1
|
Song S, Rhee S. CKAP4 is a potential therapeutic target to overcome resistance to EGFR-TKIs in lung adenocarcinoma. Genes Genomics 2025; 47:331-340. [PMID: 39704929 DOI: 10.1007/s13258-024-01606-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 12/04/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND Epidermal growth factor receptor-tyrosine kinase inhibitors (EGFR-TKIs) are standard treatments for non-small cell lung cancer (NSCLC) patients with EGFR mutations; however, drug resistance limits their efficacy. Cytoskeleton-associated protein 4 (CKAP4) has been linked to cancer progression, but its role in EGFR-TKI resistance remains unclear. OBJECTIVE This study investigates the clinical relevance of CKAP4 as a therapeutic target to overcome EGFR-TKI resistance in lung adenocarcinoma (LUAD) patients. METHODS GEO datasets were analyzed to identify 24 differentially expressed genes associated with EGFR-TKI resistance, with CKAP4 selected via functional annotation and scoring using the VarElect tool. The prognostic significance of CKAP4 was evaluated using public databases, and its upregulation was confirmed in osimertinib-tolerant H1975 cells through quantitative reverse transcription-polymerase chain reaction. RESULTS Integrated bioinformatics analysis identified CKAP4 as strongly associated with EGFR-TKI resistance. Elevated CKAP4 expression was particularly linked to poorer clinical outcomes in LUAD patients. Notably, osimertinib-tolerant cells exhibited high CKAP4 expression, correlating positively with increased half-maximal inhibitory concentrations of EGFR-TKIs. LUAD patients with upregulated CKAP4 showed significantly reduced overall and relapse-free survival. CONCLUSION This study underscores the prognostic value of CKAP4 in EGFR-mutated LUAD and highlights its potential as a therapeutic target to counter EGFR-TKI resistance.
Collapse
Affiliation(s)
- Seongeun Song
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Sangmyung Rhee
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea.
| |
Collapse
|
2
|
Hdedeh O, Mercier C, Poitout A, Martinière A, Zelazny E. Membrane nanodomains to shape plant cellular functions and signaling. THE NEW PHYTOLOGIST 2025; 245:1369-1385. [PMID: 39722237 PMCID: PMC11754938 DOI: 10.1111/nph.20367] [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: 08/16/2024] [Accepted: 11/19/2024] [Indexed: 12/28/2024]
Abstract
Plasma membrane (PM) nanodomains have emerged as pivotal elements in the regulation of plant cellular functions and signal transduction. These nanoscale membrane regions, enriched in specific lipids and proteins, behave as regulatory/signaling hubs spatially and temporally coordinating critical cellular functions. In this review, we first examine the mechanisms underlying the formation and maintenance of PM nanodomains in plant cells, highlighting the roles of PM lipid composition, protein oligomerization and interactions with cytoskeletal and cell wall components. Then, we discuss how nanodomains act as organizing centers by mediating protein-protein interactions that orchestrate essential processes such as symbiosis, defense against pathogens, ion transport or hormonal and reactive oxygen species (ROS) signaling. Finally, we introduce the concept of nanoenvironments, where localized physicochemical variations are generated in the very close proximity of PM nanodomains, in response to stimuli. After decoding by a dedicated machinery likely localized in the vicinity of nanodomains, this enrichment of secondary messengers, such as ROS or Ca2+, would allow specific downstream cellular responses. This review provides insights into the dynamic nature of nanodomains and proposes future research to better understand their contribution to the intricate signaling networks that govern plant development and stress responses.
Collapse
Affiliation(s)
- Omar Hdedeh
- IPSiM, Univ Montpellier, CNRS, INRAE, Institut AgroMontpellier34000France
| | - Caroline Mercier
- IPSiM, Univ Montpellier, CNRS, INRAE, Institut AgroMontpellier34000France
| | - Arthur Poitout
- IPSiM, Univ Montpellier, CNRS, INRAE, Institut AgroMontpellier34000France
| | | | - Enric Zelazny
- IPSiM, Univ Montpellier, CNRS, INRAE, Institut AgroMontpellier34000France
| |
Collapse
|
3
|
Falahati H, Wu Y, Fang M, De Camilli P. Ectopic reconstitution of a spine-apparatus-like structure provides insight into mechanisms underlying its formation. Curr Biol 2025; 35:265-276.e4. [PMID: 39626668 PMCID: PMC11753949 DOI: 10.1016/j.cub.2024.11.010] [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: 06/10/2024] [Revised: 09/25/2024] [Accepted: 11/06/2024] [Indexed: 12/11/2024]
Abstract
The endoplasmic reticulum (ER) is a continuous cellular endomembrane network that displays focal specializations. Most notable examples of such specializations include the spine apparatus of neuronal dendrites and the cisternal organelle of axonal initial segments. Both organelles exhibit stacks of smooth ER sheets with a narrow lumen, interconnected by a dense protein matrix. The actin-binding protein synaptopodin is required for their formation, but the underlying mechanisms remain unknown. Here, we report that the spine apparatus and synaptopodin are conserved from flies to mammals and that a highly conserved region of this protein is necessary, but not sufficient, for its association with ER. We reveal a dual role of synaptopodin in generating actin bundles and in linking them to the ER. Expression of a synaptopodin construct constitutively anchored to the ER in non-neuronal cells is sufficient to generate stacked ER cisterns resembling the spine apparatus. Cisterns within these stacks are molecularly distinct from the surrounding ER and are connected to each other by an actin-based matrix that contains proteins also found at the spine apparatus of neuronal spines. Our findings shed light on mechanisms governing the biogenesis of this peculiar structure and represent a step toward understanding the elusive properties of this organelle.
Collapse
Affiliation(s)
- Hanieh Falahati
- Departments of Neuroscience and of Cell Biology, Howard Hughes Medical Institute, Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale School of Medicine, New Haven, CT 06510, USA
| | - Yumei Wu
- Departments of Neuroscience and of Cell Biology, Howard Hughes Medical Institute, Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale School of Medicine, New Haven, CT 06510, USA
| | - Mumu Fang
- Departments of Neuroscience and of Cell Biology, Howard Hughes Medical Institute, Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale School of Medicine, New Haven, CT 06510, USA
| | - Pietro De Camilli
- Departments of Neuroscience and of Cell Biology, Howard Hughes Medical Institute, Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
4
|
Zheng J, Cardoen B, Ortiz-Silva M, Hamarneh G, Nabi IR. Comparative Analysis of SPLICS and MCS-DETECT for Detecting Mitochondria-ER Contact Sites (MERCs). CONTACT (THOUSAND OAKS (VENTURA COUNTY, CALIF.)) 2025; 8:25152564251313721. [PMID: 40115170 PMCID: PMC11923443 DOI: 10.1177/25152564251313721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 11/26/2024] [Accepted: 12/23/2024] [Indexed: 03/23/2025]
Abstract
Detection of mitochondria-ER contacts (MERCs) from diffraction limited confocal images commonly uses fluorescence colocalization analysis of mitochondria and endoplasmic reticulum (ER) as well as split fluorescent probes, such as the split-GFP-based contact site sensor (SPLICS). However, inter-organelle distances (∼10-60 nm) for MERCs are lower than the 200-250 nm diffraction limited resolution obtained by standard confocal microscopy. Super-resolution microscopy of 3D volume analysis provides a two-fold resolution improvement (∼120 nm XY; 250 nm Z), which remains unable to resolve MERCs. MCS-DETECT, a membrane contact site (MCS) detection algorithm faithfully detects elongated ribosome-studded riboMERCs when applied to 3D STED super-resolution image volumes. Here, we expressed the SPLICSL reporter in HeLa cells co-transfected with the ER reporter RFP-KDEL and label fixed cells with antibodies to RFP and the mitochondrial protein TOM20. MCS-DETECT analysis of 3D STED volumes was compared to contacts determined by co-occurrence colocalization analysis of mitochondria and ER or the SPLICSL probe. Percent mitochondria coverage by MCS-DETECT derived contacts was significantly smaller than those obtained for colocalization analysis or SPLICSL, and more closely matched contact site metrics obtained by 3D electron microscopy. Further, STED analysis localized a subset of the SPLICSL label to mitochondria with some SPLICSL puncta observed to be completely enveloped by mitochondria in 3D views. These data suggest that MCS-DETECT reports on a limited set of MERCs that more closely corresponds to those observed by EM.
Collapse
Affiliation(s)
- Jieyi Zheng
- Department of Cellular & Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Ben Cardoen
- School of Computing Science, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Milene Ortiz-Silva
- Department of Cellular & Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Ghassan Hamarneh
- School of Computing Science, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Ivan R Nabi
- Department of Cellular & Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|
5
|
Htet PH, Avezov E, Lauga E. Fluid mechanics of luminal transport in actively contracting endoplasmic reticulum. eLife 2024; 13:RP93518. [PMID: 39671235 PMCID: PMC11643642 DOI: 10.7554/elife.93518] [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] [Indexed: 12/14/2024] Open
Abstract
The endoplasmic reticulum (ER), the largest cellular compartment, harbours the machinery for the biogenesis of secretory proteins and lipids, calcium storage/mobilisation, and detoxification. It is shaped as layered membranous sheets interconnected with a network of tubules extending throughout the cell. Understanding the influence of the ER morphology dynamics on molecular transport may offer clues to rationalising neuro-pathologies caused by ER morphogen mutations. It remains unclear, however, how the ER facilitates its intra-luminal mobility and homogenises its content. It has been recently proposed that intra-luminal transport may be enabled by active contractions of ER tubules. To surmount the barriers to empirical studies of the minuscule spatial and temporal scales relevant to ER nanofluidics, here we exploit the principles of viscous fluid dynamics to generate a theoretical physical model emulating in silico the content motion in actively contracting nanoscopic tubular networks. The computational model reveals the luminal particle speeds, and their impact in facilitating active transport, of the active contractile behaviour of the different ER components along various time-space parameters. The results of the model indicate that reproducing transport with velocities similar to those reported experimentally in single-particle tracking would require unrealistically high values of tubule contraction site length and rate. Considering further nanofluidic scenarios, we show that width contractions of the ER's flat domains (perinuclear sheets) generate local flows with only a short-range effect on luminal transport. Only contractions of peripheral sheets can reproduce experimental measurements, provided they are able to contract fast enough.
