1
|
Liu W, Zhao Y, Rao Y, Wu Z, Peng Y, Gong L. Frontiers and hotspot evolution in research on Alzheimer's disease and hypertension: a bibliometric analysis from 2004 to 2023. Front Neurol 2025; 16:1514054. [PMID: 40356629 PMCID: PMC12066472 DOI: 10.3389/fneur.2025.1514054] [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: 10/19/2024] [Accepted: 04/09/2025] [Indexed: 05/15/2025] Open
Abstract
Background Alzheimer's disease (AD) is a neurodegenerative disease that imposes a heavy burden on patients and their families. Hypertension is an important risk factor for AD, but the specific mechanism of its impact is still unclear. This study thus aimed to analyze the relationship and trend changes between AD and hypertension through bibliometric methods. Methods Literature on AD and hypertension was retrieved from the Web of Science Core Collection (WoSCC) database between 2004 and 2023. Data regarding countries, institutions, authors and journals were sourced from WoSCC. CiteSpace and VOSviewer were used for data visualization, including author collaboration, timelines view of references, reference bursts and overlay visualization maps of keywords. Excel 2018 software was used for the statistical analysis. Results A total of 1,833 publications were ultimately included. From 2004 to 2023, the number of publications per year basically showed an increasing trend. The United States (United States) not only had the largest output of publications and the highest H-index but also had the seven highest frequencies of publication institutions. Kehoe, Patrick ranked first with the most articles among 9,330 authors. The journal with the most published articles was the Journal of Alzheimer's Disease. Reference analysis revealed a hotspot in the exploration of the pathophysiological association between AD and hypertension. Second, the treatment effects and potential risks of antihypertensive drugs (AHDs) on AD are also the focus of research. Researchers have carried out a series of studies ranging from basic research to clinical research on AHDs for the treatment of AD. Finally, personalized treatment strategies will also become one of the hotspots of future research. Controlling hypertension through lifestyle changes and medication interventions in AD patients is a promising strategy. The analysis of keywords revealed that "amyloid deposition," "preeclampsia," "Corona Virus Disease 2019 (COVID-19)" and "biomarkers" have been research hotspots in recent years. Conclusion By analyzing the references and keywords, we summarized the hot topics and research trends in this field. These findings provide useful information for researchers to explore the relationship between hypertension and AD further, with the hope of providing more effective treatments for AD patients to delay disease progression and improve quality of life.
Collapse
Affiliation(s)
- Wenying Liu
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Intelligent Medical Imaging, Nanchang, China
| | - Yaya Zhao
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Intelligent Medical Imaging, Nanchang, China
| | - Yutong Rao
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Intelligent Medical Imaging, Nanchang, China
| | - Zijing Wu
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Intelligent Medical Imaging, Nanchang, China
| | - Yun Peng
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Intelligent Medical Imaging, Nanchang, China
| | - Lianggeng Gong
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Intelligent Medical Imaging, Nanchang, China
| |
Collapse
|
2
|
Chao CK, Blecha J, Polvoy I, Nillo RM, Seo Y, Wilson DM, Forsayeth JR, VanBrocklin HF, Gerdes JM. First-in-human healthy volunteer dosimetry studies of the excitatory amino acid transporter 2 (EAAT2) PET imaging tracer methyl N 4-(7-[ 18F]fluoro-9H-fluoren-2-yl)asparaginate, [ 18F]RP-115. Nucl Med Biol 2025; 142-143:108992. [PMID: 39913962 DOI: 10.1016/j.nucmedbio.2025.108992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/21/2024] [Accepted: 01/08/2025] [Indexed: 05/04/2025]
Abstract
OBJECTIVE AND BACKGROUND The objective of this first-in-human study was to investigate the radiosynthesis, and the preliminary safety, biodistribution, and organ radiation dosimetry of the positron emission tomography (PET) imaging tracer methyl N4-([18F]7-fluoro-9H-fluoren-2-yl)asparaginate, known as [18F]RP-115, in a small cohort (n=8) of healthy volunteers. The [18F]RP-115 tracer is a methyl ester prodrug and undergoes metabolic saponification in the central nervous system to generate the corresponding carboxylic acid form that selectively binds to the excitatory amino acid transporter 2 (EAAT2) protein. PROCEDURES AND METHODS A multi-step high molar activity tracer radiosynthesis was devised to produce doses. Eight healthy human participants (four male and four female), aged 56-75, received a bolus intravenous injection of [18F]RP-115 (administered activity range: 70.3-355 MBq) prior to a total of 94 min of PET-MR scanning performed as three sequential scanning sessions. Regional tissue volumes of interest were defined, time-integrated activity coefficients (TIAC) were derived, and then estimates of organ and tissue activities and effective doses (whole body) were calculated, with two versions of OLINDA software (1.1 and 2.0) that incorporated two tissue weighting factor sets (ICRP-60 and -103), respectively. MAIN FINDINGS Tracer was routinely produced in good radiochemical yields and as suitable high molar activity doses for injection. The [18F]RP-115 injections and PET-MR scans were well-tolerated and no adverse events were reported (≤48 h). Radioactivity was widely biodistributed with good organ uptake. TIACs and estimated radiation organ doses were determined, for which a few statistically significant estimated organ dose differences between males and females were noted. The kidneys were identified as the critical target organ. PRINCIPAL CONCLUSIONS Injection of [18F]RP-115 was considered safe. The estimated kidney radiation doses relative to administered radioactivity identified a more optimal human [18F]RP-115 tracer injected amount of <211 MBq. This more optimal [18F]RP-115 tracer injected activity definition is similar to the amounts used for other established [18F]labeled clinical PET tracers such as [18F]FDG, and it will be used in future RP-115 clinical PET imaging studies.
Collapse
Affiliation(s)
- Chih-Kai Chao
- Rio Pharmaceuticals, Inc., 18 Elsie St., San Francisco, CA 94110, USA.
| | - Joseph Blecha
- Department of Radiology and Biomedical Imaging, University of California San Francisco, 185 Berry St., Suite 350, San Francisco, CA 94107, USA.
| | - Ilona Polvoy
- Department of Radiology and Biomedical Imaging, University of California San Francisco, 185 Berry St., Suite 350, San Francisco, CA 94107, USA
| | - Ryan Michael Nillo
- Department of Radiology and Biomedical Imaging, University of California San Francisco, 185 Berry St., Suite 350, San Francisco, CA 94107, USA.
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California San Francisco, 185 Berry St., Suite 350, San Francisco, CA 94107, USA.
| | - David M Wilson
- Department of Radiology and Biomedical Imaging, University of California San Francisco, 185 Berry St., Suite 350, San Francisco, CA 94107, USA.
| | - John R Forsayeth
- Rio Pharmaceuticals, Inc., 18 Elsie St., San Francisco, CA 94110, USA.
| | - Henry F VanBrocklin
- Department of Radiology and Biomedical Imaging, University of California San Francisco, 185 Berry St., Suite 350, San Francisco, CA 94107, USA.
| | - John M Gerdes
- Rio Pharmaceuticals, Inc., 18 Elsie St., San Francisco, CA 94110, USA.
| |
Collapse
|
3
|
Dugan R, Ramakrishnapillai S, Amant JS, Murray K, McKlveen K, Naseri M, Madden K, Bazzano L, Carmichael O. Lifespan cardiometabolic exposures are associated with midlife white matter microstructure: The Bogalusa Heart Study. Neuroimage 2025; 307:121034. [PMID: 39826773 DOI: 10.1016/j.neuroimage.2025.121034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025] Open
Abstract
INTRODUCTION Compartment model analysis of diffusion MRI data provides unique information on the microstructural properties of white matter. However, studies relating compartment model microstructural measures to longitudinal cardiometabolic health data are rare. METHODS 130 cognitively healthy participants in the Bogalusa Heart Study completed diffusion MRI scans. Compartment model analysis was performed, and summary metrics were measured in organized and diffuse white matter. Multiple linear regression models were used to relate the white matter microstructure metrics to demographics and cardiometabolic risk factors. RESULTS In both organized and diffuse white matter, age was associated with worse diffusion metrics, women had better diffusion metrics than men, and African American participants had worse diffusion metrics compared to White participants. Greater blood pressure in pre-adulthood was associated with worse diffusion metrics in midlife. DISCUSSION Summary metrics from compartment model analyses of diffusion MRI data were associated cardiometabolic risk factors from youth to midlife as well as demographic factors.
Collapse
Affiliation(s)
- Reagan Dugan
- Department of Physics & Astronomy, Louisiana State University, 202 Nicholson Hall, Baton Rouge, LA, 70803, USA; Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA.
| | - Sreekrishna Ramakrishnapillai
- Department of Neurological Surgery, University of Pittsburgh, 200 Lothrop St suite b-400, Pittsburgh, PA, 15213, USA
| | - Julia St Amant
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA
| | - Kori Murray
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA
| | - Kevin McKlveen
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA
| | - Maryam Naseri
- Department of Physics & Astronomy, Louisiana State University, 202 Nicholson Hall, Baton Rouge, LA, 70803, USA; Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA
| | - Kaitlyn Madden
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA
| | - Lydia Bazzano
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, 1440 Canal Street, Suite 2000, New Orleans, LA, 70112, USA
| | - Owen Carmichael
- Pennington Biomedical Research Center, 6400 Perkins Road, Baton Rouge, LA, 70808, USA
| |
Collapse
|
4
|
Patil S, Ayubcha C, Teichner E, Subtirelu R, Cho JH, Ghonim M, Ghonim M, Werner TJ, Høilund-Carlsen PF, Alavi A, Newberg AB. Clinical Applications of PET Imaging in Alzheimer's Disease. PET Clin 2025; 20:89-100. [PMID: 39547733 DOI: 10.1016/j.cpet.2024.09.015] [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] [Indexed: 11/17/2024]
Abstract
Alzheimer's disease (AD) involves a complex pathophysiology of neurodegeneration that leads to severe cognitive deficiencies. Understanding the molecular alterations that underlie this disease is fundamental to clinical management and therapeutic innovation. Functional imaging with positron emission tomography (PET) enables a visualization of these impaired pathways, such as cerebral hypometabolism, amyloid and tau accumulation, and neurotransmitter dysfunction. This review discusses the clinical applications of PET in AD and mild cognitive impairment, focusing on relevant original research studies for disease diagnosis, progression, and treatment response.
Collapse
Affiliation(s)
- Shiv Patil
- Department of Radiology, Hospital of the University of Pennsylvania, PA, USA; Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Cyrus Ayubcha
- Harvard Medical School, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Eric Teichner
- Department of Radiology, Hospital of the University of Pennsylvania, PA, USA; Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA
| | - Robert Subtirelu
- Department of Radiology, Hospital of the University of Pennsylvania, PA, USA
| | | | - Mohanad Ghonim
- Department of Radiology, Hospital of the University of Pennsylvania, PA, USA; Department of Radiology, Ain Shams University, Cairo, Egypt
| | - Mohamed Ghonim
- Department of Radiology, Hospital of the University of Pennsylvania, PA, USA; Department of Radiology, Ain Shams University, Cairo, Egypt
| | - Thomas J Werner
- Department of Radiology, Hospital of the University of Pennsylvania, PA, USA
| | | | - Abass Alavi
- Department of Radiology, Hospital of the University of Pennsylvania, PA, USA
| | - Andrew B Newberg
- Marcus Institute of Integrative Health, Thomas Jefferson University, Philadelphia, PA 19107, USA; Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
5
|
Bao YW, Wang ZJ, Shea YF, Chiu PKC, Kwan JS, Chan FHW, Mak HKF. Combined Quantitative amyloid-β PET and Structural MRI Features Improve Alzheimer's Disease Classification in Random Forest Model - A Multicenter Study. Acad Radiol 2024; 31:5154-5163. [PMID: 39003227 DOI: 10.1016/j.acra.2024.06.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/18/2024] [Accepted: 06/24/2024] [Indexed: 07/15/2024]
Abstract
RATIONALE AND OBJECTIVES Prior to clinical presentations of Alzheimer's Disease (AD), neuropathological changes, such as amyloid-β and brain atrophy, have accumulated at the earlier stages of the disease. The combination of such biomarkers assessed by multiple modalities commonly improves the likelihood of AD etiology. We aimed to explore the discriminative ability of Aβ PET features and whether combining Aβ PET and structural MRI features can improve the classification performance of the machine learning model in older healthy control (OHC) and mild cognitive impairment (MCI) from AD. MATERIAL AND METHODS We collected 94 AD patients, 82 MCI patients, and 85 OHC from three different cohorts. 17 global/regional Aβ features in Centiloid, 122 regional volume, and 68 regional cortical thickness were extracted as imaging features. Single or combined modality features were used to train the random forest model on the testing set. The top 10 features were sorted based on the Gini index in each binary classification. RESULTS The results showed that AUC scores were 0.81/0.86 and 0.69/0.68 using sMRI/Aβ PET features on the testing set in differentiating OHC and MCI from AD. The performance was improved while combining two-modality features with an AUC of 0.89 and an AUC of 0.71 in two classifications. Compared to sMRI features, particular Aβ PET features contributed more to differentiating AD from others. CONCLUSION Our study demonstrated the discriminative ability of Aβ PET features in differentiating AD from OHC and MCI. A combination of Aβ PET and structural MRI features can improve the RF model performance.
Collapse
Affiliation(s)
- Yi-Wen Bao
- Department of Medical Imaging Center, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China (Y-W.B.)
| | - Zuo-Jun Wang
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China (Z-J.W., H.K-F.M.)
| | - Yat-Fung Shea
- Department of Medicine, Queen Mary Hospital, Hong Kong SAR, China (Y-F.S., P.K-C.C., J.S.K., F.H-W.C.)
| | - Patrick Ka-Chun Chiu
- Department of Medicine, Queen Mary Hospital, Hong Kong SAR, China (Y-F.S., P.K-C.C., J.S.K., F.H-W.C.)
| | - Joseph Sk Kwan
- Department of Medicine, Queen Mary Hospital, Hong Kong SAR, China (Y-F.S., P.K-C.C., J.S.K., F.H-W.C.)
| | - Felix Hon-Wai Chan
- Department of Medicine, Queen Mary Hospital, Hong Kong SAR, China (Y-F.S., P.K-C.C., J.S.K., F.H-W.C.)
| | - Henry Ka-Fung Mak
- Department of Diagnostic Radiology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China (Z-J.W., H.K-F.M.).
| |
Collapse
|
6
|
Rudroff T, Rainio O, Klén R. AI for the prediction of early stages of Alzheimer's disease from neuroimaging biomarkers - A narrative review of a growing field. Neurol Sci 2024; 45:5117-5127. [PMID: 38866971 DOI: 10.1007/s10072-024-07649-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 06/10/2024] [Indexed: 06/14/2024]
Abstract
OBJECTIVES The objectives of this narrative review are to summarize the current state of AI applications in neuroimaging for early Alzheimer's disease (AD) prediction and to highlight the potential of AI techniques in improving early AD diagnosis, prognosis, and management. METHODS We conducted a narrative review of studies using AI techniques applied to neuroimaging data for early AD prediction. We examined single-modality studies using structural MRI and PET imaging, as well as multi-modality studies integrating multiple neuroimaging techniques and biomarkers. Furthermore, they reviewed longitudinal studies that model AD progression and identify individuals at risk of rapid decline. RESULTS Single-modality studies using structural MRI and PET imaging have demonstrated high accuracy in classifying AD and predicting progression from mild cognitive impairment (MCI) to AD. Multi-modality studies, integrating multiple neuroimaging techniques and biomarkers, have shown improved performance and robustness compared to single-modality approaches. Longitudinal studies have highlighted the value of AI in modeling AD progression and identifying individuals at risk of rapid decline. However, challenges remain in data standardization, model interpretability, generalizability, clinical integration, and ethical considerations. CONCLUSION AI techniques applied to neuroimaging data have the potential to improve early AD diagnosis, prognosis, and management. Addressing challenges related to data standardization, model interpretability, generalizability, clinical integration, and ethical considerations is crucial for realizing the full potential of AI in AD research and clinical practice. Collaborative efforts among researchers, clinicians, and regulatory agencies are needed to develop reliable, robust, and ethical AI tools that can benefit AD patients and society.
