1
|
Madsen LS, Ismail R, Parbo P, Kjeldsen PL, Schaldemose JL, Hansen KV, Gottrup H, Aanerud J, Eskildsen SF, Brooks DJ. Microglial responses partially mediate the effect of Aβ on cognition in Alzheimer's disease. Alzheimers Dement 2024; 20:8028-8037. [PMID: 39392185 DOI: 10.1002/alz.14298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/24/2024] [Accepted: 09/07/2024] [Indexed: 10/12/2024]
Abstract
INTRODUCTION Microglial responses are an integral part of Alzheimer's disease (AD) pathology and are associated with amyloid beta (Aβ) deposition. This study aimed to investigate the effects of Aβ and microglial responses on global cognitive impairment. METHODS In this longitudinal study, 28 patients with mild cognitive impairment and 11 healthy controls underwent 11C-PK11195 and 11C-Pittsburgh compound B positron emission tomography (PET), structural magnetic resonance imaging scans, and global cognitive ratings at baseline and 2-year follow-up. Correlations between PET uptake and global cognition were assessed. Additionally, the mediation effect of the microglial response on the association between Aβ load and global cognition was assessed. RESULTS Aβ load and the microglial response were both independently detrimental to global cognitive performance at baseline; however, at 2-year follow-up the association between Aβ load and global cognitive ratings was partially mediated by the microglial response. DISCUSSION As AD progresses, the associated microglial response partially mediates the detrimental effect of aggregated Aβ on cognition. HIGHLIGHTS This was a longitudinal study of amyloid beta (Aβ), microglial responses, and global cognitive performance. Aβ and microglial responses both affect cognition in early Alzheimer's disease. Microglial response partially mediates the effect of Aβ on cognition in later stages.
Collapse
Affiliation(s)
- Lasse S Madsen
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience, Aarhus University, Aarhus, Denmark
| | - Rola Ismail
- Department of Nuclear Medicine, Sygehus Lillebaelt, Vejle, Denmark
| | - Peter Parbo
- Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
| | - Pernille L Kjeldsen
- Department of Nuclear Medicine and PET-Centre, Aarhus University Hospital, Aarhus, Denmark
- Department of Neurology, Aalborg University Hospital, Aalborg, Denmark
| | - Jeppe L Schaldemose
- Department of Nuclear Medicine and PET-Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Kim V Hansen
- Department of Nuclear Medicine and PET-Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Hanne Gottrup
- Department of Neurology, Aarhus University Hospital, Aarhus, Denmark
| | - Joel Aanerud
- Department of Nuclear Medicine and PET-Centre, Aarhus University Hospital, Aarhus, Denmark
| | - Simon F Eskildsen
- Department of Clinical Medicine, Center of Functionally Integrative Neuroscience, Aarhus University, Aarhus, Denmark
| | - David J Brooks
- Department of Nuclear Medicine and PET-Centre, Aarhus University Hospital, Aarhus, Denmark
- Institute of Translational and Clinical Research, University of Newcastle upon Tyne, Newcastle upon Tyne, UK
| |
Collapse
|
2
|
Gaire BP, Koronyo Y, Fuchs DT, Shi H, Rentsendorj A, Danziger R, Vit JP, Mirzaei N, Doustar J, Sheyn J, Hampel H, Vergallo A, Davis MR, Jallow O, Baldacci F, Verdooner SR, Barron E, Mirzaei M, Gupta VK, Graham SL, Tayebi M, Carare RO, Sadun AA, Miller CA, Dumitrascu OM, Lahiri S, Gao L, Black KL, Koronyo-Hamaoui M. Alzheimer's disease pathophysiology in the Retina. Prog Retin Eye Res 2024; 101:101273. [PMID: 38759947 PMCID: PMC11285518 DOI: 10.1016/j.preteyeres.2024.101273] [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: 02/11/2023] [Revised: 04/23/2024] [Accepted: 05/10/2024] [Indexed: 05/19/2024]
Abstract
The retina is an emerging CNS target for potential noninvasive diagnosis and tracking of Alzheimer's disease (AD). Studies have identified the pathological hallmarks of AD, including amyloid β-protein (Aβ) deposits and abnormal tau protein isoforms, in the retinas of AD patients and animal models. Moreover, structural and functional vascular abnormalities such as reduced blood flow, vascular Aβ deposition, and blood-retinal barrier damage, along with inflammation and neurodegeneration, have been described in retinas of patients with mild cognitive impairment and AD dementia. Histological, biochemical, and clinical studies have demonstrated that the nature and severity of AD pathologies in the retina and brain correspond. Proteomics analysis revealed a similar pattern of dysregulated proteins and biological pathways in the retina and brain of AD patients, with enhanced inflammatory and neurodegenerative processes, impaired oxidative-phosphorylation, and mitochondrial dysfunction. Notably, investigational imaging technologies can now detect AD-specific amyloid deposits, as well as vasculopathy and neurodegeneration in the retina of living AD patients, suggesting alterations at different disease stages and links to brain pathology. Current and exploratory ophthalmic imaging modalities, such as optical coherence tomography (OCT), OCT-angiography, confocal scanning laser ophthalmoscopy, and hyperspectral imaging, may offer promise in the clinical assessment of AD. However, further research is needed to deepen our understanding of AD's impact on the retina and its progression. To advance this field, future studies require replication in larger and diverse cohorts with confirmed AD biomarkers and standardized retinal imaging techniques. This will validate potential retinal biomarkers for AD, aiding in early screening and monitoring.
Collapse
Affiliation(s)
- Bhakta Prasad Gaire
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yosef Koronyo
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Dieu-Trang Fuchs
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Haoshen Shi
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Altan Rentsendorj
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ron Danziger
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jean-Philippe Vit
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Nazanin Mirzaei
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jonah Doustar
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Julia Sheyn
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Harald Hampel
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Andrea Vergallo
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Miyah R Davis
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ousman Jallow
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Filippo Baldacci
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Department of Clinical and Experimental Medicine, Neurology Unit, University of Pisa, Pisa, Italy
| | | | - Ernesto Barron
- Department of Ophthalmology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA; Doheny Eye Institute, Los Angeles, CA, USA
| | - Mehdi Mirzaei
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Vivek K Gupta
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Stuart L Graham
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia; Department of Clinical Medicine, Macquarie University, Sydney, NSW, Australia
| | - Mourad Tayebi
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Roxana O Carare
- Department of Clinical Neuroanatomy, University of Southampton, Southampton, UK
| | - Alfredo A Sadun
- Department of Ophthalmology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA; Doheny Eye Institute, Los Angeles, CA, USA
| | - Carol A Miller
- Department of Pathology Program in Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - Shouri Lahiri
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Liang Gao
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Keith L Black
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Biomedical Sciences, Division of Applied Cell Biology and Physiology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
3
|
Aguero C, Dhaynaut M, Amaral AC, Moon SH, Neelamegam R, Scapellato M, Carazo-Casas C, Kumar S, El Fakhri G, Johnson K, Frosch MP, Normandin MD, Gómez-Isla T. Head-to-head comparison of [ 18F]-Flortaucipir, [ 18F]-MK-6240 and [ 18F]-PI-2620 postmortem binding across the spectrum of neurodegenerative diseases. Acta Neuropathol 2024; 147:25. [PMID: 38280071 PMCID: PMC10822013 DOI: 10.1007/s00401-023-02672-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 12/19/2023] [Accepted: 12/19/2023] [Indexed: 01/29/2024]
Abstract
We and others have shown that [18F]-Flortaucipir, the most validated tau PET tracer thus far, binds with strong affinity to tau aggregates in Alzheimer's (AD) but has relatively low affinity for tau aggregates in non-AD tauopathies and exhibits off-target binding to neuromelanin- and melanin-containing cells, and to hemorrhages. Several second-generation tau tracers have been subsequently developed. [18F]-MK-6240 and [18F]-PI-2620 are the two that have garnered most attention. Our recent data indicated that the binding pattern of [18F]-MK-6240 closely parallels that of [18F]-Flortaucipir. The present study aimed at the direct comparison of the autoradiographic binding properties and off-target profile of [18F]-Flortaucipir, [18F]-MK-6240 and [18F]-PI-2620 in human tissue specimens, and their potential binding to monoamine oxidases (MAO). Phosphor-screen and high resolution autoradiographic patterns of the three tracers were studied in the same postmortem tissue material from AD and non-AD tauopathies, cerebral amyloid angiopathy, synucleopathies, transactive response DNA-binding protein 43 (TDP-43)-frontotemporal lobe degeneration and controls. Our results show that the three tracers show nearly identical autoradiographic binding profiles. They all strongly bind to neurofibrillary tangles in AD but do not seem to bind to a significant extent to tau aggregates in non-AD tauopathies pointing to their limited utility for the in vivo detection of non-AD tau lesions. None of them binds to lesions containing β-amyloid, α-synuclein or TDP-43 but they all show strong off-target binding to neuromelanin and melanin-containing cells, as well as weaker binding to areas of hemorrhage. The autoradiographic binding signals of the three tracers are only weakly displaced by competing concentrations of selective MAO-B inhibitor deprenyl but not by MAO-A inhibitor clorgyline suggesting that MAO enzymes do not appear to be a significant binding target of any of them. These findings provide relevant insights for the correct interpretation of the in vivo behavior of these three tau PET tracers.
Collapse
Affiliation(s)
- Cinthya Aguero
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
| | - Maeva Dhaynaut
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Ana C Amaral
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
| | - S-H Moon
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Ramesh Neelamegam
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Margaret Scapellato
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
| | - Carlos Carazo-Casas
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
| | - Sunny Kumar
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
| | - Georges El Fakhri
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Keith Johnson
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Matthew P Frosch
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA
- C.S. Kubik Laboratory for Neuropathology, Massachusetts General Hospital, Boston, MA, USA
| | - Marc D Normandin
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| | - Teresa Gómez-Isla
- MassGeneral Institute for NeuroDegenerative Disease, Charlestown, MA, USA.
