1
|
Goossens E, Deblock L, Caboor L, Eynden DVD, Josipovic I, Isaacura PR, Maksimova E, Van Impe M, Bonnin A, Segers P, Cornillie P, Boone MN, Van Driessche I, De Spiegelaere W, De Roo J, Sips P, De Buysser K. From Corrosion Casting to Virtual Dissection: Contrast-Enhanced Vascular Imaging using Hafnium Oxide Nanocrystals. SMALL METHODS 2024; 8:e2301499. [PMID: 38200600 DOI: 10.1002/smtd.202301499] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Indexed: 01/12/2024]
Abstract
Vascular corrosion casting is a method used to visualize the three dimensional (3D) anatomy and branching pattern of blood vessels. A polymer resin is injected in the vascular system and, after curing, the surrounding tissue is removed. The latter often deforms or even fractures the fragile cast. Here, a method is proposed that does not require corrosion, and is based on in situ micro computed tomography (micro-CT) scans. To overcome the lack of CT contrast between the polymer cast and the animals' surrounding soft tissue, hafnium oxide nanocrystals (HfO2 NCs) are introduced as CT contrast agents into the resin. The NCs dramatically improve the overall CT contrast of the cast and allow for straightforward segmentation in the CT scans. Careful design of the NC surface chemistry ensures the colloidal stability of the NCs in the casting resin. Using only 5 m% of HfO2 NCs, high-quality cardiovascular casts of both zebrafish and mice can be automatically segmented using CT imaging software. This allows to differentiate even μ $\umu$ m-scale details without having to alter the current resin injection methods. This new method of virtual dissection by visualizing casts in situ using contrast-enhanced CT imaging greatly expands the application potential of the technique.
Collapse
Affiliation(s)
- Eline Goossens
- Department of Chemistry, Ghent University, Ghent, 9000, Belgium
- Department of Chemistry, University of Basel, Basel, 4058, Switzerland
| | - Loren Deblock
- Department of Chemistry, Ghent University, Ghent, 9000, Belgium
| | - Lisa Caboor
- Department of Biomolecular Medicine, Ghent University, Ghent, 9000, Belgium
| | - Dietger Van den Eynden
- Department of Chemistry, Ghent University, Ghent, 9000, Belgium
- Department of Chemistry, University of Basel, Basel, 4058, Switzerland
| | - Iván Josipovic
- Center for X-ray Tomography, Ghent University, Ghent, 9000, Belgium
| | - Pablo Reyes Isaacura
- Laboratory of Veterinary Morphology, Ghent University, Merelbeke, 9820, Belgium
- Centre for Polymer Material Technologies, Ghent University, Ghent, 9052, Belgium
- Laboratory for Chemical Technology, Ghent University, Ghent, 9052, Belgium
| | - Elizaveta Maksimova
- Department of Chemistry, University of Basel, Basel, 4058, Switzerland
- Swiss Light Source, Paul Scherrer Institut, Villigen PSI, 5232, Switzerland
- Swiss Nanoscience Institute, University of Basel, Basel, 4056, Switzerland
| | - Matthias Van Impe
- Institute of Biomedical Engineering and Technology, Ghent University, Ghent, 9000, Belgium
| | - Anne Bonnin
- Swiss Light Source, Paul Scherrer Institut, Villigen PSI, 5232, Switzerland
| | - Patrick Segers
- Institute of Biomedical Engineering and Technology, Ghent University, Ghent, 9000, Belgium
| | - Pieter Cornillie
- Laboratory of Veterinary Morphology, Ghent University, Merelbeke, 9820, Belgium
| | - Matthieu N Boone
- Center for X-ray Tomography, Ghent University, Ghent, 9000, Belgium
| | | | - Ward De Spiegelaere
- Laboratory of Veterinary Morphology, Ghent University, Merelbeke, 9820, Belgium
| | - Jonathan De Roo
- Department of Chemistry, University of Basel, Basel, 4058, Switzerland
| | - Patrick Sips
- Department of Biomolecular Medicine, Ghent University, Ghent, 9000, Belgium
| | | |
Collapse
|
2
|
Taylor JL, Baudel MMA, Nieves-Cintron M, Navedo MF. Vascular Function and Ion Channels in Alzheimer's Disease. Microcirculation 2024; 31:e12881. [PMID: 39190776 PMCID: PMC11498901 DOI: 10.1111/micc.12881] [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: 07/04/2024] [Accepted: 08/06/2024] [Indexed: 08/29/2024]
Abstract
This review paper explores the critical role of vascular ion channels in the regulation of cerebral artery function and examines the impact of Alzheimer's disease (AD) on these processes. Vascular ion channels are fundamental in controlling vascular tone, blood flow, and endothelial function in cerebral arteries. Dysfunction of these channels can lead to impaired cerebral autoregulation, contributing to cerebrovascular pathologies. AD, characterized by the accumulation of amyloid beta (Aβ) plaques and neurofibrillary tangles, has been increasingly linked to vascular abnormalities, including altered vascular ion channel activity. Here, we briefly review the role of vascular ion channels in cerebral blood flow control and neurovascular coupling. We then examine the vascular defects in AD, the current understanding of how AD pathology affects vascular ion channel function, and how these changes may lead to compromised cerebral blood flow and neurodegenerative processes. Finally, we provide future perspectives and conclusions. Understanding this topic is important as ion channels may be potential therapeutic targets for improving cerebrovascular health and mitigating AD progression.
Collapse
Affiliation(s)
- Jade L. Taylor
- Department of Pharmacology, University of California Davis, Davis CA, 95616, USA
| | | | | | - Manuel F. Navedo
- Department of Pharmacology, University of California Davis, Davis CA, 95616, USA
| |
Collapse
|
3
|
Neuroimaging of Mouse Models of Alzheimer’s Disease. Biomedicines 2022; 10:biomedicines10020305. [PMID: 35203515 PMCID: PMC8869427 DOI: 10.3390/biomedicines10020305] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/25/2022] [Accepted: 01/25/2022] [Indexed: 12/23/2022] Open
Abstract
Magnetic resonance imaging (MRI) and positron emission tomography (PET) have made great strides in the diagnosis and our understanding of Alzheimer’s Disease (AD). Despite the knowledge gained from human studies, mouse models have and continue to play an important role in deciphering the cellular and molecular evolution of AD. MRI and PET are now being increasingly used to investigate neuroimaging features in mouse models and provide the basis for rapid translation to the clinical setting. Here, we provide an overview of the human MRI and PET imaging landscape as a prelude to an in-depth review of preclinical imaging in mice. A broad range of mouse models recapitulate certain aspects of the human AD, but no single model simulates the human disease spectrum. We focused on the two of the most popular mouse models, the 3xTg-AD and the 5xFAD models, and we summarized all known published MRI and PET imaging data, including contrasting findings. The goal of this review is to provide the reader with broad framework to guide future studies in existing and future mouse models of AD. We also highlight aspects of MRI and PET imaging that could be improved to increase rigor and reproducibility in future imaging studies.
