1
|
Singh S A, Ansari MN, M. Elossaily G, Vellapandian C, Prajapati B. Investigating the Potential Impact of Air Pollution on Alzheimer's Disease and the Utility of Multidimensional Imaging for Early Detection. ACS OMEGA 2024; 9:8615-8631. [PMID: 38434844 PMCID: PMC10905749 DOI: 10.1021/acsomega.3c06328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 12/25/2023] [Accepted: 01/30/2024] [Indexed: 03/05/2024]
Abstract
Pollution is ubiquitous, and much of it is anthropogenic in nature, which is a severe risk factor not only for respiratory infections or asthma sufferers but also for Alzheimer's disease, which has received a lot of attention recently. This Review aims to investigate the primary environmental risk factors and their profound impact on Alzheimer's disease. It underscores the pivotal role of multidimensional imaging in early disease identification and prevention. Conducting a comprehensive review, we delved into a plethora of literature sources available through esteemed databases, including Science Direct, Google Scholar, Scopus, Cochrane, and PubMed. Our search strategy incorporated keywords such as "Alzheimer Disease", "Alzheimer's", "Dementia", "Oxidative Stress", and "Phytotherapy" in conjunction with "Criteria Pollutants", "Imaging", "Pathology", and "Particulate Matter". Alzheimer's disease is not only a result of complex biological factors but is exacerbated by the infiltration of airborne particles and gases that surreptitiously breach the nasal defenses to traverse the brain, akin to a Trojan horse. Various imaging modalities and noninvasive techniques have been harnessed to identify disease progression in its incipient stages. However, each imaging approach possesses inherent limitations, prompting exploration of a unified technique under a single umbrella. Multidimensional imaging stands as the linchpin for detecting and forestalling the relentless march of Alzheimer's disease. Given the intricate etiology of the condition, identifying a prospective candidate for Alzheimer's disease may take decades, rendering the development of a multimodal imaging technique an imperative. This research underscores the pressing need to recognize the chronic ramifications of invisible particulate matter and to advance our understanding of the insidious environmental factors that contribute to Alzheimer's disease.
Collapse
Affiliation(s)
- Ankul Singh S
- Department
of Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology (SRMIST), Kattankulathur, Tamil Nadu 603203, India
| | - Mohd Nazam Ansari
- Department
of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Gehan M. Elossaily
- Department
of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 13713, Saudi Arabia
| | - Chitra Vellapandian
- Department
of Pharmacology, SRM College of Pharmacy, SRM Institute of Science and Technology (SRMIST), Kattankulathur, Tamil Nadu 603203, India
| | - Bhupendra Prajapati
- Department
of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy,
Shree S.K. Patel College of Pharmaceutical Education and Research, Ganpat University, Gozaria Highway, Mehsana, North Gujarat 384012, India
| |
Collapse
|
2
|
Stamenkovic S, Li Y, Waters J, Shih A. Deep Imaging to Dissect Microvascular Contributions to White Matter Degeneration in Rodent Models of Dementia. Stroke 2023; 54:1403-1415. [PMID: 37094035 PMCID: PMC10460612 DOI: 10.1161/strokeaha.122.037156] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2023]
Abstract
The increasing socio-economic burden of Alzheimer disease (AD) and AD-related dementias has created a pressing need to define targets for therapeutic intervention. Deficits in cerebral blood flow and neurovascular function have emerged as early contributors to disease progression. However, the cause, progression, and consequence of small vessel disease in AD/AD-related dementias remains poorly understood, making therapeutic targets difficult to pinpoint. Animal models that recapitulate features of AD/AD-related dementias may provide mechanistic insight because microvascular pathology can be studied as it develops in vivo. Recent advances in in vivo optical and ultrasound-based imaging of the rodent brain facilitate this goal by providing access to deeper brain structures, including white matter and hippocampus, which are more vulnerable to injury during cerebrovascular disease. Here, we highlight these novel imaging approaches and discuss their potential for improving our understanding of vascular contributions to AD/AD-related dementias.
Collapse
Affiliation(s)
- Stefan Stamenkovic
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Yuandong Li
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, USA
| | - Jack Waters
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Andy Shih
- Center for Developmental Biology and Regenerative Medicine, Seattle Children’s Research Institute, Seattle, WA, USA
- Department of Pediatrics, University of Washington, Seattle, WA, USA
- Allen Institute for Brain Science, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| |
Collapse
|
3
|
Shih NC, Kurniawan ND, Cabeen RP, Korobkova L, Wong E, Chui HC, Clark KA, Miller CA, Hawes D, Jones KT, Sepehrband F. Microstructural mapping of dentate gyrus pathology in Alzheimer's disease: A 16.4 Tesla MRI study. Neuroimage Clin 2023; 37:103318. [PMID: 36630864 PMCID: PMC9841366 DOI: 10.1016/j.nicl.2023.103318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/07/2023]
Abstract
The dentate gyrus (DG) is an integral portion of the hippocampal formation, and it is composed of three layers. Quantitative magnetic resonance (MR) imaging has the capability to map brain tissue microstructural properties which can be exploited to investigate neurodegeneration in Alzheimer's disease (AD). However, assessing subtle pathological changes within layers requires high resolution imaging and histological validation. In this study, we utilized a 16.4 Tesla scanner to acquire ex vivo multi-parameter quantitative MRI measures in human specimens across the layers of the DG. Using quantitative diffusion tensor imaging (DTI) and multi-parameter MR measurements acquired from AD (N = 4) and cognitively normal control (N = 6) tissues, we performed correlation analyses with histological measurements. Here, we found that quantitative MRI measures were significantly correlated with neurofilament and phosphorylated Tau density, suggesting sensitivity to layer-specific changes in the DG of AD tissues.
Collapse
Affiliation(s)
- Nien-Chu Shih
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Nyoman D Kurniawan
- Center for Advanced Imaging, The University of Queensland, Brisbane 4072, Australia
| | - Ryan P Cabeen
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Laura Korobkova
- Neuroscience Graduate Program, University of Southern California, Los Angeles, CA 90089. USA
| | - Ellen Wong
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Neurology, Rancho Los Amigos National Rehabilitation Center, Downey, CA 90242, USA
| | - Helena C Chui
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Kristi A Clark
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Carol A Miller
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Debra Hawes
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA; Department of Pathology and Laboratory Medicine, Children's Hospital of Los Angeles, Los Angeles, CA 90033, USA
| | - Kymry T Jones
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Farshid Sepehrband
- USC Stevens Neuroimaging and Informatics Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
4
|
Falangola MF, Nie X, Voltin J, Ward R, Dhiman S, Nietert PJ, Jensen JH. Brain microstructure abnormalities in the 3xTg-AD mouse - A diffusion MRI and morphology correlation study. Magn Reson Imaging 2022; 94:48-55. [PMID: 36116712 PMCID: PMC9695071 DOI: 10.1016/j.mri.2022.09.002] [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: 07/06/2022] [Revised: 08/26/2022] [Accepted: 09/13/2022] [Indexed: 10/14/2022]
Abstract
The widely studied triple transgenic (3xTg-AD) mouse provides a robust model of Alzheimer's disease (AD) with region dependent patterns of progressive amyloid-β (Aß) and tau pathology. Using diffusion MRI (dMRI), we investigated the sensitivity of dMRI measures in capturing AD pathology associated microstructure alterations in older 3xTg-AD mice, and the degree to which dMRI changes correlate with measurements of Aβ and tau pathology. 3xTg-AD and normal control (NC) mice, 15 to 21 months of age, were used in this study. In vivo dMRI data were acquired for the generation of diffusion tensor (DT) and diffusional kurtosis (DK) measures within the hippocampus and fimbria (Fi). For these same brain regions, Aβ and tau pathology were quantified by morphological analysis of Aß1-42 and AT8 immunoreactivity. Two-tailed, two-sample t-tests were performed to assess group differences in each brain region of interest (ROI), with the Benjamini-Hochberg false discovery rate (FDR) method being applied to adjust for multiple comparisons. Spearman correlation coefficients were calculated to investigate associations between diffusion and morphological measures. Our results revealed, depending on the brain region, DT and DK measures were able to detect group differences. In the dorsal hippocampus (HD), fractional anisotropy (FA) was significantly higher in the 3xTg-AD mice compared with NC mice. In the subiculum (SUB), FA, axial diffusivity (D||) and radial kurtosis (K┴) were significantly higher in 3xTg-AD mice compared with NC mice. Morphological quantification of Aß1-42 and AT8 immunoreactivity showed elevated Aß and tau in the Fi, ventral hippocampus (HV) and SUB of 3xTg-AD mice. The presence of Aβ and tau was significantly correlated with several DT and DK measures, particularly in the SUB, where an increase in tau correlated with an increase in mean kurtosis (MK) and K┴. This work demonstrates significant dMRI differences between older 3xTg-AD and NC mice in the hippocampus and Fi. Significant correlations were found between dMRI and morphological measures of Aβ and tau pathology. These results support the potential of dMRI-derived parameters as biomarkers of AD pathology. Since the imaging methods employed here are easily translatable to clinical MRI, our results are also relevant for human AD patients.
Collapse
Affiliation(s)
- Maria Fatima Falangola
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA; Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC, USA.
| | - Xingju Nie
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA; Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC, USA
| | - Joshua Voltin
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA; Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC, USA
| | - Ralph Ward
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Siddhartha Dhiman
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA
| | - Paul J Nietert
- Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - Jens H Jensen
- Department of Neuroscience, Medical University of South Carolina, Charleston, SC, USA; Center for Biomedical Imaging, Medical University of South Carolina, Charleston, SC, USA; Department of Radiology and Radiological Science, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
5
|
Sarkar D, Kang J, Wassie AT, Schroeder ME, Peng Z, Tarr TB, Tang AH, Niederst ED, Young JZ, Su H, Park D, Yin P, Tsai LH, Blanpied TA, Boyden ES. Revealing nanostructures in brain tissue via protein decrowding by iterative expansion microscopy. Nat Biomed Eng 2022; 6:1057-1073. [PMID: 36038771 PMCID: PMC9551354 DOI: 10.1038/s41551-022-00912-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Accepted: 06/22/2022] [Indexed: 12/25/2022]
Abstract
Many crowded biomolecular structures in cells and tissues are inaccessible to labelling antibodies. To understand how proteins within these structures are arranged with nanoscale precision therefore requires that these structures be decrowded before labelling. Here we show that an iterative variant of expansion microscopy (the permeation of cells and tissues by a swellable hydrogel followed by isotropic hydrogel expansion, to allow for enhanced imaging resolution with ordinary microscopes) enables the imaging of nanostructures in expanded yet otherwise intact tissues at a resolution of about 20 nm. The method, which we named 'expansion revealing' and validated with DNA-probe-based super-resolution microscopy, involves gel-anchoring reagents and the embedding, expansion and re-embedding of the sample in homogeneous swellable hydrogels. Expansion revealing enabled us to use confocal microscopy to image the alignment of pre-synaptic calcium channels with post-synaptic scaffolding proteins in intact brain circuits, and to uncover periodic amyloid nanoclusters containing ion-channel proteins in brain tissue from a mouse model of Alzheimer's disease. Expansion revealing will enable the further discovery of previously unseen nanostructures within cells and tissues.
Collapse
Affiliation(s)
- Deblina Sarkar
- Media Lab, MIT, Cambridge, MA, 02139, USA.,MIT Center for Neurobiological Engineering, MIT, Cambridge, MA, 02139, USA.,These authors contributed equally
| | - Jinyoung Kang
- MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, 02139, USA.,These authors contributed equally
| | - Asmamaw T Wassie
- Department of Biological Engineering, MIT, Cambridge, MA, 02139, USA.,These authors contributed equally
| | - Margaret E. Schroeder
- MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, 02139, USA.,Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, 02139, USA
| | - Zhuyu Peng
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, 02139, USA.,The Picower Institute for Learning and Memory, MIT, Cambridge, MA, 02139, USA
| | - Tyler B. Tarr
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - Ai-Hui Tang
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,CAS Key Laboratory of Brain Function and Disease, Biomedical Sciences and Health Laboratory of Anhui Province, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230031, China
| | - Emily D. Niederst
- The Picower Institute for Learning and Memory, MIT, Cambridge, MA, 02139, USA
| | - Jennie, Z. Young
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, 02139, USA.,The Picower Institute for Learning and Memory, MIT, Cambridge, MA, 02139, USA
| | - Hanquan Su
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA.,Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Demian Park
- MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, 02139, USA
| | - Peng Yin
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02115, USA.,Department of Systems Biology, Harvard Medical School, Boston, MA, 02115, USA
| | - Li-Huei Tsai
- Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, USA. .,The Picower Institute for Learning and Memory, MIT, Cambridge, MA, USA. .,Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Thomas A. Blanpied
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.,Correspondence and requests for materials should be addressed to Li-Huei Tsai, Thomas A. Blanpied or Edward S. Boyden. , ,
| | - Edward S. Boyden
- MIT Center for Neurobiological Engineering, MIT, Cambridge, MA, 02139, USA.,MIT McGovern Institute for Brain Research, MIT, Cambridge, MA, 02139, USA.,Department of Biological Engineering, MIT, Cambridge, MA, 02139, USA.,Department of Brain and Cognitive Sciences, MIT, Cambridge, MA, 02139, USA.,Koch Institute, MIT, Cambridge, MA, 02139, USA.,Howard Hughes Medical Institute, Cambridge, MA, 02139, USA.,Media Arts and Sciences, MIT, Cambridge, MA, 02139, USA.,K. Lisa Yang Center for Bionics, MIT, Cambridge, MA, 02139, USA.,Correspondence and requests for materials should be addressed to Li-Huei Tsai, Thomas A. Blanpied or Edward S. Boyden. , ,
| |
Collapse
|
6
|
Ahn K, Lee SJ, Mook-Jung I. White matter-associated microglia: New players in brain aging and neurodegenerative diseases. Ageing Res Rev 2022; 75:101574. [PMID: 35093614 DOI: 10.1016/j.arr.2022.101574] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 12/09/2021] [Accepted: 01/24/2022] [Indexed: 12/20/2022]
Abstract
There has been growing interest in brain aging and rejuvenation. It is well known that brain aging is one of the leading causes of neurodegenerative diseases, such as Alzheimer's disease, but brain aging alone can cause cognitive decline. Microglia are thought to act as 'conductors' of white matter aging by modulating diverse glial cells and phagocytosing white matter-derived myelin debris. A recent study identified a specific subpopulation of microglia in the white matter of aged mice, termed white matter-associated microglia (WAM). Additionally, senescent microglia show impaired phagocytic function and altered lipid metabolism, which cause accumulation of lipid metabolites and eventually lead to myelin sheath degeneration. These results suggest that senescent WAM could be pivotal players in axonal loss during brain aging. The aim of this review is to assess the current state of knowledge on brain aging, with an emphasis on the roles of the white matter and microglia, and suggest potential approaches for rejuvenating the aged brain.
