1
|
Cho H, Huh KM, Shim MS, Cho YY, Lee JY, Lee HS, Kwon YJ, Kang HC. Selective delivery of imaging probes and therapeutics to the endoplasmic reticulum or Golgi apparatus: Current strategies and beyond. Adv Drug Deliv Rev 2024; 212:115386. [PMID: 38971180 DOI: 10.1016/j.addr.2024.115386] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 06/14/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
To maximize therapeutic effects and minimize unwanted effects, the interest in drug targeting to the endoplasmic reticulum (ER) or Golgi apparatus (GA) has been recently growing because two organelles are distributing hubs of cellular building/signaling components (e.g., proteins, lipids, Ca2+) to other organelles and the plasma membrane. Their structural or functional damages induce organelle stress (i.e., ER or GA stress), and their aggravation is strongly related to diseases (e.g., cancers, liver diseases, brain diseases). Many efforts have been developed to image (patho)physiological functions (e.g., oxidative stress, protein/lipid-related processing) and characteristics (e.g., pH, temperature, biothiols, reactive oxygen species) in the target organelles and to deliver drugs for organelle disruption using organelle-targeting moieties. Therefore, this review will overview the structure, (patho)physiological functions/characteristics, and related diseases of the organelles of interest. Future direction on ER or GA targeting will be discussed by understanding current strategies and investigations on targeting, imaging/sensing, and therapeutic systems.
Collapse
Affiliation(s)
- Hana Cho
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Kang Moo Huh
- Departments of Polymer Science and Engineering & Materials Science and Engineering, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Min Suk Shim
- Division of Bioengineering, Incheon National University, Incheon 22012, Republic of Korea
| | - Yong-Yeon Cho
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Republic of Korea; Research Institute for Controls and Materials of Regulated Cell Death, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Joo Young Lee
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Republic of Korea; Research Institute for Controls and Materials of Regulated Cell Death, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Hye Suk Lee
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Republic of Korea; Research Institute for Controls and Materials of Regulated Cell Death, The Catholic University of Korea, Bucheon 14662, Republic of Korea
| | - Young Jik Kwon
- Department of Pharmaceutical Sciences, University of California, Irvine, CA 92697, USA
| | - Han Chang Kang
- Department of Pharmacy, College of Pharmacy, The Catholic University of Korea, Bucheon 14662, Republic of Korea; Research Institute for Controls and Materials of Regulated Cell Death, The Catholic University of Korea, Bucheon 14662, Republic of Korea.
| |
Collapse
|
2
|
Zhu M, Fang Z, Wu Y, Dong F, Wang Y, Zheng F, Ma X, Ma S, He J, Liu X, Yao X, Fu C. A KDELR-mediated ER-retrieval system modulates mitochondrial functions via the unfolded protein response in fission yeast. J Biol Chem 2024; 300:105754. [PMID: 38360270 PMCID: PMC10938167 DOI: 10.1016/j.jbc.2024.105754] [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: 11/17/2023] [Revised: 01/23/2024] [Accepted: 02/08/2024] [Indexed: 02/17/2024] Open
Abstract
KDELR (Erd2 [ER retention defective 2] in yeasts) is a receptor protein that retrieves endoplasmic reticulum (ER)-resident proteins from the Golgi apparatus. However, the role of the KDELR-mediated ER-retrieval system in regulating cellular homeostasis remains elusive. Here, we show that the absence of Erd2 triggers the unfolded protein response (UPR) and enhances mitochondrial respiration and reactive oxygen species in an UPR-dependent manner in the fission yeast Schizosaccharomyces pombe. Moreover, we perform transcriptomic analysis and find that the expression of genes related to mitochondrial respiration and the tricarboxylic acid cycle is upregulated in a UPR-dependent manner in cells lacking Erd2. The increased mitochondrial respiration and reactive oxygen species production is required for cell survival in the absence of Erd2. Therefore, our findings reveal a novel role of the KDELR-Erd2-mediated ER-retrieval system in modulating mitochondrial functions and highlight its importance for cellular homeostasis in the fission yeast.
Collapse
Affiliation(s)
- Mengdan Zhu
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Zheng Fang
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Yifan Wu
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Fenfen Dong
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Yuzhou Wang
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Fan Zheng
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Xiaopeng Ma
- Division of Life Sciences and Medicine, Department of General Surgery, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Shisong Ma
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Jiajia He
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Xing Liu
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Xuebiao Yao
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China.
| | - Chuanhai Fu
- MOE Key Laboratory for Cellular Dynamics & Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China; Anhui Key Laboratory of Cellular Dynamics and Chemical Biology & Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
3
|
Jia J, Zhu L, Yue X, Tang S, Jing S, Tan C, Du Y, Gao J, Lee I, Qian Y. Crosstalk between KDEL receptor and EGF receptor mediates cell proliferation and migration via STAT3 signaling. Cell Commun Signal 2024; 22:140. [PMID: 38378560 PMCID: PMC10880305 DOI: 10.1186/s12964-024-01517-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 02/07/2024] [Indexed: 02/22/2024] Open
Abstract
Hostile microenvironment of cancer cells provoke a stressful condition for endoplasmic reticulum (ER) and stimulate the expression and secretion of ER chaperones, leading to tumorigenic effects. However, the molecular mechanism underlying these effects is largely unknown. In this study, we reveal that the last four residues of ER chaperones, which are recognized by KDEL receptor (KDELR), is required for cell proliferation and migration induced by secreted chaperones. By combining proximity-based mass spectrometry analysis, split venus imaging and membrane yeast two hybrid assay, we present that EGF receptor (EGFR) may be a co-receptor for KDELR on the surface. Prior to ligand addition, KDELR spontaneously oligomerizes and constantly undergoes recycling near the plasma membrane. Upon KDEL ligand binding, the interactions of KDELR with itself and with EGFR increase rapidly, leading to augmented internalization of KDELR and tyrosine phosphorylation in the C-terminus of EGFR. STAT3, which binds the phosphorylated tyrosine motif on EGFR, is subsequently activated by EGFR and mediates cell growth and migration. Taken together, our results suggest that KDELR serves as a bona fide cell surface receptor for secreted ER chaperones and transactivates EGFR-STAT3 signaling pathway.
Collapse
Affiliation(s)
- Jie Jia
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Lianhui Zhu
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Xihua Yue
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Shuocheng Tang
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Shuaiyang Jing
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
- Present address: Department of Pharmacology, School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Chuanting Tan
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Yulei Du
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Jingkai Gao
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Intaek Lee
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China.
| | - Yi Qian
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China.
| |
Collapse
|
4
|
Jia J, Tang S, Yue X, Jing S, Zhu L, Tan C, Gao J, Du Y, Lee I, Qian Y. An A-Kinase Anchoring Protein (ACBD3) Coordinates Traffic-Induced PKA Activation At The Golgi. J Biol Chem 2023; 299:104696. [PMID: 37044218 DOI: 10.1016/j.jbc.2023.104696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 03/23/2023] [Accepted: 03/25/2023] [Indexed: 04/14/2023] Open
Abstract
KDEL receptor (KDELR) is a key protein that recycles escaped ER resident proteins from the Golgi apparatus back to the ER and maintains a dynamic balance between these two organelles in the early secretory pathway. Studies have shown that this retrograde transport pathway is partly regulated by two KDELR-interacting proteins, Acyl-CoA-binding domain-containing 3 (ACBD3), and cyclic AMP-dependent protein kinase A (PKA). However, whether Golgi-localized ACBD3, which was first discovered as a PKA-anchoring protein in mitochondria, directly interacts with PKA at the Golgi and coordinates its signaling in Golgi-to-ER traffic has remained unclear. In this study, we showed that the GOLD domain of ACBD3 directly interacts with the regulatory subunit II (RII) of PKA and effectively recruits PKA holoenzyme to the Golgi. Forward trafficking of proteins from the ER triggers activation of PKA by releasing the catalytic subunit from RII. Furthermore, we determined that depletion of ACBD3 reduces the Golgi fraction of RII, resulting in moderate, but constitutive activation of PKA and KDELR retrograde transport, independent of cargo influx from the ER. Taken together, these data demonstrate that ACBD3 coordinates the protein secretory pathway at the Golgi by facilitating KDELR/PKA-containing protein complex formation.
Collapse
Affiliation(s)
- Jie Jia
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Shuocheng Tang
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Xihua Yue
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Shuaiyang Jing
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Lianhui Zhu
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Chuanting Tan
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Jingkai Gao
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yulei Du
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China; University of Chinese Academy of Sciences, Beijing, China
| | - Intaek Lee
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China.
| | - Yi Qian
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China.
| |
Collapse
|
5
|
Tan C, Du Y, Zhu L, Jing S, Gao J, Qian Y, Yue X, Lee I. KDEL Receptor Trafficking to the Plasma Membrane Is Regulated by ACBD3 and Rab4A-GTP. Cells 2023; 12:cells12071079. [PMID: 37048152 PMCID: PMC10093020 DOI: 10.3390/cells12071079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/01/2023] [Accepted: 03/14/2023] [Indexed: 04/07/2023] Open
Abstract
KDEL receptor-1 maintains homeostasis in the early secretory pathway by capturing and retrieving ER chaperones to the ER during heavy secretory activity. Unexpectedly, a fraction of the receptor is also known to reside in the plasma membrane (PM), although it is largely unknown exactly how the KDEL receptor gets exported from the Golgi and travels to the PM. We have previously shown that a Golgi scaffolding protein (ACBD3) facilitates KDEL receptor localization at the Golgi via the regulating cargo wave-induced cAMP/PKA-dependent signaling pathway. Upon endocytosis, surface-expressed KDEL receptor undergoes highly complex itineraries through the Golgi and the endo-lysosomal compartments, where the endocytosed receptor utilizes Rab14A- and Rab11A-positive recycling endosomes and clathrin-decorated tubulovesicular carriers. In this study, we sought to investigate the mechanism through which the KDEL receptor gets exported from the Golgi en route to the PM. We report here that ACBD3 depletion results in greatly increased trafficking of KDEL receptor to the PM via Rab4A-positive tubular carriers emanating from the Golgi. Expression of constitutively activated Rab4A mutant (Q72L) increases the surface expression of KDEL receptor up to 2~3-fold, whereas Rab4A knockdown or the expression of GDP-locked Rab4A mutant (S27N) inhibits KDEL receptor targeting of the PM. Importantly, KDELR trafficking from the Golgi to the PM is independent of PKA- and Src kinase-mediated mechanisms. Taken together, these results reveal that ACBD3 and Rab4A play a key role in regulating KDEL receptor trafficking to the cell surface.
Collapse
Affiliation(s)
- Chuanting Tan
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Yulei Du
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Lianhui Zhu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Shuaiyang Jing
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Jingkai Gao
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Yi Qian
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xihua Yue
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Intaek Lee
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
6
|
Alvim JC, Bolt RM, An J, Kamisugi Y, Cuming A, Silva-Alvim FAL, Concha JO, daSilva LLP, Hu M, Hirsz D, Denecke J. The K/HDEL receptor does not recycle but instead acts as a Golgi-gatekeeper. Nat Commun 2023; 14:1612. [PMID: 36959220 PMCID: PMC10036638 DOI: 10.1038/s41467-023-37056-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 02/24/2023] [Indexed: 03/25/2023] Open
Abstract
Accurately measuring the ability of the K/HDEL receptor (ERD2) to retain the ER cargo Amy-HDEL has questioned earlier results on which the popular receptor recycling model is based upon. Here we demonstrate that ERD2 Golgi-retention, rather than fast ER export supports its function. Ligand-induced ERD2 redistribution is only observed when the C-terminus is masked or mutated, compromising the signal that prevents Golgi-to-ER transport of the receptor. Forcing COPI mediated retrograde transport destroys receptor function, but introducing ER-to-Golgi export or cis-Golgi retention signals re-activate ERD2 when its endogenous Golgi-retention signal is masked or deleted. We propose that ERD2 remains fixed as a Golgi gatekeeper, capturing K/HDEL proteins when they arrive and releasing them again into a subdomain for retrograde transport back to the ER. An in vivo ligand:receptor ratio far greater than 100 to 1 strongly supports this model, and the underlying mechanism appears to be extremely conserved across kingdoms.