Collapse
Affiliation(s)
- Thorsten Rudroff
- Department of Health and Human Physiology, University of Iowa, Iowa City, IA, 52242, USA.
- Department of Neurology, University of Iowa Hospitals and Clinics, Iowa City, IA, 52242, USA.
| | - Oona Rainio
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| | - Riku Klén
- Turku PET Centre, University of Turku and Turku University Hospital, Turku, Finland
| |
Collapse
|
7
|
Honda G, Nagamachi S, Takahashi M, Higuma Y, Tani T, Hida K, Yoshimitsu K, Ogomori K, Tsuboi Y. The usefulness of combined analysis using CIScore and VSRAD parameters for differentiating between dementia with Lewy body and Alzheimer's disease. Jpn J Radiol 2024; 42:1206-1212. [PMID: 38856880 PMCID: PMC11442568 DOI: 10.1007/s11604-024-01604-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 05/26/2024] [Indexed: 06/11/2024]
Abstract
PURPOSE The Cingulate Island score (CIScore) is useful index for differentiating between dementia with Lewy body (DLB) and Alzheimer's disease (AD) using regional cerebral blood flow (rCBF) SPECT. The Z score standing for medial temporal lobe (MTL) atrophy and the ratio of Z score between dorsal brain stem (DBS) to MTL are useful indices for differentiating between DLB and AD using MRI with VSRAD. The current study investigated the diagnostic ability by the combined use of rCBF SPECT and MRI in the differentiation between AD and DLB. MATERIALS AND METHODS In cases with 42 AD and 28 DLB undertaken Tc-99m-ECD SPECT and MRI, we analyzed differential diagnostic ability between AD and DLB among following conditions by single or combined settings. Namely, they were (1) the CIScore as a parameter of rCBF SPECT (DLB ≦ 0.25), (2) Z score value of MTL atrophy (DLB ≦ 2.05), (3) the ratio of Z score of DBS to medial temporal gray matter as a parameter of brain atrophy using VSRAD (DLB ≧ 0.38). Also, we analyzed them both including and omitting the elderly (over 75 years old). RESULTS The accuracy of differential diagnosis in this condition was 74% for (1), 69% for (2), and 67% for (3). The accuracy by combination condition was 84% for (1) and (2), 81% for (1) and (3), and 67% for (2) and (3), respectively. The combination method by CIScore and the Z score of MTL showed the best accuracy. When we confined condition to ages younger than 75 years, the accuracy improved to 94% in the combination method. CONCLUSION The combined use of CIScore and Z score of MTL was suggested to be useful in the differential diagnosis between DLB and AD particularly in younger than 75 years old.
Collapse
Affiliation(s)
- Gaku Honda
- Department of Radiology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan.
| | - Shigeki Nagamachi
- Department of Radiology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Mai Takahashi
- Department of Radiology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Yukie Higuma
- Department of Radiology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Tomonobu Tani
- Department of Radiology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Kosuke Hida
- Department of Radiology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Kengo Yoshimitsu
- Department of Radiology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Koji Ogomori
- Department of Psychiatry, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| | - Yoshio Tsuboi
- Department of Neurology, Faculty of Medicine, Fukuoka University, Fukuoka, Japan
| |
Collapse
|
8
|
Duman Sastim D, Elboga G, Elboga U, Gungor K. Evaluation of the relationship between FDG-PET hypometabolism and retinal layer thickness in patients with Alzheimer's disease. Acta Neurol Belg 2024; 124:987-993. [PMID: 38546932 DOI: 10.1007/s13760-024-02511-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 02/23/2024] [Indexed: 06/01/2024]
Abstract
We aimed to investigate the diagnostic value of Optical coherence tomography (OCT) in Alzheimer's disease (AD) and to assess the correlation between OCT and fluorodeoxyglucose (FDG)-positron emission tomography (PET) which shows high diagnostic agreement with findings from postmortem histopathology-the gold standard method. Patients who were diagnosed with AD-related dementia were selected for the study. Patients with a mini mental test (MMT) score between 18 and 23 were included in the study (n = 31). Volunteers with MMT ≥ 28 and no cognitive impairment were included in the study as the control group (n = 31). OCT imaging was performed in the patient and control groups after detailed ophthalmological examinations including visual acuity and intraocular pressure measurements. Brain glucose metabolism measurement was performed using 18 F-FDG PET/computed tomography. When adjusted for age and sex, mean retinal nerve fiber layer thickness (RNFL) thickness showed a significant difference between groups and the RNFL thickness in the superior temporal and superior nasal quadrants in AD-related mild dementia group showed a significant difference (p < 0.05). Furthermore, only the RNFL thickness in the inferior nasal quadrant of the right eye showed a significant difference between the groups (p = 0.016). It is thought that OCT is a promising imaging method in the elderly population due to its low-cost, non-invasive and easily applicability, and therefore, it may contribute in the future as a tool in the periodic follow-up of patients diagnosed with AD.
Collapse
Affiliation(s)
- Demet Duman Sastim
- Department of Psychiatry, Tunceli State Hospital, Merkez, 62000, Merkez/Tunceli, Turkey.
| | - Gulcin Elboga
- Faculty of Medicine, Department of Psychiatry, Gaziantep University, Gaziantep, Turkey
| | - Umut Elboga
- Faculty of Medicine, Department of Nuclear Medicine, Gaziantep University, Gaziantep, Turkey
| | - Kivanc Gungor
- Faculty of Medicine, Department of Ophthalmology, Gaziantep University, Gaziantep, Turkey
| |
Collapse
|
9
|
Castellano G, Esposito A, Lella E, Montanaro G, Vessio G. Automated detection of Alzheimer's disease: a multi-modal approach with 3D MRI and amyloid PET. Sci Rep 2024; 14:5210. [PMID: 38433282 PMCID: PMC10909869 DOI: 10.1038/s41598-024-56001-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 02/28/2024] [Indexed: 03/05/2024] Open
Abstract
Recent advances in deep learning and imaging technologies have revolutionized automated medical image analysis, especially in diagnosing Alzheimer's disease through neuroimaging. Despite the availability of various imaging modalities for the same patient, the development of multi-modal models leveraging these modalities remains underexplored. This paper addresses this gap by proposing and evaluating classification models using 2D and 3D MRI images and amyloid PET scans in uni-modal and multi-modal frameworks. Our findings demonstrate that models using volumetric data learn more effective representations than those using only 2D images. Furthermore, integrating multiple modalities enhances model performance over single-modality approaches significantly. We achieved state-of-the-art performance on the OASIS-3 cohort. Additionally, explainability analyses with Grad-CAM indicate that our model focuses on crucial AD-related regions for its predictions, underscoring its potential to aid in understanding the disease's causes.
Collapse
Affiliation(s)
| | - Andrea Esposito
- Department of Computer Science, University of Bari Aldo Moro, Bari, Italy
| | - Eufemia Lella
- Sirio - Research & Innovation, Sidea Group, Bari, Italy
| | | | - Gennaro Vessio
- Department of Computer Science, University of Bari Aldo Moro, Bari, Italy.
| |
Collapse
|
10
|
Brugnolo A, Orso B, Girtler N, Ferraro PM, Arnaldi D, Mattioli P, Massa F, Famà F, Argenti L, Biffa G, Morganti W, Buonopane S, Uccelli A, Morbelli S, Pardini M. Tracking the progression of Alzheimer's disease: Insights from metabolic patterns of SOMI stages. Cortex 2024; 171:413-422. [PMID: 38113612 DOI: 10.1016/j.cortex.2023.11.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 08/14/2023] [Accepted: 11/08/2023] [Indexed: 12/21/2023]
Abstract
BACKGROUND SOMI (Stages of Objective Memory Impairment) is a novel classification that identifies six stages of memory decline in Alzheimer's Disease (AD) using the Free and Cued Selective Reminding Test (FCSRT). However, the relationship between SOMI stages and brain metabolism remains unexplored. This study aims to investigate the metabolic correlates of SOMI stages using FDG-PET in Mild Cognitive Impairment due to AD (MCI-AD) and early AD patients. METHODS One hundred twenty-nine-patients (99 aMCI-AD and 30 AD), and 42 healthy controls (HCs) (MMSE = 29.2 ± .8; age:69.1 ± 8.6 years; education:10.7 ± 3.8 years) who underwent an extensive neuropsychological battery including FCSRT and brain FDG-PET were enrolled. According to their clinical relevance and available sample sizes, SOMI-4 (N = 24 subjects; MMSE score:26.6 ± 2.6: age:75.4 ± 3.2; education:9.9 ± 4.5) and SOMI-5 groups (N = 97; MMSE:25.3 ± 2.6; age:73.9 ± 5.8; education:9.4 ± 4.1) were investigated. RESULTS Compared to HCs, SOMI-4 showed hypometabolism in the precuneus, medial temporal gyrus bilaterally, right pecuneus and angular gyrus. SOMI-5 exhibited broader hypometabolism, extending to the left posterior cingulate and medial frontal gyrus bilaterally. The conjunction analysis revealed overlapping areas in the precuneus, medial temporal gyrus bilaterally, and in the right angular gyrus and cuneus. The disjunction analysis identified SOMI-5 specific hypometabolism encompassing left inferior temporal gyrus, uncus and parahippocampal gyrus, and medial frontal gyrus bilaterally (p < .001, p-value (FWE) < .05). DISCUSSION SOMI-4 relates to posterior hypometabolism, while SOMI-5 to more extensive hypometabolism further encompassing frontal cortices, suggesting SOMI as a biologically relevant classification system of memory decline. CONCLUSION Memory decline staged with SOMI is associated with hypometabolism spreading in amnesic MCI-AD/AD, suggesting its usefulness as a clinical marker of increasing neurodegeneration.
Collapse
Affiliation(s)
- Andrea Brugnolo
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Mother-Child Health (DINOGMI), University of Genoa, Italy; Clinical Psychology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Beatrice Orso
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Mother-Child Health (DINOGMI), University of Genoa, Italy.
| | - Nicola Girtler
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Mother-Child Health (DINOGMI), University of Genoa, Italy; Clinical Psychology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | | | - Dario Arnaldi
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Mother-Child Health (DINOGMI), University of Genoa, Italy; Neurology Clinics, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Pietro Mattioli
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Mother-Child Health (DINOGMI), University of Genoa, Italy; Neurology Clinics, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Federico Massa
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Mother-Child Health (DINOGMI), University of Genoa, Italy; Neurology Clinics, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Francesco Famà
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Mother-Child Health (DINOGMI), University of Genoa, Italy; Neurology Clinics, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Lucia Argenti
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Mother-Child Health (DINOGMI), University of Genoa, Italy.
| | - Gabriella Biffa
- Clinical Psychology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Wanda Morganti
- Geriatrics Unit, Department of Geriatric Care, Orthogeriatrics and Rehabilitation, E.O. Galliera Hospital, Genoa, Italy.
| | - Silvia Buonopane
- Geriatrics Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Antonio Uccelli
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Mother-Child Health (DINOGMI), University of Genoa, Italy; IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Silvia Morbelli
- Department of Health Sciences, University of Genoa, Italy; Nuclear Medicine Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Matteo Pardini
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, and Mother-Child Health (DINOGMI), University of Genoa, Italy; Neurology Clinics, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| |
Collapse
|
11
|
Rathee S, Sen D, Pandey V, Jain SK. Advances in Understanding and Managing Alzheimer's Disease: From Pathophysiology to Innovative Therapeutic Strategies. Curr Drug Targets 2024; 25:752-774. [PMID: 39039673 DOI: 10.2174/0113894501320096240627071400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 05/30/2024] [Accepted: 06/04/2024] [Indexed: 07/24/2024]
Abstract
Alzheimer's disease (AD) is a debilitating neurodegenerative disorder characterized by the presence of amyloid-β (Aβ) plaques and tau-containing neurofibrillary tangles, leading to cognitive and physical decline. Representing the majority of dementia cases, AD poses a significant burden on healthcare systems globally, with onset typically occurring after the age of 65. While most cases are sporadic, about 10% exhibit autosomal forms associated with specific gene mutations. Neurofibrillary tangles and Aβ plaques formed by misfolded tau proteins and Aβ peptides contribute to neuronal damage and cognitive impairment. Currently, approved drugs, such as acetylcholinesterase inhibitors and N-methyl D-aspartate receptor agonists, offer only partial symptomatic relief without altering disease progression. A promising development is using lecanemab, a humanized IgG1 monoclonal antibody, as an immune therapeutic approach. Lecanemab demonstrates selectivity for polymorphic Aβ variants and binds to large soluble Aβ aggregates, providing a potential avenue for targeted treatment. This shift in understanding the role of the adaptive immune response in AD pathogenesis opens new possibilities for therapeutic interventions aiming to address the disease's intricate mechanisms. This review aims to summarize recent advancements in understanding Alzheimer's disease pathophysiology and innovative therapeutic approaches, providing valuable insights for both researchers and clinicians.
Collapse
Affiliation(s)
- Sunny Rathee
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh, 470003, India
| | - Debasis Sen
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh, 470003, India
| | - Vishal Pandey
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh, 470003, India
| | - Sanjay K Jain
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour Vishwavidyalaya (A Central University), Sagar, Madhya Pradesh, 470003, India
| |
Collapse
|
12
|
Mir M, Khosravani P, Ramezannezhad E, Saadabad FP, Falahati M, Ghanbarian M, Saberian P, Sadeghi M, Niknam N, Ghejelou SE, Jafari M, Gulisashvili D, Mayeli M. Associations Between Metabolomics Findings and Brain Hypometabolism in Mild Cognitive Impairment and Alzheimer's Disease. Curr Alzheimer Res 2024; 21:679-689. [PMID: 39878109 DOI: 10.2174/0115672050350196250110092338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 11/19/2024] [Accepted: 11/26/2024] [Indexed: 01/31/2025]
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disease with rising prevalence due to the aging global population. Existing methods for diagnosing AD are struggling to detect the condition in its earliest and most treatable stages. One early indicator of AD is a substantial decrease in the brain's glucose metabolism. Metabolomics can detect disturbances in biofluids, which may be advantageous for early detection of some AD-related changes. The study aims to predict brain hypometabolism in Alzheimer's disease using metabolomics findings and develop a predictive model based on metabolomic data. METHODS The data used in this study were acquired from the Alzheimer's Disease Neuroimaging Initiative (ADNI) project. We conducted a longitudinal study with three assessment time points to investigate the predictive power of baseline metabolomics for modeling longitudinal fluorodeoxyglucose- positron emission tomography (FDG-PET) trajectory changes in AD patients. A total of 44 participants with AD were included. The Alzheimer's Disease Assessment Scale (ADAS), the Mini-Mental State Examination (MMSE), and the Clinical Dementia Rating Scale-Sum of Boxes (CDR-SB) were used for cognitive assessments. A single global brain hypo-metabolism index was used as the outcome variable. RESULTS Across models, we observed consistent positive relationships between specific cholesterol esters - CE (20:3) (p = 0.005) and CE (18:3) (p = 0.0039) - and FDG-PET metrics, indicating these baseline metabolites may be valuable indicators of future PET score changes. Selected triglycerides like DG-O (16:0-20:4) also showed time-specific positive associations (p = 0.017). CONCLUSION This research provides new insights into the disruptions in the metabolic network linked to AD pathology. These findings could pave the way for identifying novel biomarkers and potential treatment targets for AD.