Collapse
Affiliation(s)
- Kyusik Ahn
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea
| | - Seung-Jae Lee
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; SNU Dementia Research Center, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Inhee Mook-Jung
- Department of Biomedical Sciences, College of Medicine, Seoul National University, Seoul 03080, Republic of Korea; Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, Republic of Korea; SNU Dementia Research Center, Seoul National University College of Medicine, Seoul 03080, Korea.
| |
Collapse
|
7
|
Ni R. Magnetic Resonance Imaging in Animal Models of Alzheimer's Disease Amyloidosis. Int J Mol Sci 2021; 22:12768. [PMID: 34884573 PMCID: PMC8657987 DOI: 10.3390/ijms222312768] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/18/2021] [Accepted: 11/23/2021] [Indexed: 02/07/2023] Open
Abstract
Amyloid-beta (Aβ) plays an important role in the pathogenesis of Alzheimer's disease. Aberrant Aβ accumulation induces neuroinflammation, cerebrovascular alterations, and synaptic deficits, leading to cognitive impairment. Animal models recapitulating the Aβ pathology, such as transgenic, knock-in mouse and rat models, have facilitated the understanding of disease mechanisms and the development of therapeutics targeting Aβ. There is a rapid advance in high-field MRI in small animals. Versatile high-field magnetic resonance imaging (MRI) sequences, such as diffusion tensor imaging, arterial spin labeling, resting-state functional MRI, anatomical MRI, and MR spectroscopy, as well as contrast agents, have been developed for preclinical imaging in animal models. These tools have enabled high-resolution in vivo structural, functional, and molecular readouts with a whole-brain field of view. MRI has been used to visualize non-invasively the Aβ deposits, synaptic deficits, regional brain atrophy, impairment in white matter integrity, functional connectivity, and cerebrovascular and glymphatic system in animal models of Alzheimer's disease amyloidosis. Many of the readouts are translational toward clinical MRI applications in patients with Alzheimer's disease. In this review, we summarize the recent advances in MRI for visualizing the pathophysiology in amyloidosis animal models. We discuss the outstanding challenges in brain imaging using MRI in small animals and propose future outlook in visualizing Aβ-related alterations in the brains of animal models.
Collapse
Affiliation(s)
- Ruiqing Ni
- Institute for Biomedical Engineering, ETH Zurich & University of Zurich, 8093 Zurich, Switzerland;
- Institute for Regenerative Medicine, University of Zurich, 8952 Zurich, Switzerland
| |
Collapse
|
8
|
Kara F, Belloy ME, Voncken R, Sarwari Z, Garima Y, Anckaerts C, Langbeen A, Leysen V, Shah D, Jacobs J, Hamaide J, Bols P, Van Audekerke J, Daans J, Guglielmetti C, Kantarci K, Prevot V, Roßner S, Ponsaerts P, Van der Linden A, Verhoye M. Long-term ovarian hormone deprivation alters functional connectivity, brain neurochemical profile and white matter integrity in the Tg2576 amyloid mouse model of Alzheimer's disease. Neurobiol Aging 2021; 102:139-150. [PMID: 33765427 PMCID: PMC8312737 DOI: 10.1016/j.neurobiolaging.2021.02.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 02/05/2021] [Accepted: 02/16/2021] [Indexed: 01/18/2023]
Abstract
Premenopausal bilateral ovariectomy is considered to be one of the risk factors of Alzheimer's disease (AD). However, the underlying mechanisms remain unclear. Here, we aimed to investigate long-term neurological consequences of ovariectomy in a rodent AD model, TG2576 (TG), and wild-type mice (WT) that underwent an ovariectomy or sham-operation, using in vivo MRI biomarkers. An increase in osmoregulation and energy metabolism biomarkers in the hypothalamus, a decrease in white matter integrity, and a decrease in the resting-state functional connectivity was observed in ovariectomized TG mice compared to sham-operated TG mice. In addition, we observed an increase in functional connectivity in ovariectomized WT mice compared to sham-operated WT mice. Furthermore, genotype (TG vs. WT) effects on imaging markers and GFAP immunoreactivity levels were observed, but there was no effect of interaction (Genotype × Surgery) on amyloid-beta-and GFAP immunoreactivity levels. Taken together, our results indicated that both genotype and ovariectomy alters imaging biomarkers associated with AD.
Collapse
Affiliation(s)
- Firat Kara
- Bio-imaging Lab- Member of INMIND consortium, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
| | - Michael E Belloy
- Bio-imaging Lab- Member of INMIND consortium, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Rick Voncken
- Bio-imaging Lab- Member of INMIND consortium, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Zahra Sarwari
- Bio-imaging Lab- Member of INMIND consortium, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Yadav Garima
- Bio-imaging Lab- Member of INMIND consortium, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Cynthia Anckaerts
- Bio-imaging Lab- Member of INMIND consortium, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - An Langbeen
- Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Valerie Leysen
- Univ. Lille, Inserm, CHU Lille, Development and Plasticity of the Neuroendocrine Brain, Lille Neurosciences and Cognition, UMR-S1172, DistalZ, Lille, France
| | - Disha Shah
- Bio-imaging Lab- Member of INMIND consortium, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Jules Jacobs
- University of Nijmegen, Nijmegen, the Netherlands
| | - Julie Hamaide
- Bio-imaging Lab- Member of INMIND consortium, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Peter Bols
- Veterinary Physiology and Biochemistry, Department of Veterinary Sciences, University of Antwerp, Antwerp, Belgium
| | - Johan Van Audekerke
- Bio-imaging Lab- Member of INMIND consortium, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Jasmijn Daans
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | | | - Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Vincent Prevot
- Univ. Lille, Inserm, CHU Lille, Development and Plasticity of the Neuroendocrine Brain, Lille Neurosciences and Cognition, UMR-S1172, DistalZ, Lille, France
| | - Steffen Roßner
- Paul Flechsig Institute of Brain Research, Leipzig University, Leipzig, Germany
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Antwerp, Belgium
| | - Annemie Van der Linden
- Bio-imaging Lab- Member of INMIND consortium, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Marleen Verhoye
- Bio-imaging Lab- Member of INMIND consortium, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
9
|
Wang P, Zhou B, Yao H, Xie S, Feng F, Zhang Z, Guo Y, An N, Zhou Y, Zhang X, Liu Y. Aberrant Hippocampal Functional Connectivity Is Associated with Fornix White Matter Integrity in Alzheimer's Disease and Mild Cognitive Impairment. J Alzheimers Dis 2021; 75:1153-1168. [PMID: 32390630 DOI: 10.3233/jad-200066] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is the most common cause of dementia in older individuals, and amnestic mild cognitive impairment (aMCI) is currently considered the prodromal stage of AD. The hippocampus and fornix interact functionally and structurally, with the fornix being the major efferent white matter tract from the hippocampus. OBJECTIVE The main aim of this study was to examine the impairments present in subjects with AD or aMCI and the relationship of these impairments with the microstructure of the fornix and the functional connectivity (FC) and gray matter volume of the hippocampus. METHODS Forty-four AD, 34 aMCI, and 41 age- and gender-matched normal controls (NCs) underwent neuropsychological assessments and multimode MRI. We chose the bilateral hippocampi as the region of interest in which gray matter alterations and FC with the whole brain were assessed and the fornix body as the region of interest in which the microstructural integrity of the white matter was observed. We also evaluated the relationship among gray matter alterations, the abnormal FC of the hippocampus and the integrity of the fornix in AD/aMCIResults:Compared to the NC group, the AD and aMCI groups demonstrated decreased gray matter volume, reduced FC between the bilateral hippocampi and several brain regions in the default mode network and control network, and damaged integrity of the fornix body (decreased fractional anisotropy and increased diffusivity). We also found that left hippocampal FC with some regions, the integrity of the fornix body, and cognition ability were significantly correlated. Therefore, our findings suggest that damage to white matter integrity may partially explain the reduced resting-state FC of the hippocampus in AD and aMCI. CONCLUSION AD and aMCI are diseases of disconnectivity including not only functional but also structural disconnectivity. Damage to white matter integrity may partially explain the reduced resting-state FC in AD and aMCI. These findings have significant implications for diagnostics and modeling and provide insights for understanding the disconnection syndrome in AD.
Collapse
Affiliation(s)
- Pan Wang
- Department of Neurology, Tianjin Huanhu Hospital, Nankai University, Tianjin, China.,Department of Neurology, The Second Medical Centre, National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Bo Zhou
- Department of Neurology, The Second Medical Centre, National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Hongxiang Yao
- Department of Radiology, The Second Medical Centre, National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Sangma Xie
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing, China.,National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China.,College of Life Information Science and Instrument Engineering, Hangzhou Dianzi University, Hangzhou, China
| | - Feng Feng
- Department of Neurology, The Second Medical Centre, National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Zengqiang Zhang
- Hainan Hospital of Chinese PLA General Hospital, Sanya, China
| | - Yan'e Guo
- Department of Neurology, The Second Medical Centre, National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Ningyu An
- Department of Radiology, The Second Medical Centre, National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Yuying Zhou
- Department of Neurology, Tianjin Huanhu Hospital, Nankai University, Tianjin, China
| | - Xi Zhang
- Department of Neurology, The Second Medical Centre, National Clinical Research Centre for Geriatric Diseases, Chinese PLA General Hospital, Beijing, China
| | - Yong Liu
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing, China.,National Laboratory of Pattern Recognition, Institute of Automation, Chinese Academy of Sciences, Beijing, China.,CAS Center for Excellence in Brain Science and Intelligence Technology, Institute of Automation, Chinese Academy of Sciences, Beijing, China.,School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
10
|
Chen Q, Baran TM, Rooks B, O'Banion MK, Mapstone M, Zhang Z, Lin F. Cognitively supernormal older adults maintain a unique structural connectome that is resistant to Alzheimer's pathology. Neuroimage Clin 2020; 28:102413. [PMID: 32971466 PMCID: PMC7511768 DOI: 10.1016/j.nicl.2020.102413] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 08/30/2020] [Accepted: 09/02/2020] [Indexed: 11/20/2022]
Abstract
Studying older adults with excellent cognitive capacities (Supernormals) provides a unique opportunity for identifying factors related to cognitive success - a critical topic across lifespan. There is a limited understanding of Supernormals' neural substrates, especially whether any of them attends shaping and supporting superior cognitive function or confer resistance to age-related neurodegeneration such as Alzheimer's disease (AD). Here, applying a state-of-the-art diffusion imaging processing pipeline and finite mixture modelling, we longitudinally examine the structural connectome of Supernormals. We find a unique structural connectome, containing the connections between frontal, cingulate, parietal, temporal, and subcortical regions in the same hemisphere that remains stable over time in Supernormals, relatively to typical agers. The connectome significantly classifies positive vs. negative AD pathology at 72% accuracy in a new sample mixing Supernormals, typical agers, and AD risk [amnestic mild cognitive impairment (aMCI)] subjects. Among this connectome, the mean diffusivity of the connection between right isthmus cingulate cortex and right precuneus most robustly contributes to predicting AD pathology across samples. The mean diffusivity of this connection links negatively to global cognition in those Supernormals with positive AD pathology. But this relationship does not exist in typical agers or aMCI. Our data suggest the presence of a structural connectome supporting cognitive success. Cingulate to precuneus white matter integrity may be useful as a structural marker for monitoring neurodegeneration and may provide critical information for understanding how some older adults maintain or excel cognitively in light of significant AD pathology.
Collapse
Affiliation(s)
- Quanjing Chen
- Elaine C. Hubbard Center for Nursing Research on Aging, School of Nursing, University of Rochester Medical Center, United States; Department of Psychiatry, School of Medicine and Dentistry, University of Rochester Medical Center, United States.
| | - Timothy M Baran
- Department of Imaging Sciences, School of Medicine and Dentistry, University of Rochester Medical Center, United States; Department of Biomedical Engineering, University of Rochester, United States
| | - Brian Rooks
- Department of Biostatistics and Computational Biology, School of Medicine and Dentistry, University of Rochester Medical Center, United States
| | - M Kerry O'Banion
- Department of Neuroscience, School of Medicine and Dentistry, University of Rochester Medical Center, United States
| | - Mark Mapstone
- Department of Neurology, University of California-Irvine, United States
| | - Zhengwu Zhang
- Department of Biostatistics and Computational Biology, School of Medicine and Dentistry, University of Rochester Medical Center, United States
| | - Feng Lin
- Elaine C. Hubbard Center for Nursing Research on Aging, School of Nursing, University of Rochester Medical Center, United States; Department of Psychiatry, School of Medicine and Dentistry, University of Rochester Medical Center, United States; Department of Neuroscience, School of Medicine and Dentistry, University of Rochester Medical Center, United States; Department of Neurology, School of Medicine and Dentistry, University of Rochester Medical Center, United States; Department of Brain and Cognitive Sciences, University of Rochester, United States.
| |
Collapse
|
11
|
Müller HP, Roselli F, Rasche V, Kassubek J. Diffusion Tensor Imaging-Based Studies at the Group-Level Applied to Animal Models of Neurodegenerative Diseases. Front Neurosci 2020; 14:734. [PMID: 32982659 PMCID: PMC7487414 DOI: 10.3389/fnins.2020.00734] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 06/22/2020] [Indexed: 12/11/2022] Open
Abstract
The understanding of human and non-human microstructural brain alterations in the course of neurodegenerative diseases has substantially improved by the non-invasive magnetic resonance imaging (MRI) technique of diffusion tensor imaging (DTI). Animal models (including disease or knockout models) allow for a variety of experimental manipulations, which are not applicable to humans. Thus, the DTI approach provides a promising tool for cross-species cross-sectional and longitudinal investigations of the neurobiological targets and mechanisms of neurodegeneration. This overview with a systematic review focuses on the principles of DTI analysis as used in studies at the group level in living preclinical models of neurodegeneration. The translational aspect from in-vivo animal models toward (clinical) applications in humans is covered as well as the DTI-based research of the non-human brains' microstructure, the methodological aspects in data processing and analysis, and data interpretation at different abstraction levels. The aim of integrating DTI in multiparametric or multimodal imaging protocols will allow the interrogation of DTI data in terms of directional flow of information and may identify the microstructural underpinnings of neurodegeneration-related patterns.