Collapse
Affiliation(s)
- Jonas C Alvim
- Centre for Plant Sciences, School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
- Laboratory of Plant Physiology and Biophysics, Bower Building, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Robert M Bolt
- Centre for Plant Sciences, School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Jing An
- Centre for Plant Sciences, School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Yasuko Kamisugi
- Centre for Plant Sciences, School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Andrew Cuming
- Centre for Plant Sciences, School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Fernanda A L Silva-Alvim
- Centre for Plant Sciences, School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
- Laboratory of Plant Physiology and Biophysics, Bower Building, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Juan O Concha
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Luis L P daSilva
- Department of Cell and Molecular Biology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Meiyi Hu
- Centre for Plant Sciences, School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Dominique Hirsz
- Centre for Plant Sciences, School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK
| | - Jurgen Denecke
- Centre for Plant Sciences, School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, UK.
| |
Collapse
|
7
|
Yue X, Qian Y, Zhu L, Gim B, Bao M, Jia J, Jing S, Wang Y, Tan C, Bottanelli F, Ziltener P, Choi S, Hao P, Lee I. ACBD3 modulates KDEL receptor interaction with PKA for its trafficking via tubulovesicular carrier. BMC Biol 2021; 19:194. [PMID: 34493279 PMCID: PMC8424950 DOI: 10.1186/s12915-021-01137-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Accepted: 08/30/2021] [Indexed: 11/10/2022] Open
Abstract
Background KDEL receptor helps establish cellular equilibrium in the early secretory pathway by recycling leaked ER-chaperones to the ER during secretion of newly synthesized proteins. Studies have also shown that KDEL receptor may function as a signaling protein that orchestrates membrane flux through the secretory pathway. We have recently shown that KDEL receptor is also a cell surface receptor, which undergoes highly complex itinerary between trans-Golgi network and the plasma membranes via clathrin-mediated transport carriers. Ironically, however, it is still largely unknown how KDEL receptor is distributed to the Golgi at steady state, since its initial discovery in late 1980s. Results We used a proximity-based in vivo tagging strategy to further dissect mechanisms of KDEL receptor trafficking. Our new results reveal that ACBD3 may be a key protein that regulates KDEL receptor trafficking via modulation of Arf1-dependent tubule formation. We demonstrate that ACBD3 directly interact with KDEL receptor and form a functionally distinct protein complex in ArfGAPs-independent manner. Depletion of ACBD3 results in re-localization of KDEL receptor to the ER by inducing accelerated retrograde trafficking of KDEL receptor. Importantly, this is caused by specifically altering KDEL receptor interaction with Protein Kinase A and Arf1/ArfGAP1, eventually leading to increased Arf1-GTP-dependent tubular carrier formation at the Golgi. Conclusions These results suggest that ACBD3 may function as a negative regulator of PKA activity on KDEL receptor, thereby restricting its retrograde trafficking in the absence of KDEL ligand binding. Since ACBD3 was originally identified as PAP7, a PBR/PKA-interacting protein at the Golgi/mitochondria, we propose that Golgi-localization of KDEL receptor is likely to be controlled by its interaction with ACBD3/PKA complex at steady state, providing a novel insight for establishment of cellular homeostasis in the early secretory pathway. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01137-7.
Collapse
Affiliation(s)
- Xihua Yue
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Yi Qian
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Lianhui Zhu
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Bopil Gim
- School of Physical Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Mengjing Bao
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Jie Jia
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Shuaiyang Jing
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yijing Wang
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Chuanting Tan
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Francesca Bottanelli
- Institut für Biochemie, Freie Universität Berlin, Thielallee 63, 14195, Berlin, Germany
| | - Pascal Ziltener
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, USA
| | - Sunkyu Choi
- Proteomics Core, Weill Cornell Medicine-Qatar, Doha, Qatar
| | - Piliang Hao
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China
| | - Intaek Lee
- School of Life Science and Technology, ShanghaiTech University, Pudong, Shanghai, China. .,Shanghai Institute for Advanced Immunochemical Studies, Shanghai, China.
| |
Collapse
|
8
|
Blagotinšek Cokan K, Mavri M, Rutland CS, Glišić S, Senćanski M, Vrecl M, Kubale V. Critical Impact of Different Conserved Endoplasmic Retention Motifs and Dopamine Receptor Interacting Proteins (DRIPs) on Intracellular Localization and Trafficking of the D 2 Dopamine Receptor (D 2-R) Isoforms. Biomolecules 2020; 10:biom10101355. [PMID: 32977535 PMCID: PMC7598153 DOI: 10.3390/biom10101355] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/09/2020] [Accepted: 09/18/2020] [Indexed: 01/13/2023] Open
Abstract
The type 2 dopamine receptor D2 (D2-R), member of the G protein-coupled receptor (GPCR) superfamily, exists in two isoforms, short (D2S-R) and long (D2L-R). They differ by an additional 29 amino acids (AA) in the third cytoplasmic loop (ICL3) of the D2L-R. These isoforms differ in their intracellular localization and trafficking functionality, as D2L-R possesses a larger intracellular pool, mostly in the endoplasmic reticulum (ER). This review focuses on the evolutionarily conserved motifs in the ICL3 of the D2-R and proteins interacting with the ICL3 of both isoforms, specifically with the 29 AA insert. These motifs might be involved in D2-R exit from the ER and have an impact on cell-surface and intracellular localization and, therefore, also play a role in the function of dopamine receptor signaling, ligand binding and possible homo/heterodimerization. Our recent bioinformatic data on potential new interaction partners for the ICL3 of D2-Rs are also presented. Both are highly relevant, and have clinical impacts on the pathophysiology of several diseases such as Parkinson’s disease, schizophrenia, Tourette’s syndrome, Huntington’s disease, manic depression, and others, as they are connected to a variety of essential motifs and differences in communication with interaction partners.
Collapse
Affiliation(s)
- Kaja Blagotinšek Cokan
- Department of Anatomy, Histology with Embryology and Cytology, Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000 Ljubljana, Slovenia; (K.B.C.); (M.M.); (M.V.)
| | - Maša Mavri
- Department of Anatomy, Histology with Embryology and Cytology, Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000 Ljubljana, Slovenia; (K.B.C.); (M.M.); (M.V.)
| | - Catrin Sian Rutland
- School of Veterinary Medicine and Science, Medical Faculty, University of Nottingham, Sutton, Bonington Campus, Loughborough LE12 5RD, UK;
| | - Sanja Glišić
- Center for Multidisciplinary Research, Institute of Nuclear Sciences VINCA, University of Belgrade, Mike Petrovića Alasa 12-14, 11351 Vinča, Belgrade, Serbia; (S.G.); (M.S.)
| | - Milan Senćanski
- Center for Multidisciplinary Research, Institute of Nuclear Sciences VINCA, University of Belgrade, Mike Petrovića Alasa 12-14, 11351 Vinča, Belgrade, Serbia; (S.G.); (M.S.)
| | - Milka Vrecl
- Department of Anatomy, Histology with Embryology and Cytology, Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000 Ljubljana, Slovenia; (K.B.C.); (M.M.); (M.V.)
| | - Valentina Kubale
- Department of Anatomy, Histology with Embryology and Cytology, Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000 Ljubljana, Slovenia; (K.B.C.); (M.M.); (M.V.)
- Correspondence:
| |
Collapse
|
9
|
Mao H, Nian J, Wang Z, Li X, Huang C. KDELR2 is an unfavorable prognostic biomarker and regulates CCND1 to promote tumor progression in glioma. Pathol Res Pract 2020; 216:152996. [PMID: 32534703 DOI: 10.1016/j.prp.2020.152996] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/07/2020] [Accepted: 04/22/2020] [Indexed: 01/09/2023]
Abstract
BACKGROUND The KDEL receptor is a seven-transmembrane-domain protein, which plays a key role in ER quality control and in the ER stress response, KDELR2 involved in regulation of cellular functions, including cell proliferation, survival, promotes glioblastoma tumorigenesis. The aim of this study was to investigate the clinicpathological value and biological role of KDELR2 in glioma. METHODS We studied the expression of KEDLR2 and its association with the prognosis through the TCGA, CGGA, and GSE16011 database. To explore the role of KDELR2 in glioma, KDELR2 siRNA was constructed and transfected into U87 glioma cells. CCK-8, colony formation and Transwell assays were used to investigate the roles of KDELR2 on GBM cell proliferation. We further studied the effect of KDELR2 on tumorigenesis in animal model. Additionally, flow cytometry was used to monitor the changes in the cell cycle and apoptosis following transfection with KDELR2 siRNA. We applied GeneChip primeview expression array to analysis the differential gene expression profiling. Ingenuity Pathway Analysis to show that KDELR2 has a significant impact in canonical pathway in cell cycle regulation and participate in multiple pathways. And we detected the cell cycle proteins CCND1 expression by Western blot analysis. RESULTS Our results showed that KDELR2 was up-regulated in glioma tissue and cell lines. Knockdown KDELR2 was able to reduce cell viability, promote cell cycle arrest at the G1 phase, and induce apoptotic cell death. Moreover, our results suggested that KDELR2 regulated the cellular functions of U87 cells by targeting CCND1. Therefore, we demonstrated that KDELR2 is a novel biomarker in glioma. CONCLUSIONS KDELR2 is highly expressed in human glioma tissues and cell lines, a higher expression of KDELR2 is associated with a poor prognosis of glioma patients. Moreover, KDELR2 regulated the cellular functions of U87 cells by targeting CCND1. The KDELR2/CCND1 axis may provide a new therapeutic target for the treatment of glioma and deepen our understanding of glioma mechanisms.
Collapse
Affiliation(s)
- Hui Mao
- Department of Neurosurgery, First Affiliated Hospital of Jishou University, Jishou 416000, Hunan, China
| | - Jiang Nian
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zhao Wang
- Department of Neurosurgery, First Affiliated Hospital of Jishou University, Jishou 416000, Hunan, China
| | - XueJun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - ChunHai Huang
- Department of Neurosurgery, First Affiliated Hospital of Jishou University, Jishou 416000, Hunan, China.
| |
Collapse
|
10
|
Khan ES, Sankaran S, Llontop L, Del Campo A. Exogenous supply of Hsp47 triggers fibrillar collagen deposition in skin cell cultures in vitro. BMC Mol Cell Biol 2020; 21:22. [PMID: 32228452 PMCID: PMC7106624 DOI: 10.1186/s12860-020-00267-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 03/20/2020] [Indexed: 12/17/2022] Open
Abstract
Background Collagen is a structural protein that provides mechanical stability and defined architectures to skin. In collagen-based skin disorders this stability is lost, either due to mutations in collagens or in the chaperones involved in collagen assembly. This leads to chronic wounds, skin fragility, and blistering. Existing approaches to treat such conditions rely on administration of small molecules to simulate collagen production, like 4-phenylbutyrate (4-PBA) or growth factors like TGF-β. However, these molecules are not specific for collagen synthesis, and result in unsolicited side effects. Hsp47 is a collagen-specific chaperone with a major role in collagen biosynthesis. Expression levels of Hsp47 correlate with collagen deposition. This article explores the stimulation of collagen deposition by exogenously supplied Hsp47 (collagen specific chaperone) to skin cells, including specific collagen subtypes quantification. Results Here we quantify the collagen deposition level and the types of deposited collagens after Hsp47 stimulation in different in vitro cultures of cells from human skin tissue (fibroblasts NHDF, keratinocytes HaCat and endothelial cells HDMEC) and mouse fibroblasts (L929 and MEF). We find upregulated deposition of fibrillar collagen subtypes I, III and V after Hsp47 delivery. Network collagen IV deposition was enhanced in HaCat and HDMECs, while fibril-associated collagen XII was not affected by the increased intracellular Hsp47 levels. The deposition levels of fibrillar collagen were cell-dependent i.e. Hsp47-stimulated fibroblasts deposited significantly higher amount of fibrillar collagen than Hsp47-stimulated HaCat and HDMECs. Conclusions A 3-fold enhancement of collagen deposition was observed in fibroblasts upon repeated dosage of Hsp47 within the first 6 days of culture. Our results provide fundamental understanding towards the idea of using Hsp47 as therapeutic protein to treat collagen disorders.
