1
|
Ma F, Longo M, Meroni M, Bhattacharya D, Paolini E, Mughal S, Hussain S, Anand SK, Gupta N, Zhu Y, Navarro-Corcuera A, Li K, Prakash S, Cogliati B, Wang S, Huang X, Wang X, Yurdagul A, Rom O, Wang L, Fried SK, Dongiovanni P, Friedman SL, Cai B. EHBP1 suppresses liver fibrosis in metabolic dysfunction-associated steatohepatitis. Cell Metab 2025; 37:1152-1170.e7. [PMID: 40015280 PMCID: PMC12058419 DOI: 10.1016/j.cmet.2025.01.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 11/27/2024] [Accepted: 01/22/2025] [Indexed: 03/01/2025]
Abstract
Excess cholesterol accumulation contributes to fibrogenesis in metabolic dysfunction-associated steatohepatitis (MASH), but how hepatic cholesterol metabolism becomes dysregulated in MASH is not completely understood. We show that human fibrotic MASH livers have decreased EH-domain-binding protein 1 (EHBP1), a genome-wide association study (GWAS) locus associated with low-density lipoprotein (LDL) cholesterol, and that EHBP1 loss- and gain-of-function increase and decrease MASH fibrosis in mice, respectively. Mechanistic studies reveal that EHBP1 promotes sortilin-mediated PCSK9 secretion, leading to LDL receptor (LDLR) degradation, decreased LDL uptake, and reduced TAZ, a fibrogenic effector. At a cellular level, EHBP1 deficiency affects the intracellular localization of retromer, a protein complex required for sortilin stabilization. Our therapeutic approach to stabilizing retromer is effective in mitigating MASH fibrosis. Moreover, we show that the tumor necrosis factor alpha (TNF-α)/peroxisome proliferator-activated receptor alpha (PPARα) pathway suppresses EHBP1 in MASH. These data not only provide mechanistic insights into the role of EHBP1 in cholesterol metabolism and MASH fibrosis but also elucidate an interplay between inflammation and EHBP1-mediated cholesterol metabolism.
Collapse
Affiliation(s)
- Fanglin Ma
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Miriam Longo
- Medicine and Metabolic Diseases, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Marica Meroni
- Medicine and Metabolic Diseases, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Dipankar Bhattacharya
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Erika Paolini
- Medicine and Metabolic Diseases, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Shama Mughal
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Syed Hussain
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Sumit Kumar Anand
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA 71103, USA
| | - Neha Gupta
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Yiwei Zhu
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Amaia Navarro-Corcuera
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Kenneth Li
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Satya Prakash
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bruno Cogliati
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Shuang Wang
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Xin Huang
- Columbia Center for Human Development, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Xiaobo Wang
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Arif Yurdagul
- Department of Molecular and Cellular Physiology, Louisiana State University Health Shreveport, Shreveport, LA 71103, USA
| | - Oren Rom
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Shreveport, Shreveport, LA 71103, USA
| | - Liheng Wang
- Institute of Cardiovascular Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Susan K Fried
- Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Paola Dongiovanni
- Medicine and Metabolic Diseases, Fondazione Ca' Granda IRCCS Ospedale Maggiore Policlinico, Milano 20122, Italy
| | - Scott L Friedman
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bishuang Cai
- Division of Liver Diseases, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
2
|
Trachsel-Moncho L, Veroni C, Mathai BJ, Lapao A, Singh S, Asp NT, Schultz SW, Pankiv S, Simonsen A. SNX10 functions as a modulator of piecemeal mitophagy and mitochondrial bioenergetics. J Cell Biol 2025; 224:e202404009. [PMID: 40052924 PMCID: PMC11893173 DOI: 10.1083/jcb.202404009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 12/20/2024] [Accepted: 02/04/2025] [Indexed: 03/12/2025] Open
Abstract
We here identify the endosomal protein SNX10 as a negative regulator of piecemeal mitophagy of OXPHOS machinery components. In control conditions, SNX10 localizes to early endocytic compartments in a PtdIns3P-dependent manner and modulates endosomal trafficking but also shows dynamic connections with mitochondria. Upon hypoxia-mimicking conditions, SNX10 localizes to late endosomal structures containing selected mitochondrial proteins, including COX-IV and SAMM50, and the autophagy proteins SQSTM1/p62 and LC3B. The turnover of COX-IV was enhanced in SNX10-depleted cells, with a corresponding reduced mitochondrial respiration and citrate synthase activity. Importantly, zebrafish larvae lacking Snx10 show reduced levels of Cox-IV, as well as elevated ROS levels and ROS-mediated cell death in the brain, demonstrating the in vivo relevance of SNX10-mediated modulation of mitochondrial bioenergetics.
Collapse
Affiliation(s)
- Laura Trachsel-Moncho
- Department of Molecular Medicine, Institute of Basic Medical Sciences Faculty of Medicine, University of Oslo, Oslo, Norway
- Center for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Chiara Veroni
- Department of Molecular Medicine, Institute of Basic Medical Sciences Faculty of Medicine, University of Oslo, Oslo, Norway
- Center for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Benan John Mathai
- Department of Molecular Medicine, Institute of Basic Medical Sciences Faculty of Medicine, University of Oslo, Oslo, Norway
- Center for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Ana Lapao
- Department of Molecular Medicine, Institute of Basic Medical Sciences Faculty of Medicine, University of Oslo, Oslo, Norway
- Center for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Sakshi Singh
- Department of Molecular Medicine, Institute of Basic Medical Sciences Faculty of Medicine, University of Oslo, Oslo, Norway
- Center for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Nagham Theres Asp
- Department of Molecular Medicine, Institute of Basic Medical Sciences Faculty of Medicine, University of Oslo, Oslo, Norway
- Center for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Sebastian W. Schultz
- Center for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Serhiy Pankiv
- Department of Molecular Medicine, Institute of Basic Medical Sciences Faculty of Medicine, University of Oslo, Oslo, Norway
- Center for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Anne Simonsen
- Department of Molecular Medicine, Institute of Basic Medical Sciences Faculty of Medicine, University of Oslo, Oslo, Norway
- Center for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
3
|
Zhang Z, Fu X, Wright N, Wang W, Ye Y, Asbury J, Li Y, Zhu C, Wu R, Wang S, Sun S. PTPσ-mediated PI3P regulation modulates neurodegeneration in C9ORF72-ALS/FTD. Neuron 2025; 113:1190-1205.e9. [PMID: 40073860 PMCID: PMC12005967 DOI: 10.1016/j.neuron.2025.02.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 12/09/2024] [Accepted: 02/06/2025] [Indexed: 03/14/2025]
Abstract
The most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) is the repeat expansion in C9ORF72. Dipeptide repeat (DPR) proteins translated from both sense and antisense repeats, especially arginine-rich DPRs (R-DPRs), contribute to neurodegeneration. Through CRISPR interference (CRISPRi) screening in human-derived neurons, we identified receptor-type tyrosine-protein phosphatase S (PTPσ) as a strong modifier of poly-GR-mediated toxicity. We showed that reducing PTPσ promotes the survival of both poly-GR- and poly-PR-expressing neurons by elevating phosphatidylinositol 3-phosphate (PI3P), accompanied by restored early endosomes and lysosomes. Remarkably, PTPσ knockdown or inhibition substantially rescues the PI3P-endolysosomal defects and improves the survival of C9ORF72-ALS/FTD patient-derived neurons. Furthermore, the PTPσ inhibitor diminishes GR toxicity and rescues pathological and behavioral phenotypes in mice. Overall, these findings emphasize the critical role of PI3P-mediated endolysosomal deficits induced by R-DPRs in disease pathogenesis and reveal the therapeutic potential of targeting PTPσ in C9ORF72-ALS/FTD.
Collapse
Affiliation(s)
- Zhe Zhang
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Xiujuan Fu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Noelle Wright
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Cellular and Molecular Physiology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Weiren Wang
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Biotechology Master Program, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Yingzhi Ye
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Cellular and Molecular Physiology Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Julie Asbury
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Notre Dame of Maryland University, Baltimore, MD 21210, USA
| | - Yini Li
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Chengzhang Zhu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Cellular and Molecular Medicine Graduate Program, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rong Wu
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shaopeng Wang
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Shuying Sun
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Brain Science Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; The Solomon H. Snyder Department of Neuroscience and Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
4
|
Wang Y, Cheng Y, Sun H, Wang Z, Chen N, Shi C, Liu H, Yang J, Xu Y. Genome editing in spinocerebellar ataxia type 3 cells improves Golgi apparatus structure. Sci Rep 2025; 15:12106. [PMID: 40204795 PMCID: PMC11982189 DOI: 10.1038/s41598-025-93369-8] [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: 09/12/2024] [Accepted: 03/06/2025] [Indexed: 04/11/2025] Open
Abstract
Spinocerebellar ataxia type 3 (SCA3) is an autosomal dominant neurodegenerative disease caused by repeat expansion of the CAG trinucleotide within exon 10 of the ATXN3 gene. This mutation results in the production of an abnormal ataxin-3 protein containing an extended polyglutamine tract, referred to as mutant ataxin-3. In this study, we investigated the therapeutic potential of CRISPR/Cas9-mediated genome editing for SCA3. First, we designed a specific single-guide RNA targeting the ATXN3 gene and constructed the corresponding targeting vector. Induced pluripotent stem cells (iPSCs) derived from a SCA3 patient were then electroporated with the CRISPR/Cas9 components. Positive clones were screened and validated by PCR and Sanger sequencing to obtain genome-editing iPSCs (GE-iPSCs). Subsequently, the pluripotency of GE-iPSCs was confirmed, and the effects of genome editing on mutant ataxin-3 protein expression and Golgi apparatus morphology were assessed using Western blotting and immunofluorescence analyses. Our results demonstrated that targeted insertion of polyadenylation signals (PAS) upstream of the abnormal CAG repeats effectively suppressed the production of mutant ataxin-3. This intervention also reduced the formation of neuronal nuclear inclusions in differentiated neurons, restored the structural integrity of the Golgi apparatus (which exhibited a loose and enlarged morphology in SCA3 cells), and increased the expression levels of Golgi structural proteins (GM130 and GORASP2). In conclusion, our findings indicate that the targeted insertion of PAS upstream of the abnormal CAG repeats in the ATXN3 gene represents a promising therapeutic strategy for SCA3 through genome editing.
Collapse
Affiliation(s)
- Yanlin Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, 50 Jianshe Road, Zhengzhou, 450000, Henan, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Disease, Zhengzhou, 450000, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases (Zhengzhou University), Zhengzhou, 450000, Henan, China
| | - Yunan Cheng
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, 50 Jianshe Road, Zhengzhou, 450000, Henan, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Disease, Zhengzhou, 450000, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases (Zhengzhou University), Zhengzhou, 450000, Henan, China
| | - Huifang Sun
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, 50 Jianshe Road, Zhengzhou, 450000, Henan, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Disease, Zhengzhou, 450000, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases (Zhengzhou University), Zhengzhou, 450000, Henan, China
| | - Zhuoya Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, 50 Jianshe Road, Zhengzhou, 450000, Henan, China
| | - Na Chen
- Henan University of Traditional Chinese Medicine, Zhengzhou, 450000, Henan, China
| | - Changhe Shi
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, 50 Jianshe Road, Zhengzhou, 450000, Henan, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Disease, Zhengzhou, 450000, Henan, China
- Henan Key Laboratory of Cerebrovascular Diseases (Zhengzhou University), Zhengzhou, 450000, Henan, China
| | - Han Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, 50 Jianshe Road, Zhengzhou, 450000, Henan, China
| | - Jing Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, 50 Jianshe Road, Zhengzhou, 450000, Henan, China.
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Disease, Zhengzhou, 450000, Henan, China.
- Henan Key Laboratory of Cerebrovascular Diseases (Zhengzhou University), Zhengzhou, 450000, Henan, China.
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou University, 50 Jianshe Road, Zhengzhou, 450000, Henan, China.
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Disease, Zhengzhou, 450000, Henan, China.
- Henan Key Laboratory of Cerebrovascular Diseases (Zhengzhou University), Zhengzhou, 450000, Henan, China.
| |
Collapse
|
5
|
Long Y, Chen X, Chen J, Zhang H, Lin Y, Cheng S, Pu N, Zhou X, Sheng R, Abubakar YS, Zheng H, Yun Y, Lu G, Wang Z, Zheng W. Golgi-associated retrograde protein (GARP) complex recruits retromer to trans-Golgi network for FgKex2 and FgSnc1 recycling, necessary for the development and pathogenicity of Fusarium graminearum. THE NEW PHYTOLOGIST 2025; 246:666-688. [PMID: 39953835 DOI: 10.1111/nph.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Accepted: 01/21/2025] [Indexed: 02/17/2025]
Abstract
In eukaryotes, the retromer complex plays a crucial role in the sorting and retrograde transport of cargo proteins from endosomes to the trans-Golgi network (TGN). Despite its importance, the molecular details of this intracellular transport process remain unclear. Here, we have identified a Golgi-associated retrograde protein (GARP) complex as a mediator of vesicle transport that facilitates the recruitment of the retromer complex to the TGN to exert its functions. The GARP complex is mainly localized in the TGN where it interacts with the retromer complex. This interaction is evolutionarily conserved across species. Furthermore, we identified FgKex2 and FgSnc1 as cargo proteins in the GARP/retromer-mediated recycling pathway. Loss of GARP or retromer results in a complete missorting of FgKex2 and FgSnc1 into the vacuolar degradation pathway, which affects the growth, development, biogenesis of toxisomes and pathogenicity of Fusarium graminearum. In summary, we demonstrate for the first time that GARP promotes the recruitment of retromer from endosomes to the TGN, thereby establishing a GARP/retromer transport pathway that coordinates the recycling of cargo proteins FgKex2 and FgSnc1. This process is essential for maintaining sustained growth and development and significantly contributes to the pathogenicity of F. graminearum.