Collapse
Affiliation(s)
| | - Francesco Roselli
- Department of Neurology, University of Ulm, Ulm, Germany.,German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| | - Volker Rasche
- Core Facility Small Animal MRI, University of Ulm, Ulm, Germany
| | - Jan Kassubek
- Department of Neurology, University of Ulm, Ulm, Germany
| |
Collapse
|
12
|
Caballero MÁA, Song Z, Rubinski A, Duering M, Dichgans M, Park DC, Ewers M. Age‐dependent amyloid deposition is associated with white matter alterations in cognitively normal adults during the adult life span. Alzheimers Dement 2020; 16:651-661. [DOI: 10.1002/alz.12062] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 12/10/2019] [Accepted: 01/03/2020] [Indexed: 01/01/2023]
Affiliation(s)
| | - Zhuang Song
- Center for Vital LongevityUniversity of Texas at Dallas Dallas Texas
| | - Anna Rubinski
- Institute for Stroke and Dementia ResearchUniversity HospitalLMU Munich Munich Germany
| | - Marco Duering
- Institute for Stroke and Dementia ResearchUniversity HospitalLMU Munich Munich Germany
| | - Martin Dichgans
- Institute for Stroke and Dementia ResearchUniversity HospitalLMU Munich Munich Germany
- Munich Cluster for Systems Neurology (SyNergy) Munich Germany
| | - Denise C. Park
- Center for Vital LongevityUniversity of Texas at Dallas Dallas Texas
| | - Michael Ewers
- Institute for Stroke and Dementia ResearchUniversity HospitalLMU Munich Munich Germany
| |
Collapse
|
13
|
Multiple inflammatory profiles of microglia and altered neuroimages in APP/PS1 transgenic AD mice. Brain Res Bull 2020; 156:86-104. [DOI: 10.1016/j.brainresbull.2020.01.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/14/2019] [Accepted: 01/03/2020] [Indexed: 12/11/2022]
|
14
|
Haller S, Montandon ML, Rodriguez C, Garibotto V, Lilja J, Herrmann FR, Giannakopoulos P. Amyloid Load, Hippocampal Volume Loss, and Diffusion Tensor Imaging Changes in Early Phases of Brain Aging. Front Neurosci 2019; 13:1228. [PMID: 31803008 PMCID: PMC6872975 DOI: 10.3389/fnins.2019.01228] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 10/30/2019] [Indexed: 01/23/2023] Open
Abstract
Background and Purpose Amyloid imaging, gray matter (GM) morphometry and diffusion tensor imaging (DTI) have all been used as predictive biomarkers in dementia. Our objective was to define the imaging profile of healthy elderly controls as a function of their cognitive trajectories and explore whether amyloid burden and white matter (WM) microstructure changes are associated with subtle decrement of neuropsychological performances in old age. Materials and Methods We performed a 4.5-year longitudinal study in 133 elderly individuals who underwent cognitive testing at inclusion and follow-up, amyloid PET, MRI including DTI sequences at inclusion, and APOE epsilon 4 genotyping. All cases were assessed using a continuous cognitive score (CCS) taking into account the global evolution of neuropsychological performances. Data processing included region of interest analysis of amyloid PET analysis, GM densities and tract-based spatial statistics (TBSS)-DTI. Regression models were built to explore the association between the CCS and imaging parameters controlling for significant demographic and clinical covariates. Results Amyloid uptake was not related to the cognitive outcome. In contrast, GM densities in bilateral hippocampus were associated with worst CCS at follow-up. In addition, radial and axial diffusivities in left hippocampus were negatively associated with CCS. Amyloid load was associated with decreased VBM and increased radial and axial diffusivity in the same area. These associations persisted when adjusting for gender and APOE4 genotype. Importantly, they were absent in amygdala and neocortical areas studied. Conclusion The progressive decrement of neuropsychological performances in normal aging is associated with volume loss and WM microstructure changes in hippocampus long before the emergence of clinically overt symptoms. Higher amyloid load in hippocampus is compatible with cognitive preservation in cases with better preservation of GM densities and WM microstructure in this area.
Collapse
Affiliation(s)
- Sven Haller
- CIRD Centre d'Imagerie Rive Droite, Geneva, Switzerland.,Department of Surgical Sciences, Radiology, Uppsala University, Uppsala, Sweden.,Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Marie-Louise Montandon
- Department of Rehabilitation and Geriatrics, Geneva University Hospitals, University of Geneva, Geneva, Switzerland.,Department of Psychiatry, University of Geneva, Geneva, Switzerland
| | - Cristelle Rodriguez
- Department of Psychiatry, University of Geneva, Geneva, Switzerland.,Division of Institutional Measures, Medical Direction, Geneva University Hospitals, Geneva, Switzerland
| | - Valentina Garibotto
- Division of Nuclear Medicine and Molecular Imaging, Department of Diagnostic, Geneva University Hospitals, Geneva, Switzerland
| | - Johan Lilja
- Department of Surgical Sciences, Radiology, Uppsala University, Uppsala, Sweden.,Hermes Medical Solutions, Stockholm, Sweden
| | - François R Herrmann
- Department of Rehabilitation and Geriatrics, Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| | - Panteleimon Giannakopoulos
- Department of Psychiatry, University of Geneva, Geneva, Switzerland.,Division of Institutional Measures, Medical Direction, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
15
|
Tezel G, Timur SS, Bozkurt İ, Türkoğlu ÖF, Eroğlu İ, Nemutlu E, Öner L, Eroğlu H. A Snapshot on the Current Status of Alzheimer’s Disease, Treatment Perspectives, in-Vitro and in-Vivo Research Studies and Future Opportunities. Chem Pharm Bull (Tokyo) 2019; 67:1030-1041. [DOI: 10.1248/cpb.c19-00511] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Gizem Tezel
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University
| | - Selin Seda Timur
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University
| | | | - Ö. Faruk Türkoğlu
- Department of Neurosurgery, Ankara Atatürk Research and Education Hospital
| | - İpek Eroğlu
- Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Hacettepe University
| | - Emirhan Nemutlu
- Department of Analytical Chemistry, Faculty of Pharmacy, Hacettepe University
| | - Levent Öner
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University
| | - Hakan Eroğlu
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University
| |
Collapse
|
16
|
Neurodegeneration and Neuro-Regeneration-Alzheimer's Disease and Stem Cell Therapy. Int J Mol Sci 2019; 20:ijms20174272. [PMID: 31480448 PMCID: PMC6747457 DOI: 10.3390/ijms20174272] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/27/2019] [Accepted: 08/28/2019] [Indexed: 12/17/2022] Open
Abstract
Aging causes many changes in the human body, and is a high risk for various diseases. Dementia, a common age-related disease, is a clinical disorder triggered by neurodegeneration. Brain damage caused by neuronal death leads to cognitive decline, memory loss, learning inabilities and mood changes. Numerous disease conditions may cause dementia; however, the most common one is Alzheimer’s disease (AD), a futile and yet untreatable illness. Adult neurogenesis carries the potential of brain self-repair by an endogenous formation of newly-born neurons in the adult brain; however it also declines with age. Strategies to improve the symptoms of aging and age-related diseases have included different means to stimulate neurogenesis, both pharmacologically and naturally. Finally, the regulatory mechanisms of stem cells neurogenesis or a functional integration of newborn neurons have been explored to provide the basis for grafted stem cell therapy. This review aims to provide an overview of AD pathology of different neural and glial cell types and summarizes current strategies of experimental stem cell treatments and their putative future use in clinical settings.
Collapse
|
17
|
Sex-Specific Differences in Fat Storage, Development of Non-Alcoholic Fatty Liver Disease and Brain Structure in Juvenile HFD-Induced Obese Ldlr-/-.Leiden Mice. Nutrients 2019; 11:nu11081861. [PMID: 31405127 PMCID: PMC6723313 DOI: 10.3390/nu11081861] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 08/01/2019] [Accepted: 08/07/2019] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Sex-specific differences play a role in metabolism, fat storage in adipose tissue, and brain structure. At juvenile age, brain function is susceptible to the effects of obesity; little is known about sex-specific differences in juvenile obesity. Therefore, this study examined sex-specific differences in adipose tissue and liver of high-fat diet (HFD)-induced obese mice, and putative alterations between male and female mice in brain structure in relation to behavioral changes during the development of juvenile obesity. METHODS In six-week-old male and female Ldlr-/-.Leiden mice (n = 48), the impact of 18 weeks of HFD-feeding was examined. Fat distribution, liver pathology and brain structure and function were analyzed imunohisto- and biochemically, in cognitive tasks and with MRI. RESULTS HFD-fed female mice were characterized by an increased perigonadal fat mass, pronounced macrovesicular hepatic steatosis and liver inflammation. Male mice on HFD displayed an increased mesenteric fat mass, pronounced adipose tissue inflammation and microvesicular hepatic steatosis. Only male HFD-fed mice showed decreased cerebral blood flow and reduced white matter integrity. CONCLUSIONS At young age, male mice are more susceptible to the detrimental effects of HFD than female mice. This study emphasizes the importance of sex-specific differences in obesity, liver pathology, and brain function.
Collapse
|
18
|
Nishioka C, Liang HF, Barsamian B, Sun SW. Amyloid-beta induced retrograde axonal degeneration in a mouse tauopathy model. Neuroimage 2019; 189:180-191. [PMID: 30630081 DOI: 10.1016/j.neuroimage.2019.01.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 12/27/2018] [Accepted: 01/04/2019] [Indexed: 12/12/2022] Open
Abstract
White matter abnormalities, revealed by Diffusion Tensor Imaging (DTI), are observed in patients with Alzheimer's Disease (AD), representing neural network deficits that underlie gradual cognitive decline in patients. However, how DTI changes related to the development of Amyloid beta (Aβ) and tau pathology, two key hallmarks of AD, remain elusive. We hypothesized that tauopathy induced by Aβ could initiate an axonal degeneration, leading to DTI-detectable white matter abnormalities. We utilized the visual system of the transgenic p301L tau mice as a model system. Aβ was injected in Lateral Geniculate Nucleus (LGN), where the Retinal Ganglion Cell (RGC) axons terminate. Longitudinal DTI was conducted to detect changes in the optic tract (OT) and optic nerve (ON), containing the distal and proximal segments of RGC axons, respectively. Our results showed DTI changes in OT (significant 13.2% reduction in axial diffusion, AxD vs. vehicle controls) followed by significant alterations in ON AxD and fractional anisotropy, FA. Histology data revealed loss of synapses, RGC axons and cell bodies resulting from the Aβ injection. We further tested whether microtubule-stabilizing compound Epothilone D (EpoD) could ameliorate the damage. EpoD co-treatment with Aβ was sufficient to prevent Aβ-induced axon and cell loss. Using an acute injection paradigm, our data suggest that EpoD may mediate its protective effect by blocking localized, acute Aβ-induced tau phosphorylation. This study demonstrates white matter disruption resulting from localized Aβ, the importance of tau pathology induction to changes in white matter connectivity, and the use of EpoD as a potential therapeutic avenue to prevent the axon loss in AD.
Collapse
Affiliation(s)
- Christopher Nishioka
- Basic Sciences, School of Medicine, Loma Linda University, CA, USA; Neuroscience Graduate Program, University of California, Riverside, USA
| | - Hsiao-Fang Liang
- Basic Sciences, School of Medicine, Loma Linda University, CA, USA
| | - Barsam Barsamian
- Basic Sciences, School of Medicine, Loma Linda University, CA, USA; Neuroscience Graduate Program, University of California, Riverside, USA
| | - Shu-Wei Sun
- Basic Sciences, School of Medicine, Loma Linda University, CA, USA; Neuroscience Graduate Program, University of California, Riverside, USA; Pharmaceutical Science, School of Pharmacy, Loma Linda University, CA, USA.
| |
Collapse
|
19
|
Nirzhor SSR, Khan RI, Neelotpol S. The Biology of Glial Cells and Their Complex Roles in Alzheimer's Disease: New Opportunities in Therapy. Biomolecules 2018; 8:biom8030093. [PMID: 30201881 PMCID: PMC6164719 DOI: 10.3390/biom8030093] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 08/28/2018] [Accepted: 09/06/2018] [Indexed: 01/01/2023] Open
Abstract
Even though Alzheimer's disease (AD) is of significant interest to the scientific community, its pathogenesis is very complicated and not well-understood. A great deal of progress has been made in AD research recently and with the advent of these new insights more therapeutic benefits may be identified that could help patients around the world. Much of the research in AD thus far has been very neuron-oriented; however, recent studies suggest that glial cells, i.e., microglia, astrocytes, oligodendrocytes, and oligodendrocyte progenitor cells (NG2 glia), are linked to the pathogenesis of AD and may offer several potential therapeutic targets against AD. In addition to a number of other functions, glial cells are responsible for maintaining homeostasis (i.e., concentration of ions, neurotransmitters, etc.) within the central nervous system (CNS) and are crucial to the structural integrity of neurons. This review explores the: (i) role of glial cells in AD pathogenesis; (ii) complex functionalities of the components involved; and (iii) potential therapeutic targets that could eventually lead to a better quality of life for AD patients.
Collapse
|
20
|
Shen Z, Lei J, Li X, Wang Z, Bao X, Wang R. Multifaceted assessment of the APP/PS1 mouse model for Alzheimer's disease: Applying MRS, DTI, and ASL. Brain Res 2018; 1698:114-120. [PMID: 30077647 DOI: 10.1016/j.brainres.2018.08.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/28/2018] [Accepted: 08/01/2018] [Indexed: 01/01/2023]
Abstract
Transgenic animal models of Alzheimer's disease (AD) can mimic pathological and behavioral changes occurring in AD patients, and are usually viewed as the first choice for testing novel therapeutics. Validated biomarkers, particularly non-invasive ones, are urgently needed for AD diagnosis or evaluation of treatment results. However, there are few studies that systematically characterize pathological changes in AD animal models. Here, we investigated the brain of 8-month-old amyloid precursor protein/presenilin 1 (APP/PS1) transgenic and wild-type (WT) mice, employing 7.0-T magnetic resonance imaging (MRI). Magnetic resonance spectroscopy (MRS), diffusion tensor imaging (DTI), and arterial spin labeling (ASL) were obtained through micro-MRI scanning. After MRI examination in both transgenic (n = 12) and WT (n = 12) mice, immunohistochemical staining and ultrastructural analysis were subsequently performed. Cerebral blood flow (CBF) was significantly decreased in the left hippocampus, left thalamus, and right cortex of AD mice (P < 0.05). Moreover, MRS showed significantly changed NAA/Cr, Glu/Cr, and mI/Cr ratios in the hippocampus of transgenic mice. While only NAA/Cr and mI/Cr ratios varied significantly in the cortex of transgenic mice. Regarding DTI imaging, however, the values of FA, MD, DA and DR were not significantly different between transgenic and WT mice. Finally, it is worth noting that pathological damage of metabolism, CBF, and white matter was more distinct between transgenic and WT mice by pathological examination. Altogether, our results suggest that intravital imaging evaluation of 8-month-old APP/PS1 transgenic mice by MRS and ASL is an alternative tool for AD research.