Collapse
Affiliation(s)
- Essak S Khan
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany.,Chemistry Department, Saarland University, 66123, Saarbrücken, Germany
| | | | - Lorena Llontop
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany
| | - Aránzazu Del Campo
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123, Saarbrücken, Germany. .,Chemistry Department, Saarland University, 66123, Saarbrücken, Germany.
| |
Collapse
|
11
|
Muñoz-Talavera A, Gómez-Lim MÁ, Salazar-Olivo LA, Reinders J, Lim K, Escobedo-Moratilla A, López-Calleja AC, Islas-Carbajal MC, Rincón-Sánchez AR. Expression of the Biologically Active Insulin Analog SCI-57 in Nicotiana Benthamiana. Front Pharmacol 2019; 10:1335. [PMID: 31798448 PMCID: PMC6868099 DOI: 10.3389/fphar.2019.01335] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 10/18/2019] [Indexed: 11/13/2022] Open
Abstract
Diabetes mellitus is a growing problem worldwide; however, only 23% of low-income countries have access to insulin, and ironically it costs higher in such countries than high-income ones. Therefore, new strategies for insulin and insulin analogs production are urgently required to improve low-cost access to therapeutic products, so as to contain the diabetes epidemic. SCI-57 is an insulin analog with a greater affinity for the insulin receptor and lower thermal degradation than native insulin. It also shows native mitogenicity and insulin-like biological activity. In this work, SCI-57 was transiently expressed in the Nicotiana benthamiana (Nb) plant, and we also evaluated some of its relevant biological effects. An expression plasmid was engineered to translate an N-terminal ubiquitin and C-terminal endoplasmic reticulum-targeting signal KDEL, in order to increase protein expression and stability. Likewise, the effect of co-expression of influenza M2 ion channel (M2) on the expression of insulin analog SCI-57 (SCI-57/M2) was evaluated. Although using M2 increases yield, it tends to alter the SCI-57 amino acid sequence, possibly promoting the formation of oligomers. Purification of SCI-57 was achieved by FPLC cation exchange and ultrafiltration of N. benthamiana leaf extract (NLE). SCI-57 exerts its anti-diabetic properties by stimulating glucose uptake in adipocytes, without affecting the lipid accumulation process. Expression of the insulin analog in agroinfiltrated plants was confirmed by SDS-PAGE, RP-HPLC, and MS. Proteome changes related to the expression of heterologous proteins on N. benthamiana were not observed; up-regulated proteins were related to the agroinfiltration process. Our results demonstrate the potential for producing a biologically active insulin analog, SCI-57, by transient expression in Nb.
Collapse
Affiliation(s)
- Adriana Muñoz-Talavera
- Department of Physiology, Institute of Experimental and Clinical Therapeutics, University Center for Health Sciences, University of Guadalajara, Guadalajara, Mexico
| | - Miguel Ángel Gómez-Lim
- Department of Genetic Engineering, Center for Research and Advanced Studies of the National Polytechnic Institute, Irapuato, Mexico
| | - Luis A Salazar-Olivo
- Division of Molecular Biology, Institute for Scientific and Technological Research of San Luis Potosí, San Luis Potosí, Mexico
| | - Jörg Reinders
- Scientific Support Unit Analytical Chemistry, Leibniz Research Centre for Working Environment and Human Factors, Dortmund, Germany
| | - Katharina Lim
- Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Abraham Escobedo-Moratilla
- CONACYT-Consortium for Research, Innovation, and Development of the Drylands (CIIDZA), IPICYT, San Luis Potosí, Mexico
| | - Alberto Cristian López-Calleja
- Department of Genetic Engineering, Center for Research and Advanced Studies of the National Polytechnic Institute, Irapuato, Mexico
| | - María Cristina Islas-Carbajal
- Department of Physiology, Institute of Experimental and Clinical Therapeutics, University Center for Health Sciences, University of Guadalajara, Guadalajara, Mexico
| | - Ana Rosa Rincón-Sánchez
- Institute of Molecular Biology and Gene Therapy, Department of Molecular Biology and Genomic, University Center for Health Sciences, University of Guadalajara, Guadalajara, Mexico
| |
Collapse
|
12
|
Kokubun H, Jin H, Aoe T. Pathogenic Effects of Impaired Retrieval between the Endoplasmic Reticulum and Golgi Complex. Int J Mol Sci 2019; 20:ijms20225614. [PMID: 31717602 PMCID: PMC6888596 DOI: 10.3390/ijms20225614] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 10/31/2019] [Accepted: 11/07/2019] [Indexed: 12/15/2022] Open
Abstract
Cellular activities, such as growth and secretion, are dependent on correct protein folding and intracellular protein transport. Injury, like ischemia, malnutrition, and invasion of toxic substances, affect the folding environment in the endoplasmic reticulum (ER). The ER senses this information, following which cells adapt their response to varied situations through the unfolded protein response. Activation of the KDEL receptor, resulting from the secretion from the ER of chaperones containing the KDEL sequence, plays an important role in this adaptation. The KDEL receptor was initially shown to be necessary for the retention of KDEL sequence-containing proteins in the ER. However, it has become clear that the activated KDEL receptor also regulates bidirectional transport between the ER and the Golgi complex, as well as from the Golgi to the secretory pathway. In addition, it has been suggested that the signal for KDEL receptor activation may also affect several other cellular activities. In this review, we discuss KDEL receptor-mediated bidirectional transport and signaling and describe disease models and human diseases related to KDEL receptor dysfunction.
Collapse
Affiliation(s)
- Hiroshi Kokubun
- Department of Anesthesiology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Hisayo Jin
- Department of Anesthesiology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Tomohiko Aoe
- Department of Medicine, Pain Center, Chiba Medical Center, Teikyo University, Ichihara 299-0111, Japan
- Correspondence: ; Tel.: +81-436-62-1211
| |
Collapse
|
13
|
Royal JM, Oh YJ, Grey MJ, Lencer WI, Ronquillo N, Galandiuk S, Matoba N. A modified cholera toxin B subunit containing an ER retention motif enhances colon epithelial repair via an unfolded protein response. FASEB J 2019; 33:13527-13545. [PMID: 31560862 DOI: 10.1096/fj.201901255r] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Cholera toxin B subunit (CTB) exhibits broad-spectrum biologic activity upon mucosal administration. Here, we found that a recombinant CTB containing an endoplasmic reticulum (ER) retention motif (CTB-KDEL) induces colon epithelial wound healing in colitis via the activation of an unfolded protein response (UPR) in colon epithelial cells. In a Caco2 cell wound healing model, CTB-KDEL, but not CTB or CTB-KDE, facilitated cell migration via interaction with the KDEL receptor, localization in the ER, UPR activation, and subsequent TGF-β signaling. Inhibition of the inositol-requiring enzyme 1/X-box binding protein 1 arm of UPR abolished the cell migration effect of CTB-KDEL, indicating that the pathway is indispensable for the activity. CTB-KDEL's capacity to induce UPR and epithelial restitution or wound healing was corroborated in a dextran sodium sulfate-induced acute colitis mouse model. Furthermore, CTB-KDEL induced a UPR, up-regulated wound healing pathways, and maintained viable crypts in colon explants from patients with inflammatory bowel disease (IBD). In summary, CTB-KDEL exhibits unique wound healing effects in the colon that are mediated by its localization to the ER and subsequent activation of UPR in epithelial cells. The results provide implications for a novel therapeutic approach for mucosal healing, a significant unmet need in IBD treatment.-Royal, J. M., Oh, Y. J., Grey, M. J., Lencer, W. I., Ronquillo, N., Galandiuk, S., Matoba, N. A modified cholera toxin B subunit containing an ER retention motif enhances colon epithelial repair via an unfolded protein response.
Collapse
Affiliation(s)
- Joshua M Royal
- Department of Pharmacology and Toxicology, James Graham Brown Cancer Center, Center for Predictive Medicine, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Young Jun Oh
- Department of Pharmacology and Toxicology, James Graham Brown Cancer Center, Center for Predictive Medicine, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Michael J Grey
- Division of Gastroenterology, Nutrition, and Hepatology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Digestive Disease Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Wayne I Lencer
- Division of Gastroenterology, Nutrition, and Hepatology, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics, Harvard Digestive Disease Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Nemencio Ronquillo
- Division of Anatomic Pathology, Department of Pathology and Laboratory Medicine, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Susan Galandiuk
- The Hiram C. Polk Jr., M.D. Department of Surgery, Price Institute of Surgical Research, University of Louisville, Louisville, Kentucky, USA
| | - Nobuyuki Matoba
- Department of Pharmacology and Toxicology, James Graham Brown Cancer Center, Center for Predictive Medicine, University of Louisville School of Medicine, Louisville, Kentucky, USA
| |
Collapse
|
14
|
Luo PM, Boyce M. Directing Traffic: Regulation of COPI Transport by Post-translational Modifications. Front Cell Dev Biol 2019; 7:190. [PMID: 31572722 PMCID: PMC6749011 DOI: 10.3389/fcell.2019.00190] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 08/23/2019] [Indexed: 12/12/2022] Open
Abstract
The coat protein complex I (COPI) is an essential, highly conserved pathway that traffics proteins and lipids between the endoplasmic reticulum (ER) and the Golgi. Many aspects of the COPI machinery are well understood at the structural, biochemical and genetic levels. However, we know much less about how cells dynamically modulate COPI trafficking in response to changing signals, metabolic state, stress or other stimuli. Recently, post-translational modifications (PTMs) have emerged as one common theme in the regulation of the COPI pathway. Here, we review a range of modifications and mechanisms that govern COPI activity in interphase cells and suggest potential future directions to address as-yet unanswered questions.
Collapse
Affiliation(s)
- Peter M Luo
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, United States
| | - Michael Boyce
- Department of Biochemistry, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
15
|
Efficient engulfment of necroptotic and pyroptotic cells by nonprofessional and professional phagocytes. Cell Discov 2019; 5:39. [PMID: 31632688 PMCID: PMC6796833 DOI: 10.1038/s41421-019-0108-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2018] [Revised: 04/18/2019] [Accepted: 06/05/2019] [Indexed: 12/20/2022] Open
|
16
|
Abstract
All proteins end with a carboxyl terminus that has unique biophysical properties and is often disordered. Although there are examples of important C-termini functions, a more global role for the C-terminus is not yet established. In this review, we summarize research on C-termini, a unique region in proteins that cells exploit. Alternative splicing and proteolysis increase the diversity of proteins and peptides in cells with unique C-termini. The C-termini of proteins contain minimotifs, short peptides with an encoded function generally characterized as binding, posttranslational modifications, and trafficking. Many of these activities are specific to minimotifs on the C-terminus. Approximately 13% of C-termini in the human proteome have a known minimotif, and the majority, if not all of the remaining termini have conserved motifs inferring a function that remains to be discovered. C-termini, their predictions, and their functions are collated in the C-terminome, Proteus, and Terminus Oriented Protein Function INferred Database (TopFIND) database/web systems. Many C-termini are well conserved, and some have a known role in health and disease. We envision that this summary of C-termini will guide future investigation of their biochemical and physiological significance.
