1
|
Fourriere L, Gleeson PA. Organelle perturbation in Alzheimer's disease: do intracellular amyloid-ß and the fragmented Golgi mediate early intracellular neurotoxicity? Front Cell Dev Biol 2025; 13:1550211. [PMID: 40302938 PMCID: PMC12037564 DOI: 10.3389/fcell.2025.1550211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 03/17/2025] [Indexed: 05/02/2025] Open
Abstract
Alzheimer's disease is a devastating and incurable neurological disease. Most of the current research has focused on developing drugs to clear the extracellular amyloid plaques in the brain of Alzheimer's disease patients. However, this approach is limited as it does not treat the underlying cause of the disease. In this review, we highlight the evidence in the field showing that the accumulation of intracellular toxic amyloid-ß could underpin very early events in neuronal death in both familial early-onset and sporadic late-onset alzheimer's disease. Indeed, intracellular amyloid-ß, which is produced within intracellular compartments, has been shown to perturb endosomal and secretory organelles, in different neuronal models, and the brain of Alzheimer's patients, leading to membrane trafficking defects and perturbation of neuronal function associated with cognition defects. The Golgi apparatus is a central transport and signaling hub at the crossroads of the secretory and endocytic pathways and perturbation of the Golgi ribbon structure is a hallmark of Alzheimer's disease. Here, we discuss the role of the Golgi as a major player in the regulation of amyloid-β production and propose that the Golgi apparatus plays a key role in a cellular network which can seed the onset of Alzheimer's disease. Moreover, we propose that the Golgi is central in an intracellular feedback loop leading to an enhanced level of amyloid-β production resulting in early neuronal defects before the appearance of clinical symptoms. Further advances in defining the molecular pathways of this intracellular feedback loop could support the design of new therapeutic strategies to target a primary source of neuronal toxicity in this disease.
Collapse
|
2
|
Meng Y, Xu L, Cheng G. Bioelectronics hydrogels for implantable cardiac and brain disease medical treatment application. Int J Biol Macromol 2025; 299:139945. [PMID: 39837454 DOI: 10.1016/j.ijbiomac.2025.139945] [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: 10/20/2024] [Revised: 12/28/2024] [Accepted: 01/14/2025] [Indexed: 01/23/2025]
Abstract
Hydrogel-based bioelectronic systems offer significant benefits for point-of-care diagnosis, treatment of cardiac and cerebral disease, surgical procedures, and other medical applications, ushering in a new era of advancements in medical technology. Progress in hydrogel-based bioelectronics has advanced from basic instrument and sensing capabilities to sophisticated multimodal perceptions and feedback systems. Addressing challenges related to immune responses and inflammation regulation after implantation, physiological dynamic mechanism, biological toxicology as well as device size, power consumption, stability, and signal conversion is crucial for the practical implementation of hydrogel-based bioelectronics in medical implants. Therefore, further exploration of hydrogel-based bioelectronics is imperative, and a comprehensive review is necessary to steer the development of these technologies for use in implantable therapies for cardiac and brain/neural conditions. In this review, a concise overview is provided on the fundamental principles underlying ionic electronic and ionic bioelectronic mechanisms. Additionally, a comprehensive examination is conducted on various bioelectronic materials integrated within hydrogels for applications in implantable medical treatments. The analysis encompasses a detailed discussion on the representative structures and physical attributes of hydrogels. This includes an exploration of their intrinsic properties such as mechanical strength, dynamic capabilities, shape-memory features, stability, stretchability, and water retention characteristics. Moreover, the discussion extends to properties related to interactions with tissues or the environment, such as adhesiveness, responsiveness, and degradability. The intricate relationships between the structure and properties of hydrogels are thoroughly examined, along with an elucidation of how these properties influence their applications in implantable medical treatments. The review also delves into the processing techniques and characterization methods employed for hydrogels. Furthermore, recent breakthroughs in the applications of hydrogels are logically explored, covering aspects such as materials, structure, properties, functions, fabrication procedures, and hybridization with other materials. Finally, the review concludes by outlining the future prospects and challenges associated with hydrogels-based bioelectronics systems.
Collapse
Affiliation(s)
- Yanfang Meng
- School of Mechanical Engineering, Jiangsu University, No.301 Xuefu Road, Zhenjiang 212013, Jiangsu Province, China
| | - Lin Xu
- School of Mechanical Engineering, Jiangsu University, No.301 Xuefu Road, Zhenjiang 212013, Jiangsu Province, China.
| | - Guanggui Cheng
- School of Mechanical Engineering, Jiangsu University, No.301 Xuefu Road, Zhenjiang 212013, Jiangsu Province, China.
| |
Collapse
|
3
|
Yavari M, Kalupahana NS, Harris BN, Ramalingam L, Zu Y, Kahathuduwa CN, Moustaid-Moussa N. Mechanisms Linking Obesity, Insulin Resistance, and Alzheimer's Disease: Effects of Polyphenols and Omega-3 Polyunsaturated Fatty Acids. Nutrients 2025; 17:1203. [PMID: 40218960 PMCID: PMC11990358 DOI: 10.3390/nu17071203] [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: 03/05/2025] [Revised: 03/23/2025] [Accepted: 03/24/2025] [Indexed: 04/14/2025] Open
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder characterized by progressive cognitive decline, memory loss, and behavioral changes. It poses a significant global health challenge. AD is associated with the accumulation of amyloid-β (Aβ) plaques and neurofibrillary tangles (NFTs) in the brain, along with chronic inflammation, dysfunctional neurons, and synapse loss. While the prevalence of AD continues to rise, the current FDA-approved drugs offer only limited effectiveness. Emerging evidence suggests that obesity, insulin resistance (IR), and type 2 diabetes mellitus (T2DM) are also implicated in AD pathogenesis, with epidemiological studies and animal models confirming the impact of IR on Aβ accumulation, and high-fat diets also exacerbating Aβ accumulation. Since neuroinflammation activated by Aβ involves the nuclear factor kappa-light-chain-enhancer of the activated B cell (NF-κB) pathway, the inhibition of NF-κB and NLRP3 inflammasome activation are potential therapeutic strategies in AD. Bioactive compounds, including polyphenols (resveratrol, epigallocatechin-3-gallate, curcumin, and quercetin), and omega-3 polyunsaturated fatty acids, show promising results in animal studies and clinical trials for reducing Aβ levels, improving cognition and modulating the signaling pathways implicated in AD. This review explores the interplay between obesity, IR, inflammation, and AD pathology, emphasizing the potential of dietary compounds and their role in reducing inflammation, oxidative stress, and cognitive decline, as viable strategies for AD prevention and treatment. By integrating epidemiological findings, observational studies, and clinical trials, this review aims to provide a comprehensive understating of how metabolic dysfunctions and bioactive compounds influence AD progression.
Collapse
Affiliation(s)
- Mahsa Yavari
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA; (M.Y.); (L.R.); (Y.Z.)
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX 79409, USA; (B.N.H.); (C.N.K.)
| | - Nishan Sudheera Kalupahana
- Department of Nutrition and Health, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates;
| | - Breanna N. Harris
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX 79409, USA; (B.N.H.); (C.N.K.)
- Department of Biological Sciences, Texas Tech University, Lubbock, TX 79409, USA
- Institute for One Health Innovation, Offices of Research & Innovation, Texas Tech University, Texas Tech Health Sciences Center, Lubbock, TX 79409, USA
| | - Latha Ramalingam
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA; (M.Y.); (L.R.); (Y.Z.)
| | - Yujiao Zu
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA; (M.Y.); (L.R.); (Y.Z.)
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX 79409, USA; (B.N.H.); (C.N.K.)
| | - Chanaka Nadeeshan Kahathuduwa
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX 79409, USA; (B.N.H.); (C.N.K.)
- Department of Neurology, Texas Tech University Health Sciences Center, El Paso, TX 79409, USA
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX 79409, USA; (M.Y.); (L.R.); (Y.Z.)
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX 79409, USA; (B.N.H.); (C.N.K.)
- Institute for One Health Innovation, Offices of Research & Innovation, Texas Tech University, Texas Tech Health Sciences Center, Lubbock, TX 79409, USA
| |
Collapse
|
4
|
Peñalva DA, Munafó JP, Antollini SS. Cholesterol´s role in membrane organization and nicotinic acetylcholine receptor function: Implications for aging and Alzheimer's disease. Chem Phys Lipids 2025; 269:105484. [PMID: 40147619 DOI: 10.1016/j.chemphyslip.2025.105484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/25/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025]
Abstract
Biological membranes are complex entities composed of various molecules exhibiting lateral and transbilayer lipid asymmetries, along with a selective spatial distribution of different membrane proteins. This dynamic orchestration is crucial for proper physiological functions, undergoes changes with aging, and is disturbed in several neurological disorders. In this review, we analyze the impact of disruption in this equilibrium on physiological aging and the onset of pathological conditions. Alzheimer´s disease (AD) is a multifactorial neurodegenerative disorder in the elderly, characterized by the increased presence of the Aβ peptide, which supports the amyloid hypothesis of the disease. However, AD also involves a progressive loss of cholinergic innervation, leading to the cholinergic hypothesis of the disease. Nicotinic acetylcholine receptors (nAChRs) are transmembrane proteins, and Aβ peptides, their oligomeric and fibrillar species, which increase in hydrophobicity as they develop, interact with membranes. Therefore, a membrane hypothesis of the disease emerges as a bridge between the other two. Here, we discuss the impact of the membrane environment, through direct or indirect mechanisms, on cholinergic signaling and Aβ formation and subsequent incorporation into the membrane, with a special focus on the crucial role of cholesterol in these processes.
Collapse
Affiliation(s)
- Daniel A Peñalva
- Instituto de Investigaciones Bioquímicas de Bahía Blanca CONICET-UNS, Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, Bahía Blanca, Argentina
| | - Juan Pablo Munafó
- Instituto de Investigaciones Bioquímicas de Bahía Blanca CONICET-UNS, Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, Bahía Blanca, Argentina
| | - Silvia S Antollini
- Instituto de Investigaciones Bioquímicas de Bahía Blanca CONICET-UNS, Departamento de Biología, Bioquímica y Farmacia, Universidad Nacional del Sur, Bahía Blanca, Argentina.
| |
Collapse
|
5
|
Ziegler M, Böger C, Alecu JE, Kim HM, Saffari A, Davies AK, Sahin M, Ebrahimi-Fakhari D. Arrayed CRISPR/Cas9 Loss-Of-Function Screen in a Neuronal Model of Adaptor Protein Complex 4 Deficiency Identifies Modulators of ATG9A Trafficking. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.22.639634. [PMID: 40027661 PMCID: PMC11870607 DOI: 10.1101/2025.02.22.639634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Biallelic loss-of-function variants in the adaptor protein complex 4 (AP-4) disrupt trafficking of transmembrane proteins at the trans -Golgi network, including the autophagy-related protein 9A (ATG9A), leading to childhood-onset hereditary spastic paraplegia (AP-4-HSP). AP-4-HSP is characterized by features of both a neurodevelopmental and degenerative neurological disease. To investigate the molecular mechanisms underlying AP-4-HSP and identify potential therapeutic targets, we conducted an arrayed CRISPR/Cas9 loss-of-function screen of 8,478 genes, targeting the 'druggable genome', in a human neuronal model of AP-4 deficiency. Through this phenotypic screen and subsequent experiments, key modulators of ATG9A trafficking were identified, and complementary pathway analyses provided insights into the regulatory landscape of ATG9A transport. Knockdown of ANPEP and NPM1 enhanced ATG9A availability outside the trans -Golgi network, suggesting they regulate ATG9A localization. These findings deepen our understanding of ATG9A trafficking in the context of AP-4 deficiency and offer a framework for the development of targeted interventions for AP-4-HSP.
Collapse
|
6
|
Cantón-Suárez A, Sánchez-Valdeón L, Bello-Corral L, Cuevas MJ, Estébanez B. Understanding the Molecular Impact of Physical Exercise on Alzheimer's Disease. Int J Mol Sci 2024; 25:13576. [PMID: 39769339 PMCID: PMC11677557 DOI: 10.3390/ijms252413576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Alzheimer's disease is one of the most common neurodegenerative diseases, characterized by a wide range of neurological symptoms that begin with personality changes and psychiatric symptoms, progress to mild cognitive impairment, and eventually lead to dementia. Physical exercise is part of the non-pharmacological treatments used in Alzheimer's disease, as it has been shown to delay the neurodegenerative process by improving the redox state in brain tissue, providing anti-inflammatory effects or stimulating the release of the brain-derived neurotrophic factor that enhances the brain structure and cognitive performance. Here, we reviewed the results obtained from studies conducted in both animal models and human subjects to comprehend how physical exercise interventions can exert changes in the molecular mechanisms underlying the pathophysiological processes in Alzheimer's disease: amyloid β-peptide pathology, tau pathology, neuroglial changes, mitochondrial dysfunction, and oxidative stress. Physical exercise seems to have a protective effect against Alzheimer's disease, since it has been shown to induce positive changes in some of the biomarkers related to the pathophysiological processes of the disease. However, additional studies in humans are necessary to address the current lack of conclusive evidence.
Collapse
Affiliation(s)
| | - Leticia Sánchez-Valdeón
- Health Research Nursing Group (GREIS), University of Leon, 24071 Leon, Spain; (L.S.-V.); (L.B.-C.)
- Department of Nursing and Physiotherapy, University of Leon, 24071 Leon, Spain
| | - Laura Bello-Corral
- Health Research Nursing Group (GREIS), University of Leon, 24071 Leon, Spain; (L.S.-V.); (L.B.-C.)