Collapse
Affiliation(s)
- Moein Mir
- Golestan Rheumatology Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Parinaz Khosravani
- Student's Scientific Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Fatemeh Pourali Saadabad
- Department of Psychology, Faculty of Humanities and Social Sciences, Khayyam University of Mashhad, Mashhad, Iran
| | - Marjan Falahati
- Department of Pharmaceutical Science, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mahsa Ghanbarian
- Department of Health Psychology, Faculty of Medical Sciences, Gorgan Branch, Islamic Azad University, Gorgan, Iran
| | - Parsa Saberian
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mohammad Sadeghi
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nafise Niknam
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sanaz Eskandari Ghejelou
- Department of Clinical Psychology, Faculty of Psychology and Educational Sciences, Tabriz branch, Islamic Azad University, Tabriz, Iran
| | - Masoumeh Jafari
- Student's Scientific Research Center, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - David Gulisashvili
- Department of Diagnostic Radiology & Nuclear Medicine, University of Maryland, School of Medicine, Baltimore, Maryland, USA
| | - Mahsa Mayeli
- Department of Diagnostic Radiology & Nuclear Medicine, University of Maryland, School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
13
|
Triumbari EKA, Chiaravalloti A, Schillaci O, Mercuri NB, Liguori C. Positron Emission Tomography/Computed Tomography Imaging in Therapeutic Clinical Trials in Alzheimer's Disease: An Overview of the Current State of the Art of Research. J Alzheimers Dis 2024; 101:S603-S628. [PMID: 39422956 DOI: 10.3233/jad-240349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
The integration of positron emission tomography/computed tomography (PET/CT) has revolutionized the landscape of Alzheimer's disease (AD) research and therapeutic interventions. By combining structural and functional imaging, PET/CT provides a comprehensive understanding of disease pathology and response to treatment assessment. PET/CT, particularly with 2-deoxy-2-[fluorine-18]fluoro-D-glucose (18F-FDG), facilitates the visualization of glucose metabolism in the brain, enabling early diagnosis, staging, and monitoring of neurodegenerative disease progression. The advent of amyloid and tau PET imaging has further propelled the field forward, offering invaluable tools for tracking pathological hallmarks, assessing treatment response, and predicting clinical outcomes. While some therapeutic interventions targeting amyloid plaque load showed promising results with the reduction of cerebral amyloid accumulation over time, others failed to demonstrate a significant impact of anti-amyloid agents for reducing the amyloid plaques burden in AD brains. Tau PET imaging has conversely fueled the advent of disease-modifying therapeutic strategies in AD by supporting the assessment of neurofibrillary tangles of tau pathology deposition over time. Looking ahead, PET imaging holds immense promise for studying additional targets such as neuroinflammation, cholinergic deficit, and synaptic dysfunction. Advances in radiotracer development, dedicated brain PET/CT scanners, and Artificial Intelligence-powered software are poised to enhance the quality, sensitivity, and diagnostic power of molecular neuroimaging. Consequently, PET/CT remains at the forefront of AD research, offering unparalleled opportunities for unravelling the complexities of the disease and advancing therapeutic interventions, although it is not yet enough alone to allow patients' recruitment in therapeutic clinical trials.
Collapse
Affiliation(s)
| | - Agostino Chiaravalloti
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- IRCCS Neuromed, Pozzilli, Italy
| | - Orazio Schillaci
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Nicola Biagio Mercuri
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Neurology Unit, University Hospital of Rome "Tor Vergata", Rome, Italy
| | - Claudio Liguori
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
- Neurology Unit, University Hospital of Rome "Tor Vergata", Rome, Italy
| |
Collapse
|
14
|
Yu H, Wang F, Wu J, Gong J, Bi S, Mao Y, Jia D, Chai G. Integrated transcriptomics reveals the brain and blood biomarkers in Alzheimer's disease. CNS Neurosci Ther 2023; 29:3943-3951. [PMID: 37334737 PMCID: PMC10651972 DOI: 10.1111/cns.14316] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 06/05/2023] [Accepted: 06/07/2023] [Indexed: 06/20/2023] Open
Abstract
BACKGROUND The systematic molecular associations between the peripheral blood cells and brain in Alzheimer's disease (AD) remains unclear, which hinders our understanding of AD pathological mechanisms and the exploration of new diagnostic biomarkers. METHODS Here, we performed an integrated analysis of the brain and peripheral blood cells transcriptomics to establish peripheral biomarkers of AD. By employing multiple statistical analyses plus machine learning, we identified and validated multiple regulated central and peripheral network in patients with AD. RESULTS By bioinformatics analysis, a total of 243 genes were differentially expressed in the central and peripheral systems, mainly enriched in three modules: immune response, glucose metabolism and lysosome. In addition, lysosome related gene ATP6V1E1 and immune response related genes (IL2RG, OSM, EVI2B TNFRSF1A, CXCR4, STAT5A) were significantly correlated with Aβ or Tau pathology. Finally, receiver operating characteristic (ROC) analysis revealed that ATP6V1E1 showed high-diagnostic potential for AD. CONCLUSION Taken together, our data identified the main pathological pathways in AD progression, particularly the systemic dysregulation of the immune response, and provided peripheral biomarkers for AD diagnosis.
Collapse
Affiliation(s)
- Haitao Yu
- Department of Fundamental Medicine, Wuxi School of MedicineJiangnan UniversityWuxiChina
| | - Fangzhou Wang
- Department of Fundamental Medicine, Wuxi School of MedicineJiangnan UniversityWuxiChina
| | - Jia‐jun Wu
- Department of Fundamental Medicine, Wuxi School of MedicineJiangnan UniversityWuxiChina
| | - Juan Gong
- Department of Fundamental Medicine, Wuxi School of MedicineJiangnan UniversityWuxiChina
| | - Shuguang Bi
- Department of Fundamental Medicine, Wuxi School of MedicineJiangnan UniversityWuxiChina
| | - Yumin Mao
- Department of Fundamental Medicine, Wuxi School of MedicineJiangnan UniversityWuxiChina
| | - Dongdong Jia
- The Affiliated Mental Health Center of Jiangnan UniversityWuxi Central Rehabilitation HospitalWuxiChina
| | - Gao‐shang Chai
- Department of Fundamental Medicine, Wuxi School of MedicineJiangnan UniversityWuxiChina
| |
Collapse
|
15
|
Kepp KP, Robakis NK, Høilund-Carlsen PF, Sensi SL, Vissel B. The amyloid cascade hypothesis: an updated critical review. Brain 2023; 146:3969-3990. [PMID: 37183523 DOI: 10.1093/brain/awad159] [Citation(s) in RCA: 76] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 04/20/2023] [Accepted: 04/23/2023] [Indexed: 05/16/2023] Open
Abstract
Results from recent clinical trials of antibodies that target amyloid-β (Aβ) for Alzheimer's disease have created excitement and have been heralded as corroboration of the amyloid cascade hypothesis. However, while Aβ may contribute to disease, genetic, clinical, imaging and biochemical data suggest a more complex aetiology. Here we review the history and weaknesses of the amyloid cascade hypothesis in view of the new evidence obtained from clinical trials of anti-amyloid antibodies. These trials indicate that the treatments have either no or uncertain clinical effect on cognition. Despite the importance of amyloid in the definition of Alzheimer's disease, we argue that the data point to Aβ playing a minor aetiological role. We also discuss data suggesting that the concerted activity of many pathogenic factors contribute to Alzheimer's disease and propose that evolving multi-factor disease models will better underpin the search for more effective strategies to treat the disease.
Collapse
Affiliation(s)
- Kasper P Kepp
- Section of Biophysical and Biomedicinal chemistry, DTU Chemistry, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | - Nikolaos K Robakis
- Icahn School of Medicine at Mount Sinai Medical Center, New York, NY 10029, USA
| | - Poul F Høilund-Carlsen
- Department of Nuclear Medicine, Odense University Hospital, 5000 Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, 5000 Odense C, Denmark
| | - Stefano L Sensi
- Center for Advanced Studies and Technology-CAST, and Institute for Advanced Biotechnology (ITAB), University G. d'Annunzio of Chieti-Pescara, Chieti, 66013, Italy
- Department of Neuroscience, Imaging, and Clinical Sciences, University G. d'Annunzio of Chieti-Pescara, Chieti, 66013, Italy
| | - Bryce Vissel
- St Vincent's Hospital Centre for Applied Medical Research, St Vincent's Hospital, Sydney, 2010, Australia
- School of Clinical Medicine, UNSW Medicine and Health, St Vincent's Healthcare Clinical Campus, Faculty of Medicine and Health, Sydney, NSW 2052, Australia
| |
Collapse
|
16
|
Rani S, Dhar SB, Khajuria A, Gupta D, Jaiswal PK, Singla N, Kaur M, Singh G, Barnwal RP. Advanced Overview of Biomarkers and Techniques for Early Diagnosis of Alzheimer's Disease. Cell Mol Neurobiol 2023; 43:2491-2523. [PMID: 36847930 PMCID: PMC11410160 DOI: 10.1007/s10571-023-01330-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 02/15/2023] [Indexed: 03/01/2023]
Abstract
The development of early non-invasive diagnosis methods and identification of novel biomarkers are necessary for managing Alzheimer's disease (AD) and facilitating effective prognosis and treatment. AD has multi-factorial nature and involves complex molecular mechanism, which causes neuronal degeneration. The primary challenges in early AD detection include patient heterogeneity and lack of precise diagnosis at the preclinical stage. Several cerebrospinal fluid (CSF) and blood biomarkers have been proposed to show excellent diagnosis ability by identifying tau pathology and cerebral amyloid beta (Aβ) for AD. Intense research endeavors are being made to develop ultrasensitive detection techniques and find potent biomarkers for early AD diagnosis. To mitigate AD worldwide, understanding various CSF biomarkers, blood biomarkers, and techniques that can be used for early diagnosis is imperative. This review attempts to provide information regarding AD pathophysiology, genetic and non-genetic factors associated with AD, several potential blood and CSF biomarkers, like neurofilament light, neurogranin, Aβ, and tau, along with biomarkers under development for AD detection. Besides, numerous techniques, such as neuroimaging, spectroscopic techniques, biosensors, and neuroproteomics, which are being explored to aid early AD detection, have been discussed. The insights thus gained would help in finding potential biomarkers and suitable techniques for the accurate diagnosis of early AD before cognitive dysfunction.
Collapse
Affiliation(s)
- Shital Rani
- Department of Biophysics, Panjab University, Chandigarh, 160014, India
| | - Sudhrita Basu Dhar
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, 160014, India
| | - Akhil Khajuria
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, 160014, India
| | - Dikshi Gupta
- JoyScore Inc., 2440 Cerritos Ave, Signal Hill, CA, 90755, USA
| | - Pradeep Kumar Jaiswal
- Department of Biochemistry and Biophysics, Texas A & M University, College Station, TX, 77843, USA
| | - Neha Singla
- Department of Biophysics, Panjab University, Chandigarh, 160014, India
| | - Mandeep Kaur
- Department of Biophysics, Panjab University, Chandigarh, 160014, India.
| | - Gurpal Singh
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, 160014, India.
| | | |
Collapse
|
17
|
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: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [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
|
18
|
Ma XY, Yang TT, Liu L, Peng XC, Qian F, Tang FR. Ependyma in Neurodegenerative Diseases, Radiation-Induced Brain Injury and as a Therapeutic Target for Neurotrophic Factors. Biomolecules 2023; 13:754. [PMID: 37238624 PMCID: PMC10216700 DOI: 10.3390/biom13050754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/03/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
The neuron loss caused by the progressive damage to the nervous system is proposed to be the main pathogenesis of neurodegenerative diseases. Ependyma is a layer of ciliated ependymal cells that participates in the formation of the brain-cerebrospinal fluid barrier (BCB). It functions to promotes the circulation of cerebrospinal fluid (CSF) and the material exchange between CSF and brain interstitial fluid. Radiation-induced brain injury (RIBI) shows obvious impairments of the blood-brain barrier (BBB). In the neuroinflammatory processes after acute brain injury, a large amount of complement proteins and infiltrated immune cells are circulated in the CSF to resist brain damage and promote substance exchange through the BCB. However, as the protective barrier lining the brain ventricles, the ependyma is extremely vulnerable to cytotoxic and cytolytic immune responses. When the ependyma is damaged, the integrity of BCB is destroyed, and the CSF flow and material exchange is affected, leading to brain microenvironment imbalance, which plays a vital role in the pathogenesis of neurodegenerative diseases. Epidermal growth factor (EGF) and other neurotrophic factors promote the differentiation and maturation of ependymal cells to maintain the integrity of the ependyma and the activity of ependymal cilia, and may have therapeutic potential in restoring the homeostasis of the brain microenvironment after RIBI or during the pathogenesis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Xin-Yu Ma
- Department of Physiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Ting-Ting Yang
- Department of Physiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Lian Liu
- Department of Pharmacology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Xiao-Chun Peng
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Feng Qian
- Department of Physiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Feng-Ru Tang
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore 138602, Singapore
| |
Collapse
|
19
|
Yang Z, Cummings JL, Kinney JW, Cordes D, the Alzheimer’s Disease Neuroimaging Initiative. Accelerated hypometabolism with disease progression associated with faster cognitive decline among amyloid positive patients. Front Neurosci 2023; 17:1151820. [PMID: 37123373 PMCID: PMC10140339 DOI: 10.3389/fnins.2023.1151820] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 03/22/2023] [Indexed: 05/02/2023] Open
Abstract
Objective To evaluate the progression of brain glucose metabolism among participants with biological signature of Alzheimer's disease (AD) and its relevance to cognitive decline. Method We studied 602 amyloid positive individuals who underwent 18F-fluorodeoxyglucose PET (FDG-PET) scan, 18F-AV-45 amyloid PET (AV45-PET) scan, structural MRI scan and neuropsychological examination, including 116 cognitively normal (CN) participants, 314 participants diagnosed as mild cognitive impairment (MCI), and 172 participants diagnosed as AD dementia. The first FDG-PET scan satisfying the inclusion criteria was considered as the baseline scan. Cross-sectional analysis were conducted with the baseline FDG-PET data to compare the regional differences between diagnostic groups after adjusting confounding factors. Among these participants, 229 participants (55 CN, 139 MCI, and 35 AD dementia) had two-year follow-up FDG-PET data available. Regional glucose metabolism was computed and the progression rates of regional glucose metabolism were derived from longitudinal FDG-PET scans. Then the group differences of regional progression rates were examined to assess whether glucose metabolism deficit accelerates or becomes stable with disease progression. The association of cognitive decline rate with baseline regional glucose metabolism, and progression rate in longitudinal data, were evaluated. Results Participants with AD dementia showed substantial glucose metabolism deficit than CN and MCI at left hippocampus, in addition to the traditionally reported frontal and parietal-temporal lobe. More substantial metabolic change was observed with the contrast AD - MCI than the contrast MCI - CN, even after adjusting time duration since cognitive symptom onset. With the longitudinal data, glucose metabolism was observed to decline the most rapidly in the AD dementia group and at a slower rate in MCI. Lower regional glucose metabolism was correlated to faster cognitive decline rate with mild-moderate correlations, and the progression rate was correlated to cognitive decline rate with moderate-large correlations. Discussion and conclusion Hippocampus was identified to experience hypometabolism in AD pathology. Hypometabolism accelerates with disease progression toward AD dementia. FDG-PET, particularly longitudinal scans, could potentially help predict how fast cognition declines and assess the impact of treatment in interventional trials.