Collapse
|
4
|
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: 4.0] [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
|
5
|
Wälchli T, Bisschop J, Miettinen A, Ulmann-Schuler A, Hintermüller C, Meyer EP, Krucker T, Wälchli R, Monnier PP, Carmeliet P, Vogel J, Stampanoni M. Hierarchical imaging and computational analysis of three-dimensional vascular network architecture in the entire postnatal and adult mouse brain. Nat Protoc 2021; 16:4564-4610. [PMID: 34480130 DOI: 10.1038/s41596-021-00587-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 06/08/2021] [Indexed: 02/08/2023]
Abstract
The formation of new blood vessels and the establishment of vascular networks are crucial during brain development, in the adult healthy brain, as well as in various diseases of the central nervous system. Here, we describe a step-by-step protocol for our recently developed method that enables hierarchical imaging and computational analysis of vascular networks in postnatal and adult mouse brains. The different stages of the procedure include resin-based vascular corrosion casting, scanning electron microscopy, synchrotron radiation and desktop microcomputed tomography imaging, and computational network analysis. Combining these methods enables detailed visualization and quantification of the 3D brain vasculature. Network features such as vascular volume fraction, branch point density, vessel diameter, length, tortuosity and directionality as well as extravascular distance can be obtained at any developmental stage from the early postnatal to the adult brain. This approach can be used to provide a detailed morphological atlas of the entire mouse brain vasculature at both the postnatal and the adult stage of development. Our protocol allows the characterization of brain vascular networks separately for capillaries and noncapillaries. The entire protocol, from mouse perfusion to vessel network analysis, takes ~10 d.
Collapse
Affiliation(s)
- Thomas Wälchli
- Group of CNS Angiogenesis and Neurovascular Link, Neuroscience Center Zurich, and Division of Neurosurgery, University and University Hospital Zurich, Zurich, Switzerland.
- Division of Neurosurgery, University Hospital Zurich, Zurich, Switzerland.
- Group Brain Vasculature and Perivascular Niche, Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada.
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, University of Toronto, Toronto, Ontario, Canada.
| | - Jeroen Bisschop
- Group of CNS Angiogenesis and Neurovascular Link, Neuroscience Center Zurich, and Division of Neurosurgery, University and University Hospital Zurich, Zurich, Switzerland
- Division of Neurosurgery, University Hospital Zurich, Zurich, Switzerland
- Group Brain Vasculature and Perivascular Niche, Division of Experimental and Translational Neuroscience, Krembil Brain Institute, Krembil Research Institute, Toronto Western Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
- Division of Neurosurgery, Department of Surgery, Toronto Western Hospital, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Arttu Miettinen
- Swiss Light Source, Paul Scherrer Institute, Villigen, Switzerland
- Institute for Biomedical Engineering, University and ETH Zurich, Zurich, Switzerland
- Department of Physics, University of Jyväskylä, Jyväskylä, Finland
| | | | | | - Eric P Meyer
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | - Thomas Krucker
- Novartis Institutes for BioMedical Research Inc, Emeryville, CA, USA
| | - Regula Wälchli
- Department of Dermatology, Pediatric Skin Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Philippe P Monnier
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Krembil Research Institute, Vision Division, Krembil Discovery Tower, Toronto, Ontario, Canada
- Department of Ophthalmology and Vision Sciences, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology, KU Leuven, Leuven, Belgium
- Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, VIB, Leuven, Belgium
| | - Johannes Vogel
- Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Marco Stampanoni
- Swiss Light Source, Paul Scherrer Institute, Villigen, Switzerland
- Institute for Biomedical Engineering, University and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
6
|
Szu JI, Obenaus A. Cerebrovascular phenotypes in mouse models of Alzheimer's disease. J Cereb Blood Flow Metab 2021; 41:1821-1841. [PMID: 33557692 PMCID: PMC8327123 DOI: 10.1177/0271678x21992462] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/16/2020] [Accepted: 01/06/2021] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a devastating neurological degenerative disorder and is the most common cause of dementia in the elderly. Clinically, AD manifests with memory and cognitive decline associated with deposition of hallmark amyloid beta (Aβ) plaques and neurofibrillary tangles (NFTs). Although the mechanisms underlying AD remains unclear, two hypotheses have been proposed. The established amyloid hypothesis states that Aβ accumulation is the basis of AD and leads to formation of NFTs. In contrast, the two-hit vascular hypothesis suggests that early vascular damage leads to increased accumulation of Aβ deposits in the brain. Multiple studies have reported significant morphological changes of the cerebrovasculature which can result in severe functional deficits. In this review, we delve into known structural and functional vascular alterations in various mouse models of AD and the cellular and molecular constituents that influence these changes to further disease progression. Many studies shed light on the direct impact of Aβ on the cerebrovasculature and how it is disrupted during the progression of AD. However, more research directed towards an improved understanding of how the cerebrovasculature is modified over the time course of AD is needed prior to developing future interventional strategies.
Collapse
Affiliation(s)
- Jenny I Szu
- Institute for Memory Impairments and Neurological Disorders, University of California Irvine, Irvine, CA, USA
| | - André Obenaus
- Department of Pediatrics, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
7
|
Steinman J, Sun HS, Feng ZP. Microvascular Alterations in Alzheimer's Disease. Front Cell Neurosci 2021; 14:618986. [PMID: 33536876 PMCID: PMC7849053 DOI: 10.3389/fncel.2020.618986] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/17/2020] [Indexed: 12/27/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder associated with continual decline in cognition and ability to perform routine functions such as remembering familiar places or understanding speech. For decades, amyloid beta (Aβ) was viewed as the driver of AD, triggering neurodegenerative processes such as inflammation and formation of neurofibrillary tangles (NFTs). This approach has not yielded therapeutics that cure the disease or significant improvements in long-term cognition through removal of plaques and Aβ oligomers. Some researchers propose alternate mechanisms that drive AD or act in conjunction with amyloid to promote neurodegeneration. This review summarizes the status of AD research and examines research directions including and beyond Aβ, such as tau, inflammation, and protein clearance mechanisms. The effect of aging on microvasculature is highlighted, including its contribution to reduced blood flow that impairs cognition. Microvascular alterations observed in AD are outlined, emphasizing imaging studies of capillary malfunction. The review concludes with a discussion of two therapies to protect tissue without directly targeting Aβ for removal: (1) administration of growth factors to promote vascular recovery in AD; (2) inhibiting activity of a calcium-permeable ion channels to reduce microglial activation and restore cerebral vascular function.
Collapse
Affiliation(s)
- Joe Steinman
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| | - Hong-Shuo Sun
- Department of Physiology, University of Toronto, Toronto, ON, Canada.,Department of Surgery, University of Toronto, Toronto, ON, Canada
| | - Zhong-Ping Feng
- Department of Physiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
8
|
Hill LK, Hoang DM, Chiriboga LA, Wisniewski T, Sadowski MJ, Wadghiri YZ. Detection of Cerebrovascular Loss in the Normal Aging C57BL/6 Mouse Brain Using in vivo Contrast-Enhanced Magnetic Resonance Angiography. Front Aging Neurosci 2020; 12:585218. [PMID: 33192479 PMCID: PMC7606987 DOI: 10.3389/fnagi.2020.585218] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 09/11/2020] [Indexed: 12/28/2022] Open
Abstract
Microvascular rarefaction, or the decrease in vascular density, has been described in the cerebrovasculature of aging humans, rats, and, more recently, mice in the presence and absence of age-dependent diseases. Given the wide use of mice in modeling age-dependent human diseases of the cerebrovasculature, visualization, and quantification of the global murine cerebrovasculature is necessary for establishing the baseline changes that occur with aging. To provide in vivo whole-brain imaging of the cerebrovasculature in aging C57BL/6 mice longitudinally, contrast-enhanced magnetic resonance angiography (CE-MRA) was employed using a house-made gadolinium-bearing micellar blood pool agent. Enhancement in the vascular space permitted quantification of the detectable, or apparent, cerebral blood volume (aCBV), which was analyzed over 2 years of aging and compared to histological analysis of the cerebrovascular density. A significant loss in the aCBV was detected by CE-MRA over the aging period. Histological analysis via vessel-probing immunohistochemistry confirmed a significant loss in the cerebrovascular density over the same 2-year aging period, validating the CE-MRA findings. While these techniques use widely different methods of assessment and spatial resolutions, their comparable findings in detected vascular loss corroborate the growing body of literature describing vascular rarefaction aging. These findings suggest that such age-dependent changes can contribute to cerebrovascular and neurodegenerative diseases, which are modeled using wild-type and transgenic laboratory rodents.
