1
|
Gu X, Fan M, Zhou Y, Zhang Y, Wang L, Gao W, Li T, Wang H, Si N, Wei X, Bian B, Zhao H. Intestinal endogenous metabolites affect neuroinflammation in 5×FAD mice by mediating "gut-brain" axis and the intervention with Chinese Medicine. Alzheimers Res Ther 2024; 16:222. [PMID: 39396997 PMCID: PMC11472645 DOI: 10.1186/s13195-024-01587-5] [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: 06/11/2023] [Accepted: 09/29/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Emerging evidence suggested the association between gut dysbiosis and Alzheimer's disease (AD) progression. However, it remained unclear how the gut microbiome and neuroinflammation in the brain mutually interact or how these interactions affect brain functioning and cognition. Here we hypothesized that "gut-brain" axis mediated by microbial derived metabolites was expected to novel breakthroughs in the fields of AD research and development. METHODS Multiple technologies, such as immunofluorescence, 16s rDNA sequencing, mass spectrometry-based metabolomics (LC-QQQ-MS and GC-MS), were used to reveal potential link between gut microbiota and the metabolism and cognition of the host. RESULTS Microbial depletion induced by the antibiotics mix (ABX) verified that "gut-brain" can transmit information bidirectionally. Short-chain fatty acid-producing (SCFAs-producing) bacteria and amino acid-producing bacteria fluctuated greatly in 5×FAD mice, especially the reduction sharply of the Bifidobacteriaceae and the increase of the Lachnospiraceae family. Concentrations of several Tryptophan-kynurenine intermediates, lactic acid, CD4+ cell, and CD8+ cells were higher in serum of 5×FAD mice, whilst TCA cycle intermediates and Th1/Th2 were lower. In addition, the levels of iso-butyric acid (IBA) in feces, serum, and brain of 5×FAD mice were increased compared with WT-M mice, especially in serum. And IBA in the brain was positively correlated with Aβ and proinflammatory factors. CONCLUSION Together, our finding highlighted that the alternation in gut microbiota affected the effective communication between the "gut-brain" axis in 5×FAD mice by regulating the immune system, carbohydrate, and energy metabolism.
Collapse
Affiliation(s)
- Xinru Gu
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
- The Neurology Department, The First People's Hospital of Lianyungang, Lianyungang, 222000, China
| | - Miaoxuan Fan
- Beijing Drug Package Test Institute, Beijing, 100700, China
| | - Yanyan Zhou
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yan Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Linna Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Wenya Gao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Tao Li
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Hongjie Wang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Nan Si
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Xiaolu Wei
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Baolin Bian
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Haiyu Zhao
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| |
Collapse
|
2
|
Frost OG, Ramkilawan P, Rebbaa A, Stolzing A. A systematic review of lifespan studies in rodents using stem cell transplantations. Ageing Res Rev 2024; 97:102295. [PMID: 38588866 DOI: 10.1016/j.arr.2024.102295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 03/27/2024] [Accepted: 04/03/2024] [Indexed: 04/10/2024]
Abstract
Organismal aging involves the progressive decline in organ function and increased susceptibility to age-associated diseases. Regardless of its origin, cellular aging is consequently reflected at the level of organ and associated systems dysfunction. Aging of stem cell populations within the body and their decreased ability to self-renew, differentiate, and regenerate damaged tissues, is a key contributor to organismal decline. Based on this, supplementing young stem cells may delay tissue aging, improve frailty and extend health and lifespan. This review investigates studies in rodents using stem cell transplantation from either mice or human donors. The aim is to consolidate available information on the efficacy of stem cell therapies in rodent models and provide insights to guide further research efforts. Out of the 21 studies included in this review, the methodology varied significantly including the lifespan measurement. To enable comparison the median lifespan was calculated using WebPlotDigitizer 4.6 if not provided by the literature. A total of 18 out of 21 studies evidenced significant lifespan extension post stem cell transplant, with 7 studies demonstrating benefits in reduced frailty and other aging complications.
Collapse
Affiliation(s)
- Oliver G Frost
- Centre for Biological Engineering, School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough LE11 3TU, UK; SENS Research Foundation, Mountain View, CA 94041, USA
| | | | | | - Alexandra Stolzing
- Centre for Biological Engineering, School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Loughborough LE11 3TU, UK.
| |
Collapse
|
3
|
Li H, Zhao Z, Fassini A, Lee HK, Green RJ, Gomperts SN. Impaired Hippocampal Reactivation Preceding Robust Aβ Deposition in a Model of Alzheimer's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.26.595168. [PMID: 38853978 PMCID: PMC11160633 DOI: 10.1101/2024.05.26.595168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Current therapeutic strategies for Alzheimer's disease (AD) target amyloid-beta (Aβ) fibrils and high molecular weight protofibrils associated with plaques, but other bioactive species may directly contribute to neural systems failure in AD. Employing hippocampal electrophysiological recordings and dynamic calcium imaging across the sleep-wake cycle in young mice expressing human Aβ and Aβ oligomers, we reveal marked impairments of hippocampal function long before amyloid plaques predominate. In slow wave sleep (SWS), Aβ increased the proportion of hypoactive cells and reduced place-cell reactivation. During awake behavior, Aβ impaired theta-gamma phase-amplitude coupling (PAC) and drove excessive synchronization of place cell calcium fluctuations with hippocampal theta. Remarkably, the on-line impairment of hippocampal theta-gamma PAC correlated with the SWS impairment of place-cell reactivation. Together, these results identify toxic effects of Aβ on memory encoding and consolidation processes before robust plaque deposition and support targeting soluble Aβ-related species to treat and prevent AD.
Collapse
Affiliation(s)
- Hanyan Li
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Zhuoyang Zhao
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Aline Fassini
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Han K Lee
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Reese J Green
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Stephen N Gomperts
- Department of Neurology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
4
|
Daudelin D, Westerhaus A, Zhang N, Leyder E, Savonenko A, Sockanathan S. Loss of GDE2 leads to complex behavioral changes including memory impairment. BEHAVIORAL AND BRAIN FUNCTIONS : BBF 2024; 20:7. [PMID: 38575965 PMCID: PMC10993612 DOI: 10.1186/s12993-024-00234-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 03/26/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND Alzheimer's disease (AD) and amyotrophic lateral sclerosis/frontotemporal dementia (ALS/FTD) are debilitating neurodegenerative diseases for which there are currently no cures. Familial cases with known genetic causes make up less than 10% of these diseases, and little is known about the underlying mechanisms that contribute to sporadic disease. Accordingly, it is important to expand investigations into possible pathways that may contribute to disease pathophysiology. Glycerophosphodiester phosphodiesterase 2 (GDE2 or GDPD5) is a membrane-bound enzyme that acts at the cell surface to cleave the glycosylphosphatidylinositol (GPI)-anchor that tethers distinct proteins to the membrane. GDE2 abnormally accumulates in intracellular compartments in the brain of patients with AD, ALS, and ALS/FTD, indicative of GDE2 dysfunction. Mice lacking GDE2 (Gde2KO) show neurodegenerative changes such as neuronal loss, reduced synaptic proteins and synapse loss, and increased Aβ deposition, raising the possibility that GDE2 disruption in disease might contribute to disease pathophysiology. However, the effect of GDE2 loss on behavioral function and learning/memory has not been characterized. RESULTS Here, we show that GDE2 is expressed throughout the adult mouse brain in areas including the cortex, hippocampus, habenula, thalamus, and amygdala. Gde2KO and WT mice were tested in a set of behavioral tasks between 7 and 16 months of age. Compared to WT, Gde2KO mice display moderate hyperactivity that becomes more pronounced with age across a variety of behavioral tests assessing novelty-induced exploratory activity. Additionally, Gde2KO mice show reduced startle response, with females showing additional defects in prepulse inhibition. No changes in anxiety-associated behaviors were found, but Gde2KOs show reduced sociability. Notably, aged Gde2KO mice demonstrate impaired short/long-term spatial memory and cued fear memory/secondary contextual fear acquisition. CONCLUSIONS Taken together, these observations suggest that loss of GDE2 leads to behavioral deficits, some of which are seen in neurodegenerative disease models, implying that loss of GDE2 may be an important contributor to phenotypes associated with neurodegeneration.
Collapse
Affiliation(s)
- Daniel Daudelin
- The Solomon Snyder Department of Neuroscience, The Johns Hopkins School of Medicine, PCTB 1004, 725 N. Wolfe Street, Baltimore, MD, 21205, USA
| | - Anna Westerhaus
- The Solomon Snyder Department of Neuroscience, The Johns Hopkins School of Medicine, PCTB 1004, 725 N. Wolfe Street, Baltimore, MD, 21205, USA
| | - Nan Zhang
- The Solomon Snyder Department of Neuroscience, The Johns Hopkins School of Medicine, PCTB 1004, 725 N. Wolfe Street, Baltimore, MD, 21205, USA
| | - Erica Leyder
- Department of Pathology, The Johns Hopkins University School of Medicine, 558 Ross Research Building, 720 Rutland Avenue, Baltimore, MD, 21205, USA
- Molecular Microbiology and Immunology Graduate Program in Life Sciences, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD, 21201, USA
| | - Alena Savonenko
- Department of Pathology, The Johns Hopkins University School of Medicine, 558 Ross Research Building, 720 Rutland Avenue, Baltimore, MD, 21205, USA.
- Sensory-Motor Neuroscience (SMN), Center for Scientific Review, ICN Review Branch, National Institutes of Health, 6701 Rockledge Drive, Suite 1010-F, Bethesda, MD, 20892 , USA.
| | - Shanthini Sockanathan
- The Solomon Snyder Department of Neuroscience, The Johns Hopkins School of Medicine, PCTB 1004, 725 N. Wolfe Street, Baltimore, MD, 21205, USA.
| |
Collapse
|
5
|
Bartman S, Coppotelli G, Ross JM. Mitochondrial Dysfunction: A Key Player in Brain Aging and Diseases. Curr Issues Mol Biol 2024; 46:1987-2026. [PMID: 38534746 DOI: 10.3390/cimb46030130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/28/2024] Open
Abstract
Mitochondria are thought to have become incorporated within the eukaryotic cell approximately 2 billion years ago and play a role in a variety of cellular processes, such as energy production, calcium buffering and homeostasis, steroid synthesis, cell growth, and apoptosis, as well as inflammation and ROS production. Considering that mitochondria are involved in a multitude of cellular processes, mitochondrial dysfunction has been shown to play a role within several age-related diseases, including cancers, diabetes (type 2), and neurodegenerative diseases, although the underlying mechanisms are not entirely understood. The significant increase in lifespan and increased incidence of age-related diseases over recent decades has confirmed the necessity to understand the mechanisms by which mitochondrial dysfunction impacts the process of aging and age-related diseases. In this review, we will offer a brief overview of mitochondria, along with structure and function of this important organelle. We will then discuss the cause and consequence of mitochondrial dysfunction in the aging process, with a particular focus on its role in inflammation, cognitive decline, and neurodegenerative diseases, such as Huntington's disease, Parkinson's disease, and Alzheimer's disease. We will offer insight into therapies and interventions currently used to preserve or restore mitochondrial functioning during aging and neurodegeneration.
Collapse
Affiliation(s)
- Sydney Bartman
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI 02881, USA
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Giuseppe Coppotelli
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI 02881, USA
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| | - Jaime M Ross
- George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI 02881, USA
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI 02881, USA
| |
Collapse
|
6
|
Frolinger T, Korkmaz F, Sims S, Sen F, Sultana F, Laurencin V, Cullen L, Pallapati AR, Liu A, Rojekar S, Pevnev G, Cheliadinova U, Vasilyeva D, Burganova G, Macdonald A, Saxena M, Goosens K, Rosen C, Barak O, Lizneva D, Gumerova A, Ye K, Ryu V, Yuen T, Zaidi M. Gene-Dose-Dependent Reduction Fshr Expression Improves Spatial Memory Deficits in Alzheimer's Mice. RESEARCH SQUARE 2024:rs.3.rs-3964789. [PMID: 38463956 PMCID: PMC10925392 DOI: 10.21203/rs.3.rs-3964789/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Alzheimer's disease (AD) is a major progressive neurodegenerative disorder of the aging population. High post-menopausal levels of the pituitary gonadotropin follicle-stimulating hormone (FSH) are strongly associated with the onset of AD, and we have shown recently that FSH directly activates the hippocampal Fshr to drive AD-like pathology and memory loss in mice. To establish a role for FSH in memory loss, we used female 3xTg;Fshr+/+, 3xTg;Fshr+/- and 3xTg;Fshr-/- mice that were either left unoperated or underwent sham surgery or ovariectomy at 8 weeks of age. Unoperated and sham-operated 3xTg;Fshr-/- mice were implanted with 17β-estradiol pellets to normalize estradiol levels. Morris Water Maze and Novel Object Recognition behavioral tests were performed to study deficits in spatial and recognition memory, respectively, and to examine the effects of Fshr depletion. 3xTg;Fshr+/+ mice displayed impaired spatial memory at 5 months of age; both the acquisition and retrieval of the memory were ameliorated in 3xTg;Fshr-/- mice and, to a lesser extent, in 3xTg;Fshr+/- mice- -thus documenting a clear gene-dose-dependent prevention of hippocampal-dependent spatial memory impairment. At 5 and 10 months, sham-operated 3xTg;Fshr-/- mice showed better memory performance during the acquasition and/or retrieval phases, suggesting that Fshr deletion prevented the progression of spatial memory deficits with age. However, this prevention was not seen when mice were ovariectomized, except in the 10-month-old 3xTg;Fshr-/- mice. In the Novel Object Recognition test performed at 10 months, all groups of mice, except ovariectomized 3xTg;Fshr-/- mice showed a loss of recognition memory. Consistent with the neurobehavioral data, there was a gene-dose-dependent reduction mainly in the amyloid β40 isoform in whole brain extracts. Finally, serum FSH levels < 8 ng/mL in 16-month-old APP/PS1 mice were associated with better retrieval of spatial memory. Collectively, the data provide compelling genetic evidence for a protective effect of inhibiting FSH signaling on the progression of spatial and recognition memory deficits in mice, and lay a firm foundation for the use of an FSH-blocking agent for the early prevention of cognitive decline in postmenopausal women.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Avi Liu
- Icahn School of Medicine at Mount Sinai
| | | | | | | | | | | | | | | | | | | | | | | | | | - Keqiang Ye
- Shenzhen Institute of Advanced Technology
| | | | | | | |
Collapse
|
7
|
Zhang S, Ai H, Wang J, Liu T, Zheng X, Tian X, Bai W. Reduced Prefrontal-Thalamic Theta Flow During Working Memory Retrieval in APP/PS1 Mice. J Alzheimers Dis 2024; 97:1737-1749. [PMID: 38306044 PMCID: PMC10894573 DOI: 10.3233/jad-231078] [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] [Accepted: 12/05/2023] [Indexed: 02/03/2024]
Abstract
Background Working memory deficits in Alzheimer's disease (AD) are linked to impairments in the retrieval of stored memory information. However, research on the mechanism of impaired working memory retrieval in Alzheimer's disease is still lacking. Objective The medial prefrontal cortex (mPFC) and mediodorsal thalamus (MD) are involved in memory retrieval. The purpose of this study is to investigate the functional interactions and information transmission between mPFC and MD in the AD model. Methods We recorded local field potentials from mPFC and MD while the mice (APP/PS1 transgenic model and control) performed a T-maze spatial working memory task. The temporal dynamics of oscillatory activity and bidirectional information flow between mPFC and MD were assessed during the task phases. Results We mainly found a significant decrease in theta flow from mPFC to MD in APP/PS1 mice during retrieval. Conclusions Our results indicate an important role of the mPFC-MD input for retrieval and the disrupted information transfer from mPFC to MD may be the underlying mechanism of working memory deficits in APP/PS1 mice.