Collapse
Affiliation(s)
- Pyae Hein Htet
- Department of Applied Mathematics and Theoretical Physics, University of CambridgeCambridgeUnited Kingdom
| | - Edward Avezov
- UK Dementia Research Institute at University of Cambridge, Department of Clinical Neurosciences, University of CambridgeCambridgeUnited Kingdom
| | - Eric Lauga
- Department of Applied Mathematics and Theoretical Physics, University of CambridgeCambridgeUnited Kingdom
| |
Collapse
|
6
|
Zou HM, Yu J, Ruan YY, Xie Y, An XM, Chen PL, Luo YQ, Shi MJ, Liu M, Xu LF, Liu J, Guo B, Zhang F. HNF-1β alleviates podocyte injury in lupus nephritis by maintaining endoplasmic reticulum homeostasis. Lupus Sci Med 2024; 11:e001349. [PMID: 39608815 PMCID: PMC11603702 DOI: 10.1136/lupus-2024-001349] [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: 08/09/2024] [Accepted: 11/08/2024] [Indexed: 11/30/2024]
Abstract
OBJECTIVE The current study aims to elucidate the critical function of hepatocyte nuclear factor 1-beta (HNF1-β) in lupus nephritis (LN) by investigating its modulation of the Derlin-1/valosin-containing protein (VCP)/VCP-interacting membrane selenoprotein (VIMP) complex, endoplasmic reticulum (ER) stress and podocyte apoptosis. METHODS In vitro and in vivo models of LN were established using glomerular podocytes treated with LN serum and MRL/lpr mice, respectively. The expression levels of HNF1-β were analysed in kidney tissues from patients with LN and MRL/lpr mice. To assess the effects of HNF1-β inhibition, an adeno-associated virus vector carrying HNF1-β short hairpin was administered to MRL/lpr mice. In vitro, glomerular podocytes were transfected with HNF1-β small interfering RNA (siRNA) or HNF1-β overexpression plasmids to explore their regulatory effects on the Derlin-1/VCP/VIMP complex and podocyte apoptosis. Dual-luciferase reporter assays and chromatin immunoprecipitation (ChIP) assays were performed to investigate the transcriptional activation of Derlin-1 and VCP promoters by HNF1-β. RESULTS A significant decrease in HNF1-β levels was observed in kidney tissues from patients with LN while MRL/lpr mice exhibited an initial compensatory increase followed by a subsequent decrease in renal HNF1-β expression. Overexpression of HNF1-β transcriptionally upregulated Derlin-1 and VCP mitigating LN serum-induced ER stress and podocyte apoptosis. In contrast, HNF1-β inhibition exacerbated renal dysfunction and structural damage in MRL/lpr mice. Interestingly, HNF1-β inhibition transcriptionally repressed ERP44, leading to calcium ions (Ca²+) release-mediated disruption and inactivation of the Derlin-1/VCP/VIMP complex. This finding suggests that HNF1-β not only regulates the expression of key proteins in the Derlin-1/VCP/VIMP complex but also influences their assembly through Ca²+ release regulation. CONCLUSION This study provides novel insights into the regulatory mechanisms of HNF1-β in LN emphasising its impact on the Derlin-1/VCP/VIMP complex, ER stress and podocyte apoptosis. These findings have the potential to inform the development of new diagnostic tools and therapeutic strategies for LN.
Collapse
Affiliation(s)
- Hui-mei Zou
- Department of Pathophysiology, Guizhou Medical University, Guiyang, China
- School of Nursing, Guizhou Medical University, Guiyang, China
| | - Jie Yu
- Department of Pathophysiology, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, China
| | - Yuan-yuan Ruan
- Department of Pathophysiology, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, China
| | - Ying Xie
- Department of Pathophysiology, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, China
| | - Xiao-min An
- Department of Pathophysiology, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, China
| | - Pei-lei Chen
- Department of Pathophysiology, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, China
| | - Ying-qin Luo
- Department of Pathophysiology, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, China
| | - Ming-jun Shi
- Department of Pathophysiology, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, China
| | - Miao Liu
- Department of Urinary Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Li-fen Xu
- Department of Pathology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Jun Liu
- Department of Rheumatology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Bing Guo
- Department of Pathophysiology, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, China
| | - Fan Zhang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, China
- Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, China
| |
Collapse
|
7
|
Sun Q, Yu L, Donnelly SC, Fradin C, Thompson RT, Prato FS, Goldhawk DE. Essential magnetosome proteins MamI and MamL from magnetotactic bacteria interact in mammalian cells. Sci Rep 2024; 14:26292. [PMID: 39487238 PMCID: PMC11530650 DOI: 10.1038/s41598-024-77591-4] [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: 02/20/2024] [Accepted: 10/23/2024] [Indexed: 11/04/2024] Open
Abstract
To detect cellular activities deep within the body using magnetic resonance platforms, magnetosomes are the ideal model of genetically-encoded nanoparticles. These membrane-bound iron biominerals produced by magnetotactic bacteria are highly regulated by approximately 30 genes; however, the number of magnetosome genes that are essential and/or constitute the root structure upon which biominerals form is largely undefined. To examine the possibility that key magnetosome genes may interact in a foreign environment, we expressed mamI and mamL as fluorescent fusion proteins in mammalian cells. Localization and potential protein-protein interaction(s) were investigated using confocal microscopy and fluorescence correlation spectroscopy (FCS). Enhanced green fluorescent protein (EGFP)-MamI and the red fluorescent Tomato-MamL displayed distinct intracellular localization, with net-like and punctate fluorescence, respectively. Remarkably, co-expression revealed co-localization of both fluorescent fusion proteins in the same punctate pattern. An interaction between MamI and MamL was confirmed by co-immunoprecipitation. In addition, changes in EGFP-MamI distribution were accompanied by acquisition of intracellular mobility which all Tomato-MamL structures displayed. Analysis of extracts from these cells by FCS was consistent with an interaction between fluorescent fusion proteins, including an increase in particle radius. Co-localization and interaction of MamI and MamL demonstrate that select magnetosome proteins may associate in mammalian cells.
Collapse
Affiliation(s)
- Qin Sun
- Imaging, Lawson Research Institute, London, ON, Canada
- Medical Biophysics, Western University, London, ON, Canada
- Collaborative Graduate Program in Molecular Imaging, Western University, London, ON, Canada
| | - Liu Yu
- Physics & Astronomy, McMaster University, Hamilton, ON, Canada
| | | | - Cécile Fradin
- Physics & Astronomy, McMaster University, Hamilton, ON, Canada
- Biochemistry & Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - R Terry Thompson
- Imaging, Lawson Research Institute, London, ON, Canada
- Medical Biophysics, Western University, London, ON, Canada
- Medical Imaging, Western University, London, ON, Canada
- Physics & Astronomy, Western University, London, ON, Canada
| | - Frank S Prato
- Imaging, Lawson Research Institute, London, ON, Canada
- Medical Biophysics, Western University, London, ON, Canada
- Collaborative Graduate Program in Molecular Imaging, Western University, London, ON, Canada
- Medical Imaging, Western University, London, ON, Canada
| | - Donna E Goldhawk
- Imaging, Lawson Research Institute, London, ON, Canada.
- Medical Biophysics, Western University, London, ON, Canada.
- Collaborative Graduate Program in Molecular Imaging, Western University, London, ON, Canada.
- Imaging Program, Lawson Research Institute, 268 Grosvenor St. , PO Box 5777 Station B, London, ON, N6A 4V2, Canada.
| |
Collapse
|
8
|
Zhang LL, Zhang DJ, Shi JX, Huang MY, Yu JM, Chen XJ, Wei X, Zou L, Lu JJ. Immunogenic cell death inducers for cancer therapy: An emerging focus on natural products. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155828. [PMID: 38905847 DOI: 10.1016/j.phymed.2024.155828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/03/2024] [Accepted: 06/12/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Immunogenic cell death (ICD) is a specific form of regulated cell death induced by a variety of stressors. During ICD, the dying cancer cells release damage-associated molecular patterns (DAMPs), which promote dendritic cell maturation and tumor antigen presentation, subsequently triggering a T-cell-mediated anti-tumor immune response. In recent years, a growing number of studies have demonstrated the potential of natural products to induce ICD and enhance tumor cell immunogenicity. Moreover, there is an increasing interest in identifying new ICD inducers from natural products. PURPOSE This study aimed to emphasize the potential of natural products and their derivatives as ICD inducers to promote research on using natural products in cancer therapy and provide ideas for future novel immunotherapies based on ICD induction. METHOD This review included a thorough search of the PubMed, Web of Science, Scopus, and Google Scholar databases to identify natural products with ICD-inducing capabilities. A comprehensive search for clinical trials on natural ICD inducers was also conducted using ClinicalTrials.gov, as well as the approved patents using the Espacenet and CNKI Patent Database. RESULTS Natural compounds that induce ICD can be categorized into several groups, such as polyphenols, flavonoids, terpenoids, and alkaloids. Natural products can induce the release of DAMPs by triggering endoplasmic reticulum stress, activation of autophagy-related pathways, and reactive oxygen species generation, etc. Ultimately, they activate anti-tumor immune response and improve the efficacy of cancer treatments. CONCLUSION A growing number of ICD inducers from natural products with promising anti-cancer potential have been identified. The detailed information presented in this review will contribute to the further development of natural ICD inducers and cancer treatment strategies based on ICD-induced responses.
