/2023] Open
Abstract
Memory generalization allows individuals to extend previously learned movement patterns to similar environments, contributing to cognitive flexibility. In Alzheimer’s disease (AD), the disturbance of generalization is responsible for the deficits of episodic memory, causing patients with AD to forget or misplace things, even lose track of the way home. Cognitive training can effectively improve the cognition of patients with AD through changing thinking mode and memory flexibility. In this study, a T-shaped maze was utilized to simulate cognitive training in APP/PS1 mice to elucidate the potential mechanisms of beneficial effects after cognitive training. We found that cognitive training conducted by a T-shaped maze for 4 weeks can improve the memory generalization ability of APP/PS1 mice. The results of functional magnetic resonance imaging (fMRI) showed that the functional activity of the medial prefrontal cortex (mPFC) and hippocampus was enhanced after cognitive training, and the results of magnetic resonance spectroscopy (MRS) showed that the neurochemical metabolism of N-acetyl aspartate (NAA) and glutamic acid (Glu) in mPFC, hippocampus and reuniens (Re) thalamic nucleus were escalated. Furthermore, the functional activity of mPFC and hippocampus was negatively correlated with the escape latency in memory generalization test. Therefore, these results suggested that cognitive training might improve memory generalization through enhancing the functional activity of mPFC and hippocampus and increasing the metabolism of NAA and Glu in the brain regions of mPFC, hippocampus and Re nucleus.
Collapse
Affiliation(s)
- Weilin Liu
- National-Local Joint Engineering Research Center of Rehabilitation Medicine Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jianhong Li
- 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
| | - Yuhao Zhang
- 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
| | - Shengxiang Liang
- TCM Rehabilitation Research Center of SATCM, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Long Li
- TCM Rehabilitation Research Center of SATCM, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Yaling Dai
- TCM Rehabilitation Research Center of SATCM, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Lewen Chen
- 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
| | - Xiaojun He
- TCM Rehabilitation Research Center of SATCM, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Huawei Lin
- TCM Rehabilitation Research Center of SATCM, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jing Tao
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- *Correspondence: Jing Tao,
| |
Collapse
|
21
|
Ma WY, Tian MJ, Yao Q, Li Q, Tang FY, Xiao CY, Shi JP, Chen J. Neuroimaging alterations in dementia with Lewy bodies and neuroimaging differences between dementia with Lewy bodies and Alzheimer's disease: An activation likelihood estimation meta-analysis. CNS Neurosci Ther 2021; 28:183-205. [PMID: 34873859 PMCID: PMC8739049 DOI: 10.1111/cns.13775] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 11/07/2021] [Accepted: 11/21/2021] [Indexed: 12/11/2022] Open
Abstract
Aims The aim of this study was to identify brain regions with local, structural, and functional abnormalities in dementia with Lewy bodies (DLB) and uncover the differences between DLB and Alzheimer's disease (AD). The neural networks involved in the identified abnormal brain regions were further described. Methods PubMed, Web of Science, OVID, Science Direct, and Cochrane Library databases were used to identify neuroimaging studies that included DLB versus healthy controls (HCs) or DLB versus AD. The coordinate‐based meta‐analysis and functional meta‐analytic connectivity modeling were performed using the activation likelihood estimation algorithm. Results Eleven structural studies and fourteen functional studies were included in this quantitative meta‐analysis. DLB patients showed a dysfunction in the bilateral inferior parietal lobule and right lingual gyrus compared with HC patients. DLB patients showed a relative preservation of the medial temporal lobe and a tendency of lower metabolism in the right lingual gyrus compared with AD. The frontal‐parietal, salience, and visual networks were all abnormally co‐activated in DLB, but the default mode network remained normally co‐activated compared with AD. Conclusions The convergence of local brain regions and co‐activation neural networks might be potential specific imaging markers in the diagnosis of DLB. This might provide a pathway for the neural regulation in DLB patients, and it might contribute to the development of specific interventions for DLB and AD.
Collapse
Affiliation(s)
- Wen-Ying Ma
- Department of Neurology, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Min-Jie Tian
- Department of Neurology, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qun Yao
- Department of Neurology, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qian Li
- Department of Neurology, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fan-Yu Tang
- Department of Neurology, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chao-Yong Xiao
- Department of Radiology, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jing-Ping Shi
- Department of Neurology, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.,Institute of Brain Functional Imaging, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiu Chen
- Institute of Neuropsychiatry, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu, China.,Institute of Brain Functional Imaging, Affiliated Nanjing Brain Hospital, Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
22
|
Piersson AD, Ibrahim B, Suppiah S, Mohamad M, Hassan HA, Omar NF, Ibrahim MI, Yusoff AN, Ibrahim N, Saripan MI, Razali RM. Multiparametric MRI for the improved diagnostic accuracy of Alzheimer's disease and mild cognitive impairment: Research protocol of a case-control study design. PLoS One 2021; 16:e0252883. [PMID: 34547018 PMCID: PMC8454976 DOI: 10.1371/journal.pone.0252883] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 05/18/2021] [Indexed: 11/19/2022] Open
Abstract
Background Alzheimer’s disease (AD) is a major neurocognitive disorder identified by memory loss and a significant cognitive decline based on previous level of performance in one or more cognitive domains that interferes in the independence of everyday activities. The accuracy of imaging helps to identify the neuropathological features that differentiate AD from its common precursor, mild cognitive impairment (MCI). Identification of early signs will aid in risk stratification of disease and ensures proper management is instituted to reduce the morbidity and mortality associated with AD. Magnetic resonance imaging (MRI) using structural MRI (sMRI), functional MRI (fMRI), diffusion tensor imaging (DTI), and magnetic resonance spectroscopy (1H-MRS) performed alone is inadequate. Thus, the combination of multiparametric MRI is proposed to increase the accuracy of diagnosing MCI and AD when compared to elderly healthy controls. Methods This protocol describes a non-interventional case control study. The AD and MCI patients and the healthy elderly controls will undergo multi-parametric MRI. The protocol consists of sMRI, fMRI, DTI, and single-voxel proton MRS sequences. An eco-planar imaging (EPI) will be used to perform resting-state fMRI sequence. The structural images will be analysed using Computational Anatomy Toolbox-12, functional images will be analysed using Statistical Parametric Mapping-12, DPABI (Data Processing & Analysis for Brain Imaging), and Conn software, while DTI and 1H-MRS will be analysed using the FSL (FMRIB’s Software Library) and Tarquin respectively. Correlation of the MRI results and the data acquired from the APOE genotyping, neuropsychological evaluations (i.e. Montreal Cognitive Assessment [MoCA], and Mini–Mental State Examination [MMSE] scores) will be performed. The imaging results will also be correlated with the sociodemographic factors. The diagnosis of AD and MCI will be standardized and based on the DSM-5 criteria and the neuropsychological scores. Discussion The combination of sMRI, fMRI, DTI, and MRS sequences can provide information on the anatomical and functional changes in the brain such as regional grey matter volume atrophy, impaired functional connectivity among brain regions, and decreased metabolite levels specifically at the posterior cingulate cortex/precuneus. The combination of multiparametric MRI sequences can be used to stratify the management of MCI and AD patients. Accurate imaging can decide on the frequency of follow-up at memory clinics and select classifiers for machine learning that may aid in the disease identification and prognostication. Reliable and consistent quantification, using standardised protocols, are crucial to establish an optimal diagnostic capability in the early detection of Alzheimer’s disease.