Collapse
Affiliation(s)
- Zhengshi Yang
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, United States
- Department of Brain Health, University of Nevada, Las Vegas, Las Vegas, NV, United States
| | - Jeffrey L. Cummings
- Department of Brain Health, University of Nevada, Las Vegas, Las Vegas, NV, United States
- Chambers-Grundy Center for Transformative Neuroscience, Pam Quirk Brain Health and Biomarker Laboratory, Department of Brain Health, School of Integrated Health Sciences, University of Nevada, Las Vegas, Las Vegas, NV, United States
| | - Jefferson W. Kinney
- Department of Brain Health, University of Nevada, Las Vegas, Las Vegas, NV, United States
- Chambers-Grundy Center for Transformative Neuroscience, Pam Quirk Brain Health and Biomarker Laboratory, Department of Brain Health, School of Integrated Health Sciences, University of Nevada, Las Vegas, Las Vegas, NV, United States
| | - Dietmar Cordes
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, United States
- Department of Brain Health, University of Nevada, Las Vegas, Las Vegas, NV, United States
- Department of Psychology and Neuroscience, University of Colorado Boulder, Boulder, CO, United States
| | | |
Collapse
|
20
|
Torok J, Anand C, Verma P, Raj A. Connectome-based biophysics models of Alzheimer's disease diagnosis and prognosis. Transl Res 2023; 254:13-23. [PMID: 36031051 PMCID: PMC11019890 DOI: 10.1016/j.trsl.2022.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/08/2022] [Indexed: 11/22/2022]
Abstract
With the increasing prevalence of Alzheimer's disease (AD) among aging populations and the limited therapeutic options available to slow or reverse its progression, the need has never been greater for improved diagnostic tools for identifying patients in the preclinical and prodomal phases of AD. Biophysics models of the connectome-based spread of amyloid-beta (Aβ) and microtubule-associated protein tau (τ) have enjoyed recent success as tools for predicting the time course of AD-related pathological changes. However, given the complex etiology of AD, which involves not only connectome-based spread of protein pathology but also the interactions of many molecular and cellular players over multiple spatiotemporal scales, more robust, complete biophysics models are needed to better understand AD pathophysiology and ultimately provide accurate patient-specific diagnoses and prognoses. Here we discuss several areas of active research in AD whose insights can be used to enhance the mathematical modeling of AD pathology as well as recent attempts at developing improved connectome-based biophysics models. These efforts toward a comprehensive yet parsimonious mathematical description of AD hold great promise for improving both the diagnosis of patients at risk for AD and our mechanistic understanding of how AD progresses.
Collapse
Affiliation(s)
- Justin Torok
- Department of Radiology, University of California, San Francisco, San Francisco, California.
| | - Chaitali Anand
- Department of Radiology, University of California, San Francisco, San Francisco, California
| | - Parul Verma
- Department of Radiology, University of California, San Francisco, San Francisco, California
| | - Ashish Raj
- Department of Radiology, University of California, San Francisco, San Francisco, California; Department of Bioengineering, University of California, Berkeley and University of California, San Francisco, Berkeley, California; Department of Radiology, Weill Cornell Medicine, New York, New York.
| |
Collapse
|
21
|
Marcisz A, for the Alzheimer’s Disease Neuroimaging Initiative, Polanska J. Can T1-Weighted Magnetic Resonance Imaging Significantly Improve Mini-Mental State Examination-Based Distinguishing Between Mild Cognitive Impairment and Early-Stage Alzheimer's Disease? J Alzheimers Dis 2023; 92:941-957. [PMID: 36806505 PMCID: PMC10116132 DOI: 10.3233/jad-220806] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2023] [Indexed: 02/19/2023]
Abstract
BACKGROUND Detecting early-stage Alzheimer's disease (AD) is still problematic in clinical practice. This work aimed to find T1-weighted MRI-based markers for AD and mild cognitive impairment (MCI) to improve the screening process. OBJECTIVE Our assumption was to build a screening model that would be accessible and easy to use for physicians in their daily clinical routine. METHODS The multinomial logistic regression was used to detect status: AD, MCI, and normal control (NC) combined with the Bayesian information criterion for model selection. Several T1-weighted MRI-based radiomic features were considered explanatory variables in the prediction model. RESULTS The best radiomic predictor was the relative brain volume. The proposed method confirmed its quality by achieving a balanced accuracy of 95.18%, AUC of 93.25%, NPV of 97.93%, and PPV of 90.48% for classifying AD versus NC for the European DTI Study on Dementia (EDSD). The comparison of the two models: with the MMSE score only as an independent variable and corrected for the relative brain value and age, shows that the addition of the T1-weighted MRI-based biomarker improves the quality of MCI detection (AUC: 67.04% versus 71.08%) while maintaining quality for AD (AUC: 93.35% versus 93.25%). Additionally, among MCI patients predicted as AD inconsistently with the original diagnosis, 60% from ADNI and 76.47% from EDSD were re-diagnosed as AD within a 48-month follow-up. It shows that our model can detect AD patients a few years earlier than a standard medical diagnosis. CONCLUSION The created method is non-invasive, inexpensive, clinically accessible, and efficiently supports AD/MCI screening.
Collapse
Affiliation(s)
- Anna Marcisz
- Department of Data Science and Engineering, The Silesian University of Technology, Gliwice, Poland
| | | | - Joanna Polanska
- Department of Data Science and Engineering, The Silesian University of Technology, Gliwice, Poland
| |
Collapse
|
22
|
Sheng C, Chu X, He Y, Ding Q, Jia S, Shi Q, Sun R, Song L, Du W, Liang Y, Chen N, Yang Y, Wang X. Alterations in Peripheral Metabolites as Key Actors in Alzheimer's Disease. Curr Alzheimer Res 2023; 20:379-393. [PMID: 37622711 DOI: 10.2174/1567205020666230825091147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 06/24/2023] [Accepted: 07/20/2023] [Indexed: 08/26/2023]
Abstract
Growing evidence supports that Alzheimer's disease (AD) could be regarded as a metabolic disease, accompanying central and peripheral metabolic disturbance. Nowadays, exploring novel and potentially alternative hallmarks for AD is needed. Peripheral metabolites based on blood and gut may provide new biochemical insights about disease mechanisms. These metabolites can influence brain energy homeostasis, maintain gut mucosal integrity, and regulate the host immune system, which may further play a key role in modulating the cognitive function and behavior of AD. Recently, metabolomics has been used to identify key AD-related metabolic changes and define metabolic changes during AD disease trajectory. This review aims to summarize the key blood- and microbial-derived metabolites that are altered in AD and identify the potential metabolic biomarkers of AD, which will provide future targets for precision therapeutic modulation.
Collapse
Affiliation(s)
- Can Sheng
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining, 272000, China
| | - Xu Chu
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining, 272000, China
| | - Yan He
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining, 272000, China
| | - Qingqing Ding
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining, 272000, China
| | - Shulei Jia
- Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Qiguang Shi
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining, 272000, China
| | - Ran Sun
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining, 272000, China
| | - Li Song
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining, 272000, China
| | - Wenying Du
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Yuan Liang
- Department of Clinical Medicine, Jining Medical University, Jining, 272067, China
| | - Nian Chen
- Department of Clinical Medicine, Jining Medical University, Jining, 272067, China
| | - Yan Yang
- Department of Neurology, The Affiliated Hospital of Jining Medical University, Jining, 272000, China
| | - Xiaoni Wang
- Department of Neurology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310020, China
| |
Collapse
|
23
|
Chiam K, Lee L, Kuo PH, Gaudet VC, Black SE, Zukotynski KA. Brain PET and Cerebrovascular Disease. PET Clin 2023; 18:115-122. [PMID: 36718716 DOI: 10.1016/j.cpet.2022.09.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Cerebrovascular disease encompasses a broad spectrum of diseases such as stroke, hemorrhage, and cognitive decline associated with vascular narrowing, obstruction, rupture, and inflammation, among other issues. Recent advances in hardware and software have led to improvements in brain PET. Although still in its infancy, machine learning using convolutional neural networks is gaining traction in this area, often with a focus on providing high-quality images with reduced noise using a shorter acquisition time or less radiation exposure for the patient.
Collapse
Affiliation(s)
- Katarina Chiam
- Division of Engineering Science, University of Toronto, 40 St. George St., Toronto, ON M5S 2E4, Canada
| | - Louis Lee
- Department of Electrical and Computer Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada
| | - Phillip H Kuo
- Departments of Medical Imaging, Medicine, Biomedical Engineering, University of Arizona, 1501 N. Campbell, Tucson, AZ 85724, USA
| | - Vincent C Gaudet
- Department of Electrical and Computer Engineering, University of Waterloo, 200 University Avenue West, Waterloo, ON N2L 3G1, Canada
| | - Sandra E Black
- Departments of Neurology, Sunnybrook Health Sciences Centre, University of Toronto, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada; Departments of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, 2075 Bayview Avenue, Toronto, ON M4N 3M5, Canada
| | - Katherine A Zukotynski
- Departments of Medicine and Radiology, McMaster University, 1200 Main Street West, Hamilton, ON L9G 4X5, Canada.
| |
Collapse
|
24
|
Rogojin A, Gorbet DJ, Hawkins KM, Sergio LE. Differences in resting state functional connectivity underlie visuomotor performance declines in older adults with a genetic risk (APOE ε4) for Alzheimer’s disease. Front Aging Neurosci 2022; 14:1054523. [DOI: 10.3389/fnagi.2022.1054523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/15/2022] [Indexed: 12/04/2022] Open
Abstract
IntroductionNon-standard visuomotor integration requires the interaction of large networks in the brain. Previous findings have shown that non-standard visuomotor performance is impaired in individuals with specific dementia risk factors (family history of dementia and presence of the APOE ε4 allele) in advance of any cognitive impairments. These findings suggest that visuomotor impairments are associated with early dementia-related brain changes. The current study examined the underlying resting state functional connectivity (RSFC) associated with impaired non-standard visuomotor performance, as well as the impacts of dementia family history, sex, and APOE status.MethodsCognitively healthy older adults (n = 48) were tested on four visuomotor tasks where reach and gaze were increasingly spatially dissociated. Participants who had a family history of dementia or the APOE ε4 allele were considered to be at an increased risk for AD. To quantify RSFC within networks of interest, an EPI sequence sensitive to BOLD contrast was collected. The networks of interest were the default mode network (DMN), somatomotor network (SMN), dorsal attention network (DAN), ventral attention network (VAN), and frontoparietal control network (FPN).ResultsIndividuals with the ε4 allele showed abnormalities in RSFC between posterior DMN nodes that predicted poorer non-standard visuomotor performance. Specifically, multiple linear regression analyses revealed lower RSFC between the precuneus/posterior cingulate cortex and the left inferior parietal lobule as well as the left parahippocampal cortex. Presence of the APOE ε4 allele also modified the relationship between mean DAN RSFC and visuomotor control, where lower mean RSFC in the DAN predicted worse non-standard visuomotor performance only in APOE ε4 carriers. There were otherwise no effects of family history, APOE ε4 status, or sex on the relationship between RSFC and visuomotor performance for any of the other resting networks.ConclusionThe preliminary findings provide insight into the impact of APOE ε4-related genetic risk on neural networks underlying complex visuomotor transformations, and demonstrate that the non-standard visuomotor task paradigm discussed in this study may be used as a non-invasive, easily accessible assessment tool for dementia risk.
Collapse
|
25
|
Yu H, Gao Y, He T, Li M, Zhang Y, Zheng J, Jiang B, Chen C, Ke D, Liu Y, Wang JZ. Discovering new peripheral plasma biomarkers to identify cognitive decline in type 2 diabetes. Front Cell Dev Biol 2022; 10:818141. [PMID: 36506101 PMCID: PMC9729784 DOI: 10.3389/fcell.2022.818141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 11/03/2022] [Indexed: 11/25/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) is an independent risk factor of Alzheimer's disease (AD), and thus identifying who among the increasing T2DM populations may develop into AD is important for early intervention. By using TMT-labeling coupled high-throughput mass spectrometry, we conducted a comprehensive plasma proteomic analysis in none-T2DM people (Ctrl, n = 30), and the age-/sex-matched T2DM patients with mild cognitive impairment (T2DM-MCI, n = 30) or T2DM without MCI (T2DM-nMCI, n = 25). The candidate biomarkers identified by proteomics and bioinformatics analyses were verified by ELISA, and their diagnostic capabilities were evaluated with machine learning. A total of 53 differentially expressed proteins (DEPs) were identified in T2DM-MCI compared with T2DM-nMCI patients. These DEPs were significantly enriched in multiple biological processes, such as amyloid neuropathies, CNS disorders, and metabolic acidosis. Among the DEPs, alpha-1-antitrypsin (SERPINA1), major viral protein (PRNP), and valosin-containing protein (VCP) showed strong correlation with AD high-risk genes APP, MAPT, APOE, PSEN1, and PSEN2. Also, the levels of PP2A cancer inhibitor (CIP2A), PRNP, corticotropin-releasing factor-binding protein (CRHBP) were significantly increased, while the level of VCP was decreased in T2DM-MCI patients compared with that of the T2DM-nMCI, and these changes were correlated with the Mini-Mental State Examination (MMSE) score. Further machine learning data showed that increases in PRNP, CRHBP, VCP, and rGSK-3β(T/S9) (ratio of total to serine-9-phosphorylated glycogen synthase kinase-3β) had the greatest power to identify mild cognitive decline in T2DM patients.
Collapse
Affiliation(s)
- Haitao Yu
- Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Yang Gao
- Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting He
- Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mengzhu Li
- Department of Neurosurgery, Wuhan Central Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yao Zhang
- Key Laboratory of Ministry of Education for Neurological Disorders, Li Yuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jie Zheng
- Key Laboratory of Basic Pharmacology of Ministry of Education, Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Key Laboratory of Basic Pharmacology of Guizhou Province, Department of Pharmacology, Zunyi Medical University, Zunyi, China
| | - Bijun Jiang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chongyang Chen
- Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dan Ke
- Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yanchao Liu
- Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jian-Zhi Wang
- Key Laboratory of Education Ministry of China/Hubei Province for Neurological Disorders, Department of Pathophysiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| |
Collapse
|
26
|
Kandiah N, Choi SH, Hu CJ, Ishii K, Kasuga K, Mok VC. Current and Future Trends in Biomarkers for the Early Detection of Alzheimer's Disease in Asia: Expert Opinion. J Alzheimers Dis Rep 2022; 6:699-710. [PMID: 36606209 PMCID: PMC9741748 DOI: 10.3233/adr-220059] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 10/23/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer's disease (AD) poses a substantial healthcare burden in the rapidly aging Asian population. Early diagnosis of AD, by means of biomarkers, can lead to interventions that might alter the course of the disease. The amyloid, tau, and neurodegeneration (AT[N]) framework, which classifies biomarkers by their core pathophysiological features, is a biomarker measure of amyloid plaques and neurofibrillary tangles. Our current AD biomarker armamentarium, comprising neuroimaging biomarkers and cerebrospinal fluid biomarkers, while clinically useful, may be invasive and expensive and hence not readily available to patients. Several studies have also investigated the use of blood-based measures of established core markers for detection of AD, such as amyloid-β and phosphorylated tau. Furthermore, novel non-invasive peripheral biomarkers and digital biomarkers could potentially expand access to early AD diagnosis to patients in Asia. Despite the multiplicity of established and potential biomarkers in AD, a regional framework for their optimal use to guide early AD diagnosis remains lacking. A group of experts from five regions in Asia gathered at a meeting in March 2021 to review the current evidence on biomarkers in AD diagnosis and discuss best practice around their use, with the goal of developing practical guidance that can be implemented easily by clinicians in Asia to support the early diagnosis of AD. This article summarizes recent key evidence on AD biomarkers and consolidates the experts' insights into the current and future use of these biomarkers for the screening and early diagnosis of AD in Asia.