Collapse
Affiliation(s)
- Le-Le Zhang
- School of Basic Medical Sciences, Chengdu University, Chengdu 610106, China; State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China; Hubei Key Laboratory of Wudang Local Chinese Medicine Research, Hubei University of Medicine, Shiyan 442000, China
| | - Du-Juan Zhang
- College of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Jia-Xin Shi
- College of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Mu-Yang Huang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China
| | - Jia-Mei Yu
- College of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Xu-Jia Chen
- College of Pharmacy, Chengdu University, Chengdu 610106, China
| | - Xiao Wei
- School of Basic Medical Sciences, Chengdu University, Chengdu 610106, China
| | - Liang Zou
- Key Laboratory of Coarse Cereal Processing, Ministry of Agriculture and Rural Affairs, Sichuan Engineering & Technology Research Center of Coarse Cereal Industrialization, Chengdu University, Chengdu 610106, China.
| | - Jin-Jian Lu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao 999078, China; Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao 999078, China.
| |
Collapse
|
9
|
Nabi IR, Cardoen B, Khater IM, Gao G, Wong TH, Hamarneh G. AI analysis of super-resolution microscopy: Biological discovery in the absence of ground truth. J Cell Biol 2024; 223:e202311073. [PMID: 38865088 PMCID: PMC11169916 DOI: 10.1083/jcb.202311073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 04/02/2024] [Accepted: 05/21/2024] [Indexed: 06/13/2024] Open
Abstract
Super-resolution microscopy, or nanoscopy, enables the use of fluorescent-based molecular localization tools to study molecular structure at the nanoscale level in the intact cell, bridging the mesoscale gap to classical structural biology methodologies. Analysis of super-resolution data by artificial intelligence (AI), such as machine learning, offers tremendous potential for the discovery of new biology, that, by definition, is not known and lacks ground truth. Herein, we describe the application of weakly supervised paradigms to super-resolution microscopy and its potential to enable the accelerated exploration of the nanoscale architecture of subcellular macromolecules and organelles.
Collapse
Affiliation(s)
- Ivan R. Nabi
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| | - Ben Cardoen
- School of Computing Science, Simon Fraser University, Burnaby, Canada
| | - Ismail M. Khater
- School of Computing Science, Simon Fraser University, Burnaby, Canada
- Department of Electrical and Computer Engineering, Faculty of Engineering and Technology, Birzeit University, Birzeit, Palestine
| | - Guang Gao
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Timothy H. Wong
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Ghassan Hamarneh
- School of Computing Science, Simon Fraser University, Burnaby, Canada
| |
Collapse
|
10
|
Konno T, Parutto P, Crapart CC, Davì V, Bailey DMD, Awadelkareem MA, Hockings C, Brown AI, Xiang KM, Agrawal A, Chambers JE, Vander Werp MJ, Koning KM, Elfari LM, Steen S, Metzakopian E, Westrate LM, Koslover EF, Avezov E. Endoplasmic reticulum morphology regulation by RTN4 modulates neuronal regeneration by curbing luminal transport. Cell Rep 2024; 43:114357. [PMID: 38955182 DOI: 10.1016/j.celrep.2024.114357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/01/2024] [Accepted: 05/29/2024] [Indexed: 07/04/2024] Open
Abstract
Cell functions rely on intracellular transport systems distributing bioactive molecules with high spatiotemporal accuracy. The endoplasmic reticulum (ER) tubular network constitutes a system for delivering luminal solutes, including Ca2+, across the cell periphery. How the ER structure enables this nanofluidic transport system is unclear. Here, we show that ER membrane-localized reticulon 4 (RTN4/Nogo) is sufficient to impose neurite outgrowth inhibition in human cortical neurons while acting as an ER morphoregulator. Improving ER transport visualization methodologies combined with optogenetic Ca2+ dynamics imaging and in silico modeling, we observed that ER luminal transport is modulated by ER tubule narrowing and dilation, proportional to the amount of RTN4. Excess RTN4 limited ER luminal transport and Ca2+ release, while RTN4 elimination reversed the effects. The described morphoregulatory effect of RTN4 defines the capacity of the ER for peripheral Ca2+ delivery for physiological releases and thus may constitute a mechanism for controlling the (re)generation of neurites.
Collapse
Affiliation(s)
- Tasuku Konno
- UK Dementia Research Institute at University of Cambridge, Department of Clinical Neurosciences, Cambridge CB2 0AH, UK
| | - Pierre Parutto
- UK Dementia Research Institute at University of Cambridge, Department of Clinical Neurosciences, Cambridge CB2 0AH, UK
| | - Cécile C Crapart
- UK Dementia Research Institute at University of Cambridge, Department of Clinical Neurosciences, Cambridge CB2 0AH, UK
| | - Valentina Davì
- UK Dementia Research Institute at University of Cambridge, Department of Clinical Neurosciences, Cambridge CB2 0AH, UK
| | | | - Mosab Ali Awadelkareem
- UK Dementia Research Institute at University of Cambridge, Department of Clinical Neurosciences, Cambridge CB2 0AH, UK; Department of Neuroscience Biomedicine and Movement Sciences, University of Verona, Verona, Italy
| | - Colin Hockings
- UK Dementia Research Institute at University of Cambridge, Department of Clinical Neurosciences, Cambridge CB2 0AH, UK
| | - Aidan I Brown
- Department of Physics, University of California, San Diego, 9500 Gilman Dr. #0374, La Jolla, CA 92093-0374, USA; Department of Physics, Toronto Metropolitan University, Toronto, ON M5B 2K3, Canada
| | | | - Anamika Agrawal
- Department of Physics, University of California, San Diego, 9500 Gilman Dr. #0374, La Jolla, CA 92093-0374, USA
| | - Joseph E Chambers
- Cambridge Institute for Medical Research (CIMR), Department of Medicine, University of Cambridge, The Keith Peters Building, Hills Road, Cambridge CB2 0XY, UK
| | - Molly J Vander Werp
- Department of Chemistry and Biochemistry, Calvin University, Grand Rapids, MI 49546, USA
| | - Katherine M Koning
- Department of Chemistry and Biochemistry, Calvin University, Grand Rapids, MI 49546, USA
| | - Louis Mounir Elfari
- Wellcome-MRC Cambridge Stem Cell Institute Advanced Imaging Facility, Puddicombe Way, Cambridge CB2 0AW, UK
| | - Sam Steen
- Department of Chemistry and Biochemistry, Calvin University, Grand Rapids, MI 49546, USA
| | - Emmanouil Metzakopian
- UK Dementia Research Institute at University of Cambridge, Department of Clinical Neurosciences, Cambridge CB2 0AH, UK
| | - Laura M Westrate
- Department of Chemistry and Biochemistry, Calvin University, Grand Rapids, MI 49546, USA
| | - Elena F Koslover
- Department of Physics, University of California, San Diego, 9500 Gilman Dr. #0374, La Jolla, CA 92093-0374, USA.
| | - Edward Avezov
- UK Dementia Research Institute at University of Cambridge, Department of Clinical Neurosciences, Cambridge CB2 0AH, UK.
| |
Collapse
|
11
|
Falahati H, Wu Y, De Camilli P. Ectopic Reconstitution of a Spine-Apparatus Like Structure Provides Insight into Mechanisms Underlying Its Formation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.16.589782. [PMID: 38659799 PMCID: PMC11042382 DOI: 10.1101/2024.04.16.589782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
The endoplasmic reticulum (ER) is a continuous cellular endomembrane network that displays focal specializations. Most notable examples of such specializations include the spine apparatus of neuronal dendrites, and the cisternal organelle of axonal initial segments. Both organelles exhibit stacks of smooth ER sheets with a narrow lumen and interconnected by a dense protein matrix. The actin-binding protein synaptopodin is required for their formation. Here, we report that expression in non-neuronal cells of a synaptopodin construct targeted to the ER is sufficient to generate stacked ER cisterns resembling the spine apparatus with molecular properties distinct from the surrounding ER. Cisterns within these stacks are connected to each other by an actin-based matrix that contains proteins also found at the spine apparatus of neuronal spines. These findings reveal a critical role of a synaptopodin-dependent actin matrix in generating cisternal stacks. These ectopically generated structures provide insight into spine apparatus morphogenesis.
Collapse
Affiliation(s)
- Hanieh Falahati
- HHMI; Departments of Neuroscience and Cell Biology; Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, 100 College Street, New Haven, 06511, CT, USA
| | - Yumei Wu
- HHMI; Departments of Neuroscience and Cell Biology; Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, 100 College Street, New Haven, 06511, CT, USA
| | - Pietro De Camilli
- HHMI; Departments of Neuroscience and Cell Biology; Program in Cellular Neuroscience, Neurodegeneration, and Repair, Yale University School of Medicine, 100 College Street, New Haven, 06511, CT, USA
| |
Collapse
|
12
|
Mann MJ, Melendez-Suchi C, Vorndran HE, Sukhoplyasova M, Flory AR, Irvine MC, Iyer AR, Guerriero CJ, Brodsky JL, Hendershot LM, Buck TM. Loss of Grp170 results in catastrophic disruption of endoplasmic reticulum function. Mol Biol Cell 2024; 35:ar59. [PMID: 38446639 PMCID: PMC11064666 DOI: 10.1091/mbc.e24-01-0012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 02/20/2024] [Accepted: 02/29/2024] [Indexed: 03/08/2024] Open
Abstract
GRP170 (Hyou1) is required for mouse embryonic development, and its ablation in kidney nephrons leads to renal failure. Unlike most chaperones, GRP170 is the lone member of its chaperone family in the ER lumen. However, the cellular requirement for GRP170, which both binds nonnative proteins and acts as nucleotide exchange factor for BiP, is poorly understood. Here, we report on the isolation of mouse embryonic fibroblasts obtained from mice in which LoxP sites were engineered in the Hyou1 loci (Hyou1LoxP/LoxP). A doxycycline-regulated Cre recombinase was stably introduced into these cells. Induction of Cre resulted in depletion of Grp170 protein which culminated in cell death. As Grp170 levels fell we observed a portion of BiP fractionating with insoluble material, increased binding of BiP to a client with a concomitant reduction in its turnover, and reduced solubility of an aggregation-prone BiP substrate. Consistent with disrupted BiP functions, we observed reactivation of BiP and induction of the unfolded protein response (UPR) in futile attempts to provide compensatory increases in ER chaperones and folding enzymes. Together, these results provide insights into the cellular consequences of controlled Grp170 loss and provide hypotheses as to why mutations in the Hyou1 locus are linked to human disease.
