1
|
Wolfstetter G, Masudi T, Uçkun E, Zhu JY, Yi M, Anthonydhason V, Guan J, Sonnenberg H, Han Z, Palmer RH. Alk Tango reveals a role for Jeb/Alk signaling in the Drosophila heart. Cell Commun Signal 2025; 23:229. [PMID: 40382638 DOI: 10.1186/s12964-025-02150-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 03/11/2025] [Indexed: 05/20/2025] Open
Abstract
Anaplastic lymphoma kinase (Alk) signaling is important in a variety of biological contexts such as cell type specification, regulation of metabolic and endocrine programs, behavior, and cancer. In this work, we generated a Tango GPCR assay-based, dimerization-sensitive Alk activity reporter (AlkTango) and followed receptor activation throughout Drosophila development. AlkTango reports Alk activation in embryonic and larval tissues previously linked to Alk signaling. Remarkably, AlkTango was active in the heart of Drosophila larvae and adult flies. We show that cardiomyocytes express Alk from late embryonic stages to adulthood, while jeb expression in pericardial cells coincided with AlkTango activity. Perturbation of cardiac Alk signaling leads to decreased adult survival as well as lower fitness and increased lethality in response to heat stress. In keeping with a role for Alk, heart measurements reveal arrythmia and irregular muscle contraction upon ligand stimulation. Finally, activation of cardiac Alk signaling induces hyperplasia in the accessory wing hearts of adult flies.
Collapse
Affiliation(s)
- G Wolfstetter
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine at the Sahlgrenska Academy, University of Gothenburg, 41390, Gothenburg, Sweden.
| | - T Masudi
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine at the Sahlgrenska Academy, University of Gothenburg, 41390, Gothenburg, Sweden
| | - E Uçkun
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine at the Sahlgrenska Academy, University of Gothenburg, 41390, Gothenburg, Sweden
| | - J Y Zhu
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - M Yi
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine at the Sahlgrenska Academy, University of Gothenburg, 41390, Gothenburg, Sweden
| | - V Anthonydhason
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine at the Sahlgrenska Academy, University of Gothenburg, 41390, Gothenburg, Sweden
- School of Biomedical Sciences and Pharmacy, College of Health, Medicine and Wellbeing, The University of Newcastle, Callaghan, NSW, 2308, Australia
| | - J Guan
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine at the Sahlgrenska Academy, University of Gothenburg, 41390, Gothenburg, Sweden
| | - H Sonnenberg
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine at the Sahlgrenska Academy, University of Gothenburg, 41390, Gothenburg, Sweden
| | - Z Han
- Center for Precision Disease Modeling, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
- Division of Endocrinology, Diabetes and Nutrition, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, 21201, USA
| | - R H Palmer
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine at the Sahlgrenska Academy, University of Gothenburg, 41390, Gothenburg, Sweden.
| |
Collapse
|
2
|
Li S, Li X, Ma L, Luo Z, Yin F, Zhang Y, Chen Y, Wan S, Zhou H, Wang X, Kong L. Polypharmacological Drug Design Guided by Integrating Phenotypic and Restricted Fragment Docking Strategies. J Med Chem 2024; 67:21049-21069. [PMID: 39300597 DOI: 10.1021/acs.jmedchem.4c01731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
Polypharmacological drugs are of great value for treating complex human diseases by the combinative modulation of several biological targets. However, multitarget drug design with more than two targets is challenging and generally discovered by serendipity. Herein, a restricted fragment docking (RFD) computational method combined with a phenotypic discovery approach was developed for rational polypharmacological drug design. Via genetic and drug combination studies in a microglial phenotype, we first identified novel synergistic effects by triple target modulation toward RIPK1, MAP4K4, and ALK. Drawing on the RFD method to explore virtual chemical spaces in three target pockets, we identified a lead compound, LP-10d, that precisely modulated RIPK1/MAP4K4/ALK for synergistic microglial protection with low nanomolar potency. LP-10d showed polypharmacology against multiple neuropathologies in the 3xTg Alzheimer's disease mouse model. Our study revealed a potential application of the RFD method, which is valuable to further polypharmacological drug discovery involved in clinical studies for treating complex human diseases.