- Department of Neurology, Massachusetts General Hospital, WACC Suite 715, 15th Parkman St., Boston, MA, 02114, USA.
| |
Collapse
|
4
|
Etekochay MO, Amaravadhi AR, González GV, Atanasov AG, Matin M, Mofatteh M, Steinbusch HW, Tesfaye T, Praticò D. Unveiling New Strategies Facilitating the Implementation of Artificial Intelligence in Neuroimaging for the Early Detection of Alzheimer's Disease. J Alzheimers Dis 2024; 99:1-20. [PMID: 38640152 DOI: 10.3233/jad-231135] [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: 04/21/2024]
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder with a global impact. The past few decades have witnessed significant strides in comprehending the underlying pathophysiological mechanisms and developing diagnostic methodologies for AD, such as neuroimaging approaches. Neuroimaging techniques, including positron emission tomography and magnetic resonance imaging, have revolutionized the field by providing valuable insights into the structural and functional alterations in the brains of individuals with AD. These imaging modalities enable the detection of early biomarkers such as amyloid-β plaques and tau protein tangles, facilitating early and precise diagnosis. Furthermore, the emerging technologies encompassing blood-based biomarkers and neurochemical profiling exhibit promising results in the identification of specific molecular signatures for AD. The integration of machine learning algorithms and artificial intelligence has enhanced the predictive capacity of these diagnostic tools when analyzing complex datasets. In this review article, we will highlight not only some of the most used diagnostic imaging approaches in neurodegeneration research but focus much more on new tools like artificial intelligence, emphasizing their application in the realm of AD. These advancements hold immense potential for early detection and intervention, thereby paving the way for personalized therapeutic strategies and ultimately augmenting the quality of life for individuals affected by AD.
Collapse
Affiliation(s)
| | - Amoolya Rao Amaravadhi
- Internal Medicine, Malla Reddy Institute of Medical Sciences, Jeedimetla, Hyderabad, India
| | - Gabriel Villarrubia González
- Expert Systems and Applications Laboratory (ESALAB), Faculty of Science, University of Salamanca, Salamanca, Spain
| | - Atanas G Atanasov
- Department of Biotechnology and Nutrigenomics, Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, Poland
- Ludwig Boltzmann Institute Digital Health and Patient Safety, Medical University of Vienna, Vienna, Austria
| | - Maima Matin
- Ludwig Boltzmann Institute Digital Health and Patient Safety, Medical University of Vienna, Vienna, Austria
| | - Mohammad Mofatteh
- School of Medicine, Dentistry, and Biomedical Sciences, Queen's University Belfast, Belfast, UK
| | - Harry Wilhelm Steinbusch
- Department of Cellular and Translational Neuroscience, School for Mental Health and Neuroscience, Faculty of Health Medicine and Life Sciences, Maastricht University, Netherlands
| | - Tadele Tesfaye
- CareHealth Medical Practice, Jimma Road, Addis Ababa, Ethiopia
| | - Domenico Praticò
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
5
|
Rani N, Alm KH, Corona-Long CA, Speck CL, Soldan A, Pettigrew C, Zhu Y, Albert M, Bakker A. Tau PET burden in Brodmann areas 35 and 36 is associated with individual differences in cognition in non-demented older adults. Front Aging Neurosci 2023; 15:1272946. [PMID: 38161595 PMCID: PMC10757623 DOI: 10.3389/fnagi.2023.1272946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 10/23/2023] [Indexed: 01/03/2024] Open
Abstract
Introduction The accumulation of neurofibrillary tau tangles, a neuropathological hallmark of Alzheimer's disease (AD), occurs in medial temporal lobe (MTL) regions early in the disease process, with some of the earliest deposits localized to subregions of the entorhinal cortex. Although functional specialization of entorhinal cortex subregions has been reported, few studies have considered functional associations with localized tau accumulation. Methods In this study, stepwise linear regressions were used to examine the contributions of regional tau burden in specific MTL subregions, as measured by 18F-MK6240 PET, to individual variability in cognition. Dependent measures of interest included the Clinical Dementia Rating Sum of Boxes (CDR-SB), Mini Mental State Examination (MMSE), and composite scores of delayed episodic memory and language. Other model variables included age, sex, education, APOE4 status, and global amyloid burden, indexed by 11C-PiB. Results Tau burden in right Brodmann area 35 (BA35), left and right Brodmann area 36 (BA36), and age each uniquely contributed to the proportion of explained variance in CDR-SB scores, while right BA36 and age were also significant predictors of MMSE scores, and right BA36 was significantly associated with delayed episodic memory performance. Tau burden in both left and right BA36, along with education, uniquely contributed to the proportion of explained variance in language composite scores. Importantly, the addition of more inclusive ROIs, encompassing less granular segmentation of the entorhinal cortex, did not significantly contribute to explained variance in cognition across any of the models. Discussion These findings suggest that the ability to quantify tau burden in more refined MTL subregions may better account for individual differences in cognition, which may improve the identification of non-demented older adults who are on a trajectory of decline due to AD.
Collapse
Affiliation(s)
- Nisha Rani
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Kylie H. Alm
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Caitlin A. Corona-Long
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Caroline L. Speck
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Anja Soldan
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Corinne Pettigrew
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Yuxin Zhu
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Marilyn Albert
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Arnold Bakker
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
6
|
Aramadaka S, Mannam R, Sankara Narayanan R, Bansal A, Yanamaladoddi VR, Sarvepalli SS, Vemula SL. Neuroimaging in Alzheimer's Disease for Early Diagnosis: A Comprehensive Review. Cureus 2023; 15:e38544. [PMID: 37273363 PMCID: PMC10239271 DOI: 10.7759/cureus.38544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/03/2023] [Indexed: 06/06/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in the elderly, affecting roughly half of those over the age of 85. We briefly discussed the risk factors, epidemiology, and treatment options for AD. The development of therapeutic therapies operating very early in the disease cascade has been spurred by the realization that the disease process begins at least a decade or more before the manifestation of symptoms. Thus, the clinical significance of early diagnosis was emphasized. Using various keywords, a literature search was carried out using PubMed and other databases. For inclusion, pertinent articles were chosen and reviewed. This article has reviewed different neuroimaging techniques that are considered advanced tools to aid in establishing a diagnosis and highlighted the advantages as well as disadvantages of those techniques. Besides, the prevalence of several in vivo biomarkers aided in discriminating affected individuals from healthy controls in the early stages of the disease. Each imaging method has its advantages and disadvantages, hence no single imaging approach can be the optimum modality for diagnosis. This article also commented on a better approach to using these techniques to increase the likelihood of an early diagnosis.
Collapse
Affiliation(s)
| | - Raam Mannam
- Research, Narayana Medical College, Nellore, IND
| | | | - Arpit Bansal
- Research, Narayana Medical College, Nellore, IND
| | | | | | | |
Collapse
|
7
|
Sarabia-Vallejo Á, López-Alvarado P, Menéndez JC. Small-molecule theranostics in Alzheimer's disease. Eur J Med Chem 2023; 255:115382. [PMID: 37141706 DOI: 10.1016/j.ejmech.2023.115382] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 05/06/2023]
Abstract
Alzheimer's Disease (AD) remains one of the most challenging health-related issues for our society. It is becoming increasingly prevalent, especially in developed countries, due to the rising life expectancy and, moreover, represents a considerable economic burden worldwide. All efforts at the discovery of new diagnostic and therapeutic tools in the last decades have invariably met with failure, making AD an incurable illness and underscoring the need for new approaches. In recent years, theranostic agents have emerged as an interesting strategy. They are molecules able to simultaneously provide diagnostic information and deliver therapeutic activity, allowing for the assessment of the molecule activity, the organism response and the pharmacokinetics. This makes these compounds promising for streamlining research on AD drugs and for their application in personalized medicine. We review here the field of small-molecule theranostic agents as promising tools for the development of novel diagnostic and therapeutic resources against AD, highlighting the positive and significant impact that theranostics can be expected to have in the near future in clinical practice.
Collapse
Affiliation(s)
- Álvaro Sarabia-Vallejo
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, 28040, Madrid, Spain
| | - Pilar López-Alvarado
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, 28040, Madrid, Spain
| | - J Carlos Menéndez
- Unidad de Química Orgánica y Farmacéutica, Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense, 28040, Madrid, Spain.
| |
Collapse
|
8
|
Song J, Kambari Y, Amaev A, Ueno F, Torres Carmona E, De Luca V, Pollock B, Flint A, Ishrat Husain M, Graff-Guerrero A, Gerretsen P. Psilocybin to promote synaptogenesis in the brains of patients with mild cognitive impairment. Med Hypotheses 2023. [DOI: 10.1016/j.mehy.2023.111068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2023]
|
9
|
Seibyl JP, DuBois JM, Racine A, Collins J, Guo Q, Wooten D, Stage E, Cheng D, Gunn RN, Porat L, Whittington A, Kuo PH, Ichise M, Comley R, Martarello L, Salinas C. A Visual Interpretation Algorithm for Assessing Brain Tauopathy with 18F-MK-6240 PET. J Nucl Med 2023; 64:444-451. [PMID: 36175137 PMCID: PMC10071795 DOI: 10.2967/jnumed.122.264371] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 09/21/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022] Open
Abstract
In vivo characterization of pathologic deposition of tau protein in the human brain by PET imaging is a promising tool in drug development trials of Alzheimer disease (AD). 6-(fluoro-18F)-3-(1H-pyrrolo[2,3-c]pyridin-1-yl)isoquinolin-5-amine (18F-MK-6240) is a radiotracer with high selectivity and subnanomolar affinity for neurofibrillary tangles that shows favorable nonspecific brain penetration and excellent kinetic properties. The purpose of the present investigation was to develop a visual assessment method that provides both an overall assessment of brain tauopathy and regional characterization of abnormal tau deposition. Methods: 18F-MK-6240 scans from 102 participants (including cognitively normal volunteers and patients with AD or other neurodegenerative disorders) were reviewed by an expert nuclear medicine physician masked to each participant's diagnosis to identify common patterns of brain uptake. This initial visual read method was field-tested in a separate, nonoverlapping cohort of 102 participants, with 2 additional naïve readers trained on the method. Visual read outcomes were compared with semiquantitative assessments using volume-of-interest SUV ratio. Results: For the visual read, the readers assessed 8 gray-matter regions per hemisphere as negative (no abnormal uptake) or positive (1%-25% of the region involved, 25%-75% involvement, or >75% involvement) and then characterized the tau binding pattern as positive or negative for evidence of tau and, if positive, whether brain uptake was in an AD pattern. The readers demonstrated agreement 94% of the time for overall positivity or negativity. Concordance on the determination of regional binary outcomes (negative or positive) showed agreement of 74.3% and a Fleiss κ of 0.912. Using clinical diagnosis as the ground truth, the readers demonstrated a sensitivity of 73%-79% and specificity of 91%-93%, with a combined reader-concordance sensitivity of 80% and specificity of 93%. The average SUV ratio in cortical regions showed a robust correlation with visually derived ratings of regional involvement (r = 0.73, P < 0.0001). Conclusion: We developed a visual read algorithm for 18F-MK-6240 PET offering determination of both scan positivity and the regional degree of cortical involvement. These cross-sectional results show strong interreader concordance on both binary and regional assessments of tau deposition, as well as good sensitivity and excellent specificity supporting use as a tool for clinical trials.