Collapse
Affiliation(s)
- Lindsay K. Hill
- Department of Chemical and Biomolecular Engineering, NYU Tandon School of Engineering, Brooklyn, NY, United States
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI2R), NYU Grossman School of Medicine, New York, NY, United States
- Department of Radiology, Bernard and Irene Schwartz Center for Biomedical Imaging, NYU Grossman School of Medicine, New York, NY, United States
- Department of Biomedical Engineering, SUNY Downstate Medical Center, Brooklyn, NY, United States
| | - Dung Minh Hoang
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI2R), NYU Grossman School of Medicine, New York, NY, United States
- Department of Radiology, Bernard and Irene Schwartz Center for Biomedical Imaging, NYU Grossman School of Medicine, New York, NY, United States
| | - Luis A. Chiriboga
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, United States
| | - Thomas Wisniewski
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, United States
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, United States
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, United States
| | - Martin J. Sadowski
- Department of Neurology, NYU Grossman School of Medicine, New York, NY, United States
- Department of Psychiatry, NYU Grossman School of Medicine, New York, NY, United States
- Department of Biochemistry and Molecular Pharmacology, NYU Grossman School of Medicine, New York, NY, United States
| | - Youssef Z. Wadghiri
- Department of Radiology, Center for Advanced Imaging Innovation and Research (CAI2R), NYU Grossman School of Medicine, New York, NY, United States
- Department of Radiology, Bernard and Irene Schwartz Center for Biomedical Imaging, NYU Grossman School of Medicine, New York, NY, United States
| |
Collapse
|
9
|
Quintana DD, Lewis SE, Anantula Y, Garcia JA, Sarkar SN, Cavendish JZ, Brown CM, Simpkins JW. The cerebral angiome: High resolution MicroCT imaging of the whole brain cerebrovasculature in female and male mice. Neuroimage 2019; 202:116109. [PMID: 31446129 PMCID: PMC6942880 DOI: 10.1016/j.neuroimage.2019.116109] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 08/15/2019] [Accepted: 08/17/2019] [Indexed: 01/09/2023] Open
Abstract
The cerebrovascular system provides crucial functions that maintain metabolic and homeostatic states of the brain. Despite its integral role of supporting cerebral viability, the topological organization of these networks remains largely uncharacterized. This void in our knowledge surmises entirely from current technological limitations that prevent the capturing of data through the entire depth of the brain. We report high-resolution reconstruction and analysis of the complete vascular network of the entire brain at the capillary level in adult female and male mice using a vascular corrosion cast procedure. Vascular network analysis of the whole brain revealed sex-related differences of vessel hierarchy. In addition, region-specific network analysis demonstrated different patterns of angioarchitecture between brain subregions and sex. Furthermore, our group is the first to provide a three-dimensional analysis of the angioarchitecture and network organization in a single reconstructed tomographic data set that encompasses all hierarchy of vessels in the brain of the adult mouse.
Collapse
Affiliation(s)
- D D Quintana
- Department of Physiology and Pharmacology, Center for Basic Translational and Stroke Research, West Virginia University, Morgantown, WV, 26506, USA
| | - S E Lewis
- Department of Physiology and Pharmacology, Center for Basic Translational and Stroke Research, West Virginia University, Morgantown, WV, 26506, USA
| | - Y Anantula
- Department of Neuroscience, Center for Basic Translational and Stroke Research, West Virginia University, Morgantown, WV, 26506, USA
| | - J A Garcia
- Department of Neuroscience, Center for Basic Translational and Stroke Research, West Virginia University, Morgantown, WV, 26506, USA
| | - S N Sarkar
- Department of Physiology and Pharmacology, Center for Basic Translational and Stroke Research, West Virginia University, Morgantown, WV, 26506, USA
| | - J Z Cavendish
- Department of Physiology and Pharmacology, Center for Basic Translational and Stroke Research, West Virginia University, Morgantown, WV, 26506, USA
| | - C M Brown
- Department of Neuroscience, Center for Basic Translational and Stroke Research, West Virginia University, Morgantown, WV, 26506, USA
| | - J W Simpkins
- Department of Physiology and Pharmacology, Center for Basic Translational and Stroke Research, West Virginia University, Morgantown, WV, 26506, USA.
| |
Collapse
|
10
|
Li HC, Chen PY, Cheng HF, Kuo YM, Huang CC. In Vivo Visualization of Brain Vasculature in Alzheimer's Disease Mice by High-Frequency Micro-Doppler Imaging. IEEE Trans Biomed Eng 2019; 66:3393-3401. [PMID: 30872220 DOI: 10.1109/tbme.2019.2904702] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Cerebrovascular disorders are associated with Alzheimer's disease (AD). Functional analysis of the cerebral vasculature requires an in vivo approach to visualize the blood flow in small animal brains. This paper proposes a high-frequency micro-Doppler imaging (HFμDI) technology for mapping mouse cerebral vasculature. METHODS HFμDI used a 40-MHz transducer with an ultrafast ultrasound imaging technology that enabled in vivo visualization of the mouse brain up to 3 mm in depth; furthermore, a minimal vessel diameter of 48 μm could be determined. RESULTS Animal experiments determined that the cortical and hippocampal vessel density in young wild-type (WT) mice was similar to that in middle-aged WT mice. However, compared with the vessel density in middle-aged WT mice, that in middle-aged mice with AD was significantly lower, particularly in the hippocampus. DISCUSSION In vivo observation of cerebral vasculature demonstrated the effectiveness of HFμDI for the preclinical study of AD, and a potential way for human diagnosis was provided.
Collapse
|
11
|
Wälchli T, Ulmann-Schuler A, Hintermüller C, Meyer E, Stampanoni M, Carmeliet P, Emmert MY, Bozinov O, Regli L, Schwab ME, Vogel J, Hoerstrup SP. Nogo-A regulates vascular network architecture in the postnatal brain. J Cereb Blood Flow Metab 2017; 37:614-631. [PMID: 27927704 PMCID: PMC5381465 DOI: 10.1177/0271678x16675182] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Recently, we discovered a new role for the well-known axonal growth inhibitory molecule Nogo-A as a negative regulator of angiogenesis in the developing central nervous system. However, how Nogo-A affected the three-dimensional (3D) central nervous system (CNS) vascular network architecture remained unknown. Here, using vascular corrosion casting, hierarchical, synchrotron radiation μCT-based network imaging and computer-aided network analysis, we found that genetic ablation of Nogo-A significantly increased the three-dimensional vascular volume fraction in the postnatal day 10 (P10) mouse brain. More detailed analysis of the cerebral cortex revealed that this effect was mainly due to an increased number of capillaries and capillary branchpoints. Interestingly, other vascular parameters such as vessel diameter, -length, -tortuosity, and -volume were comparable between both genotypes for non-capillary vessels and capillaries. Taken together, our three-dimensional data showing more vessel segments and branchpoints at unchanged vessel morphology suggest that stimulated angiogenesis upon Nogo-A gene deletion results in the insertion of complete capillary micro-networks and not just single vessels into existing vascular networks. These findings significantly enhance our understanding of how angiogenesis, vascular remodeling, and three-dimensional vessel network architecture are regulated during central nervous system development. Nogo-A may therefore be a potential novel target for angiogenesis-dependent central nervous system pathologies such as brain tumors or stroke.