Collapse
Affiliation(s)
- Nagaendran Kandiah
- Dementia Research Centre (Singapore), Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore,Correspondence to: Nagaendran Kandiah, Dementia Research Centre, Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore 308232. Tel.: +65 6592 2653; Fax: +65 6339 2889; E-mail: ; ORCID: 0000-0001-9244-4298
| | - Seong Hye Choi
- Department of Neurology, Inha University School of Medicine, Incheon, Republic of Korea
| | - Chaur-Jong Hu
- Department of Neurology, Dementia Center, Shuang Ho Hospital, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Kenji Ishii
- Team for Neuroimaging Research, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Kensaku Kasuga
- Department of Molecular Genetics, Center for Bioresources, Brain Research Institute, Niigata University, Niigata, Japan
| | - Vincent C.T. Mok
- Division of Neurology, Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China,Li Ka Shing Institute of Health Sciences, Gerald Choa Neuroscience Institute, Lui Che Woo Institute of Innovative Medicine, Therese Pei Fong Chow Research Centre for Prevention of Dementia, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
27
|
Correlation of Global and Regional Amyloid Burden by 18F-Florbetaben PET/CT With Cognitive Impairment Profile and Severity. Clin Nucl Med 2022; 47:923-930. [DOI: 10.1097/rlu.0000000000004370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
28
|
Nabizadeh F, Balabandian M, Rostami MR, Ward RT, Ahmadi N, Pourhamzeh M. Plasma p-tau181 associated with structural changes in mild cognitive impairment. Aging Clin Exp Res 2022; 34:2139-2147. [PMID: 35648357 DOI: 10.1007/s40520-022-02148-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 05/02/2022] [Indexed: 01/29/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease associated with dementia and is a serious concern for the health of individuals and government health care systems worldwide. Gray matter atrophy and white matter damage are major contributors to cognitive deficits in AD patients, as demonstrated by magnetic resonance imaging (MRI). Many of these brain changes associated with AD begin to occur about 15 years before the onset of initial clinical symptoms. Therefore, it is critical to find biomarkers reflective of these brain changes associated with AD to identify this disease and monitor its prognosis and development. The increased plasma level of hyperphosphorylated tau 181 (p-tau181) has been recently considered a novel biomarker for the diagnosis of AD, preclinical AD, and mild cognitive impairment (MCI). In the current study, we examined the association of cerebrospinal fluid (CSF) and plasma levels of p-tau181 with structural brain changes in cortical thickness, cortical volume, surface area, and subcortical volume in MCI patients. In this cross-sectional study, we included the information of 461 MCI patients from the Alzheimer's Disease Neuroimaging Initiative (ADNI) cohort. The results of voxel-wise partial correlation analyses showed a significant negative correlation between the increased levels of plasma p-tau181, CSF total tau, and CSF p-tau181 with structural changes in widespread brain regions. These results provide evidence for the use of plasma p-tau181 as a diagnostic marker for structural changes in the brain associated with the early stages of AD and neurodegeneration.
Collapse
Affiliation(s)
- Fardin Nabizadeh
- Neuroscience Research Group (NRG), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| | - Mohammad Balabandian
- Neuroscience Research Group (NRG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mohammad Reza Rostami
- Neuroscience Research Group (NRG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Richard T Ward
- Center for the Study of Emotion and Attention, University of Florida, Florida, USA
- Department of Psychology, University of Florida, Florida, USA
| | - Niloufar Ahmadi
- Student Research Committee, School of Nursing and Midwifery, Iran University of Medical Sciences, Tehran, Iran
| | - Mahsa Pourhamzeh
- Division of Neuroscience, Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
29
|
Zakharova NV, Bugrova AE, Indeykina MI, Fedorova YB, Kolykhalov IV, Gavrilova SI, Nikolaev EN, Kononikhin AS. Proteomic Markers and Early Prediction of Alzheimer's Disease. BIOCHEMISTRY. BIOKHIMIIA 2022; 87:762-776. [PMID: 36171657 DOI: 10.1134/s0006297922080089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 06/16/2023]
Abstract
Alzheimer's disease (AD) is the most common socially significant neurodegenerative pathology, which currently affects more than 30 million elderly people worldwide. Since the number of patients grows every year and may exceed 115 million by 2050, and due to the lack of effective therapies, early prediction of AD remains a global challenge, solution of which can contribute to the timely appointment of a preventive therapy in order to avoid irreversible changes in the brain. To date, clinical assays for the markers of amyloidosis in cerebrospinal fluid (CSF) have been developed, which, in conjunction with the brain MRI and PET studies, are used either to confirm the diagnosis based on obligate clinical criteria or to predict the risk of AD developing at the stage of mild cognitive impairment (MCI). However, the problem of predicting AD at the asymptomatic stage remains unresolved. In this regard, the search for new protein markers and studies of proteomic changes in CSF and blood plasma are of particular interest and may consequentially identify particular pathways involved in the pathogenesis of AD. Studies of specific proteomic changes in blood plasma deserve special attention and are of increasing interest due to the much less invasive method of sample collection as compared to CSF, which is important when choosing the object for large-scale screening. This review briefly summarizes the current knowledge on proteomic markers of AD and considers the prospects of developing reliable methods for early identification of AD risk factors based on the proteomic profile.
Collapse
Affiliation(s)
- Natalia V Zakharova
- Emanuel Institute for Biochemical Physics, Russian Academy of Sciences, Moscow, 119334, Russia.
| | - Anna E Bugrova
- Emanuel Institute for Biochemical Physics, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Maria I Indeykina
- Emanuel Institute for Biochemical Physics, Russian Academy of Sciences, Moscow, 119334, Russia
| | | | | | | | - Evgeny N Nikolaev
- Skolkovo Institute of Science and Technology, Moscow, 121205, Russia
| | | |
Collapse
|
30
|
Newberg AB, Coble R, Khosravi M, Alavi A. Positron Emission Tomography-Based Assessment of Cognitive Impairment and Dementias, Critical Role of Fluorodeoxyglucose in such Settings. PET Clin 2022; 17:479-494. [PMID: 35717103 DOI: 10.1016/j.cpet.2022.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Positron emission tomography (PET) has been a key component in the diagnostic armamentarium for assessing neurodegenerative diseases such as Alzheimer or Parkinson disease. PET imaging has been useful for diagnosing these disorders, identifying their pathophysiology, and following their treatment. Further, PET imaging has been extensively used for both clinical and research purposes, particularly for helping with potential therapeutic approaches for managing neurodegenerative diseases. This article will review the current literature regarding PET imaging in patients with neurodegenerative disorders. This includes an evaluation of the most commonly used tracer fluorodeoxyglucose that measures cerebral glucose metabolism, tracers that assess neurotransmitter systems, and tracers designed to reveal disease-specific pathophysiological processes. With the continuing development of an expanding variety of radiopharmaceuticals, PET imaging will likely play a prominent role in future research and clinical applications for neurodegenerative diseases.
Collapse
Affiliation(s)
- Andrew B Newberg
- Marcus Institute of Integrative Health, Thomas Jefferson University, 789 East Lancaster Avenue, Suite 110, Villanova, PA 19085, USA; Department of Radiology, Thomas Jefferson University, Philadelphia, PA, USA.
| | - Roger Coble
- Marcus Institute of Integrative Health, Thomas Jefferson University, 789 East Lancaster Avenue, Suite 110, Villanova, PA 19085, USA; University of California Berkeley, Berkeley, CA, USA
| | - Mohsen Khosravi
- Marcus Institute of Integrative Health, Thomas Jefferson University, 789 East Lancaster Avenue, Suite 110, Villanova, PA 19085, USA
| | - Abass Alavi
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
31
|
Saleem TJ, Zahra SR, Wu F, Alwakeel A, Alwakeel M, Jeribi F, Hijji M. Deep Learning-Based Diagnosis of Alzheimer's Disease. J Pers Med 2022; 12:815. [PMID: 35629237 PMCID: PMC9143671 DOI: 10.3390/jpm12050815] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/15/2022] [Accepted: 05/16/2022] [Indexed: 12/27/2022] Open
Abstract
Alzheimer's disease (AD), the most familiar type of dementia, is a severe concern in modern healthcare. Around 5.5 million people aged 65 and above have AD, and it is the sixth leading cause of mortality in the US. AD is an irreversible, degenerative brain disorder characterized by a loss of cognitive function and has no proven cure. Deep learning techniques have gained popularity in recent years, particularly in the domains of natural language processing and computer vision. Since 2014, these techniques have begun to achieve substantial consideration in AD diagnosis research, and the number of papers published in this arena is rising drastically. Deep learning techniques have been reported to be more accurate for AD diagnosis in comparison to conventional machine learning models. Motivated to explore the potential of deep learning in AD diagnosis, this study reviews the current state-of-the-art in AD diagnosis using deep learning. We summarize the most recent trends and findings using a thorough literature review. The study also explores the different biomarkers and datasets for AD diagnosis. Even though deep learning has shown promise in AD diagnosis, there are still several challenges that need to be addressed.
Collapse
Affiliation(s)
- Tausifa Jan Saleem
- Department of Computer Science and Engineering, National Institute of Technology Srinagar, Srinagar 190006, J&K, India; (T.J.S.); (S.R.Z.)
| | - Syed Rameem Zahra
- Department of Computer Science and Engineering, National Institute of Technology Srinagar, Srinagar 190006, J&K, India; (T.J.S.); (S.R.Z.)
| | - Fan Wu
- Department of Computer Science, Tuskegee University, Tuskegee, AL 36088, USA;
| | - Ahmed Alwakeel
- Sensor Network and Cellular Systems Research Center, University of Tabuk, Tabuk 71491, Saudi Arabia
- Faculty of Computers & Information Technology, University of Tabuk, Tabuk 71491, Saudi Arabia; (M.A.); (M.H.)
| | - Mohammed Alwakeel
- Faculty of Computers & Information Technology, University of Tabuk, Tabuk 71491, Saudi Arabia; (M.A.); (M.H.)
| | - Fathe Jeribi
- College of Computer Science and Information Technology, Jazan University, Jazan 45142, Saudi Arabia;
| | - Mohammad Hijji
- Faculty of Computers & Information Technology, University of Tabuk, Tabuk 71491, Saudi Arabia; (M.A.); (M.H.)
| |
Collapse
|
32
|
Kersting D, Settelmeier S, Mavroeidi IA, Herrmann K, Seifert R, Rischpler C. Shining Damaged Hearts: Immunotherapy-Related Cardiotoxicity in the Spotlight of Nuclear Cardiology. Int J Mol Sci 2022; 23:3802. [PMID: 35409161 PMCID: PMC8998973 DOI: 10.3390/ijms23073802] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 03/20/2022] [Accepted: 03/25/2022] [Indexed: 11/30/2022] Open
Abstract
The emerging use of immunotherapies in cancer treatment increases the risk of immunotherapy-related cardiotoxicity. In contrast to conventional chemotherapy, these novel therapies have expanded the forms and presentations of cardiovascular damage to a broad spectrum from asymptomatic changes to fulminant short- and long-term complications in terms of cardiomyopathy, arrythmia, and vascular disease. In cancer patients and, particularly, cancer patients undergoing (immune-)therapy, cardio-oncological monitoring is a complex interplay between pretherapeutic risk assessment, identification of impending cardiotoxicity, and post-therapeutic surveillance. For these purposes, the cardio-oncologist can revert to a broad spectrum of nuclear cardiological diagnostic workup. The most promising commonly used nuclear medicine imaging techniques in relation to immunotherapy will be discussed in this review article with a special focus on the continuous development of highly specific molecular markers and steadily improving methods of image generation. The review closes with an outlook on possible new developments of molecular imaging and advanced image evaluation techniques in this exciting and increasingly growing field of immunotherapy-related cardiotoxicity.
Collapse
Affiliation(s)
- David Kersting
- Department of Nuclear Medicine, University Hospital Essen, West German Cancer Center (WTZ), University of Duisburg-Essen, 45147 Essen, Germany; (K.H.); (R.S.); (C.R.)
- German Cancer Consortium (DKTK, Partner Site Essen/Düsseldorf), 45147 Essen, Germany;
| | - Stephan Settelmeier
- Department of Cardiology and Vascular Medicine, University Hospital Essen, West German Heart and Vascular Center, University of Duisburg-Essen, 45147 Essen, Germany;
| | - Ilektra-Antonia Mavroeidi
- German Cancer Consortium (DKTK, Partner Site Essen/Düsseldorf), 45147 Essen, Germany;
- Clinic for Internal Medicine (Tumor Research), University Hospital Essen, West German Cancer Center (WTZ), University of Duisburg-Essen, 45147 Essen, Germany
| | - Ken Herrmann
- Department of Nuclear Medicine, University Hospital Essen, West German Cancer Center (WTZ), University of Duisburg-Essen, 45147 Essen, Germany; (K.H.); (R.S.); (C.R.)
- German Cancer Consortium (DKTK, Partner Site Essen/Düsseldorf), 45147 Essen, Germany;
| | - Robert Seifert
- Department of Nuclear Medicine, University Hospital Essen, West German Cancer Center (WTZ), University of Duisburg-Essen, 45147 Essen, Germany; (K.H.); (R.S.); (C.R.)
- German Cancer Consortium (DKTK, Partner Site Essen/Düsseldorf), 45147 Essen, Germany;
| | - Christoph Rischpler
- Department of Nuclear Medicine, University Hospital Essen, West German Cancer Center (WTZ), University of Duisburg-Essen, 45147 Essen, Germany; (K.H.); (R.S.); (C.R.)
- German Cancer Consortium (DKTK, Partner Site Essen/Düsseldorf), 45147 Essen, Germany;
| |
Collapse
|
33
|
Raikes AC, Hernandez GD, Matthews DC, Lukic AS, Law M, Shi Y, Schneider LS, Brinton RD. Exploratory imaging outcomes of a phase 1b/2a clinical trial of allopregnanolone as a regenerative therapeutic for Alzheimer's disease: Structural effects and functional connectivity outcomes. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12258. [PMID: 35310526 PMCID: PMC8919249 DOI: 10.1002/trc2.12258] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 12/18/2021] [Accepted: 12/21/2021] [Indexed: 01/14/2023]
Abstract
Introduction Allopregnanolone (ALLO), an endogenous neurosteroid, promoted neurogenesis and oligogenesis and restored cognitive function in animal models of Alzheimer's disease (AD). Based on these discovery research findings, we conducted a randomized-controlled phase 1b/2a multiple ascending dose trial of ALLO in persons with early AD (NCT02221622) to assess safety, tolerability, and pharmacokinetics. Exploratory imaging outcomes to determine whether ALLO impacted hippocampal structure, white matter integrity, and functional connectivity are reported. Methods Twenty-four individuals participated in the trial (n = 6 placebo; n = 18 ALLO) and underwent brain magnetic resonance imaging (MRI) before and after 12 weeks of treatment. Hippocampal atrophy rate was determined from volumetric MRI, computed as rate of change, and qualitatively assessed between ALLO and placebo sex, apolipoprotein E (APOE) ε4 allele, and ALLO dose subgroups. White matter microstructural integrity was compared between placebo and ALLO using fractional and quantitative anisotropy (QA). Changes in local, inter-regional, and network-level functional connectivity were also compared between groups using resting-state functional MRI. Results Rate of decline in hippocampal volume was slowed, and in some cases reversed, in the ALLO group compared to placebo. Gain of hippocampal volume was evident in APOE ε4 carriers (range: 0.6% to 7.8% increased hippocampal volume). Multiple measures of white matter integrity indicated evidence of preserved or improved integrity. ALLO significantly increased fractional anisotropy (FA) in 690 of 690 and QA in 1416 of 1888 fiber tracts, located primarily in the corpus callosum, bilateral thalamic radiations, and bilateral corticospinal tracts. Consistent with structural changes, ALLO strengthened local, inter-regional, and network level functional connectivity in AD-vulnerable regions, including the precuneus and posterior cingulate, and network connections between the default mode network and limbic system. Discussion Indicators of regeneration from previous preclinical studies and these exploratory MRI-based outcomes from this phase 1b/2a clinical cohort support advancement to a phase 2 proof-of-concept efficacy clinical trial of ALLO as a regenerative therapeutic for mild AD (REGEN-BRAIN study; NCT04838301).
