1
|
Vredevoogd DW, Apriamashvili G, Levy PL, Sinha S, Huinen ZR, Visser NL, de Bruijn B, Boshuizen J, van Hal-van Veen SE, Ligtenberg MA, Bleijerveld OB, Lin CP, Díaz-Gómez J, Sánchez SD, Markovits E, Simon Nieto J, van Vliet A, Krijgsman O, Markel G, Besser MJ, Altelaar M, Ruppin E, Peeper DS. TMED inhibition suppresses cell surface PD-1 expression and overcomes T cell dysfunction. J Immunother Cancer 2024; 12:e010145. [PMID: 39510795 PMCID: PMC11552591 DOI: 10.1136/jitc-2024-010145] [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] [Accepted: 08/09/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND Blockade of the programmed cell death protein 1 (PD-1) immune checkpoint (ICB) is revolutionizing cancer therapy, but little is known about the mechanisms governing its expression on CD8 T cells. Because PD-1 is induced during activation of T cells, we set out to uncover regulators whose inhibition suppresses PD-1 abundance without adversely impacting on T cell activation. METHODS To identify PD-1 regulators in an unbiased fashion, we performed a whole-genome, fluorescence-activated cell sorting (FACS)-based CRISPR-Cas9 screen in primary murine CD8 T cells. A dual-readout design using the activation marker CD137 allowed us to uncouple genes involved in PD-1 regulation from those governing general T cell activation. RESULTS We found that the inactivation of one of several members of the TMED/EMP24/GP25L/p24 family of transport proteins, most prominently TMED10, reduced PD-1 cell surface abundance, thereby augmenting T cell activity. Another client protein was cytotoxic T lymphocyte-associated protein 4 (CTLA-4), which was also suppressed by TMED inactivation. Treatment with TMED inhibitor AGN192403 led to lysosomal degradation of the TMED-PD-1 complex and reduced PD-1 abundance in tumor-infiltrating CD8 T cells (TIL) in mice, thus reversing T cell dysfunction. Clinically corroborating these findings, single-cell RNA analyses revealed a positive correlation between TMED expression in CD8 TIL, and both a T cell dysfunction signature and lack of ICB response. Similarly, patients receiving a TIL product with high TMED expression had a shorter overall survival. CONCLUSION Our results uncover a novel mechanism of PD-1 regulation, and identify a pharmacologically tractable target whose inhibition suppresses PD-1 abundance and T cell dysfunction.
Collapse
Affiliation(s)
- David W Vredevoogd
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Georgi Apriamashvili
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Pierre L Levy
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Sanju Sinha
- Cancer Data Science Laboratory, National Cancer Institute Center for Cancer Research, Bethesda, Maryland, USA
| | - Zowi R Huinen
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Nils L Visser
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Beaunelle de Bruijn
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Julia Boshuizen
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Susan E van Hal-van Veen
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Maarten A Ligtenberg
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Onno B Bleijerveld
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Chun-Pu Lin
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Judit Díaz-Gómez
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Santiago Duro Sánchez
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Ettai Markovits
- Ella Lemelbaum Institute for Immuno-oncology, Sheba Medical Center, Tel Hashomer, Israel
- Department of Clinical Microbiology and Immunology, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Juan Simon Nieto
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Alex van Vliet
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Oscar Krijgsman
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Gal Markel
- Department of Clinical Microbiology and Immunology, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Davidoff Center and Samueli Integrative Cancer Pioneering Center, Rabin Medical Center, Petah Tikva, Israel
| | - Michal J Besser
- Department of Clinical Microbiology and Immunology, Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Davidoff Center and Samueli Integrative Cancer Pioneering Center, Rabin Medical Center, Petah Tikva, Israel
- Felsenstein Medical Research Center, The Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Maarten Altelaar
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, The Netherlands
| | - Eytan Ruppin
- Cancer Data Science Laboratory, National Cancer Institute Center for Cancer Research, Bethesda, Maryland, USA
| | - Daniel S Peeper
- Department of Molecular oncology and immunology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| |
Collapse
|
2
|