Collapse
Affiliation(s)
- Thomas Wälchli
- 1 Group of CNS Angiogenesis and Neurovascular Link, and Physician-Scientist Program, Institute for Regenerative Medicine, Neuroscience Center Zurich, and Division of Neurosurgery, University and University Hospital Zurich, Switzerland, and Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland.,2 Division of Neurosurgery and Laboratory of Molecular Neuro-Oncology, University Hospital Zurich, Zurich, Switzerland.,3 Brain Research Institute, University of Zurich and Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | | | | | - Eric Meyer
- 3 Brain Research Institute, University of Zurich and Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Marco Stampanoni
- 6 Swiss Light Source, Paul Scherrer Institute, Villigen, Switzerland.,7 Institute for Biomedical Engineering, University and ETH Zurich, Zurich, Switzerland
| | - Peter Carmeliet
- 8 Laboratory of Angiogenesis and Vascular Metabolism, Vesalius Research Center, Leuven, Belgium.,9 Department of Oncology, Laboratory of Angiogenesis and Neurovascular Link, Leuven, Belgium
| | - Maximilian Y Emmert
- 10 Institute for Regenerative Medicine and Clinic for Cardiovascular Surgery, University Hospital Zurich.,11 Wyss Translational Center Zurich, University of Zurich and Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Oliver Bozinov
- 2 Division of Neurosurgery and Laboratory of Molecular Neuro-Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Luca Regli
- 2 Division of Neurosurgery and Laboratory of Molecular Neuro-Oncology, University Hospital Zurich, Zurich, Switzerland
| | - Martin E Schwab
- 3 Brain Research Institute, University of Zurich and Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| | - Johannes Vogel
- 12 Institute of Veterinary Physiology, Vetsuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Simon P Hoerstrup
- 10 Institute for Regenerative Medicine and Clinic for Cardiovascular Surgery, University Hospital Zurich.,11 Wyss Translational Center Zurich, University of Zurich and Swiss Federal Institute of Technology (ETH) Zurich, Zurich, Switzerland
| |
Collapse
|
12
|
Synchrotron Radiation X-Ray Phase-Contrast Tomography Visualizes Microvasculature Changes in Mice Brains after Ischemic Injury. Neural Plast 2016; 2016:3258494. [PMID: 27563468 PMCID: PMC4983401 DOI: 10.1155/2016/3258494] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 06/02/2016] [Accepted: 06/23/2016] [Indexed: 12/15/2022] Open
Abstract
Imaging brain microvasculature is important in plasticity studies of cerebrovascular diseases. Applying contrast agents, traditional μCT and μMRI methods gain imaging contrast for vasculature. The aim of this study is to develop a synchrotron radiation X-ray inline phase-contrast tomography (SRXPCT) method for imaging the intact mouse brain (micro)vasculature in high resolution (~3.7 μm) without contrast agent. A specific preparation protocol was proposed to enhance the phase contrast of brain vasculature by using density difference over gas-tissue interface. The CT imaging system was developed and optimized to obtain 3D brain vasculature of adult male C57BL/6 mice. The SRXPCT method was further applied to investigate the microvasculature changes in mouse brains (n = 14) after 14-day reperfusion from transient middle cerebral artery occlusion (tMCAO). 3D reconstructions of brain microvasculature demonstrated that the branching radius ratio (post- to preinjury) of small vessels (radius < 7.4 μm) in the injury group was significantly smaller than that in the sham group (p < 0.05). This result revealed the active angiogenesis in the recovery brain after stroke. As a high-resolution and contrast-agent-free method, the SRXPCT method demonstrates higher potential in investigations of functional plasticity in cerebrovascular diseases.
Collapse
|
13
|
Tracing Mercox Injected at Acupuncture Points Under the Protocol of Partial Body Macerations in Mice. J Acupunct Meridian Stud 2015; 8:314-20. [DOI: 10.1016/j.jams.2015.08.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 08/03/2015] [Accepted: 08/07/2015] [Indexed: 11/18/2022] Open
|
14
|
Kelly P, McClean PL, Ackermann M, Konerding MA, Hölscher C, Mitchell CA. Restoration of cerebral and systemic microvascular architecture in APP/PS1 transgenic mice following treatment with Liraglutide™. Microcirculation 2015; 22:133-45. [PMID: 25556713 DOI: 10.1111/micc.12186] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Accepted: 12/23/2014] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Cerebral microvascular impairments occurring in AD may reduce Aβ peptide clearance and impact upon circulatory ultrastructure and function. We hypothesized that microvascular pathologies occur in organs responsible for systemic Aβ peptide clearance in a model of AD and that Liraglutide (Victoza(®)) improves vessel architecture. METHODS Seven-month-old APP/PS1 and age-matched wild-type mice received once-daily intraperitoneal injections of either Liraglutide or saline (n = 4 per group) for eight weeks. Casts of cerebral, splenic, hepatic, and renal microanatomy were analyzed using SEM. RESULTS Casts from wild-type mice showed regularly spaced microvasculature with smooth lumenal profiles, whereas APP/PS1 mice revealed evidence of microangiopathies including cerebral microanuerysms, intracerebral microvascular leakage, extravasation from renal glomerular microvessels, and significant reductions in both splenic sinus density (p = 0.0286) and intussusceptive microvascular pillars (p = 0.0412). Quantification of hepatic vascular ultrastructure in APP/PS1 mice revealed that vessel parameters (width, length, branching points, intussusceptive pillars and microaneurysms) were not significantly different from wild-type mice. Systemic administration of Liraglutide reduced the incidence of cerebral microanuerysms and leakage, restored renal microvascular architecture and significantly increased both splenic venous sinus number (p = 0.0286) and intussusceptive pillar formation (p = 0.0129). CONCLUSION Liraglutide restores cerebral, splenic, and renal architecture in APP/PS1 mice.
Collapse
Affiliation(s)
- Patricia Kelly
- School of Biomedical Sciences, University of Ulster, Coleraine, UK
| | | | | | | | | | | |
Collapse
|
15
|
Klohs J, Rudin M, Shimshek DR, Beckmann N. Imaging of cerebrovascular pathology in animal models of Alzheimer's disease. Front Aging Neurosci 2014; 6:32. [PMID: 24659966 PMCID: PMC3952109 DOI: 10.3389/fnagi.2014.00032] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 02/19/2014] [Indexed: 01/04/2023] Open
Abstract
In Alzheimer's disease (AD), vascular pathology may interact with neurodegeneration and thus aggravate cognitive decline. As the relationship between these two processes is poorly understood, research has been increasingly focused on understanding the link between cerebrovascular alterations and AD. This has at last been spurred by the engineering of transgenic animals, which display pathological features of AD and develop cerebral amyloid angiopathy to various degrees. Transgenic models are versatile for investigating the role of amyloid deposition and vascular dysfunction, and for evaluating novel therapeutic concepts. In addition, research has benefited from the development of novel imaging techniques, which are capable of characterizing vascular pathology in vivo. They provide vascular structural read-outs and have the ability to assess the functional consequences of vascular dysfunction as well as to visualize and monitor the molecular processes underlying these pathological alterations. This article focusses on recent in vivo small animal imaging studies addressing vascular aspects related to AD. With the technical advances of imaging modalities such as magnetic resonance, nuclear and microscopic imaging, molecular, functional and structural information related to vascular pathology can now be visualized in vivo in small rodents. Imaging vascular and parenchymal amyloid-β (Aβ) deposition as well as Aβ transport pathways have been shown to be useful to characterize their dynamics and to elucidate their role in the development of cerebral amyloid angiopathy and AD. Structural and functional imaging read-outs have been employed to describe the deleterious affects of Aβ on vessel morphology, hemodynamics and vascular integrity. More recent imaging studies have also addressed how inflammatory processes partake in the pathogenesis of the disease. Moreover, imaging can be pivotal in the search for novel therapies targeting the vasculature.