Collapse
Affiliation(s)
- Albert Dayor Piersson
- Diagnostic Imaging and Radiotherapy Programme, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
- Department of Imaging Technology & Sonography, School of Allied Health Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Buhari Ibrahim
- Faculty of Medicine and Health Sciences, Department of Radiology, Universiti Putra Malaysia, Seri Kembangan, Malaysia
- Faculty of Medicine and Health Sciences, Neuroscience Laboratory for Cognitive Function and Behavioural Imaging (NeuroCoB), Universiti Putra Malaysia, Seri Kembangan, Malaysia
- Faculty of Basic Medical Sciences, Department of Physiology, Bauchi State University PMB 65, Gadau, Nigeria
| | - Subapriya Suppiah
- Faculty of Medicine and Health Sciences, Department of Radiology, Universiti Putra Malaysia, Seri Kembangan, Malaysia
- Faculty of Medicine and Health Sciences, Neuroscience Laboratory for Cognitive Function and Behavioural Imaging (NeuroCoB), Universiti Putra Malaysia, Seri Kembangan, Malaysia
- * E-mail:
| | - Mazlyfarina Mohamad
- Diagnostic Imaging and Radiotherapy Programme, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Hasyma Abu Hassan
- Faculty of Medicine and Health Sciences, Department of Radiology, Universiti Putra Malaysia, Seri Kembangan, Malaysia
| | - Nur Farhayu Omar
- Faculty of Medicine and Health Sciences, Department of Radiology, Universiti Putra Malaysia, Seri Kembangan, Malaysia
| | - Mohd Izuan Ibrahim
- Diagnostic Imaging and Radiotherapy Programme, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Ahmad Nazlim Yusoff
- Diagnostic Imaging and Radiotherapy Programme, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Normala Ibrahim
- Faculty of Medicine and Health Sciences, Department of Psychiatry, Universiti Putra Malaysia, Seri Kembangan, Malaysia
| | - M. Iqbal Saripan
- Faculty of Engineering, Department of Computer & Communication Systems, University Putra Malaysia, Seri Kembangan, Malaysia
| | - Rizah Mazzuin Razali
- Gerontology Unit, Department of Medicine, Hospital Kuala Lumpur, Kuala Lumpur, Malaysia
| |
Collapse
|
23
|
Montal V, Barroeta I, Bejanin A, Pegueroles J, Carmona-Iragui M, Altuna M, Benejam B, Videla L, Fernández S, Padilla C, Aranha MR, Iulita MF, Vidal-Piñeiro D, Alcolea D, Blesa R, Lleó A, Fortea J. Metabolite Signature of Alzheimer's Disease in Adults with Down Syndrome. Ann Neurol 2021; 90:407-416. [PMID: 34309066 DOI: 10.1002/ana.26178] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 12/14/2022]
Abstract
OBJECTIVE The purpose of this study was to examine the Alzheimer's disease metabolite signature through magnetic resonance spectroscopy in adults with Down syndrome and its relation with Alzheimer's disease biomarkers and cortical thickness. METHODS We included 118 adults with Down syndrome from the Down Alzheimer Barcelona Imaging Initiative and 71 euploid healthy controls from the Sant Pau Initiative on Neurodegeneration cohort. We measured the levels of myo-inositol (a marker of neuroinflammation) and N-acetyl-aspartate (a marker of neuronal integrity) in the precuneus using magnetic resonance spectroscopy. We investigated the changes with age and along the disease continuum (asymptomatic, prodromal Alzheimer's disease, and Alzheimer's disease dementia stages). We assessed the relationship between these metabolites and Aβ42 /Aβ40 ratio, phosphorylated tau-181, neurofilament light (NfL), and YKL-40 cerebrospinal fluid levels as well as amyloid positron emission tomography uptake using Spearman correlations controlling for multiple comparisons. Finally, we computed the relationship between cortical thickness and metabolite levels using Freesurfer. RESULTS Asymptomatic adults with Down syndrome had a 27.5% increase in the levels of myo-inositol, but equal levels of N-acetyl-aspartate compared to euploid healthy controls. With disease progression, myo-inositol levels increased, whereas N-acetyl-aspartate levels decreased in symptomatic stages of the disease. Myo-inositol was associated with amyloid, tau, and neurodegeneration markers, mainly at symptomatic stages of the disease, whereas N-acetyl-aspartate was related to neurodegeneration biomarkers in symptomatic stages. Both metabolites were significantly associated with cortical thinning, mainly in symptomatic participants. INTERPRETATION Magnetic resonance spectroscopy detects Alzheimer's disease related inflammation and neurodegeneration, and could be a good noninvasive disease-stage biomarker in Down syndrome. ANN NEUROL 2021.
Collapse
Affiliation(s)
- Victor Montal
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Isabel Barroeta
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Alexandre Bejanin
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Jordi Pegueroles
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - María Carmona-Iragui
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Barcelona Down Medical Center. Fundació Catalana Síndrome de Down, Barcelona, Spain
| | - Miren Altuna
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Barcelona Down Medical Center. Fundació Catalana Síndrome de Down, Barcelona, Spain
| | - Bessy Benejam
- Barcelona Down Medical Center. Fundació Catalana Síndrome de Down, Barcelona, Spain
| | - Laura Videla
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Barcelona Down Medical Center. Fundació Catalana Síndrome de Down, Barcelona, Spain
| | - Susana Fernández
- Barcelona Down Medical Center. Fundació Catalana Síndrome de Down, Barcelona, Spain
| | - Concepcion Padilla
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Mateus Rozalem Aranha
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Maria Florencia Iulita
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Didac Vidal-Piñeiro
- Department of Psychology, Centre for Lifespan Changes in Brain and Cognition, University of Oslo, Oslo, Norway
| | - Daniel Alcolea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Rafael Blesa
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Alberto Lleó
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Juan Fortea
- Sant Pau Memory Unit, Department of Neurology, Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain.,Center of Biomedical Investigation Network for Neurodegenerative Diseases (CIBERNED), Madrid, Spain.,Barcelona Down Medical Center. Fundació Catalana Síndrome de Down, Barcelona, Spain
| | | |
Collapse
|
24
|
Wong D, Atiya S, Fogarty J, Montero-Odasso M, Pasternak SH, Brymer C, Borrie MJ, Bartha R. Reduced Hippocampal Glutamate and Posterior Cingulate N-Acetyl Aspartate in Mild Cognitive Impairment and Alzheimer's Disease Is Associated with Episodic Memory Performance and White Matter Integrity in the Cingulum: A Pilot Study. J Alzheimers Dis 2021; 73:1385-1405. [PMID: 31958093 DOI: 10.3233/jad-190773] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Identification of biological changes underlying the early symptoms of Alzheimer's disease (AD) will help to identify and stage individuals prior to symptom onset. The limbic system, which supports episodic memory and is impaired early in AD, is a primary target. In this study, brain metabolism and microstructure evaluated by high field (7 Tesla) proton magnetic resonance spectroscopy (1H-MRS) and diffusion tensor imaging (DTI) were evaluated in the limbic system of eight individuals with mild cognitive impairment (MCI), nine with AD, and sixteen normal elderly controls (NEC). Left hippocampal glutamate and posterior cingulate N-acetyl aspartate concentrations were reduced in MCI and AD compared to NEC. Differences in DTI metrics indicated volume and white matter loss along the cingulum in AD compared to NEC. Metabolic and microstructural changes were associated with episodic memory performance assessed using Craft Story 21 Recall and Benson Complex Figure Copy. The current study suggests that metabolite concentrations measured using 1H-MRS may provide insight into the underlying metabolic and microstructural processes of episodic memory impairment.