Collapse
Affiliation(s)
- Yunfei Long
- State Key Laboratory of Agricultural and Forestry Biosecurity, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
- Key Laboratory of Bio-pesticide and Chemistry Biology, Ministry of Education, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350000, China
| | - Xin Chen
- State Key Laboratory of Agricultural and Forestry Biosecurity, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
- Key Laboratory of Bio-pesticide and Chemistry Biology, Ministry of Education, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350000, China
| | - Jia Chen
- State Key Laboratory of Agricultural and Forestry Biosecurity, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
- Key Laboratory of Bio-pesticide and Chemistry Biology, Ministry of Education, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350000, China
| | - Haoran Zhang
- State Key Laboratory of Agricultural and Forestry Biosecurity, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
- Key Laboratory of Bio-pesticide and Chemistry Biology, Ministry of Education, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350000, China
| | - Ying Lin
- State Key Laboratory of Agricultural and Forestry Biosecurity, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
- Key Laboratory of Bio-pesticide and Chemistry Biology, Ministry of Education, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350000, China
| | - Shuyuan Cheng
- Jiangxi Agricultural University, College of Agriculture, Nanchang, Jiangxi, 330000, China
| | - Neng Pu
- Agricultural and Rural Comprehensive Service Center, Shuitang Town, Xinping County, Yunnan, 653400, China
| | - Xuandong Zhou
- State Key Laboratory of Agricultural and Forestry Biosecurity, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
- Key Laboratory of Bio-pesticide and Chemistry Biology, Ministry of Education, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350000, China
| | - Renzhi Sheng
- State Key Laboratory of Agricultural and Forestry Biosecurity, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
- Key Laboratory of Bio-pesticide and Chemistry Biology, Ministry of Education, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350000, China
| | - Yakubu Saddeeq Abubakar
- State Key Laboratory of Agricultural and Forestry Biosecurity, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
- Key Laboratory of Bio-pesticide and Chemistry Biology, Ministry of Education, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350000, China
- Department of Biochemistry, Faculty of Life Sciences, Ahmadu Bello University, Zaria, 810281, Nigeria
| | - Huawei Zheng
- Institute of Oceanography, Minjiang University, Fuzhou, 350108, China
| | - Yingzi Yun
- State Key Laboratory of Agricultural and Forestry Biosecurity, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
- Key Laboratory of Bio-pesticide and Chemistry Biology, Ministry of Education, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350000, China
| | - Guodong Lu
- State Key Laboratory of Agricultural and Forestry Biosecurity, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
- Key Laboratory of Bio-pesticide and Chemistry Biology, Ministry of Education, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350000, China
| | - Zonghua Wang
- State Key Laboratory of Agricultural and Forestry Biosecurity, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
- Institute of Oceanography, Minjiang University, Fuzhou, 350108, China
| | - Wenhui Zheng
- State Key Laboratory of Agricultural and Forestry Biosecurity, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, 350002, China
- Key Laboratory of Bio-pesticide and Chemistry Biology, Ministry of Education, College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou, Fujian, 350000, China
| |
Collapse
|
6
|
Xu P, Yue J. Protocol for monitoring the endosomal trafficking of membrane proteins in mammalian cells. STAR Protoc 2025; 6:103686. [PMID: 40057947 PMCID: PMC11928761 DOI: 10.1016/j.xpro.2025.103686] [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: 10/09/2024] [Revised: 12/12/2024] [Accepted: 02/19/2025] [Indexed: 03/25/2025] Open
Abstract
Here, we present a protocol to study epidermal growth factor (EGF) receptor (EGFR) or transferrin trafficking in mammalian cells. We describe steps for using fluorescent ligands or antibodies, confocal imaging, and quantitative analysis to track their movement. We detail procedures for cell culture preparation, labeling membrane proteins, optimizing imaging, and isolating cell lysates for the biochemical analysis of EGFR degradation after EGF treatment. This protocol is adaptable to various cell types and for assessing genetic or pharmacological impacts on endosomal trafficking. For complete details on the use and execution of this protocol, please refer to Ye et al.,1 Ye et al.,2 and Wang et al.3.
Collapse
Affiliation(s)
- Peng Xu
- Division of Natural and Applied Sciences, Synear Molecular Biology Lab, Global Health Research Center, Jiangsu Provincial University Key (Construction) Laboratory for Smart Diagnosis and Treatment of Lung Cancer, Duke Kunshan University, Kunshan 215316, China.
| | - Jianbo Yue
- Division of Natural and Applied Sciences, Synear Molecular Biology Lab, Global Health Research Center, Jiangsu Provincial University Key (Construction) Laboratory for Smart Diagnosis and Treatment of Lung Cancer, Duke Kunshan University, Kunshan 215316, China.
| |
Collapse
|
7
|
Takeo Y, Crite M, Mehmood K, DiMaio D. γ-secretase facilitates retromer-mediated retrograde transport. J Cell Sci 2025; 138:JCS263538. [PMID: 39865938 PMCID: PMC11883284 DOI: 10.1242/jcs.263538] [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: 09/12/2024] [Accepted: 01/14/2025] [Indexed: 01/28/2025] Open
Abstract
Retromer mediates retrograde transport of protein cargoes from endosomes to the trans-Golgi network (TGN). γ-secretase is a protease that cleaves the transmembrane domain of its target proteins. Although retromer can form a stable complex with γ-secretase, the functional consequences of this interaction are not known. Here, we report that retromer-mediated retrograde protein trafficking in cultured human epithelial cells is impaired by the γ-secretase inhibitor XXI or by knockout of PS1 (also known as PSEN1), the catalytic subunit of γ-secretase. These treatments inhibited endosome-to-TGN trafficking of retromer-dependent retrograde cellular cargoes, divalent metal transporter 1 isoform II, cation-independent mannose-6-phosphate receptor and shiga toxin, whereas trafficking of retromer-independent cargoes, cholera toxin and a mutant CIMPR unable to bind retromer was not affected. Moreover, we found that γ-secretase associates with retromer cargoes even in the absence of retromer. XXI treatment and PS1 knockout did not inhibit the ability of retromer or γ-secretase to associate with cargo and did not affect the expression of retromer subunits or Rab7-GTP, which regulates retromer-cargo interaction. These results imply that the γ-secretase-retromer interaction facilitates retromer-mediated retrograde trafficking of cellular transmembrane proteins.
Collapse
Affiliation(s)
- Yuka Takeo
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Mac Crite
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Kashif Mehmood
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
| | - Daniel DiMaio
- Department of Genetics, Yale School of Medicine, New Haven, CT 06510, USA
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA
- Department of Therapeutic Radiology, Yale School of Medicine, New Haven, CT 06510, USA
- Yale Cancer Center, Yale School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
8
|
Naderi Yeganeh P, Kwak SS, Jorfi M, Koler K, Kalatturu T, von Maydell D, Liu Z, Guo K, Choi Y, Park J, Abarca N, Bakiasi G, Cetinbas M, Sadreyev R, Griciuc A, Quinti L, Choi SH, Xia W, Tanzi RE, Hide W, Kim DY. Integrative pathway analysis across humans and 3D cellular models identifies the p38 MAPK-MK2 axis as a therapeutic target for Alzheimer's disease. Neuron 2025; 113:205-224.e8. [PMID: 39610246 PMCID: PMC11757051 DOI: 10.1016/j.neuron.2024.10.029] [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: 10/17/2023] [Revised: 08/29/2024] [Accepted: 10/31/2024] [Indexed: 11/30/2024]
Abstract
Alzheimer's disease (AD) presents a complex pathological landscape, posing challenges to current therapeutic strategies that primarily target amyloid-β (Aβ). Using a novel integrative pathway activity analysis (IPAA), we identified 83 dysregulated pathways common between both post-mortem AD brains and three-dimensional AD cellular models showing robust Aβ42 accumulation. p38 mitogen-activated protein kinase (MAPK) was the most upregulated common pathway. Active p38 MAPK levels increased in the cellular models, human brains, and 5XFAD mice and selectively localized to presynaptic dystrophic neurites. Unbiased phosphoproteomics confirmed increased phosphorylation of p38 MAPK substrates. Downstream activation of MAPK-activated protein kinase 2 (MK2) plays a crucial role in Aβ42-p38 MAPK-mediated tau pathology. Therapeutic targeting of the p38 MAPK-MK2 axis with selective inhibitors significantly reduced Aβ42-driven tau pathology and neuronal loss. IPAA prioritizes the best models to derisk target-drug discovery by integrating human tissue gene expression with functional readouts from cellular models, enabling the identification and validation of high-confidence AD therapeutic targets.
Collapse
Affiliation(s)
- Pourya Naderi Yeganeh
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Sang Su Kwak
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Mehdi Jorfi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Katjuša Koler
- Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, Sheffield, UK
| | - Thejesh Kalatturu
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Djuna von Maydell
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Zhiqing Liu
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA
| | | | - Younjung Choi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | - Joseph Park
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Nelson Abarca
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Grisilda Bakiasi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Murat Cetinbas
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Ruslan Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Ana Griciuc
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Luisa Quinti
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Se Hoon Choi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Weiming Xia
- Department of Pharmacology, Physiology and Biophysics, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, USA; Geriatric Research Education and Clinical Center, Bedford VA Healthcare System, Bedford, MA, USA; Department of Biological Sciences, University of Massachusetts Kennedy College of Science, Lowell, MA, USA
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA.
| | - Winston Hide
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Sheffield Institute for Translational Neuroscience, Department of Neuroscience, University of Sheffield, Sheffield, UK.
| | - Doo Yeon Kim
- Genetics and Aging Research Unit, MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA; McCance Center for Brain Health, Massachusetts General Hospital, Boston, MA 02114, USA.
| |
Collapse
|
9
|
Tacke C, Landgraf P, Dieterich DC, Kröger A. The fate of neuronal synapse homeostasis in aging, infection, and inflammation. Am J Physiol Cell Physiol 2024; 327:C1546-C1563. [PMID: 39495249 DOI: 10.1152/ajpcell.00466.2024] [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: 07/12/2024] [Revised: 10/18/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024]
Abstract
Neuroplasticity is the brain's ability to reorganize and modify its neuronal connections in response to environmental stimuli, experiences, learning, and disease processes. This encompasses a variety of mechanisms, including changes in synaptic strength and connectivity, the formation of new synapses, alterations in neuronal structure and function, and the generation of new neurons. Proper functioning of synapses, which facilitate neuron-to-neuron communication, is crucial for brain activity. Neuronal synapse homeostasis, which involves regulating and maintaining synaptic strength and function in the central nervous system (CNS), is vital for this process. Disruptions in synaptic balance, due to factors like inflammation, aging, or infection, can lead to impaired brain function. This review highlights the main aspects and mechanisms underlying synaptic homeostasis, particularly in the context of aging, infection, and inflammation.
Collapse
Affiliation(s)
- Charlotte Tacke
- Institute of Medical Microbiology and Hospital Hygiene, Molecular Microbiology Group, Otto-von-Guericke University, Magdeburg, Germany
| | - Peter Landgraf
- Institute of Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany
| | - Daniela C Dieterich
- Institute of Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| | - Andrea Kröger
- Institute of Medical Microbiology and Hospital Hygiene, Molecular Microbiology Group, Otto-von-Guericke University, Magdeburg, Germany
- Helmholtz Center for Infection Research, Innate Immunity and Infection Group, Braunschweig, Germany
- Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University, Magdeburg, Germany
| |
Collapse
|
10
|
Ajoolabady A, Pratico D, Tang D, Zhou S, Franceschi C, Ren J. Immunosenescence and inflammaging: Mechanisms and role in diseases. Ageing Res Rev 2024; 101:102540. [PMID: 39395575 DOI: 10.1016/j.arr.2024.102540] [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: 04/28/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/14/2024]
Abstract
Age-related changes initiate a cascade of cellular and molecular alterations that lead to immune system dysfunction or abnormal activation, predisposing individuals to age-related diseases. This phenomenon, commonly referred to as immunosenescence, highlighting aging-associated progressive decline of the immune system. Moreover, mounting evidence suggests that immunosenescence contributes to a related pathological phenomenon known as inflammaging. Inflammaging refers to chronic, low-grade, and systemic inflammation associated with aging, occurring despite the absence of overt stimuli. In the body, inflammation is typically activated in response to overt stimuli such as bacterial/microbial invasion or a pathological state, however, inflammaging occurrence and its underpinning mechanisms seem to be independent and in the absence of such stimuli. Despite recent advancements in molecular characterization and the scrutiny of disease relevance, these two interconnected concepts have remained largely unexplored and unrecognized. In this comprehensive review, we aim to shed light on the mechanistic and cellular aspects of immunosenescence and inflammaging, as well as their pivotal roles in the pathogenesis of aging-related diseases, including cancer, infections, dementia, and neurodegenerative disorders.
Collapse
Affiliation(s)
- Amir Ajoolabady
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL 35294, USA
| | - Domenico Pratico
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shuqin Zhou
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine Tongji University, Shanghai 200072, China
| | - Claudio Franceschi
- IRCCS Institute of Neurological Sciences of Bologna, Bologna, Italy; Department of Applied Mathematics and Laboratory of Systems Biology of Aging, Lobachevsky University, Nizhny Novgorod, Russia.