Chen X, Zhang W, Han X, Li X, Xia L, Wu Y, Zhou Y. TMED3 stabilizes SMAD2 by counteracting NEDD4-mediated ubiquitination to promote ovarian cancer. Mol Carcinog 2024; 63:803-816. [PMID: 38411267 DOI: 10.1002/mc.23689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 12/04/2023] [Accepted: 01/11/2024] [Indexed: 02/28/2024]
Abstract
Ovarian cancer is a major cause of death among cancer patients. Recent research has shown that the transmembrane emp24 domain (TMED) protein family plays a role in the progression of various types of cancer. In this study, we investigated the expression of TMED3 in ovarian cancer tumors compared to nontumor tissues using immunohistochemical staining. We found that TMED3 was overexpressed in ovarian cancer tumors, and its high expression was associated with poor disease-free and overall survival. To understand the functional implications of TMED3 overexpression in ovarian cancer, we conducted experiments to knockdown TMED3 using short hairpin RNA (shRNA). We observed that TMED3 knockdown resulted in reduced cell viability and migration, as well as increased cell apoptosis. Additionally, in subcutaneous xenograft models in BALB-c nude mice, TMED3 knockdown inhibited tumor growth. Further investigation revealed that SMAD family member 2 (SMAD2) was a downstream target of TMED3, driving ovarian cancer progression. TMED3 stabilized SMAD2 by inhibiting the E3 ligase NEDD4-mediated ubiquitination of SMAD2. To confirm the importance of SMAD2 in TMED3-mediated ovarian cancer, we performed functional rescue experiments and found that SMAD2 played a critical role in this process. Moreover, we discovered that the PI3K-AKT pathway was involved in the promoting effects of TMED3 overexpression on ovarian cancer cells. Overall, our study identifies TMED3 as a prognostic indicator and tumor promoter in ovarian cancer. Its function is likely mediated through the regulation of the SMAD2 and PI3K-AKT signaling pathway. These findings contribute to our understanding of the molecular mechanisms underlying ovarian cancer progression and provide potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Xiaojun Chen
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Wei Zhang
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiaotian Han
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Xiaoqi Li
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Lingfang Xia
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yong Wu
- Department of Gynecologic Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yang Zhou
- Department of Obstetrics and Gynaecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
3
|
Zhang Y, Lu M, Huang J, Tian X, Liang M, Wang M, Song X, Xu L, Yan R, Li X. Identification and characterization of the receptors of a microneme adhesive repeat domain of Eimeria maxima microneme protein 3 in chicken intestine epithelial cells. Poult Sci 2024; 103:103486. [PMID: 38350385 PMCID: PMC10874745 DOI: 10.1016/j.psj.2024.103486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/03/2024] [Accepted: 01/17/2024] [Indexed: 02/15/2024] Open
Abstract
Eimeria maxima microneme protein 3 (EmMIC3) is pivotal in the initial recognition and attachment of E. maxima sporozoites to host cells. EmMIC3 comprises 5 tandem Type I microneme adhesive repeat (MAR) domains, among which MAR2 of EmMIC3 (EmMAR2) has been identified as the primary determinant of EmMIC3-mediated tissue tropism. Nonetheless, the mechanisms through which EmMAR2 guides the parasite to its invasion site through interactions with host receptors remained largely uncharted. In this study, we employed yeast two-hybrid (YTH) screening assays and shotgun LC-MS/MS analysis to identify EmMAR2 receptors in chicken intestine epithelial cells. ATPase H+ transporting V1 subunit G1 (ATP6V1G1), receptor accessory protein 5 (REEP5), transmembrane p24 trafficking protein (TMED2), and delta 4-desaturase sphingolipid 1 (DEGS1) were characterized as the 4 receptors of EmMAR2 by both assays. By blocking the interaction of EmMAR2 with each receptor using specific antibodies, we observed varying levels of inhibition on the invasion of E. maxima sporozoites, and the combined usage of all 4 antibodies resulted in the most pronounced inhibitory effect. Additionally, the spatio-temporal expression profiles of ATP6V1G1, REEP5, TMED2, and DEGS1 were assessed. The tissue-specific expression patterns of EmMAR2 receptors throughout E. maxima infection suggested that ATP6V1G1 and DEGS1 might play a role in early-stage invasion, whereas TMED2 could be involved in middle and late-stage invasion and REEP5 and DEGS1 may participate primarily in late-stage invasion. Consequently, E. maxima may employ a multitude of ligand-receptor interactions to drive invasion during different stages of infection. This study marks the first report of EmMAR2 receptors at the interface between E. maxima and the host, providing insights into the invasion mechanisms of E. maxima and the pathogenesis of coccidiosis.