Collapse
Affiliation(s)
- Dickson Wong
- Department of Medical Biophysics, University of Western Ontario, London, ON, Canada
| | - Samir Atiya
- Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Jennifer Fogarty
- Parkwood Institute Research Program, Lawson Health Research Institute, London, ON, Canada
| | - Manuel Montero-Odasso
- Parkwood Institute Research Program, Lawson Health Research Institute, London, ON, Canada.,Geriatric Medicine, University of Western Ontario, London, ON, Canada.,Gait and Brain Lab, Parkwood Institute, Lawson Health Research Institute, London, ON, Canada.,Department of Epidemiology and Biostatistics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Stephen H Pasternak
- Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.,Parkwood Institute Research Program, Lawson Health Research Institute, London, ON, Canada
| | - Chris Brymer
- Geriatric Medicine, University of Western Ontario, London, ON, Canada
| | - Michael J Borrie
- Parkwood Institute Research Program, Lawson Health Research Institute, London, ON, Canada.,Geriatric Medicine, University of Western Ontario, London, ON, Canada
| | - Robert Bartha
- Department of Medical Biophysics, University of Western Ontario, London, ON, Canada.,Robarts Research Institute, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| |
Collapse
|
25
|
Zhao L, Teng J, Mai W, Su J, Yu B, Nong X, Li C, Wei Y, Duan G, Deng X, Deng D, Chen S. A Pilot Study on the Cutoff Value of Related Brain Metabolite in Chinese Elderly Patients With Mild Cognitive Impairment Using MRS. Front Aging Neurosci 2021; 13:617611. [PMID: 33897404 PMCID: PMC8063036 DOI: 10.3389/fnagi.2021.617611] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 02/12/2021] [Indexed: 11/27/2022] Open
Abstract
Objective: This cross-sectional study aimed to distinguish patients with mild cognitive impairment (MCI) from patients with normal controls (NCs) by measuring the levels of N-acetyl aspartate (NAA), total creatinine (tCr), and choline (Cho) in their hippocampus (HIP) and their posterior cingulate gyrus (PCG) by using proton magnetic resonance spectroscopy (MRS) and to predict the cutoff value on the ratios of metabolites. We further aimed to provide a reference for the diagnosis of MCI in elderly patients in China. Methods: About 69 patients who underwent a clinical diagnosis of the MCI group and 67 patients with NCs, the Mini-Mental Status Examination (MMSE) score, the Montreal Cognitive Assessment (MoCA) score, and MRS of the bilateral HIP and bilateral PCG were considered. The ratio of NAA/tCr and Cho/tCr in the bilateral HIP and bilateral PCG was calculated. The relationship between the ratios of metabolites and the scores of MMSE and MoCA was analyzed, and the possible brain metabolite cutoff point for the diagnosis of MCI was evaluated. Results: Compared with the NC group, the scores of MMSE and MoCA in the MCI group decreased significantly (p < 0.05); the ratio of NAA/tCr in the bilateral HIP and bilateral PCG and the ratio of Cho/tCr at the right HIP in the MCI group decreased significantly (p < 0.05); however, there was no significant difference in the ratio of Cho/tCr in the left HIP and bilateral PCG between the two groups (p > 0.05). The correlation coefficient between MMSE/MoCA and the ratio of NAA/tCr was 0.49–0.56 in the bilateral HIP (p < 0.01). The best cutoff value of NAA/creatine (Cr) in the left HIP and the right HIP was 1.195 and 1.19. Sensitivity, specificity, and the Youden index (YDI) in the left HIP and the right HIP were (0.725, 0.803, 0.528) and (0.754, 0.803, 0.557), respectively. Conclusion: The level of metabolites in the HIP and the PCG of patients with MCI and of those with normal subjects has a certain correlation with the score of their MMSE and MoCA. When the value of NAA/tCr in the left HIP and right HIP is <1.19, it suggests that MCI may have occurred. According to this cutoff point, elderly patients with MCI in China could be screened.
Collapse
Affiliation(s)
- Lihua Zhao
- Department of Acupuncture, First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, China
| | - Jinlong Teng
- Department of Acupuncture, First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, China
| | - Wei Mai
- Department of Acupuncture, First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, China
| | - Jiahui Su
- Department of Acupuncture, First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, China
| | - Bihan Yu
- Department of Acupuncture, First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, China
| | - Xiucheng Nong
- Department of Acupuncture, First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, China
| | - Chong Li
- Department of Acupuncture, First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, China
| | - Yichen Wei
- Department of Radiology, First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, China
| | - Gaoxiong Duan
- Department of Radiology, First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, China
| | - Xiangming Deng
- Department of Acupuncture, First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, China
| | - Demao Deng
- Department of Radiology, First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, China
| | - Shangjie Chen
- Department of Rehabilitation, Bao'an Hospital, Southern Medical University, Shenzhen, China
| |
Collapse
|
26
|
Li Y, Wang Z, Sun R, Lam F. Separation of Metabolites and Macromolecules for Short-TE 1H-MRSI Using Learned Component-Specific Representations. IEEE TRANSACTIONS ON MEDICAL IMAGING 2021; 40:1157-1167. [PMID: 33395390 PMCID: PMC8049099 DOI: 10.1109/tmi.2020.3048933] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Short-echo-time (TE) proton magnetic resonance spectroscopic imaging (MRSI) allows for simultaneously mapping a number of molecules in the brain, and has been recognized as an important tool for studying in vivo biochemistry in various neuroscience and disease applications. However, separation of the metabolite and macromolecule (MM) signals present in the short-TE data with significant spectral overlaps remains a major technical challenge. This work introduces a new approach to solve this problem by integrating imaging physics and representation learning. Specifically, a mixed unsupervised and supervised learning-based strategy was developed to learn the metabolite and MM-specific low-dimensional representations using deep autoencoders. A constrained reconstruction formulation is proposed to integrate the MRSI spatiospectral encoding model and the learned representations as effective constraints for signal separation. An efficient algorithm was developed to solve the resulting optimization problem with provable convergence. Simulation and experimental results have been obtained to demonstrate the component-specific representation power of the learned models and the capability of the proposed method in separating metabolite and MM signals for practical short-TE [Formula: see text]-MRSI data.
Collapse
|
27
|
Early Detection of Radiation-Induced Injury and Prediction of Cognitive Deficit by MRS Metabolites in Radiotherapy of Low-Grade Glioma. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6616992. [PMID: 34258272 PMCID: PMC8260313 DOI: 10.1155/2021/6616992] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/04/2021] [Accepted: 02/21/2021] [Indexed: 12/18/2022]
Abstract
Purpose To compare the sensitivity of MRS metabolites and MoCA and ACE-R cognitive tests in the detection of radiation-induced injury in low grade glioma (LGG) patients in early and early delayed postradiation stages. Methods MRS metabolite ratios of NAA/Cr and Cho/Cr, ACE-R and MoCA cognitive tests, and dosimetric parameters in corpus callosum were analyzed during RT and up to 6-month post-RT for ten LGG patients. Results Compared to pre RT baseline, a significant decline in both NAA/Cr and Cho/Cr in the corpus callosum was seen at the 4th week of RT, 1, 3, and 6-month post-RT. These declines were detected at least 3 months before the detection of declines in cognitive functions by ACE-R and MoCA tools. Moreover, NAA/Cr alterations at 4th week of RT and 1-month post-RT were significantly negatively correlated with the mean dose received by the corpus callosum, as well as the corpus callosum 40 Gy dose volume, i.e., the volume of the corpus callosum receiving a dose greater than 40 Gy. Conclusion MRS-based biomarkers may be more sensitive than the state-of-the-art cognitive tests in the prediction of postradiation cognitive impairments. They would be utilized in treatment planning and dose sparing protocols, with a specific focus on the corpus callosum in the radiation therapy of LGG patients.
Collapse
|
28
|
Mitochondrial Dysfunction in Alzheimer's Disease: A Biomarker of the Future? Biomedicines 2021; 9:biomedicines9010063. [PMID: 33440662 PMCID: PMC7827030 DOI: 10.3390/biomedicines9010063] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/05/2021] [Accepted: 01/07/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia worldwide and is characterised pathologically by the accumulation of amyloid beta and tau protein aggregates. Currently, there are no approved disease modifying therapies for clearance of either of these proteins from the brain of people with AD. As well as abnormalities in protein aggregation, other pathological changes are seen in this condition. The function of mitochondria in both the nervous system and rest of the body is altered early in this disease, and both amyloid and tau have detrimental effects on mitochondrial function. In this review article, we describe how the function and structure of mitochondria change in AD. This review summarises current imaging techniques that use surrogate markers of mitochondrial function in both research and clinical practice, but also how mitochondrial functions such as ATP production, calcium homeostasis, mitophagy and reactive oxygen species production are affected in AD mitochondria. The evidence reviewed suggests that the measurement of mitochondrial function may be developed into a future biomarker for early AD. Further work with larger cohorts of patients is needed before mitochondrial functional biomarkers are ready for clinical use.