- Department of Nursing and Physiotherapy, University of Leon, 24071 Leon, Spain
| | - María J. Cuevas
- Institute of Biomedicine (IBIOMED), University of León, 24071 Leon, Spain;
| | - Brisamar Estébanez
- Institute of Biomedicine (IBIOMED), University of León, 24071 Leon, Spain;
| |
Collapse
|
7
|
Haessler A, Gier S, Jung N, Windbergs M. The Aβ 42:Aβ 40 ratio modulates aggregation in beta-amyloid oligomers and drives metabolic changes and cellular dysfunction. Front Cell Neurosci 2024; 18:1516093. [PMID: 39717390 PMCID: PMC11664223 DOI: 10.3389/fncel.2024.1516093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 11/25/2024] [Indexed: 12/25/2024] Open
Abstract
The pathophysiological role of Aβ42 oligomers in the onset of Alzheimer's disease (AD) is heavily disputed, pivoting research toward investigating mixed oligomers composed of Aβ42 and Aβ40, which is more abundant but less aggregation-prone. This study investigates Aβ42:Aβ40 oligomers in different ratios, examining their adverse effects on endothelial cells, neurons, astroglia, and microglia, as well as in a human blood-brain barrier (BBB) model. Combining label-free Raman microscopy with complementary imaging techniques and biochemical assays, we show the prominent impact of Aβ40 on Aβ42 fibrillation, suggesting an inhibitory effect on aggregation. Mixed oligomers, especially with low proportions of Aβ42, were equally detrimental as pure Aβ42 oligomers regarding cell viability, functionality, and metabolism. They also differentially affected lipid droplet metabolism in BBB-associated microglia, indicating distinct pathophysiological responses. Our findings demonstrate the overarching significance of the Aβ42:Aβ40 ratio in Aβ oligomers, challenging the traditional focus on Aβ42 in AD research.
Collapse
Affiliation(s)
| | | | | | - Maike Windbergs
- Institute of Pharmaceutical Technology, Goethe University Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
8
|
Wang J, Fourriere L, Gleeson PA. Advances in the cell biology of the trafficking and processing of amyloid precursor protein: impact of familial Alzheimer's disease mutations. Biochem J 2024; 481:1297-1325. [PMID: 39302110 PMCID: PMC11555708 DOI: 10.1042/bcj20240056] [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: 05/21/2024] [Revised: 09/02/2024] [Accepted: 09/05/2024] [Indexed: 09/22/2024]
Abstract
The production of neurotoxic amyloid-β peptides (Aβ) is central to the initiation and progression of Alzheimer's disease (AD) and involves sequential cleavage of the amyloid precursor protein (APP) by β- and γ-secretases. APP and the secretases are transmembrane proteins and their co-localisation in the same membrane-bound sub-compartment is necessary for APP cleavage. The intracellular trafficking of APP and the β-secretase, BACE1, is critical in regulating APP processing and Aβ production and has been studied in several cellular systems. Here, we summarise the intracellular distribution and transport of APP and its secretases, and the intracellular location for APP cleavage in non-polarised cells and neuronal models. In addition, we review recent advances on the potential impact of familial AD mutations on APP trafficking and processing. This is critical information in understanding the molecular mechanisms of AD progression and in supporting the development of novel strategies for clinical treatment.
Collapse
Affiliation(s)
- Jingqi Wang
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Lou Fourriere
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Paul A. Gleeson
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria 3010, Australia
| |
Collapse
|
9
|
Ali Z, Al-Ghouti MA, Abou-Saleh H, Rahman MM. Unraveling the Omega-3 Puzzle: Navigating Challenges and Innovations for Bone Health and Healthy Aging. Mar Drugs 2024; 22:446. [PMID: 39452854 PMCID: PMC11509197 DOI: 10.3390/md22100446] [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: 08/27/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024] Open
Abstract
Omega-3 polyunsaturated fatty acids (ω-3 PUFAs, n-3 PUFAs), including eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA), and alpha-linolenic acid (ALA), are essential polyunsaturated fats primarily obtained from fatty fish and plant-based sources. Compelling evidence from preclinical and epidemiological studies consistently suggests beneficial effects of ω-3 PUFAs on bone health and healthy aging processes. However, clinical trials have yielded mixed results, with some failing to replicate these benefits seen in preclinical models. This contraindication is mainly due to challenges such as low bioavailability, potential adverse effects with higher doses, and susceptibility to oxidation of ω-3 fatty acids, hindering their clinical effectiveness. This review comprehensively discusses recent findings from a clinical perspective, along with preclinical and epidemiological studies, emphasizing the role of ω-3 PUFAs in promoting bone health and supporting healthy aging. Additionally, it explores strategies to improve ω-3 PUFA efficacy, including nanoparticle encapsulation and incorporation of specialized pro-resolving mediators (SPM) derived from DHA and EPA, to mitigate oxidation and enhance solubility, thereby improving therapeutic potential. By consolidating evidence from various studies, this review underscores current insights and future directions in leveraging ω-3 PUFAs for therapeutic applications.
Collapse
Affiliation(s)
- Zayana Ali
- Biological Science Program, Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Mohammad Ahmed Al-Ghouti
- Environmental Science Program, Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Haissam Abou-Saleh
- Biomedical Sciences Department, College of Health Sciences, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Md Mizanur Rahman
- Biological Science Program, Department of Biological and Environmental Sciences, College of Arts and Sciences, Qatar University, Doha P.O. Box 2713, Qatar;
| |
Collapse
|
10
|
Puranik N, Song M. Insights into the Role of microRNAs as Clinical Tools for Diagnosis, Prognosis, and as Therapeutic Targets in Alzheimer's Disease. Int J Mol Sci 2024; 25:9936. [PMID: 39337429 PMCID: PMC11431957 DOI: 10.3390/ijms25189936] [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: 08/12/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Neurodegenerative diseases (NDDs) are a diverse group of neurological disorders characterized by alterations in the structure and function of the central nervous system. Alzheimer's disease (AD), characterized by impaired memory and cognitive abilities, is the most prevalent type of senile dementia. Loss of synapses, intracellular aggregation of hyperphosphorylated tau protein, and extracellular amyloid-β peptide (Aβ) plaques are the hallmarks of AD. MicroRNAs (miRNAs/miRs) are single-stranded ribonucleic acid (RNA) molecules that bind to the 3' and 5' untranslated regions of target genes to cause post-transcriptional gene silencing. The brain expresses over 70% of all experimentally detected miRNAs, and these miRNAs are crucial for synaptic function and particular signals during memory formation. Increasing evidence suggests that miRNAs play a role in AD pathogenesis and we provide an overview of the role of miRNAs in synapse formation, Aβ synthesis, tau protein accumulation, and brain-derived neurotrophic factor-associated AD pathogenesis. We further summarize and discuss the role of miRNAs as potential therapeutic targets and biomarkers for AD detection and differentiation between early- and late-stage AD, based on recent research. In conclusion, altered expression of miRNAs in the brain and peripheral circulation demonstrates their potential as biomarkers and therapeutic targets in AD.
Collapse
Affiliation(s)
- Nidhi Puranik
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea
| | - Minseok Song
- Department of Life Sciences, Yeungnam University, Gyeongsan 38541, Republic of Korea
| |
Collapse
|
11
|
Cao Y, Zhao LW, Chen ZX, Li SH. New insights in lipid metabolism: potential therapeutic targets for the treatment of Alzheimer's disease. Front Neurosci 2024; 18:1430465. [PMID: 39323915 PMCID: PMC11422391 DOI: 10.3389/fnins.2024.1430465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 08/14/2024] [Indexed: 09/27/2024] Open
Abstract
Alzheimer's disease (AD) is increasingly recognized as being intertwined with the dysregulation of lipid metabolism. Lipids are a significant class of nutrients vital to all organisms, playing crucial roles in cellular structure, energy storage, and signaling. Alterations in the levels of various lipids in AD brains and dysregulation of lipid pathways and transportation have been implicated in AD pathogenesis. Clinically, evidence for a high-fat diet firmly links disrupted lipid metabolism to the pathogenesis and progression of AD, although contradictory findings warrant further exploration. In view of the significance of various lipids in brain physiology, the discovery of complex and diverse mechanisms that connect lipid metabolism with AD-related pathophysiology will bring new hope for patients with AD, underscoring the importance of lipid metabolism in AD pathophysiology, and promising targets for therapeutic intervention. Specifically, cholesterol, sphingolipids, and fatty acids have been shown to influence amyloid-beta (Aβ) accumulation and tau hyperphosphorylation, which are hallmarks of AD pathology. Recent studies have highlighted the potential therapeutic targets within lipid metabolism, such as enhancing apolipoprotein E lipidation, activating liver X receptors and retinoid X receptors, and modulating peroxisome proliferator-activated receptors. Ongoing clinical trials are investigating the efficacy of these strategies, including the use of ketogenic diets, statin therapy, and novel compounds like NE3107. The implications of these findings suggest that targeting lipid metabolism could offer new avenues for the treatment and management of AD. By concentrating on alterations in lipid metabolism within the central nervous system and their contribution to AD development, this review aims to shed light on novel research directions and treatment approaches for combating AD, offering hope for the development of more effective management strategies.
Collapse
Affiliation(s)
- Yuan Cao
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, Zhengzhou, China
- Clinical Systems Biology Laboratories, Translation Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Lin-Wei Zhao
- Department of Cardiology, People’s Hospital of Zhengzhou University, Henan Provincial People’s Hospital, Zhengzhou University Central China Fuwai Hospital, Zhengzhou, China
| | - Zi-Xin Chen
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, Zhengzhou, China
- Clinical Systems Biology Laboratories, Translation Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| | - Shao-Hua Li
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, Zhengzhou, China
- Clinical Systems Biology Laboratories, Translation Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
12
|
Aranda-Abreu GE, Rojas-Durán F, Hernández-Aguilar ME, Herrera-Covarrubias D, Chí-Castañeda LD, Toledo-Cárdenas MR, Suárez-Medellín JM. Alzheimer's Disease: Cellular and Pharmacological Aspects. Geriatrics (Basel) 2024; 9:86. [PMID: 39051250 PMCID: PMC11270425 DOI: 10.3390/geriatrics9040086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/23/2024] [Accepted: 06/21/2024] [Indexed: 07/27/2024] Open
Abstract
Alzheimer's disease was described more than 100 years ago and despite the fact that several molecules are being tested for its treatment, which are in phase III trials, the disease continues to progress. The main problem is that these molecules function properly in healthy neurons, while neuronal pathology includes plasma membrane disruption, malfunction of various organelles, and hyperphosphorylation of Tau and amyloid plaques. The main objective of this article is the discussion of a neuronal restoration therapy, where molecules designed for the treatment of Alzheimer's disease would probably be more effective, and the quality of life of people would be better.
Collapse
Affiliation(s)
- Gonzalo Emiliano Aranda-Abreu
- Instituto de Investigaciones Cerebrales, Universidad Veracruzana, Xalapa 91192, Mexico; (F.R.-D.); (M.E.H.-A.); (D.H.-C.); (L.D.C.-C.); (M.R.T.-C.); (J.M.S.-M.)
| | | | | | | | | | | | | |
Collapse
|
13
|
Yubolphan R, Pratchayasakul W, Koonrungsesomboon N, Chattipakorn N, Chattipakorn SC. Potential links between platelets and amyloid-β in the pathogenesis of Alzheimer's disease: Evidence from in vitro, in vivo, and clinical studies. Exp Neurol 2024; 374:114683. [PMID: 38211684 DOI: 10.1016/j.expneurol.2024.114683] [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: 08/25/2023] [Revised: 12/28/2023] [Accepted: 01/04/2024] [Indexed: 01/13/2024]
Abstract
Cerebral amyloid angiopathy (CAA) is a prevalent comorbidity among patients with Alzheimer's disease (AD), present in up to 80% of cases with varying levels of severity. There is evidence to suggest that CAA might intensify cognitive deterioration in AD patients, thereby accelerating the development of AD pathology. As a source of amyloids, it has been postulated that platelets play a significant role in the pathogenesis of both AD and CAA. Although several studies have demonstrated that platelet activation plays an important role in the pathogenesis of AD and CAA, a clear understanding of the mechanisms involved in the three steps: platelet activation, platelet adhesion, and platelet aggregation in AD pathogenesis still remains elusive. Moreover, potential therapeutic targets in platelet-mediated AD pathogenesis have not been explicitly addressed. Therefore, the aim of this review is to collate and discuss the in vitro, in vivo, and clinical evidence related to platelet dysfunction, including associated activation, adhesion, and aggregation, with specific reference to amyloid-related AD pathogenesis. Potential therapeutic targets of platelet-mediated AD pathogenesis are also discussed. By enriching the understanding of the intricate relationship between platelet dysfunction and onset of AD, researchers may unveil new therapeutic targets or strategies to tackle this devastating neurodegeneration.