Collapse
Affiliation(s)
- Shengnan Zhang
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| | - Hongrui Ai
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| | - Jia Wang
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| | - Tiaotiao Liu
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| | - Xuyuan Zheng
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| | - Xin Tian
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| | - Wenwen Bai
- School of Biomedical Engineering and Technology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
8
|
Chilton PM, Ghare SS, Charpentier BT, Myers SA, Rao AV, Petrosino JF, Hoffman KL, Greenwell JC, Tyagi N, Behera J, Wang Y, Sloan LJ, Zhang J, Shields CB, Cooper GE, Gobejishvili L, Whittemore SR, McClain CJ, Barve SS. Age-associated temporal decline in butyrate-producing bacteria plays a key pathogenic role in the onset and progression of neuropathology and memory deficits in 3×Tg-AD mice. Gut Microbes 2024; 16:2389319. [PMID: 39182227 PMCID: PMC11346541 DOI: 10.1080/19490976.2024.2389319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/16/2024] [Accepted: 08/01/2024] [Indexed: 08/27/2024] Open
Abstract
Alterations in the gut-microbiome-brain axis are increasingly being recognized to be involved in Alzheimer's disease (AD) pathogenesis. However, the functional consequences of enteric dysbiosis linking gut microbiota and brain pathology in AD progression remain largely undetermined. The present work investigated the causal role of age-associated temporal decline in butyrate-producing bacteria and butyrate in the etiopathogenesis of AD. Longitudinal metagenomics, neuropathological, and memory analyses were performed in the 3×Tg-AD mouse model. Metataxonomic analyses showed a significant temporal decline in the alpha diversity marked by a decrease in butyrate-producing bacterial communities and a concurrent reduction in cecal butyrate production. Inferred metagenomics analysis identified the bacterial acetyl-CoA pathway as the main butyrate synthesis pathway impacted. Concomitantly, there was an age-associated decline in the transcriptionally permissive acetylation of histone 3 at lysines 9 and 14 (H3K9/K14-Ac) in hippocampal neurons. Importantly, these microbiome-gut-brain changes preceded AD-related neuropathology, including oxidative stress, tau hyperphosphorylation, memory deficits, and neuromuscular dysfunction, which manifest by 17-18 months. Initiation of oral administration of tributyrin, a butyrate prodrug, at 6 months of age mitigated the age-related decline in butyrate-producing bacteria, protected the H3K9/K14-Ac status, and attenuated the development of neuropathological and cognitive changes associated with AD pathogenesis. These data causally implicate age-associated decline in butyrate-producing bacteria as a key pathogenic feature of the microbiome-gut-brain axis affecting the onset and progression of AD. Importantly, the regulation of butyrate-producing bacteria and consequent butyrate synthesis could be a significant therapeutic strategy in the prevention and treatment of AD.
Collapse
Affiliation(s)
- Paula M. Chilton
- Department of Medicine, Division of Gastroenterology, Hepatology & Nutrition, University of Louisville School of Medicine, Louisville, KY, USA
- UofL Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY, USA
- Norton Neuroscience Institute, 4915 Norton Healthcare Blvd, Louisville, KY, USA
| | - Smita S. Ghare
- Department of Medicine, Division of Gastroenterology, Hepatology & Nutrition, University of Louisville School of Medicine, Louisville, KY, USA
- UofL Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY, USA
- Norton Neuroscience Institute, 4915 Norton Healthcare Blvd, Louisville, KY, USA
- UofL Hepatobiology COBRE, University of Louisville School of Medicine, Louisville, KY, USA
| | - Benjamin T. Charpentier
- Department of Medicine, Division of Gastroenterology, Hepatology & Nutrition, University of Louisville School of Medicine, Louisville, KY, USA
- UofL Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY, USA
- Department of Anatomical Sciences & Neurobiology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Scott A. Myers
- Department of Medicine, Division of Gastroenterology, Hepatology & Nutrition, University of Louisville School of Medicine, Louisville, KY, USA
- UofL Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY, USA
- Department of Anatomical Sciences & Neurobiology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Aakarsha V. Rao
- Department of Medicine, Division of Gastroenterology, Hepatology & Nutrition, University of Louisville School of Medicine, Louisville, KY, USA
- UofL Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY, USA
| | - Joseph F. Petrosino
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Kristi L. Hoffman
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - John C. Greenwell
- Department of Medicine, Division of Gastroenterology, Hepatology & Nutrition, University of Louisville School of Medicine, Louisville, KY, USA
- UofL Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY, USA
| | - Neetu Tyagi
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Jyotirmaya Behera
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Yali Wang
- Department of Medicine, Division of Gastroenterology, Hepatology & Nutrition, University of Louisville School of Medicine, Louisville, KY, USA
- UofL Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY, USA
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Lucy J. Sloan
- Department of Medicine, Division of Gastroenterology, Hepatology & Nutrition, University of Louisville School of Medicine, Louisville, KY, USA
- UofL Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY, USA
- Department of Pharmacology & Toxicology, University of Louisville School of Medicine, Louisville, KY, USA
| | - JingWen Zhang
- Department of Medicine, Division of Gastroenterology, Hepatology & Nutrition, University of Louisville School of Medicine, Louisville, KY, USA
- UofL Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY, USA
| | - Christopher B. Shields
- Norton Neuroscience Institute, 4915 Norton Healthcare Blvd, Louisville, KY, USA
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, USA
| | - Gregory E. Cooper
- Norton Neuroscience Institute, 4915 Norton Healthcare Blvd, Louisville, KY, USA
| | - Leila Gobejishvili
- Department of Medicine, Division of Gastroenterology, Hepatology & Nutrition, University of Louisville School of Medicine, Louisville, KY, USA
- UofL Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY, USA
- UofL Hepatobiology COBRE, University of Louisville School of Medicine, Louisville, KY, USA
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, USA
| | - Scott R. Whittemore
- Department of Anatomical Sciences & Neurobiology, University of Louisville School of Medicine, Louisville, KY, USA
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, USA
| | - Craig J. McClain
- Department of Medicine, Division of Gastroenterology, Hepatology & Nutrition, University of Louisville School of Medicine, Louisville, KY, USA
- UofL Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY, USA
- UofL Hepatobiology COBRE, University of Louisville School of Medicine, Louisville, KY, USA
- Department of Medicine, Robley Rex VA Medical Center, Louisville, KY, USA
| | - Shirish S. Barve
- Department of Medicine, Division of Gastroenterology, Hepatology & Nutrition, University of Louisville School of Medicine, Louisville, KY, USA
- UofL Alcohol Research Center, University of Louisville School of Medicine, Louisville, KY, USA
- Norton Neuroscience Institute, 4915 Norton Healthcare Blvd, Louisville, KY, USA
- UofL Hepatobiology COBRE, University of Louisville School of Medicine, Louisville, KY, USA
| |
Collapse
|
9
|
Marzi SJ, Schilder BM, Nott A, Frigerio CS, Willaime-Morawek S, Bucholc M, Hanger DP, James C, Lewis PA, Lourida I, Noble W, Rodriguez-Algarra F, Sharif JA, Tsalenchuk M, Winchester LM, Yaman Ü, Yao Z, Ranson JM, Llewellyn DJ. Artificial intelligence for neurodegenerative experimental models. Alzheimers Dement 2023; 19:5970-5987. [PMID: 37768001 DOI: 10.1002/alz.13479] [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: 04/17/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 09/29/2023]
Abstract
INTRODUCTION Experimental models are essential tools in neurodegenerative disease research. However, the translation of insights and drugs discovered in model systems has proven immensely challenging, marred by high failure rates in human clinical trials. METHODS Here we review the application of artificial intelligence (AI) and machine learning (ML) in experimental medicine for dementia research. RESULTS Considering the specific challenges of reproducibility and translation between other species or model systems and human biology in preclinical dementia research, we highlight best practices and resources that can be leveraged to quantify and evaluate translatability. We then evaluate how AI and ML approaches could be applied to enhance both cross-model reproducibility and translation to human biology, while sustaining biological interpretability. DISCUSSION AI and ML approaches in experimental medicine remain in their infancy. However, they have great potential to strengthen preclinical research and translation if based upon adequate, robust, and reproducible experimental data. HIGHLIGHTS There are increasing applications of AI in experimental medicine. We identified issues in reproducibility, cross-species translation, and data curation in the field. Our review highlights data resources and AI approaches as solutions. Multi-omics analysis with AI offers exciting future possibilities in drug discovery.
Collapse
Affiliation(s)
- Sarah J Marzi
- UK Dementia Research Institute, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Brian M Schilder
- UK Dementia Research Institute, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Alexi Nott
- UK Dementia Research Institute, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | | | | | - Magda Bucholc
- School of Computing, Engineering & Intelligent Systems, Ulster University, Derry, UK
| | - Diane P Hanger
- Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | | | - Patrick A Lewis
- Royal Veterinary College, London, UK
- Department of Neurodegenerative Disease, UCL Queen Square Institute of Neurology, London, UK
| | | | - Wendy Noble
- Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | | | - Jalil-Ahmad Sharif
- UK Dementia Research Institute, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | - Maria Tsalenchuk
- UK Dementia Research Institute, Imperial College London, London, UK
- Department of Brain Sciences, Imperial College London, London, UK
| | | | - Ümran Yaman
- UK Dementia Research Institute at UCL, London, UK
| | | | | | - David J Llewellyn
- University of Exeter Medical School, Exeter, UK
- Alan Turing Institute, London, UK
| |
Collapse
|
10
|
Cao Q, Kumar M, Frazier A, Williams JB, Zhao S, Yan Z. Longitudinal characterization of behavioral, morphological and transcriptomic changes in a tauopathy mouse model. Aging (Albany NY) 2023; 15:11697-11719. [PMID: 37925173 PMCID: PMC10683589 DOI: 10.18632/aging.205057] [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: 03/01/2023] [Accepted: 09/02/2023] [Indexed: 11/06/2023]
Abstract
Neurodegenerative disorders, such as Alzheimer's disease (AD), have the gradual onset of neurobiological changes preceding clinical diagnosis by decades. To elucidate how brain dysfunction proceeds in neurodegenerative disorders, we performed longitudinal characterization of behavioral, morphological, and transcriptomic changes in a tauopathy mouse model, P301S transgenic mice. P301S mice exhibited cognitive deficits as early as 3 months old, and deficits in social preference and social cognition at 5-6 months. They had a significant decrease of arborization in basal dendrites of hippocampal pyramidal neurons from 3 months and apical dendrites of PFC pyramidal neurons at 9 months. Transcriptomic analysis of genome-wide changes revealed the enrichment of synaptic gene upregulation at 3 months of age, while most of these synaptic genes were downregulated in PFC and hippocampus of P301S mice at 9 months. These time-dependent changes in gene expression may lead to progressive alterations of neuronal structure and function, resulting in the manifestation of behavioral symptoms in tauopathies.
Collapse
Affiliation(s)
- Qing Cao
- Department of Physiology and Biophysics, State University of New York at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
| | - Manasa Kumar
- Department of Physiology and Biophysics, State University of New York at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
| | - Allea Frazier
- Department of Physiology and Biophysics, State University of New York at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
| | - Jamal B. Williams
- Department of Physiology and Biophysics, State University of New York at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
| | - Shengkai Zhao
- Department of Physiology and Biophysics, State University of New York at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
| | - Zhen Yan
- Department of Physiology and Biophysics, State University of New York at Buffalo, Jacobs School of Medicine and Biomedical Sciences, Buffalo, NY 14203, USA
| |
Collapse
|
11
|
Balu D, Valencia-Olvera AC, Islam Z, Mielczarek C, Hansen A, Perez Ramos TM, York J, LaDu MJ, Tai LM. APOE genotype and sex modulate Alzheimer's disease pathology in aged EFAD transgenic mice. Front Aging Neurosci 2023; 15:1279343. [PMID: 38020764 PMCID: PMC10644540 DOI: 10.3389/fnagi.2023.1279343] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 10/10/2023] [Indexed: 12/01/2023] Open
Abstract
Increasing evidence supports that age, APOE and sex interact to modulate Alzheimer's disease (AD) risk, however the underlying pathways are unclear. One way that AD risk factors may modulate cognition is by impacting amyloid beta (Aβ) accumulation as plaques, and/or neuroinflammation Therefore, the goal of the present study was to evaluate the extent to which age, APOE and sex modulate Aβ pathology, neuroinflammation and behavior in vivo. To achieve this goal, we utilized the EFAD mice, which express human APOE3 or APOE4 and have five familial AD mutations (FAD) that result in Aβ42 overproduction. We assessed Aβ levels, reactive glia and Morris water maze performance in 6-, 10-, 14-, and 18-month-old EFAD mice. Female APOE4 mice had the highest Aβ deposition, fibrillar amyloid deposits and neuroinflammation as well as earlier behavior deficits. Interestingly, we found that female APOE3 mice and male APOE4 mice had similar levels of pathology. Collectively our data support that the combination of APOE4 and female sex is the most detrimental combination for AD, and that at older ages, female sex may be equivalent to APOE4 genotype.
Collapse
Affiliation(s)
- Deebika Balu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Ana C. Valencia-Olvera
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Zarak Islam
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
- University of Illinois College of Medicine, Chicago, IL, United States
| | - Clare Mielczarek
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Allison Hansen
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
- University of Illinois College of Medicine, Peoria, IL, United States
| | - Tamara M. Perez Ramos
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
- School of Medicine, St. George’s University, St. George’s, Grenada
| | - Jason York
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| | - Leon M. Tai
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
12
|
Aleksandrova Y, Neganova M. Deciphering the Mysterious Relationship between the Cross-Pathogenetic Mechanisms of Neurodegenerative and Oncological Diseases. Int J Mol Sci 2023; 24:14766. [PMID: 37834214 PMCID: PMC10573395 DOI: 10.3390/ijms241914766] [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: 08/10/2023] [Revised: 09/22/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023] Open
Abstract
The relationship between oncological pathologies and neurodegenerative disorders is extremely complex and is a topic of concern among a growing number of researchers around the world. In recent years, convincing scientific evidence has accumulated that indicates the contribution of a number of etiological factors and pathophysiological processes to the pathogenesis of these two fundamentally different diseases, thus demonstrating an intriguing relationship between oncology and neurodegeneration. In this review, we establish the general links between three intersecting aspects of oncological pathologies and neurodegenerative disorders, i.e., oxidative stress, epigenetic dysregulation, and metabolic dysfunction, examining each process in detail to establish an unusual epidemiological relationship. We also focus on reviewing the current trends in the research and the clinical application of the most promising chemical structures and therapeutic platforms that have a modulating effect on the above processes. Thus, our comprehensive analysis of the set of molecular determinants that have obvious cross-functional pathways in the pathogenesis of oncological and neurodegenerative diseases can help in the creation of advanced diagnostic tools and in the development of innovative pharmacological strategies.
Collapse
Affiliation(s)
- Yulia Aleksandrova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, 142432 Chernogolovka, Russia;
| | - Margarita Neganova
- Institute of Physiologically Active Compounds at Federal Research Center of Problems of Chemical Physics and Medicinal Chemistry, Russian Academy of Sciences, 142432 Chernogolovka, Russia;
- Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, 420088 Kazan, Russia
| |
Collapse
|
13
|
Pavon MV, Navakkode S, Wong LW, Sajikumar S. Inhibition of Nogo-A rescues synaptic plasticity and associativity in APP/PS1 animal model of Alzheimer's disease. Semin Cell Dev Biol 2023; 139:111-120. [PMID: 35431138 DOI: 10.1016/j.semcdb.2022.04.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/30/2022] [Accepted: 04/06/2022] [Indexed: 12/31/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by memory loss and cognitive decline. Synaptic impairment is one of the first events to occur in the progression of this disease. Synaptic plasticity and cellular association of various plastic events have been shown to be affected in AD models. Nogo-A, a well-known axonal growth inhibitor with a recently discovered role as a plasticity suppressor, and its main receptor Nogo-66 receptor 1 (NGR1) have been found to be overexpressed in the hippocampus of Alzheimer's patients. However, the role of Nogo-A and its receptor in the pathology of AD is still widely unknown. In this work we set out to investigate whether Nogo-A is working as a plasticity suppressor in AD. Our results show that inhibition of the Nogo-A pathway via the Nogo-R antibody in an Alzheimer's mouse model, APP/PS1, leads to the restoration of both synaptic plasticity and associativity in a protein synthesis and NMDR-dependent manner. We also show that inhibition of the p75NTR pathway, which is strongly associated with NGR1, restores synaptic plasticity as well. Mechanistically, we propose that the restoration of synaptic plasticity in APP/PS1 via inhibition of the Nogo-A pathway is due to the modulation of the RhoA-ROCK2 pathway and increase in plasticity related proteins. Our study identifies Nogo-A as a plasticity suppressor in AD models hence targeting Nogo-A could be a promising strategy to understanding AD pathology.