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China.
| |
Collapse
|
11
|
Cheng S, Long Y, Zhang X, Liu B, Song S, Li G, Hu Y, Du L, Wang Q, Jiang J, Xiong G. The Sorting and Transport of the Cargo Protein CcSnc1 by the Retromer Complex Regulate the Growth, Development, and Pathogenicity of Corynespora cassiicola. J Fungi (Basel) 2024; 10:714. [PMID: 39452666 PMCID: PMC11508248 DOI: 10.3390/jof10100714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/27/2024] [Accepted: 10/12/2024] [Indexed: 10/26/2024] Open
Abstract
In eukaryotes, the retromer complex is critical for the transport of cargo proteins from endosomes to the trans-Golgi network (TGN). Despite its importance, there is a lack of research on the retromer-mediated transport of cargo proteins regulating the growth, development, and pathogenicity of filamentous fungi. In the present study, transcriptome analysis showed that the expression levels of the retromer complex (CcVPS35, CcVPS29 and CcVPS26) were significantly elevated during the early stages of Corynespora cassiicola invasion. Gene knockout and complementation analyses further highlighted the critical role of the retromer complex in C. cassiicola infection. Subcellular localization analysis showed that the retromer complex was mainly localized to the vacuolar membrane and partially to endosomes and the TGN. Further research found that the retromer core subunit CcVps35 can interact with the cargo protein CcSnc1. Subcellular localization showed that CcSnc1 is mainly located at the hyphal tip and partially in endosomes and the Golgi apparatus. Deletion of CcVPS35 resulted in the missorting of CcSnc1 into the vacuolar degradation pathway, indicating that the retromer can sort CcSnc1 from endosomes and transport it to the TGN. Additionally, gene knockout and complementation analyses demonstrated that CcSnc1 is critical for the growth, development, and pathogenicity of C. cassiicola. In summary, the vesicular transport pathway involving the retromer complex regulates the sorting and transport of the cargo protein CcSnc1, which is important for the growth, development and pathogenicity of C. cassiicola.
Collapse
Affiliation(s)
- Shuyuan Cheng
- College of Agronomy, Jiangxi Agricultural University, Nanchang 330045, China; (S.C.); (B.L.); (S.S.); (G.L.); (Y.H.); (L.D.); (Q.W.)
- Jiangxi Province Key Laboratory of Vegetable Cultivation and Utilization, Jiangxi Agricultural University, Nanchang 330045, China
| | - Yunfei Long
- College of Plant Protection, Fujian Agriculture and Forestry University, Fuzhou 350002, China;
| | - Xiaoyang Zhang
- Jiujiang Agricultural Technology Extension Center, Jiujiang 332000, China;
| | - Bing Liu
- College of Agronomy, Jiangxi Agricultural University, Nanchang 330045, China; (S.C.); (B.L.); (S.S.); (G.L.); (Y.H.); (L.D.); (Q.W.)
- Jiangxi Province Key Laboratory of Vegetable Cultivation and Utilization, Jiangxi Agricultural University, Nanchang 330045, China
| | - Shuilin Song
- College of Agronomy, Jiangxi Agricultural University, Nanchang 330045, China; (S.C.); (B.L.); (S.S.); (G.L.); (Y.H.); (L.D.); (Q.W.)
| | - Genghua Li
- College of Agronomy, Jiangxi Agricultural University, Nanchang 330045, China; (S.C.); (B.L.); (S.S.); (G.L.); (Y.H.); (L.D.); (Q.W.)
| | - Yuzhuan Hu
- College of Agronomy, Jiangxi Agricultural University, Nanchang 330045, China; (S.C.); (B.L.); (S.S.); (G.L.); (Y.H.); (L.D.); (Q.W.)
| | - Lei Du
- College of Agronomy, Jiangxi Agricultural University, Nanchang 330045, China; (S.C.); (B.L.); (S.S.); (G.L.); (Y.H.); (L.D.); (Q.W.)
| | - Quanxing Wang
- College of Agronomy, Jiangxi Agricultural University, Nanchang 330045, China; (S.C.); (B.L.); (S.S.); (G.L.); (Y.H.); (L.D.); (Q.W.)
| | - Junxi Jiang
- College of Agronomy, Jiangxi Agricultural University, Nanchang 330045, China; (S.C.); (B.L.); (S.S.); (G.L.); (Y.H.); (L.D.); (Q.W.)
- Jiangxi Province Key Laboratory of Vegetable Cultivation and Utilization, Jiangxi Agricultural University, Nanchang 330045, China
| | - Guihong Xiong
- College of Agronomy, Jiangxi Agricultural University, Nanchang 330045, China; (S.C.); (B.L.); (S.S.); (G.L.); (Y.H.); (L.D.); (Q.W.)
- Jiangxi Province Key Laboratory of Vegetable Cultivation and Utilization, Jiangxi Agricultural University, Nanchang 330045, China
| |
Collapse
|
12
|
Zhu Q, Zhang R, Gu X, Zhao Z, Gao Q, Chen M, Wu Q, Xie T, Sui X. Honokiol enhances the sensitivity of cetuximab in KRAS G13D mutant colorectal cancer through destroying SNX3-retromer complex. Theranostics 2024; 14:5443-5460. [PMID: 39310106 PMCID: PMC11413778 DOI: 10.7150/thno.97180] [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: 04/10/2024] [Accepted: 08/15/2024] [Indexed: 09/25/2024] Open
Abstract
Rationale : the proto-oncogene KRAS is frequently mutated in colorectal cancer (CRC), leading to inherent resistance against monoclonal antibodies targeting the epidermal growth factor receptor (EGFR), such as cetuximab. Therefore, addressing the primary resistance and expanding the indications for target therapy have become critical challenges. Methods : the screening of a natural product library against KRAS mutant CRC cells was conducted, leading to the discovery of a small molecule compound that sensitive to the KRASG13D mutation site. The anti-tumor activity of this small molecule compound in combination with cetuximab was evaluated using the KRASG13D mutant CRC models both in vivo and in vitro. This evaluation includes an examination of its effects on cell proliferation, viability, apoptosis, cell cycle progression, and tumor growth. Furthermore, RNA sequencing, western blot analysis, immunofluorescence, real-time quantitative PCR, and pull-down assays were employed to explore the molecular mechanisms underlying the synergistic anti-tumor effect of this small molecule compound in combination with cetuximab. Results : our study screened 882 compounds in KRAS mutant CRC cells and identified honokiol, a small molecule compound that exhibits specific sensitivity to KRASG13D mutant CRC cells. Furthermore, we revealed that the synergistic augmentation of cetuximab's sensitivity in vivo and in vitro models of KRASG13D mutant CRC in combination with honokiol. Mechanistically, honokiol suppresses SNX3-retromer mediated trafficking, thereby impeding lysosomal proteolytic capacity and inhibiting autophagy and macropinocytosis fluxes. Moreover, honokiol inhibits the conversion of RAS GDP to RAS GTP, heightening the susceptibility of KRASG13D CRC mutant cells to cetuximab. Conclusions : honokiol enhances the sensitivity of cetuximab by destroying SNX3 retromer in KRASG13D mutant CRC preclinical model. These findings present a promising strategy for expanding the indications of target therapy in KRAS mutant colorectal cancer patients.
Collapse
Affiliation(s)
- Qianru Zhu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, P.R. China
| | - Ruonan Zhang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Xiaoqing Gu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
| | - Ziming Zhao
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, P.R. China
| | - Quan Gao
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, P.R. China
| | - Min Chen
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, P.R. China
| | - Qibiao Wu
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, P.R. China
| | - Tian Xie
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, P.R. China
| | - Xinbing Sui
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, Zhejiang, China
- State Key Laboratory of Quality Research in Chinese Medicines, Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, P.R. China
- Department of Medical Oncology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| |
Collapse
|
13
|
Takeo Y, Crite M, DiMaio D. γ-secretase facilitates retromer-mediated retrograde transport. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.07.597932. [PMID: 38895404 PMCID: PMC11185792 DOI: 10.1101/2024.06.07.597932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
The retromer complex mediates retrograde transport of protein cargos from endosomes to the trans-Golgi network (TGN). γ-secretase is a multisubunit protease that cleaves the transmembrane domain of its target proteins. Mutations in genes encoding subunits of retromer or γ-secretase can cause familial Alzheimer disease (AD) and other degenerative neurological diseases. It has been reported that retromer interacts with γ-secretase, but the consequences of this interaction are not known. Here, we report that retromer-mediated retrograde protein trafficking in cultured human epithelial cells is impaired by inhibition of γ-secretase activity or by genetic elimination of γ-secretase. γ-secretase inhibitor XXI and knockout of PS1, the catalytic subunit of γ-secretase, inhibit endosome to TGN trafficking of retromer-dependent retrograde cargos, divalent metal transporter 1 isoform II (DMT1-II), cation-independent mannose-6-phosphate receptor (CIMPR), and shiga toxin. Trafficking of retromer-independent cargos, such as cholera toxin and a CIMPR mutant that does not bind to retromer was not affected by γ-secretase inhibition. XXI treatment and PS1 KO inhibit interaction of γ-secretase with retromer but do not inhibit the association of cargo with retromer or with γ-secretase in intact cells. Similarly, these treatments do not affect the level of Rab7-GTP, which regulates retromer-cargo interaction. These results suggest that the γ-secretase-retromer interaction facilitates retromer-mediated retrograde trafficking.
Collapse
Affiliation(s)
- Yuka Takeo
- Department of Genetics, Yale School of Medicine
| | - Mac Crite
- Department of Genetics, Yale School of Medicine
- Current affiliation: American University
| | - Daniel DiMaio
- Department of Genetics, Yale School of Medicine
- Department of Molecular Biophysics and Biochemistry, Yale University
- Department of Therapeutic Radiology, Yale School of Medicine
- Yale Cancer Center, Yale School of Medicine
| |
Collapse
|
14
|
Cooper JM, Lathuiliere A, Su EJ, Song Y, Torrente D, Jo Y, Weinrich N, Sales JD, Migliorini M, Sisson TH, Lawrence DA, Hyman BT, Strickland DK. SORL1 is a receptor for tau that promotes tau seeding. J Biol Chem 2024; 300:107313. [PMID: 38657864 PMCID: PMC11145553 DOI: 10.1016/j.jbc.2024.107313] [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: 01/05/2024] [Revised: 03/28/2024] [Accepted: 04/15/2024] [Indexed: 04/26/2024] Open
Abstract
Sortilin-related receptor 1 (SORL1) is an intracellular sorting receptor genetically implicated in Alzheimer's disease (AD) that impacts amyloid precursor protein trafficking. The objective of these studies was to test the hypothesis that SORL1 binds tau, modulates its cellular trafficking and impacts the aggregation of cytoplasmic tau induced by pathological forms of tau. Using surface plasmon resonance measurements, we observed high-affinity binding of tau to SORL1 and the vacuolar protein sorting 10 domain of SORL1. Interestingly, unlike LDL receptor-related protein 1, SORL1 binds tau at both pH 7.4 and pH 5.5, revealing its ability to bind tau at endosomal pH. Immunofluorescence studies confirmed that exogenously added tau colocalized with SORL1 in H4 neuroglioma cells, while overexpression of SORL1 in LDL receptor-related protein 1-deficient Chinese hamster ovary (CHO) cells resulted in a marked increase in the internalization of tau, indicating that SORL1 can bind and mediate the internalization of monomeric forms of tau. We further demonstrated that SORL1 mediates tau seeding when tau RD P301S FRET biosensor cells expressing SORL1 were incubated with high molecular weight forms of tau isolated from the brains of patients with AD. Seeding in H4 neuroglioma cells is significantly reduced when SORL1 is knocked down with siRNA. Finally, we demonstrate that the N1358S mutant of SORL1 significantly increases tau seeding when compared to WT SORL1, identifying for the first time a potential mechanism that connects this specific SORL1 mutation to Alzheimer's disease. Together, these studies identify SORL1 as a receptor that contributes to trafficking and seeding of pathogenic tau.
Collapse
Affiliation(s)
- Joanna M Cooper
- The Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Aurelien Lathuiliere
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA; Department of Rehabilitation and Geriatrics, Memory Center, Geneva University Hospital and University of Geneva, Geneva, Switzerland
| | - Enming J Su
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Yuyu Song
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Daniel Torrente
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, New York, USA
| | - Youhwa Jo
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA
| | - Nicholas Weinrich
- The Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Jennifer Diaz Sales
- The Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Mary Migliorini
- The Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Thomas H Sisson
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Daniel A Lawrence
- Department of Internal Medicine, University of Michigan School of Medicine, Ann Arbor, Michigan, USA
| | - Bradley T Hyman
- Alzheimer Research Unit, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts, USA.
| | - Dudley K Strickland
- The Center for Vascular and Inflammatory Diseases, University of Maryland School of Medicine, Baltimore, Maryland, USA; Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland, USA; Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
15
|
Peng KY, Liemisa B, Pasato J, D'Acunzo P, Pawlik M, Heguy A, Penikalapati SC, Labuza A, Pidikiti H, Alldred MJ, Ginsberg SD, Levy E, Mathews PM. Apolipoprotein E2 Expression Alters Endosomal Pathways in a Mouse Model With Increased Brain Exosome Levels During Aging. Traffic 2024; 25:e12937. [PMID: 38777335 PMCID: PMC11141728 DOI: 10.1111/tra.12937] [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: 12/13/2022] [Revised: 04/14/2024] [Accepted: 04/15/2024] [Indexed: 05/25/2024]
Abstract
The polymorphic APOE gene is the greatest genetic determinant of sporadic Alzheimer's disease risk: the APOE4 allele increases risk, while the APOE2 allele is neuroprotective compared with the risk-neutral APOE3 allele. The neuronal endosomal system is inherently vulnerable during aging, and APOE4 exacerbates this vulnerability by driving an enlargement of early endosomes and reducing exosome release in the brain of humans and mice. We hypothesized that the protective effects of APOE2 are, in part, mediated through the endosomal pathway. Messenger RNA analyses showed that APOE2 leads to an enrichment of endosomal pathways in the brain when compared with both APOE3 and APOE4. Moreover, we show age-dependent alterations in the recruitment of key endosomal regulatory proteins to vesicle compartments when comparing APOE2 to APOE3. In contrast to the early endosome enlargement previously shown in Alzheimer's disease and APOE4 models, we detected similar morphology and abundance of early endosomes and retromer-associated vesicles within cortical neurons of aged APOE2 targeted-replacement mice compared with APOE3. Additionally, we observed increased brain extracellular levels of endosome-derived exosomes in APOE2 compared with APOE3 mice during aging, consistent with enhanced endosomal cargo clearance by exosomes to the extracellular space. Our findings thus demonstrate that APOE2 enhances an endosomal clearance pathway, which has been shown to be impaired by APOE4 and which may be protective due to APOE2 expression during brain aging.