Collapse
Affiliation(s)
- Shang Li
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Xinxin Li
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Liangliang Ma
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Zhongwen Luo
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Fucheng Yin
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Yonglei Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Yifan Chen
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Siyuan Wan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Han Zhou
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Xiaobing Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| | - Lingyi Kong
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Bioactive Natural Product Research, Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 210009, People's Republic of China
| |
Collapse
|
3
|
Yaghoobi A, Malekpour SA. Unraveling the genetic architecture of blood unfolded p-53 among non-demented elderlies: novel candidate genes for early Alzheimer's disease. BMC Genomics 2024; 25:440. [PMID: 38702606 PMCID: PMC11067101 DOI: 10.1186/s12864-024-10363-6] [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: 09/15/2023] [Accepted: 04/29/2024] [Indexed: 05/06/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is a heritable neurodegenerative disease whose long asymptomatic phase makes the early diagnosis of it pivotal. Blood U-p53 has recently emerged as a superior predictive biomarker for AD in the early stages. We hypothesized that genetic variants associated with blood U-p53 could reveal novel loci and pathways involved in the early stages of AD. RESULTS We performed a blood U-p53 Genome-wide association study (GWAS) on 484 healthy and mild cognitively impaired subjects from the ADNI cohort using 612,843 Single nucleotide polymorphisms (SNPs). We performed a pathway analysis and prioritized candidate genes using an AD single-cell gene program. We fine-mapped the intergenic SNPs by leveraging a cell-type-specific enhancer-to-gene linking strategy using a brain single-cell multimodal dataset. We validated the candidate genes in an independent brain single-cell RNA-seq and the ADNI blood transcriptome datasets. The rs279686 between AASS and FEZF1 genes was the most significant SNP (p-value = 4.82 × 10-7). Suggestive pathways were related to the immune and nervous systems. Twenty-three candidate genes were prioritized at 27 suggestive loci. Fine-mapping of 5 intergenic loci yielded nine cell-specific candidate genes. Finally, 15 genes were validated in the independent single-cell RNA-seq dataset, and five were validated in the ADNI blood transcriptome dataset. CONCLUSIONS We underlined the importance of performing a GWAS on an early-stage biomarker of AD and leveraging functional omics datasets for pinpointing causal genes in AD. Our study prioritized nine genes (SORCS1, KIF5C, TMEFF2, TMEM63C, HLA-E, ATAT1, TUBB, ARID1B, and RUNX1) strongly implicated in the early stages of AD.
Collapse
Affiliation(s)
- Arash Yaghoobi
- School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, 19395-5746, Iran
| | - Seyed Amir Malekpour
- School of Biological Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, 19395-5746, Iran.