Collapse
Affiliation(s)
- Maria Vazquez Pavon
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; Life Sciences Institute, Neurobiology Programme, National University of Singapore, Singapore 117456, Singapore
| | - Sheeja Navakkode
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
| | - Lik-Wei Wong
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; Life Sciences Institute, Neurobiology Programme, National University of Singapore, Singapore 117456, Singapore
| | - Sreedharan Sajikumar
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; Life Sciences Institute, Neurobiology Programme, National University of Singapore, Singapore 117456, Singapore; Healthy Longevity Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore.
| |
Collapse
|
14
|
Sai K, Nakanishi A, Scofield KM, Tokarz DA, Linder KE, Cohen TJ, Ninomiya-Tsuji J. Aberrantly activated TAK1 links neuroinflammation and neuronal loss in Alzheimer's disease mouse models. J Cell Sci 2023; 136:jcs260102. [PMID: 36912451 PMCID: PMC10112982 DOI: 10.1242/jcs.260102] [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: 04/08/2022] [Accepted: 01/27/2023] [Indexed: 03/14/2023] Open
Abstract
Neuroinflammation is causally associated with Alzheimer's disease (AD) pathology. Reactive glia cells secrete various neurotoxic factors that impair neuronal homeostasis eventually leading to neuronal loss. Although the glial activation mechanism in AD has been relatively well studied, how it perturbs intraneuronal signaling, which ultimately leads to neuronal cell death, remains poorly understood. Here, we report that compound stimulation with the neurotoxic factors TNF and glutamate aberrantly activates neuronal TAK1 (also known as MAP3K7), which promotes the pathogenesis of AD in mouse models. Glutamate-induced Ca2+ influx shifts TNF signaling to hyper-activate TAK1 enzymatic activity through Ca2+/calmodulin-dependent protein kinase II, which leads to necroptotic cellular damage. Genetic ablation and pharmacological inhibition of TAK1 ameliorated AD-associated neuronal loss and cognitive impairment in the AD model mice. Our findings provide a molecular mechanism linking cytokines, Ca2+ signaling and neuronal necroptosis in AD.
Collapse
Affiliation(s)
- Kazuhito Sai
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695-7633, USA
| | - Aoi Nakanishi
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695-7633, USA
| | - Kimberly M. Scofield
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695-7633, USA
| | - Debra A. Tokarz
- Center for Human Health and the Environment, Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
| | - Keith E. Linder
- Center for Human Health and the Environment, Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA
| | - Todd J. Cohen
- Department of Neurology, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jun Ninomiya-Tsuji
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695-7633, USA
| |
Collapse
|
15
|
Wen Y, Zhang L, Li N, Tong A, Zhao C. Nutritional assessment models for Alzheimer's disease: Advances and perspectives. FOOD FRONTIERS 2023. [DOI: 10.1002/fft2.216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023] Open
Affiliation(s)
- Yuxi Wen
- College of Marine Sciences Fujian Agriculture and Forestry University Fuzhou China
- Universidade de Vigo, Nutrition and Bromatology Group, Department of Analytical and Food Chemistry Faculty of Sciences Ourense Spain
| | - Lizhu Zhang
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Na Li
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Aijun Tong
- College of Food Science Fujian Agriculture and Forestry University Fuzhou China
| | - Chao Zhao
- College of Marine Sciences Fujian Agriculture and Forestry University Fuzhou China
- Key Laboratory of Marine Biotechnology of Fujian Province, Institute of Oceanology Fujian Agriculture and Forestry University Fuzhou China
| |
Collapse
|
16
|
Xu QQ, Su ZR, Yang W, Zhong M, Xian YF, Lin ZX. Patchouli alcohol attenuates the cognitive deficits in a transgenic mouse model of Alzheimer's disease via modulating neuropathology and gut microbiota through suppressing C/EBPβ/AEP pathway. J Neuroinflammation 2023; 20:19. [PMID: 36717922 PMCID: PMC9887791 DOI: 10.1186/s12974-023-02704-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Accepted: 01/22/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a chronic neurodegenerative disease characterized by progressive cognitive dysfunctions and behavioral impairments. Patchouli alcohol (PA), isolated from Pogostemonis Herba, exhibits multiple pharmacological properties, including neuroprotective effects. This study aimed to investigate the therapeutic effects of PA against AD using the TgCRND8 transgenic AD mouse model, and to explore the underlying mechanisms targeting CCAAT/enhancer-binding protein β/asparagine endopeptidase (C/EBPβ/AEP) signaling pathway. METHODS After genotyping to confirm the transgenicity, drug treatments were administered intragastrically once daily to 3-month-old TgCRND8 mice for 4 consecutive months. Several behavioral tests were applied to assess different aspects of neurological functions. Then the brain and colon tissues were harvested for in-depth mechanistic studies. To further verify whether PA exerts anti-AD effects via modulating C/EBPβ/AEP signaling pathway in TgCRND8 mice, adeno-associated virus (AAV) vectors encoding CEBP/β were bilaterally injected into the hippocampal CA1 region in TgCRND8 mice to overexpress C/EBPβ. Additionally, the fecal microbiota transplantation (FMT) experiment was performed to verify the potential role of gut microbiota on the anti-AD effects of PA. RESULTS Our results showed that PA treatment significantly improved activities of daily living (ADL), ameliorated the anxiety-related behavioral deficits and cognitive impairments in TgCRND8 mice. PA modulated the amyloid precursor protein (APP) processing. PA also markedly reduced the levels of beta-amyloid (Aβ) 40 and Aβ42, suppressed Aβ plaque burdens, inhibited tau protein hyperphosphorylation at several sites and relieved neuroinflammation in the brains of TgCRND8 mice. Moreover, PA restored gut dysbiosis and inhibited the activation of the C/EBPβ/AEP signaling pathway in the brain and colon tissues of TgCRND8 mice. Interestingly, PA strikingly alleviated the AD-like pathologies induced by the overexpression of C/EBPβ in TgCRND8 mice. Additionally, the FMT of fecal microbiota from the PA-treated TgCRND8 mice significantly alleviated the cognitive impairments and AD-like pathologies in the germ-free TgCRND8 mice. CONCLUSION All these findings amply demonstrated that PA could ameliorate the cognitive deficits in TgCRND8 mice via suppressing Aβ plaques deposition, hyperphosphorylation of tau protein, neuroinflammation and gut dysbiosis through inhibiting the activation of C/EBPβ/AEP pathway, suggesting that PA is a promising naturally occurring chemical worthy of further development into the pharmaceutical treatment of AD.
Collapse
Affiliation(s)
- Qing-Qing Xu
- grid.10784.3a0000 0004 1937 0482School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China
| | - Zi-Ren Su
- grid.411866.c0000 0000 8848 7685Guangdong Provincial Key Laboratory of New Drug Development and Research of Chinese Medicine, Mathematical Engineering Academy of Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Wen Yang
- grid.10784.3a0000 0004 1937 0482School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China
| | - Mei Zhong
- grid.10784.3a0000 0004 1937 0482School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China
| | - Yan-Fang Xian
- grid.10784.3a0000 0004 1937 0482School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China
| | - Zhi-Xiu Lin
- grid.10784.3a0000 0004 1937 0482School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China ,grid.10784.3a0000 0004 1937 0482Hong Kong Institute of Integrative Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China ,grid.10784.3a0000 0004 1937 0482Li Dak Sum Yip Yio Chin R&D Centre for Chinese Medicine, The Chinese University of Hong Kong, Shatin, N.T., Hong Kong SAR, People’s Republic of China
| |
Collapse
|
17
|
Xiao QY, Ye TY, Wang XL, Qi DM, Cheng XR. Effects of Qi-Fu-Yin on aging of APP/PS1 transgenic mice by regulating the intestinal microbiome. Front Cell Infect Microbiol 2023; 12:1048513. [PMID: 36710967 PMCID: PMC9880330 DOI: 10.3389/fcimb.2022.1048513] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 12/23/2022] [Indexed: 01/14/2023] Open
Abstract
Introduction Alzheimer's disease is the most common form of dementia and closely related to aging. Qi-Fu-Yin is widely used to treat dementia, but its anti-aging effects is unknown. Methods We used 11-month-old APP/PS1 transgenic mice for behavioral tests to observe the changes in cognitive function and age-related symptoms after Qi-Fu-Yin treatment. Fecal samples were collected for 16sRNA sequencing and metagenomic sequencing. Differences among the groups of intestinal microbiota and the associations with aging and intestinal microbiota were analyzed based on the results. Results Here we found that Qi-Fu-Yin improved the ability of motor coordination, raised survival rate and prolonged the survival days under cold stress stimulation in aged APP/ PS1 transgenic mice. Our data from 16sRNA and metagenomic sequencing showed that at the Family level, the intestinal microbiota was significantly different among wild-type mice, APP/PS1 transgenic mice and the Qi-Fu-Yin group by PCA analysis. Importantly, Qi-Fu-Yin improved the functional diversity of the major KEGG pathways, carbohydrate-active enzymes, and major virulence factors in the intestinal flora of APP/PS1 transgenic mice. Among them, the functions of eight carbohydrate-active enzymes (GT2_Glycos_transf_2, GT4, GT41, GH2, CE1, CE10, CE3, and GH24) and the functions of top three virulence factors (defensive virulence factors, offensive virulence factors and nonspecific virulence factors) were significantly and positively correlated with the level of grasping ability. We further indicated that the Qi-Fu-Yin significantly reduced the plasma levels of IL-6. Conclusion Our results indicated that the effects of Qi-Fu-Yin anti-aging of APP/PS1 transgenic mice might be through the regulation of intestinal flora diversity, species richness and the function of major active enzymes.
Collapse
Affiliation(s)
- Qiu-yue Xiao
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tian-yuan Ye
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiao-long Wang
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dong-mei Qi
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiao-rui Cheng
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
18
|
Luxem K, Mocellin P, Fuhrmann F, Kürsch J, Miller SR, Palop JJ, Remy S, Bauer P. Identifying behavioral structure from deep variational embeddings of animal motion. Commun Biol 2022; 5:1267. [PMID: 36400882 PMCID: PMC9674640 DOI: 10.1038/s42003-022-04080-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 10/06/2022] [Indexed: 11/19/2022] Open
Abstract
Quantification and detection of the hierarchical organization of behavior is a major challenge in neuroscience. Recent advances in markerless pose estimation enable the visualization of high-dimensional spatiotemporal behavioral dynamics of animal motion. However, robust and reliable technical approaches are needed to uncover underlying structure in these data and to segment behavior into discrete hierarchically organized motifs. Here, we present an unsupervised probabilistic deep learning framework that identifies behavioral structure from deep variational embeddings of animal motion (VAME). By using a mouse model of beta amyloidosis as a use case, we show that VAME not only identifies discrete behavioral motifs, but also captures a hierarchical representation of the motif's usage. The approach allows for the grouping of motifs into communities and the detection of differences in community-specific motif usage of individual mouse cohorts that were undetectable by human visual observation. Thus, we present a robust approach for the segmentation of animal motion that is applicable to a wide range of experimental setups, models and conditions without requiring supervised or a-priori human interference.
Collapse
Affiliation(s)
- Kevin Luxem
- Leibniz Institute for Neurobiology (LIN), Department of Cellular Neuroscience, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Petra Mocellin
- Leibniz Institute for Neurobiology (LIN), Department of Cellular Neuroscience, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Falko Fuhrmann
- Leibniz Institute for Neurobiology (LIN), Department of Cellular Neuroscience, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Johannes Kürsch
- Leibniz Institute for Neurobiology (LIN), Department of Cellular Neuroscience, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Stephanie R Miller
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Jorge J Palop
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Stefan Remy
- Leibniz Institute for Neurobiology (LIN), Department of Cellular Neuroscience, Magdeburg, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
- Center for Behavioral Brain Sciences (CBBS), Magdeburg, Germany.
- German Center for Mental Health (DZPG), Magdeburg, Germany.
| | - Pavol Bauer
- Leibniz Institute for Neurobiology (LIN), Department of Cellular Neuroscience, Magdeburg, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| |
Collapse
|
19
|
Cummings J, Ortiz A, Castellino J, Kinney J. Diabetes: Risk factor and translational therapeutic implications for Alzheimer's disease. Eur J Neurosci 2022; 56:5727-5757. [PMID: 35128745 PMCID: PMC9393901 DOI: 10.1111/ejn.15619] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 12/31/2022]
Abstract
Type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) commonly co-occur. T2DM increases the risk for AD by approximately twofold. Animal models provide one means of interrogating the relationship of T2DM to AD and investigating brain insulin resistance in the pathophysiology of AD. Animal models show that persistent hyperglycaemia results in chronic low-grade inflammation that may contribute to the development of neuroinflammation and accelerate the pathobiology of AD. Epidemiological studies suggest that patients with T2DM who received treatment with specific anti-diabetic agents have a decreased risk for the occurrence of AD and all-cause dementia. Agents such as metformin ameliorate T2DM and may have other important systemic effects that lower the risk of AD. Glucagon-like peptide 1 (GLP-1) agonists have been associated with a decreased risk for AD in patients with T2DM. Both insulin and non-insulin anti-diabetic treatments have been evaluated for the treatment of AD in clinical trials. In most cases, patients included in the trials have clinical features of AD but do not have T2DM. Many of the trials were conducted prior to the use of diagnostic biomarkers for AD. Trials have had a wide range of durations and population sizes. Many of the agents used to treat T2DM do not cross the blood brain barrier, and the effects are posited to occur via lowering of peripheral hyperglycaemia and reduction of peripheral and central inflammation. Clinical trials of anti-diabetic agents to treat AD are ongoing and will provide insight into the therapeutic utility of these agents.
Collapse
Affiliation(s)
- Jeffrey Cummings
- Chambers‐Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health SciencesUniversity of Nevada Las Vegas (UNLV)Las VegasNevadaUSA
| | - Andrew Ortiz
- Department of Brain Health, School of Integrated Health SciencesUniversity of Nevada Las Vegas (UNLV)Las VegasNevadaUSA
| | | | - Jefferson Kinney
- Chambers‐Grundy Center for Transformative Neuroscience, Department of Brain Health, School of Integrated Health SciencesUniversity of Nevada Las Vegas (UNLV)Las VegasNevadaUSA,Department of Brain Health, School of Integrated Health SciencesUniversity of Nevada Las Vegas (UNLV)Las VegasNevadaUSA
| |
Collapse
|
20
|
Buendía D, Guncay T, Oyanedel M, Lemus M, Weinstein A, Ardiles ÁO, Marcos J, Fernandes A, Zângaro R, Muñoz P. The Transcranial Light Therapy Improves Synaptic Plasticity in the Alzheimer’s Disease Mouse Model. Brain Sci 2022; 12:brainsci12101272. [PMID: 36291206 PMCID: PMC9599908 DOI: 10.3390/brainsci12101272] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/10/2022] [Accepted: 09/15/2022] [Indexed: 11/16/2022] Open
Abstract
Alzheimer’s disease (AD) is the main cause of dementia worldwide. Emerging non-invasive treatments such as photobiomodulation target the mitochondria to minimize brain damage, improving cognitive functions. In this work, an experimental design was carried out to evaluate the effect of transcranial light therapy (TLTC) on synaptic plasticity (SP) and cognitive functions in an AD animal model. Twenty-three mice were separated into two general groups: an APP/PS1 (ALZ) transgenic group and a wild-type (WT) group. Each group was randomly subdivided into two subgroups: mice with and without TLTC, depending on whether they would undergo treatment with TLTC. Cognitive function, measured through an object recognition task, showed non-significant improvement after TLTC. SP, on the other hand, was evaluated using four electrophysiological parameters from the Schaffer-CA1 collateral hippocampal synapses: excitatory field potentials (fEPSP), paired pulse facilitation (PPF), long-term depression (LTD), and long-term potentiation (LTP). An improvement was observed in subjects treated with TLTC, showing higher levels of LTP than those transgenic mice that were not exposed to the treatment. Therefore, the results obtained in this work showed that TLTC could be an efficient non-invasive treatment for AD-associated SP deficits.