Collapse
Affiliation(s)
- John P Seibyl
- Institute for Neurodegenerative Disorders, New Haven, Connecticut;
- Invicro, New Haven, Connecticut
| | | | | | | | - Qi Guo
- AbbVie, North Chicago, Illinois
| | | | | | | | | | | | | | | | - Masanori Ichise
- National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, Japan
| | | | | | | |
Collapse
|
10
|
Serrano ME, Kim E, Petrinovic MM, Turkheimer F, Cash D. Imaging Synaptic Density: The Next Holy Grail of Neuroscience? Front Neurosci 2022; 16:796129. [PMID: 35401097 PMCID: PMC8990757 DOI: 10.3389/fnins.2022.796129] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 02/15/2022] [Indexed: 12/19/2022] Open
Abstract
The brain is the central and most complex organ in the nervous system, comprising billions of neurons that constantly communicate through trillions of connections called synapses. Despite being formed mainly during prenatal and early postnatal development, synapses are continually refined and eliminated throughout life via complicated and hitherto incompletely understood mechanisms. Failure to correctly regulate the numbers and distribution of synapses has been associated with many neurological and psychiatric disorders, including autism, epilepsy, Alzheimer’s disease, and schizophrenia. Therefore, measurements of brain synaptic density, as well as early detection of synaptic dysfunction, are essential for understanding normal and abnormal brain development. To date, multiple synaptic density markers have been proposed and investigated in experimental models of brain disorders. The majority of the gold standard methodologies (e.g., electron microscopy or immunohistochemistry) visualize synapses or measure changes in pre- and postsynaptic proteins ex vivo. However, the invasive nature of these classic methodologies precludes their use in living organisms. The recent development of positron emission tomography (PET) tracers [such as (18F)UCB-H or (11C)UCB-J] that bind to a putative synaptic density marker, the synaptic vesicle 2A (SV2A) protein, is heralding a likely paradigm shift in detecting synaptic alterations in patients. Despite their limited specificity, novel, non-invasive magnetic resonance (MR)-based methods also show promise in inferring synaptic information by linking to glutamate neurotransmission. Although promising, all these methods entail various advantages and limitations that must be addressed before becoming part of routine clinical practice. In this review, we summarize and discuss current ex vivo and in vivo methods of quantifying synaptic density, including an evaluation of their reliability and experimental utility. We conclude with a critical assessment of challenges that need to be overcome before successfully employing synaptic density biomarkers as diagnostic and/or prognostic tools in the study of neurological and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Maria Elisa Serrano
- Department of Neuroimaging, The BRAIN Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, United Kingdom
| | - Eugene Kim
- Department of Neuroimaging, The BRAIN Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, United Kingdom
| | - Marija M Petrinovic
- Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,MRC Centre for Neurodevelopmental Disorders, King's College London, London, United Kingdom
| | - Federico Turkheimer
- Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, United Kingdom
| | - Diana Cash
- Department of Neuroimaging, The BRAIN Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, United Kingdom.,Department of Neuroimaging, Institute of Psychiatry, Psychology and Neuroscience, Kings College London, London, United Kingdom
| |
Collapse
|
11
|
Murchison CF, Kennedy RE, McConathy JE, Roberson ED. Racial Differences in Alzheimer's Disease Specialist Encounters Are Associated with Usage of Molecular Imaging and Dementia Medications: An Enterprise-Wide Analysis Using i2b2. J Alzheimers Dis 2021; 79:543-557. [PMID: 33337364 PMCID: PMC7902957 DOI: 10.3233/jad-200796] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background: African Americans are at increased risk for Alzheimer’s disease (AD) but barriers to optimal clinical care are unclear. Objective: To comprehensively evaluate potential racial differences in the diagnosis and treatment of AD in an academic medical center. Methods: We used the clinical informatics tool, i2b2, to analyze all patient encounters for AD or mild cognitive impairment (MCI) in the University of Alabama at Birmingham Health System over a three-year period, examining neuroimaging rates and dementia-related medication use by race and clinic site using ratio tests on contingency tables of stratified patient counts. Results: Enterprise-wide, African Americans were not underrepresented among outpatients seen for AD/MCI. However, there were differences in the clinic setting where visits occurred, with African Americans overrepresented in Geriatrics and primary care clinics and underrepresented in Memory Disorders specialty clinics. There were no racial differences in the rates at which any clinic ordered PET neuroimaging tests or dementia-related medications. However, unsurprisingly, specialty clinics ordered both PET neuroimaging and dementia-related medications at a higher rate than primary care clinics, and overall across the medical enterprise, African Americans were statistically less likely to have PET neuroimaging or dementia-related medications ordered. Conclusion: African Americans with AD/MCI were not underrepresented at this academic medical center but were somewhat less likely to have PET neuroimaging or to be on dementia-related medications, potentially in part from underrepresentation in the specialty clinics where these orders are more likely. The reasons for this underrepresentation in specialty clinics are likely multifactorial and important to better understand.
Collapse
Affiliation(s)
- Charles F Murchison
- Alzheimer's Disease Research Center, Department of Neurology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.,Department of Biostatistics, School of Public Health, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Richard E Kennedy
- Alzheimer's Disease Research Center, Department of Neurology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.,Integrative Center for Aging Research, Department of Medicine, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jonathan E McConathy
- Molecular Imaging and Therapeutics, Department of Radiology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Erik D Roberson
- Alzheimer's Disease Research Center, Department of Neurology, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA.,Center for Neurodegeneration and Experimental Therapeutics, Department of Neurobiology, School of Medicine, Birmingham, AL, USA
| |
Collapse
|
12
|
Dartora CM, Borelli WV, Koole M, Marques da Silva AM. Cognitive Decline Assessment: A Review From Medical Imaging Perspective. Front Aging Neurosci 2021; 13:704661. [PMID: 34489675 PMCID: PMC8416532 DOI: 10.3389/fnagi.2021.704661] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 07/19/2021] [Indexed: 11/13/2022] Open
Abstract
Aging is a complex process that involves changes at both molecular and morphological levels. However, our understanding of how aging affects brain anatomy and function is still poor. In addition, numerous biomarkers and imaging markers, usually associated with neurodegenerative diseases such as Alzheimer's disease (AD), have been clinically used to study cognitive decline. However, the path of cognitive decline from healthy aging to a mild cognitive impairment (MCI) stage has been studied only marginally. This review presents aspects of cognitive decline assessment based on the imaging differences between individuals cognitively unimpaired and in the decline spectrum. Furthermore, we discuss the relationship between imaging markers and the change in their patterns with aging by using neuropsychological tests. Our goal is to delineate how aging has been studied by using medical imaging tools and further explore the aging brain and cognitive decline. We find no consensus among the biomarkers to assess the cognitive decline and its relationship with the cognitive decline trajectory. Brain glucose hypometabolism was found to be directly related to aging and indirectly to cognitive decline. We still need to understand how to quantify an expected hypometabolism during cognitive decline during aging. The Aβ burden should be longitudinally studied to achieve a better consensus on its association with changes in the brain and cognition decline with aging. There exists a lack of standardization of imaging markers that highlight the need for their further improvement. In conclusion, we argue that there is a lot to investigate and understand cognitive decline better and seek a window for a suitable and effective treatment strategy.
Collapse
Affiliation(s)
- Caroline Machado Dartora
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul, PUCRS, Porto Alegre, Brazil
| | - Wyllians Vendramini Borelli
- Neurology Department, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil.,Brain Institute of Rio Grande do Sul, BraIns, Porto Alegre, Brazil
| | - Michel Koole
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Ana Maria Marques da Silva
- School of Medicine, Pontifical Catholic University of Rio Grande do Sul, PUCRS, Porto Alegre, Brazil.,Brain Institute of Rio Grande do Sul, BraIns, Porto Alegre, Brazil.,Medical Image Computing Laboratory, School of Technology, Pontifical Catholic University of Rio Grande do Sul, PUCRS, Porto Alegre, Brazil
| |
Collapse
|
13
|
Liang Z, Wu L, Gong S, Liu X. The cognitive dysfunction related to Alzheimer disease or cerebral small vessel disease: What's the differences. Medicine (Baltimore) 2021; 100:e26967. [PMID: 34449462 PMCID: PMC8389965 DOI: 10.1097/md.0000000000026967] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 06/14/2021] [Accepted: 08/01/2021] [Indexed: 01/04/2023] Open
Abstract
ABSTRACT Alzheimer disease (AD) and sporadic cerebral small vessel disease (CSVD) are common cognitive disorders. Both AD and CSVD have mental symptoms including chronic progressive cognitive impairment, dysfunction, and behavioral abnormalities. However, the differences on the cognitive dysfunction of AD and CSVD remain unclear. It is necessary to elucidate the cognitive dysfunction differences of AD and CSVD, and to identify the potential risk factors.AD or sporadic CSVD patients treated in our hospital from December 1, 2018 to May 31, 2019 were included. And we selected healthy participants as controls. The mini-mental state examination and Montreal Cognitive Assessment Scale were used for neuropsychological assessment, and related medical information were collected and compared.A total of 190 patients were included. The total mini-mental state examination scores in AD, CSVD group were significantly less than that of control group, there were significant differences in the domains of directional ability, attention and computing ability, delayed recall, and visual perception (all P < .05); the total Montreal Cognitive Assessment Scale scores in AD, CSVD group were significantly less than that of control group. There were significant differences in the domains of visual space and execution, immediate remember, attention and computing ability, language, delayed recall, and directional ability (all P < .05); diabetes was a risk factor both for AD (hazard ratio = 1.63, 95% confidence interval: 1.35-1.97) and CSVD (hazard ratio = 1.15, 95% confidence interval: 1.08-1.27).The cognitive dysfunctions of AD are difference to that of CSVD patients, and diabetes is the risk factor both for AD and CSVD, future studies are needed to further identify the prevention and treatment of AD and CSVD.