Collapse
Affiliation(s)
- Jan Klohs
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich Zurich, Switzerland ; Neuroscience Center Zurich, University of Zurich and ETH Zurich Zurich, Switzerland
| | - Markus Rudin
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich Zurich, Switzerland ; Neuroscience Center Zurich, University of Zurich and ETH Zurich Zurich, Switzerland ; Institute of Pharmacology and Toxicology, University of Zurich Zurich, Switzerland
| | - Derya R Shimshek
- Autoimmunity, Transplantation and Inflammation/Neuroinflammation Department, Novartis Institutes for BioMedical Research Basel, Switzerland
| | - Nicolau Beckmann
- Analytical Sciences and Imaging, Novartis Institutes for BioMedical Research Basel, Switzerland
| |
Collapse
|
16
|
Trotier AJ, Lefrançois W, Ribot EJ, Thiaudiere E, Franconi JM, Miraux S. Time-resolved TOF MR angiography in mice using a prospective 3D radial double golden angle approach. Magn Reson Med 2014; 73:984-94. [DOI: 10.1002/mrm.25201] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 02/11/2014] [Accepted: 02/11/2014] [Indexed: 11/09/2022]
Affiliation(s)
- Aurelien J. Trotier
- Centre de Résonance Magnétique des Systèmes Biologiques, UMR 5536 CNRS/Université Bordeaux Segalen; Bordeaux Cedex France
| | - William Lefrançois
- Centre de Résonance Magnétique des Systèmes Biologiques, UMR 5536 CNRS/Université Bordeaux Segalen; Bordeaux Cedex France
| | - Emeline J. Ribot
- Centre de Résonance Magnétique des Systèmes Biologiques, UMR 5536 CNRS/Université Bordeaux Segalen; Bordeaux Cedex France
| | - Eric Thiaudiere
- Centre de Résonance Magnétique des Systèmes Biologiques, UMR 5536 CNRS/Université Bordeaux Segalen; Bordeaux Cedex France
| | - Jean-Michel Franconi
- Centre de Résonance Magnétique des Systèmes Biologiques, UMR 5536 CNRS/Université Bordeaux Segalen; Bordeaux Cedex France
| | - Sylvain Miraux
- Centre de Résonance Magnétique des Systèmes Biologiques, UMR 5536 CNRS/Université Bordeaux Segalen; Bordeaux Cedex France
| |
Collapse
|
17
|
Zerbi V, Jansen D, Dederen PJ, Veltien A, Hamans B, Liu Y, Heerschap A, Kiliaan AJ. Microvascular cerebral blood volume changes in aging APPswe/PS1dE9 AD mouse model: a voxel-wise approach. Brain Struct Funct 2012; 218:1085-98. [DOI: 10.1007/s00429-012-0448-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 07/30/2012] [Indexed: 10/28/2022]
|
18
|
Contrast-enhanced magnetic resonance microangiography reveals remodeling of the cerebral microvasculature in transgenic ArcAβ mice. J Neurosci 2012; 32:1705-13. [PMID: 22302811 DOI: 10.1523/jneurosci.5626-11.2012] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Amyloid-β (Aβ) deposition in the cerebral vasculature is accompanied by remodeling which has a profound influence on vascular integrity and function. In the current study we have quantitatively assessed the age-dependent changes of the cortical vasculature in the arcAβ model of cerebral amyloidosis. To estimate the density of the cortical microvasculature in vivo, we used contrast-enhanced magnetic resonance microangiography (CE-μMRA). Three-dimensional gradient echo datasets with 60 μm isotropic resolution were acquired in 4- and 24-month-old arcAβ mice and compared with wild-type (wt) control mice of the same age before and after administration of superparamagnetic iron oxide nanoparticles. After segmentation of the cortical vasculature from difference images, an automated algorithm was applied for assessing the number and size distribution of intracortical vessels. With CE-μMRA, cerebral arteries and veins with a diameter of less than the nominal pixel resolution (60 μm) can be visualized. A significant age-dependent reduction in the number of functional intracortical microvessels (radii of 20-80 μm) has been observed in 24-month-old arcAβ mice compared with age-matched wt mice, whereas there was no difference between transgenic and wt mice of 4 months of age. Immunohistochemistry demonstrated strong fibrinogen and Aβ deposition in small- and medium-sized vessels, but not in large cerebral arteries, of 24-month-old arcAβ mice. The reduced density of transcortical vessels may thus be attributed to impaired perfusion and vascular occlusion caused by deposition of Aβ and fibrin. The study demonstrated that remodeling of the cerebrovasculature can be monitored noninvasively with CE-μMRA in mice.
Collapse
|
19
|
Lloyd DJ, Helmering J, Kaufman SA, Turk J, Silva M, Vasquez S, Weinstein D, Johnston B, Hale C, Véniant MM. A volumetric method for quantifying atherosclerosis in mice by using microCT: comparison to en face. PLoS One 2011; 6:e18800. [PMID: 21533112 PMCID: PMC3078927 DOI: 10.1371/journal.pone.0018800] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Accepted: 03/15/2011] [Indexed: 12/02/2022] Open
Abstract
Precise quantification of atherosclerotic plaque in preclinical models of atherosclerosis requires the volumetric assessment of the lesion(s) while maintaining in situ architecture. Here we use micro-computed tomography (microCT) to detect ex vivo aortic plaque established in three dyslipidemic mouse models of atherosclerosis. All three models lack the low-density lipoprotein receptor (Ldlr−/−), each differing in plaque severity, allowing the evaluation of different plaque volumes using microCT technology. From clearly identified lesions in the thoracic aorta from each model, we were able to determine plaque volume (0.04–3.1 mm3), intimal surface area (0.5–30 mm2), and maximum plaque (intimal-medial) thickness (0.1–0.7 mm). Further, quantification of aortic volume allowed calculation of vessel occlusion by the plaque. To validate microCT for future preclinical studies, we compared microCT data to intimal surface area (by using en face methodology). Both plaque surface area and plaque volume were in excellent correlation between microCT assessment and en face surface area (r2 = 0.99, p<0.0001 and r2 = 0.95, p<0.0001, respectively). MicroCT also identified internal characteristics of the lipid core and fibrous cap, which were confirmed pathologically as Stary type III-V lesions. These data validate the use of microCT technology to provide a more exact empirical measure of ex vivo plaque volume throughout the entire intact aorta in situ for the quantification of atherosclerosis in preclinical models.
Collapse
Affiliation(s)
- David J Lloyd
- Department of Metabolic Disorders, Amgen Inc., Thousand Oaks, California, United States of America.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Trancikova A, Ramonet D, Moore DJ. Genetic Mouse Models of Neurodegenerative Diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2011; 100:419-82. [DOI: 10.1016/b978-0-12-384878-9.00012-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
21
|
Howles GP, Ghaghada KB, Qi Y, Mukundan S, Johnson GA. High-resolution magnetic resonance angiography in the mouse using a nanoparticle blood-pool contrast agent. Magn Reson Med 2010; 62:1447-56. [PMID: 19902507 DOI: 10.1002/mrm.22154] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
High-resolution magnetic resonance angiography is already a useful tool for studying mouse models of human disease. Magnetic resonance angiography in the mouse is typically performed using time-of-flight contrast. In this work, a new long-circulating blood-pool contrast agent-a liposomal nanoparticle with surface-conjugated gadolinium (SC-Gd liposomes)-was evaluated for use in mouse neurovascular magnetic resonance angiography. A total of 12 mice were imaged. Scan parameters were optimized for both time-of-flight and SC-Gd contrast. Compared to time-of-flight contrast, SC-Gd liposomes (0.08 mmol/kg) enabled improved small-vessel contrast-to-noise ratio, larger field of view, shorter scan time, and imaging of venous structures. For a limited field of view, time-of-flight and SC-Gd were not significantly different; however, SC-Gd provided better contrast-to-noise ratio when the field of view encompassed the whole brain (P < 0.001) or the whole neurovascular axis (P < 0.001). SC-Gd allowed acquisition of high-resolution magnetic resonance angiography (52 x 52 x 100 micrometer(3) or 0.27 nL), with 123% higher (P < 0.001) contrast-to-noise ratio in comparable scan time ( approximately 45 min). Alternatively, SC-Gd liposomes could be used to acquire high-resolution magnetic resonance angiography (0.27 nL) with 32% higher contrast-to-noise ratio (P < 0.001) in 75% shorter scan time (12 min).