Collapse
Affiliation(s)
- Melissa J. Mann
- Department of Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, TN 30105
| | - Chris Melendez-Suchi
- Department of Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, TN 30105
| | - Hannah E. Vorndran
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260
| | - Maria Sukhoplyasova
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260
| | - Ashley R. Flory
- Department of Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, TN 30105
| | - Mary Carson Irvine
- Department of Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, TN 30105
| | - Anuradha R. Iyer
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260
| | | | - Jeffrey L. Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260
| | - Linda M. Hendershot
- Department of Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, TN 30105
| | - Teresa M. Buck
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260
| |
Collapse
|
13
|
Cardoen B, Vandevoorde KR, Gao G, Ortiz-Silva M, Alan P, Liu W, Tiliakou E, Vogl AW, Hamarneh G, Nabi IR. Membrane contact site detection (MCS-DETECT) reveals dual control of rough mitochondria-ER contacts. J Cell Biol 2024; 223:e202206109. [PMID: 37948126 PMCID: PMC10638097 DOI: 10.1083/jcb.202206109] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 09/20/2022] [Accepted: 10/23/2023] [Indexed: 11/12/2023] Open
Abstract
Identification and morphological analysis of mitochondria-ER contacts (MERCs) by fluorescent microscopy is limited by subpixel resolution interorganelle distances. Here, the membrane contact site (MCS) detection algorithm, MCS-DETECT, reconstructs subpixel resolution MERCs from 3D super-resolution image volumes. MCS-DETECT shows that elongated ribosome-studded riboMERCs, present in HT-1080 but not COS-7 cells, are morphologically distinct from smaller smooth contacts and larger contacts induced by mitochondria-ER linker expression in COS-7 cells. RiboMERC formation is associated with increased mitochondrial potential, reduced in Gp78 knockout HT-1080 cells and induced by Gp78 ubiquitin ligase activity in COS-7 and HeLa cells. Knockdown of riboMERC tether RRBP1 eliminates riboMERCs in both wild-type and Gp78 knockout HT-1080 cells. By MCS-DETECT, Gp78-dependent riboMERCs present complex tubular shapes that intercalate between and contact multiple mitochondria. MCS-DETECT of 3D whole-cell super-resolution image volumes, therefore, identifies novel dual control of tubular riboMERCs, whose formation is dependent on RRBP1 and size modulated by Gp78 E3 ubiquitin ligase activity.
Collapse
Affiliation(s)
- Ben Cardoen
- School of Computing Science, Simon Fraser University, Burnaby, Canada
| | - Kurt R. Vandevoorde
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Guang Gao
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Milene Ortiz-Silva
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Parsa Alan
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - William Liu
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Ellie Tiliakou
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - A. Wayne Vogl
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Ghassan Hamarneh
- School of Computing Science, Simon Fraser University, Burnaby, Canada
| | - Ivan R. Nabi
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, Canada
| |
Collapse
|
14
|
Mann MJ, Melendez-Suchi C, Sukhoplyasova M, Flory AR, Carson Irvine M, Iyer AR, Vorndran H, Guerriero CJ, Brodsky JL, Hendershot LM, Buck TM. Loss of Grp170 results in catastrophic disruption of endoplasmic reticulum functions. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.19.563191. [PMID: 37905119 PMCID: PMC10614942 DOI: 10.1101/2023.10.19.563191] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
GRP170, a product of the Hyou1 gene, is required for mouse embryonic development, and its ablation in kidney nephrons leads to renal failure. Unlike most chaperones, GRP170 is the lone member of its chaperone family in the ER lumen. However, the cellular requirement for GRP170, which both binds non-native proteins and acts as nucleotide exchange factor for BiP, is poorly understood. Here, we report on the isolation of embryonic fibroblasts from mice in which LoxP sites were engineered in the Hyou1 loci ( Hyou1 LoxP/LoxP ). A doxycycline-regulated Cre recombinase was also stably introduced into these cells. Induction of Cre resulted in excision of Hyou1 and depletion of Grp170 protein, culminating in apoptotic cell death. As Grp170 levels fell we observed increased steady-state binding of BiP to a client, slowed degradation of a misfolded BiP substrate, and BiP accumulation in NP40-insoluble fractions. Consistent with disrupted BiP functions, we observed reactivation of BiP storage pools and induction of the unfolded protein response (UPR) in futile attempts to provide compensatory increases in ER chaperones and folding enzymes. Together, these results provide insights into the cellular consequences of controlled Grp170 loss and insights into mutations in the Hyou1 locus and human disease.
Collapse
|
15
|
Wang P, Duckney P, Gao E, Hussey PJ, Kriechbaumer V, Li C, Zang J, Zhang T. Keep in contact: multiple roles of endoplasmic reticulum-membrane contact sites and the organelle interaction network in plants. THE NEW PHYTOLOGIST 2023; 238:482-499. [PMID: 36651025 DOI: 10.1111/nph.18745] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 12/02/2022] [Indexed: 06/17/2023]
Abstract
Functional regulation and structural maintenance of the different organelles in plants contribute directly to plant development, reproduction and stress responses. To ensure these activities take place effectively, cells have evolved an interconnected network amongst various subcellular compartments, regulating rapid signal transduction and the exchange of biomaterial. Many proteins that regulate membrane connections have recently been identified in plants, and this is the first step in elucidating both the mechanism and function of these connections. Amongst all organelles, the endoplasmic reticulum is the key structure, which likely links most of the different subcellular compartments through membrane contact sites (MCS) and the ER-PM contact sites (EPCS) have been the most intensely studied in plants. However, the molecular composition and function of plant MCS are being found to be different from other eukaryotic systems. In this article, we will summarise the most recent advances in this field and discuss the mechanism and biological relevance of these essential links in plants.
Collapse
Affiliation(s)
- Pengwei Wang
- National Key Laboratory for Germplasm Innovation & Utilization of Horticultural Crops, College of Horticulture & Forestry Sciences, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Patrick Duckney
- Department of Biosciences, Durham University, South Road, Durham, DH1 3LE, UK
| | - Erlin Gao
- National Key Laboratory for Germplasm Innovation & Utilization of Horticultural Crops, College of Horticulture & Forestry Sciences, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Patrick J Hussey
- Department of Biosciences, Durham University, South Road, Durham, DH1 3LE, UK
| | - Verena Kriechbaumer
- Endomembrane Structure and Function Research Group, Department of Biological and Medical Sciences, Oxford Brookes University, Oxford, OX3 0BP, UK
| | - Chengyang Li
- National Key Laboratory for Germplasm Innovation & Utilization of Horticultural Crops, College of Horticulture & Forestry Sciences, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Jingze Zang
- National Key Laboratory for Germplasm Innovation & Utilization of Horticultural Crops, College of Horticulture & Forestry Sciences, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
| | - Tong Zhang
- National Key Laboratory for Germplasm Innovation & Utilization of Horticultural Crops, College of Horticulture & Forestry Sciences, Huazhong Agricultural University, Wuhan, 430070, China
- Hubei Hongshan Laboratory, Wuhan, 430070, China
| |
Collapse
|
16
|
Han K, Huang S, Kong J, Yang Y, Shi L, Ci Y. A novel fluorescent endoplasmic reticulum marker for super-resolution imaging in live cells. FEBS Lett 2023; 597:693-701. [PMID: 36694281 DOI: 10.1002/1873-3468.14581] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 01/26/2023]
Abstract
Endoplasmic reticulum (ER) is a highly complicated and dynamic organelle that actively changes its shape and communicates with other organelles. Visualization of ER in live cells is of great importance to understand cellular activities. Here, we designed a novel ER marker, RR-mNeonGreen, which comprised an N-terminal ER retention signal, a bright fluorescent protein (mNeonGreen), and a C-terminal transmembrane region. Colocalization of RR-mNeonGreen with mCherry-KDEL verified that RR-mNeonGreen perfectly labeled the ER. RR-mNeonGreen showed better continuity of ER tubules when imaged by super-resolution microscopy. Moreover, RR-mNeonGreen is competent for live-cell imaging of ER dynamics and tracing of the interaction between ER and mitochondria at high spatiotemporal resolution. In summary, RR-mNeonGreen is a novel ER marker for super-resolution live-cell imaging with multiple merits.
Collapse
Affiliation(s)
- Kai Han
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Shuhan Huang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Jie Kong
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Yang Yang
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Lei Shi
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Yali Ci
- State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
- Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, China
| |
Collapse
|
17
|
Sandoz PA, Denhardt-Eriksson RA, Abrami L, Abriata LA, Spreemann G, Maclachlan C, Ho S, Kunz B, Hess K, Knott G, S Mesquita F, Hatzimanikatis V, van der Goot FG. Dynamics of CLIMP-63 S-acylation control ER morphology. Nat Commun 2023; 14:264. [PMID: 36650170 PMCID: PMC9844198 DOI: 10.1038/s41467-023-35921-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 01/06/2023] [Indexed: 01/19/2023] Open
Abstract
The complex architecture of the endoplasmic reticulum (ER) comprises distinct dynamic features, many at the nanoscale, that enable the coexistence of the nuclear envelope, regions of dense sheets and a branched tubular network that spans the cytoplasm. A key player in the formation of ER sheets is cytoskeleton-linking membrane protein 63 (CLIMP-63). The mechanisms by which CLIMP-63 coordinates ER structure remain elusive. Here, we address the impact of S-acylation, a reversible post-translational lipid modification, on CLIMP-63 cellular distribution and function. Combining native mass-spectrometry, with kinetic analysis of acylation and deacylation, and data-driven mathematical modelling, we obtain in-depth understanding of the CLIMP-63 life cycle. In the ER, it assembles into trimeric units. These occasionally exit the ER to reach the plasma membrane. However, the majority undergoes S-acylation by ZDHHC6 in the ER where they further assemble into highly stable super-complexes. Using super-resolution microscopy and focused ion beam electron microscopy, we show that CLIMP-63 acylation-deacylation controls the abundance and fenestration of ER sheets. Overall, this study uncovers a dynamic lipid post-translational regulation of ER architecture.