Collapse
|
29
|
Lepping RJ, Montgomery RN, Sharma P, Mahnken JD, Vidoni ED, Choi IY, Sarnak MJ, Brooks WM, Burns JM, Gupta A. Normalization of Cerebral Blood Flow, Neurochemicals, and White Matter Integrity after Kidney Transplantation. J Am Soc Nephrol 2021; 32:177-187. [PMID: 33067382 PMCID: PMC7894653 DOI: 10.1681/asn.2020050584] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 09/06/2020] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND CKD is associated with abnormalities in cerebral blood flow, cerebral neurochemical concentrations, and white matter integrity. Each of these is associated with adverse clinical consequences in the non-CKD population, which may explain the high prevalence of dementia and stroke in ESKD. Because cognition improves after kidney transplantation, comparing these brain abnormalities before and after kidney transplantation may identify potential reversibility in ESKD-associated brain abnormalities. METHODS In this study of patients with ESKD and age-matched healthy controls, we used arterial spin labeling to assess the effects of kidney transplantation on cerebral blood flow and magnetic resonance spectroscopic imaging to measure cerebral neurochemical concentrations (N-acetylaspartate, choline, glutamate, glutamine, myo-inositol, and total creatine). We also assessed white matter integrity measured by fractional anisotropy (FA) and mean diffusivity (MD) with diffusion tensor imaging. We used a linear mixed model analysis to compare longitudinal, repeated brain magnetic resonance imaging measurements before, 3 months after, and 12 months after transplantation and compared these findings with those of healthy controls. RESULTS Study participants included 29 patients with ESKD and 19 controls; 22 patients completed post-transplant magnetic resonance imaging. Cerebral blood flow, which was higher in patients pretransplant compared with controls (P=0.003), decreased post-transplant (P<0.001) to values in controls. Concentrations of neurochemicals choline and myo-inositol that were higher pretransplant compared with controls (P=0.001 and P<0.001, respectively) also normalized post-transplant (P<0.001 and P<0.001, respectively). FA increased (P=0.001) and MD decreased (P<0.001) post-transplant. CONCLUSIONS Certain brain abnormalities in CKD are reversible and normalize with kidney transplantation. Further studies are needed to understand the mechanisms underlying these brain abnormalities and to explore interventions to mitigate them even in patients who cannot be transplanted. CLINICAL TRIAL REGISTRY NAME AND REGISTRATION NUMBER Cognitive Impairment and Imaging Correlates in End Stage Renal Disease, NCT01883349.
Collapse
Affiliation(s)
- Rebecca J. Lepping
- Hoglund Biomedical Imaging Center, Kansas City, Kansas,University of Kansas Alzheimer’s Disease Center, Fairway, Kansas
| | - Robert N. Montgomery
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas
| | - Palash Sharma
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas
| | - Jonathan D. Mahnken
- University of Kansas Alzheimer’s Disease Center, Fairway, Kansas,Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas
| | - Eric D. Vidoni
- University of Kansas Alzheimer’s Disease Center, Fairway, Kansas,Department of Neurology, University of Kansas Medical Center, Kansas City, Kansas
| | - In-Young Choi
- Hoglund Biomedical Imaging Center, Kansas City, Kansas,Department of Neurology, University of Kansas Medical Center, Kansas City, Kansas
| | - Mark J. Sarnak
- Division of Nephrology and Hypertension, Department of Internal Medicine, Tufts Medical Center, Boston, Massachusetts
| | - William M. Brooks
- Hoglund Biomedical Imaging Center, Kansas City, Kansas,University of Kansas Alzheimer’s Disease Center, Fairway, Kansas,Department of Neurology, University of Kansas Medical Center, Kansas City, Kansas,Frontiers: University of Kanas Clinical and Translational Science Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Jeffrey M. Burns
- University of Kansas Alzheimer’s Disease Center, Fairway, Kansas,Department of Neurology, University of Kansas Medical Center, Kansas City, Kansas,Frontiers: University of Kanas Clinical and Translational Science Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Aditi Gupta
- University of Kansas Alzheimer’s Disease Center, Fairway, Kansas,Division of Nephrology and Hypertension, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas,The Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|
30
|
Tang L, Zhao Y, Li Y, Guo R, Clifford B, El Fakhri G, Ma C, Liang ZP, Luo J. Accelerated J-resolved 1 H-MRSI with limited and sparse sampling of ( k , t 1 , t 2 -space. Magn Reson Med 2020; 85:30-41. [PMID: 32726510 DOI: 10.1002/mrm.28413] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 06/13/2020] [Accepted: 06/15/2020] [Indexed: 01/18/2023]
Abstract
PURPOSE To accelerate the acquisition of J-resolved proton magnetic resonance spectroscopic imaging (1 H-MRSI) data for high-resolution mapping of brain metabolites and neurotransmitters. METHODS The proposed method used a subspace model to represent multidimensional spatiospectral functions, which significantly reduced the number of parameters to be determined from J-resolved 1 H-MRSI data. A semi-LASER-based (Localization by Adiabatic SElective Refocusing) echo-planar spectroscopic imaging (EPSI) sequence was used for data acquisition. The proposed data acquisition scheme sampled k , t 1 , t 2 -space in variable density, where t1 and t2 specify the J-coupling and chemical-shift encoding times, respectively. Selection of the J-coupling encoding times (or, echo time values) was based on a Cramer-Rao lower bound analysis, which were optimized for gamma-aminobutyric acid (GABA) detection. In image reconstruction, parameters of the subspace-based spatiospectral model were determined by solving a constrained optimization problem. RESULTS Feasibility of the proposed method was evaluated using both simulated and experimental data from a spectroscopic phantom. The phantom experimental results showed that the proposed method, with a factor of 12 acceleration in data acquisition, could determine the distribution of J-coupled molecules with expected accuracy. In vivo study with healthy human subjects also showed that 3D maps of brain metabolites and neurotransmitters can be obtained with a nominal spatial resolution of 3.0 × 3.0 × 4.8 mm3 from J-resolved 1 H-MRSI data acquired in 19.4 min. CONCLUSIONS This work demonstrated the feasibility of highly accelerated J-resolved 1 H-MRSI using limited and sparse sampling of k , t 1 , t 2 -space and subspace modeling. With further development, the proposed method may enable high-resolution mapping of brain metabolites and neurotransmitters in clinical applications.
Collapse
Affiliation(s)
- Lihong Tang
- Institute for Medical Imaging Technology, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yibo Zhao
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA.,Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Yudu Li
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA.,Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Rong Guo
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA.,Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Bryan Clifford
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA.,Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Georges El Fakhri
- Department of Radiology, Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Chao Ma
- Department of Radiology, Gordon Center for Medical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Zhi-Pei Liang
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA.,Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Jie Luo
- Institute for Medical Imaging Technology, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
31
|
Frontal lobe metabolic alterations characterizing Parkinson's disease cognitive impairment. Neurol Sci 2020; 42:1053-1064. [PMID: 32729012 DOI: 10.1007/s10072-020-04626-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/21/2020] [Indexed: 10/23/2022]
Abstract
BACKGROUND AND PURPOSE Diagnosis of Parkinson's disease (PD) cognitive impairment at early stages is challenging compared to the stage of PD dementia where functional impairment is apparent and easily diagnosed. Hence, to evaluate potential early stage cognitive biomarkers, we assessed frontal lobe metabolic alterations using in vivo multi-voxel proton magnetic resonance spectroscopic imaging (1H-MRSI). METHOD Frontal metabolism was studied in patients with PD with normal cognition (PD-CN) (n = 26), with cognitive impairment (PD-CI) (n = 27), and healthy controls (HC) (n = 30) using a single slice (two-dimensional) 1H-MRSI at 3 T. The acquired spectra were post-processed distinctly for voxels corresponding to the bilateral middle/superior frontal gray matter (GM) and frontal white matter (WM) regions (delineated employing neuromorphometrics atlas) using the LC-Model software. RESULT Significant (post hoc p < 0.016) reduction in the concentration of N-acetyl aspartate (NAA) in the middle and superior frontal GMs and total choline (tCho) and total creatine (tCr) in the frontal WM was observed in PD-CI compared to PD-CN and HC, while that in HC and PD-CN groups were comparable. The NAA and tCr/tCho metabolite concentrations showed significant (p < 0.05) positive correlations with cognitive test scores in the frontal GM and WM, respectively. The receiver operating curve (ROC) analysis revealed significant (p < 0.05) "area under curve" for NAA/tNAA in the frontal GM and tCho in the frontal WM. CONCLUSION The frontal metabolic profile is altered in cognitively impaired PD compared with cognitively normal PD. Neuronal function loss (NAA), altered energy metabolism (Cr), and cholinergic (Cho) neural transmission are implicated in PD cognitive pathology. Frontal neuro-metabolism may promisingly serve as PD cognitive biomarker.