Collapse
Affiliation(s)
- Zhiwei Shen
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jianfeng Lei
- Center for Medical Experiments and Testing, Capital Medical University, Beijing, China
| | - Xueyuan Li
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhanjing Wang
- Center for Medical Experiments and Testing, Capital Medical University, Beijing, China
| | - Xinjie Bao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| | - Renzhi Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.
| |
Collapse
|
21
|
Zhao YS, Zhang LH, Yu PP, Gou YJ, Zhao J, You LH, Wang ZY, Zheng X, Yan LJ, Yu P, Chang YZ. Ceruloplasmin, a Potential Therapeutic Agent for Alzheimer's Disease. Antioxid Redox Signal 2018; 28:1323-1337. [PMID: 28874056 DOI: 10.1089/ars.2016.6883] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
AIMS Ceruloplasmin (CP), a ferrous oxidase enzyme, plays an important role in regulating iron metabolism and redox reactions. Previous studies showed that CP deficiency contributes to Parkinson's disease by increasing iron accumulation and oxidative stress in the substantia nigra. However, the role of CP in Alzheimer's disease (AD) is unclear. We hypothesized that the lack of CP gene expression would affect the pathogenesis and damage of AD by promoting abnormal iron levels and oxidative stress. RESULTS AD mouse models were induced in CP knockout mouse either by injection of Aβ25-35 into the lateral ventricle of the brain or transgenic APP expression. CP levels were decreased significantly in the hippocampus of AD patients, as well as Aβ-CP+/+ and APP-CP+/+ mice. Compared to control AD mice, CP gene deletion increased memory impairment and iron accumulation, which could be associated with elevated reactive oxygen species (ROS) levels and lead to cell apoptosis mediated through the Bcl-2/Bax and Erk/p38 signaling pathways in Aβ-CP-/- and APP-CP-/- mice. In contrast, the restoration of CP expression to CP-/- mice through injection of an exogenous expression plasmid into the brain ventricle alleviated Aβ-induced neuronal damage in the hippocampus. INNOVATION CP alterations in iron contents were mediated through DMT1(-IRE) and changes in ROS levels, which in turn attenuated the progression of AD through the Erk/p38 and Bcl-2/Bax signaling pathways. CONCLUSION Our results show a protective role of CP in AD and suggest that regulating CP expression in the hippocampus may provide a new neuroprotective strategy for AD. Antioxid. Redox Signal. 28, 1323-1337.
Collapse
Affiliation(s)
- Ya-Shuo Zhao
- 1 Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University , Shijiazhuang, China .,2 Scientific Research Center, Hebei University of Chinese Medicine , Shijiazhuang, China
| | - Li-Hong Zhang
- 1 Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University , Shijiazhuang, China
| | - Pan-Pan Yu
- 1 Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University , Shijiazhuang, China
| | - Yu-Jing Gou
- 1 Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University , Shijiazhuang, China
| | - Jing Zhao
- 1 Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University , Shijiazhuang, China
| | - Lin-Hao You
- 1 Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University , Shijiazhuang, China
| | - Zhan-You Wang
- 3 College of Life and Health Sciences, Northeastern University , Shenyang, China
| | - Xin Zheng
- 1 Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University , Shijiazhuang, China
| | - Liang-Jun Yan
- 4 Department of Pharmaceutical Sciences, UNIT System College of Pharmacy, University of North Texas Health Science Center , Fort Worth, Texas
| | - Peng Yu
- 1 Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University , Shijiazhuang, China
| | - Yan-Zhong Chang
- 1 Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, College of Life Sciences, Hebei Normal University , Shijiazhuang, China .,5 Instrumental Analysis Center, Hebei Normal University , Shijiazhuang, China
| |
Collapse
|
22
|
Snow WM, Dale R, O'Brien-Moran Z, Buist R, Peirson D, Martin M, Albensi BC. In Vivo Detection of Gray Matter Neuropathology in the 3xTg Mouse Model of Alzheimer's Disease with Diffusion Tensor Imaging. J Alzheimers Dis 2018; 58:841-853. [PMID: 28505976 DOI: 10.3233/jad-170136] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
A diagnosis of Alzheimer's disease (AD), a neurodegenerative disorder accompanied by severe functional and cognitive decline, is based on clinical findings, with final confirmation of the disease at autopsy by the presence of amyloid-β (Aβ) plaques and neurofibrillary tangles. Given that microstructural brain alterations occur years prior to clinical symptoms, efforts to detect brain changes early could significantly enhance our ability to diagnose AD sooner. Diffusion tensor imaging (DTI), a type of MRI that characterizes the magnitude, orientation, and anisotropy of the diffusion of water in tissues, has been used to infer neuropathological changes in vivo. Its utility in AD, however, is still under investigation. The current study used DTI to examine brain regions susceptible to AD-related pathology; the cerebral cortex, entorhinal cortex, and hippocampus, in 12-14-month-old 3xTg AD mice that possess both Aβ plaques and neurofibrillary tangles. Mean diffusivity did not differ between 3xTg and control mice in any region. Decreased fractional anisotropy (p < 0.01) and axial diffusivity (p < 0.05) were detected only in the hippocampus, in which both congophilic Aβ plaques and hyperphosphorylated tau accumulation, consistent with neurofibrillary tangle formation, were detected. Pathological tau accumulation was seen in the cortex. The entorhinal cortex was largely spared from AD-related neuropathology. This is the first study to demonstrate DTI abnormalities in gray matter in a mouse model of AD in which both pathological hallmarks are present, suggesting the feasibility of DTI as a non-invasive means of detecting brain pathology in vivo in early-stage AD.
Collapse
Affiliation(s)
- Wanda M Snow
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada.,Department of Pharmacology & Therapeutics, University of Manitoba, Winnipeg, MB, Canada
| | - Ryan Dale
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada.,Department of Pharmacology & Therapeutics, University of Manitoba, Winnipeg, MB, Canada
| | | | - Richard Buist
- Department of Radiology, University of Manitoba, Winnipeg, MB, Canada
| | - Danial Peirson
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada.,Department of Pharmacology & Therapeutics, University of Manitoba, Winnipeg, MB, Canada
| | - Melanie Martin
- Department of Pharmacology & Therapeutics, University of Manitoba, Winnipeg, MB, Canada.,Department of Physics, University of Winnipeg, Winnipeg, MB, Canada.,Department of Radiology, University of Manitoba, Winnipeg, MB, Canada
| | - Benedict C Albensi
- Division of Neurodegenerative Disorders, St. Boniface Hospital Albrechtsen Research Centre, Winnipeg, MB, Canada.,Department of Pharmacology & Therapeutics, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
23
|
Whittaker HT, Zhu S, Di Curzio DL, Buist R, Li XM, Noy S, Wiseman FK, Thiessen JD, Martin M. T 1, diffusion tensor, and quantitative magnetization transfer imaging of the hippocampus in an Alzheimer's disease mouse model. Magn Reson Imaging 2018; 50:26-37. [PMID: 29545212 DOI: 10.1016/j.mri.2018.03.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 03/10/2018] [Indexed: 01/08/2023]
Abstract
Alzheimer's disease (AD) pathology causes microstructural changes in the brain. These changes, if quantified with magnetic resonance imaging (MRI), could be studied for use as an early biomarker for AD. The aim of our study was to determine if T1 relaxation, diffusion tensor imaging (DTI), and quantitative magnetization transfer imaging (qMTI) metrics could reveal changes within the hippocampus and surrounding white matter structures in ex vivo transgenic mouse brains overexpressing human amyloid precursor protein with the Swedish mutation. Delineation of hippocampal cell layers using DTI color maps allows more detailed analysis of T1-weighted imaging, DTI, and qMTI metrics, compared with segmentation of gross anatomy based on relaxation images, and with analysis of DTI or qMTI metrics alone. These alterations are observed in the absence of robust intracellular Aβ accumulation or plaque deposition as revealed by histology. This work demonstrates that multiparametric quantitative MRI methods are useful for characterizing changes within the hippocampal substructures and surrounding white matter tracts of mouse models of AD.
Collapse
Affiliation(s)
- Heather T Whittaker
- Biopsychology, University of Winnipeg, Winnipeg, MB R3B 2N2, Canada; Neurodegenerative Disease, University College London Institute of Neurology, London WC1N 3BG, United Kingdom.
| | - Shenghua Zhu
- Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB R3E 0T6, Canada
| | | | - Richard Buist
- Radiology, University of Manitoba, Winnipeg, MB R3E 0T6, Canada
| | - Xin-Min Li
- Psychiatry, University of Alberta, Alberta T6G 2R3, Canada
| | - Suzanna Noy
- Neurodegenerative Disease, University College London Institute of Neurology, London WC1N 3BG, United Kingdom
| | - Frances K Wiseman
- Neurodegenerative Disease, University College London Institute of Neurology, London WC1N 3BG, United Kingdom
| | - Jonathan D Thiessen
- Imaging Program, Lawson Health Research Institute, London, ON N6A 4V2, Canada; Medical Biophysics, Western University, London, Ontario, Canada
| | - Melanie Martin
- Pharmacology and Therapeutics, University of Manitoba, Winnipeg, MB R3E 0T6, Canada; Radiology, University of Manitoba, Winnipeg, MB R3E 0T6, Canada; Physics, University of Winnipeg, R3B 2N2, Canada
| |
Collapse
|
24
|
Wu D, Tang X, Gu LH, Li XL, Qi XY, Bai F, Chen XC, Wang JZ, Ren QG, Zhang ZJ. LINGO-1 antibody ameliorates myelin impairment and spatial memory deficits in the early stage of 5XFAD mice. CNS Neurosci Ther 2018; 24:381-393. [PMID: 29427384 DOI: 10.1111/cns.12809] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 12/03/2017] [Accepted: 01/03/2018] [Indexed: 12/14/2022] Open
Abstract
AIMS Multiple evidence has indicated that myelin injury is common in Alzheimer's disease (AD). However, whether myelin injury is an early event in AD and the relationship between it and cognitive function is still elusive. METHODS Spatial memory of 5XFAD mice was determined by Morris water maze at 1 and 3 months old. Meanwhile, the deposition of Aβ, the expression of myelin basic protein (MBP), LINGO-1, NgR, and myelin ultrastructure in many memory-associated brain regions were detected in one-month-old and three-month-old mice (before and after LINGO-1 antibody administration) using immunostaining, Western blot (WB), and transmission electron microscopy (TEM), respectively. RESULTS No abnormal Aβ deposition was found in one-month-old 5XFAD mice. However, spatial memory deficits were proved in accordance with an obvious demyelination in memory-associated brain regions in one-month-old mice and both deteriorated with age. Administration of LINGO-1 antibody could obviously restore the myelin impairments in CA1 and DG region and partially ameliorate spatial memory deficits. CONCLUSIONS Our results demonstrated that myelin injury was an early event in 5XFAD mice even prior to emergence of deposition of Aβ. Intervention with the LINGO-1 antibody could attenuate impaired spatial memory deficits by remyelination, which suggested that myelin injury was involved in spatial memory deficits and remyelination may be a potential therapeutic strategy in early stage of AD or mild cognitive impairments.
Collapse
Affiliation(s)
- Di Wu
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute, School of Medicine, Southeast University, Nanjing, China
| | - Xiang Tang
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute, School of Medicine, Southeast University, Nanjing, China
| | - Li-Hua Gu
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute, School of Medicine, Southeast University, Nanjing, China
| | - Xiao-Li Li
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute, School of Medicine, Southeast University, Nanjing, China
| | - Xin-Yang Qi
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute, School of Medicine, Southeast University, Nanjing, China
| | - Feng Bai
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute, School of Medicine, Southeast University, Nanjing, China
| | - Xiao-Chun Chen
- Department of Neurology and Geriatrics, Fujian Institute of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, China
| | - Jian-Zhi Wang
- Pathophysiology Department, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qing-Guo Ren
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute, School of Medicine, Southeast University, Nanjing, China
| | - Zhi-Jun Zhang
- Department of Neurology, Affiliated ZhongDa Hospital, Neuropsychiatric Institute, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
25
|
Liu LX, Du D, Wang ZQ, Fang Y, Zheng T, Dong YC, Shi QL, Zhao M, Xiao F, Du J. Differences in brain pathological changes between rotenone and 6-hydroxydopamine Parkinson's disease models. Neural Regen Res 2018; 13:1276-1280. [PMID: 30028338 PMCID: PMC6065241 DOI: 10.4103/1673-5374.235076] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Rotenone and 6-hydroxydopamine are two drugs commonly used to generate Parkinson's disease animal models. They not only achieve degenerative changes of dopaminergic neurons in the substantia nigra, but also satisfy the requirements for iron deposition. However, few studies have compared the characteristics of these two models by magnetic resonance imaging. In this study, rat models of Parkinson's disease were generated by injection of 3 μg rotenone or 10 μg 6-hydroxydopamine into the right substantia nigra. At 1, 2, 4, and 6 weeks after injection, coronal whole-brain T2-weighted imaging, transverse whole-brain T2-weighted imaging, and coronal diffusion tensor weighted imaging were conducted to measure fractional anisotropy and T2* values at the injury site. The fractional anisotropy value on the right side of the substantia nigra was remarkably lower at 6 weeks than at other time points in the rotenone group. In the 6-hydroxydopamine group, the fractional anisotropy value was decreased, but T2* values were increased on the right side of the substantia nigra at 1 week. Our findings confirm that the 6-hydroxydopamine-induced model is suitable for studying dopaminergic neurons over short periods, while the rotenone-induced model may be appropriate for studying the pathological and physiological processes of Parkinson's disease over long periods.