Collapse
Affiliation(s)
- Débora Buendía
- Programa de Engenharia Biomédica, Instituto de Engenharia Biomédica, Universidade Anhembi Morumbi—UAM, Rua Casa do Ator, 294, Sao Paulo 04546-001, Brazil
- Escuela de Ingeniería Civil Biomédica, Facultad de Ingeniería, Universidad de Valparaíso, General Cruz 222, Valparaíso 2362905, Chile
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Centro de Inovação, Tecnología e Educação—CITÉ, Parque Tecnológico de São José dos Campos, Estrada Dr. Altino Bondesan 500, São José dos Campos 12247-016, Brazil
| | - Tatiana Guncay
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
| | - Macarena Oyanedel
- Escuela de Ingeniería Civil Biomédica, Facultad de Ingeniería, Universidad de Valparaíso, General Cruz 222, Valparaíso 2362905, Chile
| | - Makarena Lemus
- Escuela de Ingeniería Civil Biomédica, Facultad de Ingeniería, Universidad de Valparaíso, General Cruz 222, Valparaíso 2362905, Chile
| | - Alejandro Weinstein
- Escuela de Ingeniería Civil Biomédica, Facultad de Ingeniería, Universidad de Valparaíso, General Cruz 222, Valparaíso 2362905, Chile
| | - Álvaro O. Ardiles
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile
- Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Angamos 655, Viña del Mar 2540064, Chile
| | - José Marcos
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Escuela de Ciencias Agrícolas y Veterinarias, Universidad Viña del Mar, Viña del Mar 2572007, Chile
| | - Adriana Fernandes
- Programa de Engenharia Biomédica, Instituto de Engenharia Biomédica, Universidade Anhembi Morumbi—UAM, Rua Casa do Ator, 294, Sao Paulo 04546-001, Brazil
- Centro de Inovação, Tecnología e Educação—CITÉ, Parque Tecnológico de São José dos Campos, Estrada Dr. Altino Bondesan 500, São José dos Campos 12247-016, Brazil
| | - Renato Zângaro
- Programa de Engenharia Biomédica, Instituto de Engenharia Biomédica, Universidade Anhembi Morumbi—UAM, Rua Casa do Ator, 294, Sao Paulo 04546-001, Brazil
- Centro de Inovação, Tecnología e Educação—CITÉ, Parque Tecnológico de São José dos Campos, Estrada Dr. Altino Bondesan 500, São José dos Campos 12247-016, Brazil
- Correspondence: (R.Z.); (P.M.); Tel.: +55-12-997830843 (R.Z.); +56-969028160 (P.M.)
| | - Pablo Muñoz
- Centro de Neurología Traslacional, Facultad de Medicina, Universidad de Valparaíso, Valparaíso 2341386, Chile
- Escuela de Medicina, Facultad de Medicina, Universidad de Valparaíso, Angamos 655, Viña del Mar 2540064, Chile
- Centro de Investigaciones Biomédicas, Facultad de Medicina, Universidad de Valparaíso, Angamos 655, Viña del Mar 2540064, Chile
- Correspondence: (R.Z.); (P.M.); Tel.: +55-12-997830843 (R.Z.); +56-969028160 (P.M.)
| |
Collapse
|
21
|
Ezzat MI, Issa MY, Sallam IE, Zaafar D, Khalil HMA, Mousa MR, Sabry D, Gawish AY, Elghandour AH, Mohsen E. Impact of different processing methods on the phenolics and neuroprotective activity of Fragaria ananassa Duch. extracts in a D-galactose and aluminum chloride-induced rat model of aging. Food Funct 2022; 13:7794-7812. [PMID: 35766389 DOI: 10.1039/d2fo00645f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/16/2023]
Abstract
Age-related diseases, including dementia, are a major health concern affecting daily human life. Strawberry (Fragaria ananassa Duch.) is the most eaten fruit worldwide due to its exceptional aroma and flavor. However, it's rapid softening and decay limit its shelf-life. Freezing and boiling represent the well-known conservation methods to extend its shelf-life. Therefore, we aimed to discover the phytochemical content differences of fresh and processed strawberries associated with investigating and comparing their neuroprotective effects in a rat model of aging. Female Wistar rats were orally pretreated with fresh, boiled, and frozen F. ananassa methanolic extracts (250 mg kg-1) for 2 weeks, and then these extracts were concomitantly exposed to D-galactose [65 mg kg-1, subcutaneously (S/C)] and AlCl3 (200 mg kg-1, orally) for 6 weeks to develop aging-like symptoms. The results of UPLC/ESI-MS phytochemical profiling revealed 36 secondary metabolites, including phenolics, flavonoids, and their glycoside derivatives. Compared with boiled and frozen extracts, the fresh extract ameliorated the behavioral deficits including anxiety and cognitive dysfunction, upregulated brain HO-1 and Nrf2 levels, and markedly reduced caspase-3 and PPAR-γ levels. Moreover, LDH and miRNA-9, 124 and 132 protein expressions were reduced. The histological architecture of the brain hippocampus was restored and glial fibrillary acidic protein (GFAP) immunoexpression was downregulated. In conclusion, the fresh extract has neuroprotective activity that could have a promising role in ameliorating age-related neurodegeneration.
Collapse
Affiliation(s)
- Marwa I Ezzat
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Kasr El-Ainy Street, Cairo, 11562, Egypt.
| | - Marwa Y Issa
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Kasr El-Ainy Street, Cairo, 11562, Egypt.
| | - Ibrahim E Sallam
- Pharmacognosy Department, College of Pharmacy, October University for Modern Sciences and Arts (MSA), 6th of October City, 12566, Egypt
| | - Dalia Zaafar
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Modern University for Technology and Information, el-Mokattam, Cairo, 11581, Egypt
| | - Heba M A Khalil
- Veterinary Hygiene and Management Department, Faculty of Vet. Medicine, Cairo University, Giza, 12211, Egypt
| | - Mohamed R Mousa
- Pathology Department, Faculty of Vet. Medicine, Cairo University, Giza, 12211, Egypt
| | - Dina Sabry
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, 11562, Egypt
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Badr University, 11829, Egypt
| | - Aya Y Gawish
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Modern University for Technology and Information, el-Mokattam, Cairo, 11581, Egypt
| | - Ahmed H Elghandour
- Communication Department, Military Technical College, Cairo, 11766, Egypt
| | - Engy Mohsen
- Pharmacognosy Department, Faculty of Pharmacy, Cairo University, Kasr El-Ainy Street, Cairo, 11562, Egypt.
| |
Collapse
|
22
|
Zajac DJ, Shaw BC, Braun DJ, Green SJ, Morganti JM, Estus S. Exogenous Short Chain Fatty Acid Effects in APP/PS1 Mice. Front Neurosci 2022; 16:873549. [PMID: 35860296 PMCID: PMC9289923 DOI: 10.3389/fnins.2022.873549] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 06/13/2022] [Indexed: 11/13/2022] Open
Abstract
Elucidating the impact of the gut microbiome on Alzheimer’s Disease (AD) is an area of intense interest. Short chain fatty acids (SCFAs) are major microbiota metabolites that have been implicated as a mediator of gut microbiome effects in the brain. Here, we tested the effects of SCFA-treated water vs. saline-treated water on APPswe/PSEN1dE9 mice maintained under standard laboratory conditions. Mice were treated with SCFAs from five months of age until ten months of age, when they were evaluated for microbiome profile, impaired spatial memory as evaluated with the radial arm water maze, astrocyte activation as measured by Gfap expression and amyloid burden as assessed by histochemistry and MSD ELISA. We report that SCFA treatment increased alpha-diversity and impacted the gut microbiome profile by increasing, in part, the relative abundance of several bacteria that typically produce SCFAs. However, SCFA treatment did not significantly affect behavior. Similarly, SCFAs did not affect cortical or hippocampal astrocyte activation observed in the APP/PS1 mice. Lastly, although robust levels of soluble and insoluble amyloid were present in the APP/PS1 mice, SCFA treatment had no effect on these indices. Overall, our findings are that SCFA treatment modifies the microbiome in a fashion that may increase further SCFA production. However, SCFA treatment did not alter behavior, astrocyte activation, nor amyloid neuropathology in APP/PS1 mice maintained with a conventional microbiome.
Collapse
Affiliation(s)
- Diana J. Zajac
- Department of Physiology and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
| | - Benjamin C. Shaw
- Department of Physiology and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
| | - David J. Braun
- Department of Neuroscience and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
| | - Stefan J. Green
- Genome Research Core, Research Resources Center, University of Illinois at Chicago, Chicago, IL, United States
| | - Joshua M. Morganti
- Department of Neuroscience and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
| | - Steven Estus
- Department of Physiology and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY, United States
- *Correspondence: Steven Estus,
| |
Collapse
|
23
|
Ahmed S, Jing Y, Mockett BG, Zhang H, Abraham WC, Liu P. Partial Endothelial Nitric Oxide Synthase Deficiency Exacerbates Cognitive Deficit and Amyloid Pathology in the APPswe/PS1ΔE9 Mouse Model of Alzheimer’s Disease. Int J Mol Sci 2022; 23:ijms23137316. [PMID: 35806318 PMCID: PMC9266765 DOI: 10.3390/ijms23137316] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 06/29/2022] [Accepted: 06/30/2022] [Indexed: 01/25/2023] Open
Abstract
Increasing evidence implicates endothelial dysfunction in the pathogenesis of Alzheimer’s disease (AD). Nitric oxide (NO) derived from endothelial NO synthase (eNOS) is essential in maintaining cerebrovascular function and can modulate the production and clearance of amyloid beta (Aβ). APPswe/PSdE1 (APP/PS1) mice display age-related Aβ accumulation and memory deficits. In order to make the model more clinically relevant with an element of endothelial dysfunction, we generated APP/PS1/eNOS+/− mice by crossing complete eNOS deficient (eNOS−/−) mice and APP/PS1 mice. APP/PS1/eNOS+/− mice at 8 months of age displayed a more severe spatial working memory deficit relative to age-matched APP/PS1 mice. Moreover, immunohistochemistry and immunoblotting revealed significantly increased Aβ plaque load in the brains of APP/PS1/eNOS+/− mice, concomitant with upregulated BACE-1 (hence increased Aβ production), downregulated insulin-degrading enzyme (hence reduced Aβ clearance) and increased immunoreactivity and expression of microglia. The present study, for the first time, demonstrated that partial eNOS deficiency exacerbated behavioral dysfunction, Aβ brain deposition, and microglial pathology in APP/PS1 mice, further implicating endothelial dysfunction in the pathogenesis of AD. The present findings also provide the scientific basis for developing preventive and/or therapeutic strategies by targeting endothelial dysfunction.
Collapse
Affiliation(s)
- Sara Ahmed
- Department of Anatomy, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, Dunedin 9054, New Zealand; (S.A.); (Y.J.)
| | - Yu Jing
- Department of Anatomy, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, Dunedin 9054, New Zealand; (S.A.); (Y.J.)
| | - Bruce G. Mockett
- Department of Psychology, Brain Health Research Centre, University of Otago, Dunedin 9054, New Zealand; (B.G.M.); (W.C.A.)
| | - Hu Zhang
- School of Pharmacy, Brain Health Research Centre, University of Otago, Dunedin 9054, New Zealand;
| | - Wickliffe C. Abraham
- Department of Psychology, Brain Health Research Centre, University of Otago, Dunedin 9054, New Zealand; (B.G.M.); (W.C.A.)
| | - Ping Liu
- Department of Anatomy, School of Biomedical Sciences, Brain Health Research Centre, University of Otago, Dunedin 9054, New Zealand; (S.A.); (Y.J.)
- Correspondence:
| |
Collapse
|
24
|
Yu Y, Jiang X, Fang X, Wang Y, Liu P, Ling J, Yu L, Jiang M, Tang C. Transauricular Vagal Nerve Stimulation at 40 Hz Inhibits Hippocampal P2X7R/NLRP3/Caspase-1 Signaling and Improves Spatial Learning and Memory in 6-Month-Old APP/PS1 Mice. Neuromodulation 2022; 26:589-600. [PMID: 35595603 DOI: 10.1016/j.neurom.2022.03.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/04/2022] [Accepted: 03/14/2022] [Indexed: 01/01/2023]
Abstract
OBJECTIVES Transauricular vagal nerve stimulation (taVNS) at 40 Hz attenuates hippocampal amyloid load in 6-month-old amyloid precursor protein/presenilin 1 (APP/PS1) transgenic mice, but it is unclear whether 40-Hz taVNS can improve cognition in these mice. Moreover, the underlying mechanisms are still unclear. MATERIALS AND METHODS 6-month-old C57BL/6 (wild type [WT]) and APP/PS1 mice were subjected to 40-Hz taVNS. Novel Object Recognition and the Morris Water Maze were used to evaluate cognition. Hippocampal amyloid-β (Aβ)1-40, Aβ1-42, pro-interleukin (IL)-1β, and pro-IL-18 were measured using enzyme-linked immunosorbent assays. Hippocampal Aβ42, purinergic 2X7 receptor (P2X7R), nucleotide-binding oligomerization domain-like receptor pyrin domain containing 3 (NLRP3), Caspase-1, IL-1β, and IL-18 expression were evaluated by western blotting. Histologic assessments including immunofluorescence, immunohistochemistry, Nissl staining, and Congo red staining were used to assess microglial phagocytosis, neuroprotective effects, and Aβ plaque load. RESULTS 40-Hz taVNS improved spatial memory and learning in 6-month-old APP/PS1 mice but did not affect recognition memory. There were no effects on the cognitive behaviors of 6-month-old WT mice. taVNS at 40 Hz modulated microglia; significantly decreased levels of Aβ1-40, Aβ1-42, pro-IL-1β, and pro-IL-18; inhibited Aβ42, P2X7R, NLRP3, Caspase-1, IL-1β, and IL-18 expression; reduced Aβ deposits; and had neuroprotective effects in the hippocampus of 6-month-old APP/PS1 mice. These changes were not observed in 6-month-old WT mice. CONCLUSION Our results show that 40-Hz taVNS inhibits the hippocampal P2X7R/NLRP3/Caspase-1 signaling and improves spatial learning and memory in 6-month-old APP/PS1 mice.
Collapse
Affiliation(s)
- Yutian Yu
- Acupuncture Department, Beijing Shijitan Hospital, Capital Medical University, Beijing, China; Ninth School of Clinical Medicine, Peking University, Beijing, China.
| | - Xuejiao Jiang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Xian Fang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Yu Wang
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Science, Beijing, China
| | - Pengfei Liu
- Ninth School of Clinical Medicine, Peking University, Beijing, China; Department of Anesthesiology, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Jing Ling
- Department of Gynecology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, China
| | - Lingling Yu
- Department of Chinese and Western Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Jiang
- Acupuncture Department, Beijing Shijitan Hospital, Capital Medical University, Beijing, China; Ninth School of Clinical Medicine, Peking University, Beijing, China.
| | - Chunzhi Tang
- Clinical Medical College of Acupuncture, Moxibustion, and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, China.
| |
Collapse
|
25
|
Vidal-Palencia L, Ramon-Duaso C, González-Parra JA, Busquets-Garcia A. Gene Expression Analysis of the Endocannabinoid System in Presymptomatic APP/PS1 Mice. Front Pharmacol 2022; 13:864591. [PMID: 35370697 PMCID: PMC8971609 DOI: 10.3389/fphar.2022.864591] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/02/2022] [Indexed: 11/21/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common type of dementia and neurodegeneration. The actual cause of AD progression is still unknown and no curative treatment is available. Recently, findings in human samples and animal models pointed to the endocannabinoid system (ECS) as a promising therapeutic approach against AD. However, the specific mechanisms by which cannabinoid drugs induce potential beneficial effects are still undefined. For this reason, it is required a full characterization of the ECS at different time points of AD progression considering important factors such as sex or the analysis of different brain regions to improve future cannabinoid-dependent therapies in AD. Thus, the main aim of the present study is to expand our knowledge of the status of the ECS in a presymptomatic period (3 months of age) using the AD mouse model APP/PS1 mice. First, we evaluated different behavioral domains including anxiety, cognitive functions, and social interactions in male and female APP/PS1 mice at 4 months of age. Although a mild working memory impairment was observed in male APP/PS1 mice, in most of the behaviors assessed we found no differences between genotypes. At 3 months of age, we performed a characterization of the ECS in different brain regions of the APP/PS1 mice considering the sex variable. We assessed the expression of the ECS components by quantitative Real-Time Polymerase Chain Reaction in the hippocampus, prefrontal cortex, hypothalamus, olfactory bulb, and cerebellum. Interestingly, gene expression levels of the type-1 and type-2 cannabinoid receptors and the anabolic and catabolic enzymes, differed depending on the brain region and the sex analyzed. For example, CB1R expression levels decreased in both hippocampus and prefrontal cortex of male APP/PS1 mice but increased in female mice. In contrast, CB2R expression was decreased in females, whereas males tended to have higher levels. Overall, our data indicated that the ECS is already altered in APP/PS1 mice at the presymptomatic stage, suggesting that it could be an early event contributing to the pathophysiology of AD or being a potential predictive biomarker.