Collapse
Affiliation(s)
- Katherine Y Peng
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York, USA
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, New York, USA
| | - Braison Liemisa
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, New York, USA
| | - Jonathan Pasato
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, New York, USA
| | - Pasquale D'Acunzo
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York, USA
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, New York, USA
| | - Monika Pawlik
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, New York, USA
| | - Adriana Heguy
- Genome Technology Center, New York University Grossman School of Medicine, New York, New York, USA
- Department of Pathology, New York University Grossman School of Medicine, New York, New York, USA
| | - Sai C Penikalapati
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, New York, USA
| | - Amanda Labuza
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York, USA
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, New York, USA
| | - Harshitha Pidikiti
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, New York, USA
| | - Melissa J Alldred
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York, USA
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, New York, USA
| | - Stephen D Ginsberg
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York, USA
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, New York, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, New York, USA
- Department of Neuroscience & Physiology, New York University Grossman School of Medicine, New York, New York, USA
| | - Efrat Levy
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York, USA
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, New York, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, New York, USA
- Department of Biochemistry & Molecular Pharmacology, New York University Grossman School of Medicine, New York, New York, USA
| | - Paul M Mathews
- Department of Psychiatry, New York University Grossman School of Medicine, New York, New York, USA
- Center for Dementia Research, Nathan S. Kline Institute, Orangeburg, New York, USA
- Neuroscience Institute, New York University Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
16
|
Guise AJ, Misal SA, Carson R, Chu JH, Boekweg H, Van Der Watt D, Welsh NC, Truong T, Liang Y, Xu S, Benedetto G, Gagnon J, Payne SH, Plowey ED, Kelly RT. TDP-43-stratified single-cell proteomics of postmortem human spinal motor neurons reveals protein dynamics in amyotrophic lateral sclerosis. Cell Rep 2024; 43:113636. [PMID: 38183652 PMCID: PMC10926001 DOI: 10.1016/j.celrep.2023.113636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 11/02/2023] [Accepted: 12/14/2023] [Indexed: 01/08/2024] Open
Abstract
A limitation of conventional bulk-tissue proteome studies in amyotrophic lateral sclerosis (ALS) is the confounding of motor neuron (MN) signals by admixed non-MN proteins. Here, we leverage laser capture microdissection and nanoPOTS single-cell mass spectrometry-based proteomics to query changes in protein expression in single MNs from postmortem ALS and control tissues. In a follow-up analysis, we examine the impact of stratification of MNs based on cytoplasmic transactive response DNA-binding protein 43 (TDP-43)+ inclusion pathology on the profiles of 2,238 proteins. We report extensive overlap in differentially abundant proteins identified in ALS MNs with or without overt TDP-43 pathology, suggesting early and sustained dysregulation of cellular respiration, mRNA splicing, translation, and vesicular transport in ALS. Together, these data provide insights into proteome-level changes associated with TDP-43 proteinopathy and begin to demonstrate the utility of pathology-stratified trace sample proteomics for understanding single-cell protein dynamics in human neurologic diseases.
Collapse
Affiliation(s)
| | - Santosh A Misal
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA
| | - Richard Carson
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA
| | | | - Hannah Boekweg
- Biology Department, Brigham Young University, Provo, UT 84602, USA
| | | | | | - Thy Truong
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA
| | - Yiran Liang
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA
| | | | | | | | - Samuel H Payne
- Biology Department, Brigham Young University, Provo, UT 84602, USA
| | | | - Ryan T Kelly
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602, USA.
| |
Collapse
|
17
|
Wilson KA, Bar S, Dammer EB, Carrera EM, Hodge BA, Hilsabeck TAU, Bons J, Brownridge GW, Beck JN, Rose J, Granath-Panelo M, Nelson CS, Qi G, Gerencser AA, Lan J, Afenjar A, Chawla G, Brem RB, Campeau PM, Bellen HJ, Schilling B, Seyfried NT, Ellerby LM, Kapahi P. OXR1 maintains the retromer to delay brain aging under dietary restriction. Nat Commun 2024; 15:467. [PMID: 38212606 PMCID: PMC10784588 DOI: 10.1038/s41467-023-44343-3] [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: 08/02/2023] [Accepted: 12/07/2023] [Indexed: 01/13/2024] Open
Abstract
Dietary restriction (DR) delays aging, but the mechanism remains unclear. We identified polymorphisms in mtd, the fly homolog of OXR1, which influenced lifespan and mtd expression in response to DR. Knockdown in adulthood inhibited DR-mediated lifespan extension in female flies. We found that mtd/OXR1 expression declines with age and it interacts with the retromer, which regulates trafficking of proteins and lipids. Loss of mtd/OXR1 destabilized the retromer, causing improper protein trafficking and endolysosomal defects. Overexpression of retromer genes or pharmacological restabilization with R55 rescued lifespan and neurodegeneration in mtd-deficient flies and endolysosomal defects in fibroblasts from patients with lethal loss-of-function of OXR1 variants. Multi-omic analyses in flies and humans showed that decreased Mtd/OXR1 is associated with aging and neurological diseases. mtd/OXR1 overexpression rescued age-related visual decline and tauopathy in a fly model. Hence, OXR1 plays a conserved role in preserving retromer function and is critical for neuronal health and longevity.
Collapse
Affiliation(s)
- Kenneth A Wilson
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Sudipta Bar
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Eric B Dammer
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | | | - Brian A Hodge
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Tyler A U Hilsabeck
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
| | - Joanna Bons
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | | | - Jennifer N Beck
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Jacob Rose
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | | | | | - Grace Qi
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | | | - Jianfeng Lan
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
- Guanxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, The Afilliated Hospital of Guilin Medican University, Guilin, 541001, Guanxi, China
| | - Alexandra Afenjar
- Assistance Publique des Hôpitaux de Paris, Unité de Génétique Clinique, Hôpital Armand Trousseau, Groupe Hospitalier Universitaire, Paris, 75012, France
- Département de Génétique et Embryologie Médicale, CRMR des Malformations et Maladies Congénitales du Cervelet, GRC ConCer-LD, Sorbonne Universités, Hôpital Trousseau, Paris, 75012, France
| | - Geetanjali Chawla
- RNA Biology Laboratory, Department of Life Sciences, School of Natural Sciences, Shiv Nadar Institute of Eminence, NH91, Tehsil Dadri, G. B. Nagar, 201314, Uttar Pradesh, India
| | - Rachel B Brem
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA
- Department of Plant and Microbial Biology, University of California, Berkeley, 111 Koshland Hall, Berkeley, CA, 94720, USA
| | - Philippe M Campeau
- Centre Hospitalier Universitaire Saint-Justine Research Center, CHU Sainte-Justine, Montreal, QC, H3T 1J4, Canada
| | - Hugo J Bellen
- Departments of Molecular and Human Genetics and Neuroscience, Neurological Research Institute, Texas Children's Hospital, Baylor College of Medicine, Houston, TX, 77030, USA
| | | | - Nicholas T Seyfried
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Lisa M Ellerby
- Buck Institute for Research on Aging, Novato, CA, 94945, USA.
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA.
| | - Pankaj Kapahi
- Buck Institute for Research on Aging, Novato, CA, 94945, USA.
- Leonard Davis School of Gerontology, University of Southern California, Los Angeles, CA, 90089, USA.
| |
Collapse
|
18
|
Li D, Yan J, Li K, Yang Q, Bian L, Lin B, Liu X, Xi Z. Identification of potential glioma drug resistance target proteins based on ultra-performance liquid chromatography-mass spectrometry differential proteomics. PeerJ 2023; 11:e16426. [PMID: 38054015 PMCID: PMC10695112 DOI: 10.7717/peerj.16426] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 10/18/2023] [Indexed: 12/07/2023] Open
Abstract
In this study, to screen for candidate markers of temozolomide (TMZ) resistance in glioblastoma, we artificially established TMZ drug-resistant glioblastoma (GBM) cell lines, U251-TMZ and U87-TMZ. In the U251-TMZ and U87-TMZ cell lines, we screened and analyzed differentially expressed proteins using ultra-performance liquid chromatography-mass spectrometry (UPLC-MS) differential proteomics. Compared with the U251 and U87 control cell lines, 95 differential proteins were screened in the U251-TMZ and U87-TMZ cell lines, of which 28 proteins were upregulated and 67 proteins were down-regulated. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses of the co-upregulated proteins showed that most of the differentially expressed proteins were located in the cytoplasm and were significantly upregulated in the biological processes related to vesicular transport in the intimal system and inflammatory response mediated by myeloid leukocytes. Seven candidates were identified as potential GBM markers of TMZ resistance. Combined with existing research findings, our study supports that UAP1L1 and BCKDK are promising potential markers of TMZ resistance in GBM. This is important for further understanding the molecular mechanisms that drive the development and enhancement of TMZ resistance.
Collapse
Affiliation(s)
- Da Li
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Jun Yan
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Kang Li
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Qingcheng Yang
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Liping Bian
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Bencheng Lin
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Xiaohua Liu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| | - Zhuge Xi
- Tianjin Institute of Environmental and Operational Medicine, Tianjin, China
| |
Collapse
|
19
|
Guise AJ, Misal SA, Carson R, Boekweg H, Watt DVD, Truong T, Liang Y, Chu JH, Welsh NC, Gagnon J, Payne SH, Plowey ED, Kelly RT. TDP-43-stratified single-cell proteomic profiling of postmortem human spinal motor neurons reveals protein dynamics in amyotrophic lateral sclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.08.544233. [PMID: 37333094 PMCID: PMC10274884 DOI: 10.1101/2023.06.08.544233] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Unbiased proteomics has been employed to interrogate central nervous system (CNS) tissues (brain, spinal cord) and fluid matrices (CSF, plasma) from amyotrophic lateral sclerosis (ALS) patients; yet, a limitation of conventional bulk tissue studies is that motor neuron (MN) proteome signals may be confounded by admixed non-MN proteins. Recent advances in trace sample proteomics have enabled quantitative protein abundance datasets from single human MNs (Cong et al., 2020b). In this study, we leveraged laser capture microdissection (LCM) and nanoPOTS (Zhu et al., 2018c) single-cell mass spectrometry (MS)-based proteomics to query changes in protein expression in single MNs from postmortem ALS and control donor spinal cord tissues, leading to the identification of 2515 proteins across MNs samples (>900 per single MN) and quantitative comparison of 1870 proteins between disease groups. Furthermore, we studied the impact of enriching/stratifying MN proteome samples based on the presence and extent of immunoreactive, cytoplasmic TDP-43 inclusions, allowing identification of 3368 proteins across MNs samples and profiling of 2238 proteins across TDP-43 strata. We found extensive overlap in differential protein abundance profiles between MNs with or without obvious TDP-43 cytoplasmic inclusions that together point to early and sustained dysregulation of oxidative phosphorylation, mRNA splicing and translation, and retromer-mediated vesicular transport in ALS. Our data are the first unbiased quantification of single MN protein abundance changes associated with TDP-43 proteinopathy and begin to demonstrate the utility of pathology-stratified trace sample proteomics for understanding single-cell protein abundance changes in human neurologic diseases.
Collapse
Affiliation(s)
| | - Santosh A. Misal
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602
| | - Richard Carson
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602
| | - Hannah Boekweg
- Biology Department, Brigham Young University, Provo, UT 84602, USA
| | | | - Thy Truong
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602
| | - Yiran Liang
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602
| | | | | | | | - Samuel H. Payne
- Biology Department, Brigham Young University, Provo, UT 84602, USA
| | | | - Ryan T. Kelly
- Department of Chemistry and Biochemistry, Brigham Young University, Provo, UT 84602
| |
Collapse
|
20
|
Curtis ME, Smith T, Nenov M, Blass BE, Praticò D. Retromer Stabilization Improves Cognitive Function and Synaptic Plasticity in a Mouse Model of Down Syndrome. J Alzheimers Dis 2023:JAD230205. [PMID: 37334603 PMCID: PMC10357169 DOI: 10.3233/jad-230205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
BACKGROUND Retromer complex proteins are decreased in postmortem brain tissues from Down syndrome subjects and inversely correlate with the Alzheimer's disease-like neuropathology. However, whether targeting in vivo the retromer system affects cognitive deficits and synaptic function in Down syndrome remains unknown. OBJECTIVE The aim of the current study was to examine the effects of pharmacological retromer stabilization on cognitive and synaptic functions in a mouse model of Down syndrome. METHODS Ts65dn mice were administered the pharmacological chaperone, TPT-172, or vehicle from 4 to 9 months of age and then assessed for changes in cognitive function. To assess the effects of TPT-172 on synaptic plasticity, hippocampal slices from Ts65dn mice were incubated in TPT-172 and used for field potential recordings. RESULTS Chronic TPT-172 treatment improved performance in cognitive function tests, its incubation with hippocampal slices ameliorated synaptic function response. CONCLUSION Pharmacological stabilization of the retromer complex improves synaptic plasticity and memory in a mouse model of Down syndrome. These results support the therapeutic potential of pharmacological retromer stabilization for individual with Down syndrome.
