1
|
Li W, Cheng J, Liu C, Zhang N, Lin H, He F, Gan Z, Zhang P, Qin M, Hou Y. Shine and darkle the blood vessels: Multiparameter hypersensitive MR angiography for diagnosis of panvascular diseases. SCIENCE ADVANCES 2024; 10:eadq4082. [PMID: 39365870 PMCID: PMC11451532 DOI: 10.1126/sciadv.adq4082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 08/28/2024] [Indexed: 10/06/2024]
Abstract
Magnetic resonance angiography (MRA) is pivotal for diagnosing panvascular diseases. However, single-modality MRA falls short in diagnosing diverse vascular abnormalities. Thus, contrast agents combining T1 and T2 effects are sought for multiparameter MRA with clinical promise, yet achieving a balance in T1 and T2 contrast enhancement effects remains a scientific challenge. Herein, we developed a hypersensitive multiparameter MRA strategy using dual-modality NaGdF4 nanoparticles. Because of the longer tumbling time (τR), NaGdF4 nanoparticles can improve the longitudinal relaxivity (r1), brightening vessels in T1-weighted sequences. Simultaneously, the regular arrangement of Gd3+ in the crystal induces magnetic anisotropy, creating local static magnetic field heterogeneity and generating negative signals in T2-weighted sequences. Consequently, the efficacy of NaGdF4-enhanced high-resolution multiparameter MRA has been validated in diagnosing ischemic stroke and Alzheimer's disease in rodent models. In addition, the dual-contrast imaging has been realized on swine with a clinical 3.0-T magnetic resonance imaging scanner, highly emphasizing the clinical translation prospect.
Collapse
Affiliation(s)
- Wenyue Li
- College of Materials Science and Engineering and College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Junwei Cheng
- College of Materials Science and Engineering and College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Chuang Liu
- College of Materials Science and Engineering and College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Ni Zhang
- Department of Psychiatry and Center for Preclinical Safety Evaluation of Drugs, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Hua Lin
- Department of Psychiatry and Center for Preclinical Safety Evaluation of Drugs, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Fangfei He
- College of Materials Science and Engineering and College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Zhihua Gan
- College of Materials Science and Engineering and College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Peisen Zhang
- College of Materials Science and Engineering and College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Meng Qin
- Department of Psychiatry and Center for Preclinical Safety Evaluation of Drugs, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yi Hou
- College of Materials Science and Engineering and College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| |
Collapse
|
2
|
Sharma M, Pal P, Gupta SK. Advances in Alzheimer's disease: A multifaceted review of potential therapies and diagnostic techniques for early detection. Neurochem Int 2024; 177:105761. [PMID: 38723902 DOI: 10.1016/j.neuint.2024.105761] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 04/20/2024] [Accepted: 05/06/2024] [Indexed: 06/04/2024]
Abstract
Alzheimer's disease (AD) remains one of the most formidable neurological disorders, affecting millions globally. This review provides a holistic overview of the therapeutic strategies, both conventional and novel, aimed at mitigating the impact of AD. Initially, we delve into the conventional approach, emphasizing the role of Acetylcholinesterase (AChE) inhibition, which has been a cornerstone in AD management. As our understanding of AD evolves, several novel potential approaches emerge. We discuss the promising roles of Butyrylcholinesterase (BChE) inhibition, Tau Protein inhibitors, COX-2 inhibition, PPAR-γ agonism, and FAHH inhibition, among others. The potential of the endocannabinoids (eCB) system, cholesterol-lowering drugs, metal chelators, and MMPs inhibitors are also explored, culminating in the exploration of the pivotal role of microRNA in AD progression. Parallel to these therapeutic insights, we shed light on the novel tools and methodologies revolutionizing AD research. From the quantitative analysis of gene expression by qRTPCR to the evaluation of mitochondrial function using induced pluripotent stem cells (iPSCs), the advances in diagnostic and research tools offer renewed hope. Moreover, we explore the current landscape of clinical trials, highlighting the leading drug interventions and their respective stages of development. This comprehensive review concludes with a look into the future perspectives, capturing the potential breakthroughs and innovations on the horizon. Through a synthesis of current knowledge and emerging research, this article aims to provide a consolidated resource for clinicians, researchers, and academicians in the realm of Alzheimer's disease.
Collapse
Affiliation(s)
- Monika Sharma
- Faculty of Pharmacy, Department of Pharmacology, Swami Vivekanand Subharti University, Meerut, Uttar Pradesh, India
| | - Pankaj Pal
- Department of Pharmacy, Banasthali Vidyapith, Rajasthan, India.
| | - Sukesh Kumar Gupta
- KIET School of Pharmacy, KIET Group of Institutions, Ghaziabad, Uttar Pradesh, India; Department of Ophthalmology, Visual and Anatomical Sciences (OVAS), School of Medicine, Wayne State University, USA.
| |
Collapse
|
3
|
Collins HM, Greenfield S. Rodent Models of Alzheimer's Disease: Past Misconceptions and Future Prospects. Int J Mol Sci 2024; 25:6222. [PMID: 38892408 PMCID: PMC11172947 DOI: 10.3390/ijms25116222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 05/28/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease with no effective treatments, not least due to the lack of authentic animal models. Typically, rodent models recapitulate the effects but not causes of AD, such as cholinergic neuron loss: lesioning of cholinergic neurons mimics the cognitive decline reminiscent of AD but not its neuropathology. Alternative models rely on the overexpression of genes associated with familial AD, such as amyloid precursor protein, or have genetically amplified expression of mutant tau. Yet transgenic rodent models poorly replicate the neuropathogenesis and protein overexpression patterns of sporadic AD. Seeding rodents with amyloid or tau facilitates the formation of these pathologies but cannot account for their initial accumulation. Intracerebral infusion of proinflammatory agents offer an alternative model, but these fail to replicate the cause of AD. A novel model is therefore needed, perhaps similar to those used for Parkinson's disease, namely adult wildtype rodents with neuron-specific (dopaminergic) lesions within the same vulnerable brainstem nuclei, 'the isodendritic core', which are the first to degenerate in AD. Site-selective targeting of these nuclei in adult rodents may recapitulate the initial neurodegenerative processes in AD to faithfully mimic its pathogenesis and progression, ultimately leading to presymptomatic biomarkers and preventative therapies.
Collapse
Affiliation(s)
- Helen M. Collins
- Neuro-Bio Ltd., Building F5 The Culham Campus, Abingdon OX14 3DB, UK;
| | | |
Collapse
|
4
|
Hayden MR. A Closer Look at the Perivascular Unit in the Development of Enlarged Perivascular Spaces in Obesity, Metabolic Syndrome, and Type 2 Diabetes Mellitus. Biomedicines 2024; 12:96. [PMID: 38255202 PMCID: PMC10813073 DOI: 10.3390/biomedicines12010096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 12/28/2023] [Accepted: 12/28/2023] [Indexed: 01/24/2024] Open
Abstract
The recently described perivascular unit (PVU) resides immediately adjacent to the true capillary neurovascular unit (NVU) in the postcapillary venule and contains the normal-benign perivascular spaces (PVS) and pathological enlarged perivascular spaces (EPVS). The PVS are important in that they have recently been identified to be the construct and the conduit responsible for the delivery of metabolic waste from the interstitial fluid to the ventricular cerebrospinal fluid for disposal into the systemic circulation, termed the glymphatic system. Importantly, the outermost boundary of the PVS is lined by protoplasmic perivascular astrocyte endfeet (pvACef) that communicate with regional neurons. As compared to the well-recognized and described neurovascular unit (NVU) and NVU coupling, the PVU is less well understood and remains an emerging concept. The primary focus of this narrative review is to compare the similarities and differences between these two units and discuss each of their structural and functional relationships and how they relate not only to brain homeostasis but also how they may relate to the development of multiple clinical neurological disease states and specifically how they may relate to obesity, metabolic syndrome, and type 2 diabetes mellitus. Additionally, the concept and importance of a perisynaptic astrocyte coupling to the neuronal synapses with pre- and postsynaptic neurons will also be considered as a perisynaptic unit to provide for the creation of the information transfer in the brain via synaptic transmission and brain homeostasis. Multiple electron microscopic images and illustrations will be utilized in order to help explain these complex units.