| |
Collapse
|
4
|
Hu Y, Chang L, Zhu Y, Geng X, Liu Z, Wang R, Wang Y, Zhao BQ, Fan W. Inhibition of Anaplastic Lymphoma Kinase Protects From Ischemic Stroke. Stroke 2024; 55:1075-1085. [PMID: 38445502 DOI: 10.1161/strokeaha.123.045991] [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: 09/06/2023] [Accepted: 02/01/2024] [Indexed: 03/07/2024]
Abstract
BACKGROUND Ischemic stroke is often accompanied by oxidative stress and inflammatory response, both of which work synergistically to exacerbate the disruption of the blood-brain barrier and ischemic brain injury. ALK (anaplastic lymphoma kinase), a cancer-associated receptor tyrosine kinase, was found to play a role in oxidative stress and inflammation. In this study, we investigated the role of ALK inhibition in a murine model of ischemic stroke. METHODS Focal cerebral ischemia was induced by temporary occlusion of the right middle cerebral artery in mice with a filament. The ALK inhibitor alectinib was administered following the stroke. ALOX15 (arachidonic acid 15-lipoxygenase) was overexpressed by adenovirus injection. The immunohistochemistry, Western blot, oxidative stress, inflammation, blood-brain barrier leakage, infarct volume, and functional outcomes were determined. RESULTS We found that the expression of ALK was markedly increased in the neurovascular unit after cerebral ischemia. Treatment with the ALK inhibitor alectinib reduced the accumulation of reactive oxygen species, lipid peroxidation, and oxidative DNA, increased the vascular levels of antioxidant enzymes, inactivated the vascular NLRP3 (nucleotide-binding oligomerization domain-like receptor protein 3) inflammasome pathway, and reduced vascular inflammation (ICAM-1 [intercellular adhesion molecule-1] and MCP-1 [monocyte chemoattractant protein-1]) after ischemia. Moreover, alectinib reduced the loss of cerebrovascular integrity and blood-brain barrier damage, consequently decreasing brain infarction and neurological deficits. Furthermore, alectinib reduced stroke-evoked ALOX15 expression, whereas virus-mediated overexpression of ALOX15 abolished alectinib-dependent inhibition of oxidative stress and vascular inflammation, blood-brain barrier protection, and neuroprotection, suggesting the protective effects of alectinib for stroke may involve ALOX15. CONCLUSIONS Our findings demonstrated that alectinib protects from stroke by regulating ischemic signaling cascades and suggest that ALK may be a novel therapeutic target for ischemic stroke.
Collapse
Affiliation(s)
- Yue Hu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, China (Y.H., L.C., Y.Z., X.G., Z.L., R.W., B.-Q.Z., W.F.)
| | - Luping Chang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, China (Y.H., L.C., Y.Z., X.G., Z.L., R.W., B.-Q.Z., W.F.)
| | - Yuanbo Zhu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, China (Y.H., L.C., Y.Z., X.G., Z.L., R.W., B.-Q.Z., W.F.)
| | - Xue Geng
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, China (Y.H., L.C., Y.Z., X.G., Z.L., R.W., B.-Q.Z., W.F.)
| | - Zhongwang Liu
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, China (Y.H., L.C., Y.Z., X.G., Z.L., R.W., B.-Q.Z., W.F.)
| | - Ranran Wang
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, China (Y.H., L.C., Y.Z., X.G., Z.L., R.W., B.-Q.Z., W.F.)
| | - Yiheng Wang
- Institute of Neuroscience and Third Affiliated Hospital, Zhengzhou University, China (Y.W.)
| | - Bing-Qiao Zhao
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, China (Y.H., L.C., Y.Z., X.G., Z.L., R.W., B.-Q.Z., W.F.)
| | - Wenying Fan
- Department of Translational Neuroscience, Jing'an District Centre Hospital of Shanghai, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Fudan University, China (Y.H., L.C., Y.Z., X.G., Z.L., R.W., B.-Q.Z., W.F.)
| |
Collapse
|
5
|
Dourlen P. Identification of Tau Toxicity Modifiers in the Drosophila Eye. Methods Mol Biol 2024; 2754:483-498. [PMID: 38512684 DOI: 10.1007/978-1-0716-3629-9_27] [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] [Indexed: 03/23/2024]
Abstract
Drosophila is a powerful model to study human diseases thanks to its genetic tools and ease of screening. Human genes can be expressed in targeted organs and their toxicity assessed on easily scorable external phenotypes that can be used as readouts to perform genetic screens of toxicity modifiers. In this chapter, I describe how to express human Tau protein in the Drosophila eye, assess protein expression by Western blot, assess Tau toxicity by quantifying the size of the Tau-induced rough eye, and perform a genetic screen of modifiers of Tau toxicity in the Drosophila eye.