Collapse
|
26
|
Therapeutic treatment with the anti-inflammatory drug candidate MW151 may partially reduce memory impairment and normalizes hippocampal metabolic markers in a mouse model of comorbid amyloid and vascular pathology. PLoS One 2022; 17:e0262474. [PMID: 35081152 PMCID: PMC8791470 DOI: 10.1371/journal.pone.0262474] [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: 05/07/2021] [Accepted: 12/24/2021] [Indexed: 12/03/2022] Open
Abstract
Alzheimer’s disease (AD) is the leading cause of dementia in the elderly, but therapeutic options are lacking. Despite long being able to effectively treat the ill-effects of pathology present in various rodent models of AD, translation of these strategies to the clinic has so far been disappointing. One potential contributor to this situation is the fact that the vast majority of AD patients have other dementia-contributing comorbid pathologies, the most common of which are vascular in nature. This situation is modeled relatively infrequently in basic AD research, and almost never in preclinical studies. As part of our efforts to develop small molecule, anti-inflammatory therapeutics for neurological injury and disease, we have recently been exploring potentially promising treatments in preclinical multi-morbidity contexts. In the present study, we generated a mouse model of mixed amyloid and hyperhomocysteinemia (HHcy) pathology in which to test the efficacy of one of our anti-inflammatory compounds, MW151. HHcy can cause cerebrovascular damage and is an independent risk factor for both AD dementia and vascular contributions to cognitive impairment and dementia. We found that MW151 was able to partially rescue hippocampal-dependent spatial memory and learning deficits in this comorbidity context, and further, that the benefit is associated with a normalization of hippocampal metabolites detectable via magnetic resonance spectroscopy. These findings provide evidence that MW151 in particular, and potentially anti-inflammatory treatment more generally, may be beneficial in AD patients with comorbid vascular pathology.
Collapse
|
27
|
The microRNA-455 Null Mouse Has Memory Deficit and Increased Anxiety, Targeting Key Genes Involved in Alzheimer's Disease. Int J Mol Sci 2022; 23:ijms23010554. [PMID: 35008980 PMCID: PMC8745123 DOI: 10.3390/ijms23010554] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/24/2021] [Accepted: 12/30/2021] [Indexed: 12/21/2022] Open
Abstract
The complete molecular mechanisms underlying the pathophysiology of Alzheimer's disease (AD) remain to be elucidated. Recently, microRNA-455-3p has been identified as a circulating biomarker of early AD, with increased expression in post-mortem brain tissue of AD patients. MicroRNA-455-3p also directly targets and down-regulates APP, with the overexpression of miR-455-3p suppressing its toxic effects. Here, we show that miR-455-3p expression decreases with age in the brains of wild-type mice. We generated a miR-455 null mouse utilising CRISPR-Cas9 to explore its function further. Loss of miR-455 resulted in increased weight gain, potentially indicative of metabolic disturbances. Furthermore, performance on the novel object recognition task diminished significantly in miR-455 null mice (p = 0.004), indicating deficits in recognition memory. A slight increase in anxiety was also captured on the open field test. BACE1 and TAU were identified as new direct targets for miR-455-3p, with overexpression of miR-455-3p leading to a reduction in the expression of APP, BACE1 and TAU in neuroblastoma cells. In the hippocampus of miR-455 null mice at 14 months of age, the levels of protein for APP, BACE1 and TAU were all increased. Such findings reinforce the involvement of miR-455 in AD progression and demonstrate its action on cognitive performance.
Collapse
|
28
|
Yuan L, Zhang J, Guo JH, Holscher C, Yang JT, Wu MN, Wang ZJ, Cai HY, Han LN, Shi H, Han YF, Qi JS. DAla2-GIP-GLU-PAL Protects Against Cognitive Deficits and Pathology in APP/PS1 Mice by Inhibiting Neuroinflammation and Upregulating cAMP/PKA/CREB Signaling Pathways. J Alzheimers Dis 2021; 80:695-713. [PMID: 33579843 DOI: 10.3233/jad-201262] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive decline in cognitive function. Type 2 diabetes mellitus (T2DM) is an important risk factor for AD. Glucose-dependent insulinotropic polypeptide (GIP) has been identified to be effective in T2DM treatment and neuroprotection. OBJECTIVE The present study investigated the neuroprotective effects and possible mechanisms of DAla2GIP-Glu-PAL, a novel long-lasting GIP analogue, in APP/PS1 AD mice. METHODS Multiple behavioral tests were performed to examine the cognitive function of mice. In vivo hippocampus late-phase long-term potentiation (L-LTP) was recorded to reflect synaptic plasticity. Immunohistochemistry and immunofluorescence were used to examine the Aβ plaques and neuroinflammation in the brain. IL-1β, TNF-α, and cAMP/PKA/CREB signal molecules were also detected by ELISA or western blotting. RESULTS DAla2GIP-Glu-PAL increased recognition index (RI) of APP/PS1 mice in novel object recognition test, elevated spontaneous alternation percentage of APP/PS1 mice in Y maze test, and increased target quadrant swimming time of APP/PS1 mice in Morris water maze test. DAla2GIP-Glu-PAL treatment enhanced in vivo L-LTP of APP/PS1 mice. DAla2GIP-Glu-PAL significantly reduced Aβ deposition, inhibited astrocyte and microglia proliferation, and weakened IL-1β and TNF-α secretion. DAla2GIP-Glu-PAL also upregulated cAMP/PKA/CREB signal transduction and inhibited NF-κB activation in the hippocampus of APP/PS1 mice. CONCLUSION DAla2GIP-Glu-PAL can improve cognitive behavior, synaptic plasticity, and central pathological damage in APP/PS1 mice, which might be associated with the inhibition of neuroinflammation, as well as upregulation of cAMP-/PKA/CREB signaling pathway. This study suggests a potential benefit of DAla2GIP-Glu-PAL in the treatment of AD.
Collapse
Affiliation(s)
- Li Yuan
- Department of Physiology, Changzhi Medical College, Changzhi, Shanxi, PR China.,Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Jun Zhang
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Jun-Hong Guo
- Department of Neurology, First Hospital, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Christian Holscher
- Research and Experimental Center, Henan University of Chinese Medicine, Zhengzhou, Henan, PR China
| | - Jun-Ting Yang
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Mei-Na Wu
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Zhao-Jun Wang
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Hong-Yan Cai
- Department of Immunology and Microbiology, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Ling-Na Han
- Department of Physiology, Changzhi Medical College, Changzhi, Shanxi, PR China
| | - Hui Shi
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Yu-Fei Han
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| | - Jin-Shun Qi
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, Shanxi, PR China
| |
Collapse
|
29
|
Uddin O, Arakawa K, Raver C, Garagusi B, Keller A. Patterns of cognitive decline and somatosensory processing in a mouse model of amyloid accumulation. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2021; 10:100076. [PMID: 34820549 PMCID: PMC8599510 DOI: 10.1016/j.ynpai.2021.100076] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 10/28/2021] [Accepted: 11/01/2021] [Indexed: 01/13/2023]
Abstract
Despite copious amyloid plaques, 5XFAD mice show modest signs of cognitive decline. At ages 2 to 13 months old 5XFAD mice show no signs of sensory or pain dysfunctions. 5XFAD mice may not be a valid model for pain abnormalities in the context of AD.
Pain and cognitive decline increase with age. In particular, there is a troubling relationship between dementia and pain, with some studies showing higher prevalence and inadequate treatment of pain in this population. Alzheimer’s disease (AD) is one of the most common causes of dementia in older adults. Amyloid plaques are a hallmark of AD. The downstream processes these plaques promote are believed to affect neuronal and glial health and activity. There is a need to better understand how the neuropathological changes of AD shape neural activity and pain sensitivity. Here, we use the 5XFAD mouse model, in which dense amyloid accumulations occur at early ages, and in which previous studies reported signs of cognitive decline. We hypothesized that 5XFAD mice develop sensory and pain processing dysfunctions. Although amyloid burden was high throughout the brain, including in regions involved with sensory processing, we identified no functionally significant differences in reflexive or spontaneous signs of pain. Furthermore, expected signs of cognitive decline were modest; a finding consistent with variable results in the literature. These data suggest that models recapitulating other pathological features of Alzheimer’s disease might be better suited to studying differences in pain perception in this disease.
Collapse
Affiliation(s)
- Olivia Uddin
- Department of Anatomy and Neurobiology, Program in Neuroscience, University of Maryland School of Medicine, 20 Penn Street, Baltimore, MD 21201, United States
| | - Keiko Arakawa
- Department of Anatomy and Neurobiology, Program in Neuroscience, University of Maryland School of Medicine, 20 Penn Street, Baltimore, MD 21201, United States
| | - Charles Raver
- Department of Anatomy and Neurobiology, Program in Neuroscience, University of Maryland School of Medicine, 20 Penn Street, Baltimore, MD 21201, United States
| | - Brendon Garagusi
- Department of Anatomy and Neurobiology, Program in Neuroscience, University of Maryland School of Medicine, 20 Penn Street, Baltimore, MD 21201, United States
| | - Asaf Keller
- Department of Anatomy and Neurobiology, Program in Neuroscience, University of Maryland School of Medicine, 20 Penn Street, Baltimore, MD 21201, United States
| |
Collapse
|
30
|
Rechnitz O, Slutsky I, Morris G, Derdikman D. Hippocampal sub-networks exhibit distinct spatial representation deficits in Alzheimer's disease model mice. Curr Biol 2021; 31:3292-3302.e6. [PMID: 34146487 DOI: 10.1016/j.cub.2021.05.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 01/03/2021] [Accepted: 05/18/2021] [Indexed: 12/25/2022]
Abstract
Not much is known about how the dentate gyrus (DG) and hippocampal CA3 networks, critical for memory and spatial processing, malfunction in Alzheimer's disease (AD). While studies of associative memory deficits in AD have focused mainly on behavior, here, we directly measured neurophysiological network dysfunction. We asked what the pattern of deterioration of different networks is during disease progression. We investigated how the associative memory-processing capabilities in different hippocampal subfields are affected by familial AD (fAD) mutations leading to amyloid-β dyshomeostasis. Specifically, we focused on the DG and CA3, which are known to be involved in pattern completion and separation and are susceptible to pathological alterations in AD. To identify AD-related deficits in neural-ensemble dynamics, we recorded single-unit activity in wild-type (WT) and fAD model mice (APPSwe+PSEN1/ΔE9) in a novel tactile morph task, which utilizes the extremely developed somatosensory modality of mice. As expected from the sub-network regional specialization, we found that tactile changes induced lower rate map correlations in the DG than in CA3 of WT mice. This reflects DG pattern separation and CA3 pattern completion. In contrast, in fAD model mice, we observed pattern separation deficits in the DG and pattern completion deficits in CA3. This demonstration of region-dependent impairments in fAD model mice contributes to understanding of brain networks deterioration during fAD progression. Furthermore, it implies that the deterioration cannot be studied generally throughout the hippocampus but must be researched at a finer resolution of microcircuits. This opens novel systems-level approaches for analyzing AD-related neural network deficits.
Collapse
Affiliation(s)
- Ohad Rechnitz
- Department of Neuroscience, Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, 31096 Haifa, Israel
| | - Inna Slutsky
- Department of Physiology and Pharmacology, Sackler Faculty of Medicine, Tel Aviv University, 69978 Tel Aviv, Israel; Sagol School of Neuroscience, Tel Aviv University, 69978 Tel Aviv, Israel
| | - Genela Morris
- Department of Neuroscience, Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, 31096 Haifa, Israel
| | - Dori Derdikman
- Department of Neuroscience, Rappaport Faculty of Medicine and Research Institute, Technion - Israel Institute of Technology, 31096 Haifa, Israel.
| |
Collapse
|
31
|
Pentkowski NS, Rogge-Obando KK, Donaldson TN, Bouquin SJ, Clark BJ. Anxiety and Alzheimer's disease: Behavioral analysis and neural basis in rodent models of Alzheimer's-related neuropathology. Neurosci Biobehav Rev 2021; 127:647-658. [PMID: 33979573 DOI: 10.1016/j.neubiorev.2021.05.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 04/28/2021] [Accepted: 05/05/2021] [Indexed: 11/29/2022]
Abstract
Alzheimer's disease (AD) pathology is commonly associated with cognitive decline but is also composed of neuropsychiatric symptoms including psychological distress and alterations in mood, including anxiety and depression. Emotional dysfunction in AD is frequently modeled using tests of anxiety-like behavior in transgenic rodents. These tests often include the elevated plus-maze, light/dark test and open field test. In this review, we describe prototypical behavioral paradigms used to examine emotional dysfunction in transgenic models of AD, specifically anxiety-like behavior. Next, we summarize the results of studies examining anxiety-like behavior in transgenic rodents, noting that the behavioral outcomes using these paradigms have produced inconsistent results. We suggest that future research will benefit from using a battery of tests to examine emotional behavior in transgenic AD models. We conclude by discussing putative, overlapping neurobiological mechanisms underlying AD-related neuropathology, stress and anxiety-like behavior reported in AD models.
Collapse
Affiliation(s)
- Nathan S Pentkowski
- Department of Psychology, University of New Mexico, Albuquerque, NM, 87109, Mexico.
| | | | - Tia N Donaldson
- Department of Psychology, University of New Mexico, Albuquerque, NM, 87109, Mexico
| | - Samuel J Bouquin
- Department of Psychology, University of New Mexico, Albuquerque, NM, 87109, Mexico
| | - Benjamin J Clark
- Department of Psychology, University of New Mexico, Albuquerque, NM, 87109, Mexico.
| |
Collapse
|
32
|
Elahi M, Motoi Y, Shimonaka S, Ishida Y, Hioki H, Takanashi M, Ishiguro K, Imai Y, Hattori N. High-fat diet-induced activation of SGK1 promotes Alzheimer's disease-associated tau pathology. Hum Mol Genet 2021; 30:1693-1710. [PMID: 33890983 PMCID: PMC8411983 DOI: 10.1093/hmg/ddab115] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/31/2021] [Accepted: 04/18/2021] [Indexed: 12/21/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) has long been considered a risk factor for Alzheimer’s disease (AD). However, the molecular links between T2DM and AD remain obscure. Here, we reported that serum-/glucocorticoid-regulated kinase 1 (SGK1) is activated by administering a chronic high-fat diet (HFD), which increases the risk of T2DM, and thus promotes Tau pathology via the phosphorylation of tau at Ser214 and the activation of a key tau kinase, namely, GSK-3ß, forming SGK1-GSK-3ß-tau complex. SGK1 was activated under conditions of elevated glucocorticoid and hyperglycemia associated with HFD, but not of fatty acid–mediated insulin resistance. Elevated expression of SGK1 in the mouse hippocampus led to neurodegeneration and impairments in learning and memory. Upregulation and activation of SGK1, SGK1-GSK-3ß-tau complex were also observed in the hippocampi of AD cases. Our results suggest that SGK1 is a key modifier of tau pathology in AD, linking AD to corticosteroid effects and T2DM.