Collapse
Affiliation(s)
- Melvin R Hayden
- Department of Internal Medicine, Endocrinology Diabetes and Metabolism, Diabetes and Cardiovascular Disease Center, University of Missouri School of Medicine, One Hospital Drive, Columbia, MO 65211, USA
| |
Collapse
|
5
|
Zedde M, Grisendi I, Assenza F, Vandelli G, Napoli M, Moratti C, Lochner P, Seiffge DJ, Piazza F, Valzania F, Pascarella R. The Venular Side of Cerebral Amyloid Angiopathy: Proof of Concept of a Neglected Issue. Biomedicines 2023; 11:2663. [PMID: 37893037 PMCID: PMC10604278 DOI: 10.3390/biomedicines11102663] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
Small vessel diseases (SVD) is an umbrella term including several entities affecting small arteries, arterioles, capillaries, and venules in the brain. One of the most relevant and prevalent SVDs is cerebral amyloid angiopathy (CAA), whose pathological hallmark is the deposition of amyloid fragments in the walls of small cortical and leptomeningeal vessels. CAA frequently coexists with Alzheimer's Disease (AD), and both are associated with cerebrovascular events, cognitive impairment, and dementia. CAA and AD share pathophysiological, histopathological and neuroimaging issues. The venular involvement in both diseases has been neglected, although both animal models and human histopathological studies found a deposition of amyloid beta in cortical venules. This review aimed to summarize the available information about venular involvement in CAA, starting from the biological level with the putative pathomechanisms of cerebral damage, passing through the definition of the peculiar angioarchitecture of the human cortex with the functional organization and consequences of cortical arteriolar and venular occlusion, and ending to the hypothesized links between cortical venular involvement and the main neuroimaging markers of the disease.
Collapse
Affiliation(s)
- Marialuisa Zedde
- Neurology Unit, Stroke Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Ilaria Grisendi
- Neurology Unit, Stroke Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Federica Assenza
- Neurology Unit, Stroke Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Gabriele Vandelli
- Neurology Unit, Stroke Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Manuela Napoli
- Neuroradiology Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Claudio Moratti
- Neuroradiology Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Piergiorgio Lochner
- Department of Neurology, Saarland University Medical Center, 66421 Homburg, Germany;
| | - David J. Seiffge
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, 3010 Bern, Switzerland
| | - Fabrizio Piazza
- CAA and AD Translational Research and Biomarkers Laboratory, School of Medicine and Surgery, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy;
| | - Franco Valzania
- Neurology Unit, Stroke Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| | - Rosario Pascarella
- Neuroradiology Unit, AUSL-IRCCS di Reggio Emilia, Via Amendola 2, 42122 Reggio Emilia, Italy
| |
Collapse
|
6
|
Zebrafish as a Potential Model for Neurodegenerative Diseases: A Focus on Toxic Metals Implications. Int J Mol Sci 2023; 24:ijms24043428. [PMID: 36834835 PMCID: PMC9959844 DOI: 10.3390/ijms24043428] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/31/2023] [Accepted: 02/05/2023] [Indexed: 02/11/2023] Open
Abstract
In the last century, industrial activities increased and caused multiple health problems for humans and animals. At this moment, heavy metals are considered the most harmful substances for their effects on organisms and humans. The impact of these toxic metals, which have no biological role, poses a considerable threat and is associated with several health problems. Heavy metals can interfere with metabolic processes and can sometimes act as pseudo-elements. The zebrafish is an animal model progressively used to expose the toxic effects of diverse compounds and to find treatments for different devastating diseases that human beings are currently facing. This review aims to analyse and discuss the value of zebrafish as animal models used in neurological conditions, such as Alzheimer's disease (AD), and Parkinson's disease (PD), particularly in terms of the benefits of animal models and the limitations that exist.
Collapse
|
7
|
Venular amyloid accumulation in transgenic Fischer 344 Alzheimer’s disease rats. Sci Rep 2022; 12:15287. [PMID: 36088484 PMCID: PMC9464208 DOI: 10.1038/s41598-022-19549-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/31/2022] [Indexed: 11/29/2022] Open
Abstract
Strong evidence demonstrates a significant association between cerebral amyloid angiopathy (CAA) and Alzheimer’s disease (AD). For this reason, interest in understanding the underlying vascular pathologies that contribute to AD remain. CAA research has primarily focused on arterioles and capillaries, overlooking the draining venules. Therefore, this study sought to examine venular amyloid pathology and its relationship to arteriolar amyloidosis throughout AD progression in the TgF344-AD rat model. Antibodies targeting the amyloid-beta peptide (Aβ) sequence suggest morphological differences between arteriolar and venular amyloid. Mass spectrometric analyses of isolated cortical parenchymal plaques, arteriolar and venular amyloid demonstrated presence of Aβ in all three samples, as well as proteins known to be associated with AD. Histopathological analysis indicates a significant age effect for both arteriolar and venular amyloid accumulation, with accumulation initiated in the somatosensory cortex followed by the motor and cingulate cortex. Lastly, significant arteriolar amyloid accumulates relative to venular amyloid deposition in AD progression. Overall, understanding venular and arteriolar amyloid pathology provides insight into the complex connection between CAA and AD.
Collapse
|
8
|
Handa T, Sasaki H, Takao M, Tano M, Uchida Y. Proteomics-based investigation of cerebrovascular molecular mechanisms in cerebral amyloid angiopathy by the FFPE-LMD-PCT-SWATH method. Fluids Barriers CNS 2022; 19:56. [PMID: 35778717 PMCID: PMC9250250 DOI: 10.1186/s12987-022-00351-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 06/21/2022] [Indexed: 11/25/2022] Open
Abstract
Background Cerebral amyloid angiopathy (CAA) occurs in 80% of patients with Alzheimer’s disease (AD) and is mainly caused by the abnormal deposition of Aβ in the walls of cerebral blood vessels. Cerebrovascular molecular mechanisms in CAA were investigated by using comprehensive and accurate quantitative proteomics. Methods Concerning the molecular mechanisms specific to CAA, formalin-fixed paraffin-embedded (FFPE) sections were prepared from patients having AD neuropathologic change (ADNC) with severe cortical Aβ vascular deposition (ADNC +/CAA +), and from patients having ADNC without vascular deposition of Aβ (ADNC +/CAA −; so called, AD). Cerebral cortical vessels were isolated from FFPE sections using laser microdissection (LMD), processed by pressure cycling technology (PCT), and applied to SWATH (sequential window acquisition of all theoretical fragment ion spectra) proteomics. Results The protein expression levels of 17 proteins in ADNC +/CAA +/H donors (ADNC +/CAA + donors with highly abundant Aβ in capillaries) were significantly different from those in ADNC +/CAA − and ADNC −/CAA − donors. Furthermore, we identified 56 proteins showing more than a 1.5-fold difference in average expression levels between ADNC +/CAA + and ADNC −/CAA − donors, and were significantly correlated with the levels of Aβ or Collagen alpha-2(VI) chain (COL6A2) (CAA markers) in 11 donors (6 ADNC +/CAA + and 5 ADNC −/CAA −). Over 70% of the 56 proteins showed ADNC +/CAA + specific changes in protein expression. The comparative analysis with brain parenchyma showed that more than 90% of the 56 proteins were vascular-specific pathological changes. A literature-based pathway analysis showed that 42 proteins are associated with fibrosis, oxidative stress and apoptosis. This included the increased expression of Heat shock protein HSP 90-alpha, CD44 antigen and Carbonic anhydrase 1 which are inhibited by potential drugs against CAA. Conclusions The combination of LMD-based isolation of vessels from FFPE sections, PCT-assisted sample processing and SWATH analysis (FFPE-LMD-PCT-SWATH method) revealed for the first time the changes in the expression of many proteins that are involved in fibrosis, ROS production and cell death in ADNC +/CAA + (CAA patients) vessels. The findings reported herein would be useful for developing a better understanding of the pathology of CAA and for promoting the discovery and development of drugs and biomarkers for CAA. Supplementary Information The online version contains supplementary material available at 10.1186/s12987-022-00351-x.