Collapse
Affiliation(s)
- Patrick A Sandoz
- Global Health Institute, School of Life Sciences, EPFL, Lausanne, Switzerland
| | | | - Laurence Abrami
- Global Health Institute, School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Luciano A Abriata
- Laboratory for Biomolecular Modelling, Institute of Bioengineering, EPFL and Swiss Institute of Bioinformatics, Lausanne, Switzerland.,Protein Production and Structure Core Facility, School of Life Sciences, EPFL, Lausanne, Switzerland
| | | | | | - Sylvia Ho
- Global Health Institute, School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Béatrice Kunz
- Global Health Institute, School of Life Sciences, EPFL, Lausanne, Switzerland
| | - Kathryn Hess
- Brain Mind Institute, EPFL, Lausanne, Switzerland
| | - Graham Knott
- BioEM Facility, School of Life Sciences, EPFL, Lausanne, Switzerland
| | | | | | | |
Collapse
|
18
|
Cardoen B, Wong T, Alan P, Lee S, Matsubara JA, Nabi IR, Hamarneh G. SPECHT: Self-tuning Plausibility based object detection Enables quantification of Conflict in Heterogeneous multi-scale microscopy. PLoS One 2022; 17:e0276726. [PMID: 36580473 PMCID: PMC9799313 DOI: 10.1371/journal.pone.0276726] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 10/12/2022] [Indexed: 12/30/2022] Open
Abstract
Identification of small objects in fluorescence microscopy is a non-trivial task burdened by parameter-sensitive algorithms, for which there is a clear need for an approach that adapts dynamically to changing imaging conditions. Here, we introduce an adaptive object detection method that, given a microscopy image and an image level label, uses kurtosis-based matching of the distribution of the image differential to express operator intent in terms of recall or precision. We show how a theoretical upper bound of the statistical distance in feature space enables application of belief theory to obtain statistical support for each detected object, capturing those aspects of the image that support the label, and to what extent. We validate our method on 2 datasets: distinguishing sub-diffraction limit caveolae and scaffold by stimulated emission depletion (STED) super-resolution microscopy; and detecting amyloid-β deposits in confocal microscopy retinal cross-sections of neuropathologically confirmed Alzheimer's disease donor tissue. Our results are consistent with biological ground truth and with previous subcellular object classification results, and add insight into more nuanced class transition dynamics. We illustrate the novel application of belief theory to object detection in heterogeneous microscopy datasets and the quantification of conflict of evidence in a joint belief function. By applying our method successfully to diffraction-limited confocal imaging of tissue sections and super-resolution microscopy of subcellular structures, we demonstrate multi-scale applicability.
Collapse
Affiliation(s)
- Ben Cardoen
- Medical Image Analysis Laboratory, School of Computing Science, Simon Fraser University, Burnaby, British Columbia, Canada
- * E-mail: (BC); (IRN); (GH)
| | - Timothy Wong
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Parsa Alan
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sieun Lee
- Department of Ophthalmology and Visual Sciences, Eye Care Centre, University of British Columbia, Vancouver, British Columbia, Canada
- Mental Health & Clinical Neurosciences, School of Medicine, University of Nottingham, Nottingham, United Kingdom
| | - Joanne Aiko Matsubara
- Department of Ophthalmology and Visual Sciences, Eye Care Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Ivan Robert Nabi
- Department of Cellular and Physiological Sciences, Life Sciences Institute, University of British Columbia, Vancouver, British Columbia, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, British Columbia, Canada
- * E-mail: (BC); (IRN); (GH)
| | - Ghassan Hamarneh
- Medical Image Analysis Laboratory, School of Computing Science, Simon Fraser University, Burnaby, British Columbia, Canada
| |
Collapse
|
19
|
Xue Y, Jia PF, Li HJ. SUN3/4/5 proteins regulate endoplasmic reticulum tubule formation and luminal spacing in Arabidopsis. J Genet Genomics 2022; 50:370-373. [PMID: 36402306 DOI: 10.1016/j.jgg.2022.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 11/13/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022]
Affiliation(s)
- Yong Xue
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Peng-Fei Jia
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Hong-Ju Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China.
| |
Collapse
|
20
|
Bonet-Ponce L, Cookson MR. The endoplasmic reticulum contributes to lysosomal tubulation/sorting driven by LRRK2. Mol Biol Cell 2022; 33:ar124. [PMID: 36044336 PMCID: PMC9634967 DOI: 10.1091/mbc.e22-04-0139] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Lysosomes are dynamic organelles that can remodel their membrane as an adaptive response to various cell signaling events including membrane damage. Recently, we have discovered that damaged lysosomes form and sort tubules into moving vesicles. We named this process LYTL for LYsosomal Tubulation/sorting driven by LRRK2, as the Parkinson's disease protein LRRK2 promotes tubulation by recruiting the motor adaptor protein JIP4 to lysosomes via phosphorylated RAB proteins. Here we use spinning-disk microscopy combined with superresolution to further characterize LYTL after membrane damage with LLOMe (l-leucyl-l-leucine methyl ester). We identified the endoplasmic reticulum (ER) colocalizing with sites of fission of lysosome-derived tubules. In addition, modifying the morphology of the ER by reducing ER tubules leads to a decrease in LYTL sorting, suggesting that contact with tubular ER is necessary for lysosomal membrane sorting. Given the central roles of LRRK2 and lysosomal biology in Parkinson's disease, these discoveries are likely relevant to disease pathology and highlight interactions between organelles in this model.
Collapse
Affiliation(s)
- Luis Bonet-Ponce
- Cell Biology and Gene Expression Section, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892,*Address correspondence to: Mark R. Cookson (); Luis Bonet-Ponce ()
| | - Mark R. Cookson
- Cell Biology and Gene Expression Section, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892,*Address correspondence to: Mark R. Cookson (); Luis Bonet-Ponce ()
| |
Collapse
|
21
|
Janota CS, Pinto A, Pezzarossa A, Machado P, Costa J, Campinho P, Franco CA, Gomes ER. Shielding of actin by the endoplasmic reticulum impacts nuclear positioning. Nat Commun 2022; 13:2763. [PMID: 35589708 PMCID: PMC9120458 DOI: 10.1038/s41467-022-30388-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 04/28/2022] [Indexed: 11/09/2022] Open
Abstract
Nuclear position is central to cell polarization, and its disruption is associated with various pathologies. The nucleus is moved away from the leading edge of migrating cells through its connection to moving dorsal actin cables, and the absence of connections to immobile ventral stress fibers. It is unclear how these asymmetric nucleo-cytoskeleton connections are established. Here, using an in vitro wound assay, we find that remodeling of endoplasmic reticulum (ER) impacts nuclear positioning through the formation of a barrier that shields immobile ventral stress fibers. The remodeling of ER and perinuclear ER accumulation is mediated by the ER shaping protein Climp-63. Furthermore, ectopic recruitment of the ER to stress fibers restores nuclear positioning in the absence of Climp-63. Our findings suggest that the ER mediates asymmetric nucleo-cytoskeleton connections to position the nucleus.
Collapse
Affiliation(s)
- Cátia Silva Janota
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Andreia Pinto
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Royal Brompton Hospital and Harefield NHS Foundation Trust, London, UK
| | - Anna Pezzarossa
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Champalimaud Foundation, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Pedro Machado
- Electron Microscopy Core Facility (EMCF), European Molecular Biology Laboratory, Heidelberg, Germany.,Centre for Ultrastructural Imaging, King's College London, London, UK
| | - Judite Costa
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Pedro Campinho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Champalimaud Foundation, Champalimaud Centre for the Unknown, Lisbon, Portugal
| | - Cláudio A Franco
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.,Instituto de Histologia e Biologia do Desenvolvimento, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Edgar R Gomes
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal. .,Instituto de Histologia e Biologia do Desenvolvimento, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
| |
Collapse
|
22
|
Yones SA, Annett A, Stoll P, Diamanti K, Holmfeldt L, Barrenäs CF, Meadows JRS, Komorowski J. Interpretable machine learning identifies paediatric Systemic Lupus Erythematosus subtypes based on gene expression data. Sci Rep 2022; 12:7433. [PMID: 35523803 PMCID: PMC9076598 DOI: 10.1038/s41598-022-10853-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 04/13/2022] [Indexed: 11/25/2022] Open
Abstract
Transcriptomic analyses are commonly used to identify differentially expressed genes between patients and controls, or within individuals across disease courses. These methods, whilst effective, cannot encompass the combinatorial effects of genes driving disease. We applied rule-based machine learning (RBML) models and rule networks (RN) to an existing paediatric Systemic Lupus Erythematosus (SLE) blood expression dataset, with the goal of developing gene networks to separate low and high disease activity (DA1 and DA3). The resultant model had an 81% accuracy to distinguish between DA1 and DA3, with unsupervised hierarchical clustering revealing additional subgroups indicative of the immune axis involved or state of disease flare. These subgroups correlated with clinical variables, suggesting that the gene sets identified may further the understanding of gene networks that act in concert to drive disease progression. This included roles for genes (i) induced by interferons (IFI35 and OTOF), (ii) key to SLE cell types (KLRB1 encoding CD161), or (iii) with roles in autophagy and NF-κB pathway responses (CKAP4). As demonstrated here, RBML approaches have the potential to reveal novel gene patterns from within a heterogeneous disease, facilitating patient clinical and therapeutic stratification.
Collapse
Affiliation(s)
- Sara A Yones
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden.
| | - Alva Annett
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Patricia Stoll
- Department of Biosystems Science and Engineering, ETH Zurich, Zurich, Switzerland
| | - Klev Diamanti
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Linda Holmfeldt
- Science for Life Laboratory, Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Carl Fredrik Barrenäs
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden
| | - Jennifer R S Meadows
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Jan Komorowski
- Science for Life Laboratory, Department of Cell and Molecular Biology, Uppsala University, Uppsala, Sweden.
- Washington National Primate Research Center, Seattle, USA.
- Swedish Collegium for Advanced Study, Uppsala, Sweden.