Collapse
Affiliation(s)
- Gabriel P Howles
- Center for In Vivo Microscopy, Department of Radiology, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | |
Collapse
|
22
|
El Tannir El Tayara N, Delatour B, Volk A, Dhenain M. Detection of vascular alterations by in vivo magnetic resonance angiography and histology in APP/PS1 mouse model of Alzheimer’s disease. MAGNETIC RESONANCE MATERIALS IN PHYSICS BIOLOGY AND MEDICINE 2010; 23:53-64. [DOI: 10.1007/s10334-009-0194-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2009] [Revised: 12/03/2009] [Accepted: 12/09/2009] [Indexed: 10/20/2022]
|
23
|
|
24
|
Schneider P, Krucker T, Meyer E, Ulmann-Schuler A, Weber B, Stampanoni M, Müller R. Simultaneous 3D visualization and quantification of murine bone and bone vasculature using micro-computed tomography and vascular replica. Microsc Res Tech 2009; 72:690-701. [DOI: 10.1002/jemt.20720] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
25
|
Schambach SJ, Bag S, Steil V, Isaza C, Schilling L, Groden C, Brockmann MA. Ultrafast High-Resolution In Vivo Volume-CTA of Mice Cerebral Vessels. Stroke 2009; 40:1444-50. [DOI: 10.1161/strokeaha.108.521740] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Sebastian J. Schambach
- From the Departments of Neuroradiology (S.S., S.B., C.I., C.G., M.A.B.), Radiation Oncology (V.S.), and Neurosurgical Research (L.S.), University Hospital Mannheim, Germany
| | - Simona Bag
- From the Departments of Neuroradiology (S.S., S.B., C.I., C.G., M.A.B.), Radiation Oncology (V.S.), and Neurosurgical Research (L.S.), University Hospital Mannheim, Germany
| | - Volker Steil
- From the Departments of Neuroradiology (S.S., S.B., C.I., C.G., M.A.B.), Radiation Oncology (V.S.), and Neurosurgical Research (L.S.), University Hospital Mannheim, Germany
| | - Cristina Isaza
- From the Departments of Neuroradiology (S.S., S.B., C.I., C.G., M.A.B.), Radiation Oncology (V.S.), and Neurosurgical Research (L.S.), University Hospital Mannheim, Germany
| | - Lothar Schilling
- From the Departments of Neuroradiology (S.S., S.B., C.I., C.G., M.A.B.), Radiation Oncology (V.S.), and Neurosurgical Research (L.S.), University Hospital Mannheim, Germany
| | - Christoph Groden
- From the Departments of Neuroradiology (S.S., S.B., C.I., C.G., M.A.B.), Radiation Oncology (V.S.), and Neurosurgical Research (L.S.), University Hospital Mannheim, Germany
| | - Marc A. Brockmann
- From the Departments of Neuroradiology (S.S., S.B., C.I., C.G., M.A.B.), Radiation Oncology (V.S.), and Neurosurgical Research (L.S.), University Hospital Mannheim, Germany
| |
Collapse
|
26
|
Thal DR, Capetillo-Zarate E, Larionov S, Staufenbiel M, Zurbruegg S, Beckmann N. Capillary cerebral amyloid angiopathy is associated with vessel occlusion and cerebral blood flow disturbances. Neurobiol Aging 2008; 30:1936-48. [PMID: 18359131 DOI: 10.1016/j.neurobiolaging.2008.01.017] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2007] [Revised: 01/04/2008] [Accepted: 01/30/2008] [Indexed: 10/22/2022]
Abstract
The role of cerebral amyloid angiopathy (CAA) in the pathogenesis of Alzheimer's disease (AD) is not fully understood. Here, we studied whether CAA is associated with alterations in microvascularisation in transgenic mouse models and in the human brain. APP23 mice at 25-26 months of age exhibited severe CAA in thalamic vessels whereas APP51/16 mice did not. Wild-type littermates were free of CAA. We found CAA-related capillary occlusion within the thalamus of APP23 mice but not in APP51/16 and wild-type mice. Magnetic resonance angiography (MRA) showed blood flow alterations in the thalamic vessels of APP23 mice. CAA-related capillary occlusion in the branches of the thalamoperforating arteries of APP23 mice, thereby, corresponded to the occurrence of blood flow disturbances. Similarly, CAA-related capillary occlusion was observed in the human occipital cortex of AD cases but less frequently in controls. These results indicate that capillary CAA can result in capillary occlusion and is associated with cerebral blood flow disturbances providing an additional mechanism for toxic effects of the amyloid beta-protein in AD.
Collapse
|
27
|
Altered morphology and 3D architecture of brain vasculature in a mouse model for Alzheimer's disease. Proc Natl Acad Sci U S A 2008; 105:3587-92. [PMID: 18305170 DOI: 10.1073/pnas.0709788105] [Citation(s) in RCA: 191] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Substantial evidence from epidemiological, pathological, and clinical reports suggests that vascular factors are critical in the pathogenesis of Alzheimer's disease (AD), and changes in blood flow are currently the most reliable indicators of the disease. We previously reported that older APP23 transgenic (tg) mice have significant blood flow alterations correlated with structural modifications of blood vessels. For the present study, our objective was to analyze the age-dependent morphological and architectural changes of the cerebral vasculature of APP23 tg mice. To visualize the 3D arrangement of the entire brain vasculature, we used vascular corrosion casts. Already at young ages, when typically parenchymal amyloid plaques are not yet present, APP23 tg mice had significant alterations, particularly of the microvasculature, often accompanied by small deposits attached to the vessels. In older animals, vasculature abruptly ended at amyloid plaques, resulting in holes. Often, small deposits were sitting near or at the end of truncated vessels. Between such holes, the surrounding vascular array appeared more dense and showed features typical for angiogenesis. We propose that small amyloid aggregates associated with the microvasculature lead to morphological and architectural alterations of the vasculature, resulting in altered local blood flow. The characteristic early onset of vascular alterations suggests that imaging blood flow and/or vasculature architecture could be used as a tool for early diagnosis of the disease and to monitor therapies.
Collapse
|
28
|
Dorr A, Sled JG, Kabani N. Three-dimensional cerebral vasculature of the CBA mouse brain: A magnetic resonance imaging and micro computed tomography study. Neuroimage 2007; 35:1409-23. [PMID: 17369055 DOI: 10.1016/j.neuroimage.2006.12.040] [Citation(s) in RCA: 203] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2006] [Revised: 12/07/2006] [Accepted: 12/08/2006] [Indexed: 11/15/2022] Open
Abstract
Studies of mouse cerebral vasculature to date have focused on the circle of Willis without examining the morphological distribution of blood vessels through the rest of the brain. Since mouse models are frequently used in brain-related studies, there is a need for a comprehensive cerebral vasculature atlas for the mouse with an emphasis on the location of vessels with respect to neuroanatomical structures, the watershed regions associated with specific arteries, as well as a consistent nomenclature of the cerebral vessels. This article describes such an atlas, based on a combination of magnetic resonance and computed tomography technology to yield high-resolution volumetric and vasculature data on CBA mouse. This three-dimensional vasculature dataset provides an anatomical resource for future mouse studies.