Collapse
Affiliation(s)
- Takumi Handa
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Hayate Sasaki
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Masaki Takao
- Department of Neurology and Brain Bank, Mihara Memorial Hospital, Isesaki, Japan.,Department of Clinical Laboratory, National Center of Neurology and Psychiatry, National Center Hospital, Kodaira, Japan
| | - Mitsutoshi Tano
- Department of Neurology and Brain Bank, Mihara Memorial Hospital, Isesaki, Japan
| | - Yasuo Uchida
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan. .,Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aoba, Aramaki, Aoba-ku, Sendai, 980-8578, Japan.
| |
Collapse
|
9
|
Nehra G, Bauer B, Hartz AMS. Blood-brain barrier leakage in Alzheimer's disease: From discovery to clinical relevance. Pharmacol Ther 2022; 234:108119. [PMID: 35108575 PMCID: PMC9107516 DOI: 10.1016/j.pharmthera.2022.108119] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/14/2022] [Accepted: 01/18/2022] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia. AD brain pathology starts decades before the onset of clinical symptoms. One early pathological hallmark is blood-brain barrier dysfunction characterized by barrier leakage and associated with cognitive decline. In this review, we summarize the existing literature on the extent and clinical relevance of barrier leakage in AD. First, we focus on AD animal models and their susceptibility to barrier leakage based on age and genetic background. Second, we re-examine barrier dysfunction in clinical and postmortem studies, summarize changes that lead to barrier leakage in patients and highlight the clinical relevance of barrier leakage in AD. Third, we summarize signaling mechanisms that link barrier leakage to neurodegeneration and cognitive decline in AD. Finally, we discuss clinical relevance and potential therapeutic strategies and provide future perspectives on investigating barrier leakage in AD. Identifying mechanistic steps underlying barrier leakage has the potential to unravel new targets that can be used to develop novel therapeutic strategies to repair barrier leakage and slow cognitive decline in AD and AD-related dementias.
Collapse
Affiliation(s)
- Geetika Nehra
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Bjoern Bauer
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY, USA
| | - Anika M S Hartz
- Sanders-Brown Center on Aging, College of Medicine, University of Kentucky, Lexington, KY, USA; Department of Pharmacology and Nutritional Sciences, College of Medicine, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
10
|
MacKenzie JL, Ivanova N, Nell HJ, Giordano CR, Terlecky SR, Agca C, Agca Y, Walton PA, Whitehead SN, Cechetto DF. Microglial inflammation and cognitive dysfunction in comorbid rat models of striatal ischemic stroke and alzheimer’s disease: effects of antioxidant catalase-SKL on behavioral and cellular pathology. Neuroscience 2022; 487:47-65. [DOI: 10.1016/j.neuroscience.2022.01.026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/26/2022] [Accepted: 01/28/2022] [Indexed: 12/25/2022]
|
11
|
Pang K, Jiang R, Zhang W, Yang Z, Li LL, Shimozawa M, Tambaro S, Mayer J, Zhang B, Li M, Wang J, Liu H, Yang A, Chen X, Liu J, Winblad B, Han H, Jiang T, Wang W, Nilsson P, Guo W, Lu B. An App knock-in rat model for Alzheimer's disease exhibiting Aβ and tau pathologies, neuronal death and cognitive impairments. Cell Res 2022; 32:157-175. [PMID: 34789895 PMCID: PMC8807612 DOI: 10.1038/s41422-021-00582-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Accepted: 10/04/2021] [Indexed: 12/21/2022] Open
Abstract
A major obstacle in Alzheimer's disease (AD) research is the lack of predictive and translatable animal models that reflect disease progression and drug efficacy. Transgenic mice overexpressing amyloid precursor protein (App) gene manifest non-physiological and ectopic expression of APP and its fragments in the brain, which is not observed in AD patients. The App knock-in mice circumvented some of these problems, but they do not exhibit tau pathology and neuronal death. We have generated a rat model, with three familiar App mutations and humanized Aβ sequence knocked into the rat App gene. Without altering the levels of full-length APP and other APP fragments, this model exhibits pathologies and disease progression resembling those in human patients: deposit of Aβ plaques in relevant brain regions, microglia activation and gliosis, progressive synaptic degeneration and AD-relevant cognitive deficits. Interestingly, we have observed tau pathology, neuronal apoptosis and necroptosis and brain atrophy, phenotypes rarely seen in other APP models. This App knock-in rat model may serve as a useful tool for AD research, identifying new drug targets and biomarkers, and testing therapeutics.
Collapse
Affiliation(s)
- Keliang Pang
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, Beijing, China
- R&D Center for the Diagnosis and Treatment of Major Brain Diseases, Research Institute of Tsinghua University in Shenzhen, Shenzhen, Guangdong, China
- Beijing Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Richeng Jiang
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
- Department of Otorhinolaryngology Head and Neck Surgery, The First Hospital of Jilin University, Changchun, China
| | - Wei Zhang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, and Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Zhengyi Yang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Lin-Lin Li
- Research Center for Brain-inspired Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, School of Future Technology, University of CAS, and CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Makoto Shimozawa
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Simone Tambaro
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Johanna Mayer
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Baogui Zhang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Man Li
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, and Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Jiesi Wang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, and Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Hang Liu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, Beijing, China
- R&D Center for the Diagnosis and Treatment of Major Brain Diseases, Research Institute of Tsinghua University in Shenzhen, Shenzhen, Guangdong, China
- Beijing Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Ailing Yang
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, Beijing, China
| | - Xi Chen
- Research Center for Brain-inspired Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, School of Future Technology, University of CAS, and CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Jiazheng Liu
- Research Center for Brain-inspired Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, School of Future Technology, University of CAS, and CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Bengt Winblad
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
- Theme Aging, Karolinska University Hospital, Huddinge, Sweden
| | - Hua Han
- Research Center for Brain-inspired Intelligence, National Laboratory of Pattern Recognition, Institute of Automation, School of Future Technology, University of CAS, and CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Tianzi Jiang
- Brainnetome Center, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Weiwen Wang
- CAS Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, and Department of Psychology, University of Chinese Academy of Sciences, Beijing, China
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Wei Guo
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, Beijing, China.
- R&D Center for the Diagnosis and Treatment of Major Brain Diseases, Research Institute of Tsinghua University in Shenzhen, Shenzhen, Guangdong, China.
- Beijing Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
| | - Bai Lu
- School of Pharmaceutical Sciences, IDG/McGovern Institute for Brain Research, Tsinghua University-Peking University Joint Center for Life Sciences, Tsinghua University, Beijing, China.
- R&D Center for the Diagnosis and Treatment of Major Brain Diseases, Research Institute of Tsinghua University in Shenzhen, Shenzhen, Guangdong, China.