Collapse
Affiliation(s)
- Lan-Xiang Liu
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, Hebei Province, China
| | - Dan Du
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, Hebei Province, China
| | - Zhan-Qiu Wang
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, Hebei Province, China
| | - Yuan Fang
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, Hebei Province, China
| | - Tao Zheng
- Department of Magnetic Resonance Imaging, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, Hebei Province, China
| | - Yan-Chao Dong
- Department of Intervention, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, Hebei Province, China
| | | | - Min Zhao
- Department of Pathology, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, Hebei Province, China
| | - Fang Xiao
- Department of Pathology, Qinhuangdao Municipal No. 1 Hospital, Qinhuangdao, Hebei Province, China
| | - Juan Du
- Hebei Medical University, Shijiazhuang, Hebei Province, China
| |
Collapse
|
26
|
Reversal of age-related cognitive impairments in mice by an extremely low dose of tetrahydrocannabinol. Neurobiol Aging 2018; 61:177-186. [DOI: 10.1016/j.neurobiolaging.2017.09.025] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Revised: 09/18/2017] [Accepted: 09/23/2017] [Indexed: 01/28/2023]
|
27
|
Gu L, Wu D, Tang X, Qi X, Li X, Bai F, Chen X, Ren Q, Zhang Z. Myelin changes at the early stage of 5XFAD mice. Brain Res Bull 2017; 137:285-293. [PMID: 29288735 DOI: 10.1016/j.brainresbull.2017.12.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Revised: 12/08/2017] [Accepted: 12/26/2017] [Indexed: 02/08/2023]
Abstract
Previous studies have demonstrated myelin deficits in Alzheimer's disease (AD). However, it is still unclear whether myelin deficits occur at early stage of AD. Our study aimed to investigate myelin deficits in 5XFAD mice dynamically in different cognition-associated brain regions at early stage of AD. Transmission electron microscopy (TEM) was applied to detect myelin changes in late-myelinating regions such as prelimbic area (PrL), retrosplenial granular cortex (Rsg), field CA1 of hippocampus (CA1) and entorhinal cortex (ERC) respectively at different stages (1, 2, 3 and 5 months of age) in 5XFAD mouse model. In addition, we assessed spatial learning and memory with Morris water maze (MWM) in 5XFAD mice. Myelin deficits in 5XFAD mice started from 1 month of age and this deterioration continued during ageing, whereas the same myelin abnormality could only be observed in 5-month-old wild-type mice. Additionally, the g-ratio (an index associated with myelin thickness) was increased in 1-month-old 5XFAD mice in the regions including PrL, CA1 and ERC, compared to wild-type mice. As animals aged, the increased g-ratio in 5XFAD appeared in more regions of the brain. Moreover, 5XFAD mice showed spatial memory deficits from 1 month of age and spatial learning deficits from 2 months of age. In conclusion, myelin deficits occurred at an early stage and progressed with ageing in 5XFAD mouse model. Notably, a sequential myelin change was detected in cognition-associated brain regions. Combined with cognitive examinations, this study suggests that myelin changes might contribute to cognitive dysfunction.
Collapse
Affiliation(s)
- Lihua Gu
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Di Wu
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Xiang Tang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Xinyang Qi
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Xiaoli Li
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Feng Bai
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Xiaochun Chen
- Department of Neurology, Fujian Institute of Geriatrics, the Affiliated Union Hospital of Fujian Medical University, Fuzhou, Fujian, China
| | - Qingguo Ren
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China.
| | - Zhijun Zhang
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China.
| |
Collapse
|
28
|
Li J, Zhao C, Rao JS, Yang FX, Wang ZJ, Lei JF, Yang ZY, Li XG. Structural and metabolic changes in the traumatically injured rat brain: high-resolution in vivo proton magnetic resonance spectroscopy at 7 T. Neuroradiology 2017; 59:1203-1212. [PMID: 28856389 DOI: 10.1007/s00234-017-1915-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 08/22/2017] [Indexed: 11/28/2022]
Abstract
PURPOSE The understanding of microstructural and metabolic changes in the post-traumatic brain injury is the key to brain damage suppression and repair in clinics. METHODS Ten female Wistar rats were traumatically injured in the brain CA1 region and above the cortex. Next, diffusion tensor magnetic resonance imaging (DTI) and proton magnetic resonance spectroscopy (1H MRS) were used to analyze the microstructural and metabolic changes in the brain within the following 2 weeks. RESULTS Anisotropy fraction (FA) and axial diffusivity (AD) of the corpus callosum (CC) began to decrease significantly at day 1, whereas radial diffusivity (RD) significantly increased immediately after injury, reflecting the loss of white matter integrity. Compared with day 3, RD decreased significantly at day 7, implicating the angioedema reduction. In the hippocampus, FA significantly increased at day 7; the choline-containing compounds (Cho) and myo-inositol (MI) remarkably increased at day 7 compared with those at day 3, indicating the proliferation of astrocytes and radial glial cells after day 7. No significant differences between DTI and 1H MRS parameters were observed between day 1 and day 3. CONCLUSION Day 1-3 after traumatic brain injury (TBI) may serve as a relatively appropriate time window for treatment planning and the following nerve repair.
Collapse
Affiliation(s)
- Jing Li
- Beijing Key Laboratory for Biomaterials and Neural Regeneration, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Can Zhao
- Beijing Key Laboratory for Biomaterials and Neural Regeneration, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Jia-Sheng Rao
- Beijing Key Laboratory for Biomaterials and Neural Regeneration, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Fei-Xiang Yang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Zhan-Jing Wang
- Medical Experiment and Test Center, Capital Medical University, Beijing, 100069, China
| | - Jian-Feng Lei
- Medical Experiment and Test Center, Capital Medical University, Beijing, 100069, China
| | - Zhao-Yang Yang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiao-Guang Li
- Beijing Key Laboratory for Biomaterials and Neural Regeneration, School of Biological Science and Medical Engineering, Beihang University, Beijing, China. .,Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China.
| |
Collapse
|
29
|
Kantarci K, Murray ME, Schwarz CG, Reid RI, Przybelski SA, Lesnick T, Zuk SM, Raman MR, Senjem ML, Gunter JL, Boeve BF, Knopman DS, Parisi JE, Petersen RC, Jack CR, Dickson DW. White-matter integrity on DTI and the pathologic staging of Alzheimer's disease. Neurobiol Aging 2017; 56:172-179. [PMID: 28552181 DOI: 10.1016/j.neurobiolaging.2017.04.024] [Citation(s) in RCA: 133] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 04/07/2017] [Accepted: 04/25/2017] [Indexed: 11/16/2022]
Abstract
Pattern of diffusion tensor MRI (DTI) alterations were investigated in pathologically-staged Alzheimer's disease (AD) patients (n = 46). Patients with antemortem DTI studies and a range of AD pathology at autopsy were included. Patients with a high neurofibrillary tangle (NFT) stage (Braak IV-VI) had significantly elevated mean diffusivity (MD) in the crus of fornix and ventral cingulum tracts, precuneus, and entorhinal white matter on voxel-based analysis after adjusting for age and time from MRI to death (p < 0.001). Higher MD and lower fractional anisotropy in the ventral cingulum tract, entorhinal, and precuneus white matter was associated with higher Braak NFT stage and clinical disease severity. There were no MD and fractional anisotropy differences among the low (none and sparse) and high (moderate and frequent) β-amyloid neuritic plaque groups. The NFT pathology of AD is associated with DTI alterations involving the medial temporal limbic connections and medial parietal white matter. This pattern of diffusion abnormalities is also associated with clinical disease severity.
Collapse
Affiliation(s)
- Kejal Kantarci
- Department of Radiology, Mayo Clinic, Rochester, MN, USA.
| | - Melissa E Murray
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Jacksonville, FL, USA
| | | | - Robert I Reid
- Department of Information Technology, Mayo Clinic, Rochester, MN, USA
| | | | - Timothy Lesnick
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Samantha M Zuk
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Mekala R Raman
- Department of Radiology, Mayo Clinic, Rochester, MN, USA
| | - Matthew L Senjem
- Department of Information Technology, Mayo Clinic, Rochester, MN, USA
| | - Jeffrey L Gunter
- Department of Information Technology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Joseph E Parisi
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Rochester, MN, USA
| | | | | | - Dennis W Dickson
- Department of Pathology and Laboratory Medicine, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
30
|
Elman JA, Panizzon MS, Hagler DJ, Fennema-Notestine C, Eyler LT, Gillespie NA, Neale MC, Lyons MJ, Franz CE, McEvoy LK, Dale AM, Kremen WS. Genetic and environmental influences on cortical mean diffusivity. Neuroimage 2017; 146:90-99. [PMID: 27864081 PMCID: PMC5322245 DOI: 10.1016/j.neuroimage.2016.11.032] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 11/08/2016] [Accepted: 11/12/2016] [Indexed: 12/13/2022] Open
Abstract
Magnetic resonance imaging (MRI) has become an important tool in the early detection of age-related and neuropathological brain changes. Recent studies suggest that changes in mean diffusivity (MD) of cortical gray matter derived from diffusion MRI scans may be useful in detecting early effects of Alzheimer's disease (AD), and that these changes may be detected earlier than alterations associated with standard structural MRI measures such as cortical thickness. Thus, due to its potential clinical relevance, we examined the genetic and environmental influences on cortical MD in middle-aged men to provide support for the biological relevance of this measure and to guide future gene association studies. It is not clear whether individual differences in cortical MD reflect neuroanatomical variability similarly detected by other MRI measures, or whether unique features are captured. For instance, variability in cortical MD may reflect morphological variability more commonly measured by cortical thickness. Differences among individuals in cortical MD may also arise from breakdowns in myelinated fibers running through the cortical mantle. Thus, we investigated whether genetic influences on variation in cortical MD are the same or different from those influencing cortical thickness and MD of white matter (WM) subjacent to the cortical ribbon. Univariate twin analyses indicated that cortical MD is heritable in the majority of brain regions; the average of regional heritability estimates ranged from 0.38 in the cingulate cortex to 0.66 in the occipital cortex, consistent with the heritability of other MRI measures of the brain. Trivariate analyses found that, while there was some shared genetic variance between cortical MD and each of the other two measures, this overlap was not complete (i.e., the correlation was statistically different from 1). A significant amount of distinct genetic variance influences inter-individual variability in cortical MD; therefore, this measure could be useful for further investigation in studies of neurodegenerative diseases and gene association studies.
Collapse
Affiliation(s)
- Jeremy A Elman
- Department of Psychiatry, University of California San Diego, CA, USA; Center for Behavior Genetics of Aging, University of California San Diego, CA, USA.
| | - Matthew S Panizzon
- Department of Psychiatry, University of California San Diego, CA, USA; Center for Behavior Genetics of Aging, University of California San Diego, CA, USA
| | - Donald J Hagler
- Department of Radiology, University of California San Diego, CA, USA
| | - Christine Fennema-Notestine
- Department of Psychiatry, University of California San Diego, CA, USA; Department of Radiology, University of California San Diego, CA, USA
| | - Lisa T Eyler
- Department of Psychiatry, University of California San Diego, CA, USA; San Diego VA Health Care System, San Diego, CA 92161, USA
| | - Nathan A Gillespie
- Virginia Institute for Psychiatric and Behavior Genetics, Virginia Commonwealth University, VA, USA
| | - Michael C Neale
- Virginia Institute for Psychiatric and Behavior Genetics, Virginia Commonwealth University, VA, USA
| | - Michael J Lyons
- Department of Psychological and Brain Sciences, Boston University, Boston, MA, USA
| | - Carol E Franz
- Department of Psychiatry, University of California San Diego, CA, USA; Center for Behavior Genetics of Aging, University of California San Diego, CA, USA
| | - Linda K McEvoy
- Department of Radiology, University of California San Diego, CA, USA
| | - Anders M Dale
- Department of Radiology, University of California San Diego, CA, USA; Department of Neurosciences, University of California San Diego, CA, USA
| | - William S Kremen
- Department of Psychiatry, University of California San Diego, CA, USA; Center for Behavior Genetics of Aging, University of California San Diego, CA, USA; San Diego VA Health Care System, San Diego, CA 92161, USA
| |
Collapse
|
31
|
Ma W, Li M, Gao F, Zhang X, Shi L, Yu L, Zhao B, Chen W, Wang G, Wang X. DTI Analysis of Presbycusis Using Voxel-Based Analysis. AJNR Am J Neuroradiol 2016; 37:2110-2114. [PMID: 27418468 DOI: 10.3174/ajnr.a4870] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 05/09/2016] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE Presbycusis is the most common sensory deficit in the aging population. A recent study reported using a DTI-based tractography technique to identify a lack of integrity in a portion of the auditory pathway in patients with presbycusis. The aim of our study was to investigate the white matter pathology of patients with presbycusis by using a voxel-based analysis that is highly sensitive to local intensity changes in DTI data. MATERIALS AND METHODS Fifteen patients with presbycusis and 14 age- and sex-matched healthy controls were scanned on a 3T scanner. Fractional anisotropy, mean diffusivity, axial diffusivity, and radial diffusivity were obtained from the DTI data. Intergroup statistics were implemented on these measurements, which were transformed to Montreal Neurological Institute coordinates by using a nonrigid image registration method called large deformation diffeomorphic metric mapping. RESULTS Increased axial diffusivity, radial diffusivity, and mean diffusivity and decreased fractional anisotropy were found near the right-side hearing-related areas in patients with presbycusis. Increased radial diffusivity and mean diffusivity were also found near a language-related area (Broca area) in patients with presbycusis. CONCLUSIONS Our findings could be important for exploring reliable imaging evidence of presbycusis and could complement an ROI-based approach.
Collapse
Affiliation(s)
- W Ma
- From the Department of Otolaryngology (W.M., L.S.)