Collapse
Affiliation(s)
- Yang Zhang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Mingmin Lu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Jianmei Huang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Xiaowei Tian
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Meng Liang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Mingyue Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Xiaokai Song
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Lixin Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Ruofeng Yan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China
| | - Xiangrui Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, People's Republic of China.
| |
Collapse
|
4
|
Roberts BS, Mitra D, Abishek S, Beher R, Satpute-Krishnan P. The p24-family and COPII subunit SEC24C facilitate the clearance of alpha1-antitrypsin Z from the endoplasmic reticulum to lysosomes. Mol Biol Cell 2024; 35:ar45. [PMID: 38294851 PMCID: PMC10916869 DOI: 10.1091/mbc.e23-06-0257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 01/16/2024] [Accepted: 01/24/2024] [Indexed: 02/01/2024] Open
Abstract
A subpopulation of the alpha-1-antitrypsin misfolding Z mutant (ATZ) is cleared from the endoplasmic reticulum (ER) via an ER-to-lysosome-associated degradation (ERLAD) pathway. Here, we report that the COPII subunit SEC24C and the p24-family of proteins facilitate the clearance of ATZ via ERLAD. In addition to the previously reported ERLAD components calnexin and FAM134B, we discovered that ATZ coimmunoprecipitates with the p24-family members TMP21 and TMED9. This contrasts with wild type alpha1-antitrypsin, which did not coimmunoprecipitate with FAM134B, calnexin or the p24-family members. Live-cell imaging revealed that ATZ and the p24-family members traffic together from the ER to lysosomes. Using chemical inhibitors to block ER exit or autophagy, we demonstrated that p24-family members and ATZ co-accumulate at SEC24C marked ER-exit sites or in ER-derived compartments, respectively. Furthermore, depletion of SEC24C, TMP21, or TMED9 inhibited lysosomal trafficking of ATZ and resulted in the increase of intracellular ATZ levels. Conversely, overexpression of these p24-family members resulted in the reduction of ATZ levels. Intriguingly, the p24-family members coimmunoprecipitate with ATZ, FAM134B, and SEC24C. Thus, we propose a model in which the p24-family functions in an adaptor complex linking SEC24C with the ERLAD machinery for the clearance of ATZ.
Collapse
Affiliation(s)
| | - Debashree Mitra
- Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| | - Sudhanshu Abishek
- Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| | - Richa Beher
- Uniformed Services University of the Health Sciences, Bethesda, MD 20814
| | | |
Collapse
|
5
|
Holm JEJ, Soares SG, Symmons MF, Huddin AS, Moncrieffe MC, Gay NJ. Anterograde trafficking of Toll-like receptors requires the cargo sorting adaptors TMED-2 and 7. Traffic 2023; 24:508-521. [PMID: 37491993 PMCID: PMC10946956 DOI: 10.1111/tra.12912] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 06/15/2023] [Accepted: 07/03/2023] [Indexed: 07/27/2023]
Abstract
Toll-Like Receptors (TLRs) play a pivotal role in immunity by recognising conserved structural features of pathogens and initiating the innate immune response. TLR signalling is subject to complex regulation that remains poorly understood. Here we show that two small type I transmembrane receptors, TMED2 and 7, that function as cargo sorting adaptors in the early secretory pathway are required for transport of TLRs from the ER to Golgi. Protein interaction studies reveal that TMED7 interacts with TLR2, TLR4 and TLR5 but not with TLR3 and TLR9. On the other hand, TMED2 interacts with TLR2, TLR4 and TLR3. Dominant negative forms of TMED7 suppress the export of cell surface TLRs from the ER to the Golgi. By contrast TMED2 is required for the ER-export of both plasma membrane and endosomal TLRs. Together, these findings suggest that association of TMED2 and TMED7 with TLRs facilitates anterograde transport from the ER to the Golgi.
Collapse
Affiliation(s)
| | | | | | | | | | - Nicholas J. Gay
- Department of BiochemistryUniversity of CambridgeCambridgeUK
| |
Collapse
|
6
|
Zhou L, Li H, Yao H, Dai X, Gao P, Cheng H. TMED family genes and their roles in human diseases. Int J Med Sci 2023; 20:1732-1743. [PMID: 37928880 PMCID: PMC10620864 DOI: 10.7150/ijms.87272] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 10/03/2023] [Indexed: 11/07/2023] Open
Abstract
The members of the transmembrane emp24 domain-containing protein (TMED) family are summarized in human as four subfamilies, α (TMED 4, 9), β (TMED 2), γ (TMED1, 3, 5, 6, 7) and δ (TMED 10), with a total of nine members, which are important regulators of intracellular protein transport and are involved in normal embryonic development, as well as in the pathogenic processes of many human diseases. Here we systematically review the composition, structure and function of TMED family members, and describe the progress of TMED family in human diseases, including malignancies (head and neck tumors, lung cancer, breast cancer, ovarian cancer, endometrial cancer, gastrointestinal tumors, urological tumors, osteosarcomas, etc.), immune responses, diabetes, neurodegenerative diseases, and nonalcoholic fatty liver disease, dilated cardiomyopathy, mucin 1 nephropathy (MKD), and desiccation syndrome (SS). Finally, we discuss and prospect the potential of TMED for disease prognosis prediction and therapeutic targeting, with a view to laying the foundation for therapeutic research based on TMED family causative genes.