Collapse
Affiliation(s)
- Adam C. Raikes
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
| | | | - Dawn C. Matthews
- Departments of Pharmacology and Neurology, College of MedicineADM DiagnosticsNorthbrookIllinoisUSA
| | - Ana S. Lukic
- Departments of Pharmacology and Neurology, College of MedicineADM DiagnosticsNorthbrookIllinoisUSA
| | - Meng Law
- Department of RadiologyAlfred HealthDepartment of Neuroscience and Computer Systems EngineeringMonash UniversityMelbourneAustralia
| | - Yonggang Shi
- Stevens Neuroimaging and Informatics InstituteKeck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Lon S. Schneider
- Keck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Roberta D. Brinton
- Center for Innovation in Brain ScienceUniversity of ArizonaTucsonArizonaUSA
| |
Collapse
|
34
|
Duggan MR, Lu A, Foster TC, Wimmer M, Parikh V. Exosomes in Age-Related Cognitive Decline: Mechanistic Insights and Improving Outcomes. Front Aging Neurosci 2022; 14:834775. [PMID: 35299946 PMCID: PMC8921862 DOI: 10.3389/fnagi.2022.834775] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 02/09/2022] [Indexed: 02/01/2023] Open
Abstract
Aging is the most prominent risk factor for cognitive decline, yet behavioral symptomology and underlying neurobiology can vary between individuals. Certain individuals exhibit significant age-related cognitive impairments, while others maintain intact cognitive functioning with only minimal decline. Recent developments in genomic, proteomic, and functional imaging approaches have provided insights into the molecular and cellular substrates of cognitive decline in age-related neuropathologies. Despite the emergence of novel tools, accurately and reliably predicting longitudinal cognitive trajectories and improving functional outcomes for the elderly remains a major challenge. One promising approach has been the use of exosomes, a subgroup of extracellular vesicles that regulate intercellular communication and are easily accessible compared to other approaches. In the current review, we highlight recent findings which illustrate how the analysis of exosomes can improve our understanding of the underlying neurobiological mechanisms that contribute to cognitive variation in aging. Specifically, we focus on exosome-mediated regulation of miRNAs, neuroinflammation, and aggregate-prone proteins. In addition, we discuss how exosomes might be used to enhance individual patient outcomes by serving as reliable biomarkers of cognitive decline and as nanocarriers to deliver therapeutic agents to the brain in neurodegenerative conditions.
Collapse
Affiliation(s)
- Michael R. Duggan
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, United States
| | - Anne Lu
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, United States
| | - Thomas C. Foster
- Department of Neuroscience, University of Florida College of Medicine, Gainesville, FL, United States
| | - Mathieu Wimmer
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, United States
| | - Vinay Parikh
- Department of Psychology and Neuroscience Program, Temple University, Philadelphia, PA, United States
| |
Collapse
|
35
|
Emsell L, Vanhaute H, Vansteelandt K, De Winter FL, Christiaens D, Van den Stock J, Vandenberghe R, Van Laere K, Sunaert S, Bouckaert F, Vandenbulcke M. An optimized MRI and PET based clinical protocol for improving the differential diagnosis of geriatric depression and Alzheimer's disease. Psychiatry Res Neuroimaging 2022; 320:111443. [PMID: 35091333 DOI: 10.1016/j.pscychresns.2022.111443] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 09/28/2021] [Accepted: 01/12/2022] [Indexed: 11/16/2022]
Abstract
Amyloid positron emission tomography (PET) and hippocampal volume derived from magnetic resonance imaging may be useful clinical biomarkers for differentiating between geriatric depression and Alzheimer's disease (AD). Here we investigated the incremental value of using hippocampal volume and 18F-flutemetmol amyloid PET measures in tandem and sequentially to improve discrimination in unclassified participants. Two approaches were compared in 41 participants with geriatric depression and 27 participants with probable AD: (1) amyloid and hippocampal volume combined in one model and (2) classification based on hippocampal volume first and then subsequent stratification using standardized uptake value ratio (SUVR)-determined amyloid positivity. Hippocampal volume and amyloid SUVR were significant diagnostic predictors of depression (sensitivity: 95%, specificity: 89%). 51% of participants were correctly classified according to clinical diagnosis based on hippocampal volume alone, increasing to 87% when adding amyloid data (sensitivity: 94%, specificity: 78%). Our results suggest that hippocampal volume may be a useful gatekeeper for identifying depressed individuals at risk for AD who would benefit from additional amyloid biomarkers when available.
Collapse
Affiliation(s)
- Louise Emsell
- Geriatric Psychiatry, UPC KU Leuven, Leuven, Belgium; Department of Neurosciences, Neuropsychiatry, KU Leuven, Leuven, Belgium; Department of Imaging & Pathology, Translational MRI, Medical Imaging Research Center, KU Leuven, UZ Leuven (Gasthuisberg), Leuven 3000, Belgium.
| | - Heleen Vanhaute
- Geriatric Psychiatry, UPC KU Leuven, Leuven, Belgium; Department of Imaging & Pathology, Nuclear Medicine and Molecular Imaging, KU Leuven, Leuven, Belgium; Nuclear Medicine, University Hospitals Leuven, Herestraat 49, Leuven 3000, Belgium
| | - Kristof Vansteelandt
- Geriatric Psychiatry, UPC KU Leuven, Leuven, Belgium; Department of Neurosciences, Neuropsychiatry, KU Leuven, Leuven, Belgium.
| | - François-Laurent De Winter
- Geriatric Psychiatry, UPC KU Leuven, Leuven, Belgium; Department of Neurosciences, Neuropsychiatry, KU Leuven, Leuven, Belgium.
| | - Danny Christiaens
- Department of Neurosciences, Neuropsychiatry, KU Leuven, Leuven, Belgium
| | - Jan Van den Stock
- Geriatric Psychiatry, UPC KU Leuven, Leuven, Belgium; Department of Neurosciences, Neuropsychiatry, KU Leuven, Leuven, Belgium.
| | - Rik Vandenberghe
- Department of Neurosciences, Laboratory for Cognitive Neurology, KU Leuven, Leuven, Belgium.
| | - Koen Van Laere
- Department of Imaging & Pathology, Nuclear Medicine and Molecular Imaging, KU Leuven, Leuven, Belgium; Nuclear Medicine, University Hospitals Leuven, Herestraat 49, Leuven 3000, Belgium.
| | - Stefan Sunaert
- Department of Imaging & Pathology, Translational MRI, Medical Imaging Research Center, KU Leuven, UZ Leuven (Gasthuisberg), Leuven 3000, Belgium; Department of Radiology, University Hospitals Leuven, Herestraat 49, Leuven 3000, Belgium.
| | - Filip Bouckaert
- Geriatric Psychiatry, UPC KU Leuven, Leuven, Belgium; Department of Neurosciences, Neuropsychiatry, KU Leuven, Leuven, Belgium.
| | - Mathieu Vandenbulcke
- Geriatric Psychiatry, UPC KU Leuven, Leuven, Belgium; Department of Neurosciences, Neuropsychiatry, KU Leuven, Leuven, Belgium.
| |
Collapse
|
36
|
Beheshti I, Geddert N, Perron J, Gupta V, Albensi BC, Ko JH, for the Alzheimer’s Disease Neuroimaging Initiative. Monitoring Alzheimer's Disease Progression in Mild Cognitive Impairment Stage Using Machine Learning-Based FDG-PET Classification Methods. J Alzheimers Dis 2022; 89:1493-1502. [PMID: 36057825 PMCID: PMC9661333 DOI: 10.3233/jad-220585] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/02/2022] [Indexed: 01/18/2023]
Abstract
BACKGROUND We previously introduced a machine learning-based Alzheimer's Disease Designation (MAD) framework for identifying AD-related metabolic patterns among neurodegenerative subjects. OBJECTIVE We sought to assess the efficiency of our MAD framework for tracing the longitudinal brain metabolic changes in the prodromal stage of AD. METHODS MAD produces subject scores using five different machine-learning algorithms, which include a general linear model (GLM), two different approaches of scaled subprofile modeling, and two different approaches of a support vector machine. We used our pre-trained MAD framework, which was trained based on metabolic brain features of 94 patients with AD and 111 age-matched cognitively healthy (CH) individuals. The MAD framework was applied on longitudinal independent test sets including 54 CHs, 51 stable mild cognitive impairment (sMCI), and 39 prodromal AD (pAD) patients at the time of the clinical diagnosis of AD, and two years prior. RESULTS The GLM showed excellent performance with area under curve (AUC) of 0.96 in distinguishing sMCI from pAD patients at two years prior to the time of the clinical diagnosis of AD while other methods showed moderate performance (AUC: 0.7-0.8). Significant annual increment of MAD scores were identified using all five algorithms in pAD especially when it got closer to the time of diagnosis (p < 0.001), but not in sMCI. The increased MAD scores were also significantly associated with cognitive decline measured by Mini-Mental State Examination in pAD (q < 0.01). CONCLUSION These results suggest that MAD may be a relevant tool for monitoring disease progression in the prodromal stage of AD.
Collapse
Affiliation(s)
- Iman Beheshti
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Science Centre, Winnipeg, MB, Canada
| | - Natasha Geddert
- Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Science Centre, Winnipeg, MB, Canada
- St. Boniface Hospital Research, Winnipeg, MB, Canada
| | - Jarrad Perron
- Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Science Centre, Winnipeg, MB, Canada
- Graduate Program in Biomedical Engineering, Price Faculty of Engineering, University of Manitoba, Winnipeg, MB, Canada
| | - Vinay Gupta
- Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Science Centre, Winnipeg, MB, Canada
- Graduate Program in Biomedical Engineering, Price Faculty of Engineering, University of Manitoba, Winnipeg, MB, Canada
| | - Benedict C. Albensi
- Graduate Program in Biomedical Engineering, Price Faculty of Engineering, University of Manitoba, Winnipeg, MB, Canada
- Department of Pharmacology and Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- St. Boniface Hospital Research, Winnipeg, MB, Canada
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Ft. Lauderdale, FL, USA
| | - Ji Hyun Ko
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Science Centre, Winnipeg, MB, Canada
- Graduate Program in Biomedical Engineering, Price Faculty of Engineering, University of Manitoba, Winnipeg, MB, Canada
| | | |
Collapse
|
37
|
Ventoulis I, Arfaras-Melainis A, Parissis J, Polyzogopoulou E. Cognitive Impairment in Acute Heart Failure: Narrative Review. J Cardiovasc Dev Dis 2021; 8:jcdd8120184. [PMID: 34940539 PMCID: PMC8703678 DOI: 10.3390/jcdd8120184] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 12/11/2021] [Accepted: 12/12/2021] [Indexed: 12/03/2022] Open
Abstract
Cognitive impairment (CI) represents a common but often veiled comorbidity in patients with acute heart failure (AHF) that deserves more clinical attention. In the AHF setting, it manifests as varying degrees of deficits in one or more cognitive domains across a wide spectrum ranging from mild CI to severe global neurocognitive disorder. On the basis of the significant negative implications of CI on quality of life and its overwhelming association with poor outcomes, there is a compelling need for establishment of detailed consensus guidelines on cognitive screening methods to be systematically implemented in the population of patients with heart failure (HF). Since limited attention has been drawn exclusively on the field of CI in AHF thus far, the present narrative review aims to shed further light on the topic. The underlying pathophysiological mechanisms of CI in AHF remain poorly understood and seem to be multifactorial. Different pathophysiological pathways may come into play, depending on the clinical phenotype of AHF. There is some evidence that cognitive decline closely follows the perturbations incurred across the long-term disease trajectory of HF, both along the time course of stable chronic HF as well as during episodes of HF exacerbation. CI in AHF remains a rather under recognized scientific field that poses many challenges, since there are still many unresolved issues regarding cognitive changes in patients hospitalized with AHF that need to be thoroughly addressed.
Collapse
Affiliation(s)
- Ioannis Ventoulis
- Department of Occupational Therapy, University of Western Macedonia, 50200 Ptolemaida, Greece
- Correspondence: or (I.V.); (A.A.-M.); Tel.: +30-6973018788 (I.V.); +1-347-920-8875 (A.A.-M.)
| | - Angelos Arfaras-Melainis
- Heart Failure Unit and University Clinic of Emergency Medicine, Attikon University Hospital, National and Kapodistrian University of Athens Medical School, 12462 Athens, Greece; (J.P.); (E.P.)
- Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Correspondence: or (I.V.); (A.A.-M.); Tel.: +30-6973018788 (I.V.); +1-347-920-8875 (A.A.-M.)
| | - John Parissis
- Heart Failure Unit and University Clinic of Emergency Medicine, Attikon University Hospital, National and Kapodistrian University of Athens Medical School, 12462 Athens, Greece; (J.P.); (E.P.)
| | - Eftihia Polyzogopoulou
- Heart Failure Unit and University Clinic of Emergency Medicine, Attikon University Hospital, National and Kapodistrian University of Athens Medical School, 12462 Athens, Greece; (J.P.); (E.P.)
| |
Collapse
|
38
|
Andersen E, Casteigne B, Chapman WD, Creed A, Foster F, Lapins A, Shatz R, Sawyer RP. Diagnostic biomarkers in Alzheimer’s disease. Biomark Neuropsychiatry 2021. [DOI: 10.1016/j.bionps.2021.100041] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
|
39
|
Wang F, Wang F, Chen T. Secondary metabolites of Galactomyces geotrichum from Laminaria japonica ameliorate cognitive deficits and brain oxidative stress in D-galactose induced Alzheimer's disease mouse model. Nat Prod Res 2021; 35:5323-5328. [PMID: 32292060 DOI: 10.1080/14786419.2020.1753738] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/02/2020] [Accepted: 04/04/2020] [Indexed: 01/09/2023]
Abstract
Accumulating evidences have shown the beneficial effects of natural products for Alzheimer's disease (AD) treatment. The present study was designed to investigate the neuroprotective effects of secondary metabolites of Galactomyces geotrichum (SMGG) on D-galactose induced AD mice. SMGG was extracted and its toxicological evaluation was conducted. To explore the neuroprotective mechanism responsible for anti-AD activity of SMGG, spatial learning and memory behavioral, oxidative stress levels, acetylcholinesterase and choline acetyltransferase activity assays were employed. The AD mice received SMGG treatment exhibited significant improvement in cognitive performance, enhanced antioxidant capacity, decreased acetylcholinesterase activity and increased choline acetyltransferase activity. Meanwhile, SMGG had no toxicity and seven compounds were separated from it: 7,8-dimethyl-iso-alloxazine, 1-methyl-3-benzyl-6-(4-hydroxybenzyl)-2,5-piperzainedione, cyclo-(Phe-Pro), cyclo-(Leu-Pro), cyclo-(Pro-Gly), cyclo-(Gly-Leu) and uracil, respectively. Overall, these data suggested that SMGG protects the brain against D-galactose induced cognitive impairment, oxidative damages and acetylcholine content decrease in AD mice.