Collapse
Affiliation(s)
- Mary Elizabeth Curtis
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Tiffany Smith
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Miroslav Nenov
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | | | - Domenico Praticò
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
21
|
Ma XY, Yang TT, Liu L, Peng XC, Qian F, Tang FR. Ependyma in Neurodegenerative Diseases, Radiation-Induced Brain Injury and as a Therapeutic Target for Neurotrophic Factors. Biomolecules 2023; 13:754. [PMID: 37238624 PMCID: PMC10216700 DOI: 10.3390/biom13050754] [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: 03/01/2023] [Revised: 04/03/2023] [Accepted: 04/24/2023] [Indexed: 05/28/2023] Open
Abstract
The neuron loss caused by the progressive damage to the nervous system is proposed to be the main pathogenesis of neurodegenerative diseases. Ependyma is a layer of ciliated ependymal cells that participates in the formation of the brain-cerebrospinal fluid barrier (BCB). It functions to promotes the circulation of cerebrospinal fluid (CSF) and the material exchange between CSF and brain interstitial fluid. Radiation-induced brain injury (RIBI) shows obvious impairments of the blood-brain barrier (BBB). In the neuroinflammatory processes after acute brain injury, a large amount of complement proteins and infiltrated immune cells are circulated in the CSF to resist brain damage and promote substance exchange through the BCB. However, as the protective barrier lining the brain ventricles, the ependyma is extremely vulnerable to cytotoxic and cytolytic immune responses. When the ependyma is damaged, the integrity of BCB is destroyed, and the CSF flow and material exchange is affected, leading to brain microenvironment imbalance, which plays a vital role in the pathogenesis of neurodegenerative diseases. Epidermal growth factor (EGF) and other neurotrophic factors promote the differentiation and maturation of ependymal cells to maintain the integrity of the ependyma and the activity of ependymal cilia, and may have therapeutic potential in restoring the homeostasis of the brain microenvironment after RIBI or during the pathogenesis of neurodegenerative diseases.
Collapse
Affiliation(s)
- Xin-Yu Ma
- Department of Physiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Ting-Ting Yang
- Department of Physiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Lian Liu
- Department of Pharmacology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Xiao-Chun Peng
- Department of Pathophysiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Feng Qian
- Department of Physiology, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou 434023, China
| | - Feng-Ru Tang
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative, National University of Singapore, Singapore 138602, Singapore
| |
Collapse
|
22
|
Uzhytchak M, Smolková B, Lunova M, Frtús A, Jirsa M, Dejneka A, Lunov O. Lysosomal nanotoxicity: Impact of nanomedicines on lysosomal function. Adv Drug Deliv Rev 2023; 197:114828. [PMID: 37075952 DOI: 10.1016/j.addr.2023.114828] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 03/28/2023] [Accepted: 04/12/2023] [Indexed: 04/21/2023]
Abstract
Although several nanomedicines got clinical approval over the past two decades, the clinical translation rate is relatively small so far. There are many post-surveillance withdrawals of nanomedicines caused by various safety issues. For successful clinical advancement of nanotechnology, it is of unmet need to realize cellular and molecular foundation of nanotoxicity. Current data suggest that lysosomal dysfunction caused by nanoparticles is emerging as the most common intracellular trigger of nanotoxicity. This review analyzes prospect mechanisms of lysosomal dysfunction-mediated toxicity induced by nanoparticles. We summarized and critically assessed adverse drug reactions of current clinically approved nanomedicines. Importantly, we show that physicochemical properties have great impact on nanoparticles interaction with cells, excretion route and kinetics, and subsequently on toxicity. We analyzed literature on adverse reactions of current nanomedicines and hypothesized that adverse reactions might be linked with lysosomal dysfunction caused by nanomedicines. Finally, from our analysis it becomes clear that it is unjustifiable to generalize safety and toxicity of nanoparticles, since different particles possess distinct toxicological properties. We propose that the biological mechanism of the disease progression and treatment should be central in the optimization of nanoparticle design.
Collapse
Affiliation(s)
- Mariia Uzhytchak
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Barbora Smolková
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Mariia Lunova
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic; Institute for Clinical & Experimental Medicine (IKEM), 14021 Prague, Czech Republic
| | - Adam Frtús
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Milan Jirsa
- Institute for Clinical & Experimental Medicine (IKEM), 14021 Prague, Czech Republic
| | - Alexandr Dejneka
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic
| | - Oleg Lunov
- Institute of Physics of the Czech Academy of Sciences, 18221 Prague, Czech Republic.
| |
Collapse
|
23
|
Li JG, Blass BE, Praticò D. Beneficial Effect of a Small Pharmacologic Chaperone on the Established Alzheimer's Disease Phenotype. J Alzheimers Dis 2023; 91:463-469. [PMID: 36442197 DOI: 10.3233/jad-220869] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
BACKGROUND The endosomal retromer complex system is a key controller for trafficking of proteins. Downregulation of its recognition core proteins, such as VPS35, is present in Alzheimer's disease (AD) brain, whereas its normalization prevents the development of AD pathology in a transgenic model with amyloid-β deposits and tau tangles. OBJECTIVE Assess the effect of targeting VPS35 after the AD pathology and memory impairments have developed. METHODS Twelve-month-old triple transgenic mice were treated with a small pharmacological chaperone, TPT-172, or vehicle for 14 weeks. At the end of this period, the effect of the drug on their phenotype was evaluated. RESULTS While control mice had a decline of learning and memory, the group receiving the chaperone did not. Moreover, when compared with controls the treated mice had significantly less amyloid-β peptides and phosphorylated tau, elevation of post-synaptic protein, and reduction in astrocytes activation. CONCLUSION Taken together, our findings demonstrate that pharmacologic stabilization of the retromer recognition core is beneficial also after the AD-like pathologic phenotype is established.
Collapse
Affiliation(s)
- Jian-Guo Li
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Benjamin E Blass
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Philadelphia, PA, USA.,Moulder Center for Drug Discovery Research, School of Pharmacy, Temple University, Philadelphia, PA, USA
| | - Domenico Praticò
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
24
|
Matutino Santos P, Pereira Campos G, Nascimento C. Endo-Lysosomal and Autophagy Pathway and Ubiquitin-Proteasome System in Mood Disorders: A Review Article. Neuropsychiatr Dis Treat 2023; 19:133-151. [PMID: 36684613 PMCID: PMC9849791 DOI: 10.2147/ndt.s376380] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 12/08/2022] [Indexed: 01/15/2023] Open
Abstract
Mood disorders are disabling conditions that cause significant functional impairment. Due to the clinical heterogeneity and complex nature of these disorders, diagnostic and treatment strategies face challenges. The etiology of mood disorders is multifactorial, involving genetic and environmental aspects that are associated with specific biological pathways including inflammation, oxidative stress, and neuroprotection. Alterations in these pathways may reduce the cell's ability to recover from stress conditions occurring during mood episodes. The endo-lysosomal and autophagy pathway (ELAP) and the ubiquitin-proteasome system (UPS) play critical roles in protein homeostasis, impacting neuroplasticity and neurodevelopment. Thus, emerging evidence has suggested a role for these pathways in mental disorders. In the case of neurodegenerative diseases (NDDs), a deeper understanding in the role of ELAP and UPS has been critical to discover new treatment targets. Since it is suggested that NDDs and mood disorders share clinical symptomatology and risk factors, it has been hypothesized that there might be common underlying molecular pathways. Here, we review the importance of the ELAP and UPS for the central nervous system and for mood disorders. Finally, we discuss potential translational strategies for the diagnosis and treatment of major depressive disorder and bipolar disorder associated with these pathways.
Collapse
Affiliation(s)
- Petala Matutino Santos
- Center for Mathematics, Computing and Cognition (CMCC), Federal University of ABC (UFABC), São Paulo, Brazil
| | - Giovanna Pereira Campos
- Center for Mathematics, Computing and Cognition (CMCC), Federal University of ABC (UFABC), São Paulo, Brazil
| | - Camila Nascimento
- Department of Psychiatry, University of São Paulo Medical School, São Paulo, Brazil
| |
Collapse
|
25
|
Salasova A, Monti G, Andersen OM, Nykjaer A. Finding memo: versatile interactions of the VPS10p-Domain receptors in Alzheimer’s disease. Mol Neurodegener 2022; 17:74. [PMID: 36397124 PMCID: PMC9673319 DOI: 10.1186/s13024-022-00576-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 10/17/2022] [Indexed: 11/19/2022] Open
Abstract
The family of VPS10p-Domain (D) receptors comprises five members named SorLA, Sortilin, SorCS1, SorCS2 and SorCS3. While their physiological roles remain incompletely resolved, they have been recognized for their signaling engagements and trafficking abilities, navigating a number of molecules between endosome, Golgi compartments, and the cell surface. Strikingly, recent studies connected all the VPS10p-D receptors to Alzheimer’s disease (AD) development. In addition, they have been also associated with diseases comorbid with AD such as diabetes mellitus and major depressive disorder. This systematic review elaborates on genetic, functional, and mechanistic insights into how dysfunction in VPS10p-D receptors may contribute to AD etiology, AD onset diversity, and AD comorbidities. Starting with their functions in controlling cellular trafficking of amyloid precursor protein and the metabolism of the amyloid beta peptide, we present and exemplify how these receptors, despite being structurally similar, regulate various and distinct cellular events involved in AD. This includes a plethora of signaling crosstalks that impact on neuronal survival, neuronal wiring, neuronal polarity, and synaptic plasticity. Signaling activities of the VPS10p-D receptors are especially linked, but not limited to, the regulation of neuronal fitness and apoptosis via their physical interaction with pro- and mature neurotrophins and their receptors. By compiling the functional versatility of VPS10p-D receptors and their interactions with AD-related pathways, we aim to further propel the AD research towards VPS10p-D receptor family, knowledge that may lead to new diagnostic markers and therapeutic strategies for AD patients.
Collapse
|
26
|
Wu QL, Gao Y, Li JT, Ma WY, Chen NH. The Role of AMPARs Composition and Trafficking in Synaptic Plasticity and Diseases. Cell Mol Neurobiol 2022; 42:2489-2504. [PMID: 34436728 PMCID: PMC11421597 DOI: 10.1007/s10571-021-01141-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 08/11/2021] [Indexed: 11/28/2022]
Abstract
AMPA receptors are tetrameric ionic glutamate receptors, which mediate 90% fast excitatory synaptic transmission induced by excitatory glutamate in the mammalian central nervous system through the activation or inactivation of ion channels. The alternation of synaptic AMPA receptor number and subtype is thought to be one of the primary mechanisms that involve in synaptic plasticity regulation and affect the functions in learning, memory, and cognition. The increasing of surface AMPARs enhances synaptic strength during long-term potentiation, whereas the decreasing of AMPARs weakens synaptic strength during the long-term depression. It is closely related to the AMPA receptor as well as its subunits assembly, trafficking, and degradation. The dysfunction of any step in these precise regulatory processes is likely to induce the disorder of synaptic transmission and loss of neurons, or even cause neuropsychiatric diseases ultimately. Therefore, it is useful to understand how AMPARs regulate synaptic plasticity and its role in related neuropsychiatric diseases via comprehending architecture and trafficking of the receptors. Here, we reviewed the progress in structure, expression, trafficking, and relationship with synaptic plasticity of AMPA receptor, especially in anxiety, depression, neurodegenerative disorders, and cerebral ischemia.
Collapse
Affiliation(s)
- Qing-Lin Wu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Yan Gao
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Jun-Tong Li
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Wen-Yu Ma
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Nai-Hong Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, 510405, China.
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
27
|
Filippone A, Esposito E, Mannino D, Lyssenko N, Praticò D. The contribution of altered neuronal autophagy to neurodegeneration. Pharmacol Ther 2022; 238:108178. [PMID: 35351465 PMCID: PMC9510148 DOI: 10.1016/j.pharmthera.2022.108178] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 12/21/2022]
Abstract
Defects in cellular functions related to altered protein homeostasis and associated progressive accumulation of pathological intracellular material is a critical process involved in the pathogenesis of many neurodegenerative disorders, including Alzheimer's disease and Parkinson's disease. Autophagy is an essential mechanism that ensures neuronal health by removing long-lived proteins or defective organelles and by doing so prevents cell toxicity and death within the central nervous system. Abundant evidence has shown that neuronal autophagy pathways are altered in Alzheimer's disease, Parkinson's disease and traumas of the central nervous system including Spinal Cord Injury and Traumatic Brain Injury. In this review, we aimed to summarize the latest studies on the role that altered neuronal autophagy plays in brain health and these pathological conditions, and how this knowledge can be leveraged for the development of novel therapeutics against them.