- Beijing Tiantan Hospital, Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.
| |
Collapse
|
12
|
Perosa V, Rotta J, Schreiber S. Author Response: Detection of Cerebral Microbleeds With Venous Connection at 7-Tesla MRI. Neurology 2021; 97:840. [PMID: 34697211 DOI: 10.1212/wnl.0000000000012731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
13
|
Cao Y, Ao DH, Ma C, Qiu WY, Zhu YC. Immunoreactivity and a new staining method of monocarboxylate transporter 1 located in endothelial cells of cerebral vessels of human brain in distinguishing cerebral venules from arterioles. Eur J Histochem 2021; 65. [PMID: 34595897 PMCID: PMC8506011 DOI: 10.4081/ejh.2021.3306] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/08/2021] [Indexed: 11/23/2022] Open
Abstract
Distinguishing brain venules from arterioles with arteriolosclerosis is less reliable using traditional staining methods. We aimed to immunohistochemically assess the monocarboxylate transporter 1 (MCT1), a specific marker of venous endothelium found in rodent studies, in different caliber vessels in human brains. Both largeand small-caliber cerebral vessels were dissected from four autopsy donors. Immunoreactivity for MCT1 was examined in all autopsied human brain tissues, and then each vessel was identified by neuropathologists using hematoxylin and eosin stain, the Verhoeff's Van Gieson stain, immunohistochemical stain with antibodies for α-smooth muscle actin and MCT1 in sequence. A total of 61 cerebral vessels, including 29 arteries and 32 veins were assessed. Immunoreactivity for MCT1 was observed in the endothelial cells of various caliber veins as well as the capillaries, whereas that was immunenegative in the endothelium of arteries. The different labeling patterns for MCT1 could aid in distinguishing various caliber veins from arteries, whereas assessment using the vessel shape, the internal elastic lamina, and the pattern of smooth muscle fibers failed to make the distinction between small-caliber veins and sclerotic arterioles. In conclusion, MCT1 immunohistochemical staining is a sensitive and reliable method to distinguish cerebral veins from arteries.
Collapse
Affiliation(s)
- Yuan Cao
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College.
| | - Dong-Hui Ao
- Department of Neurology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing.
| | - Chao Ma
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College.
| | - Wen-Ying Qiu
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College.
| | - Yi-Cheng Zhu
- Department of Human Anatomy, Histology and Embryology, Institute of Basic Medical Sciences, Neuroscience Center, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College.
| |
Collapse
|
14
|
mTOR Attenuation with Rapamycin Reverses Neurovascular Uncoupling and Memory Deficits in Mice Modeling Alzheimer's Disease. J Neurosci 2021; 41:4305-4320. [PMID: 33888602 DOI: 10.1523/jneurosci.2144-20.2021] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 02/19/2021] [Accepted: 03/02/2021] [Indexed: 02/07/2023] Open
Abstract
Vascular dysfunction is a universal feature of aging and decreased cerebral blood flow has been identified as an early event in the pathogenesis of Alzheimer's disease (AD). Cerebrovascular dysfunction in AD includes deficits in neurovascular coupling (NVC), a mechanism that ensures rapid delivery of energy substrates to active neurons through the blood supply. The mechanisms underlying NVC impairment in AD, however, are not well understood. We have previously shown that mechanistic/mammalian target of rapamycin (mTOR) drives cerebrovascular dysfunction in models of AD by reducing the activity of endothelial nitric oxide synthase (eNOS), and that attenuation of mTOR activity with rapamycin is sufficient to restore eNOS-dependent cerebrovascular function. Here we show mTOR drives NVC impairments in an AD model through the inhibition of neuronal NOS (nNOS)- and non-NOS-dependent components of NVC, and that mTOR attenuation with rapamycin is sufficient to restore NVC and even enhance it above WT responses. Restoration of NVC and concomitant reduction of cortical amyloid-β levels effectively treated memory deficits in 12-month-old hAPP(J20) mice. These data indicate that mTOR is a critical driver of NVC dysfunction and underlies cognitive impairment in an AD model. Together with our previous findings, the present studies suggest that mTOR promotes cerebrovascular dysfunction in AD, which is associated with early disruption of nNOS activation, through its broad negative impact on nNOS as well as on non-NOS components of NVC. Our studies highlight the potential of mTOR attenuation as an efficacious treatment for AD and potentially other neurologic diseases of aging.SIGNIFICANCE STATEMENT Failure of the blood flow response to neuronal activation [neurovascular coupling (NVC)] in a model of AD precedes the onset of AD-like cognitive symptoms and is driven, to a large extent, by mammalian/mechanistic target of rapamycin (mTOR)-dependent inhibition of nitric oxide synthase activity. Our studies show that mTOR also drives AD-like failure of non-nitric oxide (NO)-mediated components of NVC. Thus, mTOR attenuation may serve to treat AD, where we find that neuronal NO synthase is profoundly reduced early in disease progression, and potentially other neurologic diseases of aging with cerebrovascular dysfunction as part of their etiology.
Collapse
|
15
|
Rotta J, Perosa V, Yakupov R, Kuijf HJ, Schreiber F, Dobisch L, Oltmer J, Assmann A, Speck O, Heinze HJ, Acosta-Cabronero J, Duzel E, Schreiber S. Detection of Cerebral Microbleeds With Venous Connection at 7-Tesla MRI. Neurology 2021; 96:e2048-e2057. [PMID: 33653897 DOI: 10.1212/wnl.0000000000011790] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 01/28/2021] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVE Cerebral microbleeds (MBs) are a common finding in patients with cerebral small vessel disease (CSVD) and Alzheimer disease as well as in healthy elderly people, but their pathophysiology remains unclear. To investigate a possible role of veins in the development of MBs, we performed an exploratory study, assessing in vivo presence of MBs with a direct connection to a vein. METHODS 7-Tesla (7T) MRI was conducted and MBs were counted on quantitative susceptibility mapping (QSM). A submillimeter resolution QSM-based venogram allowed identification of MBs with a direct spatial connection to a vein. RESULTS A total of 51 people (mean age [SD] 70.5 [8.6] years, 37% female) participated in the study: 20 had CSVD (cerebral amyloid angiopathy [CAA] with strictly lobar MBs [n = 8], hypertensive arteriopathy [HA] with strictly deep MBs [n = 5], or mixed lobar and deep MBs [n = 7], 72.4 [6.1] years, 30% female) and 31 were healthy controls (69.4 [9.9] years, 42% female). In our cohort, we counted a total of 96 MBs with a venous connection, representing 14% of all detected MBs on 7T QSM. Most venous MBs (86%, n = 83) were observed in lobar locations and all of these were cortical. Patients with CAA showed the highest ratio of venous to total MBs (19%) (HA = 9%, mixed = 18%, controls = 5%). CONCLUSION Our findings establish a link between cerebral MBs and the venous vasculature, pointing towards a possible contribution of veins to CSVD in general and to CAA in particular. Pathologic studies are needed to confirm our observations.