- The Institute of Computer Science, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
23
|
Abstract
The endoplasmic reticulum (ER) is one of the most structurally visible and functionally important organelles in the cell. Utilizing superresolution microscopy, we here unveil that in the mammalian cell, the peripheral ER adopts two distinct, well-defined tubule forms of contrasting structures, molecular signatures, and functions, with one of the two curiously being ribbon-like, ultranarrow sheets of fixed widths. With fast multicolor microscopy, we further show how the two tubule forms dynamically interconvert while differentially accommodating proteins in the living cell. The endoplasmic reticulum (ER) is a versatile organelle with diverse functions. Through superresolution microscopy, we show that the peripheral ER in the mammalian cell adopts two distinct forms of tubules. Whereas an ultrathin form, R1, is consistently covered by ER-membrane curvature-promoting proteins, for example, Rtn4 in the native cell, in the second form, R2, Rtn4 and analogs are arranged into two parallel lines at a conserved separation of ∼105 nm over long ranges. The two tubule forms together account for ∼90% of the total tubule length in the cell, with either one being dominant in different cell types. The R1–R2 dichotomy and the final tubule geometry are both coregulated by Rtn4 (and analogs) and the ER sheet–maintaining protein Climp63, which, respectively, define the edge curvature and lumen height of the R2 tubules to generate a ribbon-like structure of well-defined width. Accordingly, the R2 tubule width correlates positively with the Climp63 intraluminal size. The R1 and R2 tubules undergo active remodeling at the second/subsecond timescales as they differently accommodate proteins, with the former effectively excluding ER-luminal proteins and ER-membrane proteins with large intraluminal domains. We thus uncover a dynamic structural dichotomy for ER tubules with intriguing functional implications.
Collapse
|
24
|
Porter AW, Nguyen DN, Clayton DR, Ruiz WG, Mutchler SM, Ray EC, Marciszyn AL, Nkashama LJ, Subramanya AR, Gingras S, Kleyman TR, Apodaca G, Hendershot LM, Brodsky JL, Buck TM. The molecular chaperone GRP170 protects against ER stress and acute kidney injury in mice. JCI Insight 2022; 7:e151869. [PMID: 35104250 PMCID: PMC8983141 DOI: 10.1172/jci.insight.151869] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 01/28/2022] [Indexed: 01/26/2023] Open
Abstract
Molecular chaperones are responsible for maintaining cellular homeostasis, and one such chaperone, GRP170, is an endoplasmic reticulum (ER) resident that oversees both protein biogenesis and quality control. We previously discovered that GRP170 regulates the degradation and assembly of the epithelial sodium channel (ENaC), which reabsorbs sodium in the distal nephron and thereby regulates salt-water homeostasis and blood pressure. To define the role of GRP170 - and, more generally, molecular chaperones in kidney physiology - we developed an inducible, nephron-specific GRP170-KO mouse. Here, we show that GRP170 deficiency causes a dramatic phenotype: profound hypovolemia, hyperaldosteronemia, and dysregulation of ion homeostasis, all of which are associated with the loss of ENaC. Additionally, the GRP170-KO mouse exhibits hallmarks of acute kidney injury (AKI). We further demonstrate that the unfolded protein response (UPR) is activated in the GRP170-deficient mouse. Notably, the UPR is also activated in AKI when originating from various other etiologies, including ischemia, sepsis, glomerulonephritis, nephrotic syndrome, and transplant rejection. Our work establishes the central role of GRP170 in kidney homeostasis and directly links molecular chaperone function to kidney injury.
Collapse
Affiliation(s)
- Aidan W. Porter
- Department of Biological Sciences
- Department of Pediatrics, Nephrology Division
| | | | | | - Wily G. Ruiz
- Department of Medicine, Renal-Electrolyte Division
| | | | - Evan C. Ray
- Department of Medicine, Renal-Electrolyte Division
| | | | | | | | | | - Thomas R. Kleyman
- Department of Medicine, Renal-Electrolyte Division
- Department of Cell Biology, and
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | - Linda M. Hendershot
- Department of Tumor Cell Biology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | | | | |
Collapse
|
25
|
Zhang F, Zhou X, Zou H, Liu L, Li X, Ruan Y, Xie Y, Shi M, Xiao Y, Wang Y, Zhou Y, Wu Y, Guo B. SAA1 is transcriptionally activated by STAT3 and accelerates renal interstitial fibrosis by inducing endoplasmic reticulum stress. Exp Cell Res 2021; 408:112856. [PMID: 34597680 DOI: 10.1016/j.yexcr.2021.112856] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 09/23/2021] [Accepted: 09/24/2021] [Indexed: 02/07/2023]
Abstract
Renal interstitial fibrosis (RIF) is the common irreversible pathway by which chronic kidney disease (CKD) progresses to the end stage. The transforming growth factor-β (TGF-β)/signal transducer and activator of transcription 3 (STAT3) signaling pathway is a common factor leading to inflammation-mediated RIF, but its downstream regulatory mechanism is still unclear. Bioinformatics analysis predicted that serum amyloid A protein 1 (SAA1) was one of the target genes for transcriptional activation of STAT3 signaling. As an acute phase reaction protein, SAA1 plays an important role in many inflammatory reactions, and research has suggested that SAA1 is significantly elevated in the serum of patients with CKD. In this research, multiple experiments were performed to investigate the role of SAA1 in the process of RIF. SAA1 was abnormally highly expressed in kidney tissue from individuals who underwent unilateral ureteral obstruction (UUO) and TGF-β-induced HK2 cells, and the abnormal expression was directly related to the transcriptional activation of STAT3. Additionally, SAA1 can directly target and bind valosin-containing protein (VCP)-interacting membrane selenoprotein (VIMP) to inhibit the function of the Derlin-1/VCP/VIMP complex, preventing the transportation and degradation of the misfolded protein, resulting in endoplasmic reticulum (ER) stress characterized by an increase in glucose-regulated protein 78 (GRP78) levels and ultimately promoting the occurrence and development of RIF.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, 550025, China
| | - Xingcheng Zhou
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, 550025, China
| | - Huimei Zou
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, 550025, China; School of Nursing, Guizhou Medical University, Guiyang, 550025, China
| | - Lirong Liu
- Center for Clinical Laboratories, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550025, China; Department of Clinical Hematology, School of Clinical Laboratory Science, Guizhou Medical University, Guiyang, 550025, China
| | - Xiaoying Li
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, 550025, China; Department of Nephrology, Guiyang First People's Hospital, Guiyang, 550025, China
| | - Yuanyuan Ruan
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, 550025, China
| | - Ying Xie
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, 550025, China
| | - Mingjun Shi
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, 550025, China
| | - Ying Xiao
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, 550025, China
| | - Yuanyuan Wang
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, 550025, China
| | - Yuxia Zhou
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, 550025, China
| | - Yuansheng Wu
- Guangdong Provincial Hospital of Chinese Medicine, The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China.
| | - Bing Guo
- Department of Pathophysiology, Guizhou Medical University, Guiyang, 550025, China; Guizhou Provincial Key Laboratory of Pathogenesis and Drug Research on Common Chronic Diseases, Guizhou Medical University, Guiyang, 550025, China.
| |
Collapse
|
26
|
Maimaris G, Christodoulou A, Santama N, Lederer CW. Regulation of ER Composition and Extent, and Putative Action in Protein Networks by ER/NE Protein TMEM147. Int J Mol Sci 2021; 22:10231. [PMID: 34638576 PMCID: PMC8508377 DOI: 10.3390/ijms221910231] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 09/17/2021] [Accepted: 09/19/2021] [Indexed: 01/03/2023] Open
Abstract
Nuclear envelope (NE) and endoplasmic reticulum (ER) collaborate to control a multitude of nuclear and cytoplasmic actions. In this context, the transmembrane protein TMEM147 localizes to both NE and ER, and through direct and indirect interactions regulates processes as varied as production and transport of multipass membrane proteins, neuronal signaling, nuclear-shape, lamina and chromatin dynamics and cholesterol synthesis. Aiming to delineate the emerging multifunctionality of TMEM147 more comprehensively, we set as objectives, first, to assess potentially more fundamental effects of TMEM147 on the ER and, second, to identify significantly TMEM147-associated cell-wide protein networks and pathways. Quantifying curved and flat ER markers RTN4 and CLIMP63/CKAP4, respectively, we found that TMEM147 silencing causes area and intensity increases for both RTN4 and CLIMP63, and the ER in general, with a profound shift toward flat areas, concurrent with reduction in DNA condensation. Protein network and pathway analyses based on comprehensive compilation of TMEM147 interactors, targets and co-factors then served to manifest novel and established roles for TMEM147. Thus, algorithmically simplified significant pathways reflect TMEM147 function in ribosome binding, oxidoreductase activity, G protein-coupled receptor activity and transmembrane transport, while analysis of protein factors and networks identifies hub proteins and corresponding pathways as potential targets of TMEM147 action and of future functional studies.
Collapse
Affiliation(s)
- Giannis Maimaris
- Department of Biological Sciences, University of Cyprus, Nicosia 1678, Cyprus; (G.M.); (A.C.); (N.S.)
| | - Andri Christodoulou
- Department of Biological Sciences, University of Cyprus, Nicosia 1678, Cyprus; (G.M.); (A.C.); (N.S.)
| | - Niovi Santama
- Department of Biological Sciences, University of Cyprus, Nicosia 1678, Cyprus; (G.M.); (A.C.); (N.S.)
| | - Carsten Werner Lederer
- Department of Molecular Genetics Thalassaemia, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus
| |
Collapse
|
27
|
Garcia-Pardo ME, Simpson JC, O'Sullivan NC. A novel automated image analysis pipeline for quantifying morphological changes to the endoplasmic reticulum in cultured human cells. BMC Bioinformatics 2021; 22:427. [PMID: 34496765 PMCID: PMC8425006 DOI: 10.1186/s12859-021-04334-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 08/24/2021] [Indexed: 11/10/2022] Open
Abstract
Background In mammalian cells the endoplasmic reticulum (ER) comprises a highly complex reticular morphology that is spread throughout the cytoplasm. This organelle is of particular interest to biologists, as its dysfunction is associated with numerous diseases, which often manifest themselves as changes to the structure and organisation of the reticular network. Due to its complex morphology, image analysis methods to quantitatively describe this organelle, and importantly any changes to it, are lacking. Results In this work we detail a methodological approach that utilises automated high-content screening microscopy to capture images of cells fluorescently-labelled for various ER markers, followed by their quantitative analysis. We propose that two key metrics, namely the area of dense ER and the area of polygonal regions in between the reticular elements, together provide a basis for measuring the quantities of rough and smooth ER, respectively. We demonstrate that a number of different pharmacological perturbations to the ER can be quantitatively measured and compared in our automated image analysis pipeline. Furthermore, we show that this method can be implemented in both commercial and open-access image analysis software with comparable results. Conclusions We propose that this method has the potential to be applied in the context of large-scale genetic and chemical perturbations to assess the organisation of the ER in adherent cell cultures. Supplementary Information The online version contains supplementary material available at 10.1186/s12859-021-04334-x.