Collapse
Affiliation(s)
- Alessia Filippone
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA; Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D' Alcontres 31. 98166 Messina, Italy
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D' Alcontres 31. 98166 Messina, Italy
| | - Deborah Mannino
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D' Alcontres 31. 98166 Messina, Italy
| | - Nicholas Lyssenko
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Domenico Praticò
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA.
| |
Collapse
|
28
|
Asadzadeh J, Ruchti E, Jiao W, Limoni G, MacLachlan C, Small SA, Knott G, Santa-Maria I, McCabe BD. Retromer deficiency in Tauopathy models enhances the truncation and toxicity of Tau. Nat Commun 2022; 13:5049. [PMID: 36030267 PMCID: PMC9420134 DOI: 10.1038/s41467-022-32683-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 08/10/2022] [Indexed: 11/24/2022] Open
Abstract
Alteration of the levels, localization or post-translational processing of the microtubule associated protein Tau is associated with many neurodegenerative disorders. Here we develop adult-onset models for human Tau (hTau) toxicity in Drosophila that enable age-dependent quantitative measurement of central nervous system synapse loss and axonal degeneration, in addition to effects upon lifespan, to facilitate evaluation of factors that may contribute to Tau-dependent neurodegeneration. Using these models, we interrogate the interaction of hTau with the retromer complex, an evolutionarily conserved cargo-sorting protein assembly, whose reduced activity has been associated with both Parkinson’s and late onset Alzheimer’s disease. We reveal that reduction of retromer activity induces a potent enhancement of hTau toxicity upon synapse loss, axon retraction and lifespan through a specific increase in the production of a C-terminal truncated isoform of hTau. Our data establish a molecular and subcellular mechanism necessary and sufficient for the depletion of retromer activity to exacerbate Tau-dependent neurodegeneration. Tau and the Retromer complex are both linked to Parkinson’s and Alzheimer’s disease. Using Drosophila neurodegeneration models, this study finds that low retromer activity induces a specific increase of a highly toxic truncated form of human Tau.
Collapse
Affiliation(s)
- Jamshid Asadzadeh
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - Evelyne Ruchti
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - Wei Jiao
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - Greta Limoni
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - Catherine MacLachlan
- BioEM Facility, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - Scott A Small
- Department of Neurology, Columbia University, New York, USA.,Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, USA
| | - Graham Knott
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland.,BioEM Facility, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland
| | - Ismael Santa-Maria
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, USA.,Department of Pathology & Cell Biology, Columbia University, New York, USA.,Facultad Ciencias Experimentales, Universidad Francisco de Vitoria, Pozuelo de Alarcón, Madrid, Spain
| | - Brian D McCabe
- Brain Mind Institute, EPFL - Swiss Federal Institute of Technology Lausanne, Lausanne, Switzerland.
| |
Collapse
|
29
|
Curtis ME, Smith T, Blass BE, Praticò D. Dysfunction of the retromer complex system contributes to amyloid and tau pathology in a stem cell model of Down syndrome. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2022; 8:e12334. [PMID: 35910668 PMCID: PMC9322819 DOI: 10.1002/trc2.12334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 11/16/2022]
Abstract
Introduction Retromer complex proteins are decreased in Down syndrome (DS) brains and correlate inversely with brain amyloidosis. However, whether retromer dysfunction contributes to the amyloid beta (Aβ) and tau neuropathology of DS remains unknown. Methods Human trisomic induced Pluripotent Stem Cells (iPSCs) and isogenic controls were differentiated into forebrain neurons, and changes in retromer proteins, tau phosphorylated epitopes, and Aβ levels were assessed in euploid and trisomic neurons using western blot and enzyme-linked immunosorbent assay (ELISA). Genetic overexpression and pharmacological retromer stabilization were used to determine the functional role of the retromer complex system in modulating amyloid and tau pathology. Results Trisomic neurons developed age-dependent retromer core protein deficiency associated with accumulation of Aβ peptides and phosphorylated tau isoforms. Enhancing retromer function through overexpression or pharmacological retromer stabilization reduced amyloid and tau pathology in trisomic neurons. However, the effect was greater using a pharmacological approach, suggesting that targeting the complex stability may be more effective in addressing this neuropathology in DS. Discussion Our results demonstrate that the retromer complex is directly involved in the development of the neuropathologic phenotype in DS, and that pharmacological stabilization of the complex should be considered as a novel therapeutic tool in people with DS.
Collapse
Affiliation(s)
- Mary Elizabeth Curtis
- Alzheimer's Center at TempleLewis Katz School of MedicineTemple UniversityPhiladelphiaPennsylvaniaUSA
| | - Tiffany Smith
- Alzheimer's Center at TempleLewis Katz School of MedicineTemple UniversityPhiladelphiaPennsylvaniaUSA
| | | | - Domenico Praticò
- Alzheimer's Center at TempleLewis Katz School of MedicineTemple UniversityPhiladelphiaPennsylvaniaUSA
| |
Collapse
|
30
|
ITRAQ-based quantitative proteomic analysis reveals that VPS35 promotes the expression of MCM2-7 genes in HeLa cells. Sci Rep 2022; 12:9700. [PMID: 35690672 PMCID: PMC9188599 DOI: 10.1038/s41598-022-13934-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 05/13/2022] [Indexed: 11/25/2022] Open
Abstract
Vacuolar protein sorting 35 (VPS35) is a major component of the retromer complex that regulates endosomal trafficking in eukaryotic cells. Recent studies have shown that VPS35 promotes tumor cell proliferation and affects the nuclear accumulation of its interacting partner. In this study, isobaric tags for relative and absolute quantitation (iTRAQ)-based mass spectrometry were used to measure the changes in nuclear protein abundance in VPS35-depleted HeLa cells. A total of 47 differentially expressed proteins were identified, including 27 downregulated and 20 upregulated proteins. Gene ontology (GO) analysis showed that the downregulated proteins included several minichromosome maintenance (MCM) proteins described as cell proliferation markers, and these proteins were present in the MCM2-7 complex, which is essential for DNA replication. Moreover, we validated that loss of VPS35 reduced the mRNA and protein expression of MCM2-7 genes. Notably, re-expression of VPS35 in VPS35 knockout HeLa cells rescued the expression of these genes. Functionally, we showed that VPS35 contributes to cell proliferation and maintenance of genomic stability of HeLa cells. Therefore, these findings reveal that VPS35 is involved in the regulation of MCM2-7 gene expression and establish a link between VPS35 and cell proliferation.
Collapse
|
31
|
Birdsall V, Kirwan K, Zhu M, Imoto Y, Wilson SM, Watanabe S, Waites CL. Axonal transport of Hrs is activity dependent and facilitates synaptic vesicle protein degradation. Life Sci Alliance 2022; 5:5/10/e202000745. [PMID: 35636965 PMCID: PMC9152131 DOI: 10.26508/lsa.202000745] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 11/29/2022] Open
Abstract
This study describes an activity-dependent mechanism for transporting ESCRT-0 protein Hrs to synaptic vesicle (SV) pools, facilitating SV protein degradation in response to increased neuronal firing. Turnover of synaptic vesicle (SV) proteins is vital for the maintenance of healthy and functional synapses. SV protein turnover is driven by neuronal activity in an endosomal sorting complex required for transport (ESCRT)-dependent manner. Here, we characterize a critical step in this process: axonal transport of ESCRT-0 component Hrs, necessary for sorting proteins into the ESCRT pathway and recruiting downstream ESCRT machinery to catalyze multivesicular body (MVB) formation. We find that neuronal activity stimulates the formation of presynaptic endosomes and MVBs, as well as the motility of Hrs+ vesicles in axons and their delivery to SV pools. Hrs+ vesicles co-transport ESCRT-0 component STAM1 and comprise a subset of Rab5+ vesicles, likely representing pro-degradative early endosomes. Furthermore, we identify kinesin motor protein KIF13A as essential for the activity-dependent transport of Hrs to SV pools and the degradation of SV membrane proteins. Together, these data demonstrate a novel activity- and KIF13A-dependent mechanism for mobilizing axonal transport of ESCRT machinery to facilitate the degradation of SV membrane proteins.
Collapse
Affiliation(s)
- Veronica Birdsall
- Neurobiology and Behavior PhD Program, Columbia University, New York, NY, USA
| | - Konner Kirwan
- Neurobiology and Behavior PhD Program, Columbia University, New York, NY, USA
| | - Mei Zhu
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Yuuta Imoto
- Department of Cell Biology, Johns Hopkins University, Baltimore, MD, USA
| | - Scott M Wilson
- Department of Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shigeki Watanabe
- Department of Cell Biology, Johns Hopkins University, Baltimore, MD, USA.,Solomon H Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - Clarissa L Waites
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA .,Department of Neuroscience, Columbia University, New York, NY, USA
| |
Collapse
|
32
|
Kuijpers M. Keeping synapses in shape: degradation pathways in the healthy and aging brain. Neuronal Signal 2022; 6:NS20210063. [PMID: 35813265 PMCID: PMC9208270 DOI: 10.1042/ns20210063] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 11/17/2022] Open
Abstract
Synapses maintain their molecular composition, plasticity and function through the concerted action of protein synthesis and removal. The complex and polarized neuronal architecture poses specific challenges to the logistics of protein and organelle turnover since protein synthesis and degradation mainly happen in the cell soma. In addition, post-mitotic neurons accumulate damage over a lifetime, challenging neuronal degradative pathways and making them particularly susceptible to the effects of aging. This review will summarize the current knowledge on neuronal protein turnover mechanisms with a particular focus on the presynapse, including the proteasome, autophagy and the endolysosomal route and their roles in regulating presynaptic proteostasis and function. In addition, the author will discuss how physiological brain aging, which entails a progressive decline in cognitive functions, affects synapses and the degradative machinery.
Collapse
Affiliation(s)
- Marijn Kuijpers
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Straße 10, 13125 Berlin, Germany
- Department of Molecular Neurobiology, Donders Institute for Brain, Cognition and Behaviour and Faculty of Science, Radboud University, Nijmegen, The Netherlands
| |
Collapse
|
33
|
Abstract
Complex mechanisms govern the sorting of membrane (cargo) proteins at endosomes to ensure that protein localization to the post-Golgi endomembrane system is accurately maintained. Endosomal retrieval complexes mediate sorting by recognizing specific motifs and signals in the cytoplasmic domains of cargo proteins transiting through endosomes. In this review, the recent progress in understanding the molecular mechanisms of how the retromer complex, in conjunction with sorting nexin (SNX) proteins, operates in cargo recognition and sorting is discussed. New data revealing the importance of different SNX proteins and detailing how post-translational modifications can modulate cargo sorting to respond to changes in the environment are highlighted along with the key role that endosomal protein sorting plays in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Xin Yong
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Lejiao Mao
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610041, China
| | - Matthew N J Seaman
- Cambridge Institute for Medical Research, University of Cambridge, The Keith Peters Building, Cambridge Biomedical Campus, CB2 0XY, UK
| | - Da Jia
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Department of Paediatrics, West China Second University Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
34
|
Behl T, Kaur D, Sehgal A, Singh S, Makeen HA, Albratty M, Abdellatif AAH, Dachani SR, Bungau S. Exploring the potential role of rab5 protein in endo-lysosomal impairment in Alzheimer's disease. Biomed Pharmacother 2022; 148:112773. [PMID: 35245734 DOI: 10.1016/j.biopha.2022.112773] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/18/2022] [Accepted: 02/27/2022] [Indexed: 11/02/2022] Open
Abstract
Growing evidence suggests that neuronal dysfunction in the endo-lysosomal and autophagic processes contributes to the onset and progression of neurodegenerative diseases such as Alzheimer's disease (AD). Since they are the primary cellular systems involved in the production and clearance of aggregated amyloid plaques, endo-lysosomal or autophagic equilibrium must be maintained throughout life. As a result, variations in the autophagic and endo-lysosomal torrent, as a measure of degenerative function in these sections or pathways, may have a direct impact on disease-related processes, such as Aß clearance from the brain and interneuronal deposition of Aß and tau aggregates, thus disrupting synaptic plasticity. The discovery of several chromosomal factors for Alzheimer's disease that are clinically linked to regulation of the endocytic pathway, including protein aggregation and removal, supports the theory that the endo-lysosomal/autophagic torrent is more susceptible to impairment, especially as people age, thus catalysing the onset of disease. Although the role of endo-lysosomal/autophagic dysfunction in neurodegeneration has progressed in recent years, the field remains underdeveloped. Because of its possible therapeutic implications in Alzheimer's disease, further study is needed to explain the possibilities for effective autophagy regulation.
Collapse
Affiliation(s)
- Tapan Behl
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India.
| | - Dapinder Kaur
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India
| | - Aayush Sehgal
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India
| | - Sukhbir Singh
- Department of Pharmacology, Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India
| | - Hafiz A Makeen
- Pharmacy Practice Research Unit, Clinical Pharmacy, Department, College of Pharmacy, Jazan University, P.O. Box-114, Jazan 45142, Saudi Arabia
| | - Mohammed Albratty
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, P.O. Box 114, Jazan 45142, Saudi Arabia
| | - Ahmed A H Abdellatif
- Department of Pharmaceutics, College of Pharmacy, Qassim University, Buraydah 51452, Saudi Arabia; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
| | - Sudharshan Reddy Dachani
- Department of Pharmacy Practice & Pharmacology, College of Pharmacy, Shaqra University, Al-Dawadmi Campus, Al-Dawadmi 11961, Saudi Arabia
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania.
| |
Collapse
|
35
|
Chae CW, Choi GE, Jung YH, Lim JR, Cho JH, Yoon JH, Han HJ. High glucose-mediated VPS26a downregulation dysregulates neuronal amyloid precursor protein processing and tau phosphorylation. Br J Pharmacol 2022; 179:3934-3950. [PMID: 35297035 DOI: 10.1111/bph.15836] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 02/07/2022] [Accepted: 02/19/2022] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND AND PURPOSE The relationship between hyperglycaemia-induced retromer dysfunction impairing intracellular trafficking and AD remains unclear, although Diabetes mellitus (DM) is considered a risk factor for Alzheimer's disease (AD). Here, we investigated the effects of high glucose on the retromer, and defined the dysregulation of mechanisms of amyloid precursor protein (APP) processing and tau phosphorylation. EXPERIMENTAL APPROACH We used human induced-pluripotent stem cell-derived neuronal differentiated cells and SH-SY5Ys exposed to high glucose to identify the underlying mechanisms. Streptozotocin-induced diabetic mice were used to elucidate whether the retromer contributes to the AD-like pathology. KEY RESULTS We found that vacuolar protein sorting-associated protein 26a (VPS26a) was decreased in the hippocampus of diabetic mice and high glucose-treated human neuronal cells. High glucose downregulated VPS26a through ROS/NF-κB/DNA methyltransferase1-mediated promoter hypermethylation. VPS26a recovery blocked retention of APP and cation-independent mannose-6-phosphate receptor in endosomes and promoted transport to the trans-Golgi, which decreased Aβ levels, and improved Cathepsin D activity, reducing p-tau levels, respectively. Retromer enhancement ameliorated synaptic deficits, astrocyte over-activation, and cognitive impairment in diabetic mice. CONCLUSION AND IMPLICATIONS In conclusion, VPS26a is a promising candidate for the inhibition of DM-associated AD pathogenesis by modulating APP processing and tau phosphorylation.