Collapse
Affiliation(s)
- Johanna Rotta
- From the Department of Neurology (J.R., V.P., F.S., A.A., H.-J.H., S.S.) and Institute of Physics (O.S.), Otto-von-Guericke University; Institute of Cognitive Neurology and Dementia Research (IKND) (V.P., R.Y., J.O., H.-J.H., E.D.), Magdeburg, Germany; J. Philip Kistler Stroke Research Center (V.P.), Massachusetts General Hospital, Boston; German Center for Neurodegenerative Diseases (DZNE) (R.Y., F.S., L.D., O.S., H.-J.H., E.D., S.S.), Magdeburg, Germany; Image Sciences Institute (H.J.K.), University Medical Center Utrecht, the Netherlands; Leibniz-Institute for Neurobiology (LIN) (O.S., H.-J.H., E.D.); Center for Behavioral Brain Sciences (CBBS) (O.S., H.-J.H., E.D., S.S.), Magdeburg, Germany; Tenoke Limited (J.A.-C.), Cambridge, UK; and Institute of Cognitive Neuroscience (E.D.), University College London, UK
| | - Valentina Perosa
- From the Department of Neurology (J.R., V.P., F.S., A.A., H.-J.H., S.S.) and Institute of Physics (O.S.), Otto-von-Guericke University; Institute of Cognitive Neurology and Dementia Research (IKND) (V.P., R.Y., J.O., H.-J.H., E.D.), Magdeburg, Germany; J. Philip Kistler Stroke Research Center (V.P.), Massachusetts General Hospital, Boston; German Center for Neurodegenerative Diseases (DZNE) (R.Y., F.S., L.D., O.S., H.-J.H., E.D., S.S.), Magdeburg, Germany; Image Sciences Institute (H.J.K.), University Medical Center Utrecht, the Netherlands; Leibniz-Institute for Neurobiology (LIN) (O.S., H.-J.H., E.D.); Center for Behavioral Brain Sciences (CBBS) (O.S., H.-J.H., E.D., S.S.), Magdeburg, Germany; Tenoke Limited (J.A.-C.), Cambridge, UK; and Institute of Cognitive Neuroscience (E.D.), University College London, UK.
| | - Renat Yakupov
- From the Department of Neurology (J.R., V.P., F.S., A.A., H.-J.H., S.S.) and Institute of Physics (O.S.), Otto-von-Guericke University; Institute of Cognitive Neurology and Dementia Research (IKND) (V.P., R.Y., J.O., H.-J.H., E.D.), Magdeburg, Germany; J. Philip Kistler Stroke Research Center (V.P.), Massachusetts General Hospital, Boston; German Center for Neurodegenerative Diseases (DZNE) (R.Y., F.S., L.D., O.S., H.-J.H., E.D., S.S.), Magdeburg, Germany; Image Sciences Institute (H.J.K.), University Medical Center Utrecht, the Netherlands; Leibniz-Institute for Neurobiology (LIN) (O.S., H.-J.H., E.D.); Center for Behavioral Brain Sciences (CBBS) (O.S., H.-J.H., E.D., S.S.), Magdeburg, Germany; Tenoke Limited (J.A.-C.), Cambridge, UK; and Institute of Cognitive Neuroscience (E.D.), University College London, UK
| | - Hugo J Kuijf
- From the Department of Neurology (J.R., V.P., F.S., A.A., H.-J.H., S.S.) and Institute of Physics (O.S.), Otto-von-Guericke University; Institute of Cognitive Neurology and Dementia Research (IKND) (V.P., R.Y., J.O., H.-J.H., E.D.), Magdeburg, Germany; J. Philip Kistler Stroke Research Center (V.P.), Massachusetts General Hospital, Boston; German Center for Neurodegenerative Diseases (DZNE) (R.Y., F.S., L.D., O.S., H.-J.H., E.D., S.S.), Magdeburg, Germany; Image Sciences Institute (H.J.K.), University Medical Center Utrecht, the Netherlands; Leibniz-Institute for Neurobiology (LIN) (O.S., H.-J.H., E.D.); Center for Behavioral Brain Sciences (CBBS) (O.S., H.-J.H., E.D., S.S.), Magdeburg, Germany; Tenoke Limited (J.A.-C.), Cambridge, UK; and Institute of Cognitive Neuroscience (E.D.), University College London, UK
| | - Frank Schreiber
- From the Department of Neurology (J.R., V.P., F.S., A.A., H.-J.H., S.S.) and Institute of Physics (O.S.), Otto-von-Guericke University; Institute of Cognitive Neurology and Dementia Research (IKND) (V.P., R.Y., J.O., H.-J.H., E.D.), Magdeburg, Germany; J. Philip Kistler Stroke Research Center (V.P.), Massachusetts General Hospital, Boston; German Center for Neurodegenerative Diseases (DZNE) (R.Y., F.S., L.D., O.S., H.-J.H., E.D., S.S.), Magdeburg, Germany; Image Sciences Institute (H.J.K.), University Medical Center Utrecht, the Netherlands; Leibniz-Institute for Neurobiology (LIN) (O.S., H.-J.H., E.D.); Center for Behavioral Brain Sciences (CBBS) (O.S., H.-J.H., E.D., S.S.), Magdeburg, Germany; Tenoke Limited (J.A.-C.), Cambridge, UK; and Institute of Cognitive Neuroscience (E.D.), University College London, UK
| | - Laura Dobisch
- From the Department of Neurology (J.R., V.P., F.S., A.A., H.-J.H., S.S.) and Institute of Physics (O.S.), Otto-von-Guericke University; Institute of Cognitive Neurology and Dementia Research (IKND) (V.P., R.Y., J.O., H.-J.H., E.D.), Magdeburg, Germany; J. Philip Kistler Stroke Research Center (V.P.), Massachusetts General Hospital, Boston; German Center for Neurodegenerative Diseases (DZNE) (R.Y., F.S., L.D., O.S., H.-J.H., E.D., S.S.), Magdeburg, Germany; Image Sciences Institute (H.J.K.), University Medical Center Utrecht, the Netherlands; Leibniz-Institute for Neurobiology (LIN) (O.S., H.-J.H., E.D.); Center for Behavioral Brain Sciences (CBBS) (O.S., H.-J.H., E.D., S.S.), Magdeburg, Germany; Tenoke Limited (J.A.-C.), Cambridge, UK; and Institute of Cognitive Neuroscience (E.D.), University College London, UK
| | - Jan Oltmer
- From the Department of Neurology (J.R., V.P., F.S., A.A., H.-J.H., S.S.) and Institute of Physics (O.S.), Otto-von-Guericke University; Institute of Cognitive Neurology and Dementia Research (IKND) (V.P., R.Y., J.O., H.-J.H., E.D.), Magdeburg, Germany; J. Philip Kistler Stroke Research Center (V.P.), Massachusetts General Hospital, Boston; German Center for Neurodegenerative Diseases (DZNE) (R.Y., F.S., L.D., O.S., H.-J.H., E.D., S.S.), Magdeburg, Germany; Image Sciences Institute (H.J.K.), University Medical Center Utrecht, the Netherlands; Leibniz-Institute for Neurobiology (LIN) (O.S., H.-J.H., E.D.); Center for Behavioral Brain Sciences (CBBS) (O.S., H.-J.H., E.D., S.S.), Magdeburg, Germany; Tenoke Limited (J.A.-C.), Cambridge, UK; and Institute of Cognitive Neuroscience (E.D.), University College London, UK
| | - Anne Assmann
- From the Department of Neurology (J.R., V.P., F.S., A.A., H.-J.H., S.S.) and Institute of Physics (O.S.), Otto-von-Guericke University; Institute of Cognitive Neurology and Dementia Research (IKND) (V.P., R.Y., J.O., H.-J.H., E.D.), Magdeburg, Germany; J. Philip Kistler Stroke Research Center (V.P.), Massachusetts General Hospital, Boston; German Center for Neurodegenerative Diseases (DZNE) (R.Y., F.S., L.D., O.S., H.-J.H., E.D., S.S.), Magdeburg, Germany; Image Sciences Institute (H.J.K.), University Medical Center Utrecht, the Netherlands; Leibniz-Institute for Neurobiology (LIN) (O.S., H.-J.H., E.D.); Center for Behavioral Brain Sciences (CBBS) (O.S., H.-J.H., E.D., S.S.), Magdeburg, Germany; Tenoke Limited (J.A.-C.), Cambridge, UK; and Institute of Cognitive Neuroscience (E.D.), University College London, UK
| | - Oliver Speck