Collapse
Affiliation(s)
- Montasir Elahi
- Department of Diagnosis, Prevention and Treatment of Dementia, Juntendo University Graduate of Medicine, Tokyo, Japan
- Department of Neurology, Juntendo University Graduate of Medicine, Tokyo, Japan
- Department of Research for Parkinson's Disease, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yumiko Motoi
- Department of Diagnosis, Prevention and Treatment of Dementia, Juntendo University Graduate of Medicine, Tokyo, Japan
- Department of Neurology, Juntendo University Graduate of Medicine, Tokyo, Japan
| | - Shotaro Shimonaka
- Department of Diagnosis, Prevention and Treatment of Dementia, Juntendo University Graduate of Medicine, Tokyo, Japan
| | - Yoko Ishida
- Department of Cell Biology and Neuroscience, Juntendo University Graduate of Medicine, Tokyo, Japan
| | - Hiroyuki Hioki
- Department of Cell Biology and Neuroscience, Juntendo University Graduate of Medicine, Tokyo, Japan
| | - Masashi Takanashi
- Department of Neurology, Juntendo University Graduate of Medicine, Tokyo, Japan
| | - Koichi Ishiguro
- Department of Neurology, Juntendo University Graduate of Medicine, Tokyo, Japan
| | - Yuzuru Imai
- Department of Neurology, Juntendo University Graduate of Medicine, Tokyo, Japan
- Department of Research for Parkinson's Disease, Juntendo University Graduate School of Medicine, Tokyo, Japan
- To whom correspondence should be addressed. Tel: +81 368018332; Fax: +81 358000547;
| | - Nobutaka Hattori
- Department of Diagnosis, Prevention and Treatment of Dementia, Juntendo University Graduate of Medicine, Tokyo, Japan
- Department of Neurology, Juntendo University Graduate of Medicine, Tokyo, Japan
- Department of Research for Parkinson's Disease, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
33
|
Zhao C, Deng Y, He Y, Huang X, Wang C, Li W. Decreased Level of Exosomal miR-5121 Released from Microglia Suppresses Neurite Outgrowth and Synapse Recovery of Neurons Following Traumatic Brain Injury. Neurotherapeutics 2021; 18:1273-1294. [PMID: 33475953 PMCID: PMC8423926 DOI: 10.1007/s13311-020-00999-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2020] [Indexed: 10/22/2022] Open
Abstract
Activated microglia can suppress neurite outgrowth and synapse recovery in the acute stage following traumatic brain injury (TBI). However, the underlying mechanism has not been clearly elucidated. Exosomes derived from microglia have been reported to play a critical role in microglia-neuron interaction in healthy and pathological brains. Here, we aimed to investigate the role of microglia-derived exosomes in regulating neurite outgrowth and synapse recovery following TBI. In our study, exosomes derived from microglia were co-cultured with stretch-injured neurons in vitro and intravenously injected into mice that underwent fluid percussion injury (FPI) by tail vein injection in vivo. The results showed that microglia-derived exosomes could be absorbed by neurons in vitro and in vivo. Moreover, exosomes derived from stretch-injured microglia decreased the protein levels of GAP43, PSD-95, GluR1, and Synaptophysin and dendritic complexity in stretch-injured neurons in vitro, and reduced GAP43+ NEUN cell percentage and apical dendritic spine density in the pericontusion region in vivo. Motor coordination was also impaired in mice treated with stretch-injured microglia-derived exosomes after FPI. A microRNA microarray showed that the level of miR-5121 was decreased most greatly in exosomes derived from stretch-injured microglia. Overexpression of miR-5121 in stretch-injured microglia-derived exosomes partly reversed the suppression of neurite outgrowth and synapse recovery of neurons both in vitro and in vivo. Moreover, motor coordination in miR-5121 overexpressed exosomes treated mice was significantly improved after FPI. Following mechanistic study demonstrated that miR-5121 might promote neurite outgrowth and synapse recovery by directly targeting RGMa. In conclusion, our finding revealed a novel exosome-mediated mechanism of microglia-neuron interaction that suppressed neurite outgrowth and synapse recovery of neurons following TBI.
Collapse
Affiliation(s)
- Chengcheng Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, 3002 Sungang Road, Shenzhen, Guangdong, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| | - Yuefei Deng
- Department of Neurosurgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yi He
- Department of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, 3002 Sungang Road, Shenzhen, Guangdong, China
| | - Xianjian Huang
- Department of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, 3002 Sungang Road, Shenzhen, Guangdong, China
| | - Chuanfang Wang
- Department of Neurosurgery and Neurosurgical Disease Research Centre, The Second Affiliated Hospital of Guangzhou Medical University, 250 Changgang East Road, Guangzhou, Guangdong, China.
| | - Weiping Li
- Department of Neurosurgery, The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, 3002 Sungang Road, Shenzhen, Guangdong, China.
| |
Collapse
|
34
|
Locci A, Orellana H, Rodriguez G, Gottliebson M, McClarty B, Dominguez S, Keszycki R, Dong H. Comparison of memory, affective behavior, and neuropathology in APP NLGF knock-in mice to 5xFAD and APP/PS1 mice. Behav Brain Res 2021; 404:113192. [PMID: 33607163 DOI: 10.1016/j.bbr.2021.113192] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Revised: 01/26/2021] [Accepted: 02/14/2021] [Indexed: 02/08/2023]
Abstract
Transgenic mouse models of Aβ amyloidosis generated by knock-in of a humanized Aβ sequence can offer some advantages over the transgenic models that overexpress amyloid precursor protein (APP). However, systematic comparison of memory, behavioral, and neuropathological phenotypes between these models has not been well documented. In this study, we compared memory and affective behavior in APPNLGF mice, an APP knock-in model, to two widely used mouse models of Alzheimer's disease, 5xFAD and APP/PS1 mice, at 10 months of age. We found that, despite similar deficits in working memory, object recognition, and social recognition memory, APPNLGF and 5xFAD mice but not APP/PS1 mice show compelling anxiety- and depressive-like behavior, and exhibited a marked impairment of social interaction. We quantified corticolimbic Aβ plaques, which were lowest in APPNLGF, intermediate in APP/PS1, and highest in 5xFAD mice. Interestingly, analysis of plaque size revealed that plaques were largest in APP/PS1 mice, intermediate in 5xFAD mice, and smallest in APPNLGF mice. Finally, we observed a significantly higher percentage of the area occupied by plaques in both 5xFAD and APP/PS1 relative to APPNLGF mice. Overall, our findings suggest that the severity of Aβ neuropathology is not directly correlated with memory and affective behavior impairments between these three transgenic mouse models. Additionally, APPNLGF may represent a valid mouse model for studying AD comorbid with anxiety and depression.
Collapse
Affiliation(s)
- Andrea Locci
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Hector Orellana
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Guadalupe Rodriguez
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Meredith Gottliebson
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Bryan McClarty
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Sky Dominguez
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Rachel Keszycki
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Hongxin Dong
- Department of Psychiatry and Behavioral Sciences, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
35
|
Zhao J, Shi Q, Tian H, Li Y, Liu Y, Xu Z, Robert A, Liu Q, Meunier B. TDMQ20, a Specific Copper Chelator, Reduces Memory Impairments in Alzheimer's Disease Mouse Models. ACS Chem Neurosci 2021; 12:140-149. [PMID: 33322892 DOI: 10.1021/acschemneuro.0c00621] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Besides targeting amyloid or tau metabolisms, regulation of redox metal ions is a recognized therapeutic target for Alzheimer's disease (AD). Based on the bioinorganic chemistry of copper, we designed specific chelators of copper(II) (TDMQs) insight to regulate copper homeostasis in the brain and to inhibit the deleterious oxidative stress catalyzed by copper-amyloid complexes. An oral treatment by TDMQ20 was able to fully reverse the cognitive and behavioral impairment in three different murine models, two nontransgenic models mimicking the early stage of AD and a transgenic model representing a more advanced stage of AD. To our knowledge, such a comparative study using the same molecule has never been performed. Regular C57BL/6 mice received a single injection of human Cu-Aβ1-42 in the lateral ventricles (icv-CuAβ) or in the hippocampus (hippo-CuAβ). In both cases, mice developed a cognitive impairment similar to that of transgenic 5XFAD mice. Oral administration of TDMQ20 to icv-CuAβ or hippo-CuAβ mice within a 16-day period resulted in a significant improvement of the cognitive status. The 3-month treatment of transgenic 5XFAD mice with TDMQ20 also resulted in behavioral improvements. The consistent positive pharmacological results obtained using these different AD models correlate well with previously obtained physicochemical data of TDMQ20. The short-term novel object recognition (NOR) test was found particularly relevant to evaluate the rescue of declarative memory impairment. TDMQ20 was also able to reduce the oxidative stress in the mouse cortex. Due to its reliability and facile use, the hippo-CuAβ model can be considered as a robust nontransgenic model to evaluate the activity of potential drugs on the early stages of memory deficits.
Collapse
Affiliation(s)
- Jie Zhao
- College of Life Sciences & Oceanography, Shenzhen University, 1066 Xueyuan Boulevard, Nanshan District, Shenzhen 518055, P. R. China
- College of Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
| | - Qihui Shi
- College of Life Sciences & Oceanography, Shenzhen University, 1066 Xueyuan Boulevard, Nanshan District, Shenzhen 518055, P. R. China
| | - Hongda Tian
- School of Chemical Engineering and Light Industry, Guangdong University of Technology (GDUT), Higher Education Mega Center, 100 Waihuan Xi Road, Panyu District, Guangzhou 510006, P. R. China
| | - Youzhi Li
- School of Chemical Engineering and Light Industry, Guangdong University of Technology (GDUT), Higher Education Mega Center, 100 Waihuan Xi Road, Panyu District, Guangzhou 510006, P. R. China
| | - Yan Liu
- School of Chemical Engineering and Light Industry, Guangdong University of Technology (GDUT), Higher Education Mega Center, 100 Waihuan Xi Road, Panyu District, Guangzhou 510006, P. R. China
| | - Zhen Xu
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Anne Robert
- Laboratoire de Chimie de Coordination du CNRS (LCC-CNRS), 205 route de Narbonne, 31077 Toulouse, cedex 4, France
| | - Qiong Liu
- College of Life Sciences & Oceanography, Shenzhen University, 1066 Xueyuan Boulevard, Nanshan District, Shenzhen 518055, P. R. China
| | - Bernard Meunier
- School of Chemical Engineering and Light Industry, Guangdong University of Technology (GDUT), Higher Education Mega Center, 100 Waihuan Xi Road, Panyu District, Guangzhou 510006, P. R. China
- Laboratoire de Chimie de Coordination du CNRS (LCC-CNRS), 205 route de Narbonne, 31077 Toulouse, cedex 4, France
| |
Collapse
|
36
|
Cognitive Impairment in the 3xTg-AD Mouse Model of Alzheimer's Disease is Affected by Aβ-ImmunoTherapy and Cognitive Stimulation. Pharmaceutics 2020; 12:pharmaceutics12100944. [PMID: 33023109 PMCID: PMC7601886 DOI: 10.3390/pharmaceutics12100944] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 09/19/2020] [Accepted: 09/30/2020] [Indexed: 12/11/2022] Open
Abstract
Clinical symptoms of Alzheimer’s Disease (AD) include behavioral alterations and cognitive impairment. These functional phenotypes early occur in triple-transgenic (3xTg-AD) mice. Specifically, behavioral alterations are first detected when mice are at around 2.5 months old and cognitive impairment in between 3- and 5-month-old mice. In this work, the effect of chronic Aβ-immunotherapy on behavioral and cognitive abilities was tested by monthly administering the antibody fragment scFv-h3D6 to 3xTg-AD female mice from 5 to 9 months of age. An untreated group was used as a reference, as well as to attain some information on the effect of training during the longitudinal study. Behavioral and psychological symptoms of dementia (BPSD)-like symptoms were already evident in 5-month-old mice, in the form of neophobia and anxious-like behavior. The exploratory activity decreased over the longitudinal study, not only for 3xTgAD mice but also for the corresponding non-transgenic mice (NTg). Learning abilities of 3xTg-AD mice were not seriously compromised but an impairment in long-term spatial memory was evident at 5 months of age. Interestingly, scFv-h3D6-treatment affected the cognitive impairment displayed by 5-month-old 3xTg-AD mice. It is worth noting that training also reduced cognitive impairment of 3xTg-AD mice over the longitudinal study, suggesting that to properly quantify the isolated therapeutic potential of any drug on cognition using this model it is convenient to perform a prompt, age-matched study rather than a longitudinal study. In addition, a combination of both training and Aβ-immunotherapy could constitute a possible approach to treat Alzheimer’s disease.
Collapse
|
37
|
Johnson ECB, Ho K, Yu GQ, Das M, Sanchez PE, Djukic B, Lopez I, Yu X, Gill M, Zhang W, Paz JT, Palop JJ, Mucke L. Behavioral and neural network abnormalities in human APP transgenic mice resemble those of App knock-in mice and are modulated by familial Alzheimer's disease mutations but not by inhibition of BACE1. Mol Neurodegener 2020; 15:53. [PMID: 32921309 PMCID: PMC7489007 DOI: 10.1186/s13024-020-00393-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 07/08/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is the most frequent and costly neurodegenerative disorder. Although diverse lines of evidence suggest that the amyloid precursor protein (APP) is involved in its causation, the precise mechanisms remain unknown and no treatments are available to prevent or halt the disease. A favorite hypothesis has been that APP contributes to AD pathogenesis through the cerebral accumulation of the amyloid-β peptide (Aβ), which is derived from APP through sequential proteolytic cleavage by BACE1 and γ-secretase. However, inhibitors of these enzymes have failed in clinical trials despite clear evidence for target engagement. METHODS To further elucidate the roles of APP and its metabolites in AD pathogenesis, we analyzed transgenic mice overexpressing wildtype human APP (hAPP) or hAPP carrying mutations that cause autosomal dominant familial AD (FAD), as well as App knock-in mice that do not overexpress hAPP but have two mouse App alleles with FAD mutations and a humanized Aβ sequence. RESULTS Although these lines of mice had marked differences in cortical and hippocampal levels of APP, APP C-terminal fragments, soluble Aβ, Aβ oligomers and age-dependent amyloid deposition, they all developed cognitive deficits as well as non-convulsive epileptiform activity, a type of network dysfunction that also occurs in a substantive proportion of humans with AD. Pharmacological inhibition of BACE1 effectively reduced levels of amyloidogenic APP C-terminal fragments (C99), soluble Aβ, Aβ oligomers, and amyloid deposits in transgenic mice expressing FAD-mutant hAPP, but did not improve their network dysfunction and behavioral abnormalities, even when initiated at early stages before amyloid deposits were detectable. CONCLUSIONS hAPP transgenic and App knock-in mice develop similar pathophysiological alterations. APP and its metabolites contribute to AD-related functional alterations through complex combinatorial mechanisms that may be difficult to block with BACE inhibitors and, possibly, also with other anti-Aβ treatments.
Collapse
Affiliation(s)
- Erik C. B. Johnson
- Gladstone Institute of Neurological Disease, 1650 Owens Street, San Francisco, CA 94158 USA
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158 USA
| | - Kaitlyn Ho
- Gladstone Institute of Neurological Disease, 1650 Owens Street, San Francisco, CA 94158 USA
| | - Gui-Qiu Yu
- Gladstone Institute of Neurological Disease, 1650 Owens Street, San Francisco, CA 94158 USA
| | - Melanie Das
- Gladstone Institute of Neurological Disease, 1650 Owens Street, San Francisco, CA 94158 USA
| | - Pascal E. Sanchez
- Gladstone Institute of Neurological Disease, 1650 Owens Street, San Francisco, CA 94158 USA
| | - Biljana Djukic
- Gladstone Institute of Neurological Disease, 1650 Owens Street, San Francisco, CA 94158 USA
| | - Isabel Lopez
- Gladstone Institute of Neurological Disease, 1650 Owens Street, San Francisco, CA 94158 USA
| | - Xinxing Yu
- Gladstone Institute of Neurological Disease, 1650 Owens Street, San Francisco, CA 94158 USA
| | - Michael Gill
- Gladstone Institute of Neurological Disease, 1650 Owens Street, San Francisco, CA 94158 USA
| | - Weiping Zhang
- NHC Key Laboratory of Hormones and Development, Tianjin Institute of Endocrinology, Tianjin Medical University Metabolic Diseases Hospital, Tianjin, China
| | - Jeanne T. Paz
- Gladstone Institute of Neurological Disease, 1650 Owens Street, San Francisco, CA 94158 USA
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158 USA
| | - Jorge J. Palop
- Gladstone Institute of Neurological Disease, 1650 Owens Street, San Francisco, CA 94158 USA
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158 USA
| | - Lennart Mucke
- Gladstone Institute of Neurological Disease, 1650 Owens Street, San Francisco, CA 94158 USA
- Department of Neurology and Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA 94158 USA
| |
Collapse
|
38
|
Lychee seed polyphenol inhibits Aβ-induced activation of NLRP3 inflammasome via the LRP1/AMPK mediated autophagy induction. Biomed Pharmacother 2020; 130:110575. [PMID: 32768883 DOI: 10.1016/j.biopha.2020.110575] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 07/14/2020] [Accepted: 07/26/2020] [Indexed: 02/07/2023] Open
Abstract
Emerging evidence indicates that the enhancement of microglial autophagy inhibits the NLRP3 inflammasome mediated neuroinflammation in Alzheimer's disease (AD). Meanwhile, low density lipoprotein receptor-related protein 1 (LRP1) highly expressed in microglia is able to negatively regulate neuroinflammation and positively regulate autophagy. In addition, we have previously reported that an active lychee seed fraction enriching polyphenol (LSP) exhibits anti-neuroinflammation in Aβ-induced BV-2 cells. However, its molecular mechanism of action is still unclear. In this study, we aim to investigate whether LSP inhibits the NLRP3 inflammasome mediated neuroinflammation and clarify its molecular mechanism in Aβ-induced BV-2 cells and APP/PS1 mice. The results showed that LSP dose- and time-dependently activated autophagy by increasing the expression of Beclin 1 and LC3II in BV-2 cells, which was regulated by the upregulation of LRP1 and its mediated AMPK signaling pathway. In addition, both the Western blotting and fluorescence microscopic results demonstrated that LSP could significantly suppress the activation of NLRP3 inflammasome by inhibiting the expression of NLRP3, ASC, the cleavage of caspase-1, and the release of IL-1β in Aβ(1-42)-induced BV-2 cells. In addition, the siRNA LRP1 successfully abolished the effect of LSP on the activation of AMPK and its mediated autophagy, as well as the inhibition of NLRP3 inflammasome. Furthermore, LSP rescued PC-12 cells which were induced by the conditioned medium from Aβ(1-42)-treated BV-2 cells. Moreover, LSP improved the cognitive function and inhibited the NLRP3 inflammasome in APP/PS1 mice. Taken together, LSP inhibited the NLRP3 inflammasome-mediated neuroinflammation in the in vitro and in vivo models of AD, which was closely associated with the LRP1/AMPK-mediated autophagy. Thus, the findings from this study further provide evidences for LSP serving as a potential drug for the treatment of AD in the future.