Collapse
Affiliation(s)
- Pierre Dourlen
- Univ. Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167 - RID-AGE - Facteurs de risque et déterminants moléculaires des maladies liées au vieillissement, Lille, France.
| |
Collapse
|
6
|
Mesquita FP, Lima LB, da Silva EL, Souza PFN, de Moraes MEA, Burbano RMR, Montenegro RC. A Review on Anaplastic Lymphoma Kinase (ALK) Rearrangements and Mutations: Implications for Gastric Carcinogenesis and Target Therapy. Curr Protein Pept Sci 2024; 25:539-552. [PMID: 38424421 DOI: 10.2174/0113892037291318240130103348] [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: 11/04/2023] [Revised: 01/10/2024] [Accepted: 01/17/2024] [Indexed: 03/02/2024]
Abstract
Gastric adenocarcinoma is a complex disease with diverse genetic modifications, including Anaplastic Lymphoma Kinase (ALK) gene changes. The ALK gene is located on chromosome 2p23 and encodes a receptor tyrosine kinase that plays a crucial role in embryonic development and cellular differentiation. ALK alterations can result from gene fusion, mutation, amplification, or overexpression in gastric adenocarcinoma. Fusion occurs when the ALK gene fuses with another gene, resulting in a chimeric protein with constitutive kinase activity and promoting oncogenesis. ALK mutations are less common but can also result in the activation of ALK signaling pathways. Targeted therapies for ALK variations in gastric adenocarcinoma have been developed, including ALK inhibitors that have shown promising results in pre-clinical studies. Future studies are needed to elucidate the ALK role in gastric cancer and to identify predictive biomarkers to improve patient selection for targeted therapy. Overall, ALK alterations are a relevant biomarker for gastric adenocarcinoma treatment and targeted therapies for ALK may improve patients' overall survival.
Collapse
Affiliation(s)
- Felipe Pantoja Mesquita
- Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE, 60430-275, Brazil
| | - Luina Benevides Lima
- Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE, 60430-275, Brazil
| | - Emerson Lucena da Silva
- Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE, 60430-275, Brazil
| | - Pedro Filho Noronha Souza
- Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE, 60430-275, Brazil
| | | | - Rommel Mario Rodrigues Burbano
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belém, Brazil
- Molecular Biology Laboratory, Ophir Loyola Hospital, Belém, Brazil
| | - Raquel Carvalho Montenegro
- Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE, 60430-275, Brazil
- Latinoamericana de Implementación y Validación de guias clinicas Farmacogenomicas (RELIVAF), Brazil
| |
Collapse
|
7
|
Tian S, Ye T, Cheng X. The behavioral, pathological and therapeutic features of the triple transgenic Alzheimer's disease (3 × Tg-AD) mouse model strain. Exp Neurol 2023; 368:114505. [PMID: 37597764 DOI: 10.1016/j.expneurol.2023.114505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 08/07/2023] [Accepted: 08/15/2023] [Indexed: 08/21/2023]
Abstract
As a classic animal model of Alzheimer's disease (AD), the 3 × Tg-AD mouse not only recapitulates most of anatomical hallmarks observed in AD pathology but also displays cognitive alterations in memory and learning tasks. The 3 × Tg-AD can better show the two characteristics of AD, amyloid β (Aβ) and neurofibrillary tangles (NFT). Therefore, 3 × Tg-AD strain is widely used in AD pathogenesis research and new drug development of AD. In this paper, the construction methods, pathological changes, and treatment characteristics of 3 × Tg-AD mouse models commonly used in AD research are summarized and commented, hoping to provide reference for researchers to choose and establish experimental patterns.