Collapse
Affiliation(s)
- A Dorr
- Sunnybrook Health Sciences Centre, 3080 Yonge Street, Suite 6020, P.O. Box 89, Toronto, ON, Canada M4N 3N1
| | | | | |
Collapse
|
29
|
Beckmann N, Kneuer R, Gremlich HU, Karmouty-Quintana H, Blé FX, Müller M. In vivo mouse imaging and spectroscopy in drug discovery. NMR IN BIOMEDICINE 2007; 20:154-85. [PMID: 17451175 DOI: 10.1002/nbm.1153] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
Imaging modalities such as micro-computed tomography (micro-CT), micro-positron emission tomography (micro-PET), high-resolution MRI, optical imaging, and high-resolution ultrasound have become invaluable tools in preclinical pharmaceutical research. They can be used to non-invasively investigate, in vivo, rodent biology and metabolism, disease models, and pharmacokinetics and pharmacodynamics of drugs. The advantages and limitations of each approach usually determine its application, and therefore a small-rodent imaging laboratory in a pharmaceutical environment should ideally provide access to several techniques. In this paper we aim to illustrate how these techniques may be used to obtain meaningful information for the phenotyping of transgenic mice and for the analysis of compounds in murine models of disease.
Collapse
Affiliation(s)
- Nicolau Beckmann
- Discovery Technologies, Novartis Institutes for BioMedical Research, Lichtstrasse 35, CH-4002 Basel, Switzerland.
| | | | | | | | | | | |
Collapse
|
30
|
Strome EM, Doudet DJ. Animal Models of Neurodegenerative Disease: Insights from In vivo Imaging Studies. Mol Imaging Biol 2007; 9:186-95. [PMID: 17357857 DOI: 10.1007/s11307-007-0093-4] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Animal models have been used extensively to understand the etiology and pathophysiology of human neurodegenerative diseases, and are an essential component in the development of therapeutic interventions for these disorders. In recent years, technical advances in imaging modalities such as positron emission tomography (PET) and magnetic resonance imaging (MRI) have allowed the use of these techniques for the evaluation of functional, neurochemical, and anatomical changes in the brains of animals. Combining animal models of neurodegenerative disorders with neuroimaging provides a powerful tool to follow the disease process, to examine compensatory mechanisms, and to investigate the effects of potential treatments preclinically to derive knowledge that will ultimately inform our clinical decisions. This article reviews the literature on the use of PET and MRI in animal models of Parkinson's disease, Huntington's disease, and Alzheimer's disease, and evaluates the strengths and limitations of brain imaging in animal models of neurodegenerative diseases.
Collapse
Affiliation(s)
- Elissa M Strome
- Pacific Parkinson's Research Centre, University of British Columbia, Vancouver, Canada.
| | | |
Collapse
|
31
|
Meyer EP, Beer GM, Lang A, Manestar M, Krucker T, Meier S, Mihic-Probst D, Groscurth P. Polyurethane elastomer: A new material for the visualization of cadaveric blood vessels. Clin Anat 2007; 20:448-54. [PMID: 17072868 DOI: 10.1002/ca.20403] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
A multitude of various materials are available for the visualization of cadaveric vessels, ranging from natural materials like gelatin and latex to synthetic materials like silicone rubber or acrylates. To achieve a detailed overview of the vascular architecture in microvascular studies in experimental flap surgery, the injected material should have low viscosity to assure perfusion of even the smallest vessels. In addition, the material ideally should have either no or only minimal shrinkage, and should harden within a reasonable time, but retain sufficient elasticity and resistance to withstand tearing off the delicate vessels during subsequent dissection or casting. Because none of the available injection materials adequately combines these attributes, we evaluated the polyurethane elastomer "PU4ii" in latissimus dorsi muscles as a new material for the visualization of cadaveric vessels in comparison with the frequently used silicone rubber. The dissection of vessels injected with PU4ii proved easy largely because of its exceptional hardness. Even if not visible before dissection, the completely perfused vessels were easily palpated in the surrounding fat or muscle tissue of the microsurgical latissimus dorsi model. Despite the significantly higher hardness of PU4ii over silicone rubber (98 Sh-A vs. 12 Sh-A), PU4ii proved enough elasticity (20-25 N/mm(2) E modulus) and a high tear resistance (64-68 N/mm vs. 15 N/mm) preventing breakage during dissection even within the smallest vessels. In contrast to silicone rubber (and latex or gelatin), the high corrosion resistance and form stability of PU4ii also allowed building of casts for qualitative examination by scanning electron microscopy and quantitative analysis of the vessel density using micro-computed tomography with accurate 3D representation. In this study we show that PU4ii has physical characteristics that make it a multi-purpose material that allows at the same breath an excellent gross visualization of the architecture of cadaveric blood vessels as well as a detailed evaluation of casts by modern microscopic and or radiologic tools. Thus, the new polyurethane elastomer PU4ii is in many respects superior to the widely used silicone rubber and can be strongly recommended as a visualization material for a comprehensive evaluation of cadaveric blood vessels in microsurgery.
Collapse
Affiliation(s)
- Eric P Meyer
- Institute of Zoology, University of Zurich, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Wolf SA, Kronenberg G, Lehmann K, Blankenship A, Overall R, Staufenbiel M, Kempermann G. Cognitive and physical activity differently modulate disease progression in the amyloid precursor protein (APP)-23 model of Alzheimer's disease. Biol Psychiatry 2006; 60:1314-23. [PMID: 16806094 DOI: 10.1016/j.biopsych.2006.04.004] [Citation(s) in RCA: 214] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2005] [Revised: 12/08/2005] [Accepted: 04/12/2006] [Indexed: 12/21/2022]
Abstract
BACKGROUND In aging mice, activity maintains hippocampal plasticity and adult hippocampal neurogenesis at a level corresponding to a younger age. Here we studied whether physical exercise and environmental enrichment would also affect brain plasticity in a mouse model of Alzheimer's disease (AD). METHODS Amyloid precursor protein (APP)-23 mice were housed under standard or enriched conditions or in cages equipped with a running wheel. We assessed beta-amyloid plaque load, adult hippocampal neurogenesis, spatial learning, and mRNA levels of trophic factors in the brain. RESULTS Despite stable beta-amyloid plaque load, enriched-living mice showed improved water maze performance, an up-regulation of hippocampal neurotrophin (NT-3) and brain-derived neurotrophic factor (BDNF) and increased hippocampal neurogenesis. In contrast, despite increased bodily fitness, wheel-running APP23 mice showed no change in spatial learning and no change in adult hippocampal neurogenesis but a down-regulation of hippocampal and cortical growth factors. CONCLUSIONS We conclude that structural and molecular prerequisites for activity-dependent plasticity are preserved in mutant mice with an AD-like pathology. Our study might help explain benefits of activity for the aging brain but also demonstrates differences between physical and more cognitive activity. It also suggests a possible cellular correlate for the dissociation between structural and functional pathology often found in AD.