- From the Department of Neurology (J.R., V.P., F.S., A.A., H.-J.H., S.S.) and Institute of Physics (O.S.), Otto-von-Guericke University; Institute of Cognitive Neurology and Dementia Research (IKND) (V.P., R.Y., J.O., H.-J.H., E.D.), Magdeburg, Germany; J. Philip Kistler Stroke Research Center (V.P.), Massachusetts General Hospital, Boston; German Center for Neurodegenerative Diseases (DZNE) (R.Y., F.S., L.D., O.S., H.-J.H., E.D., S.S.), Magdeburg, Germany; Image Sciences Institute (H.J.K.), University Medical Center Utrecht, the Netherlands; Leibniz-Institute for Neurobiology (LIN) (O.S., H.-J.H., E.D.); Center for Behavioral Brain Sciences (CBBS) (O.S., H.-J.H., E.D., S.S.), Magdeburg, Germany; Tenoke Limited (J.A.-C.), Cambridge, UK; and Institute of Cognitive Neuroscience (E.D.), University College London, UK
| | - Hans-Jochen Heinze
- From the Department of Neurology (J.R., V.P., F.S., A.A., H.-J.H., S.S.) and Institute of Physics (O.S.), Otto-von-Guericke University; Institute of Cognitive Neurology and Dementia Research (IKND) (V.P., R.Y., J.O., H.-J.H., E.D.), Magdeburg, Germany; J. Philip Kistler Stroke Research Center (V.P.), Massachusetts General Hospital, Boston; German Center for Neurodegenerative Diseases (DZNE) (R.Y., F.S., L.D., O.S., H.-J.H., E.D., S.S.), Magdeburg, Germany; Image Sciences Institute (H.J.K.), University Medical Center Utrecht, the Netherlands; Leibniz-Institute for Neurobiology (LIN) (O.S., H.-J.H., E.D.); Center for Behavioral Brain Sciences (CBBS) (O.S., H.-J.H., E.D., S.S.), Magdeburg, Germany; Tenoke Limited (J.A.-C.), Cambridge, UK; and Institute of Cognitive Neuroscience (E.D.), University College London, UK
| | - Julio Acosta-Cabronero
- From the Department of Neurology (J.R., V.P., F.S., A.A., H.-J.H., S.S.) and Institute of Physics (O.S.), Otto-von-Guericke University; Institute of Cognitive Neurology and Dementia Research (IKND) (V.P., R.Y., J.O., H.-J.H., E.D.), Magdeburg, Germany; J. Philip Kistler Stroke Research Center (V.P.), Massachusetts General Hospital, Boston; German Center for Neurodegenerative Diseases (DZNE) (R.Y., F.S., L.D., O.S., H.-J.H., E.D., S.S.), Magdeburg, Germany; Image Sciences Institute (H.J.K.), University Medical Center Utrecht, the Netherlands; Leibniz-Institute for Neurobiology (LIN) (O.S., H.-J.H., E.D.); Center for Behavioral Brain Sciences (CBBS) (O.S., H.-J.H., E.D., S.S.), Magdeburg, Germany; Tenoke Limited (J.A.-C.), Cambridge, UK; and Institute of Cognitive Neuroscience (E.D.), University College London, UK
| | - Emrah Duzel
- From the Department of Neurology (J.R., V.P., F.S., A.A., H.-J.H., S.S.) and Institute of Physics (O.S.), Otto-von-Guericke University; Institute of Cognitive Neurology and Dementia Research (IKND) (V.P., R.Y., J.O., H.-J.H., E.D.), Magdeburg, Germany; J. Philip Kistler Stroke Research Center (V.P.), Massachusetts General Hospital, Boston; German Center for Neurodegenerative Diseases (DZNE) (R.Y., F.S., L.D., O.S., H.-J.H., E.D., S.S.), Magdeburg, Germany; Image Sciences Institute (H.J.K.), University Medical Center Utrecht, the Netherlands; Leibniz-Institute for Neurobiology (LIN) (O.S., H.-J.H., E.D.); Center for Behavioral Brain Sciences (CBBS) (O.S., H.-J.H., E.D., S.S.), Magdeburg, Germany; Tenoke Limited (J.A.-C.), Cambridge, UK; and Institute of Cognitive Neuroscience (E.D.), University College London, UK
| | - Stefanie Schreiber
- From the Department of Neurology (J.R., V.P., F.S., A.A., H.-J.H., S.S.) and Institute of Physics (O.S.), Otto-von-Guericke University; Institute of Cognitive Neurology and Dementia Research (IKND) (V.P., R.Y., J.O., H.-J.H., E.D.), Magdeburg, Germany; J. Philip Kistler Stroke Research Center (V.P.), Massachusetts General Hospital, Boston; German Center for Neurodegenerative Diseases (DZNE) (R.Y., F.S., L.D., O.S., H.-J.H., E.D., S.S.), Magdeburg, Germany; Image Sciences Institute (H.J.K.), University Medical Center Utrecht, the Netherlands; Leibniz-Institute for Neurobiology (LIN) (O.S., H.-J.H., E.D.); Center for Behavioral Brain Sciences (CBBS) (O.S., H.-J.H., E.D., S.S.), Magdeburg, Germany; Tenoke Limited (J.A.-C.), Cambridge, UK; and Institute of Cognitive Neuroscience (E.D.), University College London, UK
| |
Collapse
|
16
|
Jura B, Młoźniak D, Goszczyńska H, Blinowska K, Biendon N, Macrez N, Meyrand P, Bem T. Reconfiguration of the cortical-hippocampal interaction may compensate for Sharp-Wave Ripple deficits in APP/PS1 mice and support spatial memory formation. PLoS One 2020; 15:e0243767. [PMID: 33382724 PMCID: PMC7774978 DOI: 10.1371/journal.pone.0243767] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 11/25/2020] [Indexed: 12/28/2022] Open
Abstract
Hippocampal-cortical dialogue, during which hippocampal ripple oscillations support information transfer, is necessary for long-term consolidation of spatial memories. Whereas a vast amount of work has been carried out to understand the cellular and molecular mechanisms involved in the impairments of memory formation in Alzheimer's disease (AD), far less work has been accomplished to understand these memory deficiencies at the network-level interaction that may underlie memory processing. We recently demonstrated that freely moving 8 to 9-month-old APP/PS1 mice, a model of AD, are able to learn a spatial reference memory task despite a major deficit in Sharp-Wave Ripples (SWRs), the integrity of which is considered to be crucial for spatial memory formation. In order to test whether reconfiguration of hippocampal-cortical dialogue could be responsible for the maintenance of this ability for memory formation, we undertook a study to identify causal relations between hippocampal and cortical circuits in epochs when SWRs are generated in hippocampus. We analyzed the data set obtained from multielectrode intracranial recording of transgenic and wild-type mice undergoing consolidation of spatial memory reported in our previous study. We applied Directed Transfer Function, a connectivity measure based on Granger causality, in order to determine effective coupling between distributed circuits which express oscillatory activity in multiple frequency bands. Our results showed that hippocampal-cortical coupling in epochs containing SWRs was expressed in the two frequency ranges corresponding to ripple (130-180 Hz) and slow gamma (20-60 Hz) band. The general features of connectivity patterns were similar in the 8 to 9-month-old APP/PS1 and wild-type animals except that the coupling in the slow gamma range was stronger and spread to more cortical sites in APP/PS1 mice than in the wild-type group. During the occurrence of SWRs, the strength of effective coupling from the cortex to hippocampus (CA1) in the ripple band undergoes sharp increase, involving cortical areas that were different in the two groups of animals. In the wild-type group, retrosplenial cortex and posterior cingulate cortex interacted with the hippocampus most strongly, whereas in the APP/PS1 group more anterior structures interacted with the hippocampus, that is, anterior cingulate cortex and prefrontal cortex. This reconfiguration of cortical-hippocampal interaction pattern may be an adaptive mechanism responsible for supporting spatial memory consolidation in AD mice model.