Collapse
Affiliation(s)
- Ruedeemars Yubolphan
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Wasana Pratchayasakul
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Nut Koonrungsesomboon
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Nipon Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand
| | - Siriporn C Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, Thailand; Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
14
|
Wang J, Gleeson PA, Fourriere L. Spatial-Temporal Mapping Reveals the Golgi as the Major Processing Site for the Pathogenic Swedish APP Mutation: Familial APP Mutant Shifts the Major APP Processing Site. Traffic 2024; 25:e12932. [PMID: 38528836 DOI: 10.1111/tra.12932] [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: 10/16/2023] [Revised: 02/13/2024] [Accepted: 03/01/2024] [Indexed: 03/27/2024]
Abstract
Alzheimer's disease is associated with increased levels of amyloid beta (Aβ) generated by sequential intracellular cleavage of amyloid precursor protein (APP) by membrane-bound secretases. However, the spatial and temporal APP cleavage events along the trafficking pathways are poorly defined. Here, we use the Retention Using Selective Hooks (RUSH) to compare in real time the anterograde trafficking and temporal cleavage events of wild-type APP (APPwt) with the pathogenic Swedish APP (APPswe) and the disease-protective Icelandic APP (APPice). The analyses revealed differences in the trafficking profiles and processing between APPwt and the APP familial mutations. While APPwt was predominantly processed by the β-secretase, BACE1, following Golgi transport to the early endosomes, the transit of APPswe through the Golgi was prolonged and associated with enhanced amyloidogenic APP processing and Aβ secretion. A 20°C block in cargo exit from the Golgi confirmed β- and γ-secretase processing of APPswe in the Golgi. Inhibition of the β-secretase, BACE1, restored APPswe anterograde trafficking profile to that of APPwt. APPice was transported rapidly through the Golgi to the early endosomes with low levels of Aβ production. This study has revealed different intracellular locations for the preferential cleavage of APPwt and APPswe and Aβ production, and the Golgi as the major processing site for APPswe, findings relevant to understand the molecular basis of Alzheimer's disease.
Collapse
Affiliation(s)
- Jingqi Wang
- The Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Paul A Gleeson
- The Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Lou Fourriere
- The Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
15
|
Sultana OF, Hia RA, Reddy PH. A Combinational Therapy for Preventing and Delaying the Onset of Alzheimer's Disease: A Focus on Probiotic and Vitamin Co-Supplementation. Antioxidants (Basel) 2024; 13:202. [PMID: 38397800 PMCID: PMC10886126 DOI: 10.3390/antiox13020202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/27/2024] [Accepted: 02/02/2024] [Indexed: 02/25/2024] Open
Abstract
Alzheimer's disease is a progressive neurodegenerative disorder with a complex etiology, and effective interventions to prevent or delay its onset remain a global health challenge. In recent years, there has been growing interest in the potential role of probiotic and vitamin supplementation as complementary strategies for Alzheimer's disease prevention. This review paper explores the current scientific literature on the use of probiotics and vitamins, particularly vitamin A, D, E, K, and B-complex vitamins, in the context of Alzheimer's disease prevention and management. We delve into the mechanisms through which probiotics may modulate gut-brain interactions and neuroinflammation while vitamins play crucial roles in neuronal health and cognitive function. The paper also examines the collective impact of this combinational therapy on reducing the risk factors associated with Alzheimer's disease, such as oxidative stress, inflammation, and gut dysbiosis. By providing a comprehensive overview of the existing evidence and potential mechanisms, this review aims to shed light on the promise of probiotic and vitamin co-supplementation as a multifaceted approach to combat Alzheimer's disease, offering insights into possible avenues for future research and clinical application.
Collapse
Affiliation(s)
- Omme Fatema Sultana
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
| | - Raksa Andalib Hia
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA;
| | - P. Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA;
- Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA;
- Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
- Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| |
Collapse
|
16
|
Chen J, Su YH, Wang M, Zhang YC. Emerging Role of Sorting Nexin 17 in Human Health and Disease. Curr Protein Pept Sci 2024; 25:814-825. [PMID: 38874037 DOI: 10.2174/0113892037284582240522155112] [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: 02/22/2024] [Revised: 04/21/2024] [Accepted: 04/25/2024] [Indexed: 06/15/2024]
Abstract
The distortion of the cellular membrane transport pathway has a profound impact on cell dynamics and can drive serious physiological consequences during the process of cell sorting. SNX17 is a member of the Sorting Nexin (SNX) family and plays a crucial role in protein sorting and transport in the endocytic pathway. SNX17, SNX27, and SNX31 belong to the SNX-FERM subfamily and possess the FERM domain, which can assist in endocytic transport and lysosomal degradation. The binding partners of SNX27 have been discovered to number over 100, and SNX27 has been linked to the development of Alzheimer's disease progression, tumorigenesis, cancer progression, and metastasis. However, the role and potential mechanisms of SNX17 in human health and disease remain poorly understood, and the function of SNX17 has not been fully elucidated. In this review, we summarize the structure and basic functions of SNX protein, focusing on providing current evidence of the role and possible mechanism of SNX17 in human neurodegenerative diseases and cardiovascular diseases.
Collapse
Affiliation(s)
- Juan Chen
- Key Laboratory of Sports Human Science, College of Physical Education, Liaoning Normal University, Dalian, 116029, China
| | - Yan-Hong Su
- Key Laboratory of Sports Human Science, College of Physical Education, Liaoning Normal University, Dalian, 116029, China
| | - Meng Wang
- Key Laboratory of Sports Human Science, College of Physical Education, Liaoning Normal University, Dalian, 116029, China
| | - Yi-Chen Zhang
- Key Laboratory of Sports Human Science, College of Physical Education, Liaoning Normal University, Dalian, 116029, China
| |
Collapse
|
17
|
André S, Verteneuil S, Ris L, Kahvecioglu ZC, Nonclercq D, De Winter J, Vander Elst L, Laurent S, Muller RN, Burtea C. Modulation of Cytosolic Phospholipase A2 as a Potential Therapeutic Strategy for Alzheimer's Disease. J Alzheimers Dis Rep 2023; 7:1395-1426. [PMID: 38225969 PMCID: PMC10789292 DOI: 10.3233/adr-230075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 11/17/2023] [Indexed: 01/17/2024] Open
Abstract
Background Alzheimer's disease (AD) is a neurodegenerative disorder lacking any curative treatment up to now. Indeed, actual medication given to the patients alleviates only symptoms. The cytosolic phospholipase A2 (cPLA2-IVA) appears as a pivotal player situated at the center of pathological pathways leading to AD and its inhibition could be a promising therapeutic approach. Objective A cPLA2-IVA inhibiting peptide was identified in the present work, aiming to develop an original therapeutic strategy. Methods We targeted the cPLA2-IVA using the phage display technology. The hit peptide PLP25 was first validated in vitro (arachidonic acid dosage [AA], cPLA2-IVA cellular translocation) before being tested in vivo. We evaluated spatial memory using the Barnes maze, amyloid deposits by MRI and immunohistochemistry (IHC), and other important biomarkers such as the cPLA2-IVA itself, the NMDA receptor, AβPP and tau by IHC after i.v. injection in APP/PS1 mice. Results Showing a high affinity for the C2 domain of this enzyme, the peptide PLP25 exhibited an inhibitory effect on cPLA2-IVA activity by blocking its binding to its substrate, resulting in a decreased release of AA. Coupled to a vector peptide (LRPep2) in order to optimize brain access, we showed an improvement of cognitive abilities of APP/PS1 mice, which also exhibited a decreased number of amyloid plaques, a restored expression of cPLA2-IVA, and a favorable effect on NMDA receptor expression and tau protein phosphorylation. Conclusions cPLA2-IVA inhibition through PLP25 peptide could be a promising therapeutic strategy for AD.
Collapse
Affiliation(s)
- Séverine André
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
| | - Sébastien Verteneuil
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
| | - Laurence Ris
- Department of Neurosciences, University of Mons, Research Institute for Health Science and Technologies, Mons, Belgium
| | - Zehra-Cagla Kahvecioglu
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
| | | | - Julien De Winter
- Organic Synthesis and Mass Spectrometry Laboratory (SMOs), University of Mons-UMONS, Mons, Belgium
| | - Luce Vander Elst
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
| | - Sophie Laurent
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
- Center for Microscopy and Molecular Imaging, Gosselies, Belgium
| | - Robert N. Muller
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
- Center for Microscopy and Molecular Imaging, Gosselies, Belgium
| | - Carmen Burtea
- General, Organic and Biomedical Chemistry Unit, NMR and Molecular Imaging Laboratory, University of Mons, Mons, Belgium
| |
Collapse
|
18
|
Kersten N, Farías GG. A voyage from the ER: spatiotemporal insights into polarized protein secretion in neurons. Front Cell Dev Biol 2023; 11:1333738. [PMID: 38188013 PMCID: PMC10766823 DOI: 10.3389/fcell.2023.1333738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 12/04/2023] [Indexed: 01/09/2024] Open
Abstract
To function properly, neurons must maintain a proteome that differs in their somatodendritic and axonal domain. This requires the polarized sorting of newly synthesized secretory and transmembrane proteins into different vesicle populations as they traverse the secretory pathway. Although the trans-Golgi-network is generally considered to be the main sorting hub, this sorting process may already begin at the ER and continue through the Golgi cisternae. At each step in the sorting process, specificity is conferred by adaptors, GTPases, tethers, and SNAREs. Besides this, local synthesis and unconventional protein secretion may contribute to the polarized proteome to enable rapid responses to stimuli. For some transmembrane proteins, some of the steps in the sorting process are well-studied. These will be highlighted here. The universal rules that govern polarized protein sorting remain unresolved, therefore we emphasize the need to approach this problem in an unbiased, top-down manner. Unraveling these rules will contribute to our understanding of neuronal development and function in health and disease.
Collapse
Affiliation(s)
- Noortje Kersten
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Ginny G Farías
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
19
|
Mishra S, Knupp A, Kinoshita C, Williams CA, Rose SE, Martinez R, Theofilas P, Young JE. Pharmacologic enhancement of retromer rescues endosomal pathology induced by defects in the Alzheimer's gene SORL1. Stem Cell Reports 2023; 18:2434-2450. [PMID: 37949073 PMCID: PMC10724056 DOI: 10.1016/j.stemcr.2023.10.011] [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: 09/22/2022] [Revised: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 11/12/2023] Open
Abstract
The SORL1 gene (SORLA) is strongly associated with risk of developing Alzheimer's disease (AD). SORLA is a regulator of endosomal trafficking in neurons and interacts with retromer, a complex that is a "master conductor" of endosomal trafficking. Small molecules can increase retromer expression in vitro, enhancing its function. We treated hiPSC-derived cortical neurons that are either fully deficient, haploinsufficient, or that harbor one copy of SORL1 variants linked to AD with TPT-260, a retromer-enhancing molecule. We show significant increases in retromer subunit VPS26B expression. We tested whether endosomal, amyloid, and TAU pathologies were corrected. We observed that the degree of rescue by TPT-260 treatment depended on the number of copies of functional SORL1 and which SORL1 variant was expressed. Using a disease-relevant preclinical model, our work illuminates how the SORL1-retromer pathway can be therapeutically harnessed.
Collapse
Affiliation(s)
- Swati Mishra
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195, USA
| | - Allison Knupp
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195, USA
| | - Chizuru Kinoshita
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195, USA
| | - C Andrew Williams
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195, USA
| | - Shannon E Rose
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195, USA
| | - Refugio Martinez
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195, USA
| | - Panos Theofilas
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jessica E Young
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA 98195, USA.
| |
Collapse
|
20
|
Eslami S, Hosseinzadeh Shakib N, Fooladfar Z, Nasrollahian S, Baghaei S, Mosaddad SA, Motamedifar M. The role of periodontitis-associated bacteria in Alzheimer's disease: A narrative review. J Basic Microbiol 2023; 63:1059-1072. [PMID: 37311215 DOI: 10.1002/jobm.202300250] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/01/2023] [Accepted: 06/03/2023] [Indexed: 06/15/2023]
Abstract
Alzheimer's disease causes memory loss and dementia in older adults through a neurodegenerative mechanism. Despite the pathophysiological clarification of this cognitive disorder, novel molecular and cellular pathways should be identified to determine its exact mechanism. Alzheimer's disease (AD) is pathologically characterized by senile plaques comprising beta-amyloid and neurofibrillary tangles (NFTs) formed by hyperphosphorylated tau as a microtubule-associated protein with a key role in the pathogenesis of AD. Periodontitis through inflammatory pathways is a risk factor for deteriorating cognitive impairment in AD patients. Poor oral hygiene coupled with immunocompromised status in older adults causes periodontal diseases and chronic inflammations through an oral bacterial imbalance. Toxic bacterial products, including bacteria themselves, can reach the central nervous system through the bloodstream and evoke inflammatory responses. The present review was conducted to investigate relationships between AD and periodontitis-involved bacteria as a risk factor.
Collapse
Affiliation(s)
- Saba Eslami
- Research Central Laboratory, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Zahra Fooladfar
- Student Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sina Nasrollahian
- Student Research Committee, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saman Baghaei
- Student Research Committee, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Ali Mosaddad
- Student Research Committee, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Motamedifar
- HIV/AIDS Research Center, Institute of Health, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
21
|
Rivai B, Umar AK. Neuroprotective compounds from marine invertebrates. BENI-SUEF UNIVERSITY JOURNAL OF BASIC AND APPLIED SCIENCES 2023; 12:71. [DOI: 10.1186/s43088-023-00407-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 07/22/2023] [Indexed: 09/01/2023] Open
Abstract
Abstract
Background
Neuroinflammation is a key pathological feature of a wide variety of neurological disorders, including Parkinson’s, multiple sclerosis, Alzheimer’s, and Huntington’s disease. While current treatments for these disorders are primarily symptomatic, there is a growing interest in developing new therapeutics that target the underlying neuroinflammatory processes.
Main body
Marine invertebrates, such as coral, sea urchins, starfish, sponges, and sea cucumbers, have been found to contain a wide variety of biologically active compounds that have demonstrated potential therapeutic properties. These compounds are known to target various key proteins and pathways in neuroinflammation, including 6-hydroxydopamine (OHDH), caspase-3 and caspase-9, p-Akt, p-ERK, p-P38, acetylcholinesterase (AChE), amyloid-β (Aβ), HSF-1, α-synuclein, cellular prion protein, advanced glycation end products (AGEs), paraquat (PQ), and mitochondria DJ-1.