Collapse
Affiliation(s)
- M Elena Garcia-Pardo
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin 4, Ireland
| | - Jeremy C Simpson
- Cell Screening Laboratory, UCD School of Biology and Environmental Science, University College Dublin, Dublin 4, Ireland
| | - Niamh C O'Sullivan
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute, University College Dublin, Dublin 4, Ireland.
| |
Collapse
|
28
|
Perkins HT, Allan VJ, Waigh TA. Network organisation and the dynamics of tubules in the endoplasmic reticulum. Sci Rep 2021; 11:16230. [PMID: 34376706 PMCID: PMC8355327 DOI: 10.1038/s41598-021-94901-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 06/27/2021] [Indexed: 02/07/2023] Open
Abstract
The endoplasmic reticulum (ER) is a eukaryotic subcellular organelle composed of tubules and sheet-like areas of membrane connected at junctions. The tubule network is highly dynamic and undergoes rapid and continual rearrangement. There are currently few tools to evaluate network organisation and dynamics. We quantified ER network organisation in Vero and MRC5 cells, and developed an analysis workflow for dynamics of established tubules in live cells. The persistence length, tubule length, junction coordination number and angles of the network were quantified. Hallmarks of imbalances in ER tension, indications of interactions with microtubules and other subcellular organelles, and active dynamics were observed. Clear differences in dynamic behaviour were observed for established tubules at different positions within the cell using itemset mining. We found that tubules with activity-driven fluctuations were more likely to be located away from the cell periphery and a population of peripheral tubules with no signs of active motion was found.
Collapse
Affiliation(s)
- Hannah T Perkins
- Biological Physics, Department of Physics and Astronomy, Schuster Building, The University of Manchester, Oxford Road, Manchester, M13 9PL, UK
- Division of Molecular and Cellular Function, School of Biological Sciences, Michael Smith Building, The University of Manchester, Dover Street, Manchester, M13 9PT, UK
| | - Victoria J Allan
- Division of Molecular and Cellular Function, School of Biological Sciences, Michael Smith Building, The University of Manchester, Dover Street, Manchester, M13 9PT, UK.
| | - Thomas A Waigh
- Biological Physics, Department of Physics and Astronomy, Schuster Building, The University of Manchester, Oxford Road, Manchester, M13 9PL, UK.
| |
Collapse
|
29
|
Pradhan LK, Das SK. The Regulatory Role of Reticulons in Neurodegeneration: Insights Underpinning Therapeutic Potential for Neurodegenerative Diseases. Cell Mol Neurobiol 2021; 41:1157-1174. [PMID: 32504327 PMCID: PMC11448699 DOI: 10.1007/s10571-020-00893-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 05/29/2020] [Indexed: 02/06/2023]
Abstract
In the last few decades, cytoplasmic organellar dysfunction, such as that of the endoplasmic reticulum (ER), has created a new area of research interest towards the development of serious health maladies including neurodegenerative diseases. In this context, the extensively dispersed family of ER-localized proteins, i.e. reticulons (RTNs), is gaining interest because of its regulative control over neural regeneration. As most neurodegenerative diseases are pathologically manifested with the accretion of misfolded proteins with subsequent induction of ER stress, the regulatory role of RTNs in neural dysfunction cannot be ignored. With the limited information available in the literature, delineation of the functional connection between rising consequences of neurodegenerative diseases and RTNs need to be elucidated. In this review, we provide a broad overview on the recently revealed regulatory roles of reticulons in the pathophysiology of several health maladies, with special emphasis on neurodegeneration. Additionally, we have also recapitulated the decisive role of RTN4 in neurite regeneration and highlighted how neurodegeneration and proteinopathies are mechanistically linked with each other through specific RTN paralogues. With the recent findings advocating zebrafish Rtn4b (a mammalian Nogo-A homologue) downregulation following central nervous system (CNS) lesion, RTNs provides new insight into the CNS regeneration. However, there are controversies with respect to the role of Rtn4b in zebrafish CNS regeneration. Given these controversies, the connection between the unique regenerative capabilities of zebrafish CNS by distinct compensatory mechanisms and Rtn4b signalling pathway could shed light on the development of new therapeutic strategies against serious neurodegenerative diseases.
Collapse
Affiliation(s)
- Lilesh Kumar Pradhan
- Neurobiology Laboratory, Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed To Be University), Kalinga Nagar, Bhubaneswar, 751003, India
| | - Saroj Kumar Das
- Neurobiology Laboratory, Centre for Biotechnology, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan (Deemed To Be University), Kalinga Nagar, Bhubaneswar, 751003, India.
| |
Collapse
|
30
|
Morphological Heterogeneity of the Endoplasmic Reticulum within Neurons and Its Implications in Neurodegeneration. Cells 2021; 10:cells10050970. [PMID: 33919188 PMCID: PMC8143122 DOI: 10.3390/cells10050970] [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: 03/12/2021] [Revised: 04/13/2021] [Accepted: 04/19/2021] [Indexed: 12/19/2022] Open
Abstract
The endoplasmic reticulum (ER) is a multipurpose organelle comprising dynamic structural subdomains, such as ER sheets and tubules, serving to maintain protein, calcium, and lipid homeostasis. In neurons, the single ER is compartmentalized with a careful segregation of the structural subdomains in somatic and neurite (axodendritic) regions. The distribution and arrangement of these ER subdomains varies between different neuronal types. Mutations in ER membrane shaping proteins and morphological changes in the ER are associated with various neurodegenerative diseases implying significance of ER morphology in maintaining neuronal integrity. Specific neurons, such as the highly arborized dopaminergic neurons, are prone to stress and neurodegeneration. Differences in morphology and functionality of ER between the neurons may account for their varied sensitivity to stress and neurodegenerative changes. In this review, we explore the neuronal ER and discuss its distinct morphological attributes and specific functions. We hypothesize that morphological heterogeneity of the ER in neurons is an important factor that accounts for their selective susceptibility to neurodegeneration.
Collapse
|
31
|
Cremer T, Neefjes J, Berlin I. The journey of Ca 2+ through the cell - pulsing through the network of ER membrane contact sites. J Cell Sci 2020; 133:133/24/jcs249136. [PMID: 33376155 DOI: 10.1242/jcs.249136] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Calcium is the third most abundant metal on earth, and the fundaments of its homeostasis date back to pre-eukaryotic life forms. In higher organisms, Ca2+ serves as a cofactor for a wide array of (enzymatic) interactions in diverse cellular contexts and constitutes the most important signaling entity in excitable cells. To enable responsive behavior, cytosolic Ca2+ concentrations are kept low through sequestration into organellar stores, particularly the endoplasmic reticulum (ER), but also mitochondria and lysosomes. Specific triggers are then used to instigate a local release of Ca2+ on demand. Here, communication between organelles comes into play, which is accomplished through intimate yet dynamic contacts, termed membrane contact sites (MCSs). The field of MCS biology in relation to cellular Ca2+ homeostasis has exploded in recent years. Taking advantage of this new wealth of knowledge, in this Review, we invite the reader on a journey of Ca2+ flux through the ER and its associated MCSs. New mechanistic insights and technological advances inform the narrative on Ca2+ acquisition and mobilization at these sites of communication between organelles, and guide the discussion of their consequences for cellular physiology.
Collapse
Affiliation(s)
- Tom Cremer
- Department of Cell and Chemical Biology, Leiden University Medical Center LUMC, Einthovenweg 20, 2300RC Leiden, The Netherlands
| | - Jacques Neefjes
- Department of Cell and Chemical Biology, Leiden University Medical Center LUMC, Einthovenweg 20, 2300RC Leiden, The Netherlands
| | - Ilana Berlin
- Department of Cell and Chemical Biology, Leiden University Medical Center LUMC, Einthovenweg 20, 2300RC Leiden, The Netherlands
| |
Collapse
|
32
|
Long RKM, Moriarty KP, Cardoen B, Gao G, Vogl AW, Jean F, Hamarneh G, Nabi IR. Super resolution microscopy and deep learning identify Zika virus reorganization of the endoplasmic reticulum. Sci Rep 2020; 10:20937. [PMID: 33262363 PMCID: PMC7708840 DOI: 10.1038/s41598-020-77170-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/28/2020] [Indexed: 01/08/2023] Open
Abstract
The endoplasmic reticulum (ER) is a complex subcellular organelle composed of diverse structures such as tubules, sheets and tubular matrices. Flaviviruses such as Zika virus (ZIKV) induce reorganization of ER membranes to facilitate viral replication. Here, using 3D super resolution microscopy, ZIKV infection is shown to induce the formation of dense tubular matrices associated with viral replication in the central ER. Viral non-structural proteins NS4B and NS2B associate with replication complexes within the ZIKV-induced tubular matrix and exhibit distinct ER distributions outside this central ER region. Deep neural networks trained to distinguish ZIKV-infected versus mock-infected cells successfully identified ZIKV-induced central ER tubular matrices as a determinant of viral infection. Super resolution microscopy and deep learning are therefore able to identify and localize morphological features of the ER and allow for better understanding of how ER morphology changes due to viral infection.