Collapse
Affiliation(s)
- Chang Woo Chae
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 Four Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, Republic of Korea
| | - Gee Euhn Choi
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 Four Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, Republic of Korea
| | - Young Hyun Jung
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 Four Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, Republic of Korea
| | - Jae Ryong Lim
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 Four Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, Republic of Korea
| | - Ji Hyeon Cho
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 Four Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, Republic of Korea
| | - Jee Hyeon Yoon
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 Four Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, Republic of Korea
| | - Ho Jae Han
- Department of Veterinary Physiology, College of Veterinary Medicine, Research Institute for Veterinary Science, and BK21 Four Future Veterinary Medicine Leading Education & Research Center, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
36
|
Curtis ME, Smith T, Yu D, Praticò D. The association of retromer deficiency and tau pathology in Down syndrome. Ann Neurol 2022; 91:561-567. [PMID: 35150166 PMCID: PMC8940634 DOI: 10.1002/ana.26321] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/17/2022] [Accepted: 02/10/2022] [Indexed: 11/09/2022]
Abstract
Retromer deficiency is reported in Down syndrome and correlates with amyloidosis, however, its association with tau neuropathology remains unclear. Down syndrome and control brain tissues were evaluated for phosphorylated tau, tau modulators, and cathepsin-D activity. Several kinases and phosphatase PP2A were unchanged, but tau phosphorylation was elevated, and cathepsin-D activity decreased in aged Down syndrome subjects. Retromer proteins positively associated with soluble tau, whereas pathogenic tau negatively correlated with retromer proteins and cathepsin-D activity. Retromer deficiency and consequent reduction of cathepsin-D activity may contribute to pathogenic tau accumulation, thus, retromer represents a viable therapeutic target against tau pathology in Down syndrome. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Mary Elizabeth Curtis
- Alzheimer's Center at Temple, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, 19140
| | - Tiffany Smith
- Alzheimer's Center at Temple, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, 19140
| | - Daohai Yu
- Department of Clinical Sciences, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, 19140
| | - Domenico Praticò
- Alzheimer's Center at Temple, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, 19140
| |
Collapse
|
37
|
Gock N, Follett J, Rintoul GL, Beischlag TV, Lee FJ. Endosomal recycling and dopamine neurotransmission: Exploring the links between the retromer and Parkinson's disease. Synapse 2022; 76:e22224. [DOI: 10.1002/syn.22224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 12/17/2021] [Accepted: 01/23/2022] [Indexed: 11/11/2022]
Affiliation(s)
- Nathan Gock
- Faculty of Health Sciences Simon Fraser University 8888 University Dr Burnaby BC V5A 1S6 Canada
- Centre for Cell Biology, Development, and Disease Simon Fraser University 8888 University Dr Burnaby BC V5A 1S6 Canada
| | - Jordan Follett
- Laboratory of Neurogenetics and Neuroscience Department of Neurology University of Florida 1149 Newell Dr Gainesville FL 32610‐0236 United States
| | - Gordon L Rintoul
- Department of Biological Sciences Simon Fraser University 8888 University Dr Burnaby BC V5A 1S6 Canada
- Centre for Cell Biology, Development, and Disease Simon Fraser University 8888 University Dr Burnaby BC V5A 1S6 Canada
| | - Timothy V Beischlag
- Faculty of Health Sciences Simon Fraser University 8888 University Dr Burnaby BC V5A 1S6 Canada
- Centre for Cell Biology, Development, and Disease Simon Fraser University 8888 University Dr Burnaby BC V5A 1S6 Canada
| | - Frank J.S. Lee
- Faculty of Health Sciences Simon Fraser University 8888 University Dr Burnaby BC V5A 1S6 Canada
- Centre for Cell Biology, Development, and Disease Simon Fraser University 8888 University Dr Burnaby BC V5A 1S6 Canada
| |
Collapse
|
38
|
Filippone A, Smith T, Pratico D. Dysregulation of the Retromer Complex in Brain Endothelial Cells Results in Accumulation of Phosphorylated Tau. J Inflamm Res 2022; 14:7455-7465. [PMID: 35002279 PMCID: PMC8721160 DOI: 10.2147/jir.s342096] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 12/16/2021] [Indexed: 01/18/2023] Open
Abstract
Introduction Transport through endothelial cells of the blood–brain barrier (BBB) involves a complex group of structures of the endo-lysosome system such as early and late endosomes, and the retromer complex system. Studies show that neuronal dysregulation of the vacuolar protein sorting 35 (VPS35), the main component of the retromer complex recognition core, results in altered protein trafficking and degradation and is involved in neurodegeneration. Since the functional role of VPS35 in endothelial cells has not been fully investigated, in the present study we aimed at characterizing the effect of its downregulation on these pathways. Methods Genetic silencing of VPS35 in human brain endothelial cells; measurement of retromer complex system proteins, autophagy and ubiquitin-proteasome systems. Results VPS35-downregulated endothelial cells had increased expression of LC3B2/1 and more ubiquitinated products, markers of autophagy flux and impaired proteasome activity, respectively. Additionally, compared with controls VPS35 downregulation resulted in significant accumulation of tau protein and its phosphorylated isoforms. Discussion Our findings demonstrate that in brain endothelial cells retromer complex dysfunction by influencing endosome-lysosome degradation pathways results in altered proteostasis. Restoration of the retromer complex system function should be considered a novel therapeutic approach to rescue endothelial protein transport.
Collapse
Affiliation(s)
- Alessia Filippone
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Tiffany Smith
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Domenico Pratico
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| |
Collapse
|
39
|
Deng S, Gan L, Liu C, Xu T, Zhou S, Guo Y, Zhang Z, Yang GY, Tian H, Tang Y. Roles of Ependymal Cells in the Physiology and Pathology of the Central Nervous System. Aging Dis 2022; 14:468-483. [PMID: 37008045 PMCID: PMC10017161 DOI: 10.14336/ad.2022.0826-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 08/26/2022] [Indexed: 11/18/2022] Open
Abstract
Ependymal cells are indispensable components of the central nervous system (CNS). They originate from neuroepithelial cells of the neural plate and show heterogeneity, with at least three types that are localized in different locations of the CNS. As glial cells in the CNS, accumulating evidence demonstrates that ependymal cells play key roles in mammalian CNS development and normal physiological processes by controlling the production and flow of cerebrospinal fluid (CSF), brain metabolism, and waste clearance. Ependymal cells have been attached to great importance by neuroscientists because of their potential to participate in CNS disease progression. Recent studies have demonstrated that ependymal cells participate in the development and progression of various neurological diseases, such as spinal cord injury and hydrocephalus, raising the possibility that they may serve as a potential therapeutic target for the disease. This review focuses on the function of ependymal cells in the developmental CNS as well as in the CNS after injury and discusses the underlying mechanisms of controlling the functions of ependymal cells.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Yaohui Tang
- Correspondence should be addressed to: Dr. Yaohui Tang, Med-X Research Institute and School of Biomedical Engineering Shanghai Jiao Tong University, Shanghai, China. .
| |
Collapse
|
40
|
Wen MH, Xie X, Huang PS, Yang K, Chen TY. Crossroads between membrane trafficking machinery and copper homeostasis in the nerve system. Open Biol 2021; 11:210128. [PMID: 34847776 PMCID: PMC8633785 DOI: 10.1098/rsob.210128] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Imbalanced copper homeostasis and perturbation of membrane trafficking are two common symptoms that have been associated with the pathogenesis of neurodegenerative and neurodevelopmental diseases. Accumulating evidence from biophysical, cellular and in vivo studies suggest that membrane trafficking orchestrates both copper homeostasis and neural functions-however, a systematic review of how copper homeostasis and membrane trafficking interplays in neurons remains lacking. Here, we summarize current knowledge of the general trafficking itineraries for copper transporters and highlight several critical membrane trafficking regulators in maintaining copper homeostasis. We discuss how membrane trafficking regulators may alter copper transporter distribution in different membrane compartments to regulate intracellular copper homeostasis. Using Parkinson's disease and MEDNIK as examples, we further elaborate how misregulated trafficking regulators may interplay parallelly or synergistically with copper dyshomeostasis in devastating pathogenesis in neurodegenerative diseases. Finally, we explore multiple unsolved questions and highlight the existing challenges to understand how copper homeostasis is modulated through membrane trafficking.
Collapse
Affiliation(s)
- Meng-Hsuan Wen
- Department of Chemistry, University of Houston, Houston, TX 77204, USA
| | - Xihong Xie
- Department of Chemistry, University of Houston, Houston, TX 77204, USA
| | - Pei-San Huang
- Department of Chemistry, University of Houston, Houston, TX 77204, USA
| | - Karen Yang
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Tai-Yen Chen
- Department of Chemistry, University of Houston, Houston, TX 77204, USA
| |
Collapse
|
41
|
Liu Q, Zhang G, Ji Z, Lin H. Molecular and cellular mechanisms of spastin in neural development and disease (Review). Int J Mol Med 2021; 48:218. [PMID: 34664680 PMCID: PMC8547542 DOI: 10.3892/ijmm.2021.5051] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/29/2021] [Indexed: 12/26/2022] Open
Abstract
Spastin is a microtubule (MT)‑severing enzyme identified from mutations of hereditary spastic paraplegia in 1999 and extensive studies indicate its vital role in various cellular activities. In the past two decades, efforts have been made to understand the underlying molecular mechanisms of how spastin is linked to neural development and disease. Recent studies on spastin have unraveled the mechanistic processes of its MT‑severing activity and revealed that spastin acts as an MT amplifier to mediate its remodeling, thus providing valuable insight into the molecular roles of spastin under physiological conditions. In addition, recent research has revealed multiple novel molecular mechanisms of spastin in cellular biological pathways, including endoplasmic reticulum shaping, calcium trafficking, fatty acid trafficking, as well as endosomal fission and trafficking. These processes are closely involved in axonal and dendritic development and maintenance. The current review presents recent biological advances regarding the molecular mechanisms of spastin at the cellular level and provides insight into how it affects neural development and disease.
Collapse
Affiliation(s)
- Qiuling Liu
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, P.R. China
| | - Guowei Zhang
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, P.R. China
| | - Zhisheng Ji
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, P.R. China
| | - Hongsheng Lin
- Department of Orthopedics, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong 510630, P.R. China
| |
Collapse
|
42
|
Filippone A, Li JG, Praticò D. VPS35 Downregulation Alters Degradation Pathways in Neuronal Cells. J Alzheimers Dis 2021; 84:1079-1089. [PMID: 34602481 DOI: 10.3233/jad-210701] [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] [Indexed: 12/11/2022]
Abstract
BACKGROUND The vacuolar protein sorting 35 (VPS35) is the main component of the retromer recognition core complex system which regulates intracellular cargo protein sorting and trafficking. Downregulation of VPS35 has been linked to the pathogenesis of neurodegenerative disorders such Alzheimer's and Parkinson's diseases via endosome dysregulation. OBJECTIVE Here we show that the genetic manipulation of VPS35 affects intracellular degradation pathways. METHODS A neuronal cell line expressing human APP Swedish mutant was used. VPS35 silencing was performed treating cells with VPS35 siRNA or Ctr siRNA for 72 h. RESULTS Downregulation of VPS35 was associated with alteration of autophagy flux and intracellular accumulation of acidic and ubiquitinated aggregates suggesting that dysfunction of the retromer recognition core leads to a significant alteration in both pathways. CONCLUSION Taken together, our data demonstrate that besides cargo sorting and trafficking, VPS35 by supporting the integral function of the retromer complex system plays an important role also as a critical regulator of intracellular degradation pathways.