Short conclusion
This review focuses on the current state of research on the neuroprotective effects of compounds found in marine invertebrates and the potential therapeutic implications of these findings for treating neuroinflammatory disorders. We also discussed the challenges and limitations of using marine-based compounds as therapeutics, such as sourcing and sustainability concerns, and the need for more preclinical and clinical studies to establish their efficacy and safety.
Graphical abstract
Collapse
|
22
|
Xu J, Hsu SH. Self-healing hydrogel as an injectable implant: translation in brain diseases. J Biomed Sci 2023; 30:43. [PMID: 37340481 DOI: 10.1186/s12929-023-00939-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 06/13/2023] [Indexed: 06/22/2023] Open
Abstract
Tissue engineering biomaterials are aimed to mimic natural tissue and promote new tissue formation for the treatment of impaired or diseased tissues. Highly porous biomaterial scaffolds are often used to carry cells or drugs to regenerate tissue-like structures. Meanwhile, self-healing hydrogel as a category of smart soft hydrogel with the ability to automatically repair its own structure after damage has been developed for various applications through designs of dynamic crosslinking networks. Due to flexibility, biocompatibility, and ease of functionalization, self-healing hydrogel has great potential in regenerative medicine, especially in restoring the structure and function of impaired neural tissue. Recent researchers have developed self-healing hydrogel as drug/cell carriers or tissue support matrices for targeted injection via minimally invasive surgery, which has become a promising strategy in treating brain diseases. In this review, the development history of self-healing hydrogel for biomedical applications and the design strategies according to different crosslinking (gel formation) mechanisms are summarized. The current therapeutic progress of self-healing hydrogels for brain diseases is described as well, with an emphasis on the potential therapeutic applications validated by in vivo experiments. The most recent aspect as well as the design rationale of self-healing hydrogel for different brain diseases is also addressed.
Collapse
Affiliation(s)
- Junpeng Xu
- Institute of Polymer Science and Engineering, National Taiwan University, No. 1, Sec. 4 Roosevelt Road, Taipei, 106319, Taiwan, Republic of China
| | - Shan-Hui Hsu
- Institute of Polymer Science and Engineering, National Taiwan University, No. 1, Sec. 4 Roosevelt Road, Taipei, 106319, Taiwan, Republic of China.
- Institute of Cellular and System Medicine, National Health Research Institutes, No. 35 Keyan Road, Miaoli, 350401, Taiwan, Republic of China.
| |
Collapse
|
23
|
Latina V, Atlante A, Malerba F, La Regina F, Balzamino BO, Micera A, Pignataro A, Stigliano E, Cavallaro S, Calissano P, Amadoro G. The Cleavage-Specific Tau 12A12mAb Exerts an Anti-Amyloidogenic Action by Modulating the Endocytic and Bioenergetic Pathways in Alzheimer's Disease Mouse Model. Int J Mol Sci 2023; 24:ijms24119683. [PMID: 37298634 DOI: 10.3390/ijms24119683] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 05/27/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023] Open
Abstract
Beyond deficits in hippocampal-dependent episodic memory, Alzheimer's Disease (AD) features sensory impairment in visual cognition consistent with extensive neuropathology in the retina. 12A12 is a monoclonal cleavage specific antibody (mAb) that in vivo selectively neutralizes the AD-relevant, harmful N-terminal 20-22 kDa tau fragment(s) (i.e., NH2htau) without affecting the full-length normal protein. When systemically injected into the Tg2576 mouse model overexpressing a mutant form of Amyloid Precursor Protein (APP), APPK670/671L linked to early onset familial AD, this conformation-specific tau mAb successfully reduces the NH2htau accumulating both in their brain and retina and, thus, markedly alleviates the phenotype-associated signs. By means of a combined biochemical and metabolic experimental approach, we report that 12A12mAb downregulates the steady state expression levels of APP and Beta-Secretase 1 (BACE-1) and, thus, limits the Amyloid beta (Aβ) production both in the hippocampus and retina from this AD animal model. The local, antibody-mediated anti-amyloidogenic action is paralleled in vivo by coordinated modulation of the endocytic (BIN1, RIN3) and bioenergetic (glycolysis and L-Lactate) pathways. These findings indicate for the first time that similar molecular and metabolic retino-cerebral pathways are modulated in a coordinated fashion in response to 12A12mAb treatment to tackle the neurosensorial Aβ accumulation in AD neurodegeneration.
Collapse
Affiliation(s)
- Valentina Latina
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Anna Atlante
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council (CNR), Via Amendola 122/O, 70126 Bari, Italy
| | - Francesca Malerba
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Federico La Regina
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Bijorn Omar Balzamino
- Research Laboratories in Ophthalmology, IRCCS-Fondazione Bietti, Via Santo Stefano Rotondo 6, 00184 Rome, Italy
| | - Alessandra Micera
- Research Laboratories in Ophthalmology, IRCCS-Fondazione Bietti, Via Santo Stefano Rotondo 6, 00184 Rome, Italy
| | - Annabella Pignataro
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), Via Fosso del Cavaliere 100, 00133 Rome, Italy
| | - Egidio Stigliano
- Area of Pathology, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Istituto di Anatomia Patologica, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Sebastiano Cavallaro
- Institute for Biomedical Research and Innovation (IRIB), National Research Council (CNR), Via P. Gaifami 18, 95126 Catania, Italy
| | - Pietro Calissano
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
| | - Giuseppina Amadoro
- European Brain Research Institute (EBRI), Viale Regina Elena 295, 00161 Rome, Italy
- Institute of Translational Pharmacology (IFT), National Research Council (CNR), Via Fosso del Cavaliere 100, 00133 Rome, Italy
| |
Collapse
|
24
|
Ben Ahmed A, Lemaire Q, Scache J, Mariller C, Lefebvre T, Vercoutter-Edouart AS. O-GlcNAc Dynamics: The Sweet Side of Protein Trafficking Regulation in Mammalian Cells. Cells 2023; 12:1396. [PMID: 37408229 PMCID: PMC10216988 DOI: 10.3390/cells12101396] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 07/07/2023] Open
Abstract
The transport of proteins between the different cellular compartments and the cell surface is governed by the secretory pathway. Alternatively, unconventional secretion pathways have been described in mammalian cells, especially through multivesicular bodies and exosomes. These highly sophisticated biological processes rely on a wide variety of signaling and regulatory proteins that act sequentially and in a well-orchestrated manner to ensure the proper delivery of cargoes to their final destination. By modifying numerous proteins involved in the regulation of vesicular trafficking, post-translational modifications (PTMs) participate in the tight regulation of cargo transport in response to extracellular stimuli such as nutrient availability and stress. Among the PTMs, O-GlcNAcylation is the reversible addition of a single N-acetylglucosamine monosaccharide (GlcNAc) on serine or threonine residues of cytosolic, nuclear, and mitochondrial proteins. O-GlcNAc cycling is mediated by a single couple of enzymes: the O-GlcNAc transferase (OGT) which catalyzes the addition of O-GlcNAc onto proteins, and the O-GlcNAcase (OGA) which hydrolyses it. Here, we review the current knowledge on the emerging role of O-GlcNAc modification in the regulation of protein trafficking in mammalian cells, in classical and unconventional secretory pathways.
Collapse
|
25
|
Yavari M, Ramalingam L, Harris BN, Kahathuduwa CN, Chavira A, Biltz C, Mounce L, Maldonado KA, Scoggin S, Zu Y, Kalupahana NS, Yosofvand M, Moussa H, Moustaid-Moussa N. Eicosapentaenoic Acid Protects against Metabolic Impairments in the APPswe/PS1dE9 Alzheimer's Disease Mouse Model. J Nutr 2023; 153:1038-1051. [PMID: 36781072 PMCID: PMC10273166 DOI: 10.1016/j.tjnut.2023.01.030] [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: 09/03/2022] [Revised: 01/18/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is an age-related neurodegenerative disease characterized by amyloid-β (Aβ) plaques. Systemic inflammation and obesity may exacerbate AD pathogenesis. We previously reported anti-inflammatory and anti-obesity effects of EPA in mice. OBJECTIVES We aimed to determine whether EPA reduces obesity-associated metabolic dysfunctions and Aβ accumulation in AD amyloidogenic mice. METHODS Two-mo-old APPswe/PS1dE9 transgenic (TG) mice and non-TG littermates were randomly assigned to low fat (LF; 10% kcal fat), high fat (HF; 45% kcal fat), or EPA (36 g/kg)-supplemented HF diets. Body composition, glucose tolerance, and energy expenditure were measured, and serum and brain metabolic markers were tested 38 wk postintervention. Outcomes were statistically analyzed via 3-factor ANOVA, modeling genotype, sex, and diet interactions. RESULTS HF-fed males gained more weight than females (Δ = 61 mg; P < 0.001). Compared with LF, HF increased body weights of wild-type (WT) males (Δ = 31 mg; P < 0.001). EPA reduced HF-induced weight gain in WT males (Δ = 24 mg; P = 0.054) but not in females. HF mice showed decreased glucose clearance and respiratory energy compared with LF-fed groups (Δ = -1.31 g/dL; P < 0.001), with no significant effects of EPA. However, EPA conferred metabolic improvements by decreasing serum leptin and insulin (Δ = -2.51 g/mL and Δ = -0.694 ng/mL, respectively compared with HF, P ≤ 0.05) and increasing adiponectin (Δ = 21.6 ng/mL; P < 0.001). As we expected, TG mice expressed higher serum and brain Aβ than WT mice (Δ = 0.131 ng/mL; P < 0.001 and Δ = 0.56%; P < 0.01, respectively), and EPA reduced serum Aβ1-40 in TG males compared with HF (Δ = 0.053 ng/mL; P ≤ 0.05). CONCLUSIONS To our knowledge, this is the first report that EPA reduces serum Aβ1-40 in obese AD male mice, warranting further investigations into tissue-specific mechanisms of EPA in AD.
Collapse
Affiliation(s)
- Mahsa Yavari
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA; Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX, USA
| | - Latha Ramalingam
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| | - Breanna N Harris
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX, USA; Department of Biological Sciences, Texas Tech University, Lubbock, TX, USA
| | - Chanaka Nadeeshan Kahathuduwa
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX, USA; Department of Laboratory Science and Primary Care, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Angela Chavira
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| | - Caroline Biltz
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| | - Logan Mounce
- Department of Biological Sciences, Texas Tech University, Lubbock, TX, USA
| | | | - Shane Scoggin
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| | - Yujiao Zu
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA; Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX, USA
| | - Nishan Sudheera Kalupahana
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA; Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX, USA; Department of Physiology, University of Peradeniya, Sri Lanka
| | - Mohammad Yosofvand
- Department of Mechanical Engineering, Texas Tech University, Lubbock, TX, USA
| | - Hanna Moussa
- Department of Mechanical Engineering, Texas Tech University, Lubbock, TX, USA
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA; Obesity Research Institute, Office of Research & Innovation, Texas Tech University, Lubbock, TX, USA.
| |
Collapse
|
26
|
Brandimarti R, Irollo E, Meucci O. The US9-Derived Protein gPTB9TM Modulates APP Processing Without Targeting Secretase Activities. Mol Neurobiol 2023; 60:1811-1825. [PMID: 36576708 PMCID: PMC9984340 DOI: 10.1007/s12035-022-03153-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 11/29/2022] [Indexed: 12/29/2022]
Abstract
Alteration of neuronal protein processing is often associated with neurological disorders and is highly dependent on cellular protein trafficking. A prime example is the amyloidogenic processing of amyloid precursor protein (APP) in intracellular vesicles, which plays a key role in age-related cognitive impairment. Most approaches to correct this altered processing aim to limit enzymatic activities that lead to toxic products, such as protein cleavage by β-secretase and the resulting amyloid β production. A viable alternative is to direct APP to cellular compartments where non-amyloidogenic mechanisms are favored. To this end, we exploited the molecular properties of the herpes simplex virus 1 (HSV-1) transport protein US9 to guide APP interaction with preferred endogenous targets. Specifically, we generated a US9 chimeric construct that facilitates APP processing through the non-amyloidogenic pathway and tested it in primary cortical neurons. In addition to reducing amyloid β production, our approach controls other APP-dependent biochemical steps that lead to neuronal deficits, including phosphorylation of APP and tau proteins. Notably, it also promotes the release of neuroprotective soluble αAPP. In contrast to other neuroprotective strategies, these US9-driven effects rely on the activity of endogenous neuronal proteins, which lends itself well to the study of fundamental mechanisms of APP processing/trafficking. Overall, this work introduces a new method to limit APP misprocessing and its cellular consequences without directly targeting secretase activity, offering a novel tool to reduce cognitive decline in pathologies such as Alzheimer's disease and HIV-associated neurocognitive disorders.