Collapse
Affiliation(s)
- Sheng Tian
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, PR China; Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250300, China
| | - Tianyuan Ye
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250300, China
| | - Xiaorui Cheng
- Faculty of Chinese Medicine and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao 999078, PR China; Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250300, China.
| |
Collapse
|
8
|
Kim SH, Cho YS, Kim Y, Park J, Yoo SM, Gwak J, Kim Y, Gwon Y, Kam TI, Jung YK. Endolysosomal impairment by binding of amyloid beta or MAPT/Tau to V-ATPase and rescue via the HYAL-CD44 axis in Alzheimer disease. Autophagy 2023; 19:2318-2337. [PMID: 36843263 PMCID: PMC10351450 DOI: 10.1080/15548627.2023.2181614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 02/10/2023] [Accepted: 02/13/2023] [Indexed: 02/28/2023] Open
Abstract
Impaired activities and abnormally enlarged structures of endolysosomes are frequently observed in Alzheimer disease (AD) brains. However, little is known about whether and how endolysosomal dysregulation is triggered and associated with AD. Here, we show that vacuolar ATPase (V-ATPase) is a hub that mediates proteopathy of oligomeric amyloid beta (Aβ) and hyperphosphorylated MAPT/Tau (p-MAPT/Tau). Endolysosomal integrity was largely destroyed in Aβ-overloaded or p-MAPT/Tau-positive neurons in culture and AD brains, which was a necessary step for triggering neurotoxicity, and treatments with acidic nanoparticles or endocytosis inhibitors rescued the endolysosomal impairment and neurotoxicity. Interestingly, we found that the lumenal ATP6V0C and cytosolic ATP6V1B2 subunits of the V-ATPase complex bound to the internalized Aβ and cytosolic PHF-1-reactive MAPT/Tau, respectively. Their interactions disrupted V-ATPase activity and accompanying endolysosomal activity in vitro and induced neurodegeneration. Using a genome-wide functional screen, we isolated a suppressor, HYAL (hyaluronidase), which reversed the endolysosomal dysfunction and proteopathy and alleviated the memory impairment in 3xTg-AD mice. Further, we found that its metabolite hyaluronic acid (HA) and HA receptor CD44 attenuated neurotoxicity in affected neurons via V-ATPase. We propose that endolysosomal V-ATPase is a bona fide proteotoxic receptor that binds to pathogenic proteins and deteriorates endolysosomal function in AD, leading to neurodegeneration in proteopathy.Abbreviations: AAV, adeno-associated virus; Aβ, amyloid beta; AD, Alzheimer disease; APP, amyloid beta precursor protein; ATP6V0C, ATPase H+ transporting V0 subunit c; ATP6V1A, ATPase H+ transporting V1 subunit A; ATP6V1B2, ATPase H+ transporting V1 subunit B2; CD44.Fc, CD44-mouse immunoglobulin Fc fusion construct; Co-IP, co-immunoprecipitation; CTSD, cathepsin D; HA, hyaluronic acid; HMWHA, high-molecular-weight hyaluronic acid; HYAL, hyaluronidase; i.c.v, intracerebroventricular; LMWHA, low-molecular-weight hyaluronic acid; NPs, nanoparticles; p-MAPT/Tau, hyperphosphorylated microtubule associated protein tau; PI3K, phosphoinositide 3-kinase; V-ATPase, vacuolar-type H+-translocating ATPase; WT, wild-type.