Collapse
Affiliation(s)
- Fengwu Wang
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao, China
| | - Feng Wang
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao, China
| | - Tiejun Chen
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao, China
| |
Collapse
|
40
|
Garcia-Romeu A, Darcy S, Jackson H, White T, Rosenberg P. Psychedelics as Novel Therapeutics in Alzheimer's Disease: Rationale and Potential Mechanisms. Curr Top Behav Neurosci 2021; 56:287-317. [PMID: 34734390 DOI: 10.1007/7854_2021_267] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Serotonin 2A receptor (5-HT2AR) agonist "classic psychedelics" are drawing increasing interest as potential mental health treatments. Recent work suggests psychedelics can exert persisting anxiolytic and antidepressant effects lasting up to several months after a single administration. Data indicate acute subjective drug effects as important psychological factors involved in observed therapeutic benefits. Additionally, animal models have shown an important role for 5-HT2AR agonists in modulating learning and memory function with relevance for Alzheimer's Disease (AD) and related dementias. A number of biological mechanisms of action are under investigation to elucidate 5-HT2AR agonists' therapeutic potential, including enhanced neuroplasticity, anti-inflammatory effects, and alterations in brain functional connectivity. These diverse lines of research are reviewed here along with a discussion of AD pathophysiology and neuropsychiatric symptoms to highlight classic psychedelics as potential novel pharmacotherapies for patients with AD. Human clinical research suggests a possible role for high-dose psychedelic administration in symptomatic treatment of depressed mood and anxiety in early-stage AD. Preclinical data indicate a potential for low- or high-dose psychedelic treatment regimens to slow or reverse brain atrophy, enhance cognitive function, and slow progression of AD. In conclusion, rationale and potential approaches for preliminary research with psychedelics in patients with AD are presented, and ramifications of this line of investigation for development of novel AD treatments are discussed.
Collapse
Affiliation(s)
- Albert Garcia-Romeu
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Center for Psychedelic and Consciousness Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Sean Darcy
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Psychedelic and Consciousness Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hillary Jackson
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Center for Psychedelic and Consciousness Research, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Toni White
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Memory and Alzheimer's Treatment Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Paul Rosenberg
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Memory and Alzheimer's Treatment Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
41
|
Tataryn NM, Singh V, Dyke JP, Berk-Rauch HE, Clausen DM, Aronowitz E, Norris EH, Strickland S, Ahn HJ. Vascular endothelial growth factor associated dissimilar cerebrovascular phenotypes in two different mouse models of Alzheimer's Disease. Neurobiol Aging 2021; 107:96-108. [PMID: 34416494 PMCID: PMC8595520 DOI: 10.1016/j.neurobiolaging.2021.07.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 07/15/2021] [Accepted: 07/20/2021] [Indexed: 01/14/2023]
Abstract
Vascular perturbations and cerebral hypometabolism are emerging as important components of Alzheimer's disease (AD). While various in vivo imaging modalities have been designed to detect changes of cerebral perfusion and metabolism in AD patients and animal models, study results were often heterogenous with respect to imaging techniques and animal models. We therefore evaluated cerebral perfusion and glucose metabolism of two popular transgenic AD mouse strains, TgCRND8 and 5xFAD, at 7 and 12 months-of-age under identical conditions and analyzed possible molecular mechanisms underlying heterogeneous cerebrovascular phenotypes. Results revealed disparate findings in these two strains, displaying important aspects of AD progression. TgCRND8 mice showed significantly decreased cerebral blood flow and glucose metabolism with unchanged cerebral blood volume (CBV) at 12 months-of-age whereas 5xFAD mice showed unaltered glucose metabolism with significant increase in CBV at 12 months-of-age and a biphasic pattern of early hypoperfusion followed by a rebound to normal cerebral blood flow in late disease. Finally, immunoblotting assays suggested that VEGF dependent vascular tone change may restore normoperfusion and increase CBV in 5xFAD.
Collapse
Affiliation(s)
- Nicholas M Tataryn
- Tri-Institutional Training Program in Laboratory Animal Medicine and Science, Memorial Sloan Kettering Cancer Center, Weill Cornell Medicine, and The Rockefeller University, New York, New York, USA and Center for Comparative Medicine and Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, Rockefeller University, New York, NY, USA; Division of Comparative Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Vishal Singh
- Department of Pharmacology, Physiology and Neurosciences, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Jonathan P Dyke
- Citigroup Biomedical Imaging Center, Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Hanna E Berk-Rauch
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, Rockefeller University, New York, NY, USA
| | - Dana M Clausen
- Department of Pharmacology, Physiology and Neurosciences, Rutgers-New Jersey Medical School, Newark, NJ, USA
| | - Eric Aronowitz
- Citigroup Biomedical Imaging Center, Department of Radiology, Weill Cornell Medicine, New York, NY, USA
| | - Erin H Norris
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, Rockefeller University, New York, NY, USA
| | - Sidney Strickland
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, Rockefeller University, New York, NY, USA
| | - Hyung Jin Ahn
- Department of Pharmacology, Physiology and Neurosciences, Rutgers-New Jersey Medical School, Newark, NJ, USA; Brain Health Institute, Rutgers University, Piscataway, NJ, USA.
| |
Collapse
|
42
|
Park S, Kim Y. Bias-generating factors in biofluid amyloid-β measurements for Alzheimer's disease diagnosis. Biomed Eng Lett 2021; 11:287-295. [PMID: 34616582 DOI: 10.1007/s13534-021-00201-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/05/2021] [Accepted: 08/08/2021] [Indexed: 01/03/2023] Open
Abstract
Alzheimer's disease (AD) is the most prevalent cause of dementia worldwide, yet the dearth of readily accessible diagnostic biomarkers is a substantial hindrance towards progressing to effective preventive and therapeutic approaches. Due to a long delay between cerebral amyloid-β (Aβ) accumulation and the onset of cognitive impairments, biomarkers that reflect Aβ pathology and enable routine screening for disease progression are of urgent need for application in the clinical diagnosis of AD. According to accumulating evidences, cerebrospinal fluid (CSF) and plasma offer windows to the brain as they allow monitoring of biochemical changes in the brain. Considering the high availability and accuracy in depicting Aβ deposition in the brain, Aβ levels in CSF and plasma are regarded as promising fluid biomarkers for the diagnosis of AD patients at an early stage. However, clinical data with intra- and interindividual variations in the concentrations of CSF and plasma Aβ implicate the need to reevaluate current Aβ detection methods and establish a standardized operating procedure. Therefore, this review introduces three bias-generating factors in biofluid Aβ measurement that may hamper the accurate Aβ quantification and how such complications can be overcome for the widespread implementation of fluid Aβ detection in clinical practice.
Collapse
Affiliation(s)
- Sohui Park
- Department of Pharmacy, Department of Integrative Biotechnology and Translational Medicine, and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, 21983 Republic of Korea
| | - YoungSoo Kim
- Department of Pharmacy, Department of Integrative Biotechnology and Translational Medicine, and Yonsei Institute of Pharmaceutical Sciences, College of Pharmacy, Yonsei University, Incheon, 21983 Republic of Korea
| |
Collapse
|
43
|
Lee ES, Youn H, Hyung WSW, Suh S, Han CE, Eo JS, Jeong HG. The effects of cerebral amyloidopathy on regional glucose metabolism in older adults with depression and mild cognitive impairment while performing memory tasks. Eur J Neurosci 2021; 54:6663-6672. [PMID: 34528336 DOI: 10.1111/ejn.15461] [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: 06/22/2021] [Accepted: 08/31/2021] [Indexed: 11/28/2022]
Abstract
Co-occurring depression and mild cognitive impairment (MCI) in older adults are important because they have a high risk of conversion to dementia. In the present study, task-related F-18 fluorodeoxyglucose positron emission tomography (FDG-PET) was used to analyse older adults with concomitant depression and MCI. We recruited 20 older adults with simultaneous depression and MCI and 10 older adults with normal cognition (NC). The Verbal Paired Associates test and digit span test were used for the task-related FDG-PET. The 20 older adults with depression and MCI were classified into two groups based on the F-18 florbetaben PET results: depressed MCI patients with (LLD-MCI-A[+]; n = 11) and without amyloid accumulation (LLD-MCI-A[-]; n = 9). Reduced regional cerebral glucose metabolism (rCMglc) in the left superior frontal region was observed in the LLD-MCI-A(-) group compared with the NC group. Analyses of the NC and LLD-MCI-A(+) groups showed significantly decreased rCMglc in the right inferior parietal and left middle frontal regions in the LLD-MCI-A(+) group. rCMglc in the left precuneus was lower in the LLD-MCI-A(+) group than in the LLD-MCI-A(-) group. Significant correlations between the rCMglc in the right inferior parietal/left precuneus regions and memory task scores were observed based on correlation analyses of NC and LLD-MCI-A(+) groups. The findings in the present study indicate the presence of amyloid accumulation influences glucose metabolism in depressed elderly subjects with MCI while performing cognitive tasks. Task-related FDG-PET examinations may help differentiate MCI associated with depression from comorbid depression in patients with prodromal Alzheimer's disease.
Collapse
Affiliation(s)
- Eun Seong Lee
- Department of Nuclear Medicine, Korea University Guro Hospital, Seoul, South Korea
| | - HyunChul Youn
- Department of Psychiatry, Soonchunhyang University Bucheon Hospital, Bucheon, South Korea
| | | | - Sangil Suh
- Department of Radiology, Korea University Guro Hospital, Seoul, South Korea
| | - Cheol E Han
- Department of Electronics and Information Engineering, Korea University, Sejong, South Korea
| | - Jae Seon Eo
- Department of Nuclear Medicine, Korea University Guro Hospital, Seoul, South Korea
| | - Hyun-Ghang Jeong
- Department of Psychiatry, Korea University Guro Hospital, Seoul, South Korea.,Korea University Research Institute of Mental Health, Seoul, South Korea
| |
Collapse
|
44
|
Li K, Fu Z, Qi S, Luo X, Zeng Q, Xu X, Huang P, Zhang M, Calhoun VD. Polygenic Hazard Score Associated Multimodal Brain Networks Along the Alzheimer's Disease Continuum. Front Aging Neurosci 2021; 13:725246. [PMID: 34539385 PMCID: PMC8446666 DOI: 10.3389/fnagi.2021.725246] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/10/2021] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Late-onset Alzheimer's disease (AD) is a polygenic neurodegenerative disease. Identifying the neuroimaging phenotypes behind the genetic predisposition of AD is critical to the understanding of AD pathogenesis. Two major questions which previous studies have led to are: (1) should the general "polygenic hazard score" (PHS) be a good choice to identify the individual genetic risk for AD; and (2) should researchers also include inter-modality relationships in the analyses considering these may provide complementary information about the AD etiology. METHODS We collected 88 healthy controls, 77 patients with mild cognitive impairment (MCI), and 22 AD patients to simulate the AD continuum included from the ADNI database. PHS-guided multimodal fusion was used to investigate the impact of PHS on multimodal brain networks in AD-continuum by maximizing both inter-modality association and reference-modality correlation. Fractional amplitude of low frequency fluctuations, gray matter (GM) volume, and amyloid standard uptake value ratios were included as neuroimaging features. Eventually, the changes in neuroimaging features along AD continuum were investigated, and relationships between cognitive performance and identified PHS associated multimodal components were established. RESULTS We found that PHS was associated with multimodal brain networks, which showed different functional and structural impairments under increased amyloid deposits. Notably, along with AD progression, functional impairment occurred before GM atrophy, amyloid deposition started from the MCI stage and progressively increased throughout the disease continuum. CONCLUSION PHS is associated with multi-facets of brain impairments along the AD continuum, including cognitive dysfunction, pathological deposition, which might underpin the AD pathogenesis.
Collapse
Affiliation(s)
- Kaicheng Li
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia Institute of Technology, Georgia State University, Emory University, Atlanta, GA, United States
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Zening Fu
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia Institute of Technology, Georgia State University, Emory University, Atlanta, GA, United States
| | - Shile Qi
- Department of Computer Science and Engineering, Nanjing University of Aeronautics and Astronautics, Nanjing, China
| | - Xiao Luo
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Qingze Zeng
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaopei Xu
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Peiyu Huang
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Minming Zhang
- Department of Radiology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Vince D. Calhoun
- Tri-Institutional Center for Translational Research in Neuroimaging and Data Science (TReNDS), Georgia Institute of Technology, Georgia State University, Emory University, Atlanta, GA, United States
- Department of Psychology, Computer Science, Neuroscience Institute, and Physics, Georgia State University, Atlanta, GA, United States
- Department of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA, United States
| |
Collapse
|
45
|
Lee YS, Youn H, Jeong HG, Lee TJ, Han JW, Park JH, Kim KW. Cost-effectiveness of using amyloid positron emission tomography in individuals with mild cognitive impairment. COST EFFECTIVENESS AND RESOURCE ALLOCATION 2021; 19:50. [PMID: 34391439 PMCID: PMC8364075 DOI: 10.1186/s12962-021-00300-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Accepted: 07/23/2021] [Indexed: 12/02/2022] Open
Abstract
Background Amyloid positron emission tomography (PET) makes it possible to diagnose Alzheimer’s disease (AD) in its prodromal phase including mild cognitive impairment (MCI). This study evaluated the cost-effectiveness of including amyloid-PET for assessing individuals with MCI. Methods The target population was 60-year-old patients who were diagnosed with MCI. We constructed a Markov model for the natural history of AD with the amyloid positivity (AP). Because amyloid-PET can detect the AP MCI state, AD detection can be made faster by reducing the follow-up interval for a high-risk group. The health outcomes were evaluated in quality-adjusted life years (QALYs) and the final results of cost-effectiveness analysis were presented in the form of the Incremental Cost-Effectiveness Ratio (ICER). To handle parameter uncertainties, one-way sensitivity analyses for various variables were performed. Results Our model showed that amyloid-PET increased QALYs by 0.003 in individuals with MCI. The estimated additional costs for adopting amyloid-PET amounted to a total of 1250 USD per patient when compared with the cost when amyloid-PET is not adopted. The ICER was 3,71,545 USD per QALY. According to the sensitivity analyses, treatment effect of Donepezil and virtual intervention effect in MCI state were the most influential factors. Conclusions In our model, using amyloid-PET at the MCI stage was not cost-effective. Future advances in management of cognitive impairment would enhance QALYs, and consequently improve cost-effectiveness.
Collapse
Affiliation(s)
- Young-Sil Lee
- Department of Public Health Science, Graduate School of Public Health, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| | - HyunChul Youn
- Department of Psychiatry, Soonchunhyang University Bucheon Hospital, 170 Jomaru-ro, Wonmi-gu, Bucheon, 14584, Republic of Korea
| | - Hyun-Ghang Jeong
- Department of Psychiatry, Korea University Guro Hospital, Korea University College of Medicine, 148, Gurodong-Ro, Guro-gu, Seoul, 08308, Republic of Korea. .,Korea University Research Institute of Mental Health, 148, Gurodong-Ro, Guro-gu, Seoul, 08308, Republic of Korea.
| | - Tae-Jin Lee
- Department of Public Health Science, Graduate School of Public Health, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea. .,Institute of Health and Environment, Seoul National University, 1, Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea.
| | - Ji Won Han
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, 82 Gumi-ro 173beon-gil, Bundang-gu, Seongnam, 13620, Republic of Korea
| | - Joon Hyuk Park
- Department of Psychiatry, Jeju National University School of Medicine, Jeju National University Hospital, Aran 13 gil, Jeju-si, Jeju, 63241, Republic of Korea
| | - Ki Woong Kim
- Department of Neuropsychiatry, Seoul National University Bundang Hospital, 82 Gumi-ro 173beon-gil, Bundang-gu, Seongnam, 13620, Republic of Korea.,Department of Psychiatry, Seoul National University College of Medicine, 101, Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.,Department of Brain and Cognitive Science, Seoul National University College of Natural Sciences, 1, Gwanak-ro, Gwanak-gu, Seoul, 08826, Republic of Korea
| |
Collapse
|
46
|
Groot C, Risacher SL, Chen JQA, Dicks E, Saykin AJ, Mac Donald CL, Mez J, Trittschuh EH, Mukherjee S, Barkhof F, Scheltens P, van der Flier WM, Ossenkoppele R, Crane PK. Differential trajectories of hypometabolism across cognitively-defined Alzheimer's disease subgroups. NEUROIMAGE-CLINICAL 2021; 31:102725. [PMID: 34153688 PMCID: PMC8238088 DOI: 10.1016/j.nicl.2021.102725] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/28/2021] [Accepted: 06/08/2021] [Indexed: 11/26/2022]
Abstract
Cognitive-subgroups can be identified among individuals
with AD dementia. Subgroup-specific patterns and longitudinal trajectories of
hypometabolism observed. Regional hypometabolism matched respective cognitive
profiles of subgroups. Cognitive-classification yields biologically distinct
subgroups.