Collapse
Affiliation(s)
- Surbhi Sharma
- a Nevada Institute of Personalized Medicine and School of Life Sciences , University of Nevada , Las Vegas , NV , USA
| | - Martin R Schiller
- a Nevada Institute of Personalized Medicine and School of Life Sciences , University of Nevada , Las Vegas , NV , USA
| |
Collapse
|
17
|
Jin H, Komita M, Aoe T. Decreased Protein Quality Control Promotes the Cognitive Dysfunction Associated With Aging and Environmental Insults. Front Neurosci 2018; 12:753. [PMID: 30443201 PMCID: PMC6221900 DOI: 10.3389/fnins.2018.00753] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 10/01/2018] [Indexed: 11/29/2022] Open
Abstract
Background: Most neurodegenerative diseases are sporadic and develop with age. Degenerative neural tissues often contain intra- and extracellular protein aggregates, suggesting that the proteostasis network that combats protein misfolding could be dysfunctional in the setting of neurodegenerative disease. Binding immunoglobulin protein (BiP) is an endoplasmic reticulum (ER) chaperone that is crucial for protein folding and modulating the adaptive response in early secretory pathways. The interaction between BiP and unfolded proteins is mediated by the substrate-binding domain and nucleotide-binding domain with ATPase activity. The interaction facilitates protein folding and maturation. BiP has a recovery motif at the carboxyl terminus. The aim of this study is to examine cognitive function in model mice with an impaired proteostasis network by expressing a mutant form of BiP lacking the recovery motif. We also investigated if impairments of cognitive function were exacerbated by exposure to environmental insults, such as inhaled anesthetics. Methods: We examined cognitive function by performing radial maze testing with mutant BiP mice and assessed the additional impact of general anesthesia in the context of proteostasis dysfunction. Testing over 8 days was performed 10 weeks, 6 months, and 1 year after birth. Results: Age-related cognitive decline occurred in both forms of mice. The mutant BiP and anesthetic exposure promoted cognitive dysfunction prior to the senile period. After senescence, when mice were tested at 6 months of age and at 1 year old, there were no significant differences between the two genotypes in terms of the radial maze testing; furthermore, there was no significant difference when tested with and without anesthetic exposure. Conclusion: Our data suggest that aging was the predominant factor underlying the impairment of cognitive function in this study. Impairment of the proteostasis network may promote age-related neurodegeneration, and this is exacerbated by external insults.
Collapse
Affiliation(s)
- Hisayo Jin
- Department of Anesthesiology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Mari Komita
- Department of Anesthesiology, Chiba Rosai Hospital, Ichihara, Japan
| | - Tomohiko Aoe
- Department of Medicine, Pain Center, Chiba Medical Center, Teikyo University, Ichihara, Japan
| |
Collapse
|
18
|
Aiki H, Wada T, Iba K, Oki G, Sohma H, Yamashita T, Kokai Y. Proteomics analysis of site- and stage-specific protein expression after peripheral nerve injury. J Orthop Sci 2018; 23:1070-1078. [PMID: 30100211 DOI: 10.1016/j.jos.2018.07.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 06/14/2018] [Accepted: 07/18/2018] [Indexed: 01/26/2023]
Abstract
BACKGROUND The peripheral nervous system has greater regenerative potential than the CNS. This fact suggests the existence of molecules that act as key factors in nerve regeneration during molecular changes in the peripheral nervous system. METHODS The right sciatic nerve of female Sprague-Dawley rats was exposed and transected at the mid-thigh level. Animals were sacrificed at 5, 10 or 35 days after nerve transection. Proximal and distal nerve segments (1-cm in length) were dissected. We then sought to observe overall molecular changes after peripheral nerve injury using a proteomic approach. For an overview of the identified proteins, each protein was classified according to its biological and molecular functions. We identified a number of proteins showing site- and stage-specific patterns of expression. RESULTS Both proximal and distal molecular changes at 5, 10 and 35 days after nerve transection were investigated, and a total of 2353 proteins were identified. Among the various expression patterns observed, aFGF and GAP-43 were found to increase in the proximal stump at 10 days after transection, and PN-1, RPL9 and prosaposin increased in the distal stump at 5 days after transection. Among these proteins, aFGF, GAP-43, PN-1 and prosaposin were found to be associated with nerve regeneration. CONCLUSION We demonstrated that aFGF, GAP-43, PN-1 and prosaposin expression increased at specific stages and in specific sites, such as the proximal or distal stump, after nerve transection by comprehensive measurement using proteomics analysis. We believe that these specific expression patterns might play important roles during regeneration after nerve injury.
Collapse
Affiliation(s)
- Hikono Aiki
- Department of Biomedical Engineering, Sapporo Medical University School of Medicine, Sapporo, Japan; Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Takuro Wada
- Division of Orthopaedic Surgery, Saiseikai Otaru Hospital, Otaru, Japan
| | - Kousuke Iba
- Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan.
| | - Gosuke Oki
- Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Hitoshi Sohma
- Department of Educational Development, Sapporo Medical University Center for Medical Education, Sapporo, Japan
| | - Toshihiko Yamashita
- Department of Orthopaedic Surgery, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Yasuo Kokai
- Department of Biomedical Engineering, Sapporo Medical University School of Medicine, Sapporo, Japan
| |
Collapse
|
19
|
The Golgi architecture and cell sensing. Biochem Soc Trans 2018; 46:1063-1072. [DOI: 10.1042/bst20180323] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 08/06/2018] [Accepted: 08/21/2018] [Indexed: 12/23/2022]
Abstract
An array of signalling molecules are located at the Golgi apparatus, including phosphoinositides, small GTPases, kinases, and phosphatases, which are linked to multiple signalling pathways. Initially considered to be associated predominantly with membrane trafficking, signalling pathways at the Golgi are now recognised to regulate a diverse range of higher-order functions. Many of these signalling pathways are influenced by the architecture of the Golgi. In vertebrate cells, the Golgi consists of individual stacks fused together into a compact ribbon structure and the function of this ribbon structure has been enigmatic. Notably, recent advances have identified a role for the Golgi ribbon in regulation of cellular processes. Fragmentation of the Golgi ribbon results in modulation of many signalling pathways. Various diseases and disorders, including cancer and neurodegeneration, are associated with the loss of the Golgi ribbon and the appearance of a dispersed fragmented Golgi. Here, we review the emerging theme of the Golgi as a cell sensor and highlight the relationship between the morphological status of the Golgi in vertebrate cells and the modulation of signalling networks.
Collapse
|
20
|
Silva-Alvim FAL, An J, Alvim JC, Foresti O, Grippa A, Pelgrom A, Adams TL, Hawes C, Denecke J. Predominant Golgi Residency of the Plant K/HDEL Receptor Is Essential for Its Function in Mediating ER Retention. THE PLANT CELL 2018; 30:2174-2196. [PMID: 30072420 PMCID: PMC6181015 DOI: 10.1105/tpc.18.00426] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 07/02/2018] [Accepted: 07/26/2018] [Indexed: 05/08/2023]
Abstract
Accumulation of soluble proteins in the endoplasmic reticulum (ER) of plants is mediated by a receptor termed ER RETENTION DEFECTIVE2 (ERD2) or K/HDEL receptor. Using two gain-of-function assays and by complementing loss of function in Nicotiana benthamiana, we discovered that compromising the lumenal N terminus or the cytosolic C terminus with fluorescent fusions abolishes its biological function and profoundly affects its subcellular localization. Based on the confirmed asymmetrical topology of ERD2, we engineered a new fluorescent ERD2 fusion protein that retains biological activity. Using this fusion, we show that ERD2 is exclusively detected at the Golgi apparatus, unlike nonfunctional C-terminal fusions, which also label the ER. Moreover, ERD2 is confined to early Golgi compartments and does not show ligand-induced redistribution to the ER. We show that the cytosolic C terminus of ERD2 plays a crucial role in its function. Two conserved leucine residues that do not correspond to any known targeting motifs for ER-Golgi trafficking were shown to be essential for both ERD2 Golgi residency and its ability to mediate ER retention of soluble ligands. The results suggest that anterograde ER to Golgi transport of ERD2 is either extremely fast, well in excess of the bulk flow rate, or that ERD2 does not recycle in the way originally proposed.
Collapse
Affiliation(s)
- Fernanda A L Silva-Alvim
- Centre for Plant Sciences, School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Jing An
- Centre for Plant Sciences, School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Jonas C Alvim
- Centre for Plant Sciences, School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Ombretta Foresti
- Centre for Plant Sciences, School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Alexandra Grippa
- Centre for Plant Sciences, School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Alexandra Pelgrom
- Centre for Plant Sciences, School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Thomas L Adams
- Centre for Plant Sciences, School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
| | - Chris Hawes
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University, Headington, Oxford OX3 0AZ, United Kingdom
| | - Jurgen Denecke
- Centre for Plant Sciences, School of Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
| |
Collapse
|
21
|
Wang S, Xie K, Xu G, Zhou H, Guo Q, Wu J, Liao Z, Liu N, Wang Y, Liu Y. Plant G proteins interact with endoplasmic reticulum luminal protein receptors to regulate endoplasmic reticulum retrieval. JOURNAL OF INTEGRATIVE PLANT BIOLOGY 2018; 60:541-561. [PMID: 29573168 DOI: 10.1111/jipb.12648] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 03/12/2018] [Indexed: 06/08/2023]
Abstract
Maintaining endoplasmic reticulum (ER) homeostasis is essential for the production of biomolecules. ER retrieval, i.e., the retrograde transport of compounds from the Golgi to the ER, is one of the pathways that ensures ER homeostasis. However, the mechanisms underlying the regulation of ER retrieval in plants remain largely unknown. Plant ERD2-like proteins (ERD2s) were recently suggested to function as ER luminal protein receptors that mediate ER retrieval. Here, we demonstrate that heterotrimeric G protein signaling is involved in ERD2-mediated ER retrieval. We show that ERD2s interact with the heterotrimeric G protein Gα and Gγ subunits at the Golgi. Silencing of Gα, Gβ, or Gγ increased the retention of ER luminal proteins. Furthermore, overexpression of Gα, Gβ, or Gγ caused ER luminal proteins to escape from the ER, as did the co-silencing of ERD2a and ERD2b. These results suggest that G proteins interact with ER luminal protein receptors to regulate ER retrieval.
Collapse
Affiliation(s)
- Shanshan Wang
- MOE Key Laboratory of Bioinformatics, Center for Plant Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Ke Xie
- Advanced Biotechnology and Application Research Center, School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Guoyong Xu
- The Institute for Advanced Studies (IAS), Wuhan University, Wuhan 430072, China
| | - Huarui Zhou
- MOE Key Laboratory of Bioinformatics, Center for Plant Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Qiang Guo
- MOE Key Laboratory of Bioinformatics, Center for Plant Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jingyi Wu
- MOE Key Laboratory of Bioinformatics, Center for Plant Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Zengwei Liao
- MOE Key Laboratory of Bioinformatics, Center for Plant Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Na Liu
- MOE Key Laboratory of Bioinformatics, Center for Plant Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yan Wang
- MOE Key Laboratory of Bioinformatics, Center for Plant Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Yule Liu
- MOE Key Laboratory of Bioinformatics, Center for Plant Biology, Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| |
Collapse
|
22
|
Serebrenik YV, Hellerschmied D, Toure M, López-Giráldez F, Brookner D, Crews CM. Targeted protein unfolding uncovers a Golgi-specific transcriptional stress response. Mol Biol Cell 2018; 29:1284-1298. [PMID: 29851555 PMCID: PMC5994893 DOI: 10.1091/mbc.e17-11-0693] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 03/26/2018] [Accepted: 03/30/2018] [Indexed: 12/12/2022] Open
Abstract
In eukaryotic cells, organelle-specific stress-response mechanisms are vital for maintaining cellular homeostasis. The Golgi apparatus, an essential organelle of the secretory system, is the major site of protein modification and sorting within a cell and functions as a platform for spatially regulated signaling. Golgi homeostasis mechanisms that regulate organelle structure and ensure precise processing and localization of protein substrates remain poorly understood. Using a chemical biology strategy to induce protein unfolding, we uncover a Golgi-specific transcriptional response. An RNA-sequencing profile of this stress response compared with the current state-of-the-art Golgi stressors, nigericin and xyloside, demonstrates the enhanced precision of Golgi targeting achieved with our system. The data set further reveals previously uncharacterized genes that we find to be essential for Golgi structural integrity. These findings highlight the Golgi's ability to sense misfolded proteins and establish new aspects of Golgi autoregulation.