Collapse
|
32
|
Zyśk M, Sakowicz-Burkiewicz M, Pikul P, Kowalski R, Michno A, Pawełczyk T. The Impact of Acetyl-CoA and Aspartate Shortages on the N-Acetylaspartate Level in Different Models of Cholinergic Neurons. Antioxidants (Basel) 2020; 9:antiox9060522. [PMID: 32545833 PMCID: PMC7346116 DOI: 10.3390/antiox9060522] [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: 05/09/2020] [Revised: 06/03/2020] [Accepted: 06/11/2020] [Indexed: 12/31/2022] Open
Abstract
N-acetylaspartate is produced by neuronal aspartate N-acetyltransferase (NAT8L) from acetyl-CoA and aspartate. In cholinergic neurons, acetyl-CoA is also utilized in the mitochondrial tricarboxylic acid cycle and in acetylcholine production pathways. While aspartate has to be shared with the malate–aspartate shuttle, another mitochondrial machinery together with the tricarboxylic acid cycle supports the electron transport chain turnover. The main goal of this study was to establish the impact of toxic conditions on N-acetylaspartate production. SN56 cholinergic cells were exposed to either Zn2+ overload or Ca2+ homeostasis dysregulation and male adult Wistar rats’ brains were studied after 2 weeks of challenge with streptozotocin-induced hyperglycemia or daily theophylline treatment. Our results allow us to hypothesize that the cholinergic neurons from brain septum prioritized the acetylcholine over N-acetylaspartate production. This report provides the first direct evidence for Zn2+-dependent suppression of N-acetylaspartate synthesis leading to mitochondrial acetyl-CoA and aspartate shortages. Furthermore, Zn2+ is a direct concentration-dependent inhibitor of NAT8L activity, while Zn2+-triggered oxidative stress is unlikely to be significant in such suppression.
Collapse
Affiliation(s)
- Marlena Zyśk
- Department of Molecular Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (M.S.-B.); (T.P.)
- Correspondence: ; Tel.: +48-5834-927-70
| | - Monika Sakowicz-Burkiewicz
- Department of Molecular Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (M.S.-B.); (T.P.)
| | - Piotr Pikul
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Center, Polish Academy of Science, 80-308 Gdansk, Poland;
- Clinical Laboratory University Clinical Center in Gdansk, 80-211 Gdansk, Poland;
| | - Robert Kowalski
- Clinical Laboratory University Clinical Center in Gdansk, 80-211 Gdansk, Poland;
| | - Anna Michno
- Department of Laboratory Medicine, Medical University of Gdansk, 80-2011 Gdansk, Poland;
| | - Tadeusz Pawełczyk
- Department of Molecular Medicine, Medical University of Gdansk, 80-211 Gdansk, Poland; (M.S.-B.); (T.P.)
| |
Collapse
|
33
|
Oglesby RT, Lam WW, Stanisz GJ. In vitro characterization of the serotonin biosynthesis pathway by CEST MRI. Magn Reson Med 2020; 84:2389-2399. [DOI: 10.1002/mrm.28281] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 03/09/2020] [Accepted: 03/23/2020] [Indexed: 12/20/2022]
Affiliation(s)
- Ryan T. Oglesby
- Physical Sciences Sunnybrook Research Institute Toronto ON Canada
- Medical Biophysics University of Toronto Toronto ON Canada
| | - Wilfred W. Lam
- Physical Sciences Sunnybrook Research Institute Toronto ON Canada
| | - Greg J. Stanisz
- Physical Sciences Sunnybrook Research Institute Toronto ON Canada
- Medical Biophysics University of Toronto Toronto ON Canada
- Neurosurgery and Pediatric Neurosurgery Medical University of Lublin Lublin Poland
| |
Collapse
|
34
|
Chen Q, Kantarci K. Imaging Biomarkers for Neurodegeneration in Presymptomatic Familial Frontotemporal Lobar Degeneration. Front Neurol 2020; 11:80. [PMID: 32184751 PMCID: PMC7058699 DOI: 10.3389/fneur.2020.00080] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/22/2020] [Indexed: 02/05/2023] Open
Abstract
Frontotemporal lobar degeneration (FTLD) is a neurodegenerative disorder characterized by behavioral changes, language abnormality, as well as executive function deficits and motor impairment. In about 30-50% of FTLD patients, an autosomal dominant pattern of inheritance was found with major mutations in the MAPT, GRN, and the C9orf72 repeat expansion. These mutations could lead to neurodegenerative pathology years before clinical symptoms onset. With potential disease-modifying treatments that are under development, non-invasive biomarkers that help determine the early brain changes in presymptomatic FTLD patients will be critical for tracking disease progression and enrolling the right participants into the clinical trials at the right time during the disease course. In recent years, there is increasing evidence that a number of imaging biomarkers show the abnormalities during the presymptomatic stage. Imaging biomarkers of presymptomatic familial FTLD may provide insight into the underlying neurodegenerative process years before symptom onset. Structural magnetic resonance imaging (MRI) has demonstrated cortical degeneration with a mutation-specific neurodegeneration pattern years before onset of clinical symptoms in presymptomatic familial FTLD mutation carriers. In addition, diffusion tensor imaging (DTI) has shown the loss of white matter microstructural integrity in the presymptomatic stage of familial FTLD. Furthermore, proton magnetic resonance spectroscopy (1H MRS), which provides a non-invasive measurement of brain biochemistry, has identified early neurochemical abnormalities in presymptomatic MAPT mutation carriers. Positron emission tomography (PET) imaging with [18F]-fluorodeoxyglucose (FDG) has demonstrated the glucose hypometabolism in the presymptomatic stage of familial FTLD. Also, a novel PET ligand, 18F-AV-1451, has been used in this group to evaluate tau deposition in the brain. Promising imaging biomarkers for presymptomatic familial FTLD have been identified and assessed for specificity and sensitivity for accurate prediction of symptom onset and tracking disease progression during the presymptomatic stage when clinical measures are not useful. Furthermore, identifying imaging biomarkers for the presymptomatic stage is important for the design of disease-modifying trials. We review the recent progress in imaging biomarkers of the presymptomatic phase of familial FTLD and discuss the imaging techniques and analysis methods, with a focus on the potential implication of these imaging techniques and their utility in specific mutation types.
Collapse
Affiliation(s)
- Qin Chen
- Department of Neurology, West China Hospital of Sichuan University, Chengdu, China.,Department of Radiology, Mayo Clinic, Rochester, MN, United States
| | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
35
|
Conway ME. Alzheimer's disease: targeting the glutamatergic system. Biogerontology 2020; 21:257-274. [PMID: 32048098 PMCID: PMC7196085 DOI: 10.1007/s10522-020-09860-4] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Accepted: 01/29/2020] [Indexed: 12/21/2022]
Abstract
Alzheimer’s disease (AD) is a debilitating neurodegenerative disease that causes a progressive decline in memory, language and problem solving. For decades mechanism-based therapies have primarily focused on amyloid β (Aβ) processing and pathways that govern neurofibrillary tangle generation. With the potential exception to Aducanumab, a monotherapy to target Aβ, clinical trials in these areas have been challenging and have failed to demonstrate efficacy. Currently, the prescribed therapies for AD are those that target the cholinesterase and glutamatergic systems that can moderately reduce cognitive decline, dependent on the individual. In the brain, over 40% of neuronal synapses are glutamatergic, where the glutamate level is tightly regulated through metabolite exchange in neuronal, astrocytic and endothelial cells. In AD brain, Aβ can interrupt effective glutamate uptake by astrocytes, which evokes a cascade of events that leads to neuronal swelling, destruction of membrane integrity and ultimately cell death. Much work has focussed on the post-synaptic response with little insight into how glutamate is regulated more broadly in the brain and the influence of anaplerotic pathways that finely tune these mechanisms. The role of blood branched chain amino acids (BCAA) in regulating neurotransmitter profiles under disease conditions also warrant discussion. Here, we review the importance of the branched chain aminotransferase proteins in regulating brain glutamate and the potential consequence of dysregulated metabolism in the context of BCAA or glutamate accumulation. We explore how the reported benefits of BCAA supplementation or restriction in improving cognitive function in other neurological diseases may have potential application in AD. Given that memantine, the glutamate receptor agonist, shows clinical relevance it is now timely to research related pathways, an understanding of which could identify novel approaches to treatment of AD.