Collapse
Affiliation(s)
- Renato Brandimarti
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N.15th Street, Philadelphia, PA, 19102, USA.,Center for Neuroimmunology and CNS Therapeutics, Drexel University College of Medicine, 245 N.15th Street, Philadelphia, PA, 19102, USA.,Department of Pharmacy and Biotechnology, University of Bologna, Via San Giacomo,14, 40126, Bologna, Italy
| | - Elena Irollo
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N.15th Street, Philadelphia, PA, 19102, USA.,Center for Neuroimmunology and CNS Therapeutics, Drexel University College of Medicine, 245 N.15th Street, Philadelphia, PA, 19102, USA
| | - Olimpia Meucci
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N.15th Street, Philadelphia, PA, 19102, USA. .,Center for Neuroimmunology and CNS Therapeutics, Drexel University College of Medicine, 245 N.15th Street, Philadelphia, PA, 19102, USA. .,Department of Microbiology and Immunology, Drexel University College of Medicine, 245 N.15th Street, Philadelphia, PA, 19102, USA.
| |
Collapse
|
27
|
Maszka P, Kwasniak-Butowska M, Cysewski D, Slawek J, Smolenski RT, Tomczyk M. Metabolomic Footprint of Disrupted Energetics and Amino Acid Metabolism in Neurodegenerative Diseases: Perspectives for Early Diagnosis and Monitoring of Therapy. Metabolites 2023; 13:metabo13030369. [PMID: 36984809 PMCID: PMC10057046 DOI: 10.3390/metabo13030369] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 02/20/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
The prevalence of neurodegenerative diseases (NDs) is increasing due to the aging population and improved longevity. They are characterized by a range of pathological hallmarks, including protein aggregation, mitochondrial dysfunction, and oxidative stress. The aim of this review is to summarize the alterations in brain energy and amino acid metabolism in Alzheimer’s disease (AD), Parkinson’s disease (PD), and Huntington’s disease (HD). Based on our findings, we proposed a group of selected metabolites related to disturbed energy or mitochondrial metabolism as potential indicators or predictors of disease. We also discussed the hidden challenges of metabolomics studies in NDs and proposed future directions in this field. We concluded that biochemical parameters of brain energy metabolism disruption (obtained with metabolomics) may have potential application as a diagnostic tool for the diagnosis, prediction, and monitoring of the effectiveness of therapies for NDs. However, more studies are needed to determine the sensitivity of the proposed candidates. We suggested that the most valuable biomarkers for NDs studies could be groups of metabolites combined with other neuroimaging or molecular techniques. To attain clinically applicable results, the integration of metabolomics with other “omic” techniques might be required.
Collapse
Affiliation(s)
- Patrycja Maszka
- Department of Biochemistry, Medical University of Gdansk, 80-210 Gdansk, Poland
| | - Magdalena Kwasniak-Butowska
- Division of Neurological and Psychiatric Nursing, Medical University of Gdansk, 80-211 Gdansk, Poland
- Department of Neurology, St. Adalbert Hospital, 80-462 Gdansk, Poland
| | - Dominik Cysewski
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Jaroslaw Slawek
- Division of Neurological and Psychiatric Nursing, Medical University of Gdansk, 80-211 Gdansk, Poland
- Department of Neurology, St. Adalbert Hospital, 80-462 Gdansk, Poland
| | - Ryszard T. Smolenski
- Department of Biochemistry, Medical University of Gdansk, 80-210 Gdansk, Poland
- Correspondence: (R.T.S.); (M.T.)
| | - Marta Tomczyk
- Department of Biochemistry, Medical University of Gdansk, 80-210 Gdansk, Poland
- Correspondence: (R.T.S.); (M.T.)
| |
Collapse
|
28
|
The interactions of amyloid β aggregates with phospholipid membranes and the implications for neurodegeneration. Biochem Soc Trans 2023; 51:147-159. [PMID: 36629697 DOI: 10.1042/bst20220434] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 01/12/2023]
Abstract
Misfolding, aggregation and accumulation of Amyloid-β peptides (Aβ) in neuronal tissue and extracellular matrix are hallmark features of Alzheimer's disease (AD) pathology. Soluble Aβ oligomers are involved in neuronal toxicity by interacting with the lipid membrane, compromising its integrity, and affecting the function of receptors. These facts indicate that the interaction between Aβ oligomers and cell membranes may be one of the central molecular level factors responsible for the onset of neurodegeneration. The present review provides a structural understanding of Aβ neurotoxicity via membrane interactions and contributes to understanding early events in Alzheimer's disease.
Collapse
|
29
|
Amyloid β, Lipid Metabolism, Basal Cholinergic System, and Therapeutics in Alzheimer’s Disease. Int J Mol Sci 2022; 23:ijms232012092. [PMID: 36292947 PMCID: PMC9603563 DOI: 10.3390/ijms232012092] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/04/2022] [Accepted: 10/06/2022] [Indexed: 12/05/2022] Open
Abstract
The presence of insoluble aggregates of amyloid β (Aβ) in the form of neuritic plaques (NPs) is one of the main features that define Alzheimer’s disease. Studies have suggested that the accumulation of these peptides in the brain significantly contributes to extensive neuronal loss. Furthermore, the content and distribution of cholesterol in the membrane have been shown to have an important effect on the production and subsequent accumulation of Aβ peptides in the plasma membrane, contributing to dysfunction and neuronal death. The monomeric forms of these membrane-bound peptides undergo several conformational changes, ranging from oligomeric forms to beta-sheet structures, each presenting different levels of toxicity. Aβ peptides can be internalized by particular receptors and trigger changes from Tau phosphorylation to alterations in cognitive function, through dysfunction of the cholinergic system. The goal of this review is to summarize the current knowledge on the role of lipids in Alzheimer’s disease and their relationship with the basal cholinergic system, as well as potential disease-modifying therapies.
Collapse
|
30
|
Limone A, Veneruso I, D'Argenio V, Sarnataro D. Endosomal trafficking and related genetic underpinnings as a hub in Alzheimer's disease. J Cell Physiol 2022; 237:3803-3815. [PMID: 35994714 DOI: 10.1002/jcp.30864] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 07/13/2022] [Accepted: 08/08/2022] [Indexed: 01/07/2023]
Abstract
Genetic studies support the amyloid cascade as the leading hypothesis for the pathogenesis of Alzheimer's disease (AD). Although significant efforts have been made in untangling the amyloid and other pathological events in AD, ongoing interventions for AD have not been revealed efficacious for slowing down disease progression. Recent advances in the field of genetics have shed light on the etiology of AD, identifying numerous risk genes associated with late-onset AD, including genes related to intracellular endosomal trafficking. Some of the bases for the development of AD may be explained by the recently emerging AD genetic "hubs," which include the processing pathway of amyloid precursor protein and the endocytic pathway. The endosomal genetic hub may represent a common pathway through which many pathological effects can be mediated and novel, alternative biological targets could be identified for therapeutic interventions. The aim of this review is to focus on the genetic and biological aspects of the endosomal compartments related to AD progression. We report recent studies which describe how changes in endosomal genetics impact on functional events, such as the amyloidogenic and non-amyloidogenic processing, degradative pathways, and the importance of receptors related to endocytic trafficking, including the 37/67 kDa laminin-1 receptor ribosomal protein SA, and their implications for neurodegenerative diseases.
Collapse
Affiliation(s)
- Adriana Limone
- Department of Molecular Medicine and Medical Biotechnology, Federico II University, Napoli, Italy
| | - Iolanda Veneruso
- Department of Molecular Medicine and Medical Biotechnology, Federico II University, Napoli, Italy.,CEINGE-Biotecnologie Avanzate, Napoli, Italy
| | - Valeria D'Argenio
- CEINGE-Biotecnologie Avanzate, Napoli, Italy.,Department of Human Sciences and Quality of Life Promotion, San Raffaele Open University, Roma, Italy
| | - Daniela Sarnataro
- Department of Molecular Medicine and Medical Biotechnology, Federico II University, Napoli, Italy
| |
Collapse
|
31
|
Blasco Tavares Pereira Lopes F, Schlatzer D, Wang R, Li X, Feng E, Koyutürk M, Qi X, Chance MR. Temporal and Sex-Linked Protein Expression Dynamics in a Familial Model of Alzheimer's Disease. Mol Cell Proteomics 2022; 21:100280. [PMID: 35944844 PMCID: PMC9483563 DOI: 10.1016/j.mcpro.2022.100280] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 07/27/2022] [Accepted: 08/03/2022] [Indexed: 12/03/2022] Open
Abstract
Mouse models of Alzheimer's disease (AD) show progression through stages reflective of human pathology. Proteomics identification of temporal and sex-linked factors driving AD-related pathways can be used to dissect initiating and propagating events of AD stages to develop biomarkers or design interventions. In the present study, we conducted label-free proteome measurements of mouse hippocampus tissue with variables of time (3, 6, and 9 months), genetic background (5XFAD versus WT), and sex (equal males and females). These time points are associated with well-defined phenotypes with respect to the following: Aβ42 plaque deposition, memory deficits, and neuronal loss, allowing correlation of proteome-based molecular signatures with the mouse model stages. Our data show 5XFAD mice exhibit increases in known human AD biomarkers as amyloid-beta peptide, APOE, GFAP, and ITM2B are upregulated across all time points/stages. At the same time, 23 proteins are here newly associated with Alzheimer's pathology as they are also dysregulated in 5XFAD mice. At a pathways level, the 5XFAD-specific upregulated proteins are significantly enriched for DNA damage and stress-induced senescence at 3-month only, while at 6-month, the AD-specific proteome signature is altered and significantly enriched for membrane trafficking and vesicle-mediated transport protein annotations. By 9-month, AD-specific dysregulation is also characterized by significant neuroinflammation with innate immune system, platelet activation, and hyper-reactive astrocyte-related enrichments. Aside from these temporal changes, analysis of sex-linked differences in proteome signatures uncovered novel sex and AD-associated proteins. Pathway analysis revealed sex-linked differences in the 5XFAD model to be involved in the regulation of well-known human AD-related processes of amyloid fibril formation, wound healing, lysosome biogenesis, and DNA damage. Verification of the discovery results by Western blot and parallel reaction monitoring confirm the fundamental conclusions of the study and poise the 5XFAD model for further use as a molecular tool for understanding AD.
Collapse
Affiliation(s)
- Filipa Blasco Tavares Pereira Lopes
- Department of Nutrition, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA; Center for Proteomics and Bioinformatics, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Daniela Schlatzer
- Department of Nutrition, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA; Center for Proteomics and Bioinformatics, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Rihua Wang
- Department of Physiology & Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA; Center for Mitochondrial Diseases, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Xiaolin Li
- Department of Nutrition, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA; Center for Proteomics and Bioinformatics, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Emily Feng
- Department of Nutrition, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA; Center for Proteomics and Bioinformatics, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Mehmet Koyutürk
- Center for Proteomics and Bioinformatics, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA; Department of Computer and Data Sciences, Case School of Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Xin Qi
- Department of Physiology & Biophysics, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA; Center for Mitochondrial Diseases, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA
| | - Mark R Chance
- Department of Nutrition, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA; Center for Proteomics and Bioinformatics, School of Medicine, Case Western Reserve University, Cleveland, Ohio, USA.
| |
Collapse
|
32
|
Januário YC, Eden J, de Oliveira LS, De Pace R, Tavares LA, da Silva-Januário ME, Apolloni VB, Wilby EL, Altmeyer R, Burgos PV, Corrêa SAL, Gershlick DC, daSilva LLP. Clathrin adaptor AP-1-mediated Golgi export of amyloid precursor protein is crucial for the production of neurotoxic amyloid fragments. J Biol Chem 2022; 298:102172. [PMID: 35753347 PMCID: PMC9352552 DOI: 10.1016/j.jbc.2022.102172] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 06/08/2022] [Accepted: 06/09/2022] [Indexed: 11/28/2022] Open
Abstract
One of the hallmarks of Alzheimer's disease is the accumulation of toxic amyloid-β (Aβ) peptides in extracellular plaques. The direct precursor of Aβ is the carboxyl-terminal fragment β (or C99) of the amyloid precursor protein (APP). C99 is detected at elevated levels in Alzheimer's disease brains, and its intracellular accumulation has been linked to early neurotoxicity independently of Aβ. Despite this, the causes of increased C99 levels are poorly understood. Here, we demonstrate that APP interacts with the clathrin vesicle adaptor AP-1 (adaptor protein 1), and we map the interaction sites on both proteins. Using quantitative kinetic trafficking assays, established cell lines and primary neurons, we also show that this interaction is required for the transport of APP from the trans-Golgi network to endosomes. In addition, disrupting AP-1-mediated transport of APP alters APP processing and degradation, ultimately leading to increased C99 production and Aβ release. Our results indicate that AP-1 regulates the subcellular distribution of APP, altering its processing into neurotoxic fragments.
Collapse
Affiliation(s)
- Yunan C Januário
- Center for Virology Research, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Jessica Eden
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Luan S de Oliveira
- Center for Virology Research, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; School of Pharmacy and Medical Sciences, University of Bradford, Bradford, UK
| | - Raffaella De Pace
- Cell Biology and Neurobiology Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Lucas A Tavares
- Center for Virology Research, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Mara E da Silva-Januário
- Center for Virology Research, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Vinícius B Apolloni
- Center for Virology Research, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Elise L Wilby
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Randolf Altmeyer
- Statslab, Department of Pure Mathematics and Mathematical Statistics, University of Cambridgee, Cambridge, UK
| | - Patricia V Burgos
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago, Chile; Center for Aging and Regeneration (CARE), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sonia A L Corrêa
- School of Pharmacy and Medical Sciences, University of Bradford, Bradford, UK; Department of Life Sciences, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, UK
| | - David C Gershlick
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK.
| | - Luis L P daSilva
- Center for Virology Research, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil; Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
33
|
Patel VN, Chorawala MR, Shah MB, Shah KC, Dave BP, Shah MP, Patel TM. Emerging Pathophysiological Mechanisms Linking Diabetes Mellitus and Alzheimer’s Disease: An Old Wine in a New Bottle. J Alzheimers Dis Rep 2022; 6:349-357. [PMID: 35891636 PMCID: PMC9277673 DOI: 10.3233/adr-220021] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 05/30/2022] [Indexed: 01/15/2023] Open
Abstract
Type-2 diabetes mellitus (T2DM) is a chronic immuno-inflammatory and metabolic disease characterized by hyperglycemia and insulin resistance with corresponding hyperinsulinemia. On the other hand, Alzheimer’s disease (AD) is a neurodegenerative disease involving cognitive impairment, neuronal dysfunction, and memory loss. Several recently published literatures suggest a causal relationship between T2DM and AD. In this review, we have discussed several potential mechanisms underlying diabetes-induced cognitive impairment which include, abnormal insulin signaling, amyloid-β accumulation, oxidative stress, immuno-inflammation, mitochondrial dysfunction, advanced glycation end products, acetylcholinesterase and butyrylcholinesterase, advanced lipid peroxidation products, and apolipoprotein E. All these interconnected mechanisms may act either individually or synergistically which eventually leads to neurodegeneration and AD.