Collapse
Affiliation(s)
- Yevgeniy V. Serebrenik
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511
| | - Doris Hellerschmied
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511
| | - Momar Toure
- Department of Chemistry, Yale University, New Haven, CT 06511
| | | | - Dennis Brookner
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511
| | - Craig M. Crews
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511
- Department of Chemistry, Yale University, New Haven, CT 06511
- Department of Pharmacology, Yale University, New Haven, CT 06511
| |
Collapse
|
23
|
Rogne M, Chu DT, Küntziger TM, Mylonakou MN, Collas P, Tasken K. OPA1-anchored PKA phosphorylates perilipin 1 on S522 and S497 in adipocytes differentiated from human adipose stem cells. Mol Biol Cell 2018; 29:1487-1501. [PMID: 29688805 PMCID: PMC6014102 DOI: 10.1091/mbc.e17-09-0538] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Optic atrophy 1 (OPA1) is the A-kinase anchoring protein targeting the pool of protein kinase A (PKA) responsible for perilipin 1 phosphorylation, a gatekeeper for lipolysis. However, the involvement of OPA1-bound PKA in the downstream regulation of lipolysis is unknown. Here we show up-regulation and relocation of OPA1 from mitochondria to lipid droplets during adipocytic differentiation of human adipose stem cells. We employed various biochemical and immunological approaches to demonstrate that OPA1-bound PKA phosphorylates perilipin 1 at S522 and S497 on lipolytic stimulation. We show that the first 30 amino acids of OPA1 are essential for its lipid droplet localization as is OMA1-dependent processing. Finally, our results indicate that presence of OPA1 is necessary for lipolytic phosphorylation of downstream targets. Our results show for the first time, to our knowledge, how OPA1 mediates adrenergic control of lipolysis in human adipocytes by regulating phosphorylation of perilipin 1.
Collapse
Affiliation(s)
- Marie Rogne
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo, 0318 Oslo, Norway
| | - Dinh-Toi Chu
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo, 0318 Oslo, Norway
| | | | - Maria-Niki Mylonakou
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo, 0318 Oslo, Norway
| | - Philippe Collas
- Department of Molecular Medicine, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0318 Oslo, Norway.,Norewegian Center for Stem Cell Research, Department of Immunology and Transfusion Medicine, Oslo University Hospital, 0424 Oslo, Norway
| | - Kjetil Tasken
- Centre for Molecular Medicine Norway, Nordic EMBL Partnership, University of Oslo, 0318 Oslo, Norway.,Department of Cancer Immunology, Institute of Cancer Research, Oslo University Hospital, 0424 Oslo, Norway
| |
Collapse
|
24
|
Abstract
The coat protein complex I (COPI) allows the precise sorting of lipids and proteins between Golgi cisternae and retrieval from the Golgi to the ER. This essential role maintains the identity of the early secretory pathway and impinges on key cellular processes, such as protein quality control. In this Cell Science at a Glance and accompanying poster, we illustrate the different stages of COPI-coated vesicle formation and revisit decades of research in the context of recent advances in the elucidation of COPI coat structure. By calling attention to an array of questions that have remained unresolved, this review attempts to refocus the perspectives of the field.
Collapse
Affiliation(s)
- Eric C Arakel
- Department of Molecular Biology, Universitätsmedizin Göttingen, Humboldtallee 23, 37073 Göttingen, Germany
| | - Blanche Schwappach
- Department of Molecular Biology, Universitätsmedizin Göttingen, Humboldtallee 23, 37073 Göttingen, Germany .,Max-Planck Institute for Biophysical Chemistry, 37077 Göttingen, Germany
| |
Collapse
|
25
|
Kim HY, Kang JA, Ryou JH, Lee GH, Choi DS, Lee DE, Kim HS. Intracellular Protein Delivery System Using a Target-Specific Repebody and Translocation Domain of Bacterial Exotoxin. ACS Chem Biol 2017; 12:2891-2897. [PMID: 29019389 DOI: 10.1021/acschembio.7b00562] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
With the high efficacy of protein-based therapeutics and plenty of intracellular drug targets, cytosolic protein delivery in a cell-specific manner has attracted considerable attention in the field of precision medicine. Herein, we present an intracellular protein delivery system based on a target-specific repebody and the translocation domain of Pseudomonas aeruginosa exotoxin A. The delivery platform was constructed by genetically fusing an EGFR-specific repebody as a targeting moiety to the translocation domain, while a protein cargo was fused to the C-terminal end of the delivery platform. The delivery platform was revealed to efficiently translocate a protein cargo to the cytosol in a target-specific manner. We demonstrate the utility and potential of the delivery platform by showing a remarkable tumor regression with negligible toxicity in a xenograft mice model when gelonin was used as the cytotoxic protein cargo. The present platform can find wide applications to the cell-selective cytosolic delivery of diverse proteins in many areas.
Collapse
Affiliation(s)
- Hee-Yeon Kim
- Graduate School of Nanoscience and Technology (WCU), Korea Advanced Institute of Science and Technology (KAIST) , 291 Daehak-ro, Yuseong-gu, Daejeon, 305-701, Korea
| | - Jung Ae Kang
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute , 29 Geumgu-gil, Jeongeup-si, Jeollabuk-do 580-185, Korea
| | - Jeong-Hyun Ryou
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST) , 291 Daehak-ro, Yuseong-gu, Daejeon, 305-701, Korea
| | - Gyeong Hee Lee
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute , 29 Geumgu-gil, Jeongeup-si, Jeollabuk-do 580-185, Korea
| | - Dae Seong Choi
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute , 29 Geumgu-gil, Jeongeup-si, Jeollabuk-do 580-185, Korea
| | - Dong Eun Lee
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute , 29 Geumgu-gil, Jeongeup-si, Jeollabuk-do 580-185, Korea
| | - Hak-Sung Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST) , 291 Daehak-ro, Yuseong-gu, Daejeon, 305-701, Korea
| |
Collapse
|
26
|
Gosavi P, Gleeson PA. The Function of the Golgi Ribbon Structure - An Enduring Mystery Unfolds! Bioessays 2017; 39. [PMID: 28984991 DOI: 10.1002/bies.201700063] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 08/31/2017] [Indexed: 12/13/2022]
Abstract
The Golgi apparatus in vertebrate cells consists of individual Golgi stacks fused together in a continuous ribbon structure. The ribbon structure per se is not required to mediate the classical functions of this organelle and the relevance of the "ribbon" structure has been a mystery since first identified ultrastructurally in the 1950s. Recent advances recognize a role for the Golgi apparatus in a range of cellular processes, some mediated by signaling networks which are regulated at the Golgi. Here we review the cellular processes and signaling events regulated by the Golgi apparatus and, in particular, explore an emerging theme that the ribbon structure of the Golgi contributes directly to the regulation of these higher order functions.
Collapse
Affiliation(s)
- Prajakta Gosavi
- The Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, 3010, Australia
| | - Paul A Gleeson
- The Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, 3010, Australia
| |
Collapse
|
27
|
Jin H, Komita M, Aoe T. The Role of BiP Retrieval by the KDEL Receptor in the Early Secretory Pathway and its Effect on Protein Quality Control and Neurodegeneration. Front Mol Neurosci 2017; 10:222. [PMID: 28769758 PMCID: PMC5511815 DOI: 10.3389/fnmol.2017.00222] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Accepted: 06/27/2017] [Indexed: 12/14/2022] Open
Abstract
Protein quality control in the early secretory pathway is a ubiquitous eukaryotic mechanism for adaptation to endoplasmic reticulum (ER) stress. An ER molecular chaperone, immunoglobulin heavy chain-binding protein (BiP), is one of the essential components in this process. BiP interacts with nascent proteins to facilitate their folding. BiP also plays an important role in preventing aggregation of misfolded proteins and regulating the ER stress response when cells suffer various injuries. BiP is a member of the 70-kDa heat shock protein (HSP70) family of molecular chaperones that resides in the ER. Interaction between BiP and unfolded proteins is mediated by a substrate-binding domain and a nucleotide-binding domain for ATPase activity, leading to protein folding and maturation. BiP also possesses a retrieval motif in its carboxyl terminal. When BiP is secreted from the ER, the Lys-Asp-Glu-Leu (KDEL) receptor in the post-ER compartments binds with the carboxyl terminal KDEL sequence of BiP and returns BiP to the ER via coat protein complex I (COPI) vesicular transport. Although yeast studies showed that BiP retrieval by the KDEL receptor is not essential in single cells, it is crucial for multicellular organisms, where some essential proteins require retrieval to facilitate folding and maturation. Experiments in knock-in mice expressing mutant BiP with the retrieval motif deleted revealed a unique role of BiP retrieval by the KDEL receptor in neuronal development and age-related neurodegeneration.
Collapse
Affiliation(s)
- Hisayo Jin
- Department of Anesthesiology, Graduate School of Medicine, Chiba UniversityChiba, Japan
| | - Mari Komita
- Department of Anesthesiology, Chiba Rosai HospitalIchihara, Japan
| | - Tomohiko Aoe
- Pain Center, Chiba Medical Center, Teikyo UniversityIchihara, Japan
| |
Collapse
|
28
|
Pastor-Cantizano N, Montesinos JC, Bernat-Silvestre C, Marcote MJ, Aniento F. p24 family proteins: key players in the regulation of trafficking along the secretory pathway. PROTOPLASMA 2016; 253:967-985. [PMID: 26224213 DOI: 10.1007/s00709-015-0858-6] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 07/13/2015] [Indexed: 05/20/2023]
Abstract
p24 family proteins have been known for a long time, but their functions have remained elusive. However, they are emerging as essential regulators of protein trafficking along the secretory pathway, influencing the composition, structure, and function of different organelles in the pathway, especially the ER and the Golgi apparatus. In addition, they appear to modulate the transport of specific cargos, including GPI-anchored proteins, G-protein-coupled receptors, or K/HDEL ligands. As a consequence, they have been shown to play specific roles in signaling, development, insulin secretion, and the pathogenesis of Alzheimer's disease. The search of new putative ligands may open the way to discover new functions for this fascinating family of proteins.