Collapse
Affiliation(s)
- Zhenhong Liang
- School of Medicine, Taizhou University, Zhejiang Province, Taizhou 318000, China
| | - Lijuan Wu
- School of Medicine, Taizhou University, Zhejiang Province, Taizhou 318000, China
| | - Shumei Gong
- School of Nursing, The second Military Medical Universtiy, Shanghaihai 2000433, China
| | - Xiaohong Liu
- School of Nursing, The second Military Medical Universtiy, Shanghaihai 2000433, China
| |
Collapse
|
14
|
van Oostveen WM, de Lange ECM. Imaging Techniques in Alzheimer's Disease: A Review of Applications in Early Diagnosis and Longitudinal Monitoring. Int J Mol Sci 2021; 22:ijms22042110. [PMID: 33672696 PMCID: PMC7924338 DOI: 10.3390/ijms22042110] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/15/2021] [Accepted: 02/16/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder affecting many individuals worldwide with no effective treatment to date. AD is characterized by the formation of senile plaques and neurofibrillary tangles, followed by neurodegeneration, which leads to cognitive decline and eventually death. INTRODUCTION In AD, pathological changes occur many years before disease onset. Since disease-modifying therapies may be the most beneficial in the early stages of AD, biomarkers for the early diagnosis and longitudinal monitoring of disease progression are essential. Multiple imaging techniques with associated biomarkers are used to identify and monitor AD. AIM In this review, we discuss the contemporary early diagnosis and longitudinal monitoring of AD with imaging techniques regarding their diagnostic utility, benefits and limitations. Additionally, novel techniques, applications and biomarkers for AD research are assessed. FINDINGS Reduced hippocampal volume is a biomarker for neurodegeneration, but atrophy is not an AD-specific measure. Hypometabolism in temporoparietal regions is seen as a biomarker for AD. However, glucose uptake reflects astrocyte function rather than neuronal function. Amyloid-β (Aβ) is the earliest hallmark of AD and can be measured with positron emission tomography (PET), but Aβ accumulation stagnates as disease progresses. Therefore, Aβ may not be a suitable biomarker for monitoring disease progression. The measurement of tau accumulation with PET radiotracers exhibited promising results in both early diagnosis and longitudinal monitoring, but large-scale validation of these radiotracers is required. The implementation of new processing techniques, applications of other imaging techniques and novel biomarkers can contribute to understanding AD and finding a cure. CONCLUSIONS Several biomarkers are proposed for the early diagnosis and longitudinal monitoring of AD with imaging techniques, but all these biomarkers have their limitations regarding specificity, reliability and sensitivity. Future perspectives. Future research should focus on expanding the employment of imaging techniques and identifying novel biomarkers that reflect AD pathology in the earliest stages.
Collapse
Affiliation(s)
- Wieke M. van Oostveen
- Faculty of Science, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands;
| | - Elizabeth C. M. de Lange
- Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre of Drug Research, Leiden University, Einsteinweg 55, 2333 CC Leiden, The Netherlands
- Correspondence: ; Tel.: +31-71-527-6330
| |
Collapse
|
15
|
Ahmed TF, Ahmed A, Imtiaz F. History in perspective: How Alzheimer's Disease came to be where it is? Brain Res 2021; 1758:147342. [PMID: 33548268 DOI: 10.1016/j.brainres.2021.147342] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 01/18/2021] [Accepted: 01/28/2021] [Indexed: 01/03/2023]
Abstract
Treatment of Alzheimer's Disease (AD) remains an unsolved issue despite the pronounced global attention it has received from researchers over the last four decades. Determining the primary cause of the disease is challenging due to its long prodromal phase and multifactorial etiology. Regardless, academic disagreements amongst the scientific community have helped in making significant advancements in underpinning the molecular basis of disease pathogenesis. Substantial development in fluid and imaging biomarkers for AD led to a sharp turn in defining the disease as a molecular construct, dispensing its clinical definition. With conceptual progress, revisions in the diagnostic criteria of AD were made, culminating into the research framework proposed by National Institute on Aging and Alzheimer's Association in 2018 which unified different stages of the disease continuum, giving a common language of AT(N)1 classification to researchers. With realization that dementia is the final stage of AD spectrum, its early diagnosis by means of cerebrospinal fluid biomarkers, Positron Emission Tomography and Magnetic Resonance Imaging of the brain holds crucial importance in discovering ways of halting the disease progression. This article maps the insights into the pathogenesis as well as the diagnostic criteria and tests for AD as these have evolved over time. A contextualized timeline of how the understanding of AD has matured with advancing knowledge allows future research to be directed and unexplored avenues to be prioritized.
Collapse
Affiliation(s)
- Tehniat F Ahmed
- Department of Biochemistry, Institute of Biomedical Sciences, Dow University of Health Sciences, Karachi, Pakistan.
| | - Affan Ahmed
- Dow Medical College, Dow University of Health Sciences, Karachi, Pakistan
| | - Fauzia Imtiaz
- Department of Biochemistry, Dow Medical College, Dow University of Health Sciences, Karachi, Pakistan
| |
Collapse
|
16
|
Zuliani I, Lanzillotta C, Tramutola A, Francioso A, Pagnotta S, Barone E, Perluigi M, Di Domenico F. The Dysregulation of OGT/OGA Cycle Mediates Tau and APP Neuropathology in Down Syndrome. Neurotherapeutics 2021; 18:340-363. [PMID: 33258073 PMCID: PMC8116370 DOI: 10.1007/s13311-020-00978-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2020] [Indexed: 12/12/2022] Open
Abstract
Protein O-GlcNAcylation is a nutrient-related post-translational modification that, since its discovery some 30 years ago, has been associated with the development of neurodegenerative diseases. As reported in Alzheimer's disease (AD), flaws in the cerebral glucose uptake translate into reduced hexosamine biosynthetic pathway flux and subsequently lead to aberrant protein O-GlcNAcylation. Notably, the reduction of O-GlcNAcylated proteins involves also tau and APP, thus promoting their aberrant phosphorylation in AD brain and the onset of AD pathological markers. Down syndrome (DS) individuals are characterized by the early development of AD by the age of 60 and, although the two conditions present the same pathological hallmarks and share the alteration of many molecular mechanisms driving brain degeneration, no evidence has been sought on the implication of O-GlcNAcylation in DS pathology. Our study aimed to unravel for the first time the role of protein O-GlcNacylation in DS brain alterations positing the attention of potential trisomy-related mechanisms triggering the aberrant regulation of OGT/OGA cycle. We demonstrate the disruption of O-GlcNAcylation homeostasis, as an effect of altered OGT and OGA regulatory mechanism, and confirm the relevance of O-GlcNAcylation in the appearance of AD hallmarks in the brain of a murine model of DS. Furthermore, we provide evidence for the neuroprotective effects of brain-targeted OGA inhibition. Indeed, the rescue of OGA activity was able to restore protein O-GlcNAcylation, and reduce AD-related hallmarks and decreased protein nitration, possibly as effect of induced autophagy.
Collapse
Affiliation(s)
- Ilaria Zuliani
- Department of Biochemical Sciences "A. Rossi Fanelli", Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Chiara Lanzillotta
- Department of Biochemical Sciences "A. Rossi Fanelli", Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Antonella Tramutola
- Department of Biochemical Sciences "A. Rossi Fanelli", Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Antonio Francioso
- Department of Biochemical Sciences "A. Rossi Fanelli", Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Sara Pagnotta
- Department of Biochemical Sciences "A. Rossi Fanelli", Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Eugenio Barone
- Department of Biochemical Sciences "A. Rossi Fanelli", Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Marzia Perluigi
- Department of Biochemical Sciences "A. Rossi Fanelli", Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy
| | - Fabio Di Domenico
- Department of Biochemical Sciences "A. Rossi Fanelli", Laboratory affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, P.le Aldo Moro 5, 00185, Rome, Italy.
| |
Collapse
|
17
|
Ye F, Tian S, Hu H, Yu Z. Electroacupuncture reduces scopolamine-induced amnesia via mediating the miR-210/SIN3A and miR-183/SIN3A signaling pathway. Mol Med 2020; 26:107. [PMID: 33183243 PMCID: PMC7661264 DOI: 10.1186/s10020-020-00233-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 10/23/2020] [Indexed: 02/07/2023] Open
Abstract
Background The expression of SIN3A is closely correlated with electroacupuncture (EA) treatment efficacy of scopolamine-induced amnesia (SIA), but its underlying mechanisms remain to be further explored. Methods Quantitative real-time PCR was performed to analyze the expression of candidate microRNAs (miRNAs) and SIN3A mRNA in a rat model of SIA. Western blot was carried out to evaluate the differential expression of SIN3A proteins under different circumstances. Luciferase assay was used to explore the inhibitory role of certain miRNAs in SIN3A expression. A novel object recognition (NOR) test was performed to assess the memory function of SIA rats undergoing EA treatment. Immunohistochemistry was carried out to evaluate the expression of SIN3A in the hippocampus of SIA rats. Results Rno-miR-183-5p, rno-miR-34c-3p and rno-miR-210-3p were significantly up-regulated in SIA rats treated with EA. In addition, rno-miR-183-5p and rno-miR-210-3p exerted an inhibitory effect on SIN3A expression. EA treatment of SIA rats effectively restored the dysregulated expression of rno-miR-183-5p, rno-miR-210-3p and SIN3A. EA treatment also promoted the inhibited expression of neuronal IEGs including Arc, Egr1, Homer1 and Narp in the hippocampus of SIA rats. Accordingly, the NOR test also confirmed the effect of EA treatment on the improvement of memory in SIA rats. Conclusion In summary, the findings of this study demonstrated that scopolamine-induced amnesia was associated with downregulated expression of miR-210/miR-183 and upregulated expression of SIN3A. Furthermore, treatment with EA alleviated scopolamine-induced amnesia in rats and was associated with upregulated expression of miR-210/miR-183 and downregulated expression of SIN3A.