Collapse
Affiliation(s)
- Vishvas N. Patel
- Department of Pharmacology, L. M. College of Pharmacy, Ahmedabad, Gujarat, India
| | - Mehul R. Chorawala
- Department of Pharmacology, L. M. College of Pharmacy, Ahmedabad, Gujarat, India
| | - Maitri B. Shah
- Department of Pharmacology, L. M. College of Pharmacy, Ahmedabad, Gujarat, India
| | - Kashvi C. Shah
- Department of Pharmacology, L. M. College of Pharmacy, Ahmedabad, Gujarat, India
| | - Bhavarth P. Dave
- Department of Pharmacology, L. M. College of Pharmacy, Ahmedabad, Gujarat, India
| | - Manal P. Shah
- Department of Pharmacology, L. M. College of Pharmacy, Ahmedabad, Gujarat, India
| | - Tanvi M. Patel
- Department of Pharmacology, L. M. College of Pharmacy, Ahmedabad, Gujarat, India
| |
Collapse
|
34
|
Singh Y, Regmi D, Ormaza D, Ayyalasomayajula R, Vela N, Mundim G, Du D, Minond D, Cudic M. Mucin-Type O-Glycosylation Proximal to β-Secretase Cleavage Site Affects APP Processing and Aggregation Fate. Front Chem 2022; 10:859822. [PMID: 35464218 PMCID: PMC9023740 DOI: 10.3389/fchem.2022.859822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/07/2022] [Indexed: 12/04/2022] Open
Abstract
The amyloid-β precursor protein (APP) undergoes proteolysis by β- and γ-secretases to form amyloid-β peptides (Aβ), which is a hallmark of Alzheimer's disease (AD). Recent findings suggest a possible role of O-glycosylation on APP's proteolytic processing and subsequent fate for AD-related pathology. We have previously reported that Tyr681-O-glycosylation and the Swedish mutation accelerate cleavage of APP model glycopeptides by β-secretase (amyloidogenic pathway) more than α-secretase (non-amyloidogenic pathway). Therefore, to further our studies, we have synthesized additional native and Swedish-mutated (glyco)peptides with O-GalNAc moiety on Thr663 and/or Ser667 to explore the role of glycosylation on conformation, secretase activity, and aggregation kinetics of Aβ40. Our results show that conformation is strongly dependent on external conditions such as buffer ions and solvent polarity as well as internal modifications of (glyco)peptides such as length, O-glycosylation, and Swedish mutation. Furthermore, the level of β-secretase activity significantly increases for the glycopeptides containing the Swedish mutation compared to their nonglycosylated and native counterparts. Lastly, the glycopeptides impact the kinetics of Aβ40 aggregation by significantly increasing the lag phase and delaying aggregation onset, however, this effect is less pronounced for its Swedish-mutated counterparts. In conclusion, our results confirm that the Swedish mutation and/or O-glycosylation can render APP model glycopeptides more susceptible to cleavage by β-secretase. In addition, this study sheds new light on the possible role of glycosylation and/or glycan density on the rate of Aβ40 aggregation.
Collapse
Affiliation(s)
- YashoNandini Singh
- Department of Chemistry and Biochemistry, Charles E. Schmidt College of Science, Florida Atlantic University, Boca Raton, FL, United States
| | - Deepika Regmi
- Department of Chemistry and Biochemistry, Charles E. Schmidt College of Science, Florida Atlantic University, Boca Raton, FL, United States
| | - David Ormaza
- Department of Chemistry and Biochemistry, Charles E. Schmidt College of Science, Florida Atlantic University, Boca Raton, FL, United States
| | - Ramya Ayyalasomayajula
- Department of Chemistry and Biochemistry, Charles E. Schmidt College of Science, Florida Atlantic University, Boca Raton, FL, United States
| | - Nancy Vela
- Department of Chemistry and Biochemistry, Charles E. Schmidt College of Science, Florida Atlantic University, Boca Raton, FL, United States
| | - Gustavo Mundim
- Department of Chemistry and Biochemistry, Charles E. Schmidt College of Science, Florida Atlantic University, Boca Raton, FL, United States
| | - Deguo Du
- Department of Chemistry and Biochemistry, Charles E. Schmidt College of Science, Florida Atlantic University, Boca Raton, FL, United States
| | - Dmitriy Minond
- College of Pharmacy and Rumbaugh-Goodwin Institute for Cancer Research, Nova Southeastern University, Fort Lauderdale, FL, United States
| | - Maré Cudic
- Department of Chemistry and Biochemistry, Charles E. Schmidt College of Science, Florida Atlantic University, Boca Raton, FL, United States
| |
Collapse
|
35
|
Burrinha T, Cláudia GA. Aging impact on amyloid precursor protein neuronal trafficking. Curr Opin Neurobiol 2022; 73:102524. [PMID: 35303572 DOI: 10.1016/j.conb.2022.102524] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Revised: 02/06/2022] [Accepted: 02/08/2022] [Indexed: 11/03/2022]
Abstract
Neurons live a lifetime. Neuronal aging may increase the risk of Alzheimer's disease. How does neuronal membrane trafficking maintain synapse function during aging? In the normal aged brain, intraneuronal beta-amyloid (Aβ) accumulates without Alzheimer's disease mutations or risk variants. However, do changes with neuronal aging potentiate Aβ accumulation? We reviewed the membrane trafficking of the amyloid precursor protein in neurons and highlighted its importance in Aβ production. Importantly, we reviewed the evidence supporting the impact of aging on neuronal membrane trafficking, APP processing, and consequently Aβ production. Dissecting the molecular regulators of APP trafficking during neuronal aging is required to identify strategies to delay synaptic decline and protect from Alzheimer's disease.
Collapse
Affiliation(s)
- Tatiana Burrinha
- Chronic Diseases Research Center (CEDOC), NOVA Medical School (NMS), Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal. https://twitter.com/@burrinha_t
| | - Guimas Almeida Cláudia
- Chronic Diseases Research Center (CEDOC), NOVA Medical School (NMS), Universidade Nova de Lisboa, 1169-056 Lisboa, Portugal.
| |
Collapse
|
36
|
Fourriere L, Cho EHJ, Gleeson PA. Segregation of the membrane cargoes, BACE1 and amyloid precursor protein (APP) throughout the Golgi apparatus. Traffic 2022; 23:158-173. [PMID: 35076977 PMCID: PMC9303681 DOI: 10.1111/tra.12831] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/26/2021] [Accepted: 01/18/2022] [Indexed: 11/28/2022]
Abstract
The intracellular trafficking of β‐site amyloid precursor protein (APP) cleaving enzyme (BACE1) and APP regulates amyloid‐β production. Our previous work demonstrated that newly synthesized BACE1 and APP are segregated into distinct trafficking pathways from the trans‐Golgi network (TGN), and that alterations in their trafficking lead to an increase in Aβ production in non‐neuronal and neuronal cells. However, it is not known whether BACE1 and APP are transported through the Golgi stacks together and sorted at the TGN or segregated prior to arrival at the TGN. To address this question, we have used high‐resolution Airyscan technology followed by Huygens deconvolution to quantify the overlap of BACE1 and APP in Golgi subcompartments in HeLa cells and primary neurons. Here, we show that APP and BACE1 are segregated, on exit from the endoplasmic reticulum and in the cis‐Golgi and throughout the Golgi stack. In contrast, the transferrin receptor, which exits the TGN in AP‐1 mediated transport carriers as for BACE1, colocalizes with BACE1, but not APP, throughout the Golgi stack. The segregation of APP and BACE1 is independent of the Golgi ribbon structure and the cytoplasmic domain of the cargo. Overall, our findings reveal the segregation of different membrane cargoes early in the secretory pathway, a finding relevant to the regulation of APP processing events.
Collapse
Affiliation(s)
- Lou Fourriere
- The Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, Australia
| | - Ellie Hyun-Jung Cho
- The Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, Australia.,Biological Optical Microscopy Platform, The University of Melbourne, Victoria, Australia
| | - Paul A Gleeson
- The Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, Australia
| |
Collapse
|
37
|
Yuan J, Wang H, Wang Y, Wang Z, Huo Q, Dai X, Zhang J, Sun Y. Rapid Identification of 3,6'-Disinapoyl Sucrose Metabolites in Alzheimer's Disease Model Mice Using UHPLC-Orbitrap Mass Spectrometry. Molecules 2021; 27:114. [PMID: 35011346 PMCID: PMC8746568 DOI: 10.3390/molecules27010114] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is a degenerative disease of the central nervous system characterized by the progressive impairment of neural activity. Studies have shown that 3,6'-disinapoyl sucrose (DISS) can alleviate the pathological symptoms of AD through the activation of the cAMP/CREB/BDNF signaling pathway. However, the exact biochemical mechanisms of action of DISS are not clear. This study explores metabolism of DISS in an AD mouse model, induced by the microinjection of a lentiviral expression plasmid of the APPswe695 gene into CA1 of the hippocampus. After gavage administration of DISS (200 mg/kg), the kidneys, livers, brains, plasma, urine, and feces were collected for UHPLC-Orbitrap mass spectrometry analysis. Twenty metabolites, including the prototype drug of DISS, were positively or tentatively identified based on accurate mass measurements, characteristic fragmentation behaviors, and retention times. Thus, the metabolic pathways of DISS in AD mice were preliminarily elucidated through the identification of metabolites, such as ester bond cleavage, demethoxylation, demethylation, and sinapic acid-related products. Furthermore, differences in the in vivo distribution of several metabolites were observed between the model and sham control groups. These findings can provide a valuable reference for the pharmacological mechanisms and biosafety of DISS.
Collapse
Affiliation(s)
- Jiaqi Yuan
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing 100191, China; (J.Y.); (H.W.); (Y.W.); (Q.H.); (X.D.)
| | - Han Wang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing 100191, China; (J.Y.); (H.W.); (Y.W.); (Q.H.); (X.D.)
| | - Yunting Wang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing 100191, China; (J.Y.); (H.W.); (Y.W.); (Q.H.); (X.D.)
| | - Zijian Wang
- Beijing Research Institution of Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China;
| | - Qing Huo
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing 100191, China; (J.Y.); (H.W.); (Y.W.); (Q.H.); (X.D.)
| | - Xueling Dai
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing 100191, China; (J.Y.); (H.W.); (Y.W.); (Q.H.); (X.D.)
| | - Jiayu Zhang
- School of Pharmacy, Binzhou Medical University, Yantai 264003, China;
| | - Yaxuan Sun
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing 100191, China; (J.Y.); (H.W.); (Y.W.); (Q.H.); (X.D.)
| |
Collapse
|
38
|
Probiotics for Alzheimer's Disease: A Systematic Review. Nutrients 2021; 14:nu14010020. [PMID: 35010895 PMCID: PMC8746506 DOI: 10.3390/nu14010020] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 11/15/2021] [Accepted: 11/15/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer’s disease (AD) is the most common form of neurodegenerative disorders affecting mostly the elderly. It is characterized by the presence of Aβ and neurofibrillary tangles (NFT), resulting in cognitive and memory impairment. Research shows that alteration in gut microbial diversity and defects in gut brain axis are linked to AD. Probiotics are known to be one of the best preventative measures against cognitive decline in AD. Numerous in vivo trials and recent clinical trials have proven the effectiveness of selected bacterial strains in slowing down the progression of AD. It is proven that probiotics modulate the inflammatory process, counteract with oxidative stress, and modify gut microbiota. Thus, this review summarizes the current evidence, diversity of bacterial strains, defects of gut brain axis in AD, harmful bacterial for AD, and the mechanism of action of probiotics in preventing AD. A literature search on selected databases such as PubMed, Semantic Scholar, Nature, and Springer link have identified potentially relevant articles to this topic. However, upon consideration of inclusion criteria and the limitation of publication year, only 22 articles have been selected to be further reviewed. The search query includes few sets of keywords as follows. (1) Probiotics OR gut microbiome OR microbes AND (2) Alzheimer OR cognitive OR aging OR dementia AND (3) clinical trial OR in vivo OR animal study. The results evidenced in this study help to clearly illustrate the relationship between probiotic supplementation and AD. Thus, this systematic review will help identify novel therapeutic strategies in the future as probiotics are free from triggering any adverse effects in human body.