Collapse
Affiliation(s)
- Alessia Filippone
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Jian-Guo Li
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Domenico Praticò
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
43
|
Liu T, Zhang T, Nicolas M, Boussicault L, Rice H, Soldano A, Claeys A, Petrova I, Fradkin L, De Strooper B, Potier MC, Hassan BA. The amyloid precursor protein is a conserved Wnt receptor. eLife 2021; 10:69199. [PMID: 34515635 PMCID: PMC8437438 DOI: 10.7554/elife.69199] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 09/01/2021] [Indexed: 12/31/2022] Open
Abstract
The Amyloid Precursor Protein (APP) and its homologues are transmembrane proteins required for various aspects of neuronal development and activity, whose molecular function is unknown. Specifically, it is unclear whether APP acts as a receptor, and if so what its ligand(s) may be. We show that APP binds the Wnt ligands Wnt3a and Wnt5a and that this binding regulates APP protein levels. Wnt3a binding promotes full-length APP (flAPP) recycling and stability. In contrast, Wnt5a promotes APP targeting to lysosomal compartments and reduces flAPP levels. A conserved Cysteine-Rich Domain (CRD) in the extracellular portion of APP is required for Wnt binding, and deletion of the CRD abrogates the effects of Wnts on flAPP levels and trafficking. Finally, loss of APP results in increased axonal and reduced dendritic growth of mouse embryonic primary cortical neurons. This phenotype can be cell-autonomously rescued by full length, but not CRD-deleted, APP and regulated by Wnt ligands in a CRD-dependent manner.
Collapse
Affiliation(s)
- Tengyuan Liu
- Paris Brain Institute - Institut du Cerveau, Sorbonne Université, Inserm, CNRS, Hôpital Pitié-Salpêtrière, Paris, France.,Doctoral School of Biomedical Sciences, Leuven, Belgium
| | - Tingting Zhang
- Paris Brain Institute - Institut du Cerveau, Sorbonne Université, Inserm, CNRS, Hôpital Pitié-Salpêtrière, Paris, France.,Doctoral School of Biomedical Sciences, Leuven, Belgium
| | - Maya Nicolas
- Doctoral School of Biomedical Sciences, Leuven, Belgium.,Center for Brain and Disease, Leuven, Belgium.,Center for Human Genetics, University of Leuven School of Medicine, Leuven, Belgium
| | - Lydie Boussicault
- Paris Brain Institute - Institut du Cerveau, Sorbonne Université, Inserm, CNRS, Hôpital Pitié-Salpêtrière, Paris, France
| | - Heather Rice
- Center for Brain and Disease, Leuven, Belgium.,Center for Human Genetics, University of Leuven School of Medicine, Leuven, Belgium
| | - Alessia Soldano
- Center for Brain and Disease, Leuven, Belgium.,Center for Human Genetics, University of Leuven School of Medicine, Leuven, Belgium
| | - Annelies Claeys
- Center for Brain and Disease, Leuven, Belgium.,Center for Human Genetics, University of Leuven School of Medicine, Leuven, Belgium
| | - Iveta Petrova
- Laboratory of Developmental Neurobiology, Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Lee Fradkin
- Laboratory of Developmental Neurobiology, Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, Netherlands
| | - Bart De Strooper
- Center for Brain and Disease, Leuven, Belgium.,UK Dementia Research institute at University College London, London, United Kingdom
| | - Marie-Claude Potier
- Paris Brain Institute - Institut du Cerveau, Sorbonne Université, Inserm, CNRS, Hôpital Pitié-Salpêtrière, Paris, France
| | - Bassem A Hassan
- Paris Brain Institute - Institut du Cerveau, Sorbonne Université, Inserm, CNRS, Hôpital Pitié-Salpêtrière, Paris, France
| |
Collapse
|
44
|
Unveiling the cryo-EM structure of retromer. Biochem Soc Trans 2021; 48:2261-2272. [PMID: 33125482 DOI: 10.1042/bst20200552] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/10/2020] [Accepted: 09/14/2020] [Indexed: 12/29/2022]
Abstract
Retromer (VPS26/VPS35/VPS29) is a highly conserved eukaryotic protein complex that localizes to endosomes to sort transmembrane protein cargoes into vesicles and elongated tubules. Retromer mediates retrieval pathways from endosomes to the trans-Golgi network in all eukaryotes and further facilitates recycling pathways to the plasma membrane in metazoans. In cells, retromer engages multiple partners to orchestrate the formation of tubulovesicular structures, including sorting nexin (SNX) proteins, cargo adaptors, GTPases, regulators, and actin remodeling proteins. Retromer-mediated pathways are especially important for sorting cargoes required for neuronal maintenance, which links retromer loss or mutations to multiple human brain diseases and disorders. Structural and biochemical studies have long contributed to the understanding of retromer biology, but recent advances in cryo-electron microscopy and cryo-electron tomography have further uncovered exciting new snapshots of reconstituted retromer structures. These new structures reveal retromer assembles into an arch-shaped scaffold and suggest the scaffold may be flexible and adaptable in cells. Interactions with cargo adaptors, particularly SNXs, likely orient the scaffold with respect to phosphatidylinositol-3-phosphate (PtdIns3P)-enriched membranes. Pharmacological small molecule chaperones have further been shown to stabilize retromer in cultured cell and mouse models, but mechanisms by which these molecules bind remain unknown. This review will emphasize recent structural and biophysical advances in understanding retromer structure as the field moves towards a molecular view of retromer assembly and regulation on membranes.
Collapse
|
45
|
Lai SSM, Ng KY, Koh RY, Chok KC, Chye SM. Endosomal-lysosomal dysfunctions in Alzheimer's disease: Pathogenesis and therapeutic interventions. Metab Brain Dis 2021; 36:1087-1100. [PMID: 33881723 DOI: 10.1007/s11011-021-00737-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 04/08/2021] [Indexed: 12/14/2022]
Abstract
The endosomal-lysosomal system mediates the process of protein degradation through endocytic pathway. This system consists of early endosomes, late endosomes, recycling endosomes and lysosomes. Each component in the endosomal-lysosomal system plays individual crucial role and they work concordantly to ensure protein degradation can be carried out functionally. Dysregulation in the endosomal-lysosomal system can contribute to the pathogenesis of neurodegenerative diseases such as Alzheimer's disease (AD). In AD endosomal-lysosomal abnormalities are the earliest pathological features to note and hence it is important to understand the involvement of endosomal-lysosomal dysfunction in the pathogenesis of AD. In-depth understanding of this dysfunction can allow development of new therapeutic intervention to prevent and treat AD.
Collapse
Affiliation(s)
- Shereen Shi Min Lai
- School of Health Science, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Khuen Yen Ng
- School of Pharmacy, Monash University Malaysia, 47500, Selangor, Malaysia
| | - Rhun Yian Koh
- Division of Biomedical Science and Biotechnology, School of Health Science, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia
| | - Kian Chung Chok
- School of Health Science, International Medical University, 57000, Kuala Lumpur, Malaysia
| | - Soi Moi Chye
- Division of Biomedical Science and Biotechnology, School of Health Science, International Medical University, No. 126, Jalan Jalil Perkasa 19, Bukit Jalil, 57000, Kuala Lumpur, Malaysia.
| |
Collapse
|
46
|
Recent developments in membrane curvature sensing and induction by proteins. Biochim Biophys Acta Gen Subj 2021; 1865:129971. [PMID: 34333084 DOI: 10.1016/j.bbagen.2021.129971] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 07/11/2021] [Accepted: 07/25/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND Membrane-bound intracellular organelles have characteristic shapes attributed to different local membrane curvatures, and these attributes are conserved across species. Over the past decade, it has been confirmed that specific proteins control the large curvatures of the membrane, whereas many others due to their specific structural features can sense the curvatures and bind to the specific geometrical cues. Elucidating the interplay between sensing and induction is indispensable to understand the mechanisms behind various biological processes such as vesicular trafficking and budding. SCOPE OF REVIEW We provide an overview of major classes of membrane proteins and the mechanisms of curvature sensing and induction. We then discuss the importance of membrane elastic characteristics to induce the membrane shapes similar to intracellular organelles. Finally, we survey recently available assays developed for studying the curvature sensing and induction by many proteins. MAJOR CONCLUSIONS Recent theoretical/computational modeling along with experimental studies have uncovered fascinating connections between lipid membrane and protein interactions. However, the phenomena of protein localization and synchronization to generate spatiotemporal dynamics in membrane morphology are yet to be fully understood. GENERAL SIGNIFICANCE The understanding of protein-membrane interactions is essential to shed light on various biological processes. This further enables the technological applications of many natural proteins/peptides in therapeutic treatments. The studies of membrane dynamic shapes help to understand the fundamental functions of membranes, while the medicinal roles of various macromolecules (such as proteins, peptides, etc.) are being increasingly investigated.
Collapse
|
47
|
Conditional GWAS analysis to identify disorder-specific SNPs for psychiatric disorders. Mol Psychiatry 2021; 26:2070-2081. [PMID: 32398722 PMCID: PMC7657979 DOI: 10.1038/s41380-020-0705-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 12/06/2019] [Accepted: 02/24/2020] [Indexed: 01/19/2023]
Abstract
Substantial genetic liability is shared across psychiatric disorders but less is known about risk variants that are specific to a given disorder. We used multi-trait conditional and joint analysis (mtCOJO) to adjust GWAS summary statistics of one disorder for the effects of genetically correlated traits to identify putative disorder-specific SNP associations. We applied mtCOJO to summary statistics for five psychiatric disorders from the Psychiatric Genomics Consortium-schizophrenia (SCZ), bipolar disorder (BIP), major depression (MD), attention-deficit hyperactivity disorder (ADHD) and autism (AUT). Most genome-wide significant variants for these disorders had evidence of pleiotropy (i.e., impact on multiple psychiatric disorders) and hence have reduced mtCOJO conditional effect sizes. However, subsets of genome-wide significant variants had larger conditional effect sizes consistent with disorder-specific effects: 15 of 130 genome-wide significant variants for schizophrenia, 5 of 40 for major depression, 3 of 11 for ADHD and 1 of 2 for autism. We show that decreased expression of VPS29 in the brain may increase risk to SCZ only and increased expression of CSE1L is associated with SCZ and MD, but not with BIP. Likewise, decreased expression of PCDHA7 in the brain is linked to increased risk of MD but decreased risk of SCZ and BIP.
Collapse
|
48
|
Chandra M, Kendall AK, Jackson LP. Toward Understanding the Molecular Role of SNX27/Retromer in Human Health and Disease. Front Cell Dev Biol 2021; 9:642378. [PMID: 33937239 PMCID: PMC8083963 DOI: 10.3389/fcell.2021.642378] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 03/22/2021] [Indexed: 11/30/2022] Open
Abstract
Aberrations in membrane trafficking pathways have profound effects in cellular dynamics of cellular sorting processes and can drive severe physiological outcomes. Sorting nexin 27 (SNX27) is a metazoan-specific sorting nexin protein from the PX-FERM domain family and is required for endosomal recycling of many important transmembrane receptors. Multiple studies have shown SNX27-mediated recycling requires association with retromer, one of the best-known regulators of endosomal trafficking. SNX27/retromer downregulation is strongly linked to Down's Syndrome (DS) via glutamate receptor dysfunction and to Alzheimer's Disease (AD) through increased intracellular production of amyloid peptides from amyloid precursor protein (APP) breakdown. SNX27 is further linked to addiction via its role in potassium channel trafficking, and its over-expression is linked to tumorigenesis, cancer progression, and metastasis. Thus, the correct sorting of multiple receptors by SNX27/retromer is vital for normal cellular function to prevent human diseases. The role of SNX27 in regulating cargo recycling from endosomes to the cell surface is firmly established, but how SNX27 assembles with retromer to generate tubulovesicular carriers remains elusive. Whether SNX27/retromer may be a putative therapeutic target to prevent neurodegenerative disease is now an emerging area of study. This review will provide an update on our molecular understanding of endosomal trafficking events mediated by the SNX27/retromer complex on endosomes.
Collapse
Affiliation(s)
- Mintu Chandra
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, United States
- Center for Structural Biology, Vanderbilt University, Nashville, TN, United States
| | - Amy K. Kendall
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, United States
- Center for Structural Biology, Vanderbilt University, Nashville, TN, United States
| | - Lauren P. Jackson
- Department of Biological Sciences, Vanderbilt University, Nashville, TN, United States
- Center for Structural Biology, Vanderbilt University, Nashville, TN, United States
- Department of Biochemistry, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
49
|
Kawabe H, Stegmüller J. The role of E3 ubiquitin ligases in synapse function in the healthy and diseased brain. Mol Cell Neurosci 2021; 112:103602. [DOI: 10.1016/j.mcn.2021.103602] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 01/06/2021] [Accepted: 02/02/2021] [Indexed: 02/08/2023] Open
|
50
|
Leneva N, Kovtun O, Morado DR, Briggs JAG, Owen DJ. Architecture and mechanism of metazoan retromer:SNX3 tubular coat assembly. SCIENCE ADVANCES 2021; 7:7/13/eabf8598. [PMID: 33762348 PMCID: PMC7990337 DOI: 10.1126/sciadv.abf8598] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/05/2021] [Indexed: 05/12/2023]
Abstract
Retromer is a master regulator of cargo retrieval from endosomes, which is critical for many cellular processes including signaling, immunity, neuroprotection, and virus infection. The retromer core (VPS26/VPS29/VPS35) is present on cargo-transporting, tubular carriers along with a range of sorting nexins. Here, we elucidate the structural basis of membrane tubulation and coupled cargo recognition by metazoan and fungal retromer coats assembled with the non-Bin1/Amphiphysin/Rvs (BAR) sorting nexin SNX3 using cryo-electron tomography. The retromer core retains its arched, scaffolding structure but changes its mode of membrane recruitment when assembled with different SNX adaptors, allowing cargo recognition at subunit interfaces. Thus, membrane bending and cargo incorporation can be modulated to allow retromer to traffic cargoes along different cellular transport routes.
Collapse
Affiliation(s)
- Natalya Leneva
- Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK.
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - Oleksiy Kovtun
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK.
| | - Dustin R Morado
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - John A G Briggs
- MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK.
| | - David J Owen
- Cambridge Institute for Medical Research, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK.
| |
Collapse
|