Collapse
|
39
|
Lee J, Kong E, Hong S, Moon J, Kim P. In vivo longitudinal visualization of the brain neuroinflammatory response at the cellular level in LysM-GFP mice induced by 3-nitropropionic acid. BIOMEDICAL OPTICS EXPRESS 2020; 11:4835-4847. [PMID: 32923081 PMCID: PMC7449715 DOI: 10.1364/boe.393690] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 06/14/2020] [Accepted: 07/21/2020] [Indexed: 05/18/2023]
Abstract
Blood-brain barrier (BBB) dysfunction is related to the development of neuroinflammation in the central nervous system (CNS). Neuroinflammation has been implicated as one of the key factors in the pathogenesis of neurodegenerative diseases such as Alzheimer's disease, Huntington's disease, and Parkinson's disease. Despite its importance, the impacts and underlying cellular mechanisms of chronic BBB impairment in neurodegenerative diseases are poorly understood. In this work, we performed a longitudinal intravital brain imaging of mouse model with neuroinflammation induced by 3-nitropropionic acid (3-NP). For this, we obtained a transgenic LysM-GFP mouse expressing the green fluorescence protein (GFP) in a subset of leukocytes. By using intravenously injected fluorescence blood tracers, we longitudinally observed in vivo dynamic cellular behaviors and the BBB integrity through a 30-day neuroinflammatory state. Vascular leakages in the cerebral cortex reflecting BBB impairment were observed at two weeks, which persisted to the third week, followed by a severe inflammatory response with massive leukocytes infiltration at day 30. These descriptions can help in the development of novel approaches to treat neurodegenerative conditions.
Collapse
Affiliation(s)
- Jingu Lee
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, South Korea
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, South Korea
| | - Eunji Kong
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, South Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Deahak-ro, Yuseong-gu, Daejeon 34141, South Korea
| | - Sujung Hong
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, South Korea
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, South Korea
| | - Jieun Moon
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, South Korea
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, South Korea
| | - Pilhan Kim
- Graduate School of Nanoscience and Technology, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, South Korea
- KI for Health Science and Technology (KIHST), Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, South Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Deahak-ro, Yuseong-gu, Daejeon 34141, South Korea
| |
Collapse
|
40
|
Luo Y, Yang W, Li N, Yang X, Zhu B, Wang C, Hou W, Wang X, Wen H, Tian X. Anodal Transcranial Direct Current Stimulation Can Improve Spatial Learning and Memory and Attenuate Aβ 42 Burden at the Early Stage of Alzheimer's Disease in APP/PS1 Transgenic Mice. Front Aging Neurosci 2020; 12:134. [PMID: 32595486 PMCID: PMC7239315 DOI: 10.3389/fnagi.2020.00134] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 04/21/2020] [Indexed: 12/18/2022] Open
Abstract
Alzheimer’s disease (AD) is an irreversible progressive neurodegenerative disease. Intervention in the early stage of AD is a new path for AD treatment that is being explored. The behavioral and pathological effects of anodal transcranial direct current stimulation (AtDCS) at the early stage of AD in the mouse model, amyloid precursor protein (APP)/presenilin-1 (PS1) transgenic mice, were investigated based on our previous studies. Thirty-three 6-month-old male APP/PS1 mice were randomly divided into the model group (AD group), model + sham stimulation group (ADST group) and stimulation group (ADT group). Eleven 6-month-old male C57 wild-type mice were randomly selected as a control group (CTL group). The ADT group received 10 AtDCS sessions. The Morris water maze (MWM) task and novel object recognition (NOR) task were used to test mouse memory. Nissl staining, Western blot (WB), immunohistochemistry and immunofluorescence staining of β-amyloid (Aβ42), glial fibrillary acidic protein (GFAP) and NF200 were conducted for pathological analysis. The ADT group and the CTL group had a shorter escape latency and more platform-region crossings than the AD group and ADST group in the MWM. There was no significant difference in the discrimination index among the groups in the NOR task. Pathological analysis showed visible differences between the AD group and ADT group. This study revealed that early-stage APP/PS1 transgenic mice did not show recognition memory impairment. AtDCS effectively improved spatial learning and memory in the early-stage APP/PS1 transgenic mouse model of AD, alleviating Aβ burden and having a protective effect on neurons. AtDCS could improve AD-related symptoms by activating many glial cells to promote the degradation and clearance of Aβ or directly affecting production and degradation of Aβ to reduce glial activation. AtDCS is an effective means of early intervention in the early stage of AD.
Collapse
Affiliation(s)
- Yinpei Luo
- Chongqing Engineering Research Center for Medical Electronics Technology, Bioengineering College, Chongqing University, Chongqing, China
| | - Wenjuan Yang
- Chongqing Engineering Research Center for Medical Electronics Technology, Bioengineering College, Chongqing University, Chongqing, China
| | - Nian Li
- Chongqing Engineering Research Center for Medical Electronics Technology, Bioengineering College, Chongqing University, Chongqing, China
| | - Xiufang Yang
- Chongqing Engineering Research Center for Medical Electronics Technology, Bioengineering College, Chongqing University, Chongqing, China
| | - Binglian Zhu
- College of Microelectronics and Communication Engineering, Chongqing University, Chongqing, China
| | - Cong Wang
- Chongqing Engineering Research Center for Medical Electronics Technology, Bioengineering College, Chongqing University, Chongqing, China
| | - Wensheng Hou
- Chongqing Engineering Research Center for Medical Electronics Technology, Bioengineering College, Chongqing University, Chongqing, China
| | - Xing Wang
- Chongqing Engineering Research Center for Medical Electronics Technology, Bioengineering College, Chongqing University, Chongqing, China
| | - Huizhong Wen
- Department of Neurobiology, College of Basic Medical Science, Chongqing Key Laboratory of Neurobiology, Army Medical University, Chongqing, China
| | - Xuelong Tian
- Chongqing Engineering Research Center for Medical Electronics Technology, Bioengineering College, Chongqing University, Chongqing, China
| |
Collapse
|
41
|
Kosel F, Pelley JMS, Franklin TB. Behavioural and psychological symptoms of dementia in mouse models of Alzheimer's disease-related pathology. Neurosci Biobehav Rev 2020; 112:634-647. [PMID: 32070692 DOI: 10.1016/j.neubiorev.2020.02.012] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 02/08/2020] [Accepted: 02/11/2020] [Indexed: 12/12/2022]
Abstract
Transgenic mouse models have been used extensively to model the cognitive impairments arising from Alzheimer's disease (AD)-related pathology. However, less is known about the relationship between AD-related pathology and the behavioural and psychological symptoms of dementia (BPSD) commonly presented by patients. This review discusses the BPSD-like behaviours recapitulated by several mouse models of AD-related pathology, including the APP/PS1, Tg2576, 3xTg-AD, 5xFAD, and APP23 models. Current evidence suggests that social withdrawal and depressive-like behaviours increase with progressive neuropathology, and increased aggression and sleep-wake disturbances are present even at early stages; however, there is no clear evidence to support increased anxiety-like behaviours, agitation (hyperactivity), or general apathy. Overall, transgenic mouse models of AD-related pathology recapitulate some of the BPSD-like behaviours associated with AD, but these behaviours vary by model. This reflects the patient population, where AD patients typically exhibit one or more BPSD, but rarely all symptoms at once. As a result, we suggest that transgenic mouse models are an important tool to investigate the pathology underlying BPSD in human AD patients.
Collapse
Affiliation(s)
- Filip Kosel
- The Social Lab, Department of Psychology and Neuroscience, Dalhousie University, Halifax, B3H 4R2, Canada
| | - Jessica M S Pelley
- The Social Lab, Department of Psychology and Neuroscience, Dalhousie University, Halifax, B3H 4R2, Canada
| | - Tamara B Franklin
- The Social Lab, Department of Psychology and Neuroscience, Dalhousie University, Halifax, B3H 4R2, Canada.
| |
Collapse
|
42
|
He Y, Li Y, Zhou F, Qi J, Wu M. Decreased circadian fluctuation in cognitive behaviors and synaptic plasticity in APP/PS1 transgenic mice. Metab Brain Dis 2020; 35:343-352. [PMID: 31879834 DOI: 10.1007/s11011-019-00531-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 12/16/2019] [Indexed: 12/28/2022]
Abstract
Cognitive decline, memory impairment and circadian rhythm disturbance are iconic manifestations of Alzheimer's disease (AD). APPswe/PS1dE9 (APP/PS1) mice, a model of AD, show deficits in multiple learning and memory abilities, synaptic plasticity, and behavioral circadian rhythm, but whether circadian differences in cognitive performance and synaptic plasticity could be affected in AD remain unclear. Here, the cognitive behaviors of 6-month-old APP/PS1 mice were assessed by multiple behavior tests in the rest phase (light period) or active phase (dark period) of the day. The possible electrophysiological mechanism was subsequently investigated by in vivo hippocampal long-term potentiation (LTP) recording, and the locomotor activity rhythm of the mice was detected using wheel-running activities. Compared to wild-type (WT) mice, APP/PS1 mice exhibited long-term spatial memory impairment and in vivo hippocampal LTP suppression. In addition, in APP/PS1 mice, circadian differences in new object recognition memory and LTP were lost, and the circadian difference in long-term spatial memory was decreased, accompanied by a less robust locomotor activity rhythm. These results indicate that the loss of circadian differences in new object recognition memory and the decrease in the circadian difference in long-term spatial memory in APP/PS1 mice, which are closely associated with the loss of the circadian difference in LTP and less robust locomotor activity, might occur early in the course of AD.
Collapse
Affiliation(s)
- Yexin He
- Department of Radiology, Affiliated Provincial People's Hospital of Shanxi Medical University, Shanxi Provincial People's Hospital, Taiyuan, 030012, China
| | - Yiying Li
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, China
| | - Fang Zhou
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, China
| | - Jinshun Qi
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, China
| | - Meina Wu
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan, 030001, China.
| |
Collapse
|
43
|
Altered dorsal CA1 neuronal population coding in the APP/PS1 mouse model of Alzheimer's disease. Sci Rep 2020; 10:1077. [PMID: 31974405 PMCID: PMC6978514 DOI: 10.1038/s41598-020-58038-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 01/09/2020] [Indexed: 12/27/2022] Open
Abstract
While the link between amyloid β (Aβ) accumulation and synaptic degradation in Alzheimer’s disease (AD) is known, the consequences of this pathology on population coding remain unknown. We found that the entropy, a measure of the diversity of network firing patterns, was lower in the dorsal CA1 region in the APP/PS1 mouse model of Aβ pathology, relative to controls, thereby reducing the population’s coding capacity. Our results reveal a network level signature of the deficits Aβ accumulation causes to the computations performed by neural circuits.
Collapse
|
44
|
Chen Y, Chou T, Lin I, Chen C, Kao C, Huang G, Chen L, Wang P, Lin C, Tsai T. Upregulation of Cisd2 attenuates Alzheimer's-related neuronal loss in mice. J Pathol 2020; 250:299-311. [PMID: 31837018 PMCID: PMC7065100 DOI: 10.1002/path.5374] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 12/01/2019] [Accepted: 12/04/2019] [Indexed: 12/24/2022]
Abstract
CDGSH iron-sulfur domain-containing protein 2 (Cisd2), a protein that declines in an age-dependent manner, mediates lifespan in mammals. Cisd2 deficiency causes accelerated aging and shortened lifespan, whereas persistent expression of Cisd2 promotes longevity in mice. Alzheimer's disease (AD) is the most prevalent form of senile dementia and is without an effective therapeutic strategy. We investigated whether Cisd2 upregulation is able to ameliorate amyloid β (Aβ) toxicity and prevent neuronal loss using an AD mouse model. Our study makes three major discoveries. First, using the AD mouse model (APP/PS1 double transgenic mice), the dosage of Cisd2 appears to modulate the severity of AD phenotypes. Cisd2 overexpression (∼two-fold) significantly promoted survival and alleviated the pathological defects associated with AD. Conversely, Cisd2 deficiency accelerated AD pathogenesis. Secondly, Cisd2 overexpression protected against Aβ-mediated mitochondrial damage and attenuated loss of neurons and neuronal progenitor cells. Finally, an increase in Cisd2 shifted the expression profiles of a panel of genes that are dysregulated by AD toward the patterns observed in wild-type mice. These findings highlight Cisd2-based therapies as a potential disease-modifying strategy for AD. © 2019 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Yi‐Fan Chen
- The Ph.D. Program for Translational Medicine, College of Medical Science and TechnologyTaipei Medical UniversityTaipeiTaiwan
| | - Tzu‐Yu Chou
- Department of Life Sciences and Institute of Genome SciencesNational Yang‐Ming UniversityTaipeiTaiwan
| | - I‐Hsuan Lin
- TMU Research Center of Cancer Translational MedicineTaipei Medical UniversityTaipeiTaiwan
| | - Chung‐Guang Chen
- Department of Biomedical Imaging and Radiological SciencesNational Yang‐Ming UniversityTaipeiTaiwan
| | - Cheng‐Heng Kao
- Center of General EducationChang Gung UniversityTaoyuanTaiwan
| | - Guo‐Jen Huang
- Department of Biomedical ScienceChang Gung UniversityTaoyuanTaiwan
| | - Liang‐Kung Chen
- Aging and Health Research CenterNational Yang‐Ming UniversityTaipeiTaiwan
- Department of Geriatric MedicineNational Yang‐Ming UniversityTaipeiTaiwan
- Center for Geriatrics and GerontologyNeurological Institute, Taipei Veterans General HospitalTaipeiTaiwan
| | - Pei‐Ning Wang
- Aging and Health Research CenterNational Yang‐Ming UniversityTaipeiTaiwan
- Brain Research CenterNational Yang‐Ming UniversityTaipeiTaiwan
- Department of NeurologyNeurological Institute, Taipei Veterans General HospitalTaipeiTaiwan
| | - Ching‐Po Lin
- Aging and Health Research CenterNational Yang‐Ming UniversityTaipeiTaiwan
- Brain Research CenterNational Yang‐Ming UniversityTaipeiTaiwan
- Institute of NeuroscienceNational Yang‐Ming UniversityTaipeiTaiwan
| | - Ting‐Fen Tsai
- Department of Life Sciences and Institute of Genome SciencesNational Yang‐Ming UniversityTaipeiTaiwan
- Aging and Health Research CenterNational Yang‐Ming UniversityTaipeiTaiwan
- Institute of Molecular and Genomic MedicineNational Health Research InstitutesZhunanTaiwan
- Institute of Biotechnology and Pharmaceutical ResearchNational Health Research InstitutesZhunanTaiwan
| |
Collapse
|
45
|
Davtyan H, Hovakimyan A, Kiani Shabestari S, Antonyan T, Coburn MA, Zagorski K, Chailyan G, Petrushina I, Svystun O, Danhash E, Petrovsky N, Cribbs DH, Agadjanyan MG, Blurton-Jones M, Ghochikyan A. Testing a MultiTEP-based combination vaccine to reduce Aβ and tau pathology in Tau22/5xFAD bigenic mice. Alzheimers Res Ther 2019; 11:107. [PMID: 31847886 PMCID: PMC6918571 DOI: 10.1186/s13195-019-0556-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 11/11/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND Alzheimer disease (AD) is characterized by the accumulation of beta-amyloid (Aβ) plaques and neurofibrillary tangles composed of hyperphosphorylated tau, which together lead to neurodegeneration and cognitive decline. Current therapeutic approaches have primarily aimed to reduce pathological aggregates of either Aβ or tau, yet phase 3 clinical trials of these approaches have thus far failed to delay disease progression in humans. Strong preclinical evidence indicates that these two abnormally aggregated proteins interact synergistically to drive downstream neurodegeneration. Therefore, combinatorial therapies that concurrently target both Aβ and tau might be needed for effective disease modification. METHODS A combinatorial vaccination approach was designed to concurrently target both Aβ and tau pathologies. Tau22/5xFAD (T5x) bigenic mice that develop both pathological Aβ and tau aggregates were injected intramuscularly with a mixture of two MultiTEP epitope vaccines: AV-1959R and AV-1980R, targeting Aβ and tau, respectively, and formulated in AdvaxCpG, a potent polysaccharide adjuvant. Antibody responses of vaccinated animals were measured by ELISA, and neuropathological changes were determined in brain homogenates of vaccinated and control mice using ELISA and Meso Scale Discovery (MSD) multiplex assays. RESULTS T5x mice immunized with a mixture of Aβ- and tau-targeting vaccines generated high Aβ- and tau-specific antibody titers that recognized senile plaques and neurofibrillary tangles/neuropil threads in human AD brain sections. Production of these antibodies in turn led to significant reductions in the levels of soluble and insoluble total tau, and hyperphosphorylated tau as well as insoluble Aβ42, within the brains of bigenic T5x mice. CONCLUSIONS AV-1959R and AV-1980R formulated with AdvaxCpG adjuvant are immunogenic and therapeutically potent vaccines that in combination can effectively reduce both of the hallmark pathologies of AD in bigenic mice. Taken together, these findings warrant further development of this vaccine technology for ultimate testing in human AD.