Collapse
Affiliation(s)
| | | | | | - Xingliang Dai
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
| | - Peng Gao
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
| | - Hongwei Cheng
- Department of Neurosurgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, P. R. China
| |
Collapse
|
7
|
Navarro KG, Chamberlin HM. Genetic characterization of C. elegans TMED genes. Dev Dyn 2023; 252:1149-1161. [PMID: 37204056 PMCID: PMC10524739 DOI: 10.1002/dvdy.601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 05/03/2023] [Accepted: 05/07/2023] [Indexed: 05/20/2023] Open
Abstract
BACKGROUND p24/transmembrane Emp24 domain (TMED) proteins are a set of evolutionarily conserved, single pass transmembrane proteins that have been shown to facilitate protein secretion and selection of cargo proteins to transport vesicles in the cellular secretion pathway. However, their functions in animal development are incompletely understood. RESULTS The C. elegans genome encodes eight identified TMED genes, with at least one member from each defined subfamily (α, β, γ, δ). TMED gene mutants exhibit a shared set of defects in embryonic viability, animal movement, and vulval morphology. Two γ subfamily genes, tmed-1 and tmed-3, exhibit the ability to compensate for each other, as defects in movement and vulva morphology are only apparent in double mutants. TMED mutants also exhibit a delay in breakdown of basement membrane during vulva development. CONCLUSIONS The results establish a genetic and experimental framework for the study of TMED gene function in C. elegans, and argue that a functional protein from each subfamily is important for a shared set of developmental processes. A specific function for TMED genes is to facilitate breakdown of the basement membrane between the somatic gonad and vulval epithelial cells, suggesting a role for TMED proteins in tissue reorganization during animal development.
Collapse
|
8
|
Kumar S, Alam SS, Bareke E, Beauchamp MC, Dong Y, Chan W, Majewski J, Jerome-Majewska LA. Sf3b4 regulates chromatin remodeler splicing and Hox expression. Differentiation 2023; 131:59-73. [PMID: 37167859 DOI: 10.1016/j.diff.2023.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 04/27/2023] [Accepted: 04/28/2023] [Indexed: 05/13/2023]
Abstract
SF3B proteins form a heptameric complex in the U2 small nuclear ribonucleoprotein, essential for pre-mRNA splicing. Heterozygous pathogenic variants in human SF3B4 are associated with head, face, limb, and vertebrae defects. Using the CRISPR/Cas9 system, we generated mice with constitutive heterozygous deletion of Sf3b4 and showed that mutant embryos have abnormal vertebral development. Vertebrae abnormalities were accompanied by changes in levels and expression pattern of Hox genes in the somites. RNA sequencing analysis of whole embryos and somites of Sf3b4 mutant and control litter mates revealed increased expression of other Sf3b4 genes. However, the mutants exhibited few differentially expressed genes and a large number of transcripts with differential splicing events (DSE), predominantly increased exon skipping and intron retention. Transcripts with increased DSE included several genes involved in chromatin remodeling that are known to regulate Hox expression. Our study confirms that Sf3b4 is required for normal vertebrae development and shows, for the first time, that like Sf3b1, Sf3b4 also regulates Hox expression. We propose that abnormal splicing of chromatin remodelers is primarily responsible for vertebral defects found in Sf3b4 heterozygous mutant embryos.
Collapse
Affiliation(s)
- Shruti Kumar
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0G1, Canada
| | | | - Eric Bareke
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0G1, Canada
| | - Marie-Claude Beauchamp
- Research Institute of the McGill University Health Centre at Glen Site, Montreal, QC, H4A 3J1, Canada
| | - Yanchen Dong
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0G1, Canada
| | - Wesley Chan
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC, H3A 2B2, Canada
| | - Jacek Majewski
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0G1, Canada
| | - Loydie A Jerome-Majewska
- Department of Human Genetics, McGill University, Montreal, QC, H3A 0G1, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, QC, H3A 2B2, Canada; Research Institute of the McGill University Health Centre at Glen Site, Montreal, QC, H4A 3J1, Canada; Department of Pediatrics, McGill University, Montreal, QC, H4A 3J1, Canada.