Collapse
Affiliation(s)
- Myra E Conway
- Faculty of Health and Applied Sciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK. .,Faculty of Health and Life Sciences, University of the West of England, Coldharbour Lane, Bristol, BS16 1QY, UK.
| |
Collapse
|
36
|
|
37
|
Maul S, Giegling I, Rujescu D. Proton Magnetic Resonance Spectroscopy in Common Dementias-Current Status and Perspectives. Front Psychiatry 2020; 11:769. [PMID: 32848938 PMCID: PMC7424040 DOI: 10.3389/fpsyt.2020.00769] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/20/2020] [Indexed: 12/13/2022] Open
Abstract
Dementia occurs mainly in the elderly and is associated with cognitive decline and impairment of activities of daily living. The most common forms of dementia are Alzheimer's disease (AD), vascular dementia (VD), dementia with Lewy bodies (DLB), and frontotemporal dementia (FTD). To date, there are no causal options for therapy, but drug and non-drug treatments can positively modulate the course of the disease. Valid biomarkers are needed for the earliest possible and reliable diagnosis, but so far, such biomarkers have only been established for AD and require invasive and expensive procedures. In this context, proton magnetic resonance spectroscopy (1H-MRS) provides a non-invasive and widely available technique for investigating the biochemical milieu of brain tissue in vivo. Numerous studies have been conducted for AD, but for VD, DLB, and FTD the number of studies is limited. Nevertheless, MRS can detect measurable metabolic alterations in common dementias. However, most of the studies conducted are too heterogeneous to assess the potential use of MRS technology in clinical applications. In the future, technological advances may increase the value of MRS in dementia diagnosis and treatment. This review summarizes the results of MRS studies conducted in common dementias and discusses the reasons for the lack of transfer into clinical routine.
Collapse
Affiliation(s)
- Stephan Maul
- University Clinic and Outpatient Clinic for Psychiatry, Psychotherapy and Psychosomatics, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Ina Giegling
- University Clinic and Outpatient Clinic for Psychiatry, Psychotherapy and Psychosomatics, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Dan Rujescu
- University Clinic and Outpatient Clinic for Psychiatry, Psychotherapy and Psychosomatics, Martin Luther University Halle-Wittenberg, Halle, Germany
| |
Collapse
|
38
|
Chen Y, Li Y, Xu Z. Improved Low-Rank Filtering of MR Spectroscopic Imaging Data With Pre-Learnt Subspace and Spatial Constraints. IEEE Trans Biomed Eng 2019; 67:2381-2388. [PMID: 31870975 DOI: 10.1109/tbme.2019.2961698] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE To investigate the use of pre-learnt subspace and spatial constraints for denoising magnetic resonance spectroscopic imaging (MRSI) data. METHOD We exploit the partial separability or subspace structures of high-dimensional MRSI data for denoising. More specifically, we incorporate a subspace model with pre-learnt spectral basis into the low-rank approximation (LORA) method. Spectral basis is determined based on empirical prior distributions of the spectral parameters variations learnt from auxiliary training data; spatial priors are also incorporated as is done in LORA to further improve denoising performance. RESULTS The effects of the explicit subspace and spatial constraints in reducing estimation bias and variance have been analyzed using Cramér-Rao Lower bound analysis, Monte-Carlo study, and experimental study. CONCLUSION The denoising effectiveness of LORA can be significantly improved by incorporating pre-learnt spectral basis and spatial priors into LORA. SIGNIFICANCE This study provides an effective method for denoising MRSI data along with comprehensive analyses of its performance. The proposed method is expected to be useful for a wide range of studies using MRSI.
Collapse
|
39
|
Chen Q, Boeve BF, Tosakulwong N, Lesnick T, Brushaber D, Dheel C, Fields J, Forsberg L, Gavrilova R, Gearhart D, Haley D, Gunter JL, Graff-Radford J, Jones D, Knopman D, Graff-Radford N, Kraft R, Lapid M, Rademakers R, Syrjanen J, Wszolek ZK, Rosen H, Boxer AL, Kantarci K. Frontal lobe 1H MR spectroscopy in asymptomatic and symptomatic MAPT mutation carriers. Neurology 2019; 93:e758-e765. [PMID: 31315971 DOI: 10.1212/wnl.0000000000007961] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 03/26/2019] [Indexed: 02/05/2023] Open
Abstract
OBJECTIVE To determine the frontal lobe proton magnetic resonance spectroscopy (1H MRS) abnormalities in asymptomatic and symptomatic carriers of microtubule-associated protein tau (MAPT) mutations. METHODS We recruited patients with MAPT mutations from 5 individual families, who underwent single voxel 1H MRS from the medial frontal lobe at 3T (n = 19) from the Longitudinal Evaluation of Familial Frontotemporal Dementia Subjects (LEFFTDS) Study at the Mayo Clinic site. Asymptomatic MAPT mutation carriers (n = 9) had Frontotemporal Lobar Degeneration Clinical Dementia Rating Sum of Boxes (FTLD-CDR SOB) score of zero, and symptomatic MAPT mutation carriers (n = 10) had a median FTLD-CDR SOB score of 5. Noncarriers from healthy first-degree relatives of the patients were recruited as controls (n = 25). The demographic aspects and 1H MRS metabolite ratios were compared by use of the Fisher exact test for sex and linear mixed models to account for within-family correlations. We used Tukey contrasts for pair-wise comparisons. RESULTS Asymptomatic MAPT mutation carriers had lower neuronal marker N-acetylaspartate (NAA)/creatine (Cr) (p = 0.001) and lower NAA/myo-inositol (mI) (p = 0.026) than noncarriers after adjustment for age. Symptomatic MAPT mutation carriers had lower NAA/Cr (p = 0.01) and NAA/mI (p = 0.01) and higher mI/Cr (p = 0.02) compared to noncarriers after adjustment for age. Furthermore, NAA/Cr (p = 0.006) and NAA/mI (p < 0.001) ratios decreased, accompanied by an increase in mI/Cr ratio (p = 0.001), as the ages of carriers approached and passed the age at symptom onset. CONCLUSION Frontal lobe neurochemical alterations measured with 1H MRS precede the symptom onset in MAPT mutation carriers. Frontal lobe 1H MRS is a potential biomarker for early neurodegenerative processes in MAPT mutation carriers.
Collapse
Affiliation(s)
- Qin Chen
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Bradley F Boeve
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Nirubol Tosakulwong
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Timothy Lesnick
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Danielle Brushaber
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Christina Dheel
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Julie Fields
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Leah Forsberg
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Ralitza Gavrilova
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Debra Gearhart
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Dana Haley
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Jeffrey L Gunter
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Jonathan Graff-Radford
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - David Jones
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - David Knopman
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Neill Graff-Radford
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Ruth Kraft
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Maria Lapid
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Rosa Rademakers
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Jeremy Syrjanen
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Zbigniew K Wszolek
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Howie Rosen
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Adam L Boxer
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco
| | - Kejal Kantarci
- From the Department of Radiology (Q.C., J.L.G., K.K.), Department of Neurology (B.F.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K.), Department of Health Sciences Research (N.T., T.L., D.B., J.S.), Department of Psychology and Psychiatry (J.F., M.L.), Department of Clinical Genomic and Neurology (R.G.), Alzheimer's Disease Research Center (B.F.B., D.B., C.D., L.F., D.G., J.G.-R., D.J., D.K., R.K., R.R., K.K.), and Research Services (D.H.), Mayo Clinic, Rochester, MN; Department of Neurology (Q.C.), West China Hospital of Sichuan University, Chengdu, Sichuan; Departments of Neurology (N.G.-R., Z.K.W.) and Neuroscience (R.R.), Mayo Clinic, Jacksonville, FL; and Memory and Aging Center (H.R., A.L.B.), University of California San Francisco.