- the Second Hospital of Shandong University, The Central Hospital of Jinan City (W.M., L.Y.)
| | - M Li
- College of Electronics and Information Engineering (M.L.), Sichuan University, Chengdu, China
| | - F Gao
- Shandong Medical Imaging Research Institute (F.G., X.Z., B.Z., G.W.), Shandong University, Jinan, China
| | - X Zhang
- Shandong Medical Imaging Research Institute (F.G., X.Z., B.Z., G.W.), Shandong University, Jinan, China
| | - L Shi
- From the Department of Otolaryngology (W.M., L.S.)
| | - L Yu
- the Second Hospital of Shandong University, The Central Hospital of Jinan City (W.M., L.Y.)
| | - B Zhao
- Shandong Medical Imaging Research Institute (F.G., X.Z., B.Z., G.W.), Shandong University, Jinan, China
| | - W Chen
- Philips Healthcare (W.C.), Shanghai, China
| | - G Wang
- Shandong Medical Imaging Research Institute (F.G., X.Z., B.Z., G.W.), Shandong University, Jinan, China
| | - X Wang
- the Department of Radiation Oncology (X.W.), University of Nebraska Medical Center, Omaha, Nebraska
| |
Collapse
|
32
|
Longitudinal imaging of the ageing mouse. Mech Ageing Dev 2016; 160:93-116. [PMID: 27530773 DOI: 10.1016/j.mad.2016.08.001] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 07/30/2016] [Accepted: 08/04/2016] [Indexed: 12/13/2022]
Abstract
Several non-invasive imaging techniques are used to investigate the effect of pathologies and treatments over time in mouse models. Each preclinical in vivo technique provides longitudinal and quantitative measurements of changes in tissues and organs, which are fundamental for the evaluation of alterations in phenotype due to pathologies, interventions and treatments. However, it is still unclear how these imaging modalities can be used to study ageing with mice models. Almost all age related pathologies in mice such as osteoporosis, arthritis, diabetes, cancer, thrombi, dementia, to name a few, can be imaged in vivo by at least one longitudinal imaging modality. These measurements are the basis for quantification of treatment effects in the development phase of a novel treatment prior to its clinical testing. Furthermore, the non-invasive nature of such investigations allows the assessment of different tissue and organ phenotypes in the same animal and over time, providing the opportunity to study the dysfunction of multiple tissues associated with the ageing process. This review paper aims to provide an overview of the applications of the most commonly used in vivo imaging modalities used in mouse studies: micro-computed-tomography, preclinical magnetic-resonance-imaging, preclinical positron-emission-tomography, preclinical single photon emission computed tomography, ultrasound, intravital microscopy, and whole body optical imaging.
Collapse
|
33
|
Badea A, Kane L, Anderson RJ, Qi Y, Foster M, Cofer GP, Medvitz N, Buckley AF, Badea AK, Wetsel WC, Colton CA. The fornix provides multiple biomarkers to characterize circuit disruption in a mouse model of Alzheimer's disease. Neuroimage 2016; 142:498-511. [PMID: 27521741 DOI: 10.1016/j.neuroimage.2016.08.014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 06/23/2016] [Accepted: 08/09/2016] [Indexed: 12/19/2022] Open
Abstract
Multivariate biomarkers are needed for detecting Alzheimer's disease (AD), understanding its etiology, and quantifying the effect of therapies. Mouse models provide opportunities to study characteristics of AD in well-controlled environments that can help facilitate development of early interventions. The CVN-AD mouse model replicates multiple AD hallmark pathologies, and we identified multivariate biomarkers characterizing a brain circuit disruption predictive of cognitive decline. In vivo and ex vivo magnetic resonance imaging (MRI) revealed that CVN-AD mice replicate the hippocampal atrophy (6%), characteristic of humans with AD, and also present changes in subcortical areas. The largest effect was in the fornix (23% smaller), which connects the septum, hippocampus, and hypothalamus. In characterizing the fornix with diffusion tensor imaging, fractional anisotropy was most sensitive (20% reduction), followed by radial (15%) and axial diffusivity (2%), in detecting pathological changes. These findings were strengthened by optical microscopy and ultrastructural analyses. Ultrastructual analysis provided estimates of axonal density, diameters, and myelination-through the g-ratio, defined as the ratio between the axonal diameter, and the diameter of the axon plus the myelin sheath. The fornix had reduced axonal density (47% fewer), axonal degeneration (13% larger axons), and abnormal myelination (1.5% smaller g-ratios). CD68 staining showed that white matter pathology could be secondary to neuronal degeneration, or due to direct microglial attack. In conclusion, these findings strengthen the hypothesis that the fornix plays a role in AD, and can be used as a disease biomarker and as a target for therapy.
Collapse
Affiliation(s)
- Alexandra Badea
- Center for In Vivo Microscopy, Duke University Medical Center, Department of Radiology, Durham, NC 27710, USA.
| | - Lauren Kane
- Trinity College of Arts & Sciences, Duke University, Durham, NC 27710, USA
| | - Robert J Anderson
- Center for In Vivo Microscopy, Duke University Medical Center, Department of Radiology, Durham, NC 27710, USA
| | - Yi Qi
- Center for In Vivo Microscopy, Duke University Medical Center, Department of Radiology, Durham, NC 27710, USA
| | - Mark Foster
- Center for In Vivo Microscopy, Duke University Medical Center, Department of Radiology, Durham, NC 27710, USA
| | - Gary P Cofer
- Center for In Vivo Microscopy, Duke University Medical Center, Department of Radiology, Durham, NC 27710, USA
| | - Neil Medvitz
- Department of Pathology, and Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA
| | - Anne F Buckley
- Department of Pathology, and Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, NC 27710, USA
| | - Andreas K Badea
- Center for In Vivo Microscopy, Duke University Medical Center, Department of Radiology, Durham, NC 27710, USA
| | - William C Wetsel
- Departments of Psychiatry and Behavioral Sciences, Cell Biology, and Neurobiology, Duke University Medical Center, Durham, NC 27710, USA
| | - Carol A Colton
- Department of Neurology, Duke University Medical Center, Durham, NC 27710, USA
| |
Collapse
|
34
|
Kincses ZT, Király A, Veréb D, Vécsei L. Structural Magnetic Resonance Imaging Markers of Alzheimer's Disease and Its Retranslation to Rodent Models. J Alzheimers Dis 2016; 47:277-90. [PMID: 26401552 DOI: 10.3233/jad-143195] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
The importance of imaging biomarkers has been acknowledged in the diagnosis and in the follow-up of Alzheimer's disease (AD), one of the major causes of dementia. Next to the molecular biomarkers and PET imaging investigations, structural MRI approaches provide important information about the disease progression and about the pathomechanism. Furthermore,a growing body of literature retranslates these imaging biomarkers to various rodent models of the disease. The goal of this review is to provide an overview of the macro- and microstructural imaging biomarkers of AD, concentrating on atrophy measures and diffusion MRI alterations. A survey is also given of the imaging approaches used in rodent models of dementias that can promote drug development.
Collapse
Affiliation(s)
- Zsigmond Tamas Kincses
- Department of Neurology, University of Szeged, Szeged, Hungary.,International Clinical Research Center, St. Anne's University Hospital Brno, Brno, Czech Republic
| | - András Király
- Department of Neurology, University of Szeged, Szeged, Hungary
| | - Dániel Veréb
- Department of Neurology, University of Szeged, Szeged, Hungary
| | - László Vécsei
- Department of Neurology, University of Szeged, Szeged, Hungary.,MTA-SZTE Neuroscience Research Group, Szeged, Hungary
| |
Collapse
|
35
|
Febo M, Foster TC. Preclinical Magnetic Resonance Imaging and Spectroscopy Studies of Memory, Aging, and Cognitive Decline. Front Aging Neurosci 2016; 8:158. [PMID: 27468264 PMCID: PMC4942756 DOI: 10.3389/fnagi.2016.00158] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 06/16/2016] [Indexed: 01/14/2023] Open
Abstract
Neuroimaging provides for non-invasive evaluation of brain structure and activity and has been employed to suggest possible mechanisms for cognitive aging in humans. However, these imaging procedures have limits in terms of defining cellular and molecular mechanisms. In contrast, investigations of cognitive aging in animal models have mostly utilized techniques that have offered insight on synaptic, cellular, genetic, and epigenetic mechanisms affecting memory. Studies employing magnetic resonance imaging and spectroscopy (MRI and MRS, respectively) in animal models have emerged as an integrative set of techniques bridging localized cellular/molecular phenomenon and broader in vivo neural network alterations. MRI methods are remarkably suited to longitudinal tracking of cognitive function over extended periods permitting examination of the trajectory of structural or activity related changes. Combined with molecular and electrophysiological tools to selectively drive activity within specific brain regions, recent studies have begun to unlock the meaning of fMRI signals in terms of the role of neural plasticity and types of neural activity that generate the signals. The techniques provide a unique opportunity to causally determine how memory-relevant synaptic activity is processed and how memories may be distributed or reconsolidated over time. The present review summarizes research employing animal MRI and MRS in the study of brain function, structure, and biochemistry, with a particular focus on age-related cognitive decline.
Collapse
Affiliation(s)
- Marcelo Febo
- Department of Psychiatry, William L. and Evelyn F. McKnight Brain Institute, University of Florida Gainesville, FL, USA
| | - Thomas C Foster
- Department of Neuroscience, William L. and Evelyn F. McKnight Brain Institute, University of Florida Gainesville, FL, USA
| |
Collapse
|
36
|
Rieckmann A, Van Dijk KRA, Sperling RA, Johnson KA, Buckner RL, Hedden T. Accelerated decline in white matter integrity in clinically normal individuals at risk for Alzheimer's disease. Neurobiol Aging 2016; 42:177-88. [PMID: 27143434 PMCID: PMC4857135 DOI: 10.1016/j.neurobiolaging.2016.03.016] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 03/13/2016] [Accepted: 03/14/2016] [Indexed: 12/19/2022]
Abstract
Prior studies have identified white matter abnormalities in Alzheimer's disease (AD). Yet, cross-sectional studies in normal older individuals show little evidence for an association between markers of AD risk (APOE4 genotype and amyloid deposition), and white matter integrity. Here, 108 normal older adults (age, 66-87) with assessments of apolipoprotein e4 (APOE4) genotype and assessment of amyloid burden by positron emission tomography underwent diffusion tensor imaging scans for measuring white matter integrity at 2 time points, on average 2.6 years apart. Linear mixed-effects models showed that amyloid burden at baseline was associated with steeper decline in fractional anisotropy in the parahippocampal cingulum (p < 0.05). This association was not significant between baseline measures suggesting that longitudinal analyses can provide novel insights that are not detectable in cross-sectional designs. Amyloid-related changes in hippocampus volume did not explain the association between amyloid burden and change in fractional anisotropy. The results suggest that accumulation of cortical amyloid and white matter changes in parahippocampal cingulum are not independent processes in individuals at increased risk for AD.
Collapse
Affiliation(s)
- Anna Rieckmann
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA; Department of Radiation Sciences, Diagnostic Radiology, Umeå University, Umeå, Sweden
| | - Koene R A Van Dijk
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA; Department of Psychology and Center for Brain Science, Harvard University, Cambridge, MA, USA
| | - Reisa A Sperling
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA; Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Keith A Johnson
- Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Center for Alzheimer Research and Treatment, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Randy L Buckner
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA; Department of Psychology and Center for Brain Science, Harvard University, Cambridge, MA, USA; Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Trey Hedden
- Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Charlestown, MA, USA; Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
37
|
Grandjean J, Derungs R, Kulic L, Welt T, Henkelman M, Nitsch RM, Rudin M. Complex interplay between brain function and structure during cerebral amyloidosis in APP transgenic mouse strains revealed by multi-parametric MRI comparison. Neuroimage 2016; 134:1-11. [PMID: 27033685 DOI: 10.1016/j.neuroimage.2016.03.042] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 03/11/2016] [Accepted: 03/17/2016] [Indexed: 11/28/2022] Open
Abstract
Alzheimer's disease is a fatal neurodegenerative disorder affecting the aging population. Neuroimaging methods, in particular magnetic resonance imaging (MRI), have helped reveal alterations in the brain structure, metabolism, and function of patients and in groups at risk of developing AD, yet the nature of these alterations is poorly understood. Neuroimaging in mice is attractive for investigating mechanisms underlying functional and structural changes associated with AD pathology. Several preclinical murine models of AD have been generated based on transgenic insertion of human mutated APP genes. Depending on the specific mutations, mouse strains express different aspects of amyloid pathology, e.g. intracellular amyloid-β (Aβ) aggregates, parenchymal plaques, or cerebral amyloid angiopathy. We have applied multi-parametric MRI in three transgenic mouse lines to compare changes in brain function with resting-state fMRI and structure with diffusion tensor imaging and high resolution anatomical imaging. E22ΔAβ developing intracellular Aβ aggregates did not present functional or structural alterations compared to their wild-type littermates. PSAPP mice displaying parenchymal amyloid plaques displayed mild functional changes within the supplementary and barrel field cortices, and increased isocortical volume relative to controls. Extensive reduction in functional connectivity in the sensory-motor cortices and within the default mode network, as well as local volume increase in the midbrain relative to wild-type have been observed in ArcAβ mice bearing intracellular Aβ aggregates as well as parenchymal and vascular amyloid deposits. Patterns of functional and structural changes appear to be strain-specific and not directly related to amyloid deposition.
Collapse
Affiliation(s)
- Joanes Grandjean
- Institute for Biomedical Engineering, University and ETH Zürich, Zürich, Switzerland
| | - Rebecca Derungs
- Center for Neuroscience Research, University and ETH Zürich, Zürich, Switzerland; Division of Psychiatry Research, University of Zürich, Zürich, Switzerland
| | - Luka Kulic
- Center for Neuroscience Research, University and ETH Zürich, Zürich, Switzerland; Division of Psychiatry Research, University of Zürich, Zürich, Switzerland
| | - Tobias Welt
- Center for Neuroscience Research, University and ETH Zürich, Zürich, Switzerland; Division of Psychiatry Research, University of Zürich, Zürich, Switzerland
| | - Mark Henkelman
- Mouse Imaging Centre, Hospital for Sick Children, Toronto, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Canada
| | - Roger M Nitsch
- Center for Neuroscience Research, University and ETH Zürich, Zürich, Switzerland; Division of Psychiatry Research, University of Zürich, Zürich, Switzerland
| | - Markus Rudin
- Institute for Biomedical Engineering, University and ETH Zürich, Zürich, Switzerland; Center for Neuroscience Research, University and ETH Zürich, Zürich, Switzerland; Institute of Pharmacology and Toxicology, University of Zürich, Zürich, Switzerland.
| |
Collapse
|
38
|
Sun SW, Nishioka C, Labib W, Liang HF. Axonal Terminals Exposed to Amyloid-β May Not Lead to Pre-Synaptic Axonal Damage. J Alzheimers Dis 2016; 45:1139-48. [PMID: 25697704 DOI: 10.3233/jad-142154] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BACKGROUND Synaptic deficits and neuronal loss are the major pathological manifestations of Alzheimer's disease. However, the link between the early synaptic loss and subsequent neurodegeneration is not entirely clear. Cell culture studies have shown that amyloid-β (Aβ) applied to axonal terminals can cause retrograde degeneration leading to the neuronal loss, but this process has not been demonstrated in live animals. OBJECTIVE To test if Aβ applied to retinal ganglion cell axonal terminals can induce axonal damage in the optic nerve and optic tract in mice. METHODS Aβ was injected into the terminal field of the optic tract, in the left lateral geniculate nucleus of wildtype C57BL/6 mice. Following the injection, monthly diffusion tensor imaging was performed. Three months after the injection, mice underwent visual evoked potential recordings, and then sacrificed for immunohistochemical examination. RESULTS There were no significant changes seen with diffusion tensor imaging in the optic nerve and optic tract 3 months after the Aβ injection. The myelin and axons in these regions remained intact according to immunohistochemistry. The only significant changes observed in this study were delayed transduction and reduced amplitude of visual evoked potentials, although both Aβ and its reversed form caused similar changes. CONCLUSION Despite the published in vitro studies, there was no significant axonal damage in the optic nerve and optic tract after injecting Aβ onto retinal ganglion cell axonal terminals of wildtype C57BL/6 mice.