Collapse
Affiliation(s)
- Fan Ye
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No.136, Jingzhou Street, Xiangcheng District, Xiangyang, 441021, Hubei, China
| | - Shiming Tian
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No.136, Jingzhou Street, Xiangcheng District, Xiangyang, 441021, Hubei, China
| | - Huimin Hu
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No.136, Jingzhou Street, Xiangcheng District, Xiangyang, 441021, Hubei, China.
| | - Zhengwen Yu
- Department of Anesthesiology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, No.136, Jingzhou Street, Xiangcheng District, Xiangyang, 441021, Hubei, China.
| |
Collapse
|
18
|
Kong Y, Liu C, Zhou Y, Qi J, Zhang C, Sun B, Wang J, Guan Y. Progress of RAGE Molecular Imaging in Alzheimer's Disease. Front Aging Neurosci 2020; 12:227. [PMID: 32848706 PMCID: PMC7417350 DOI: 10.3389/fnagi.2020.00227] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 06/29/2020] [Indexed: 12/19/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disease characterized by senile plaques (SPs), which are caused by amyloid beta (Aβ) deposition and neurofibrillary tangles (NFTs) of abnormal hyperphosphorylated tau protein. The receptor for advanced glycation end products (RAGE) binds to advanced glycation end products deposited during vascular dysfunction. Alzheimer’s disease may occur when RAGE binds to Aβ and releases reactive oxygen species, further exacerbating Aβ deposition and eventually leading to SPs and NFTs. As it is involved in early AD, RAGE may be considered as a more potent biomarker than Aβ. Positron emission tomography provides valuable information regarding the underlying pathological processes of AD many years before the appearance of clinical symptoms. Thus, to further reveal the role of RAGE in AD pathology and for early diagnosis of AD, a tracer that targets RAGE is needed. In this review, we first describe the early diagnosis of AD and then summarize the interaction between RAGE and Aβ and Tau that is required to induce AD pathology, and finally focus on RAGE-targeting probes, highlighting the potential of RAGE to be used as an effective target. The development of RAGE probes is expected to aid in AD diagnosis and treatment.
Collapse
Affiliation(s)
- Yanyan Kong
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| | - Cuiping Liu
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Yinping Zhou
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Jingxuan Qi
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Chencheng Zhang
- Department of Neurosurgery, Center for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bomin Sun
- Department of Neurosurgery, Center for Functional Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiao Wang
- Laboratory of Molecular Neural Biology, School of Life Sciences, Shanghai University, Shanghai, China
| | - Yihui Guan
- PET Center, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
19
|
Adenosine and Metabotropic Glutamate Receptors Are Present in Blood Serum and Exosomes from SAMP8 Mice: Modulation by Aging and Resveratrol. Cells 2020; 9:cells9071628. [PMID: 32645849 PMCID: PMC7407497 DOI: 10.3390/cells9071628] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/27/2020] [Accepted: 07/02/2020] [Indexed: 12/23/2022] Open
Abstract
Adenosine (ARs) and metabotropic glutamate receptors (mGluRs) are G-protein coupled receptors (GPCRs) that are modulated in the brain of SAMP8 mice, an animal model of Alzheimer's disease (AD). In the present work, it is shown the presence of ARs and mGluRs in blood serum and derived exosomes from SAMP8 mice as well as its possible modulation by aging and resveratrol (RSV) consumption. In blood serum, adenosine A1 and A2A receptors remained unaltered from 5 to 7 months of age. However, an age-related decrease in adenosine level was observed, while 5'-Nucleotidase activity was not modulated. Regarding the glutamatergic system, it was observed a decrease in mGluR5 density and glutamate levels in older mice. In addition, dietary RSV supplementation caused an age-dependent modulation in both adenosinergic and glutamatergic systems. These GPCRs were also found in blood serum-derived exosomes, which might suggest that these receptors could be released into circulation via exosomes. Interestingly, changes elicited by age and RSV supplementation on mGluR5 density, and adenosine and glutamate levels were similar to that detected in whole-brain. Therefore, we might suggest that the quantification of these receptors, and their corresponding endogenous ligands, in blood serum could have predictive value for early diagnosis in combination with other distinctive hallmarks of AD.
Collapse
|
20
|
Gupta A, Caravan P, Price WS, Platas-Iglesias C, Gale EM. Applications for Transition-Metal Chemistry in Contrast-Enhanced Magnetic Resonance Imaging. Inorg Chem 2020; 59:6648-6678. [PMID: 32367714 DOI: 10.1021/acs.inorgchem.0c00510] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Contrast-enhanced magnetic resonance imaging (MRI) is an indispensable tool for diagnostic medicine. However, safety concerns related to gadolinium in commercial MRI contrast agents have emerged in recent years. For patients suffering from severe renal impairment, there is an important unmet medical need to perform contrast-enhanced MRI without gadolinium. There are also concerns over the long-term effects of retained gadolinium within the general patient population. Demand for gadolinium-free MRI contrast agents is driving a new wave of inorganic chemistry innovation as researchers explore paramagnetic transition-metal complexes as potential alternatives. Furthermore, advances in personalized care making use of molecular-level information have motivated inorganic chemists to develop MRI contrast agents that can detect pathologic changes at the molecular level. Recent studies have highlighted how reaction-based modulation of transition-metal paramagnetism offers a highly effective mechanism to achieve MRI contrast enhancement that is specific to biochemical processes. This Viewpoint highlights how recent advances in transition-metal chemistry are leading the way for a new generation of MRI contrast agents.
Collapse
Affiliation(s)
- Abhishek Gupta
- Nanoscale Organisation and Dynamics Group, School of Science and Health, Western Sydney University, Penrith, New South Wales 2751, Australia.,Ingham Institute of Applied Medical Research, Liverpool, New South Wales 2170, Australia
| | | | - William S Price
- Nanoscale Organisation and Dynamics Group, School of Science and Health, Western Sydney University, Penrith, New South Wales 2751, Australia.,Ingham Institute of Applied Medical Research, Liverpool, New South Wales 2170, Australia
| | - Carlos Platas-Iglesias
- Centro de Investigacións Científicas Avanzadas and Departamento de Química, Facultade de Ciencias, Universidade da Coruña, A Coruña, Galicia 15071, Spain
| | | |
Collapse
|
21
|
Qin T, Prins S, Groeneveld GJ, Van Westen G, de Vries HE, Wong YC, Bischoff LJ, de Lange EC. Utility of Animal Models to Understand Human Alzheimer's Disease, Using the Mastermind Research Approach to Avoid Unnecessary Further Sacrifices of Animals. Int J Mol Sci 2020; 21:ijms21093158. [PMID: 32365768 PMCID: PMC7247586 DOI: 10.3390/ijms21093158] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/18/2022] Open
Abstract
To diagnose and treat early-stage (preclinical) Alzheimer’s disease (AD) patients, we need body-fluid-based biomarkers that reflect the processes that occur in this stage, but current knowledge on associated processes is lacking. As human studies on (possible) onset and early-stage AD would be extremely expensive and time-consuming, we investigate the potential value of animal AD models to help to fill this knowledge gap. We provide a comprehensive overview of processes associated with AD pathogenesis and biomarkers, current knowledge on AD-related biomarkers derived from on human and animal brains and body fluids, comparisons of biomarkers obtained in human AD and frequently used animal AD models, and emerging body-fluid-based biomarkers. In human studies, amyloid beta (Aβ), hyperphosphorylated tau (P-tau), total tau (T-tau), neurogranin, SNAP-25, glial fibrillary acidic protein (GFAP), YKL-40, and especially neurofilament light (NfL) are frequently measured. In animal studies, the emphasis has been mostly on Aβ. Although a direct comparison between human (familial and sporadic) AD and (mostly genetic) animal AD models cannot be made, still, in brain, cerebrospinal fluid (CSF), and blood, a majority of similar trends are observed for human AD stage and animal AD model life stage. This indicates the potential value of animal AD models in understanding of the onset and early stage of AD. Moreover, animal studies can be smartly designed to provide mechanistic information on the interrelationships between the different AD processes in a longitudinal fashion and may also include the combinations of different conditions that may reflect comorbidities in human AD, according to the Mastermind Research approach.
Collapse
Affiliation(s)
- Tian Qin
- Predictive Pharmacology, Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre of Drug Research, Leiden University, 2333 CC Leiden, The Netherlands; (T.Q.); (L.J.M.B.)
| | - Samantha Prins
- Centre for Human Drug Research (CHDR), 2333 CL Leiden, The Netherlands; (S.P.); (G.J.G.)
| | - Geert Jan Groeneveld
- Centre for Human Drug Research (CHDR), 2333 CL Leiden, The Netherlands; (S.P.); (G.J.G.)
| | - Gerard Van Westen
- Computational Drug Discovery, Division of Drug Discovery and Safety, Leiden Academic Centre of Drug Research, Leiden University, 2333 CC Leiden, The Netherlands;
| | - Helga E. de Vries
- Neuro-immunology research group, Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, 1081 HZ Amsterdam, The Netherlands;
| | - Yin Cheong Wong
- Advanced Modelling and Simulation, UCB Celltech, Slough SL1 3WE, UK;
| | - Luc J.M. Bischoff
- Predictive Pharmacology, Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre of Drug Research, Leiden University, 2333 CC Leiden, The Netherlands; (T.Q.); (L.J.M.B.)
| | - Elizabeth C.M. de Lange
- Predictive Pharmacology, Division of Systems Biomedicine and Pharmacology, Leiden Academic Centre of Drug Research, Leiden University, 2333 CC Leiden, The Netherlands; (T.Q.); (L.J.M.B.)