| |
Collapse
|
40
|
Zhang Y, Liu Z, Ji B, Liu L, Wu S, Liu X, Wang S, Wang L. Metabolite Profile of Alzheimer's Disease in the Frontal Cortex as Analyzed by HRMAS 1H NMR. Front Aging Neurosci 2019; 10:424. [PMID: 30687076 PMCID: PMC6333733 DOI: 10.3389/fnagi.2018.00424] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 12/06/2018] [Indexed: 12/14/2022] Open
Abstract
Background: Investigation on neurochemical changes in the frontal cortex in individuals with Alzheimer’s disease (AD) and different Apolipoprotein E (APOE) genotypes, using ex vivo solid-state high-resolution NMR analysis, may lead to a better understanding of the neurochemistry associated with AD as well as new AD-specific metabolite biomarkers that might potentially improve the clinical diagnosis of AD. Methods: Intact tissue samples of the frontal cortex were obtained from 11 patients and 11 age-matched non-demented controls. Metabolite profiles in all samples were analyzed ex vivo, using solid-state high-resolution magic angle spinning (HRMAS) 600 MHz 1H nuclear magnetic resonance (NMR). A logistic regression analysis was used to rank metabolites based on their level of contribution in differentiating the AD patient tissues and the controls, and different AD-associated APOE genotypes (APOE ε4 vs. APOE ε3). Results: Tissue samples from the AD patients showed significantly lower NAA/Cr (p = 0.011), Ace/Cr (p = 0.027), GABA/Cr (p = 0.005), Asp/Cr (p < 0.0001), mI/Cr (p < 0.0001), and Tau/Cr (p = 0.021), and higher PCho/Cr (p < 0.0001), GPCho/Cr (p < 0.0001), and α&β-Glc/Cr (p < 0.0001) than the controls did. Specifically, a newly observed resonance at 3.71 ppm, referred to as α&β-Glc, was observed in 90.9% of the AD samples (10/11). Samples with APOE ε4 also exhibited higher PCho/Cr (p = 0.0002), GPCho/Cr (p = 0.0001), α&β-Glc/Cr (p < 0.0001), and lower Asp/Cr (p = 0.004) and GABA/Cr (p = 0.04) than the samples with APOE ε3 did. In the logistic regression analysis, PCho, GPCho, ASP, and α&β-Glc were found to be the most relevant metabolites for differentiating the AD patient tissues and the controls, and different APOE genotypes. Conclusion: HRMAS 1H NMR with high spectral resolution and sensitivity offers a powerful tool to gain quantitative information on AD associated neurochemical changes. There are important neurochemical differences in the frontal cortex between the AD patient tissues and the controls, and between those with different APOE genotypes. The resonance (α&β-Glc) found at 3.71 ppm in the AD patient tissues may be further investigated for its potential in the diagnosis and monitoring of AD.
Collapse
Affiliation(s)
- Yuzhong Zhang
- Department of Radiology, The People's Hospital of Longhua, Shenzhen, China
| | - Zhou Liu
- Graduate School, Medical College of Nanchang University, Nanchang, China.,Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, United States
| | - Bing Ji
- Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, United States
| | - Lijian Liu
- Graduate School, Medical College of Nanchang University, Nanchang, China
| | - Shaoxiong Wu
- Department of Chemistry, NMR Research Center, Emory University, Atlanta, GA, United States
| | - Xiaowu Liu
- Yiwei Medical Technology, Inc., Shenzhen, China
| | - Silun Wang
- Yiwei Medical Technology, Inc., Shenzhen, China
| | - Liya Wang
- Graduate School, Medical College of Nanchang University, Nanchang, China.,Department of Radiology and Imaging Sciences, Emory University School of Medicine, Atlanta, GA, United States
| |
Collapse
|
41
|
Balážová Z, Nováková M, Minsterová A, Rektorová I. Structural and Functional Magnetic Resonance Imaging of Dementia With Lewy Bodies. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2018; 144:95-141. [PMID: 30638458 DOI: 10.1016/bs.irn.2018.10.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Dementia with Lewy bodies (DLB) is the second most common cause of neurodegenerative dementia after Alzheimer's disease (AD). Although diagnosis may be challenging, there is increasing evidence that the use of biomarkers according to 2017 revised criteria for diagnosis and management of dementia with Lewy bodies can increase diagnostic accuracy. Apart from nuclear medicine techniques, various magnetic resonance imaging (MRI) techniques have been utilized in attempt to enhance diagnostic accuracy. This chapter reviews structural, functional and diffusion MRI studies in DLB cohorts being compared to healthy controls, AD or dementia in Parkinson's disease (PDD). We also included relatively new MRI methods that may have potential to identify early DLB subjects and aim at examining brain iron and neuromelanin.
Collapse
Affiliation(s)
- Zuzana Balážová
- Applied Neuroscience Research Group, Central European Institute of Technology, CEITEC MU, Masaryk University, Brno, Czech Republic; Department of Radiology and Nuclear Medicine, University Hospital Brno, Faculty of Medicine, Brno, Czech Republic
| | - Marie Nováková
- Applied Neuroscience Research Group, Central European Institute of Technology, CEITEC MU, Masaryk University, Brno, Czech Republic
| | - Alžběta Minsterová
- Applied Neuroscience Research Group, Central European Institute of Technology, CEITEC MU, Masaryk University, Brno, Czech Republic
| | - Irena Rektorová
- Applied Neuroscience Research Group, Central European Institute of Technology, CEITEC MU, Masaryk University, Brno, Czech Republic; St. Anne's University Hospital and Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| |
Collapse
|
42
|
Nanga RPR, DeBrosse C, Kumar D, Roalf D, McGeehan B, D'Aquilla K, Borthakur A, Hariharan H, Reddy D, Elliott M, Detre JA, Epperson CN, Reddy R. Reproducibility of 2D GluCEST in healthy human volunteers at 7 T. Magn Reson Med 2018; 80:2033-2039. [PMID: 29802635 PMCID: PMC6107408 DOI: 10.1002/mrm.27362] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 04/11/2018] [Accepted: 04/24/2018] [Indexed: 01/06/2023]
Abstract
PURPOSE To investigate the reproducibility of gray and white matter glutamate contrast of a brain slice among a small group of healthy volunteers by using the 2D single-slice glutamate CEST (GluCEST) imaging technique. METHODS Six healthy volunteers were scanned multiple times for within-day and between-day reproducibility. One more volunteer was scanned for within-day reproducibility at 7T MRI. Glutamate CEST contrast measurements were calculated for within subjects and among the subjects and the coefficient of variations are reported. RESULTS The GluCEST measurements were highly reproducible in the gray and white matter area of the brain slice, whether it was within-day or between-day with a coefficient of variation of less than 5%. CONCLUSION This preliminary study in a small group of healthy volunteers shows a high degree of reproducibility of GluCEST MRI in brain and holds promise for implementation in studying age-dependent changes in the brain.
Collapse
Affiliation(s)
- Ravi Prakash Reddy Nanga
- Department of RadiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvania
| | - Catherine DeBrosse
- Department of RadiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvania
| | - Dushyant Kumar
- Department of RadiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvania
| | - David Roalf
- Department of PsychiatryUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvania
| | - Brendan McGeehan
- Department of PsychiatryUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvania
| | - Kevin D'Aquilla
- Department of RadiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvania
| | - Arijitt Borthakur
- Department of RadiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvania
| | - Hari Hariharan
- Department of RadiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvania
| | - Damodara Reddy
- Department of RadiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvania
| | - Mark Elliott
- Department of RadiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvania
| | - John A. Detre
- Department of NeurologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvania
| | - Cynthia Neill Epperson
- Department of PsychiatryUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvania
| | - Ravinder Reddy
- Department of RadiologyUniversity of Pennsylvania Perelman School of MedicinePhiladelphiaPennsylvania
| |
Collapse
|
43
|
Magnetic resonance imaging in Alzheimer's disease and mild cognitive impairment. J Neurol 2018; 266:1293-1302. [PMID: 30120563 PMCID: PMC6517561 DOI: 10.1007/s00415-018-9016-3] [Citation(s) in RCA: 189] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 08/07/2018] [Accepted: 08/11/2018] [Indexed: 11/25/2022]
Abstract
Research utilizing magnetic resonance imaging (MRI) has been crucial to the understanding of the neuropathological mechanisms behind and clinical identification of Alzheimer’s disease (AD) and mild cognitive impairment (MCI). MRI modalities show patterns of brain damage that discriminate AD from other brain illnesses and brain abnormalities that are associated with risk of conversion to AD from MCI and other behavioural outcomes. This review discusses the application of various MRI techniques to and their clinical usefulness in AD and MCI. MRI modalities covered include structural MRI, diffusion tensor imaging (DTI), arterial spin labelling (ASL), magnetic resonance spectroscopy (MRS), and functional MRI (fMRI). There is much evidence supporting the validity of MRI as a biomarker for these disorders; however, only traditional structural imaging is currently recommended for routine use in clinical settings. Future research is needed to warrant the inclusion for more advanced MRI methodology in forthcoming revisions to diagnostic criteria for AD and MCI.