Disentangling biologically distinct subgroups of Alzheimer’s
disease (AD) may facilitate a deeper understanding of the neurobiology underlying
clinical heterogeneity. We employed longitudinal [18F]FDG-PET
standardized uptake value ratios (SUVRs) to map hypometabolism across
cognitively-defined AD subgroups. Participants were 384 amyloid-positive individuals
with an AD dementia diagnosis from ADNI who had a total of 1028 FDG-scans (mean time
between first and last scan: 1.6 ± 1.8 years). These participants were categorized
into subgroups on the basis of substantial impairment at time of dementia diagnosis
in a specific cognitive domain relative to the average across domains. This approach
resulted in groups of AD-Memory (n = 135), AD-Executive (n = 8), AD-Language
(n = 22), AD-Visuospatial (n = 44), AD-Multiple Domains (n = 15) and AD-No Domains
(for whom no domain showed substantial relative impairment; n = 160). Voxelwise
contrasts against controls revealed that all AD-subgroups showed progressive
hypometabolism compared to controls across temporoparietal regions at time of AD
diagnosis. Voxelwise and regions-of-interest (ROI)-based linear mixed model analyses
revealed there were also subgroup-specific hypometabolism patterns and trajectories.
The AD-Memory group had more pronounced hypometabolism compared to all other groups
in the medial temporal lobe and posterior cingulate, and faster decline in metabolism
in the medial temporal lobe compared to AD-Visuospatial. The AD-Language group had
pronounced lateral temporal hypometabolism compared to all other groups, and the
pattern of metabolism was also more asymmetrical (left < right) than all other
groups. The AD-Visuospatial group had faster decline in metabolism in parietal
regions compared to all other groups, as well as faster decline in the precuneus
compared to AD-Memory and AD-No Domains. Taken together, in addition to a common
pattern, cognitively-defined subgroups of people with AD dementia show
subgroup-specific hypometabolism patterns, as well as differences in trajectories of
metabolism over time. These findings provide support to the notion that
cognitively-defined subgroups are biologically distinct.
Collapse
Affiliation(s)
- Colin Groot
- Department of Neurology & Alzheimer Center, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.
| | | | - J Q Alida Chen
- Department of Neurology & Alzheimer Center, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.
| | - Ellen Dicks
- Department of Neurology & Alzheimer Center, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.
| | - Andrew J Saykin
- Indiana University School of Medicine, Indianapolis, IN, USA.
| | | | - Jesse Mez
- Department of Neurology, Boston University School of Medicine, Boston, MA, USA; Alzheimer's Disease Center, Boston University School of Medicine, MA, USA.
| | - Emily H Trittschuh
- Psychiatry & Behavioral Science, University of Washington, Seattle, WA, USA; Veterans Affairs Puget Sound Health Care System, Geriatric Research, Education, & Clinical Center, Seattle, WA, USA
| | | | - Frederik Barkhof
- Department of Radiology and Nuclear Medicine, Amsterdam Neuroscience Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands; University College London, Institutes of Neurology & Healthcare Engineering, London, United Kingdom.
| | - Philip Scheltens
- Department of Neurology & Alzheimer Center, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.
| | - Wiesje M van der Flier
- Department of Neurology & Alzheimer Center, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands; Epidemiology and Data Science, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands.
| | - Rik Ossenkoppele
- Department of Neurology & Alzheimer Center, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands; Lund University, Clinical Memory Research Unit, Lund, Sweden.
| | - Paul K Crane
- Department of Medicine, University of Washington, Seattle, WA, USA
| | | |
Collapse
|
47
|
Müller L, Kirschstein T, Köhling R, Kuhla A, Teipel S. Neuronal Hyperexcitability in APPSWE/PS1dE9 Mouse Models of Alzheimer's Disease. J Alzheimers Dis 2021; 81:855-869. [PMID: 33843674 DOI: 10.3233/jad-201540] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Transgenic mouse models serve a better understanding of Alzheimer's disease (AD) pathogenesis and its consequences on neuronal function. Well-known and broadly used AD models are APPswe/PS1dE9 mice, which are able to reproduce features of amyloid-β (Aβ) plaque formations as well as neuronal dysfunction as reflected in electrophysiological recordings of neuronal hyperexcitability. The most prominent findings include abnormal synaptic function and synaptic reorganization as well as changes in membrane threshold and spontaneous neuronal firing activities leading to generalized excitation-inhibition imbalances in larger neuronal circuits and networks. Importantly, these findings in APPswe/PS1dE9 mice are at least partly consistent with results of electrophysiological studies in humans with sporadic AD. This underscores the potential to transfer mechanistic insights into amyloid related neuronal dysfunction from animal models to humans. This is of high relevance for targeted downstream interventions into neuronal hyperexcitability, for example based on repurposing of existing antiepileptic drugs, as well as the use of combinations of imaging and electrophysiological readouts to monitor effects of upstream interventions into amyloid build-up and processing on neuronal function in animal models and human studies. This article gives an overview on the pathogenic and methodological basis for recording of neuronal hyperexcitability in AD mouse models and on key findings in APPswe/PS1dE9 mice. We point at several instances to the translational perspective into clinical intervention and observation studies in humans. We particularly focus on bi-directional relations between hyperexcitability and cerebral amyloidosis, including build-up as well as clearance of amyloid, possibly related to sleep and so called glymphatic system function.
Collapse
Affiliation(s)
- Luisa Müller
- Department of Psychosomatic Medicine and Psychotherapy, University of Rostock, Rostock, Germany.,Rudolf Zenker Institute for Experimental Surgery, University of Rostock, Rostock, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| | - Timo Kirschstein
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| | - Rüdiger Köhling
- Oscar Langendorff Institute of Physiology, University of Rostock, Rostock, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| | - Angela Kuhla
- Rudolf Zenker Institute for Experimental Surgery, University of Rostock, Rostock, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| | - Stefan Teipel
- Department of Psychosomatic Medicine and Psychotherapy, University of Rostock, Rostock, Germany.,German Center for Neurodegenerative Diseases (DZNE), Rostock and Greifswald, Germany.,Centre for Transdisciplinary Neurosciences Rostock (CTNR), University of Rostock, Rostock, Germany
| |
Collapse
|
48
|
Bochiński A, Sujenthiran A, Al-Hussini M, Fruhwirth GO, Shabbir M, Yap T. 18 F-FDG PET/CT use in functional assessment of the testes: A systematic review. Andrology 2021; 9:1410-1421. [PMID: 34019736 DOI: 10.1111/andr.13042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 04/29/2021] [Accepted: 05/18/2021] [Indexed: 01/01/2023]
Abstract
INTRODUCTION Our study analysed previous studies employing positron emission tomography with co-registered computer tomography (PET/CT) in andrological patient evaluation and assessed the differences in 2-[18 F]F-fluoro-2'-deoxyglucose (FDG) uptake between three groups: healthy testes, benign and malignant testicular pathology. METHODS Medline and Embase were systematically searched for studies involving FDG-PET/CT imaging of testes with results expressed as mean standardised uptake value (SUVmean ). A one-way ANOVA was used to compare SUVmean between three groups. All papers assessing andrological parameters were pooled to compare fertility data. RESULTS Seventeen studies, including three relating to fertility diagnosis, with a total of 830 patients, were included in the review. One-way ANOVA showed a statistical difference between mean values of tracer SUVmean in healthy and malignant testes (Dif. = -2.77, 95% CI = -4.32 to 1.21, p < 0.01) as well as benign and malignant (Dif. = -2.95, 95% CI = -4.33 to -1.21, p < 0.01) but no difference between healthy and benign (Dif. = 0.19, 95% CI = -0.96 to 1.33, p = 0.90). There is some evidence to suggest that FDG uptake and testicular volume are positively correlated to total sperm count, sperm concentration and sperm motility and that germ cells are likely to account for the majority of testicular FDG accumulation. CONCLUSION Our findings indicate that malignant testicular lesions demonstrate a significantly higher FDG uptake than benign testicular lesions or healthy testes. Some evidence also suggests that FDG-PET could visualise metabolic activity and thus spermatogenesis; however more studies are required to determine whether FDG-PET could also be used to diagnose infertility. Further studies should focus on correlating both sex hormone-serum levels and semen analysis results with imaging data.
Collapse
Affiliation(s)
- Antoni Bochiński
- School of Bioscience Education, Guy's Campus, King's College London, London, UK
| | | | | | - Gilbert O Fruhwirth
- Imaging Therapies and Cancer Group, School of Biomedical Engineering and Imaging Sciences, King's College London, London, UK
| | - Majed Shabbir
- Department of Urology, Guy's and St Thomas' NHS Trust, London, UK
| | - Tet Yap
- Department of Urology, Guy's and St Thomas' NHS Trust, London, UK
| |
Collapse
|
49
|
Yu H, Liu Y, He B, He T, Chen C, He J, Yang X, Wang J. Platelet biomarkers for a descending cognitive function: A proteomic approach. Aging Cell 2021; 20:e13358. [PMID: 33942972 PMCID: PMC8135080 DOI: 10.1111/acel.13358] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 02/23/2021] [Accepted: 03/15/2021] [Indexed: 12/31/2022] Open
Abstract
Memory loss is the most common clinical sign in Alzheimer's disease (AD); thus, searching for peripheral biomarkers to predict cognitive decline is promising for early diagnosis of AD. As platelets share similarities to neuron biology, it may serve as a peripheral matrix for biomarkers of neurological disorders. Here, we conducted a comprehensive and in-depth platelet proteomic analysis using TMT-LC-MS/MS in the populations with mild cognitive impairment (MCI, MMSE = 18-23), severe cognitive impairments (AD, MMSE = 2-17), and the age-/sex-matched normal cognition controls (MMSE = 29-30). A total of 360 differential proteins were detected in MCI and AD patients compared with the controls. These differential proteins were involved in multiple KEGG pathways, including AD, AMP-activated protein kinase (AMPK) pathway, telomerase RNA localization, platelet activation, and complement activation. By correlation analysis with MMSE score, three positively correlated pathways and two negatively correlated pathways were identified to be closely related to cognitive decline in MCI and AD patients. Partial least squares discriminant analysis (PLS-DA) showed that changes of nine proteins, including PHB, UQCRH, CD63, GP1BA, FINC, RAP1A, ITPR1/2, and ADAM10 could effectively distinguish the cognitively impaired patients from the controls. Further machine learning analysis revealed that a combination of four decreased platelet proteins, that is, PHB, UQCRH, GP1BA, and FINC, was most promising for predicting cognitive decline in MCI and AD patients. Taken together, our data provide a set of platelet biomarkers for predicting cognitive decline which may be applied for the early screening of AD.
Collapse
Affiliation(s)
- Haitao Yu
- Key Laboratory of Ministry of Education for Neurological DisordersSchool of Basic MedicineDepartment of PathophysiologyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Modern Toxicology of ShenzhenShenzhen Center for Disease Control and PreventionShenzhenChina
| | - Yanchao Liu
- Key Laboratory of Ministry of Education for Neurological DisordersSchool of Basic MedicineDepartment of PathophysiologyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Benrong He
- Key Laboratory of Ministry of Education for Neurological DisordersSchool of Basic MedicineDepartment of PathophysiologyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Ting He
- Key Laboratory of Ministry of Education for Neurological DisordersSchool of Basic MedicineDepartment of PathophysiologyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Chongyang Chen
- Key Laboratory of Ministry of Education for Neurological DisordersSchool of Basic MedicineDepartment of PathophysiologyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Key Laboratory of Modern Toxicology of ShenzhenShenzhen Center for Disease Control and PreventionShenzhenChina
| | - Jiahua He
- School of PhysicsHuazhong University of Science and TechnologyWuhanHubeiChina
| | - Xifei Yang
- Key Laboratory of Modern Toxicology of ShenzhenShenzhen Center for Disease Control and PreventionShenzhenChina
| | - Jian‐Zhi Wang
- Key Laboratory of Ministry of Education for Neurological DisordersSchool of Basic MedicineDepartment of PathophysiologyTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Co‐innovation Center of NeuroregenerationNantong UniversityNantongChina
| |
Collapse
|
50
|
Sarikaya I, Kamel WA, Ateyah KK, Essa NB, AlTailji S, Sarikaya A. Visual versus semiquantitative analysis of 18F- fluorodeoxyglucose-positron emission tomography brain images in patients with dementia. World J Nucl Med 2021; 20:82-89. [PMID: 33850493 PMCID: PMC8034786 DOI: 10.4103/wjnm.wjnm_53_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 05/31/2018] [Accepted: 07/19/2018] [Indexed: 11/04/2022] Open
Abstract
Various studies have reported to the superiority of semiquantitative (SQ) analysis over visual analysis in detecting metabolic changes in the brain. In this study, we aimed to determine the limitations of SQ analysis programs and the current status of 18F- fluorodeoxyglucose (FDG)-positron emission tomography (PET) scan in dementia. 18F- FDG-PET/computed tomography (CT) brain images of 39 patients with a history of dementia were analyzed both visually and semiquantitatively. Using the visually markedly abnormal 18F- FDG-PET images as standard, we wanted to test the accuracy of two commercially available SQ analysis programs. SQ analysis results were classified as matching, partially matching and nonmatching with visually markedly abnormal studies. On visual analysis, 18F- FDG-PET showed marked regional hypometabolism in 19 patients, mild abnormalities in 8 and was normal in 12 patients. SQ analysis-1 results matched with visual analysis in 8 patients (42.1%) and partially matched in 11. SQ analysis-2 findings matched with visual analysis in 11 patients (57.8%) and partially matched in 7 and did not match in 1. Marked regional hypometabolism was either on the left side of the brain or was more significant on the left than the right in 63% of patients. Preservation of metabolism in sensorimotor cortex was seen in various dementia subtypes. Reviewing images in color scale and maximum intensity projection (MIP) image was helpful in demonstrating and displaying regional abnormalities, respectively. SQ analysis provides less accurate results as compared to visual analysis by experts. Due to suboptimal image registration and selection of brain areas, SQ analysis provides inaccurate results, particularly in small areas and areas in close proximity. Image registration and selection of areas with SQ programs should be checked carefully before reporting the results.
Collapse
Affiliation(s)
- Ismet Sarikaya
- Department of Nuclear Medicine, Faculty of Medicine, Kuwait University, Kuwait University, Kuwait
| | - Walaa A Kamel
- Department of Neurology, Faculty of Medicine, Beni-Suef University, Egypt.,Department of Nuclear Medicine, Ibn Sina Hospital, Kuwait
| | | | - Nooraessa Bin Essa
- Department of Nuclear Medicine, Mubarak Al-Kabeer Hospital, Kuwait City, Kuwait
| | | | - Ali Sarikaya
- Department of Nuclear Medicine, Faculty of Medicine, Trakya University, Edirne, Turkey
| |
Collapse
|