Collapse
Affiliation(s)
- Noelia Pastor-Cantizano
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universitat de València, Avenida Vicente Andrés Estellés, s/n, E-46100, Burjassot, Valencia, Spain
| | - Juan Carlos Montesinos
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universitat de València, Avenida Vicente Andrés Estellés, s/n, E-46100, Burjassot, Valencia, Spain
| | - César Bernat-Silvestre
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universitat de València, Avenida Vicente Andrés Estellés, s/n, E-46100, Burjassot, Valencia, Spain
| | - María Jesús Marcote
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universitat de València, Avenida Vicente Andrés Estellés, s/n, E-46100, Burjassot, Valencia, Spain
| | - Fernando Aniento
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universitat de València, Avenida Vicente Andrés Estellés, s/n, E-46100, Burjassot, Valencia, Spain.
| |
Collapse
|
29
|
Tenorio MJ, Luchsinger C, Mardones GA. Protein kinase A activity is necessary for fission and fusion of Golgi to endoplasmic reticulum retrograde tubules. PLoS One 2015; 10:e0135260. [PMID: 26258546 PMCID: PMC4530959 DOI: 10.1371/journal.pone.0135260] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 07/20/2015] [Indexed: 11/18/2022] Open
Abstract
It is becoming increasingly accepted that together with vesicles, tubules play a major role in the transfer of cargo between different cellular compartments. In contrast to our understanding of the molecular mechanisms of vesicular transport, little is known about tubular transport. How signal transduction molecules regulate these two modes of membrane transport processes is also poorly understood. In this study we investigated whether protein kinase A (PKA) activity regulates the retrograde, tubular transport of Golgi matrix proteins from the Golgi to the endoplasmic reticulum (ER). We found that Golgi-to-ER retrograde transport of the Golgi matrix proteins giantin, GM130, GRASP55, GRASP65, and p115 was impaired in the presence of PKA inhibitors. In addition, we unexpectedly found accumulation of tubules containing both Golgi matrix proteins and resident Golgi transmembrane proteins. These tubules were still attached to the Golgi and were highly dynamic. Our data suggest that both fission and fusion of retrograde tubules are mechanisms regulated by PKA activity.
Collapse
Affiliation(s)
- María J. Tenorio
- Instituto de Fisiología, Facultad de Medicina, and Centro Interdisciplinario de Estudios del Sistema Nerviso, Universidad Austral de Chile, Valdivia, Chile
| | - Charlotte Luchsinger
- Instituto de Fisiología, Facultad de Medicina, and Centro Interdisciplinario de Estudios del Sistema Nerviso, Universidad Austral de Chile, Valdivia, Chile
| | - Gonzalo A. Mardones
- Instituto de Fisiología, Facultad de Medicina, and Centro Interdisciplinario de Estudios del Sistema Nerviso, Universidad Austral de Chile, Valdivia, Chile
- * E-mail:
| |
Collapse
|
30
|
Kilaparty SP, Singh A, Baltosser WH, Ali N. Computational analysis reveals a successive adaptation of multiple inositol polyphosphate phosphatase 1 in higher organisms through evolution. Evol Bioinform Online 2014; 10:239-50. [PMID: 25574123 PMCID: PMC4275298 DOI: 10.4137/ebo.s18948] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 09/29/2014] [Accepted: 10/05/2014] [Indexed: 11/05/2022] Open
Abstract
Multiple inositol polyphosphate phosphatase 1 (Minpp1) in higher organisms dephosphorylates InsP6, the most abundant inositol phosphate. It also dephosphorylates less phosphorylated InsP5 and InsP4 and more phosphorylated InsP7 or InsP8. Minpp1 is classified as a member of the histidine acid phosphatase super family of proteins with functional resemblance to phytases found in lower organisms. This study took a bioinformatics approach to explore the extent of evolutionary diversification in Minpp1 structure and function in order to understand its physiological relevance in higher organisms. The human Minpp1 amino acid (AA) sequence was BLAST searched against available national protein databases. Phylogenetic analysis revealed that Minpp1 was widely distributed from lower to higher organisms. Further, we have identified that there exist four isoforms of Minpp1. Multiple computational tools were used to identify key functional motifs and their conservation among various species. Analyses showed that certain motifs predominant in higher organisms were absent in lower organisms. Variation in AA sequences within motifs was also analyzed. We found that there is diversification of key motifs and thus their functions present in Minpp1 from lower organisms to higher organisms. Another interesting result of this analysis was the presence of a glucose-1-phosphate interaction site in Minpp1; the functional significance of which has yet to be determined experimentally. The overall findings of our study point to an evolutionary adaptability of Minpp1 functions from lower to higher life forms.
Collapse
Affiliation(s)
- Surya P Kilaparty
- Department of Biology, University of Arkansas at Little Rock, Arkansas, USA
| | - Awantika Singh
- UAMS/UALR Joint Bioinformatics Program, University of Arkansas at Little Rock, Arkansas, USA
| | | | - Nawab Ali
- Department of Biology, University of Arkansas at Little Rock, Arkansas, USA
| |
Collapse
|
31
|
Montesinos JC, Pastor-Cantizano N, Robinson DG, Marcote MJ, Aniento F. Arabidopsis p24δ5 and p24δ9 facilitate Coat Protein I-dependent transport of the K/HDEL receptor ERD2 from the Golgi to the endoplasmic reticulum. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2014; 80:1014-30. [PMID: 25312353 DOI: 10.1111/tpj.12700] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2014] [Revised: 10/02/2014] [Accepted: 10/03/2014] [Indexed: 05/18/2023]
Abstract
The p24 proteins belong to a family of type I membrane proteins which cycle between the endoplasmic reticulum (ER) and Golgi via coat protein I (COPI) and COPII vesicles. Current nomenclature classifies them into four subfamilies, although plant p24 proteins belong to either the p24β or the p24δ subfamilies. Here, we show that Arabidopsis p24δ5/δ9 and HDEL ligands shift the steady-state distribution of the K/HDEL receptor ERD2 from the Golgi to the ER. We also show that p24δ5/δ9 interact directly with ERD2. This interaction requires the Golgi dynamics (GOLD) domain in p24δ5 and is much higher at acidic than at neutral pH, consistent with both proteins interacting at the cis-Golgi. In addition, p24δ5 also inhibits the secretion of HDEL ligands, but not constitutive secretion, showing a role for p24δ5 in retrograde Golgi-to-ER transport. Both p24δ5 and ERD2 interact with ADP-ribosylation factor 1 (ARF1) and COPI subunits, mostly at acidic pH, consistent with COPI vesicles being involved in retrograde transport of both proteins. In contrast, both proteins interact with the COPII subunit Sec23, mostly at neutral pH, consistent with this interaction taking place at the ER for anterograde transport to the Golgi apparatus.
Collapse
Affiliation(s)
- Juan Carlos Montesinos
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universitat de València, València, Spain
| | | | | | | | | |
Collapse
|
32
|
Cancino J, Capalbo A, Di Campli A, Giannotta M, Rizzo R, Jung JE, Di Martino R, Persico M, Heinklein P, Sallese M, Luini A. Control systems of membrane transport at the interface between the endoplasmic reticulum and the Golgi. Dev Cell 2014; 30:280-94. [PMID: 25117681 DOI: 10.1016/j.devcel.2014.06.018] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 04/08/2014] [Accepted: 06/23/2014] [Indexed: 10/24/2022]
Abstract
A fundamental property of cellular processes is to maintain homeostasis despite varying internal and external conditions. Within the membrane transport apparatus, variations in membrane fluxes from the endoplasmic reticulum (ER) to the Golgi complex are balanced by opposite fluxes from the Golgi to the ER to maintain homeostasis between the two organelles. Here we describe a molecular device that balances transport fluxes by integrating transduction cascades with the transport machinery. Specifically, ER-to-Golgi transport activates the KDEL receptor at the Golgi, which triggers a cascade that involves Gs and adenylyl cyclase and phosphodiesterase isoforms and then PKA activation and results in the phosphorylation of transport machinery proteins. This induces retrograde traffic to the ER and balances transport fluxes between the ER and Golgi. Moreover, the KDEL receptor activates CREB1 and other transcription factors that upregulate transport-related genes. Thus, a Golgi-based control system maintains transport homeostasis through both signaling and transcriptional networks.
Collapse
Affiliation(s)
- Jorge Cancino
- Istituto di Biochimica delle Proteine, Consiglio Nazionale delle Ricerche, Via Pietro Castellino 111, 80131 Napoli, Italy; Telethon Institute of Genetics and Medicine, Via Pietro Castellino 111, 80131 Napoli, Italy; Departamento de Ciencias Biológicas, Facultad de Ciencias Biológicas, Universidad Andrés Bello, Quillota 980, Viña del Mar 2520000, Chile.
| | - Anita Capalbo
- Istituto di Biochimica delle Proteine, Consiglio Nazionale delle Ricerche, Via Pietro Castellino 111, 80131 Napoli, Italy; Telethon Institute of Genetics and Medicine, Via Pietro Castellino 111, 80131 Napoli, Italy
| | - Antonella Di Campli
- Istituto di Biochimica delle Proteine, Consiglio Nazionale delle Ricerche, Via Pietro Castellino 111, 80131 Napoli, Italy
| | - Monica Giannotta
- Consorzio Mario Negri Sud, Via Nazionale 8/A, 66030 Santa Maria Imbaro (Chieti), Italy
| | - Riccardo Rizzo
- Istituto di Biochimica delle Proteine, Consiglio Nazionale delle Ricerche, Via Pietro Castellino 111, 80131 Napoli, Italy; Telethon Institute of Genetics and Medicine, Via Pietro Castellino 111, 80131 Napoli, Italy
| | - Juan E Jung
- Istituto di Biochimica delle Proteine, Consiglio Nazionale delle Ricerche, Via Pietro Castellino 111, 80131 Napoli, Italy; Istituto di Ricovero e Cura a Carattere Scientifico, Istituto di Ricerca Diagnostica e Nucleare (SDN), 80143 Napoli, Italy
| | - Rosaria Di Martino
- Istituto di Biochimica delle Proteine, Consiglio Nazionale delle Ricerche, Via Pietro Castellino 111, 80131 Napoli, Italy
| | - Maria Persico
- Istituto di Biochimica delle Proteine, Consiglio Nazionale delle Ricerche, Via Pietro Castellino 111, 80131 Napoli, Italy; Istituto di Ricovero e Cura a Carattere Scientifico, Istituto di Ricerca Diagnostica e Nucleare (SDN), 80143 Napoli, Italy
| | - Petra Heinklein
- Institut für Biochemie Charité, Universitätsmedizin Berlin, CrossOver Charitéplatz 1/Sitz, Virchowweg 6, 10117 Berlin, Germany
| | - Michele Sallese
- Consorzio Mario Negri Sud, Via Nazionale 8/A, 66030 Santa Maria Imbaro (Chieti), Italy
| | - Alberto Luini
- Istituto di Biochimica delle Proteine, Consiglio Nazionale delle Ricerche, Via Pietro Castellino 111, 80131 Napoli, Italy; Telethon Institute of Genetics and Medicine, Via Pietro Castellino 111, 80131 Napoli, Italy.
| |
Collapse
|
33
|
Kabeiseman EJ, Cichos KH, Moore ER. The eukaryotic signal sequence, YGRL, targets the chlamydial inclusion. Front Cell Infect Microbiol 2014; 4:129. [PMID: 25309881 PMCID: PMC4161167 DOI: 10.3389/fcimb.2014.00129] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 08/28/2014] [Indexed: 11/13/2022] Open
Abstract
Understanding how host proteins are targeted to pathogen-specified organelles, like the chlamydial inclusion, is fundamentally important to understanding the biogenesis of these unique subcellular compartments and how they maintain autonomy within the cell. Syntaxin 6, which localizes to the chlamydial inclusion, contains an YGRL signal sequence. The YGRL functions to return syntaxin 6 to the trans-Golgi from the plasma membrane, and deletion of the YGRL signal sequence from syntaxin 6 also prevents the protein from localizing to the chlamydial inclusion. YGRL is one of three YXXL (YGRL, YQRL, and YKGL) signal sequences which target proteins to the trans-Golgi. We designed various constructs of eukaryotic proteins to test the specificity and propensity of YXXL sequences to target the inclusion. The YGRL signal sequence redirects proteins (e.g., Tgn38, furin, syntaxin 4) that normally do not localize to the chlamydial inclusion. Further, the requirement of the YGRL signal sequence for syntaxin 6 localization to inclusions formed by different species of Chlamydia is conserved. These data indicate that there is an inherent property of the chlamydial inclusion, which allows it to recognize the YGRL signal sequence. To examine whether this "inherent property" was protein or lipid in nature, we asked if deletion of the YGRL signal sequence from syntaxin 6 altered the ability of the protein to interact with proteins or lipids. Deletion or alteration of the YGRL from syntaxin 6 does not appreciably impact syntaxin 6-protein interactions, but does decrease syntaxin 6-lipid interactions. Intriguingly, data also demonstrate that YKGL or YQRL can successfully substitute for YGRL in localization of syntaxin 6 to the chlamydial inclusion. Importantly and for the first time, we are establishing that a eukaryotic signal sequence targets the chlamydial inclusion.