Collapse
Affiliation(s)
- Shu-Wei Sun
- Basic Sciences, Schools of Medicine, Loma Linda University, Loma Linda, CA, USA Radiation Medicine, Schools of Medicine, Loma Linda University, Loma Linda, CA, USA Pharmaceutical Science, School of Pharmacy, Loma Linda University, Loma Linda, CA, USA Neuroscience, University of California in Riverside, Riverside, CA, USA Bioengineering, University of California in Riverside, Riverside, CA, USA
| | | | - Wessam Labib
- Family Medicine, Loma Linda University Medical Center, Loma Linda, CA, USA
| | - Hsiao-Fang Liang
- Basic Sciences, Schools of Medicine, Loma Linda University, Loma Linda, CA, USA
| |
Collapse
|
39
|
Holmes HE, Colgan N, Ismail O, Ma D, Powell NM, O'Callaghan JM, Harrison IF, Johnson RA, Murray TK, Ahmed Z, Heggenes M, Fisher A, Cardoso MJ, Modat M, Walker-Samuel S, Fisher EMC, Ourselin S, O'Neill MJ, Wells JA, Collins EC, Lythgoe MF. Imaging the accumulation and suppression of tau pathology using multiparametric MRI. Neurobiol Aging 2016; 39:184-94. [PMID: 26923415 PMCID: PMC4782737 DOI: 10.1016/j.neurobiolaging.2015.12.001] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 12/08/2015] [Accepted: 12/09/2015] [Indexed: 01/30/2023]
Abstract
Mouse models of Alzheimer's disease have served as valuable tools for investigating pathogenic mechanisms relating to neurodegeneration, including tau-mediated and neurofibrillary tangle pathology-a major hallmark of the disease. In this work, we have used multiparametric magnetic resonance imaging (MRI) in a longitudinal study of neurodegeneration in the rTg4510 mouse model of tauopathy, a subset of which were treated with doxycycline at different time points to suppress the tau transgene. Using this paradigm, we investigated the sensitivity of multiparametric MRI to both the accumulation and suppression of pathologic tau. Tau-related atrophy was discernible from 5.5 months within the cortex and hippocampus. We observed markedly less atrophy in the treated rTg4510 mice, which was enhanced after doxycycline intervention from 3.5 months. We also observed differences in amide proton transfer, cerebral blood flow, and diffusion tensor imaging parameters in the rTg4510 mice, which were significantly less altered after doxycycline treatment. We propose that these non-invasive MRI techniques offer insight into pathologic mechanisms underpinning Alzheimer's disease that may be important when evaluating emerging therapeutics targeting one of more of these processes.
Collapse
Affiliation(s)
- Holly E Holmes
- Division of Medicine, Centre for Advanced Biomedical Imaging, University College London, London, UK.
| | - Niall Colgan
- Division of Medicine, Centre for Advanced Biomedical Imaging, University College London, London, UK
| | - Ozama Ismail
- Division of Medicine, Centre for Advanced Biomedical Imaging, University College London, London, UK
| | - Da Ma
- Division of Medicine, Centre for Advanced Biomedical Imaging, University College London, London, UK; Translational Imaging Group, Centre for Medical Image Computing, University College London, London, UK
| | - Nick M Powell
- Division of Medicine, Centre for Advanced Biomedical Imaging, University College London, London, UK; Translational Imaging Group, Centre for Medical Image Computing, University College London, London, UK
| | - James M O'Callaghan
- Division of Medicine, Centre for Advanced Biomedical Imaging, University College London, London, UK
| | - Ian F Harrison
- Division of Medicine, Centre for Advanced Biomedical Imaging, University College London, London, UK
| | - Ross A Johnson
- Tailored Therapeutics, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, USA
| | | | | | | | | | - M J Cardoso
- Translational Imaging Group, Centre for Medical Image Computing, University College London, London, UK
| | - Marc Modat
- Translational Imaging Group, Centre for Medical Image Computing, University College London, London, UK
| | - Simon Walker-Samuel
- Division of Medicine, Centre for Advanced Biomedical Imaging, University College London, London, UK
| | - Elizabeth M C Fisher
- Department of Neurodegenerative Diseases, Institute of Neurology, University College London, London, UK
| | - Sebastien Ourselin
- Translational Imaging Group, Centre for Medical Image Computing, University College London, London, UK
| | | | - Jack A Wells
- Division of Medicine, Centre for Advanced Biomedical Imaging, University College London, London, UK
| | - Emily C Collins
- Tailored Therapeutics, Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN, USA
| | - Mark F Lythgoe
- Division of Medicine, Centre for Advanced Biomedical Imaging, University College London, London, UK
| |
Collapse
|
40
|
Fan SM, Chen W, Xiong L, Xia Y, Xie YB, Chen J. Magnetic resonance diffusion tensor imaging study of rhesus optic nerve radiation injury caused by a single dose/fractionation scheme stereotactic radiosurgery at an early stage. J Neuroradiol 2015; 43:207-13. [PMID: 26726930 DOI: 10.1016/j.neurad.2015.10.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 10/10/2015] [Accepted: 10/24/2015] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND PURPOSE Radiation-induced optic neuropathy (RION) is a devastating late complication of radiotherapy. However, research on the imaging performance of RION is not sufficient. The aim of this study was to investigate the performance of magnetic resonance diffusion tensor imaging (DTI) early after injury of the optic nerve of rhesus monkeys by a single-dose/fractionation-scheme of stereotactic radiosurgery (SRS). MATERIALS AND METHODS The intraorbital optic nerve contour of 5 rhesus monkeys was acquired by magnetic resonance imaging (MRI). Then, the unilateral intraorbital optic nerves of 5 rhesus monkeys were injured by gamma knife surgery (GKS) with a single-dose/fractionation scheme (marginal dose of 15Gy, 50% isodose curve). DTI was performed before the irradiation and 1week, 2weeks, 4weeks, and 24weeks after injury to obtain the cross-sectional area, and the fractional anisotropy (FA), apparent diffusion coefficient (ADC), axial diffusivity (AD) and radial diffusivity (RD) values. RESULTS The cross-sectional area of the injured optic nerve exhibited significant atrophy 24weeks after SRS. FA declined 1week after injury; this value then increased slightly but remained lower than before injury (P<0.05). AD began to decline in the 2weeks after injury and gradually disappeared (P<0.05). CONCLUSION SRS with a single-dose/fractionation scheme (marginal dose of 15Gy, 50% isodose curve) on the unilateral intraorbital optic nerve can induce RION. DTI can detect RION at an early stage. FA and AD are useful indicators for RION diagnosis. In the early stage, the primary site of RION may be the vascular endothelium.
Collapse
Affiliation(s)
- Shuang Min Fan
- Department of Neurosurgery, Sichuan University, West China Hospital, Chengdu, Sichuan Province, PR China
| | - Wei Chen
- Department of Neurosurgery, Sichuan University, West China Hospital, Chengdu, Sichuan Province, PR China
| | - Li Xiong
- Department of Neurosurgery, Sichuan Provincial People's Hospital, Chengdu, Sichuan Province, PR China
| | - Yong Xia
- Department of Neurosurgery, Sichuan University, West China Hospital, Chengdu, Sichuan Province, PR China
| | - Yue Bin Xie
- Department of Neurosurgery, Sichuan University, West China Hospital, Chengdu, Sichuan Province, PR China
| | - Jing Chen
- Department of Neurosurgery, Sichuan University, West China Hospital, Chengdu, Sichuan Province, PR China.
| |
Collapse
|
41
|
Nowrangi MA, Okonkwo O, Lyketsos C, Oishi K, Mori S, Albert M, Mielke MM. Atlas-based diffusion tensor imaging correlates of executive function. J Alzheimers Dis 2015; 44:585-98. [PMID: 25318544 DOI: 10.3233/jad-141937] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Impairment in executive function (EF) is commonly found in Alzheimer's disease (AD) and mild cognitive impairment (MCI). Atlas-based diffusion tensor imaging (DTI) methods may be useful in relating regional integrity to EF measures in MCI and AD. Sixty-six participants (25 normal controls, 22 MCI, and 19 AD) received DTI scans and clinical evaluation. DTI scans were applied to a pre-segmented atlas and fractional anisotropy (FA) and mean diffusivity (MD) were calculated. ANOVA was used to assess group differences in frontal, parietal, and cerebellar regions. For regions differing between groups (p < 0.01), linear regression examined the relationship between EF scores and regional FA and MD. Anisotropy and diffusivity in frontal and parietal lobe white matter structures were associated with EF scores in MCI and only frontal lobe structures in AD. EF was more strongly associated with FA than MD. The relationship between EF and anisotropy and diffusivity was strongest in MCI. These results suggest that regional white matter integrity is compromised in MCI and AD and that FA may be a better correlate of EF than MD.
Collapse
Affiliation(s)
- Milap A Nowrangi
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine and Johns Hopkins Bayview Medical Center, Baltimore, MD, USA
| | - Ozioma Okonkwo
- Department of Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Constantine Lyketsos
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine and Johns Hopkins Bayview Medical Center, Baltimore, MD, USA
| | - Kenichi Oishi
- Department of Radiology Johns Hopkins University School of Medicine and Johns Hopkins Bayview Medical Center, Baltimore, MD, USA
| | - Susumu Mori
- Department of Radiology Johns Hopkins University School of Medicine and Johns Hopkins Bayview Medical Center, Baltimore, MD, USA
| | - Marilyn Albert
- Department of Neurology, Johns Hopkins University School of Medicine and Johns Hopkins Bayview Medical Center, Baltimore, MD, USA
| | - Michelle M Mielke
- Department of Health Sciences Research, Division of Epidemiology and Department of Neurology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
42
|
Wolf D, Fischer FU, Scheurich A, Fellgiebel A. Non-Linear Association between Cerebral Amyloid Deposition and White Matter Microstructure in Cognitively Healthy Older Adults. J Alzheimers Dis 2015; 47:117-27. [DOI: 10.3233/jad-150049] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
43
|
Fischer FU, Wolf D, Scheurich A, Fellgiebel A. Altered whole-brain white matter networks in preclinical Alzheimer's disease. NEUROIMAGE-CLINICAL 2015; 8:660-6. [PMID: 26288751 PMCID: PMC4536470 DOI: 10.1016/j.nicl.2015.06.007] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 06/17/2015] [Accepted: 06/20/2015] [Indexed: 11/28/2022]
Abstract
Surrogates of whole-brain white matter (WM) networks reconstructed using diffusion tensor imaging (DTI) are novel markers of structural brain connectivity. Global connectivity of networks has been found impaired in clinical Alzheimer's disease (AD) compared to cognitively healthy aging. We hypothesized that network alterations are detectable already in preclinical AD and investigated major global WM network properties. Other structural markers of neurodegeneration typically affected in prodromal AD but seeming largely unimpaired in preclinical AD were also examined. 12 cognitively healthy elderly with preclinical AD as classified by florbetapir-PET (mean age 73.4 ± 4.9) and 31 age-matched controls without cerebral amyloidosis (mean age 73.1 ± 6.7) from the ADNI were included. WM networks were reconstructed from DTI using tractography and graph theory. Indices of network capacity and the established imaging markers of neurodegeneration hippocampal volume, and cerebral glucose utilization as measured by fludeoxyglucose-PET were compared between the two groups. Additionally, we measured surrogates of global WM integrity (fractional anisotropy, mean diffusivity, volume). We found an increase of shortest path length and a decrease of global efficiency in preclinical AD. These results remained largely unchanged when controlling for WM integrity. In contrast, neither markers of neurodegeneration nor WM integrity were altered in preclinical AD subjects. Our results suggest an impairment of WM networks in preclinical AD that is detectable while other structural imaging markers do not yet indicate incipient neurodegeneration. Moreover, these findings are specific to WM networks and cannot be explained by other surrogates of global WM integrity.
Collapse
Affiliation(s)
- Florian Udo Fischer
- University Medical Center Mainz, Untere Zahlbacher Str. 8, Mainz 55131, Germany
| | - Dominik Wolf
- University Medical Center Mainz, Untere Zahlbacher Str. 8, Mainz 55131, Germany
| | - Armin Scheurich
- University Medical Center Mainz, Untere Zahlbacher Str. 8, Mainz 55131, Germany
| | - Andreas Fellgiebel
- University Medical Center Mainz, Untere Zahlbacher Str. 8, Mainz 55131, Germany
| | | |
Collapse
|
44
|
Wells JA, O'Callaghan JM, Holmes HE, Powell NM, Johnson RA, Siow B, Torrealdea F, Ismail O, Walker-Samuel S, Golay X, Rega M, Richardson S, Modat M, Cardoso MJ, Ourselin S, Schwarz AJ, Ahmed Z, Murray TK, O'Neill MJ, Collins EC, Colgan N, Lythgoe MF. In vivo imaging of tau pathology using multi-parametric quantitative MRI. Neuroimage 2015; 111:369-78. [PMID: 25700953 PMCID: PMC4626540 DOI: 10.1016/j.neuroimage.2015.02.023] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Revised: 02/04/2015] [Accepted: 02/10/2015] [Indexed: 12/29/2022] Open
Abstract
As the number of people diagnosed with Alzheimer's disease (AD) reaches epidemic proportions, there is an urgent need to develop effective treatment strategies to tackle the social and economic costs of this fatal condition. Dozens of candidate therapeutics are currently being tested in clinical trials, and compounds targeting the aberrant accumulation of tau proteins into neurofibrillary tangles (NFTs) are the focus of substantial current interest. Reliable, translatable biomarkers sensitive to both tau pathology and its modulation by treatment along with animal models that faithfully reflect aspects of the human disease are urgently required. Magnetic resonance imaging (MRI) is well established as a valuable tool for monitoring the structural brain changes that accompany AD progression. However the descent into dementia is not defined by macroscopic brain matter loss alone: non-invasive imaging measurements sensitive to protein accumulation, white matter integrity and cerebral haemodynamics probe distinct aspects of AD pathophysiology and may serve as superior biomarkers for assessing drug efficacy. Here we employ a multi-parametric array of five translatable MRI techniques to characterise the in vivo pathophysiological phenotype of the rTg4510 mouse model of tauopathy (structural imaging, diffusion tensor imaging (DTI), arterial spin labelling (ASL), chemical exchange saturation transfer (CEST) and glucose CEST). Tau-induced pathological changes included grey matter atrophy, increased radial diffusivity in the white matter, decreased amide proton transfer and hyperperfusion. We demonstrate that the above markers unambiguously discriminate between the transgenic group and age-matched controls and provide a comprehensive profile of the multifaceted neuropathological processes underlying the rTg4510 model. Furthermore, we show that ASL and DTI techniques offer heightened sensitivity to processes believed to precede detectable structural changes and, as such, provides a platform for the study of disease mechanisms and therapeutic intervention.