Collapse
Affiliation(s)
- Bartosz Jura
- Nałęcz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | - Dariusz Młoźniak
- Nałęcz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | - Hanna Goszczyńska
- Nałęcz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
| | - Katarzyna Blinowska
- Nałęcz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
- Department of Biomedical Physics, Faculty of Physics, University of Warsaw, Warsaw, Poland
| | - Nathalie Biendon
- Institut des Maladies Neurodégénératives, Université de Bordeaux, UMR 5293, Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | - Nathalie Macrez
- Institut des Maladies Neurodégénératives, Université de Bordeaux, UMR 5293, Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | - Pierre Meyrand
- Institut des Maladies Neurodégénératives, Université de Bordeaux, UMR 5293, Bordeaux, France
- CNRS, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
- INSERM, Neurocentre Magendie, Bordeaux, France
| | - Tiaza Bem
- Nałęcz Institute of Biocybernetics and Biomedical Engineering, Polish Academy of Sciences, Warsaw, Poland
- * E-mail:
| |
Collapse
|
17
|
Agca C, Klakotskaia D, Stopa EG, Schachtman TR, Agca Y. Ovariectomy Influences Cognition and Markers of Alzheimer's Disease. J Alzheimers Dis 2020; 73:529-541. [PMID: 31796679 DOI: 10.3233/jad-190935] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is one of the most devastating and costly diseases, and prevalence of AD increases with age. Furthermore, females are twice as likely to suffer from AD compared to males. The cessation of reproductive steroid hormone production during menopause is hypothesized to cause this difference. Two rodent AD models, APP21 and APP+PS1, and wild type (WT) rats underwent an ovariectomy or sham surgery. Changes in learning and memory, brain histology, amyloid-β (Aβ) deposition, levels of mRNAs involved in Aβ production and clearance, and synaptic and cognitive function were determined. Barnes maze results showed that regardless of ovariectomy status, APP+PS1 rats learned slower and had poor memory retention. Ovariectomy caused learning impairment only in the APP21 rats. High levels of Aβ42 and very low levels of Aβ40 were observed in the brain cortices of APP+PS1 rats indicating limited endogenous PS1. The APP+PS1 rats had 43-fold greater formic acid soluble Aβ42 than Aβ40 at 17 months. Furthermore, levels of formic acid soluble Aβ42 increased 57-fold in ovariectomized APP+PS1 rats between 12 and 17 months of age. The mRNA encoding Grin1 significantly decreased due to ovariectomy whereas levels of Bace1, Chat, and Prkcb all decreased with age. The expression levels of mRNAs involved in Aβ degradation and AβPP cleavage (Neprilysin, Ide, Adam9, and Psenen) were found to be highly correlated with each other as well as hippocampal Aβ deposition. Taken together, these results indicate that both ovariectomy and genotype influence AD markers in a complex manner.
Collapse
Affiliation(s)
- Cansu Agca
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| | - Diana Klakotskaia
- Department of Psychological Sciences, University of Missouri, Columbia, MO, USA
| | - Edward G Stopa
- Departments of Pathology and Neurosurgery, Brown University, Rhode Island Hospital, Providence, RI, USA
| | - Todd R Schachtman
- Department of Psychological Sciences, University of Missouri, Columbia, MO, USA
| | - Yuksel Agca
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, USA
| |
Collapse
|
18
|
Poon CH, Wang Y, Fung ML, Zhang C, Lim LW. Rodent Models of Amyloid-Beta Feature of Alzheimer's Disease: Development and Potential Treatment Implications. Aging Dis 2020; 11:1235-1259. [PMID: 33014535 PMCID: PMC7505263 DOI: 10.14336/ad.2019.1026] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 10/26/2019] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder worldwide and causes severe financial and social burdens. Despite much research on the pathogenesis of AD, the neuropathological mechanisms remain obscure and current treatments have proven ineffective. In the past decades, transgenic rodent models have been used to try to unravel this disease, which is crucial for early diagnosis and the assessment of disease-modifying compounds. In this review, we focus on transgenic rodent models used to study amyloid-beta pathology in AD. We also discuss their possible use as promising tools for AD research. There is still no effective treatment for AD and the development of potent therapeutics are urgently needed. Many molecular pathways are susceptible to AD, ranging from neuroinflammation, immune response, and neuroplasticity to neurotrophic factors. Studying these pathways may shed light on AD pathophysiology as well as provide potential targets for the development of more effective treatments. This review discusses the advantages and limitations of these models and their potential therapeutic implications for AD.
Collapse
Affiliation(s)
- Chi Him Poon
- 1School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yingyi Wang
- 1School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Man-Lung Fung
- 1School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Chengfei Zhang
- 2Endodontology, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
| | - Lee Wei Lim
- 1School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
19
|
Potential Role of Venular Amyloid in Alzheimer's Disease Pathogenesis. Int J Mol Sci 2020; 21:ijms21061985. [PMID: 32183293 PMCID: PMC7139584 DOI: 10.3390/ijms21061985] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 03/07/2020] [Accepted: 03/12/2020] [Indexed: 02/06/2023] Open
Abstract
Insurmountable evidence has demonstrated a strong association between Alzheimer's disease (AD) and cerebral amyloid angiopathy (CAA), along with various other cerebrovascular diseases. One form of CAA, which is the accumulation of amyloid-beta peptides (Aβ) along cerebral vessel walls, impairs perivascular drainage pathways and contributes to cerebrovascular dysfunction in AD. To date, CAA research has been primarily focused on arterial Aβ, while the accumulation of Aβ in veins and venules were to a lesser extent. In this review, we describe preclinical models and clinical studies supporting the presence of venular amyloid and potential downstream pathological mechanisms that affect the cerebrovasculature in AD. Venous collagenosis, impaired cerebrovascular pulsatility, and enlarged perivascular spaces are exacerbated by venular amyloid and increase Aβ deposition, potentially through impaired perivascular clearance. Gaining a comprehensive understanding of the mechanisms involved in venular Aβ deposition and associated pathologies will give insight to how CAA contributes to AD and its association with AD-related cerebrovascular disease. Lastly, we suggest that special consideration should be made to develop Aβ-targeted therapeutics that remove vascular amyloid and address cerebrovascular dysfunction in AD.
Collapse
|
20
|
Ivanova N, Liu Q, Agca C, Agca Y, Noble EG, Whitehead SN, Cechetto DF. White matter inflammation and cognitive function in a co-morbid metabolic syndrome and prodromal Alzheimer's disease rat model. J Neuroinflammation 2020; 17:29. [PMID: 31964387 PMCID: PMC6975033 DOI: 10.1186/s12974-020-1698-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 01/02/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Metabolic syndrome, the development of which is associated with high-caloric Western diet (HCD) intake, represent a risk factor for mild cognitive impairment (MCI) and dementia including Alzheimer's disease (AD) later in life. This study aimed to investigate the effect of diet-induced metabolic disturbances on white matter neuroinflammation and cognitive function in a transgenic (TG) Fischer 344 rat carrying a human β-amyloid precursor protein (APP) gene with Swedish and Indiana mutations (APP21 TG), a model of pre-AD and MCI. METHODS TG and wildtype (WT) rats received either a HCD with 40% kJ from fat supplemented with 20% corn syrup drink or a standard diet for 12 weeks. Body weight, caloric intake, and blood pressure were measured repeatedly. End-point changes in glucose and lipid metabolism were also assessed. Open field task was used for assessment of activity; Morris water maze was used to assess spatial learning and memory. Cerebral white matter microglia and astrocytes, hippocampal neurons, and neuronal synapses were examined using immunohistochemistry. RESULTS Rats maintained on the HCD developed significant obesity, visceral adiposity, dyslipidemia, and hyperinsulinemia, but did not become hypertensive. Impaired glucose tolerance was observed only in WT rats on the HCD. Total microglia number, activated OX-6+ microglia, as well as GFAP+ astrocytes located predominantly in the white matter were greater in the APP21 TG rat model in comparison to WT rats. HCD-driven metabolic perturbations further exacerbated white matter microgliosis and microglia cell activation in the APP21 TG rats and led to detectable changes in spatial reference memory in the comorbid prodromal AD and metabolic syndrome group compared to WT control rats. Neuronal density in the CA1 subregion of the hippocampus was not different between the experimental groups. Synaptic density in the CA1 and CA3 hippocampal subregions was lower in the TG rats compared to WT rats; however, there was no additional effect of the co-morbidity on this measure. CONCLUSIONS These results suggest that white matter neuroinflammation might be one of the possible processes of early interaction of metabolic syndrome with MCI and pre-AD and could be one of the early brain pathologies contributing to cognitive deficits observed in mild cognitive impairment and dementia, including AD cases.