Collapse
Affiliation(s)
| | | | - Elizabeth R. Moore
- Division of Basic Biomedical Sciences, Sanford School of Medicine, University of South DakotaVermillion, SD, USA
| |
Collapse
|
34
|
Gao C, Cai Y, Wang Y, Kang BH, Aniento F, Robinson DG, Jiang L. Retention mechanisms for ER and Golgi membrane proteins. TRENDS IN PLANT SCIENCE 2014; 19:508-15. [PMID: 24794130 DOI: 10.1016/j.tplants.2014.04.004] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/23/2014] [Revised: 03/27/2014] [Accepted: 04/03/2014] [Indexed: 05/18/2023]
Abstract
Unless there are mechanisms to selectively retain membrane proteins in the endoplasmic reticulum (ER) or in the Golgi apparatus, they automatically proceed downstream to the plasma or vacuole membranes. Two types of coat protein complex I (COPI)-interacting motifs in the cytosolic tails of membrane proteins seem to facilitate membrane retention in the early secretory pathway of plants: a dilysine (KKXX) motif (which is typical of p24 proteins) for the ER and a KXE/D motif (which occurs in the Arabidopsis endomembrane protein EMP12) for the Golgi apparatus. The KXE/D motif is highly conserved in all eukaryotic EMPs and is additionally present in hundreds of other proteins of unknown subcellular localization and function. This novel signal may represent a new general mechanism for Golgi targeting and the retention of polytopic integral membrane proteins.
Collapse
Affiliation(s)
- Caiji Gao
- School of Life Sciences, Centre for Cell and Developmental Biology and State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Yi Cai
- School of Life Sciences, Centre for Cell and Developmental Biology and State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | - Yejun Wang
- Vaccine and Infectious Disease Organization, University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK, S7N5E3, Canada
| | - Byung-Ho Kang
- Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| | - Fernando Aniento
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Valencia, Spain
| | - David G Robinson
- Department of Plant Cell Biology, Centre for Organismal Studies, University of Heidelberg, D-69120 Heidelberg, Germany
| | - Liwen Jiang
- School of Life Sciences, Centre for Cell and Developmental Biology and State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China.
| |
Collapse
|
35
|
Abrams EW, Cheng YL, Andrew DJ. Drosophila KDEL receptor function in the embryonic salivary gland and epidermis. PLoS One 2013; 8:e77618. [PMID: 24204897 PMCID: PMC3799842 DOI: 10.1371/journal.pone.0077618] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Accepted: 09/10/2013] [Indexed: 11/19/2022] Open
Abstract
Core components of the secretory pathway have largely been identified and studied in single cell systems such as the budding yeast S. cerevisiae or in mammalian tissue culture. These studies provide details on the molecular functions of the secretory machinery; they fail, however, to provide insight into the role of these proteins in the context of specialized organs of higher eukaryotes. Here, we identify and characterize the first loss-of-function mutations in a KDEL receptor gene from higher eukaryotes. Transcripts from the Drosophila KDEL receptor gene KdelR - formerly known as dmErd2 - are provided maternally and, at later stages, are at elevated levels in several embryonic cell types, including the salivary gland secretory cells, the fat body and the epidermis. We show that, unlike Saccharomyces cerevisiae Erd2 mutants, which are viable, KdelR mutations are early larval lethal, with homozygous mutant animals dying as first instar larvae. KdelR mutants have larval cuticle defects similar to those observed with loss-of-function mutations in other core secretory pathway genes and with mutations in CrebA, which encodes a bZip transcription factor that coordinately upregulates secretory pathway component genes in specialized secretory cell types. Using the salivary gland, we demonstrate a requirement for KdelR in maintaining the ER pool of a subset of soluble resident ER proteins. These studies underscore the utility of the Drosophila salivary gland as a unique system for studying the molecular machinery of the secretory pathway in vivo in a complex eukaryote.
Collapse
Affiliation(s)
- Elliott W. Abrams
- Department of Cell Biology, the Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Yim Ling Cheng
- Department of Cell Biology, the Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Deborah J. Andrew
- Department of Cell Biology, the Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
36
|
Regulation of Golgi signaling and trafficking by the KDEL receptor. Histochem Cell Biol 2013; 140:395-405. [DOI: 10.1007/s00418-013-1130-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/09/2013] [Indexed: 12/31/2022]
|
37
|
Peripheral nerve axons contain machinery for co-translational secretion of axonally-generated proteins. Neurosci Bull 2013; 29:493-500. [PMID: 23839054 DOI: 10.1007/s12264-013-1360-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Accepted: 05/19/2013] [Indexed: 10/26/2022] Open
Abstract
The axonal compartment of developing neurons and mature peripheral nervous system (PNS) neurons has the capacity to locally synthesize proteins. Axonally-synthesized proteins have been shown to facilitate axonal pathfinding and maintenance in developing central nervous system (CNS) and PNS neurons, and to facilitate the regeneration of mature PNS neurons. RNA-profiling studies of the axons of cultured neurons have shown a surprisingly complex population of mRNAs that encode proteins for a myriad of functions. Although classic-appearing rough endoplasmic reticulum (RER), smooth endoplasmic reticulum (ER) and Golgi apparatus have not been documented in axons by ultrastructural studies, axonal RNA profiling studies show several membrane and secreted protein-encoding mRNAs whose translation products would need access to a localized secretory mechanism. We previously showed that the axons of cultured neurons contain functional equivalents of RER and Golgi apparatus. Here, we show that markers for the signal-recognition particle, RER, ER, and Golgi apparatus are present in PNS axons in vivo. Co-localization of these proteins mirrors that seen for cultured axons where locally-translated proteins are localized to the axoplasmic membrane. Moreover, nerve injury increases the levels and/or aggregation of these proteins, suggesting that the regenerating axon has an increased capacity for membrane targeting of locally synthesized proteins.
Collapse
|
38
|
Gutiérrez-Martínez E, Fernández-Ulibarri I, Lázaro-Diéguez F, Johannes L, Pyne S, Sarri E, Egea G. Lipid phosphate phosphatase 3 participates in transport carrier formation and protein trafficking in the early secretory pathway. J Cell Sci 2013; 126:2641-55. [PMID: 23591818 DOI: 10.1242/jcs.117705] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The inhibition of phosphatidic acid phosphatase (PAP) activity by propanolol indicates that diacylglycerol (DAG) is required for the formation of transport carriers at the Golgi and for retrograde trafficking to the ER. Here we report that the PAP2 family member lipid phosphate phosphatase 3 (LPP3, also known as PAP2b) localizes in compartments of the secretory pathway from ER export sites to the Golgi complex. The depletion of human LPP3: (i) reduces the number of tubules generated from the ER-Golgi intermediate compartment and the Golgi, with those formed from the Golgi being longer in LPP3-silenced cells than in control cells; (ii) impairs the Rab6-dependent retrograde transport of Shiga toxin subunit B from the Golgi to the ER, but not the anterograde transport of VSV-G or ssDsRed; and (iii) induces a high accumulation of Golgi-associated membrane buds. LPP3 depletion also reduces levels of de novo synthesized DAG and the Golgi-associated DAG contents. Remarkably, overexpression of a catalytically inactive form of LPP3 mimics the effects of LPP3 knockdown on Rab6-dependent retrograde transport. We conclude that LPP3 participates in the formation of retrograde transport carriers at the ER-Golgi interface, where it transitorily cycles, and during its route to the plasma membrane.
Collapse
Affiliation(s)
- Enric Gutiérrez-Martínez
- Departament de Biologia Cel·lular, Immunologia i Neurociències, Facultat de Medicina, Universitat de Barcelona, 08036 Barcelona, Spain
| | | | | | | | | | | | | |
Collapse
|
39
|
Wang G, Norton AS, Pokharel D, Song Y, Hill RA. KDEL peptide gold nanoconstructs: promising nanoplatforms for drug delivery. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2013; 9:366-74. [DOI: 10.1016/j.nano.2012.09.002] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 08/29/2012] [Accepted: 09/08/2012] [Indexed: 12/20/2022]
|
40
|
Cancino J, Luini A. Signaling Circuits on the Golgi Complex. Traffic 2012; 14:121-34. [DOI: 10.1111/tra.12022] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 10/12/2012] [Accepted: 10/12/2012] [Indexed: 01/21/2023]
|
41
|
Muppirala M, Gupta V, Swarup G. Tyrosine phosphorylation of a SNARE protein, syntaxin 17: implications for membrane trafficking in the early secretory pathway. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2012; 1823:2109-19. [PMID: 23006999 DOI: 10.1016/j.bbamcr.2012.09.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Accepted: 09/13/2012] [Indexed: 10/27/2022]
Abstract
The T-cell protein tyrosine phosphatase is expressed as two splice variants - TC45, a nuclear protein, and TC48, which is localized predominantly in the ER (endoplasmic reticulum). Yeast two-hybrid screening revealed direct interaction of TC48 with Syntaxin17, a SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor) protein localized predominantly in the ER and to some extent in the ER-Golgi intermediate compartment. Syntaxin 17 did not interact with TC45. C-terminal 40 amino acids of TC48 were sufficient for interaction with syntaxin 17. Overexpressed syntaxin 17 was phosphorylated at tyrosine upon pervanadate treatment (a tyrosine phosphatase inhibitor/tyrosine kinase activator) of COS-1 cells. Mutational analysis identified Tyr156 in the cytoplasmic domain as the major site of phosphorylation. Endogenous syntaxin 17 was phosphorylated by pervanadate treatment in CHO and MIN6 cells but was not phosphorylated in a variety of other cell lines tested. c-Abl was identified as one of the kinases, which phosphorylates syntaxin 17 in MIN6 cells. Phosphorylation of endogenous and overexpressed syntaxin 17 was reduced in the presence of IGF receptor and EGF receptor kinase inhibitors. Serum depletion reduced pervanadate-induced phosphorylation of endogenous syntaxin 17. TC48 coexpression reduced phosphorylation of syntaxin 17 by pervanadate and purified TC48 directly dephosphorylated syntaxin 17. β-COP dispersal by overexpressed syntaxin 17 was reduced after pervanadate-induced phosphorylation. A phospho-mimicking mutant (Y156E) of syntaxin 17 showed reduced interaction with COPI vesicles. These results suggest that tyrosine phosphorylation of syntaxin 17 is likely to have a role in regulating syntaxin 17 dependent membrane trafficking in the early secretory pathway.