Collapse
Affiliation(s)
- Hayk Davtyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA USA
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA USA
| | - Armine Hovakimyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA USA
| | | | - Tatevik Antonyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA USA
| | - Morgan A. Coburn
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA USA
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA USA
| | - Karen Zagorski
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA USA
- Current address: Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE USA
| | - Gor Chailyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA USA
| | - Irina Petrushina
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA USA
| | - Olga Svystun
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA USA
| | - Emma Danhash
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA USA
- School of Biological Sciences, University of California, Irvine, Irvine, CA USA
| | | | - David H. Cribbs
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA USA
| | - Michael G. Agadjanyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA USA
| | - Mathew Blurton-Jones
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA USA
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA USA
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, CA USA
- School of Biological Sciences, University of California, Irvine, Irvine, CA USA
| | - Anahit Ghochikyan
- Department of Molecular Immunology, Institute for Molecular Medicine, Huntington Beach, CA USA
| |
Collapse
|
46
|
Zhang S, Zhang J, Wei D, An H, Liu W, Lai Y, Yang T, Shao W, Huang Y, Wang L, Dou F, Peng D, Zhang Z. Dengzhan Shengmai capsules and their active component scutellarin prevent cognitive decline in APP/PS1 mice by accelerating Aβ aggregation and reducing oligomers formation. Biomed Pharmacother 2019; 121:109682. [PMID: 31810113 DOI: 10.1016/j.biopha.2019.109682] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Revised: 11/01/2019] [Accepted: 11/16/2019] [Indexed: 12/19/2022] Open
Abstract
There is currently no effective treatment to prevent the progress of Alzheimer's disease (AD). The traditional Chinese herbs Dengzhan Shengmai (DZSM) capsules and their active component scutellarin possess multiple effects and are clinically used for the treatment of cerebrovascular diseases. Scutellarin has been reported to affect Aβ aggregation. However, the effects of DZSM capsules on AD remain unknown. Through in vivo experiments, our study proved that the alleviating effects of DZSM capsules on cognitive deficits of AD mice were due to the role of scutellarin, which up-regulated low toxic amyloid plaques and down-regulated highly toxic soluble Aβ42 and Aβ40 levels in cortex. In vitro, we confirmed scutellarin's role in accelerating transforming Aβ42 monomers into high-molecular-mass aggregates by biochemical assays, which supported the results observed in drug-treated APP/PS1 mice. In detail, the 1:10 ratio of scutellarin/Aβ42 mixtures promoted production of large β-sheet-rich fibrils whereas the 1:1 ratio promoted production of protofibrils. In addition, the binding between scutellarin and Aβ monomers was quantified by microscale thermophoresis test and the apparent dissociation constant (Kd) was 1284.4 ± 238.8 μM. What's more, binding regions between scutellarin and Aβ fibrils were predicted by computational docking models and scutellarin might bind parallel to the long axis of Aβ42 fibrils targeting hydrophobic grooves at residues 35-36 or 39. In conclusion, DZSM capsules protected against cognitive defects of AD through scutellarin-mediated acceleration of Aβ aggregation into fibrils or protofibrils and reduction of soluble Aβ oligomers, thus suggesting potential clinical applications of DZSM capsules and scutellarin in the treatment of AD.
Collapse
Affiliation(s)
- Shujuan Zhang
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, 100029, China; Department of Neurology, China-Japan Friendship Hospital, 2 Yinghuayuan East Street, Chaoyang District, Beijing 100029, China
| | - Jianxiang Zhang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, 100875 Beijing, China
| | - Dongfeng Wei
- BABRI Centre, Beijing Normal University, Beijing 100875, China; Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing 100700, China
| | - Haiting An
- State Key Laboratory of Cognitive Neuroscience and Learning and IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China; BABRI Centre, Beijing Normal University, Beijing 100875, China
| | - Wei Liu
- College of Chemistry, Beijing Normal University, Beijing, China
| | - Yihui Lai
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, 100875 Beijing, China
| | - Te Yang
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, 100875 Beijing, China
| | - Wen Shao
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, 100029, China; Department of Neurology, China-Japan Friendship Hospital, 2 Yinghuayuan East Street, Chaoyang District, Beijing 100029, China
| | - Yaping Huang
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, 100029, China; Department of Neurology, China-Japan Friendship Hospital, 2 Yinghuayuan East Street, Chaoyang District, Beijing 100029, China
| | - Lei Wang
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, 100029, China; Department of Neurology, China-Japan Friendship Hospital, 2 Yinghuayuan East Street, Chaoyang District, Beijing 100029, China
| | - Fei Dou
- Beijing Key Laboratory of Gene Resource and Molecular Development, College of Life Sciences, Beijing Normal University, 100875 Beijing, China
| | - Dantao Peng
- Peking University China-Japan Friendship School of Clinical Medicine, Beijing, 100029, China; Department of Neurology, China-Japan Friendship Hospital, 2 Yinghuayuan East Street, Chaoyang District, Beijing 100029, China.
| | - Zhanjun Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning and IDG/McGovern Institute for Brain Research, Beijing Normal University, Beijing 100875, China; BABRI Centre, Beijing Normal University, Beijing 100875, China.
| |
Collapse
|
47
|
Cox LM, Schafer MJ, Sohn J, Vincentini J, Weiner HL, Ginsberg SD, Blaser MJ. Calorie restriction slows age-related microbiota changes in an Alzheimer's disease model in female mice. Sci Rep 2019; 9:17904. [PMID: 31784610 PMCID: PMC6884494 DOI: 10.1038/s41598-019-54187-x] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Accepted: 11/09/2019] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) affects an estimated 5.8 million Americans, and advanced age is the greatest risk factor. AD patients have altered intestinal microbiota. Accordingly, depleting intestinal microbiota in AD animal models reduces amyloid-beta (Aβ) plaque deposition. Age-related changes in the microbiota contribute to immunologic and physiologic decline. Translationally relevant dietary manipulations may be an effective approach to slow microbiota changes during aging. We previously showed that calorie restriction (CR) reduced brain Aβ deposition in the well-established Tg2576 mouse model of AD. Presently, we investigated whether CR alters the microbiome during aging. We found that female Tg2576 mice have more substantial age-related microbiome changes compared to wildtype (WT) mice, including an increase in Bacteroides, which were normalized by CR. Specific gut microbiota changes were linked to Aβ levels, with greater effects in females than in males. In the gut, Tg2576 female mice had an enhanced intestinal inflammatory transcriptional profile, which was reversed by CR. Furthermore, we demonstrate that Bacteroides colonization exacerbates Aβ deposition, which may be a mechanism whereby the gut impacts AD pathogenesis. These results suggest that long-term CR may alter the gut environment and prevent the expansion of microbes that contribute to age-related cognitive decline.
Collapse
Affiliation(s)
- Laura M Cox
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA.
- Department of Medicine, NYU Langone Medical Center, New York, NY, USA.
| | - Marissa J Schafer
- Cellular and Molecular Biology Training Program, NYU Langone Medical Center, New York, NY, USA
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA
- Department of Physical Medicine and Rehabilitation and Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, US
| | - Jiho Sohn
- Department of Medicine, NYU Langone Medical Center, New York, NY, USA
- Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - Julia Vincentini
- Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Howard L Weiner
- Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Stephen D Ginsberg
- Psychiatry, Neuroscience & Physiology & the NYU Neuroscience Institute, NYU Langone Medical Center, New York, NY, USA
- Center for Dementia Research, Nathan Kline Institute, Orangeburg, NY, USA
| | - Martin J Blaser
- Department of Medicine, NYU Langone Medical Center, New York, NY, USA
- Center for Advanced Biotechnology and Medicine, Rutgers University, New Brunswick, NJ, USA
| |
Collapse
|
48
|
Giuliani A, Sivilia S, Baldassarro VA, Gusciglio M, Lorenzini L, Sannia M, Calzà L, Giardino L. Age-Related Changes of the Neurovascular Unit in the Cerebral Cortex of Alzheimer Disease Mouse Models: A Neuroanatomical and Molecular Study. J Neuropathol Exp Neurol 2019; 78:101-112. [PMID: 30629191 DOI: 10.1093/jnen/nly125] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
We describe age-related histological structure and molecular changes of the neurovascular unit (NVU) in the cerebral cortex of Tg2576 and age-matched wild-type (WT) mice. Major results can be summarized as follows: (i) β-amyloid (6E10)-immunoreactivity progressively increases in neurons and astrocytes of Tg2576 mice, reaching the highest concentration at 5 months and then decreasing as soon as extracellular plaque deposition begins; (ii) the synaptic puncta density of glutamatergic and GABAergic neurons in Tg2576 mice is unbalanced versus WT at all investigated ages, with a decrease in synaptophysin and VGLUT1; density of VGAT contacts is higher in 27-month-old Tg2576 versus WT mice; (iii) capillary density is higher in 5-month-old Tg2576 versus WT mice, then decreases to a lower density at 27 months, when the capillary-astrocyte interface is lower; and (iv) mRNA expression of genes involved in microvessel dynamics indicates age- and genotype-dependent changes in the expression levels of hypoxia-related genes, i.e. the highest level is in 5-month-old animals and there is impaired regulation in Tg2576. We conclude that at 5 months, when learning and memory impairment is already present in the absence of extracellular amyloid plaque deposition, Tg2576 mice display alterations in the structure and molecular regulation of the NVU.
Collapse
Affiliation(s)
- Alessandro Giuliani
- Department of Veterinary Medical Sciences (DIMEVET), University of Bologna, Bologna, Italy
| | | | - Vito Antonio Baldassarro
- Department of Veterinary Medical Sciences (DIMEVET), University of Bologna, Bologna, Italy.,IRET Foundation, Ozzano Emilia, Italy.,Pharmacy and Biotechnology Department (FaBiT), University of Bologna, Bologna, Italy
| | | | - Luca Lorenzini
- IRET Foundation, Ozzano Emilia, Italy.,Health Science and Technologies Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Bologna, Italy
| | - Michele Sannia
- Department of Veterinary Medical Sciences (DIMEVET), University of Bologna, Bologna, Italy.,Health Science and Technologies Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Bologna, Italy
| | - Laura Calzà
- IRET Foundation, Ozzano Emilia, Italy.,Health Science and Technologies Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Bologna, Italy.,Pharmacy and Biotechnology Department (FaBiT), University of Bologna, Bologna, Italy
| | - Luciana Giardino
- Department of Veterinary Medical Sciences (DIMEVET), University of Bologna, Bologna, Italy.,IRET Foundation, Ozzano Emilia, Italy.,Health Science and Technologies Interdepartmental Center for Industrial Research (HST-ICIR), University of Bologna, Bologna, Italy
| |
Collapse
|
49
|
Butterfield DA, Boyd-Kimball D. Redox proteomics and amyloid β-peptide: insights into Alzheimer disease. J Neurochem 2019; 151:459-487. [PMID: 30216447 PMCID: PMC6417976 DOI: 10.1111/jnc.14589] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 08/15/2018] [Accepted: 09/07/2018] [Indexed: 12/12/2022]
Abstract
Alzheimer disease (AD) is a progressive neurodegenerative disorder associated with aging and characterized pathologically by the presence of senile plaques, neurofibrillary tangles, and neurite and synapse loss. Amyloid beta-peptide (1-42) [Aβ(1-42)], a major component of senile plaques, is neurotoxic and induces oxidative stress in vitro and in vivo. Redox proteomics has been used to identify proteins oxidatively modified by Aβ(1-42) in vitro and in vivo. In this review, we discuss these proteins in the context of those identified to be oxidatively modified in animal models of AD, and human studies including familial AD, pre-clinical AD (PCAD), mild cognitive impairment (MCI), early AD, late AD, Down syndrome (DS), and DS with AD (DS/AD). These redox proteomics studies indicate that Aβ(1-42)-mediated oxidative stress occurs early in AD pathogenesis and results in altered antioxidant and cellular detoxification defenses, decreased energy yielding metabolism and mitochondrial dysfunction, excitotoxicity, loss of synaptic plasticity and cell structure, neuroinflammation, impaired protein folding and degradation, and altered signal transduction. Improved access to biomarker imaging and the identification of lifestyle interventions or treatments to reduce Aβ production could be beneficial in preventing or delaying the progression of AD. This article is part of the special issue "Proteomics".
Collapse
Affiliation(s)
- D. Allan Butterfield
- Department of Chemistry and Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40506
| | - Debra Boyd-Kimball
- Department of Chemistry and Biochemistry, University of Mount Union, Alliance, OH 44601
| |
Collapse
|
50
|
Joseph DJ, Liu C, Peng J, Liang G, Wei H. Isoflurane mediated neuropathological and cognitive impairments in the triple transgenic Alzheimer's mouse model are associated with hippocampal synaptic deficits in an age-dependent manner. PLoS One 2019; 14:e0223509. [PMID: 31600350 PMCID: PMC6786564 DOI: 10.1371/journal.pone.0223509] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 09/22/2019] [Indexed: 12/17/2022] Open
Abstract
Many in vivo studies suggest that inhalational anesthetics can accelerate or prevent the progression of neuropathology and cognitive impairments in Alzheimer Disease (AD), but the synaptic mechanisms mediating these ambiguous effects are unclear. Here, we show that repeated exposures of neonatal and old triple transgenic AD (3xTg) and non-transgenic (NonTg) mice to isoflurane (Iso) distinctly increased neurodegeneration as measured by S100β levels, intracellular Aβ, Tau oligomerization, and apoptotic markers. Spatial cognition measured by reference and working memory testing in the Morris Water Maze (MWM) were altered in young NonTg and 3xTg. Field recordings in the cornu ammonis 1 (CA1) hippocampus showed that neonatal control 3xTg mice exhibited hypo-excitable synaptic transmission, reduced paired-pulse facilitation (PPF), and normal long-term potentiation (LTP) compared to NonTg controls. By contrast, the old control 3xTg mice exhibited hyper-excitable synaptic transmission, enhanced PPF, and unstable LTP compared to NonTg controls. Repeated Iso exposures reduced synaptic transmission and PPF in neonatal NonTg and old 3xTg mice. LTP was normalized in old 3xTg mice, but reduced in neonates. By contrast, LTP was reduced in old but not neonatal NonTg mice. Our results indicate that Iso-mediated neuropathologic and cognitive defects in AD mice are associated with synaptic pathologies in an age-dependent manner. Based on these findings, the extent of this association with age and, possibly, treatment paradigms warrant further study.
Collapse
Affiliation(s)
- Donald J. Joseph
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Chunxia Liu
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
- Department of Anesthesiology, China-Japan Friendship Hospital, Beijing, China
| | - Jun Peng
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
- Department of Anesthesiology, sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Ge Liang
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
| | - Huafeng Wei
- Department of Anesthesiology and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States of America
- * E-mail:
| |
Collapse
|