Collapse
Affiliation(s)
- Nadezda Ivanova
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, ON, N6A 5C1, Canada.
| | - Qingfan Liu
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, ON, N6A 5C1, Canada
| | - Cansu Agca
- Department of Veterinary Pathobiology, University of Missouri College of Veterinary Medicine, Columbia, MO, USA
| | - Yuksel Agca
- Department of Veterinary Pathobiology, University of Missouri College of Veterinary Medicine, Columbia, MO, USA
| | - Earl G Noble
- School of Kinesiology, Western University, London, ON, Canada
| | - Shawn Narain Whitehead
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, ON, N6A 5C1, Canada
| | - David Floyd Cechetto
- Department of Anatomy & Cell Biology, Schulich School of Medicine & Dentistry, Western University, London, ON, N6A 5C1, Canada
| |
Collapse
|
21
|
Denver P, D’Adamo H, Hu S, Zuo X, Zhu C, Okuma C, Kim P, Castro D, Jones MR, Leal C, Mekkittikul M, Ghadishah E, Teter B, Vinters HV, Cole GM, Frautschy SA. A Novel Model of Mixed Vascular Dementia Incorporating Hypertension in a Rat Model of Alzheimer's Disease. Front Physiol 2019; 10:1269. [PMID: 31708792 PMCID: PMC6821690 DOI: 10.3389/fphys.2019.01269] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 09/19/2019] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) and mixed dementia (MxD) comprise the majority of dementia cases in the growing global aging population. MxD describes the coexistence of AD pathology with vascular pathology, including cerebral small vessel disease (SVD). Cardiovascular disease increases risk for AD and MxD, but mechanistic synergisms between the coexisting pathologies affecting dementia risk, progression and the ultimate clinical manifestations remain elusive. To explore the additive or synergistic interactions between AD and chronic hypertension, we developed a rat model of MxD, produced by breeding APPswe/PS1ΔE9 transgenes into the stroke-prone spontaneously hypertensive rat (SHRSP) background, resulting in the SHRSP/FAD model and three control groups (FAD, SHRSP and non-hypertensive WKY rats, n = 8-11, both sexes, 16-18 months of age). After behavioral testing, rats were euthanized, and tissue assessed for vascular, neuroinflammatory and AD pathology. Hypertension was preserved in the SHRSP/FAD cross. Results showed that SHRSP increased FAD-dependent neuroinflammation (microglia and astrocytes) and tau pathology, but plaque pathology changes were subtle, including fewer plaques with compact cores and slightly reduced plaque burden. Evidence for vascular pathology included a change in the distribution of astrocytic end-foot protein aquaporin-4, normally distributed in microvessels, but in SHRSP/FAD rats largely dissociated from vessels, appearing disorganized or redistributed into neuropil. Other evidence of SVD-like pathology included increased collagen IV staining in cerebral vessels and PECAM1 levels. We identified a plasma biomarker in SHRSP/FAD rats that was the only group to show increased Aqp-4 in plasma exosomes. Evidence of neuron damage in SHRSP/FAD rats included increased caspase-cleaved actin, loss of myelin and reduced calbindin staining in neurons. Further, there were mitochondrial deficits specific to SHRSP/FAD, notably the loss of complex II, accompanying FAD-dependent loss of mitochondrial complex I. Cognitive deficits exhibited by FAD rats were not exacerbated by the introduction of the SHRSP phenotype, nor was the hyperactivity phenotype associated with SHRSP altered by the FAD transgene. This novel rat model of MxD, encompassing an amyloidogenic transgene with a hypertensive phenotype, exhibits several features associated with human vascular or "mixed" dementia and may be a useful tool in delineating the pathophysiology of MxD and development of therapeutics.
Collapse
Affiliation(s)
- Paul Denver
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Heather D’Adamo
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Shuxin Hu
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Geriatric Research Education and Clinical Center, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, United States
| | - Xiaohong Zuo
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Geriatric Research Education and Clinical Center, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, United States
| | - Cansheng Zhu
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Chihiro Okuma
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Peter Kim
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Geriatric Research Education and Clinical Center, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, United States
| | - Daniel Castro
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Mychica R. Jones
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Carmen Leal
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Geriatric Research Education and Clinical Center, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, United States
| | - Marisa Mekkittikul
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Geriatric Research Education and Clinical Center, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, United States
| | - Elham Ghadishah
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Bruce Teter
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Geriatric Research Education and Clinical Center, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, United States
| | - Harry V. Vinters
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
| | - Gregory Michael Cole
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Geriatric Research Education and Clinical Center, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, United States
| | - Sally A. Frautschy
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA, United States
- Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
- Geriatric Research Education and Clinical Center, Greater Los Angeles Veterans Affairs Healthcare System, Los Angeles, CA, United States
| |
Collapse
|
22
|
Mullane K, Williams M. Preclinical Models of Alzheimer's Disease: Relevance and Translational Validity. ACTA ACUST UNITED AC 2019; 84:e57. [PMID: 30802363 DOI: 10.1002/cpph.57] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The only drugs currently approved for the treatment of Alzheimer's Disease (AD) are four acetylcholinesterase inhibitors and the NMDA antagonist memantine. Apart from these drugs, which have minimal to no clinical benefit, the 40-year search for effective therapeutics to treat AD has resulted in a clinical failure rate of 100% not only for compounds that prevent brain amyloid deposition or remove existing amyloid plaques but also those acting by a variety of other putative disease-associated mechanisms. This indicates that the preclinical data generated from current AD targets to support the selection, optimization, and translation of new chemical entities (NCEs) and biologics to clinical trials is seriously compromised. While many of these failures reflect flawed hypotheses or a lack of adequate characterization of the preclinical pharmacodynamic and pharmacokinetic (PD/PK) properties of lead NCEs-including their bioavailability and toxicity-the conceptualization, validation, and interrogation of the current animal models of AD represent key limitations. The overwhelming majority of these AD models are transgenic, based on aspects of the amyloid hypothesis and the genetics of the familial form of the disease. As a result, these generally lack construct and predictive validity for the sporadic form of the human disease. The 170 or so transgenic models, perhaps the largest number ever focused on a single disease, use rodents, mainly mice, and in addition to amyloid also address aspects of tau causality with more complex multigene models including other presumed causative factors together with amyloid. This overview discusses the current animal models of AD in the context of both the controversies surrounding the causative role of amyloid in the disease and the need to develop validated models of cognitive function/dysfunction that more appropriately reflect the phenotype(s) of human aged-related dementias. © 2019 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
| | - Michael Williams
- Department of Biological Chemistry and Pharmacology, College of Medicine, Ohio State University, Columbus, Ohio
| |
Collapse
|