Collapse
Affiliation(s)
- Rory K M Long
- Life Sciences Institute, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.,Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.,Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - Kathleen P Moriarty
- School of Computing Science, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Ben Cardoen
- School of Computing Science, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada
| | - Guang Gao
- Life Sciences Institute, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.,Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - A Wayne Vogl
- Life Sciences Institute, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.,Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada
| | - François Jean
- Life Sciences Institute, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada. .,Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| | - Ghassan Hamarneh
- School of Computing Science, Simon Fraser University, Burnaby, BC, V5A 1S6, Canada.
| | - Ivan R Nabi
- Life Sciences Institute, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada. .,Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada. .,School of Biomedical Engineering, University of British Columbia, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
33
|
Detection of immunogenic cell death and its relevance for cancer therapy. Cell Death Dis 2020; 11:1013. [PMID: 33243969 PMCID: PMC7691519 DOI: 10.1038/s41419-020-03221-2] [Citation(s) in RCA: 617] [Impact Index Per Article: 123.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 02/07/2023]
Abstract
Chemotherapy, radiation therapy, as well as targeted anticancer agents can induce clinically relevant tumor-targeting immune responses, which critically rely on the antigenicity of malignant cells and their capacity to generate adjuvant signals. In particular, immunogenic cell death (ICD) is accompanied by the exposure and release of numerous damage-associated molecular patterns (DAMPs), which altogether confer a robust adjuvanticity to dying cancer cells, as they favor the recruitment and activation of antigen-presenting cells. ICD-associated DAMPs include surface-exposed calreticulin (CALR) as well as secreted ATP, annexin A1 (ANXA1), type I interferon, and high-mobility group box 1 (HMGB1). Additional hallmarks of ICD encompass the phosphorylation of eukaryotic translation initiation factor 2 subunit-α (EIF2S1, better known as eIF2α), the activation of autophagy, and a global arrest in transcription and translation. Here, we outline methodological approaches for measuring ICD markers in vitro and ex vivo for the discovery of next-generation antineoplastic agents, the development of personalized anticancer regimens, and the identification of optimal therapeutic combinations for the clinical management of cancer.
Collapse
|
34
|
Cao Z, Wang X, Huang X, Mak HY. Are endoplasmic reticulum subdomains shaped by asymmetric distribution of phospholipids? Evidence from a C. elegans model system. Bioessays 2020; 43:e2000199. [PMID: 33169432 DOI: 10.1002/bies.202000199] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/13/2020] [Accepted: 09/18/2020] [Indexed: 12/19/2022]
Abstract
Physical contact between organelles are widespread, in part to facilitate the shuttling of protein and lipid cargoes for cellular homeostasis. How do protein-protein and protein-lipid interactions shape organelle subdomains that constitute contact sites? The endoplasmic reticulum (ER) forms extensive contacts with multiple organelles, including lipid droplets (LDs) that are central to cellular fat storage and mobilization. Here, we focus on ER-LD contacts that are highlighted by the conserved protein seipin, which promotes LD biogenesis and expansion. Seipin is enriched in ER tubules that form cage-like structures around a subset of LDs. Such enrichment is strongly dependent on polyunsaturated and cyclopropane fatty acids. Based on these findings, we speculate on molecular events that lead to the formation of seipin-positive peri-LD cages in which protein movement is restricted. We hypothesize that asymmetric distribution of specific phospholipids distinguishes cage membrane tubules from the bulk ER.
Collapse
Affiliation(s)
- Zhe Cao
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Xiaowei Wang
- Department of Chemistry, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Xuhui Huang
- Department of Chemistry, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| | - Ho Yi Mak
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong SAR, China
| |
Collapse
|
35
|
Zhao J, Hu J. Self-Association of Purified Reconstituted ER Luminal Spacer Climp63. Front Cell Dev Biol 2020; 8:500. [PMID: 32612999 PMCID: PMC7308479 DOI: 10.3389/fcell.2020.00500] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 05/25/2020] [Indexed: 01/15/2023] Open
Abstract
Membranes of the endoplasmic reticulum (ER) are shaped into cisternal sheets and cylindrical tubules. How ER sheets are generated and maintained is not clear. ER membrane protein Climp63 is enriched in sheets and routinely used as a marker of this structure. The luminal domain (LD) of Climp63 is predicted to be highly helical, and it may form bridges between parallel membranes, regulating the abundance and width of ER sheets. Here, we purified the LD and full-length (FL) Climp63 to analyze their homotypic interactions. The N-terminal tagged LD formed low-order oligomers in solution, but was extremely aggregation-prone when the GST tag was removed. Purified FL Climp63 formed detectable but moderate interactions with both the FL protein and the LD. When Climp63 was reconstituted into proteoliposomes with its LD facing out, the homotypic interactions were retained and could be competed by soluble LD, though vesicle clustering was not observed. These results demonstrate a direct self-association of Climp63, supporting its role as an ER luminal spacer.
Collapse
Affiliation(s)
- Jinghua Zhao
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Junjie Hu
- Department of Genetics and Cell Biology, College of Life Sciences, Nankai University, Tianjin, China.,National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
36
|
Alam SMD, Tsukamoto Y, Ogawa M, Senoo Y, Ikeda K, Tashima Y, Takeuchi H, Okajima T. N-Glycans on EGF domain-specific O-GlcNAc transferase (EOGT) facilitate EOGT maturation and peripheral endoplasmic reticulum localization. J Biol Chem 2020; 295:8560-8574. [PMID: 32376684 DOI: 10.1074/jbc.ra119.012280] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 04/30/2020] [Indexed: 02/03/2023] Open
Abstract
Epidermal growth factor (EGF) domain-specific O-GlcNAc transferase (EOGT) is an endoplasmic reticulum (ER)-resident protein that modifies EGF repeats of Notch receptors and thereby regulates Delta-like ligand-mediated Notch signaling. Several EOGT mutations that may affect putative N-glycosylation consensus sites are recorded in the cancer database, but the presence and function of N-glycans in EOGT have not yet been characterized. Here, we identified N-glycosylation sites in mouse EOGT and elucidated their molecular functions. Three predicted N-glycosylation consensus sequences on EOGT are highly conserved among mammalian species. Within these sites, we found that Asn-263 and Asn-354, but not Asn-493, are modified with N-glycans. Lectin blotting, endoglycosidase H digestion, and MS analysis revealed that both residues are modified with oligomannose N-glycans. Loss of an individual N-glycan on EOGT did not affect its endoplasmic reticulum (ER) localization, enzyme activity, and ability to O-GlcNAcylate Notch1 in HEK293T cells. However, simultaneous substitution of both N-glycosylation sites affected both EOGT maturation and expression levels without an apparent change in enzymatic activity, suggesting that N-glycosylation at a single site is sufficient for EOGT maturation and expression. Accordingly, a decrease in O-GlcNAc stoichiometry was observed in Notch1 co-expressed with an N263Q/N354Q variant compared with WT EOGT. Moreover, the N263Q/N354Q variant exhibited altered subcellular distribution within the ER in HEK293T cells, indicating that N-glycosylation of EOGT is required for its ER localization at the cell periphery. These results suggest critical roles of N-glycans in sustaining O-GlcNAc transferase function both by maintaining EOGT levels and by ensuring its proper subcellular localization in the ER.
Collapse
Affiliation(s)
- Sayad Md Didarul Alam
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yohei Tsukamoto
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Mitsutaka Ogawa
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yuya Senoo
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazutaka Ikeda
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan.,RIKEN, Center for Integrative Medical Sciences, Suehiro-cho, Tsurumi, Yokohama, Japan
| | - Yuko Tashima
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hideyuki Takeuchi
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tetsuya Okajima
- Department of Molecular Biochemistry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
37
|
Li X, Yang L, Mao Z, Pan X, Zhao Y, Gu X, Eckel-Mahan K, Zuo Z, Tong Q, Hartig SM, Cheng X, Du G, Moore DD, Bellen HJ, Sesaki H, Sun K. Novel role of dynamin-related-protein 1 in dynamics of ER-lipid droplets in adipose tissue. FASEB J 2020; 34:8265-8282. [PMID: 32294302 DOI: 10.1096/fj.201903100rr] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 04/06/2020] [Accepted: 04/06/2020] [Indexed: 12/17/2022]
Abstract
Dynamin-Related-Protein 1 (DRP1) critically regulates mitochondrial and peroxisomal fission in multicellular organisms. However, the impact of DRP1 on other organelles, especially its direct influence on ER functions remains largely unclear. Here, we report that DRP1 translocates to endoplasmic reticulum (ER) in response to β-adrenergic stimulation. To further investigate the function of DRP1 on ER-lipid droplet (LD) dynamics and the metabolic subsequences, we generated an adipose tissue-specific DRP1 knockout model (Adipo-Drp1flx/flx ). We found that the LDs in adipose tissues of Adipo-Drp1flx/flx mice exhibited more unilocular morphology with larger sizes, and formed less multilocular structures upon cold exposure. Mechanistically, we discovered that abnormal LD morphology occurs because newly generated micro-LDs fail to dissociate from the ER due to DRP1 ablation. Conversely, the ER retention of LDs can be rescued by the overexpressed DRP1 in the adipocytes. The alteration of LD dynamics, combined with abnormal mitochondrial and autophagy functions in adipose tissue, ultimately lead to abnormalities in lipid metabolism in Adipo-Drp1flx/flx mice.
Collapse
Affiliation(s)
- Xin Li
- Center for Metabolic and Degenerative Diseases, the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Li Yang
- Center for Metabolic and Degenerative Diseases, the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Zhengmei Mao
- Microscopy Core, the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Xueyang Pan
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, MN, USA
| | - Yueshui Zhao
- Center for Metabolic and Degenerative Diseases, the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Xue Gu
- Center for Metabolic and Degenerative Diseases, the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Kristin Eckel-Mahan
- Center for Metabolic and Degenerative Diseases, the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA.,Department of Integrative Biology and Pharmacology, Graduate Program in Cell and Regulatory Biology, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Zhongyuan Zuo
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.,Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA.,Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA.,Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Qiang Tong
- Children's Nutrition Research Center, Baylor College of Medicine, Houston, TX, USA
| | - Sean M Hartig
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Xiaodong Cheng
- Department of Integrative Biology and Pharmacology, Graduate Program in Cell and Regulatory Biology, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA.,Texas Therapeutics Institute, the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Guangwei Du
- Department of Integrative Biology and Pharmacology, Graduate Program in Cell and Regulatory Biology, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - David D Moore
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX, USA.,Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX, USA.,Program in Developmental Biology, Baylor College of Medicine, Houston, TX, USA.,Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Hiromi Sesaki
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kai Sun
- Center for Metabolic and Degenerative Diseases, the Brown Foundation Institute of Molecular Medicine for the Prevention of Human Diseases, University of Texas Health Science Center at Houston, Houston, TX, USA.,Department of Integrative Biology and Pharmacology, Graduate Program in Cell and Regulatory Biology, Graduate School of Biomedical Sciences, University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|