Collapse
|
39
|
Soto-Mercado V, Mendivil-Perez M, Velez-Pardo C, Jimenez-Del-Rio M. (-)-Epigallocatechin-3-Gallate Diminishes Intra-and Extracellular Amyloid-Induced Cytotoxic Effects on Cholinergic-like Neurons from Familial Alzheimer's Disease PSEN1 E280A. Biomolecules 2021; 11:biom11121845. [PMID: 34944489 PMCID: PMC8699501 DOI: 10.3390/biom11121845] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 11/26/2021] [Accepted: 11/26/2021] [Indexed: 12/26/2022] Open
Abstract
Alzheimer’s disease (AD) is a complex neurodegenerative disease characterized by functional disruption, death of cholinergic neurons (ChNs) because of intracellular and extracellular Aβ aggregates, and hyperphosphorylation of protein TAU (p-TAU). To date, there are no efficient therapies against AD. Therefore, new therapies for its treatment are in need. The goal of this investigation was to evaluate the effect of the polyphenol epigallocatechin-3-gallate (EGCG) on cholinergic-like neurons (ChLNs) bearing the mutation E280A in PRESENILIN 1 (PSEN1 E280A). To this aim, wild-type (WT) and PSEN1 E280A ChLNs were exposed to EGCG (5–50 μM) for 4 days. Untreated or treated neurons were assessed for biochemical and functional analysis. We found that EGCG (50 μM) significantly inhibited the aggregation of (i)sAPPβf, blocked p-TAU, increased ∆Ψm, decreased oxidation of DJ-1 at residue Cys106-SH, and inhibited the activation of transcription factor c-JUN and P53, PUMA, and CASPASE-3 in mutant ChLNs compared to WT. Although EGCG did not reduce (e)Aβ42, the polyphenol reversed Ca2+ influx dysregulation as a response to acetylcholine (ACh) stimuli in PSEN1 E280A ChLNs, inhibited the activation of transcription factor NF-κB, and reduced the secretion of pro-inflammatory IL-6 in wild-type astrocyte-like cells (ALCs) when exposed to mutant ChLNs culture supernatant. Taken together, our findings suggest that the EGCG might be a promising therapeutic approach for the treatment of FAD.
Collapse
|
40
|
Rab35 and glucocorticoids regulate APP and BACE1 trafficking to modulate Aβ production. Cell Death Dis 2021; 12:1137. [PMID: 34876559 PMCID: PMC8651661 DOI: 10.1038/s41419-021-04433-w] [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: 07/14/2021] [Revised: 11/08/2021] [Accepted: 11/23/2021] [Indexed: 11/08/2022]
Abstract
Chronic stress and elevated glucocorticoids (GCs), the major stress hormones, are risk factors for Alzheimer’s disease (AD) and promote AD pathomechanisms, including overproduction of toxic amyloid-β (Aβ) peptides and intraneuronal accumulation of hyperphosphorylated Tau protein. The latter is linked to downregulation of the small GTPase Rab35, which mediates Tau degradation via the endolysosomal pathway. Whether Rab35 is also involved in Aβ overproduction remains an open question. Here, we find that hippocampal Rab35 levels are decreased not only by stress/GC but also by aging, another AD risk factor. Moreover, we show that Rab35 negatively regulates Aβ production by sorting amyloid precursor protein (APP) and β-secretase (BACE1) out of the endosomal network, where they interact to produce Aβ. Interestingly, Rab35 coordinates distinct intracellular trafficking steps for BACE1 and APP, mediated by its effectors OCRL and ACAP2, respectively. Finally, we demonstrate that Rab35 overexpression prevents the amyloidogenic trafficking of APP and BACE1 induced by high GC levels. These studies identify Rab35 as a key regulator of APP processing and suggest that its downregulation may contribute to stress-related and AD-related amyloidogenesis.
Collapse
|
41
|
Hypoxia and the Kynurenine Pathway: Implications and Therapeutic Prospects in Alzheimer's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5522981. [PMID: 34804368 PMCID: PMC8598363 DOI: 10.1155/2021/5522981] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 06/09/2021] [Accepted: 07/06/2021] [Indexed: 02/06/2023]
Abstract
Neurodegenerative diseases (NDs) like Alzheimer's disease, multiple sclerosis, amyotrophic lateral sclerosis, Parkinson's disease, and Huntington's disease predominantly pose a significant socioeconomic burden. Characterized by progressive neural dysfunction coupled with motor or intellectual impairment, the pathogenesis of ND may result from contributions of certain environmental and molecular factors. One such condition is hypoxia, characterized by reduced organ/tissue exposure to oxygen. Reduced oxygen supply often occurs during the pathogenesis of ND and the aging process. Despite the well-established relationship between these two conditions (i.e., hypoxia and ND), the underlying molecular events or mechanisms connecting hypoxia to ND remain ill-defined. However, the relatedness may stem from the protective or deleterious effects of the transcription factor, hypoxia-inducible factor 1-alpha (HIF-1α). The upregulation of HIF-1α occurs in the pathogenesis of most NDs. The dual function of HIF-1α in acting as a "killer factor" or a "protective factor" depends on the prevailing local cellular condition. The kynurenine pathway is a metabolic pathway involved in the oxidative breakdown of tryptophan. It is essential in neurotransmission and immune function and, like hypoxia, associated with ND. Thus, a good understanding of factors, including hypoxia (i.e., the biochemical implication of HIF-1α) and kynurenine pathway activation in NDs, focusing on Alzheimer's disease could prove beneficial to new therapeutic approaches for this disease, thus the aim of this review.
Collapse
|
42
|
Chang W, Xiao D, Fang X, Wang J. Phospholipids in small extracellular vesicles: emerging regulators of neurodegenerative diseases and cancer. Cytotherapy 2021; 24:93-100. [PMID: 34742629 DOI: 10.1016/j.jcyt.2021.09.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 08/26/2021] [Accepted: 09/22/2021] [Indexed: 12/16/2022]
Abstract
Small extracellular vesicles (sEVs) are generated by almost all cell types. They have a bilayer membrane structure that is similar to cell membranes. Thus, the phospholipids contained in sEVs are the main components of cell membranes and function as structural support elements. However, as in-depth research on sEV membrane components is conducted, some phospholipids have been found to participate in cellular biological processes and function as targets for cell-cell communication. Currently, sEVs are being developed as part of drug delivery systems and diagnostic factors for various diseases, especially neurodegenerative diseases and cancer. An understanding of the physiological and pathological roles of sEV phospholipids in cellular processes is essential for their future medical application. In this review, the authors discuss phospholipid components in sEVs of different origins and summarize the roles of phospholipids in sEV biogenesis. The authors further collect the current knowledge on the functional roles of sEV phospholipids in cell-cell communication and bioactivities as signals regulating neurodegenerative diseases and cancer and the possibility of using sEV phospholipids as biomarkers or in drug delivery systems for cancer diagnosis and treatment. Knowledge of sEV phospholipids is important to help us identify directions for future studies.
Collapse
Affiliation(s)
- Wenguang Chang
- Institute for Translational Medicine, The Affiliated Hospital, College of Medicine, Qingdao University, Qingdao, China.
| | - Dandan Xiao
- Institute for Translational Medicine, The Affiliated Hospital, College of Medicine, Qingdao University, Qingdao, China; School of Basic Medical Sciences, College of Medicine, Qingdao University, Qingdao, China
| | - Xinyu Fang
- Institute for Translational Medicine, The Affiliated Hospital, College of Medicine, Qingdao University, Qingdao, China; School of Basic Medical Sciences, College of Medicine, Qingdao University, Qingdao, China
| | - Jianxun Wang
- School of Basic Medical Sciences, College of Medicine, Qingdao University, Qingdao, China
| |
Collapse
|
43
|
Chen J, Luo B, Zhong BR, Li KY, Wen QX, Song L, Xiang XJ, Zhou GF, Hu LT, Deng XJ, Ma YL, Chen GJ. Sulfuretin exerts diversified functions in the processing of amyloid precursor protein. Genes Dis 2021; 8:867-881. [PMID: 34522714 PMCID: PMC8427253 DOI: 10.1016/j.gendis.2020.11.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/14/2020] [Accepted: 11/16/2020] [Indexed: 01/03/2023] Open
Abstract
Sulfuretin is a flavonoid that protects cell from damage induced by reactive oxygen species and inflammation. In this study, we investigated the role of sulfuretin in the processing of amyloid precursor protein (APP), in association with the two catalytic enzymes the α-secretase a disintegrin and metalloproteinase (ADAM10), and the beta-site APP cleaving enzyme 1 (BACE1) that play important roles in the generation of β amyloid protein (Aβ) in Alzheimer's disease (AD). We found that sulfuretin increased the levels of the immature but not the mature form of ADAM10 protein. The enhanced ADAM10 transcription by sulfuretin was mediated by the nucleotides −444 to −300 in the promoter region, and was attenuated by silencing or mutation of transcription factor retinoid X receptor (RXR) and by GW6471, a specific inhibitor of peroxisome proliferator-activated receptor α (PPAR-α). We further found that sulfuretin preferentially increased protein levels of the immature form of APP (im-APP) but significantly reduced those of BACE1, sAPPβ and β-CTF, whereas Aβ1-42 levels were slightly increased. Finally, the effect of sulfuretin on BACE1 and im-APP was selectively attenuated by the translation inhibitor cycloheximide and by lysosomal inhibitor chloroquine, respectively. Taken together, (1) RXR/PPAR-α signaling was involved in sulfuretin-mediated ADAM10 transcription. (2) Alteration of Aβ protein level by sulfuretin was not consistent with that of ADAM10 and BACE1 protein levels, but was consistent with the elevated level of im-APP protein, suggesting that im-APP, an isoform mainly localized to trans-Golgi network, plays an important role in Aβ generation.
Collapse
Affiliation(s)
- Jian Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, PR China
| | - Biao Luo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, PR China
| | - Bi-Rou Zhong
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, PR China
| | - Kun-Yi Li
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, PR China
| | - Qi-Xin Wen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, PR China
| | - Li Song
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, PR China
| | - Xiao-Jiao Xiang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, PR China
| | - Gui-Feng Zhou
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, PR China
| | - Li-Tian Hu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, PR China.,Department of Neurology, Nanchong Central Hospital, The Second Clinical College of North Sichuan Medical College, Nanchong, Sichuan Province, 637000, PR China
| | - Xiao-Juan Deng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, PR China
| | - Yuan-Lin Ma
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, PR China
| | - Guo-Jun Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing Key Laboratory of Neurology, 1 Youyi Road, Chongqing, 400016, PR China
| |
Collapse
|
44
|
Komaki K, Takano T, Sato Y, Asada A, Ikeda S, Yamada K, Wei R, Huo A, Fukuchi A, Saito T, Ando K, Murayama S, Araki W, Kametani F, Hasegawa M, Iwatsubo T, Tomomura M, Fukuda M, Hisanaga SI. Lemur tail kinase 1 (LMTK1) regulates the endosomal localization of β-secretase BACE1. J Biochem 2021; 170:729-738. [PMID: 34523681 DOI: 10.1093/jb/mvab094] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 08/21/2021] [Indexed: 12/15/2022] Open
Abstract
Lemur tail kinase 1 (LMTK1), previously called apoptosis-associated tyrosine kinase (AATYK), is an endosomal Ser/Thr kinase. We recently reported that LMTK1 regulates axon outgrowth, dendrite arborization and spine formation via Rab11-mediated vesicle transport. Rab11, a small GTPase regulating recycling endosome trafficking, is shown to be associated with late-onset Alzheimer's disease (LOAD). In fact, genome-wide association studies identified many proteins regulating vesicle transport as risk factors for LOAD. Furthermore, LMTK1 has been reported to be a risk factor for frontotemporal dementia. Then, we hypothesized that LMTK1 contributes to AD development through vesicle transport and examined the effect of LMTK1 on the cellular localization of AD-related proteins, amyloid precursor protein (APP) and β-site APP cleaving enzyme 1 (BACE1). The β-cleavage of APP by BACE1 is the initial and rate-limiting step in Aβ generation. We found that LMTK1 accumulated BACE1, but not APP, to the perinuclear endosomal compartment, whereas the kinase-negative (kn) mutant of LMTK1A did not. The β-C-terminal fragment was prone to increase under overexpression of LMTK1A kn. Moreover, the expression level of LMTK1A was reduced in AD brains. These results suggest the possibility that LMTK1 is involved in AD development through the regulation of the proper endosomal localization of BACE1.