Collapse
Affiliation(s)
- Seo-Hyun Kim
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Young-Sin Cho
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Youbin Kim
- Interdisciplinary Program in Neuroscience, Seoul National University, Seoul, Korea
| | - Jisu Park
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Seung-Min Yoo
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Jimin Gwak
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Youngwon Kim
- School of Biological Sciences, Seoul National University, Seoul, Korea
| | - Youngdae Gwon
- School of Medicine, Sungkyunkwan University, Suwon, Korea
| | - Tae-in Kam
- Department of Neurology and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Yong-Keun Jung
- School of Biological Sciences, Seoul National University, Seoul, Korea
- Interdisciplinary Program in Neuroscience, Seoul National University, Seoul, Korea
| |
Collapse
|
9
|
Advances in Molecular Psychiatry - March 2023: mitochondrial function, stress, neuroinflammation - bipolar disorder, psychosis, and Alzheimer's disease. Mol Psychiatry 2023; 28:968-971. [PMID: 36899214 DOI: 10.1038/s41380-023-01968-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 01/17/2023] [Indexed: 03/12/2023]
|
10
|
Zhu XL, Zhang HW, Peng WJ, Gao S, Yang ZL, Zhang JQ, Liu XS. Autophagy impairment is involved in midazolam-induced lipid droplet accumulation and consequent phagocytosis decrease in BV2 cells. Biochem Biophys Res Commun 2023; 643:147-156. [PMID: 36609155 DOI: 10.1016/j.bbrc.2022.12.067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 12/22/2022] [Indexed: 01/01/2023]
Abstract
An increasing number of experimental and clinical observation suggest that the use of anaesthetics is closely associated with postoperative central nervous system (CNS) complications, such as delirium and cognitive dysfunction. Brain energy rescue is an emerging therapeutic strategy for central nervous system disease (CNSDs). However, the effect of anaesthetics on nerve cell energy utilisation, especially microglia, and its potential effects on cell function still unclear. Elucidating the effects of anaesthetics on lipid droplets, which are specific lipid storage organs, and phagocytosis of microglia is crucial to discover a new therapeutic concept for postoperative CNS complications. Here, we studied the effects of the commonly used anaesthetic midazolam on lipid droplets and phagocytosis in immortalised microglial BV2 cells. Lipid droplets were assessed by flow cytometry and triglyceride quantification. The phagocytosis of BV2 cells was evaluated by detecting their phagocytosis by latex beads. Additionally, the autophagy of BV2 cells was evaluated by western blot and observation under microscopy. Our results showed that midazolam caused lipid droplet accumulation and reduced phagocytosis in BV2 cells, and inhibition of lipid droplet accumulation partially restored phagocytosis. Furthermore, midazolam blocks autophagic degradation by increasing phosphorylated TFEB in BV2 cells, inhibition of midazolam-increased phosphorylated TFEB might contribute to the improvement of autophagic flux by rapamycin. Moreover, promoting autophagy reverse the lipid droplet accumulation and phagocytosis decrease. This study suggests autophagy is a target for attenuating lipid droplet accumulation, normal degradation of lipid droplets is important for maintaining microglia phagocytosis and attenuating the side effects of midazolam on the CNS.
Collapse
Affiliation(s)
- Xiao-Ling Zhu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, China
| | - Hui-Wen Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, China
| | - Wen-Jing Peng
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, China
| | - Shan Gao
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Zhi-Lai Yang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, China
| | - Ji-Qian Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, China.
| | - Xue-Sheng Liu
- Department of Anesthesiology, The First Affiliated Hospital of Anhui Medical University, Key Laboratory of Anesthesiology and Perioperative Medicine of Anhui Higher Education Institutes, Anhui Medical University, China.
| |
Collapse
|
11
|
Mansour HM, El-Khatib AS. Repositioning of receptor tyrosine kinase inhibitors. RECEPTOR TYROSINE KINASES IN NEURODEGENERATIVE AND PSYCHIATRIC DISORDERS 2023:353-401. [DOI: 10.1016/b978-0-443-18677-6.00010-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
12
|
Xiang J, Meng X, Zhao Y, Wu FX, Li M. HyMM: hybrid method for disease-gene prediction by integrating multiscale module structure. Brief Bioinform 2022; 23:6547263. [PMID: 35275996 DOI: 10.1093/bib/bbac072] [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: 10/20/2021] [Revised: 01/18/2022] [Accepted: 02/13/2022] [Indexed: 11/12/2022] Open
Abstract
MOTIVATION Identifying disease-related genes is an important issue in computational biology. Module structure widely exists in biomolecule networks, and complex diseases are usually thought to be caused by perturbations of local neighborhoods in the networks, which can provide useful insights for the study of disease-related genes. However, the mining and effective utilization of the module structure is still challenging in such issues as a disease gene prediction. RESULTS We propose a hybrid disease-gene prediction method integrating multiscale module structure (HyMM), which can utilize multiscale information from local to global structure to more effectively predict disease-related genes. HyMM extracts module partitions from local to global scales by multiscale modularity optimization with exponential sampling, and estimates the disease relatedness of genes in partitions by the abundance of disease-related genes within modules. Then, a probabilistic model for integration of gene rankings is designed in order to integrate multiple predictions derived from multiscale module partitions and network propagation, and a parameter estimation strategy based on functional information is proposed to further enhance HyMM's predictive power. By a series of experiments, we reveal the importance of module partitions at different scales, and verify the stable and good performance of HyMM compared with eight other state-of-the-arts and its further performance improvement derived from the parameter estimation. CONCLUSIONS The results confirm that HyMM is an effective framework for integrating multiscale module structure to enhance the ability to predict disease-related genes, which may provide useful insights for the study of the multiscale module structure and its application in such issues as a disease-gene prediction.