Collapse
Affiliation(s)
- J A Wells
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine and Institute of Child Health, University College London, UK.
| | - J M O'Callaghan
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine and Institute of Child Health, University College London, UK
| | - H E Holmes
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine and Institute of Child Health, University College London, UK
| | - N M Powell
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine and Institute of Child Health, University College London, UK; Translational Imaging Group, Centre for Medical Imaging Computing, University College London, UK
| | - R A Johnson
- Eli Lilly & Co. Ltd, Erl Wood Manor, Windlesham, Surrey GU20 6PH, UK
| | - B Siow
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine and Institute of Child Health, University College London, UK
| | - F Torrealdea
- Brain Repair & Rehabilitation, Institute of Neurology, University College London, UK
| | - O Ismail
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine and Institute of Child Health, University College London, UK
| | - S Walker-Samuel
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine and Institute of Child Health, University College London, UK
| | - X Golay
- Brain Repair & Rehabilitation, Institute of Neurology, University College London, UK
| | - M Rega
- Brain Repair & Rehabilitation, Institute of Neurology, University College London, UK
| | - S Richardson
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine and Institute of Child Health, University College London, UK
| | - M Modat
- Translational Imaging Group, Centre for Medical Imaging Computing, University College London, UK
| | - M J Cardoso
- Translational Imaging Group, Centre for Medical Imaging Computing, University College London, UK
| | - S Ourselin
- Translational Imaging Group, Centre for Medical Imaging Computing, University College London, UK
| | - A J Schwarz
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN 46285, USA
| | - Z Ahmed
- Eli Lilly & Co. Ltd, Erl Wood Manor, Windlesham, Surrey GU20 6PH, UK
| | - T K Murray
- Eli Lilly & Co. Ltd, Erl Wood Manor, Windlesham, Surrey GU20 6PH, UK
| | - M J O'Neill
- Eli Lilly & Co. Ltd, Erl Wood Manor, Windlesham, Surrey GU20 6PH, UK
| | - E C Collins
- Eli Lilly and Company, Lilly Corporate Center, Indianapolis, IN 46285, USA
| | - N Colgan
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine and Institute of Child Health, University College London, UK
| | - M F Lythgoe
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine and Institute of Child Health, University College London, UK
| |
Collapse
|
45
|
Goveas J, O'Dwyer L, Mascalchi M, Cosottini M, Diciotti S, De Santis S, Passamonti L, Tessa C, Toschi N, Giannelli M. Diffusion-MRI in neurodegenerative disorders. Magn Reson Imaging 2015; 33:853-76. [PMID: 25917917 DOI: 10.1016/j.mri.2015.04.006] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Revised: 04/18/2015] [Accepted: 04/19/2015] [Indexed: 12/11/2022]
Abstract
The ability to image the whole brain through ever more subtle and specific methods/contrasts has come to play a key role in understanding the basis of brain abnormalities in several diseases. In magnetic resonance imaging (MRI), "diffusion" (i.e. the random, thermally-induced displacements of water molecules over time) represents an extraordinarily sensitive contrast mechanism, and the exquisite structural detail it affords has proven useful in a vast number of clinical as well as research applications. Since diffusion-MRI is a truly quantitative imaging technique, the indices it provides can serve as potential imaging biomarkers which could allow early detection of pathological alterations as well as tracking and possibly predicting subtle changes in follow-up examinations and clinical trials. Accordingly, diffusion-MRI has proven useful in obtaining information to better understand the microstructural changes and neurophysiological mechanisms underlying various neurodegenerative disorders. In this review article, we summarize and explore the main applications, findings, perspectives as well as challenges and future research of diffusion-MRI in various neurodegenerative disorders including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease and degenerative ataxias.
Collapse
Affiliation(s)
- Joseph Goveas
- Department of Psychiatry and Behavioral Medicine, and Institute for Health and Society, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Laurence O'Dwyer
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, Goethe University, Frankfurt, Germany
| | - Mario Mascalchi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Florence, Italy; Quantitative and Functional Neuroradiology Research Program at Meyer Children and Careggi Hospitals of Florence, Florence, Italy
| | - Mirco Cosottini
- Department of Translational Research and New Surgical and Medical Technologies, University of Pisa, Pisa, Italy; Unit of Neuroradiology, Pisa University Hospital "Azienda Ospedaliero-Universitaria Pisana", Pisa, Italy
| | - Stefano Diciotti
- Department of Electrical, Electronic, and Information Engineering "Guglielmo Marconi", University of Bologna, Cesena, Italy
| | - Silvia De Santis
- Cardiff University Brain Research Imaging Centre (CUBRIC), School of Psychology, Cardiff University, Cardiff, UK
| | - Luca Passamonti
- Institute of Bioimaging and Molecular Physiology, National Research Council, Catanzaro, Italy; Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Carlo Tessa
- Division of Radiology, "Versilia" Hospital, AUSL 12 Viareggio, Lido di Camaiore, Italy
| | - Nicola Toschi
- Department of Biomedicine and Prevention, Medical Physics Section, University of Rome "Tor Vergata", Rome, Italy; Department of Radiology, Athinoula A. Martinos Center for Biomedical Imaging, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Marco Giannelli
- Unit of Medical Physics, Pisa University Hospital "Azienda Ospedaliero-Universitaria Pisana", Pisa, Italy.
| |
Collapse
|
46
|
Nowrangi MA, Rosenberg PB. The fornix in mild cognitive impairment and Alzheimer's disease. Front Aging Neurosci 2015; 7:1. [PMID: 25653617 PMCID: PMC4301006 DOI: 10.3389/fnagi.2015.00001] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 01/02/2015] [Indexed: 01/15/2023] Open
Abstract
The fornix is an integral white matter bundle located in the medial diencephalon and is part of the limbic structures. It serves a vital role in memory functions and as such has become the subject of recent research emphasis in Alzheimer's disease (AD) and mild cognitive impairment (MCI). As the characteristic pathological processes of AD progress, structural and functional changes to the medial temporal lobes and other regions become evident years before clinical symptoms are present. Though gray matter atrophy has been the most studied, degradation of white matter structures especially the fornix may precede these and has become detectable with use of diffusion tensor imaging (DTI) and other complimentary imaging techniques. Recent research utilizing DTI measurement of the fornix has shown good discriminability of diagnostic groups, particularly early and preclinical, as well as predictive power for incident MCI and AD. Stimulating and modulating fornix function by the way of DBS has been an exciting new area as pharmacological therapeutics has been slow to develop.
Collapse
Affiliation(s)
- Milap A Nowrangi
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine , Baltimore, MD , USA
| | - Paul B Rosenberg
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine , Baltimore, MD , USA
| |
Collapse
|
47
|
Haast RAM, Kiliaan AJ. Impact of fatty acids on brain circulation, structure and function. Prostaglandins Leukot Essent Fatty Acids 2015; 92:3-14. [PMID: 24485516 DOI: 10.1016/j.plefa.2014.01.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Accepted: 01/03/2014] [Indexed: 12/30/2022]
Abstract
The use of dietary intervention has evolved into a promising approach to prevent the onset and progression of brain diseases. The positive relationship between intake of omega-3 long chain polyunsaturated fatty acids (ω3-LCPUFAs) and decreased onset of disease- and aging-related deterioration of brain health is increasingly endorsed across epidemiological and diet-interventional studies. Promising results are found regarding to the protection of proper brain circulation, structure and functionality in healthy and diseased humans and animal models. These include enhanced cerebral blood flow (CBF), white and gray matter integrity, and improved cognitive functioning, and are possibly mediated through increased neurovascular coupling, neuroprotection and neuronal plasticity, respectively. Contrary, studies investigating diets high in saturated fats provide opposite results, which may eventually lead to irreversible damage. Studies like these are of great importance given the high incidence of obesity caused by the increased and decreased consumption of respectively saturated fats and ω3-LCPUFAs in the Western civilization. This paper will review in vivo research conducted on the effects of ω3-LCPUFAs and saturated fatty acids on integrity (circulation, structure and function) of the young, aging and diseased brain.
Collapse
Affiliation(s)
- Roy A M Haast
- Department of Anatomy, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | - Amanda J Kiliaan
- Department of Anatomy, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
| |
Collapse
|
48
|
Mhatre SD, Michelson SJ, Gomes J, Tabb LP, Saunders AJ, Marenda DR. Development and characterization of an aged onset model of Alzheimer's disease in Drosophila melanogaster. Exp Neurol 2014; 261:772-81. [DOI: 10.1016/j.expneurol.2014.08.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 08/14/2014] [Accepted: 08/19/2014] [Indexed: 01/08/2023]
|
49
|
Striatum and entorhinal cortex atrophy in AD mouse models: MRI comprehensive analysis. Neurobiol Aging 2014; 36:776-88. [PMID: 25433456 DOI: 10.1016/j.neurobiolaging.2014.10.027] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Revised: 09/19/2014] [Accepted: 10/07/2014] [Indexed: 01/24/2023]
Abstract
Alzheimer's disease is experimentally modeled in transgenic (Tg) mice overexpressing mutated forms of the human amyloid precursor protein either alone or combined with mutated presenilins and tau. In the present study, we developed a systematic approach to compare double (TASTPM) and triple (APP/PS2/Tau) Tg mice by serial magnetic resonance imaging and spectroscopy analysis from 4 to 26 months of age to define homologous biomarkers between mice and humans. Hippocampal atrophy was found in Tg mice compared with WT. In APP/PS2/Tau the effect was age-dependent, whereas in TASTPM it was detectable from the first investigated time point. Importantly, both mice displayed an age-related entorhinal cortex thinning and robust striatal atrophy, the latter associated with a significant loss of synaptophysin. Hippocampal magnetic resonance spectroscopy revealed lower glutamate levels in both Tg mice and a selective myo-inositol increase in TASTPM. This noninvasive magnetic resonance imaging analysis, revealed common biomarkers between humans and mice, and could, thus, be promoted as a fully translational tool to be adopted in the preclinical investigation of therapeutic approaches.
Collapse
|
50
|
Tuor UI, Morgunov M, Sule M, Qiao M, Clark D, Rushforth D, Foniok T, Kirton A. Cellular correlates of longitudinal diffusion tensor imaging of axonal degeneration following hypoxic-ischemic cerebral infarction in neonatal rats. NEUROIMAGE-CLINICAL 2014; 6:32-42. [PMID: 25379414 PMCID: PMC4215526 DOI: 10.1016/j.nicl.2014.08.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Revised: 07/14/2014] [Accepted: 08/04/2014] [Indexed: 02/07/2023]
Abstract
Ischemically damaged brain can be accompanied by secondary degeneration of associated axonal connections e.g. Wallerian degeneration. Diffusion tensor imaging (DTI) is widely used to investigate axonal injury but the cellular correlates of many of the degenerative changes remain speculative. We investigated the relationship of DTI of directly damaged cerebral cortex and secondary axonal degeneration in the cerebral peduncle with cellular alterations in pan-axonal neurofilament staining, myelination, reactive astrocytes, activation of microglia/macrophages and neuronal cell death. DTI measures (axial, radial and mean diffusivity, and fractional anisotropy (FA)) were acquired at hyperacute (3 h), acute (1 and 2 d) and chronic (1 and 4 week) times after transient cerebral hypoxia with unilateral ischemia in neonatal rats. The tissue pathology underlying ischemic and degenerative responses had a complex relationship with DTI parameters. DTI changes at hyperacute and subacute times were smaller in magnitude and tended to be transient and/or delayed in cerebral peduncle compared to cerebral cortex. In cerebral peduncle by 1 d post-insult, there were reductions in neurofilament staining corresponding with decreases in parallel diffusivity which were more sensitive than mean diffusivity in detecting axonal changes. Ipsilesional reductions in FA within cerebral peduncle were robust in detecting both early and chronic degenerative responses. At one or four weeks post-insult, radial diffusivity was increased ipsilaterally in the cerebral peduncle corresponding to pathological evidence of a lack of ontogenic myelination in this region. The detailed differences in progression and magnitude of DTI and histological changes reported provide a reference for identifying the potential contribution of various cellular responses to FA, and, parallel, radial, and mean diffusivity. Diffusion tensor imaging (DTI) widely used; cellular correlates often speculative Studied longitudinal DTI and histological changes following hypoxia–ischemia Compared neonatal cortex changes to those in degenerating cerebral peduncle DTI and cellular changes were often transient or delayed in cerebral peduncle. This provides a reference for potential cellular contributions to DTI changes.
Collapse
Affiliation(s)
- Ursula I Tuor
- Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, T2N 4N1, Canada ; Experimental Imaging Centre, Faculty of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada ; Departments of Physiology and Pharmacology, Faculty of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada ; Department of Clinical Neurosciences, Faculty of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Melissa Morgunov
- Experimental Imaging Centre, Faculty of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada ; Departments of Physiology and Pharmacology, Faculty of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Manasi Sule
- Experimental Imaging Centre, Faculty of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada ; Departments of Physiology and Pharmacology, Faculty of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Min Qiao
- Experimental Imaging Centre, Faculty of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada ; Departments of Physiology and Pharmacology, Faculty of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Darren Clark
- Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, T2N 4N1, Canada ; Department of Clinical Neurosciences, Faculty of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada ; Department of Medical Physics and Informatics, School of Medicine, University of Szeged, Szeged, Hungary
| | - David Rushforth
- Experimental Imaging Centre, Faculty of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Tadeusz Foniok
- Experimental Imaging Centre, Faculty of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Adam Kirton
- Hotchkiss Brain Institute, Faculty of Medicine, University of Calgary, Calgary, T2N 4N1, Canada ; Department of Clinical Neurosciences, Faculty of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada ; Department of Pediatrics, Alberta Children's Hospital Research Institute, Faculty of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada
| |
Collapse
|