Collapse
Affiliation(s)
- Keisuke Komaki
- Department of Biological Sciences, Tokyo Metropolitan University, Minami-osawa, Hachioji, Tokyo 192-0397, Japan
| | - Tetsuya Takano
- Department of Biological Sciences, Tokyo Metropolitan University, Minami-osawa, Hachioji, Tokyo 192-0397, Japan
| | - Yutaka Sato
- Department of Biological Sciences, Tokyo Metropolitan University, Minami-osawa, Hachioji, Tokyo 192-0397, Japan
| | - Akiko Asada
- Department of Biological Sciences, Tokyo Metropolitan University, Minami-osawa, Hachioji, Tokyo 192-0397, Japan
| | - Shikito Ikeda
- Department of Biological Sciences, Tokyo Metropolitan University, Minami-osawa, Hachioji, Tokyo 192-0397, Japan
| | - Kaoru Yamada
- Department of Neuropathology, Graduate School of Medicine, the University of Tokyo, Bunkyo, Tokyo 113-0033, Japan
| | - Ran Wei
- Department of Biological Sciences, Tokyo Metropolitan University, Minami-osawa, Hachioji, Tokyo 192-0397, Japan
| | - Anni Huo
- Department of Biological Sciences, Tokyo Metropolitan University, Minami-osawa, Hachioji, Tokyo 192-0397, Japan
| | - Aoi Fukuchi
- Department of Biological Sciences, Tokyo Metropolitan University, Minami-osawa, Hachioji, Tokyo 192-0397, Japan
| | - Taro Saito
- Department of Biological Sciences, Tokyo Metropolitan University, Minami-osawa, Hachioji, Tokyo 192-0397, Japan
| | - Kanae Ando
- Department of Biological Sciences, Tokyo Metropolitan University, Minami-osawa, Hachioji, Tokyo 192-0397, Japan
| | - Shigeo Murayama
- Tokyo Metropolitan Institute of Gerontology, Itabashi, Tokyo 173-0015, Japan
| | - Wataru Araki
- Department of Demyelinating Disease and Aging, National Institute of Neuroscience, NCNP, Kodaira, Tokyo 187-8502, Japan
| | - Fuyuki Kametani
- Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo 156-8506, Japan
| | - Masato Hasegawa
- Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo 156-8506, Japan
| | - Takeshi Iwatsubo
- Department of Neuropathology, Graduate School of Medicine, the University of Tokyo, Bunkyo, Tokyo 113-0033, Japan
| | - Mineko Tomomura
- Department of Oral Health Sciences, Meikai University School of Health Sciences, Urayasu, Chiba 279-9950, Japan
| | - Mitsunori Fukuda
- Department of Integrative Life Sciences, Graduate School of Life Sciences, Tohoku University, Aoba-ku, Sendai, Miyagi 980-8578, Japan
| | - Shin-Ichi Hisanaga
- Department of Biological Sciences, Tokyo Metropolitan University, Minami-osawa, Hachioji, Tokyo 192-0397, Japan.,Tokyo Metropolitan Institute of Medical Science, Setagaya, Tokyo 156-8506, Japan
| |
Collapse
|
45
|
Wang S, Ma J, Zeng Y, Zhou G, Wang Y, Zhou W, Sun X, Wu M. Icariin, an Up-and-Coming Bioactive Compound Against Neurological Diseases: Network Pharmacology-Based Study and Literature Review. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:3619-3641. [PMID: 34447243 PMCID: PMC8384151 DOI: 10.2147/dddt.s310686] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 06/25/2021] [Indexed: 12/12/2022]
Abstract
Icariin is a biologically active substance in Epimedii herba that is used for the treatment of neurologic disorders. However, a comprehensive analysis of the molecular mechanisms of icariin is lacking. In this review, we present a brief history of the use of icariin for medicinal purposes; describe the active chemical components of Epimedii herba; and examine the evidence from experimental studies that have uncovered molecular targets of icariin in different diseases. We also constructed a protein–protein interaction network and carried out Gene Ontology and Kyoto Encyclopedia of Genes and Genomes functional enrichment analyses to predict the therapeutic actions of icariin in nervous system diseases including Alzheimer disease, Parkinson disease, ischemic stroke, depressive disorder, multiple sclerosis, glioblastoma, and hereditary spastic paraplegias. The results of our analyses can guide future studies on the application of icariin to the treatment of neurologic disorders.
Collapse
Affiliation(s)
- Shuangqiu Wang
- Department of Neurology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, People's Republic of China.,Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210046, People's Republic of China.,State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, People's Republic of China
| | - Jiarui Ma
- Provincial Key Laboratory of Drug Target and Drug for Degenerative Disease, School of Medicine and Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, People's Republic of China
| | - Yanqi Zeng
- First Clinical Medical School, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, People's Republic of China
| | - Guowei Zhou
- Department of General Surgery, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, People's Republic of China
| | - Yuxuan Wang
- Department of Neurology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, People's Republic of China.,Key Laboratory of Acupuncture and Medicine Research of Ministry of Education, Nanjing University of Chinese Medicine, Nanjing, 210046, People's Republic of China.,State Key Laboratory Cultivation Base for TCM Quality and Efficacy, Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, People's Republic of China
| | - Wenjuan Zhou
- First Clinical Medical School, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, People's Republic of China
| | - Xiaohe Sun
- First Clinical Medical School, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, People's Republic of China
| | - Minghua Wu
- Department of Neurology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, People's Republic of China.,First Clinical Medical School, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210046, Jiangsu, People's Republic of China
| |
Collapse
|
46
|
Fourriere L, Gleeson PA. Amyloid β production along the neuronal secretory pathway: Dangerous liaisons in the Golgi? Traffic 2021; 22:319-327. [PMID: 34189821 DOI: 10.1111/tra.12808] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/24/2021] [Accepted: 06/27/2021] [Indexed: 12/11/2022]
Abstract
β-amyloid peptides (Aβ) are generated in intracellular compartments of neurons and secreted to form cytotoxic fibrils and plaques. Dysfunctional membrane trafficking contributes to aberrant Aβ production and Alzheimer's disease. Endosomes represent one of the major sites for Aβ production and recently the Golgi has re-emerged also as a major location for amyloid precursor protein (APP) processing and Aβ production. Based on recent findings, here we propose that APP processing in the Golgi is finely tuned by segregating newly-synthesised APP and the β-secretase BACE1 within the Golgi and into distinct trans-Golgi network transport pathways. We hypothesise that there are multiple mechanisms responsible for segregating APP and BACE1 during transit through the Golgi, and that perturbation in Golgi morphology associated with Alzheimer's disease, and or changes in cholesterol metabolism associated with Alzheimer's disease risk factors, may lead to a loss of partitioning and enhanced Aβ production.
Collapse
Affiliation(s)
- Lou Fourriere
- The Department of Biochemistry and Pharmacology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | - Paul A Gleeson
- The Department of Biochemistry and Pharmacology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
47
|
Lin T, Tjernberg LO, Schedin-Weiss S. Neuronal Trafficking of the Amyloid Precursor Protein-What Do We Really Know? Biomedicines 2021; 9:801. [PMID: 34356865 PMCID: PMC8301342 DOI: 10.3390/biomedicines9070801] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/29/2021] [Accepted: 07/03/2021] [Indexed: 11/23/2022] Open
Abstract
Alzheimer's disease (AD) is the most common type of dementia, contributing to 60-80% of cases. It is a neurodegenerative disease that usually starts symptomless in the first two to three decades and then propagates into a long-term, irreversible disease, resulting in the progressive loss of memory, reasoning, abstraction and language capabilities. It is a complex disease, involving a large number of entangled players, and there is no effective treatment to cure it or alter its progressive course. Therefore, a thorough understanding of the disease pathology and an early diagnosis are both necessary. AD has two significant pathological hallmarks: extracellular senile plaques composed of amyloid β-peptide (Aβ) and intracellular neurofibrillary tangles composed of hyperphosphorylated tau protein, and the aggregation of Aβ, which starts in earlier stages, is usually claimed to be the primary cause of AD. Secretases that cleave Aβ precursor protein (APP) and produce neurotoxic Aβ reside in distinct organelles of the cell, and current concepts suggest that APP moves between distinct intracellular compartments. Obviously, APP transport and processing are intimately related processes that cannot be dissociated from each other, and, thus, how and where APP is transported determines its processing fate. In this review, we summarize critical mechanisms underlying neuronal APP transport, which we divide into separate parts: (1) secretory pathways and (2) endocytic and autophagic pathways. We also include two lipoprotein receptors that play essential roles in APP transport: sorting-related receptor with A-type repeats and sortilin. Moreover, we consider here some major disruptions in the neuronal transport of APP that contribute to AD physiology and pathology. Lastly, we discuss current methods and technical difficulties in the studies of APP transport.
Collapse
Affiliation(s)
| | - Lars O. Tjernberg
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Stockholm, Sweden;
| | - Sophia Schedin-Weiss
- Division of Neurogeriatrics, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, 17164 Stockholm, Sweden;
| |
Collapse
|
48
|
Cunha S, Forbes B, Sousa Lobo JM, Silva AC. Improving Drug Delivery for Alzheimer's Disease Through Nose-to-Brain Delivery Using Nanoemulsions, Nanostructured Lipid Carriers (NLC) and in situ Hydrogels. Int J Nanomedicine 2021; 16:4373-4390. [PMID: 34234432 PMCID: PMC8256381 DOI: 10.2147/ijn.s305851] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/29/2021] [Indexed: 12/18/2022] Open
Abstract
Current treatments for Alzheimer's disease (AD) attenuate the progression of symptoms and aim to improve the patient's quality of life. Licensed medicines are mostly for oral administration and are limited by the difficulty in crossing the blood-brain barrier (BBB). Here in, the nasal route has been explored as an alternative pathway that allows drugs to be directly delivered to the brain via the nasal cavity. However, clearance mechanisms in the nasal cavity impair the delivery of drugs to the brain and limit their bioavailability. To optimize nose-to-brain delivery, formulations of lipid-based nanosystems, namely nanoemulsions and nanostructured lipid carriers (NLC), formulated in situ gelling hydrogels have been proposed as approaches for nose-to-brain delivery. These formulations possess characteristics that facilitate drug transport directly to the brain, minimizing side effects and maximizing therapeutic benefits. It has been recommended that the manufacture of these drug delivery systems follows the quality by design (QbD) approach based on nasal administration requirements. This review provides an insight into the current knowledge of the AD, highlighting the need for an effective drug delivery to the brain. Considering the mounting interest in the use of nanoemulsions and NLC for nose-to-brain delivery, a description of drug transport pathways in the nasal cavity and the application of these nanosystems and their in situ hydrogels through the intranasal route are presented. Relevant preclinical studies are summarised, and the future prospects for the use of lipid-based nanosystems in the treatment of AD are emphasized.
Collapse
Affiliation(s)
- Sara Cunha
- UCIBIO/REQUIMTE, MEDTECH Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, 4050-313, Portugal
| | - Ben Forbes
- Institute of Pharmaceutical Science, Faculty of Life Sciences and Medicine, King’s College London, London, SE1 9NH, UK
| | - José Manuel Sousa Lobo
- UCIBIO/REQUIMTE, MEDTECH Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, 4050-313, Portugal
| | - Ana Catarina Silva
- UCIBIO/REQUIMTE, MEDTECH Laboratory of Pharmaceutical Technology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, 4050-313, Portugal
- UFP Energy, Environment and Health Research Unit (FP ENAS), Fernando Pessoa University, Porto, 4249-004, Portugal
| |
Collapse
|
49
|
Pannuzzo M. Beta-amyloid pore linked to controlled calcium influx into the cell: A new paradigm for Alzheimer's Disease. Alzheimers Dement 2021; 18:191-196. [PMID: 34051062 PMCID: PMC9290490 DOI: 10.1002/alz.12373] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/20/2021] [Accepted: 04/16/2021] [Indexed: 11/28/2022]
Abstract
Despite tremendous worldwide efforts, clinical trials assessing Alzheimer's disease (AD)‐related therapeutics have been relentlessly unsuccessful. Hence, there is an urgent need to challenge old hypotheses with novel paradigms. An emerging concept is that the amyloid‐beta (Aβ) peptide, which was until recently deemed a major player in the cause of AD, may instead modulate synaptic plasticity and protect against excitotoxicity. The link between Aβ‐mediated synaptic plasticity and Aβ trafficking is central for understanding AD pathogenesis and remains a perplexing relationship. The crossover between Aβ pathological and physiological roles is subtle and remains controversial. Based on existing literature, as a signaling molecule, Aβ is proposed to modulate its own turnover and synaptic plasticity through what is currently believed to be the cause of AD: the transient formation of pore‐like oligomers. A change of perspective regarding how Aβ pores exert a protective function will unavoidably revolutionize the entire field of anti‐amyloid drug development.
Collapse
Affiliation(s)
- Martina Pannuzzo
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| |
Collapse
|
50
|
Walsh RB, Dresselhaus EC, Becalska AN, Zunitch MJ, Blanchette CR, Scalera AL, Lemos T, Lee SM, Apiki J, Wang S, Isaac B, Yeh A, Koles K, Rodal AA. Opposing functions for retromer and Rab11 in extracellular vesicle traffic at presynaptic terminals. J Cell Biol 2021; 220:212178. [PMID: 34019080 PMCID: PMC8144913 DOI: 10.1083/jcb.202012034] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 03/15/2021] [Accepted: 04/29/2021] [Indexed: 12/18/2022] Open
Abstract
Neuronal extracellular vesicles (EVs) play important roles in intercellular communication and pathogenic protein propagation in neurological disease. However, it remains unclear how cargoes are selectively packaged into neuronal EVs. Here, we show that loss of the endosomal retromer complex leads to accumulation of EV cargoes including amyloid precursor protein (APP), synaptotagmin-4 (Syt4), and neuroglian (Nrg) at Drosophila motor neuron presynaptic terminals, resulting in increased release of these cargoes in EVs. By systematically exploring known retromer-dependent trafficking mechanisms, we show that EV regulation is separable from several previously identified roles of neuronal retromer. Conversely, mutations in rab11 and rab4, regulators of endosome-plasma membrane recycling, cause reduced EV cargo levels, and rab11 suppresses cargo accumulation in retromer mutants. Thus, EV traffic reflects a balance between Rab4/Rab11 recycling and retromer-dependent removal from EV precursor compartments. Our data shed light on previous studies implicating Rab11 and retromer in competing pathways in Alzheimer's disease, and suggest that misregulated EV traffic may be an underlying defect.
Collapse
Affiliation(s)
- Rylie B Walsh
- Department of Biology, Brandeis University, Waltham, MA
| | | | | | | | | | - Amy L Scalera
- Department of Biology, Brandeis University, Waltham, MA
| | - Tania Lemos
- Department of Biology, Brandeis University, Waltham, MA
| | - So Min Lee
- Department of Biology, Brandeis University, Waltham, MA
| | - Julia Apiki
- Department of Biology, Brandeis University, Waltham, MA
| | - ShiYu Wang
- Department of Biology, Brandeis University, Waltham, MA
| | - Berith Isaac
- Department of Biology, Brandeis University, Waltham, MA
| | - Anna Yeh
- Department of Biology, Brandeis University, Waltham, MA
| | - Kate Koles
- Department of Biology, Brandeis University, Waltham, MA
| | | |
Collapse
|