- Correspondence: ; Tel.: +31-71-527-6330
| |
Collapse
|
22
|
Wright JW, Harding JW. Contributions by the Brain Renin-Angiotensin System to Memory, Cognition, and Alzheimer's Disease. J Alzheimers Dis 2020; 67:469-480. [PMID: 30664507 DOI: 10.3233/jad-181035] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive neuron losses in memory-associated brain structures that rob patients of their dignity and quality of life. Five drugs have been approved by the FDA to treat AD but none modify or significantly slow disease progression. New therapies are needed to delay the course of this disease with the ultimate goal of preventing neuron losses and preserving memory functioning. In this review we describe the renin-angiotensin II (AngII) system (RAS) with specific regard to its deleterious contributions to hypertension, facilitation of neuroinflammation and oxidative stress, reduced cerebral blood flow, tissue remodeling, and disruption of memory consolidation and retrieval. There is evidence that components of the RAS, AngIV and Ang(1-7), are positioned to counter such damaging influences and these systems are detailed with the goal of drawing attention to their importance as drug development targets. Ang(1-7) binds at the Mas receptor, while AngIV binds at the AT4 receptor subtype, and these receptor numbers are significantly decreased in AD patients, accompanied by declines in brain aminopeptidases A and N, enzymes essential for the synthesis of AngIV. Potent analogs may be useful to counter these changes and facilitate neuronal functioning and reduce apoptosis in memory associated brain structures of AD patients.
Collapse
Affiliation(s)
- John W Wright
- Department of Psychology, Washington State University, Pullman, WA, USA.,Department of Integrative Physiology and Neuroscience, and Program in Biotechnology, Washington State University, Pullman, WA, USA.,M3 Biotechnology, Inc., Seattle, WA, USA
| | - Joseph W Harding
- Department of Psychology, Washington State University, Pullman, WA, USA.,Department of Integrative Physiology and Neuroscience, and Program in Biotechnology, Washington State University, Pullman, WA, USA.,M3 Biotechnology, Inc., Seattle, WA, USA
| |
Collapse
|
23
|
Cha DJ, Mengel D, Mustapic M, Liu W, Selkoe DJ, Kapogiannis D, Galasko D, Rissman RA, Bennett DA, Walsh DM. miR-212 and miR-132 Are Downregulated in Neurally Derived Plasma Exosomes of Alzheimer's Patients. Front Neurosci 2019; 13:1208. [PMID: 31849573 PMCID: PMC6902042 DOI: 10.3389/fnins.2019.01208] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 10/25/2019] [Indexed: 12/14/2022] Open
Abstract
It was recently discovered that brain cells release extracellular vesicles (EV) which can pass from brain into blood. These findings raise the possibility that brain-derived EV’s present in blood can be used to monitor disease processes occurring in the cerebrum. Since the levels of certain micro-RNAs (miRNAs) have been reported to be altered in Alzheimer’s disease (AD) brain, we sought to assess miRNA dysregulation in AD brain tissue and to determine if these changes were reflected in neural EVs isolated from blood of subjects with AD. To this end, we employed high-content miRNA arrays to search for differences in miRNAs in RNA pools from brain tissue of AD (n = 5), high pathological control (HPC) (n = 5), or cognitively intact pathology-free controls (n = 5). Twelve miRNAs were altered by >1.5-fold in AD compared to controls, and six of these were also changed compared to HPCs. Analysis of hits in brain extracts from 11 AD, 7 HPCs and 9 controls revealed a similar fold difference in these six miRNAs, with three showing statistically significant group differences and one with a strong trend toward group differences. Thereafter, we focused on the four miRNAs that showed group differences and measured their content in neurally derived blood EVs isolated from 63 subjects: 16 patients with early stage dementia and a CSF Aβ42+ tau profile consistent with AD, 16 individuals with mild cognitive impairment (MCI) and an AD CSF profile, and 31 cognitively intact controls with normal CSF Aβ42+ tau levels. ROC analysis indicated that measurement of miR-132-3p in neurally-derived plasma EVs showed good sensitivity and specificity to diagnose AD, but did not effectively separate individuals with AD-MCI from controls. Moreover, when we measured the levels of a related miRNA, miR-212, we found that this miRNA was also decreased in neural EVs from AD patients compared to controls. Our results suggest that measurement of miR-132 and miR-212 in neural EVs should be further investigated as a diagnostic aid for AD and as a potential theragnostic.
Collapse
Affiliation(s)
- Diana J Cha
- Laboratory for Neurodegenerative Disease Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - David Mengel
- Laboratory for Neurodegenerative Disease Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Department of Neurodegenerative Diseases, Hertie-Institute for Clinical Brain Research and Center of Neurology, University of Tübingen, Tübingen, Germany.,German Center for Neurodegenerative Diseases, University of Tübingen, Tübingen, Germany
| | - Maja Mustapic
- Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Wen Liu
- Laboratory for Neurodegenerative Disease Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Dennis J Selkoe
- Laboratory for Neurodegenerative Disease Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Dimitrios Kapogiannis
- Laboratory of Neurosciences, National Institute on Aging, National Institutes of Health, Baltimore, MD, United States
| | - Douglas Galasko
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States
| | - Robert A Rissman
- Department of Neurosciences, University of California, San Diego, La Jolla, CA, United States.,VA San Diego Healthcare System, La Jolla, CA, United States
| | - David A Bennett
- Rush Alzheimer's Disease Center, Rush Medical College, Chicago, IL, United States
| | - Dominic M Walsh
- Laboratory for Neurodegenerative Disease Research, Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States.,Alzheimer's Disease and Dementia Research Unit, Biogen Inc., Cambridge, MA, United States
| |
Collapse
|
24
|
Ricci G. Social Aspects of Dementia Prevention from a Worldwide to National Perspective: A Review on the International Situation and the Example of Italy. Behav Neurol 2019; 2019:8720904. [PMID: 31583024 PMCID: PMC6754867 DOI: 10.1155/2019/8720904] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 08/05/2019] [Accepted: 08/23/2019] [Indexed: 11/17/2022] Open
Abstract
At the moment, dementia is affecting around 47 million people worldwide, with a forecast amount of 135 million affected people in 2050. Dementia is a growing health concern worldwide with no treatment currently available, but only symptomatic medication. Effective interventions in the prevention and management of dementia are urgently needed to contain direct and indirect costs of this disease. Indeed, the economic impact of dementia is a vast and continually growing figure, but it is still difficult to quantify. Due to an increase in both the disease spreading and its direct and indirect costs, national and international action plans have to be implemented. As a virtuous example, the Italian national plan for dementia has been summarized. Faced with an increasingly less sustainable disease impact at national and international levels, the plan suggests that it is certainly the entire welfare model that should be rethought, strengthening the network of services and providing interventions to support affected people and their caregivers. Alongside this synergistic approach, scientific research could play a crucial role for pharmacological and nonpharmacological treatments capable of delaying the state of loss of self-sufficiency of the patient, with a significant impact on social and health costs.
Collapse
|
25
|
Tiepolt S, Patt M, Aghakhanyan G, Meyer PM, Hesse S, Barthel H, Sabri O. Current radiotracers to image neurodegenerative diseases. EJNMMI Radiopharm Chem 2019; 4:17. [PMID: 31659510 PMCID: PMC6660543 DOI: 10.1186/s41181-019-0070-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 07/16/2019] [Indexed: 12/11/2022] Open
Abstract
The term of neurodegenerative diseases covers a heterogeneous group of disorders that are distinguished by progressive degeneration of the structure and function of the nervous system such as dementias, movement disorders, motor neuron disorders, as well as some prion disorders. In recent years, a paradigm shift started for the diagnosis of neurodegenerative diseases, for which successively clinical testing is supplemented by biomarker information. In research scenarios, it was even proposed recently to substitute the current syndromic by a biological definition of Alzheimer's diseases. PET examinations with various radiotracers play an important role in providing non-invasive biomarkers and co-morbidity information in neurodegeneration. Information on co-morbidity, e.g. Aβ plaques and Lewy-bodies or Aβ plaques in patients with aphasia or the absence of Aβ plaques in clinical AD patients are of interest to expand our knowledge about the pathogenesis of different phenotypically defined neurodegenerative diseases. Moreover, this information is also important in therapeutic trials targeting histopathological abnormalities.The aim of this review is to present an overview of the currently available radiotracers for imaging neurodegenerative diseases in research and in routine clinical settings. In this context, we also provide a short summary of the most frequent neurodegenerative diseases from a nuclear medicine point of view, their clinical and pathophysiological as well as nuclear imaging characteristics, and the resulting need for new radiotracers.