Collapse
Affiliation(s)
- Susanne A Wolf
- Max Delbrück Center for Molecular Medicine, Berlin-Buch, Charité University Medicine, Berlin, Germany
| | | | | | | | | | | | | |
Collapse
|
33
|
Miraux S, Franconi JM, Thiaudière E. Blood velocity assessment using 3D bright-blood time-resolved magnetic resonance angiography. Magn Reson Med 2006; 56:469-73. [PMID: 16902973 DOI: 10.1002/mrm.20990] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Blood velocity is a functional parameter that is not easily assessed noninvasively, especially in small animals. A new noninvasive method that uses magnetic resonance angiography (MRA) to measure blood flows is proposed. This method is based on the time-of-flight (TOF) phenomenon. By initially suppressing the signal from the stationary spins in the area of interest, it is possible to sequentially visualize only the signal from the moving spins entering a given volume. With this method, 3D cine images of the blood flow can be generated by positive contrast, with unparalleled spatial (<200 microm) and temporal resolutions (<10 ms/image). As a result, it is possible to measure flow in sinuous paths. The present method was applied in vivo to measure the blood velocity in mouse carotid arteries. Because of its robustness and simplicity of implementation, this method has numerous potential applications for fundamental studies in small animal models.
Collapse
Affiliation(s)
- Sylvain Miraux
- Magnetic Resonance Center, CNRS-Victor Segalen University of Bordeaux 2, Bordeaux, France
| | | | | |
Collapse
|
34
|
Bolan PJ, Yacoub E, Garwood M, Ugurbil K, Harel N. In vivo micro-MRI of intracortical neurovasculature. Neuroimage 2006; 32:62-9. [PMID: 16675271 DOI: 10.1016/j.neuroimage.2006.03.027] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2005] [Revised: 03/08/2006] [Accepted: 03/14/2006] [Indexed: 11/20/2022] Open
Abstract
This work describes a methodology for in vivo MR imaging of arteries and veins within the visual cortex of the cat brain. Very high magnetic fields (9.4 T) and small field-of-view 3D acquisitions were used to image the neurovasculature at resolutions approaching the microscopic scale. A combination of time-of-flight MR angiography and T*(2)-weighted imaging, using both endogenous BOLD contrast and an exogenous iron-oxide contrast agent, provided high specificity for distinguishing between arteries and veins within the cortex. These acquisition techniques, combined with 3D image processing and display methods, were used to detect and visualize intracortical arteries and veins with diameters smaller than 100 microm. This methodology can be used for visualizing the neurovasculature or building models of the vascular network and may benefit a variety of research applications including fMRI, cerebrovascular disease and cancer angiogenesis.
Collapse
Affiliation(s)
- Patrick J Bolan
- Center for Magnetic Resonance Research, Department of Radiology, University of Minnesota School of Medicine, 2021 Sixth Street SE, Minneapolis, MN 55455, USA.
| | | | | | | | | |
Collapse
|
35
|
Heinzer S, Krucker T, Stampanoni M, Abela R, Meyer EP, Schuler A, Schneider P, Müller R. Hierarchical microimaging for multiscale analysis of large vascular networks. Neuroimage 2006; 32:626-36. [PMID: 16697665 DOI: 10.1016/j.neuroimage.2006.03.043] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2005] [Revised: 02/15/2006] [Accepted: 03/23/2006] [Indexed: 10/24/2022] Open
Abstract
There is a wide range of diseases and normal physiological processes that are associated with alterations of the vascular system in organs. Ex vivo imaging of large vascular networks became feasible with recent developments in microcomputed tomography (microCT). Current methods permit to visualize only limited numbers of physically excised regions of interests (ROIs) from larger samples. We developed a method based on modified vascular corrosion casting (VCC), scanning electron microscopy (SEM), and desktop and synchrotron radiation microCT (SRmicroCT) technologies to image vasculature at increasing levels of resolution, also referred to as hierarchical imaging. This novel approach allows nondestructive 3D visualization and quantification of large microvascular networks, while retaining a precise anatomical context for ROIs scanned at very high resolution. Scans of entire mouse brain VCCs were performed at 16-microm resolution with a desktop microCT system. Custom-made navigation software with a ROI selection tool enabled the identification of anatomical brain structures and precise placement of multiple ROIs. These were then scanned at 1.4-microm voxel size using SRmicroCT and a local tomography setup. A framework was developed for fast sample positioning, precise selection of ROIs, and sequential high-throughput scanning of a large numbers of brain VCCs. Despite the use of local tomography, exceptional image quality was achieved with SRmicroCT. This method enables qualitative and quantitative assessment of vasculature at unprecedented resolution and volume with relatively high throughput, opening new possibilities to study vessel architecture and vascular alterations in models of disease.
Collapse
Affiliation(s)
- Stefan Heinzer
- Institute for Biomedical Engineering, University and ETH Zürich, Zürich, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Krucker T, Lang A, Meyer EP. New polyurethane-based material for vascular corrosion casting with improved physical and imaging characteristics. Microsc Res Tech 2006; 69:138-47. [PMID: 16456839 DOI: 10.1002/jemt.20263] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Vascular corrosion casting has been established as a method to reconstruct the three-dimensional (3D) structure of blood vessels of organs and tissues. After replacing the blood volume with a low viscosity resin, the surrounding tissue is removed to replicate the vascular architecture, typically using scanning electron microscopy (SEM). To date available casting resins have had significant limitations such as lack of viscosity, leading to insufficient perfusion of smallest capillaries in organs and tissues of smaller species, interaction with surrounding tissue or fragility of resulting casts. We have reported here about a new polyurethane-based casting resin (PU4ii) with superior physical and imaging characteristics. Low viscosity, timely polymerization, and minimal shrinking of PU4ii produces high quality casts, including the finest capillaries. These casts are highly elastic while retaining their original structure to facilitate postcasting tissue dissection and pruning of casts. SEM images illustrate the high reproduction quality, including endothelial cell imprints, features that allow one to discern arterial and veinal vessels. For quantitative analysis, casts from PU4ii can be imaged using micro-computed tomography to produce digital 3D reconstructions. The inherent fluorescence of PU4ii is sufficient to reproduce casts with or without tissue using confocal microscopy (CM). Because of the simplified casting procedure, the high reproducibility and the superior reproduction quality, a combination of vascular corrosion casting using PU4ii with advanced imaging technologies has great potential to support the description of vascular defects and drug effects in disease models using mutant mice.
Collapse
Affiliation(s)
- Thomas Krucker
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, California 92037, USA.
| | | | | |
Collapse
|
37
|
Van Dam D, Vloeberghs E, Abramowski D, Staufenbiel M, De Deyn PPP. APP23 mice as a model of Alzheimer's disease: an example of a transgenic approach to modeling a CNS disorder. CNS Spectr 2005; 10:207-22. [PMID: 15744222 DOI: 10.1017/s1092852900010051] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Animal models are considered essential in research ensuing elucidation of human disease processes and subsequently, testing of potential therapeutic strategies. This is especially true for neurodegenerative disorders, in which the first steps in pathogenesis are often not accessible in human patients. Alzheimer's disease is vastly becoming a major medical and socioeconomic problem in our aging society. Valid animal models for this uniquely human condition should exhibit histopathological, biochemical, cognitive, and behavioral alterations observed in Alzheimer's disease patients. Major progress has been made since the understanding of the genetic basis of Alzheimer's disease and the development and improvement of transgenic mouse models. All present Alzheimer's disease models developed are partial but nevertheless essential in further unraveling the nature and spatial and temporal development of the complex molecular pathology underlying this condition. One of the more recent transgenic attempts to model Alzheimer's disease is the APP23 transgenic mouse. This article describes the development and assessment of this human amyloid precursor protein overexpression model. We summarize histopathological and biochemical, cognitive and behavioral observations made in heterozygous APP23 mice, thereby emphasizing the model's contribution to clarification of neurodegenerative disease mechanisms. In addition, the first therapeutic interventions in the APP23 model are included.
Collapse
Affiliation(s)
- Debby Van Dam
- Laboratory of Neurochemistry and Behaviour, Institute Born-Bunge, University of Antwerp, Antwerp, Wilrijk, Belgium
| | | | | | | | | |
Collapse
|