Collapse
Affiliation(s)
- Madhavi Muppirala
- Centre for Cellular and Molecular Biology, Council of Scientific and Industrial Research, Uppal Road, Hyderabad 500 007, India
| | | | | |
Collapse
|
42
|
Balse E, Steele DF, Abriel H, Coulombe A, Fedida D, Hatem SN. Dynamic of Ion Channel Expression at the Plasma Membrane of Cardiomyocytes. Physiol Rev 2012; 92:1317-58. [DOI: 10.1152/physrev.00041.2011] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cardiac myocytes are characterized by distinct structural and functional entities involved in the generation and transmission of the action potential and the excitation-contraction coupling process. Key to their function is the specific organization of ion channels and transporters to and within distinct membrane domains, which supports the anisotropic propagation of the depolarization wave. This review addresses the current knowledge on the molecular actors regulating the distinct trafficking and targeting mechanisms of ion channels in the highly polarized cardiac myocyte. In addition to ubiquitous mechanisms shared by other excitable cells, cardiac myocytes show unique specialization, illustrated by the molecular organization of myocyte-myocyte contacts, e.g., the intercalated disc and the gap junction. Many factors contribute to the specialization of the cardiac sarcolemma and the functional expression of cardiac ion channels, including various anchoring proteins, motors, small GTPases, membrane lipids, and cholesterol. The discovery of genetic defects in some of these actors, leading to complex cardiac disorders, emphasizes the importance of trafficking and targeting of ion channels to cardiac function. A major challenge in the field is to understand how these and other actors work together in intact myocytes to fine-tune ion channel expression and control cardiac excitability.
Collapse
Affiliation(s)
- Elise Balse
- Institute of Cardiometabolism and Nutrition, Paris, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Heart and Metabolism Division, Paris, France; Institut National de la Santé et de la Recherche Médicale UMR_S956, Paris, France; Université Pierre et Marie Curie, Paris, France; Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, Canada; and Department of Clinical Research University of Bern, Bern, Switzerland
| | - David F. Steele
- Institute of Cardiometabolism and Nutrition, Paris, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Heart and Metabolism Division, Paris, France; Institut National de la Santé et de la Recherche Médicale UMR_S956, Paris, France; Université Pierre et Marie Curie, Paris, France; Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, Canada; and Department of Clinical Research University of Bern, Bern, Switzerland
| | - Hugues Abriel
- Institute of Cardiometabolism and Nutrition, Paris, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Heart and Metabolism Division, Paris, France; Institut National de la Santé et de la Recherche Médicale UMR_S956, Paris, France; Université Pierre et Marie Curie, Paris, France; Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, Canada; and Department of Clinical Research University of Bern, Bern, Switzerland
| | - Alain Coulombe
- Institute of Cardiometabolism and Nutrition, Paris, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Heart and Metabolism Division, Paris, France; Institut National de la Santé et de la Recherche Médicale UMR_S956, Paris, France; Université Pierre et Marie Curie, Paris, France; Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, Canada; and Department of Clinical Research University of Bern, Bern, Switzerland
| | - David Fedida
- Institute of Cardiometabolism and Nutrition, Paris, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Heart and Metabolism Division, Paris, France; Institut National de la Santé et de la Recherche Médicale UMR_S956, Paris, France; Université Pierre et Marie Curie, Paris, France; Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, Canada; and Department of Clinical Research University of Bern, Bern, Switzerland
| | - Stéphane N. Hatem
- Institute of Cardiometabolism and Nutrition, Paris, France; Assistance Publique-Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Heart and Metabolism Division, Paris, France; Institut National de la Santé et de la Recherche Médicale UMR_S956, Paris, France; Université Pierre et Marie Curie, Paris, France; Department of Anesthesiology, Pharmacology, and Therapeutics, University of British Columbia, Vancouver, Canada; and Department of Clinical Research University of Bern, Bern, Switzerland
| |
Collapse
|
43
|
Popoff V, Adolf F, Brügger B, Wieland F. COPI budding within the Golgi stack. Cold Spring Harb Perspect Biol 2011; 3:a005231. [PMID: 21844168 DOI: 10.1101/cshperspect.a005231] [Citation(s) in RCA: 120] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The Golgi serves as a hub for intracellular membrane traffic in the eukaryotic cell. Transport within the early secretory pathway, that is within the Golgi and from the Golgi to the endoplasmic reticulum, is mediated by COPI-coated vesicles. The COPI coat shares structural features with the clathrin coat, but differs in the mechanisms of cargo sorting and vesicle formation. The small GTPase Arf1 initiates coating on activation and recruits en bloc the stable heptameric protein complex coatomer that resembles the inner and the outer shells of clathrin-coated vesicles. Different binding sites exist in coatomer for membrane machinery and for the sorting of various classes of cargo proteins. During the budding of a COPI vesicle, lipids are sorted to give a liquid-disordered phase composition. For the release of a COPI-coated vesicle, coatomer and Arf cooperate to mediate membrane separation.
Collapse
Affiliation(s)
- Vincent Popoff
- Heidelberg University Biochemistry Center, 69120 Heidelberg, Germany
| | | | | | | |
Collapse
|
44
|
Abstract
The protein processing and trafficking function of the Golgi is intimately linked to multiple intracellular signaling pathways. Assembly of Golgi trafficking structures and lipid sorting at the Golgi complex is controlled and coordinated by specific phosphoinositide kinases and phosphatases. The intra-Golgi transport machinery is also regulated by kinases belonging to several functionally distinct families, for example, MAP kinase signaling is required for mitotic disassembly of the Golgi. However, the Golgi plays an additional, prominent role in compartmentalizing other signaling cascades that originate at the plasma membrane or at other organelles. This article summarizes recent advances in our understanding of the signaling network that converges at the Golgi.
Collapse
Affiliation(s)
- Peter Mayinger
- Division of Nephrology and Hypertension and Department of Cell and Developmental Biology, Oregon Health & Science University, Portland, OR 97239, USA.
| |
Collapse
|
45
|
Abstract
For growth, survival, communication and homeostasis, cells transport a large number of proteins to the plasma membrane and the extracellular medium by using the secretory pathway. Consequently, to adapt to the surrounding environment and the different intracellular contexts, the secretory pathway needs to accommodate and respond to a plethora of endogenous and exogenous stimuli. It is now well established that several kinases, known to be activated by environmental stimuli, signal from the plasma membrane to the secretory pathway in order to remodel its architecture and modulate the cellular secretion capacity. By contrast, membranes of the early secretory pathway, similar to the endosomal system, can also initiate and modulate signalling cascades, thereby spatially organising cellular signalling and eliciting a different cellular outcome than when signalling is localised to the plasma membrane. This Commentary highlights recent contributions to our understanding of the mutual regulation of the secretory pathway and cellular signalling.
Collapse
Affiliation(s)
- Hesso Farhan
- Biozentrum, University of Basel, Klingelbergstrasse 50/70, Basel, Switzerland.
| | | |
Collapse
|
46
|
Dobashi T, Tanabe S, Jin H, Mimura N, Yamamoto T, Nishino T, Aoe T. BiP, an endoplasmic reticulum chaperone, modulates the development of morphine antinociceptive tolerance. J Cell Mol Med 2011; 14:2816-26. [PMID: 19818092 PMCID: PMC3822731 DOI: 10.1111/j.1582-4934.2009.00932.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Morphine is a potent analgesic, but the molecular mechanism for tolerance formation after repeated use is not fully understood. Binding immunoglobulin protein (BiP) is an endoplasmic reticulum (ER) chaperone that is central to ER function. We examined knock-in mice expressing a mutant BiP with the retrieval sequence deleted in order to elucidate physiological processes that are sensitive to BiP functions. We tested the thermal antinociceptive effect of morphine in heterozygous mutant BiP mice in a hot plate test. Paw withdrawal latencies before and after a single administration of morphine were not significantly different between the wild-type and mutant BiP mice. Repeated morphine administration caused the development of morphine tolerance in the wild-type mice. The activation of glycogen synthase kinase 3b (GSK-3b) was associated with morphine tolerance, because an inhibitor of GSK-3β prevented it. On the other hand, the mutant BiP mice showed less morphine tolerance, and the activation of GSK-3b was suppressed in their brain. These results suggest that BiP may play an important role in the development of morphine tolerance. Furthermore, we found that a chemical chaperone which improves ER protein folding capacity also attenuated the development of morphine tolerance in wild-type mice, suggesting a possible clinical application of chemical chaperones in preventing morphine tolerance.
Collapse
Affiliation(s)
- Tamae Dobashi
- Department of Anesthesiology, Chiba University Graduate School of Medicine, Chiba, Japan
| | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Estimates based on proteomic analyses indicate that a third of translated proteins in eukaryotic genomes enter the secretory pathway. After folding and assembly of nascent secretory proteins in the endoplasmic reticulum (ER), the coat protein complex II (COPII) selects folded cargo for export in membrane-bound vesicles. To accommodate the great diversity in secretory cargo, protein sorting receptors are required in a number of instances for efficient ER export. These transmembrane sorting receptors couple specific secretory cargo to COPII through interactions with both cargo and coat subunits. After incorporation into COPII transport vesicles, protein sorting receptors release bound cargo in pre-Golgi or Golgi compartments, and receptors are then recycled back to the ER for additional rounds of cargo export. Distinct types of protein sorting receptors that recognize carbohydrate and/or polypeptide signals in secretory cargo have been characterized. Our current understanding of the molecular mechanisms underlying cargo receptor function are described.
Collapse
Affiliation(s)
- Julia Dancourt
- Department of Biochemistry, Dartmouth Medical School, Hanover, New Hampshire 03755, USA
| | | |
Collapse
|
48
|
Mavillard F, Hidalgo J, Megias D, Levitsky KL, Velasco A. PKA-mediated Golgi remodeling during cAMP signal transmission. Traffic 2010; 11:90-109. [PMID: 20002352 DOI: 10.1111/j.1600-0854.2009.01007.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Cyclic AMP (cAMP)-dependent protein kinase A (PKA) is part of the set of signaling proteins that are stably associated to the cytosolic surface of Golgi membranes in mammalian cells. In principle, Golgi-associated PKA could participate in either signal transduction events and/or the coordination of Golgi transport activities. Here, we show data indicating that although Golgi-associated PKA is activated fast and efficiently during cell stimulation by an extracellular ligand it does not contribute significantly to cAMP signal transmission to the nucleus. Instead, most of the PKA catalytic subunits Calphaderived from the Golgi complex remain localized in the perinuclear cytoplasm where they induce changes in Golgi structural organization. Thus, in stimulated cells the Golgi complex appears collapsed, showing increased colocalization of previously segregated markers and exhibiting merging of different proximal cisternae within a single stack. In contrast, the trans-Golgi network remains as a separate compartment. Consequently, the rate of protein transport is increased whereas glycan processing is not severely affected. This remodeling process requires the presence of PKA activity associated to the Golgi membranes. Together these data indicate that Golgi-associated PKA activity is involved in the adaptation of Golgi dynamic organization to extracellular signaling events.
Collapse
Affiliation(s)
- Fabiola Mavillard
- Department of Cell Biology, Faculty of Biology, University of Seville, Seville, Spain
| | | | | | | | | |
Collapse
|
49
|
The KDEL receptor: new functions for an old protein. FEBS Lett 2009; 583:3863-71. [PMID: 19854180 DOI: 10.1016/j.febslet.2009.10.053] [Citation(s) in RCA: 154] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2009] [Revised: 10/14/2009] [Accepted: 10/20/2009] [Indexed: 02/07/2023]
Abstract
The KDEL receptor is a seven-transmembrane-domain protein that was first described about 20 years ago. Its well-known function is to retrotransport chaperones from the Golgi complex to the endoplasmic reticulum. Recent studies, however, have suggested that the KDEL receptor has additional functions. Indeed, we have demonstrated that chaperone-bound KDEL receptor triggers the activation of Src family kinases on the Golgi complex. This activity is essential in the regulation of Golgi-to-plasma membrane transport. However, the identification of different KDEL receptor interactors that are inconsistent with these established functions opens the possibility of further receptor activities.
Collapse
|
50
|
Coordination of the secretory compartments via inter-organelle signalling. Semin Cell Dev Biol 2009; 20:801-9. [DOI: 10.1016/j.semcdb.2009.04.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2009] [Revised: 04/01/2009] [Accepted: 04/03/2009] [Indexed: 11/18/2022]
|