Collapse
Affiliation(s)
- Solveig Tiepolt
- Department of Nuclear Medicine, University of Leipzig, Liebigstraße 18, 04103 Leipzig, Germany
| | - Marianne Patt
- Department of Nuclear Medicine, University of Leipzig, Liebigstraße 18, 04103 Leipzig, Germany
| | - Gayane Aghakhanyan
- Department of Nuclear Medicine, University of Leipzig, Liebigstraße 18, 04103 Leipzig, Germany
| | - Philipp M. Meyer
- Department of Nuclear Medicine, University of Leipzig, Liebigstraße 18, 04103 Leipzig, Germany
| | - Swen Hesse
- Department of Nuclear Medicine, University of Leipzig, Liebigstraße 18, 04103 Leipzig, Germany
| | - Henryk Barthel
- Department of Nuclear Medicine, University of Leipzig, Liebigstraße 18, 04103 Leipzig, Germany
| | - Osama Sabri
- Department of Nuclear Medicine, University of Leipzig, Liebigstraße 18, 04103 Leipzig, Germany
| |
Collapse
|
26
|
Tao PF, Huang HC. Regulation of AβPP Glycosylation Modification and Roles of Glycosylation on AβPP Cleavage in Alzheimer's Disease. ACS Chem Neurosci 2019; 10:2115-2124. [PMID: 30802027 DOI: 10.1021/acschemneuro.8b00574] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The presence of senile plaques in the gray matter of the brain is one of the major pathologic features of Alzheimer's disease (AD), and amyloid-β (Aβ) is the main component of extracellular deposits of the senile plaques. Aβ derives from amyloid-β precursor protein (AβPP) cleaved by β-secretase (BACE1) and γ-secretase, and the abnormal cleavage of AβPP is an important event leading to overproduction and aggregation of Aβ species. After translation, AβPP undergoes post-translational modifications (PTMs) including glycosylation and phosphorylation in the endoplasmic reticulum (ER) and Golgi apparatus, and these modifications play an important role in regulating the cleavage of this protein. In this Review, we summarize research progress on the modification of glycosylation, especially O-GlcNAcylation and mucin-type O-linked glycosylation (also known as O-GalNAcylation), on the regulation of AβPP cleavage and on the influence of AβPP's glycosylation in the pathogenesis of AD.
Collapse
Affiliation(s)
- Peng-Fei Tao
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, 100191, China
| | - Han-Chang Huang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, 100191, China
| |
Collapse
|
27
|
Optimization of solid-phase extraction (SPE) in the preparation of [18F]D3FSP: A new PET imaging agent for mapping Aβ plaques. Nucl Med Biol 2019; 71:54-64. [DOI: 10.1016/j.nucmedbio.2019.05.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 05/09/2019] [Accepted: 05/09/2019] [Indexed: 11/21/2022]
|
28
|
Li S, Cai Z, Wu X, Holden D, Pracitto R, Kapinos M, Gao H, Labaree D, Nabulsi N, Carson RE, Huang Y. Synthesis and in Vivo Evaluation of a Novel PET Radiotracer for Imaging of Synaptic Vesicle Glycoprotein 2A (SV2A) in Nonhuman Primates. ACS Chem Neurosci 2019; 10:1544-1554. [PMID: 30396272 PMCID: PMC6810685 DOI: 10.1021/acschemneuro.8b00526] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Structural disruption and alterations of synapses are associated with many brain disorders including Alzheimer's disease, epilepsy, depression, and schizophrenia. We have previously developed the PET radiotracer 11C-UCB-J for imaging and quantification of synaptic vesicle glycoprotein 2A (SV2A) and synaptic density in nonhuman primates and humans. Here we report the synthesis of a novel radiotracer 18F-SDM-8 and its in vivo evaluation in rhesus monkeys. The in vitro binding assay of SDM-8 showed high SV2A binding affinity ( Ki = 0.58 nM). 18F-SDM-8 was prepared in high molar activity (241.7 MBq/nmol) and radiochemical purity (>98%). In the brain, 18F-SDM-8 displayed very high uptake with peak standardized uptake value (SVU) greater than 8 and fast and reversible kinetics. A displacement study with levetiracetam and blocking studies with UCB-J and levetiracetam demonstrated its binding reversibility and specificity toward SV2A. Regional binding potential values were calculated and ranged from 0.8 in the brainstem to 4.5 in the cingulate cortex. By comparing to 11C-UCB-J, 18F-SDM-8 displayed the same attractive imaging properties: very high brain uptake, appropriate tissue kinetics, and high levels of specific binding. Given the longer half-life of F-18 and the feasibility for central production and multisite distribution, 18F-SDM-8 holds promise as an excellent radiotracer for SV2A and as a biomarker for synaptic density measurement in neurodegenerative diseases and psychiatric disorders.
Collapse
Affiliation(s)
- Songye Li
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Zhengxin Cai
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Xiaoai Wu
- Department of Nuclear Medicine, West China Hospital, Laboratory of Clinical Nuclear Medicine, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Daniel Holden
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Richard Pracitto
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Michael Kapinos
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Hong Gao
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - David Labaree
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Nabeel Nabulsi
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Richard E. Carson
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| | - Yiyun Huang
- PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, Connecticut 06520, United States
| |
Collapse
|
29
|
Cai Z, Li S, Matuskey D, Nabulsi N, Huang Y. PET imaging of synaptic density: A new tool for investigation of neuropsychiatric diseases. Neurosci Lett 2019; 691:44-50. [PMID: 30075287 PMCID: PMC6339829 DOI: 10.1016/j.neulet.2018.07.038] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 07/25/2018] [Accepted: 07/26/2018] [Indexed: 12/14/2022]
Abstract
Synaptic vesicle glycoprotein 2A (SV2A) is expressed ubiquitously in neurons of the central nervous system, and is the binding target of the anti-epileptic drug levetiracetam. Because of the availability of positron emission tomography (PET) ligands targeting SV2A, there is increasing enthusiasm on the use of SV2A PET to study a variety of neuropsychiatric diseases. This review discusses the recent development of radioligands for PET imaging of SV2A and their potential use in the research and diagnosis of CNS diseases.
Collapse
Affiliation(s)
- Zhengxin Cai
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA.
| | - Songye Li
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - David Matuskey
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Nabeel Nabulsi
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| | - Yiyun Huang
- PET Center, Department of Radiology and Biomedical Imaging, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
30
|
Braun DJ, Van Eldik LJ. In vivo Brainstem Imaging in Alzheimer's Disease: Potential for Biomarker Development. Front Aging Neurosci 2018; 10:266. [PMID: 30254583 PMCID: PMC6141632 DOI: 10.3389/fnagi.2018.00266] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Accepted: 08/17/2018] [Indexed: 12/25/2022] Open
Abstract
The dearth of effective treatments for Alzheimer's disease (AD) is one of the largest public health issues worldwide, costing hundreds of billions of dollars per year. From a therapeutic standpoint, research efforts to date have met with strikingly little clinical success. One major issue is that trials begin after substantial pathological change has occurred, and it is increasingly clear that the most effective treatment regimens will need to be administered earlier in the disease process. In order to identify individuals within the long preclinical phase of AD who are likely to progress to dementia, improvements are required in biomarker development. One potential area of research that might prove fruitful in this regard is the in vivo detection of brainstem pathology. The brainstem is known to undergo pathological changes very early and progressively in AD. With an updated and harmonized AD research framework, and emerging advances in neuroimaging technology, the potential to leverage knowledge of brainstem pathology into biomarkers for AD will be discussed.
Collapse
Affiliation(s)
- David J Braun
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
| | - Linda J Van Eldik
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States.,Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, United States.,Department of Neuroscience, University of Kentucky, Lexington, KY, United States
| |
Collapse
|
31
|
Novel targets in Alzheimer's disease: A special focus on microglia. Pharmacol Res 2018; 130:402-413. [PMID: 29391235 DOI: 10.1016/j.phrs.2018.01.017] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 01/12/2018] [Accepted: 01/26/2018] [Indexed: 02/07/2023]
Abstract
Several years after the intriguing novelty in the β-amyloid (Aβ) cascade hypothesis, where the Aβ oligomers emerged as the most detrimental species in the neuropathogenic process of Alzheimer's disease (AD) in place of fibrillar plaques, more recently innate immune system have come on stage as the other prominent factor. Neuroinflammation apparently contributes to AD eziopathogenesis, in large part through overactivation of microglia cells. Genetic and experimental studies strongly support the contribution of the immune system to increasing the risk of AD and participating in its progression. Besides the central immune response mediated by resident microglial cells, peripheral immune challenges may have profound negative effects on brain physiology as well, such as those originating from the gut microbiota. Despite the initial immune response to defend the organism, perpetuation seemingly turns into a chronic detrimental phenomenon that contributes to neuronal dysfunction and exacerbation of the disease. Several new immune-druggable targets are now under investigation, but much still remains to be defined about their precise role and whether and how their physiological activity changes in the injurious context of AD. From a therapeutic perspective, we can undoubtedly consider that AD is no longer solely an Aβ pathology, but rather a multifaceted disorder calling for multi-target therapies. New therapies fighting AD must still counteract Aβ but must also restore appropriate immune defences by tempering maladaptive factors and enabling beneficial responses.
Collapse
|
32
|
Henriques AD, Benedet AL, Camargos EF, Rosa-Neto P, Nóbrega OT. Fluid and imaging biomarkers for Alzheimer's disease: Where we stand and where to head to. Exp Gerontol 2018; 107:169-177. [PMID: 29307736 DOI: 10.1016/j.exger.2018.01.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 12/29/2017] [Accepted: 01/02/2018] [Indexed: 10/18/2022]
Abstract
There is increasing evidence that a number of potentially informative biomarkers for Alzheimer disease (AD) can improve the accuracy of diagnosing this form of dementia, especially when used as a panel of diagnostic assays and interpreted in the context of neuroimaging and clinical data. Moreover, by combining the power of CSF biomarkers with neuroimaging techniques to visualize Aβ deposits (or neurodegenerative lesions), it might be possible to better identify individuals at greatest risk for developing MCI and converting to AD. The objective of this article was to review recent progress in selected imaging and chemical biomarkers for prediction, early diagnosis and progression of AD. We present our view point of a scenario that places CSF and imaging markers on the verge of general utility based on accuracy levels that already match (or even surpass) current clinical precision.