Collapse
Affiliation(s)
- Ju Xiang
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha 410083, China; Department of Basic Medical Sciences & Academician Workstation, Changsha Medical University, Changsha, Hunan 410219, China
| | - Xiangmao Meng
- School of Computer Science and Engineering, Central South University, Changsha 410083, China
| | - Yichao Zhao
- School of Computer Science and Engineering, Central South University, Changsha, Hunan 410083, China
| | - Fang-Xiang Wu
- Division of Biomedical Engineering and Department of Mechanical Engineering, University of Saskatchewan, Saskatoon, SK, S7N 5A9, Canada
| | - Min Li
- Hunan Provincial Key Lab on Bioinformatics, School of Computer Science and Engineering, Central South University, Changsha 410083, China
| |
Collapse
|
13
|
Licinio J, Wong ML. Molecular Psychiatry special issue: advances in Alzheimer's disease. Mol Psychiatry 2021; 26:5467-5470. [PMID: 35027660 DOI: 10.1038/s41380-021-01434-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 12/23/2021] [Indexed: 11/10/2022]
Affiliation(s)
- Julio Licinio
- State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA.
| | - Ma-Li Wong
- State University of New York, Upstate Medical University, Syracuse, NY, 13210, USA
| |
Collapse
|
14
|
Annadurai N, De Sanctis JB, Hajdúch M, Das V. Tau secretion and propagation: Perspectives for potential preventive interventions in Alzheimer's disease and other tauopathies. Exp Neurol 2021; 343:113756. [PMID: 33989658 DOI: 10.1016/j.expneurol.2021.113756] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/26/2021] [Accepted: 05/10/2021] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is characterised by the accumulation of intracytoplasmic aggregates of tau protein, which are suggested to spread in a prion-like manner between interconnected brain regions. This spreading is mediated by the secretion and uptake of tau from the extracellular space or direct cell-to-cell transmission through cellular protrusions. The prion-like tau then converts the endogenous, normal tau into pathological forms, resulting in neurodegeneration. The endoplasmic reticulum/Golgi-independent tau secretion through unconventional secretory pathways involves delivering misfolded and aggregated tau to the plasma membrane and its release into the extracellular space by non-vesicular and vesicular mechanisms. Although cytoplasmic tau was thought to be released only from degenerating cells, studies now show that cells constitutively secrete tau at low levels under physiological conditions. The mechanisms of secretion of tau under physiological and pathological conditions remain unclear. Therefore, a better understanding of these pathways is essential for developing therapeutic approaches that can target prion-like tau forms to prevent neurodegeneration progression in AD. This review focuses on unconventional secretion pathways involved in the spread of tau pathology in AD and presents these pathways as prospective areas for future AD drug discovery and development.
Collapse
Affiliation(s)
- Narendran Annadurai
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 5, 77900 Olomouc, Czech Republic
| | - Juan B De Sanctis
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 5, 77900 Olomouc, Czech Republic
| | - Marián Hajdúch
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 5, 77900 Olomouc, Czech Republic
| | - Viswanath Das
- Institute of Molecular and Translational Medicine, Faculty of Medicine and Dentistry, Palacký University, Hněvotínská 5, 77900 Olomouc, Czech Republic.
| |
Collapse
|