Collapse
|
44
|
Zhang L, Li M, Sui R. Correlation between cerebellar metabolism and post-stroke depression in patients with ischemic stroke. Oncotarget 2017; 8:91711-91722. [PMID: 29207680 PMCID: PMC5710960 DOI: 10.18632/oncotarget.21063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 08/31/2017] [Indexed: 12/11/2022] Open
Abstract
The neurochemical changes that occur in the brain of patients with post-stroke depression (PSD) are not fully understood. This study aims to explore the correlation between cerebellar metabolism changes and PSD using proton magnetic resonance spectroscopy (1H-MRS). Participants were assigned to 3 groups: 60 patients with PSD (PSD group), 60 stroke patients without depression (NOPSD group), and 60 healthy volunteers (HEAL group). T1 WI, T2 WI, DWI and 1H-MRS examination were performed for patients at 14 days, 3 months after the stroke, respectively, and for healthy volunteers once when included in the study. Cho/Cr and Cho/NAA ratios in the cerebellar hemisphere contralateral to the lesion were higher in the PSD group than those in the HEAL and NOPSD groups on 14th day after the stroke (P < 0.05). In PSD group, Cho/Cr and Cho/NAA ratios in the cerebellar hemisphere contralateral to the lesion were positively correlated to the HAMD scale scores at both 14 days and 3 months after stroke (P < 0.05); Higher Cho/Cr and Cho/NAA ratios, and lower NAA/Cr ratio in the cerebellar hemisphere contralateral to the lesion were observed at 3 months after stroke compared to that at 14 days after stroke. Cerebellar damage may lead to PSD, and the degree of cerebellar damage may be associated with severity of PSD.
Collapse
Affiliation(s)
- Lei Zhang
- School of Nursing, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Muzi Li
- Department of Neurology, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Rubo Sui
- Department of Neurology, First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, China
| |
Collapse
|
45
|
Lagarde J, Sarazin M, Bottlaender M. In vivo PET imaging of neuroinflammation in Alzheimer's disease. J Neural Transm (Vienna) 2017; 125:847-867. [PMID: 28516240 DOI: 10.1007/s00702-017-1731-x] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 05/01/2017] [Indexed: 12/15/2022]
Abstract
Increasing evidence suggests that neuroinflammation contributes to the pathophysiology of many neurodegenerative diseases, especially Alzheimer's disease (AD). Molecular imaging by PET may be a useful tool to assess neuroinflammation in vivo, thus helping to decipher the complex role of inflammatory processes in the pathophysiology of neurodegenerative diseases and providing a potential means of monitoring the effect of new therapeutic approaches. For this objective, the main target of PET studies is the 18 kDa translocator protein (TSPO), as it is overexpressed by activated microglia. In the present review, we describe the most widely used PET tracers targeting the TSPO, the methodological issues in tracer quantification and summarize the results obtained by TSPO PET imaging in AD, as well as in neurodegenerative disorders associated with AD, in psychiatric disorders and ageing. We also briefly describe alternative PET targets and imaging modalities to study neuroinflammation. Lastly, we question the meaning of PET imaging data in the context of a highly complex and multifaceted role of neuroinflammation in neurodegenerative diseases. This overview leads to the conclusion that PET imaging of neuroinflammation is a promising way of deciphering the enigma of the pathophysiology of AD and of monitoring the effect of new therapies.
Collapse
Affiliation(s)
- Julien Lagarde
- Unit of Neurology of Memory and Language, Centre de Psychiatrie et Neurosciences, INSERM UMR S894, Centre Hospitalier Sainte-Anne and Université Paris Descartes, Sorbonne Paris Cité, 75014, Paris, France
| | - Marie Sarazin
- Unit of Neurology of Memory and Language, Centre de Psychiatrie et Neurosciences, INSERM UMR S894, Centre Hospitalier Sainte-Anne and Université Paris Descartes, Sorbonne Paris Cité, 75014, Paris, France
| | - Michel Bottlaender
- UNIACT, NeuroSpin, Institut d'Imagerie Biomédicale, Direction de la Recherche Fondamentale, Commissariat à l'Energie Atomique, 91191, Gif-sur-Yvette, France. .,Laboratoire Imagerie Moléculaire in Vivo, UMR 1023, Service Hospitalier Frédéric Joliot, Institut d'Imagerie Biomédicale, Direction de la Recherche Fondamentale, Commissariat à l'Energie Atomique, 91400, Orsay, France.
| |
Collapse
|
46
|
Szutowicz A, Bielarczyk H, Zyśk M, Dyś A, Ronowska A, Gul-Hinc S, Klimaszewska-Łata J. Early and Late Pathomechanisms in Alzheimer's Disease: From Zinc to Amyloid-β Neurotoxicity. Neurochem Res 2017; 42:891-904. [PMID: 28039593 PMCID: PMC5357490 DOI: 10.1007/s11064-016-2154-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 12/12/2016] [Accepted: 12/19/2016] [Indexed: 11/05/2022]
Abstract
There are several systemic and intracerebral pathologic conditions, which limit provision and utilization of energy precursor metabolites in neuronal cells. Energy deficits cause excessive depolarization of neuronal cells triggering glutamate-zinc evoked excitotoxic cascade. The intracellular zinc excess hits several intraneuronal targets yielding collapse of energy balance and impairment functional and structural impairments cholinergic neurons. Disturbances in metabolism of acetyl-CoA, which is a direct precursor for energy, acetylcholine, N-acetyl-L-aspartate and acetylated proteins synthesis, play an important role in these pathomechanisms. Disruption of brain homeostasis activates slow accumulation of amyloid-β 1-42 , which extra and intracellular oligomeric deposits disrupt diverse transporting and signaling processes in all membrane structures of the cell. Both neurotoxic signals may combine aggravating detrimental effects on neuronal cell. Different neuroglial and neuronal cell types may display differential susceptibility to similar pathogenic insults depending on specific features of their energy and functional parameters. This review, basing on findings gained from cellular and animal models of Alzheimer's disease, discusses putative energy/acetyl-CoA dependent mechanism in early and late stages of neurodegeneration.
Collapse
Affiliation(s)
- Andrzej Szutowicz
- Department of Laboratory Medicine, Medical University of Gdańsk, Ul. Dębinki 7, 80-211, Gdansk, Poland.
| | - Hanna Bielarczyk
- Department of Laboratory Medicine, Medical University of Gdańsk, Ul. Dębinki 7, 80-211, Gdansk, Poland
| | - Marlena Zyśk
- Department of Laboratory Medicine, Medical University of Gdańsk, Ul. Dębinki 7, 80-211, Gdansk, Poland
| | - Aleksandra Dyś
- Department of Laboratory Medicine, Medical University of Gdańsk, Ul. Dębinki 7, 80-211, Gdansk, Poland
| | - Anna Ronowska
- Department of Laboratory Medicine, Medical University of Gdańsk, Ul. Dębinki 7, 80-211, Gdansk, Poland
| | - Sylwia Gul-Hinc
- Department of Laboratory Medicine, Medical University of Gdańsk, Ul. Dębinki 7, 80-211, Gdansk, Poland
| | - Joanna Klimaszewska-Łata
- Department of Laboratory Medicine, Medical University of Gdańsk, Ul. Dębinki 7, 80-211, Gdansk, Poland
| |
Collapse
|