Collapse
Affiliation(s)
- Adriane Dallanora Henriques
- Medical Centre for the Elderly, University Hospital, University of Brasília (UnB), 70910-900 Brasília, DF, Brazil
| | - Andrea Lessa Benedet
- Translational Neuroimaging Laboratory, Research Centre for Studies in Aging, Douglas Hospital, McGill University, H4H 1R3 Montreal, QC, Canada
| | - Einstein Francisco Camargos
- Medical Centre for the Elderly, University Hospital, University of Brasília (UnB), 70910-900 Brasília, DF, Brazil
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory, Research Centre for Studies in Aging, Douglas Hospital, McGill University, H4H 1R3 Montreal, QC, Canada; Montreal Neurological Institute, H3A 2B4 Montreal, QC, Canada
| | - Otávio Toledo Nóbrega
- Medical Centre for the Elderly, University Hospital, University of Brasília (UnB), 70910-900 Brasília, DF, Brazil.
| |
Collapse
|
33
|
Mercier J, Provins L, Valade A. Discovery and development of SV2A PET tracers: Potential for imaging synaptic density and clinical applications. DRUG DISCOVERY TODAY. TECHNOLOGIES 2017; 25:45-52. [PMID: 29233267 DOI: 10.1016/j.ddtec.2017.11.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 11/08/2017] [Accepted: 11/09/2017] [Indexed: 10/18/2022]
Abstract
Imaging synaptic density in vivo has promise for numerous research and clinical applications in the diagnosis and treatment monitoring of neurodegenerative and psychiatric diseases. Recent developments in the field of PET, such as SV2A human imaging with the novel tracers UCB-A, UCB-H and UCB-J, may help in realizing this potential and bring significant benefit for the patients suffering from these diseases. This review provides an overview of the most recent progress in the field of SV2A PET imaging, its potential for use as a biomarker of synaptic density and the future development areas.
Collapse
|
34
|
Rabiner EA. Imaging Synaptic Density: A Different Look at Neurologic Diseases. J Nucl Med 2017; 59:380-381. [PMID: 29123011 DOI: 10.2967/jnumed.117.198317] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 10/30/2017] [Indexed: 11/16/2022] Open
Affiliation(s)
- Eugenii A Rabiner
- Imanova Ltd. and Centre for Neuroimaging Sciences, Institute of Psychiatry, Psychology, and Neuroscience, King's College, London, United Kingdom
| |
Collapse
|
35
|
Liu W, Zhuo P, Li L, Jin H, Lin B, Zhang Y, Liang S, Wu J, Huang J, Wang Z, Lin R, Chen L, Tao J. Activation of brain glucose metabolism ameliorating cognitive impairment in APP/PS1 transgenic mice by electroacupuncture. Free Radic Biol Med 2017; 112:174-190. [PMID: 28756309 DOI: 10.1016/j.freeradbiomed.2017.07.024] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2016] [Revised: 07/24/2017] [Accepted: 07/25/2017] [Indexed: 02/06/2023]
Abstract
An essential feature of Alzheimer's disease (AD) is implicated in brain energy metabolic impairment that is considered underlying pathogenesis of cognitive impairment. Therefore, therapeutic interventions to allay cognitive deficits that target energy metabolism may be an efficacy strategy in AD. In this study, we found that electroacupuncture (EA) at the DU20 acupoint obviously increased glucose metabolism in specific brain regions such as cortex, hippocampus, cingulate gyrus, basal forebrain septum, brain stem, and cerebellum in APP/PS1 transgenic mice by animal 18F-Fluoro-2-deoxy-D-Glucose (18F-FDG)/positron emission tomography (PET) imaging, accompanied by cognitive improvements in the spatial reference learning and memory and memory flexibility and novel object recognition performances. Further evidence shown energy metabolism occurred in neurons or non-neuronal cells of the cortex and hippocampus in terms of the co-location of GLUT3/NeuN and GLUT1/GFAP. Simultaneously, metabolic homeostatic factors were critical for glucose metabolism, including phosphorylated adenosine monophosphate-activated protein kinase (AMPK) and AKT serine/threonine kinase. Furthermore, EA-induced phosphorylated AMPK and AKT inhibited the phosphorylation level of the mammalian target of rapamycin (mTOR) to decrease the accumulation of amyloid-beta (Aβ) in the cortex and hippocampus. These findings are concluded that EA is a potential therapeutic target for delaying memory decline and Aβ deposition of AD. The AMPK and AKT are implicated in the EA-induced cortical and hippocampal energy metabolism, which served as a contributor to improving cognitive function and Aβ deposition in a transgenic mouse model of AD.
Collapse
Affiliation(s)
- Weilin Liu
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Peiyuan Zhuo
- Fujian Key Laboratory of Rehabilitation Technology, Fuzhou, Fujian 350122, China
| | - Long Li
- Fujian Key Laboratory of Rehabilitation Technology, Fuzhou, Fujian 350122, China
| | - Hao Jin
- Fujian Key Laboratory of Rehabilitation Technology, Fuzhou, Fujian 350122, China
| | - Bingbing Lin
- Fujian Key Laboratory of Rehabilitation Technology, Fuzhou, Fujian 350122, China
| | - Yingzheng Zhang
- Fujian Key Laboratory of Rehabilitation Technology, Fuzhou, Fujian 350122, China
| | - Shengxiang Liang
- Division of Nuclear Technology and Applications, Institute of High Energy Physics, Chinese Academy of Sciences, Beijing 100049, China
| | - Jie Wu
- Fujian Key Laboratory of Rehabilitation Technology, Fuzhou, Fujian 350122, China
| | - Jia Huang
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China
| | - Zhifu Wang
- Fujian Key Laboratory of Rehabilitation Technology, Fuzhou, Fujian 350122, China
| | - Ruhui Lin
- Fujian Key Laboratory of Rehabilitation Technology, Fuzhou, Fujian 350122, China
| | - Lidian Chen
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China.
| | - Jing Tao
- College of Rehabilitation Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, China.
| |
Collapse
|
36
|
Jellinger KA. Potential clinical utility of multiple system atrophy biomarkers. Expert Rev Neurother 2017; 17:1189-1208. [DOI: 10.1080/14737175.2017.1392239] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
37
|
Sgambato-Faure V, Billard T, Météreau E, Duperrier S, Fieux S, Costes N, Tremblay L, Zimmer L. Characterization and Reliability of [ 18F]2FNQ1P in Cynomolgus Monkeys as a PET Radiotracer for Serotonin 5-HT 6 Receptors. Front Pharmacol 2017; 8:471. [PMID: 28769801 PMCID: PMC5513908 DOI: 10.3389/fphar.2017.00471] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2017] [Accepted: 07/03/2017] [Indexed: 01/08/2023] Open
Abstract
Brain serotonin-6 receptor (5-HT6R) is the one of the most recently identified serotonin receptors. Accumulating evidence suggests that it is a potent therapeutic target for psychiatric and neurological diseases. Since [18F]2FNQ1P was recently proposed as the first fluorinated positron emission tomography (PET) radioligand for this receptor, the objective of the present study was to demonstrate its suitability for 5-HT6R neuroimaging in primates. [18F]2FNQ1P was characterized by in vitro autoradiography and in vivo PET imaging in cynomolgus monkeys. Following in vivo PET imaging, tracer binding indices were computed using the simplified reference tissue model and Logan graphical model, with cerebellum as reference region. The tracer binding reproducibility was assessed by test–retest in five animals. Finally, specificity was assessed by pre-injection of a 5-HT6R antagonist, SB258585. In vitro, results showed wide cerebral distribution of the tracer with specificity toward 5-HT6Rs as binding was effectively displaced by SB258585. In vivo brain penetration was good with reproducible distribution at cortical and subcortical levels. The automated method gave the best spatial normalization. The Logan graphical model showed the best tracer binding indices, giving the highest magnitude, lowest standard deviation and best reproducibility and robustness. Finally, 5-HT6R antagonist pre-injection significantly decreased [18F]2FNQ1P binding mainly in the striatum and sensorimotor cortex. Taken together, these preclinical results show that [18F]2FNQ1P is a good candidate to address 5-HT6 receptors in clinical studies.
Collapse
Affiliation(s)
- Véronique Sgambato-Faure
- Université Claude Bernard Lyon ILyon, France.,Institut des Sciences Cognitives Marc Jeannerod, CNRS UMR5229Bron, France
| | - Thierry Billard
- Université Claude Bernard Lyon ILyon, France.,Institut de Chimie et de Biochimie, CNRS UMR5246Villeurbanne, France
| | - Elise Météreau
- Université Claude Bernard Lyon ILyon, France.,Institut des Sciences Cognitives Marc Jeannerod, CNRS UMR5229Bron, France
| | - Sandra Duperrier
- Université Claude Bernard Lyon ILyon, France.,Institut des Sciences Cognitives Marc Jeannerod, CNRS UMR5229Bron, France
| | - Sylvain Fieux
- Université Claude Bernard Lyon ILyon, France.,Centre de Recherche en Neurosciences de Lyon, CNRS UMR5292, INSERM U1028Lyon, France
| | | | - Léon Tremblay
- Université Claude Bernard Lyon ILyon, France.,Institut des Sciences Cognitives Marc Jeannerod, CNRS UMR5229Bron, France
| | - Luc Zimmer
- Université Claude Bernard Lyon ILyon, France.,Centre de Recherche en Neurosciences de Lyon, CNRS UMR5292, INSERM U1028Lyon, France.,CERMEP-Imagerie du VivantLyon, France.,Hospices Civils de LyonLyon, France
| |
Collapse
|
38
|
Lacerda S, Morfin JF, Geraldes CFGC, Tóth É. Metal complexes for multimodal imaging of misfolded protein-related diseases. Dalton Trans 2017; 46:14461-14474. [DOI: 10.1039/c7dt02371e] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Aggregation of misfolded proteins and progressive polymerization of otherwise soluble proteins is a common hallmark of several highly debilitating and increasingly prevalent diseases, including amyotrophic lateral sclerosis, cerebral amyloid angiopathy, type II diabetes and Parkinson's, Huntington's and Alzheimer's diseases.
Collapse
Affiliation(s)
- S. Lacerda
- Centre de Biophysique Moléculaire
- CNRS
- UPR 4301
- Université d'Orléans
- 45071 Orléans Cedex 2
| | - J.-F. Morfin
- Centre de Biophysique Moléculaire
- CNRS
- UPR 4301
- Université d'Orléans
- 45071 Orléans Cedex 2
| | - C. F. G. C. Geraldes
- Department of Life Sciences
- Faculty of Sciences and Technology
- University of Coimbra
- 3000-393 Coimbra
- Portugal
| | - É. Tóth
- Centre de Biophysique Moléculaire
- CNRS
- UPR 4301
- Université d'Orléans
- 45071 Orléans Cedex 2
| |
Collapse
|