| |
Collapse
|
9
|
Roberts BS, Satpute-Krishnan P. The many hats of transmembrane emp24 domain protein TMED9 in secretory pathway homeostasis. Front Cell Dev Biol 2023; 10:1096899. [PMID: 36733337 PMCID: PMC9888432 DOI: 10.3389/fcell.2022.1096899] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Accepted: 12/29/2022] [Indexed: 01/18/2023] Open
Abstract
The secretory pathway is an intracellular highway for the vesicular transport of newly synthesized proteins that spans the endoplasmic reticulum (ER), Golgi, lysosomes and the cell surface. A variety of cargo receptors, chaperones, and quality control proteins maintain the smooth flow of cargo along this route. Among these is vesicular transport protein TMED9, which belongs to the p24/transmembrane emp24 domain (TMED) family of proteins, and is expressed across vertebrate species. The TMED family is comprised of structurally-related type I transmembrane proteins with a luminal N-terminal Golgi-dynamics domain, a luminal coiled-coil domain, a transmembrane domain and a short cytosolic C-terminal tail that binds COPI and COPII coat proteins. TMED9, like other members of the TMED family, was first identified as an abundant constituent of the COPI and COPII coated vesicles that mediate traffic between the ER and the Golgi. TMED9 is typically purified in hetero-oligomers together with TMED family members, suggesting that it may function as part of a complex. Recently, TMED family members have been discovered to play various roles in secretory pathway homeostasis including secreted protein processing, quality control and degradation of misfolded proteins, and post-Golgi trafficking. In particular, TMED9 has been implicated in autophagy, lysosomal sorting, viral replication and cancer, which we will discuss in this Mini-Review.
Collapse
|
10
|
Fang Z, Song YX, Wo GQ, Zhou HL, Li L, Yang SY, Chen X, Zhang J, Tang JH. Screening of the novel immune-suppressive biomarkers of TMED family and whether knockdown of TMED2/3/4/9 inhibits cell migration and invasion in breast cancer. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1280. [PMID: 36618780 PMCID: PMC9816852 DOI: 10.21037/atm-22-5444] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 12/07/2022] [Indexed: 12/23/2022]
Abstract
Background Transmembrane p24 trafficking protein (TMED) family members are implicated in several solid tumors, but their clinical relevance for breast cancer (BC) remains unclear. This study aimed to probe their prognostic values and relations with tumor immunity in BC. Methods TMED family mRNA expression was assessed in five microarray datasets (GSE65212, GSE42568, GSE5364, GSE22820 and GSE45827) from Gene Expression Omnibus (GEO) database and invasive breast cancer (BRCA) cohort from The Cancer Genome Atlas (TCGA). Receiver operating characteristic (ROC) curve was performed to determine the predictive values of filtered members of the TMED family. The protein expressions of screen genes were validated by Clinical Proteomic Tumor Analysis Consortium (CPTAC) data from University of ALabama at Birmingham CANcer data analysis portal (UALCAN) and detected in the clinical specimens by western blot assay. Clinicopathologic variables were analyzed with bc-GenExMiner, and patient prognostic data were obtained with Kaplan-Meier Plotter. In vitro wound healing and invasion assays were performed on siRNA-transfected BC cell lines. TIMER 2.0, SangerBox, and ImmPort were used to evaluate tumor immune infiltration, immune checkpoints, and other immune-related genes. CbioPortal, Metascape, Expression2kinases, and LinkedOmics were used to explore gene regulatory network. Results BC tissues expressed TMED2/3/4/9 at a higher level than normal tissues, providing diagnostic potential. All the areas under the ROC curve for TMED2/3/4/9 were more than 0.7. TMED2/3/4/9 correlated with numerous clinical variables, including lymph node status, Scarff-Bloom-Richardson score (SBR), Nottingham Prognostic Index (NPI), estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER-2), and triple-negative breast cancer (TNBC) status, and their high expression predicted the poor prognosis of BC patients. TMED2/3/4/9 knockdown drastically inhibited the migratory and invasive capacities of MDA-MB-231 and HCC1937 cells. TMED2/3/4/9 expressions correlated negatively with the infiltration of tumor-suppressive immune cells such as CD8+ T cells, dendritic cells, and natural killer cells, and was inversely related to a variety of immune checkpoint genes, including programmed cell death 1 (PD-1) and cytotoxic T-lymphocyte associated protein 4 (CTLA4). A set of kinases, transcription factors, and microRNAs (miRNAs) may regulate TMED2/3/4/9 abnormalities at the genome level. Conclusions TMED2/3/4/9 may serve as diagnostic, prognostic, and immune-suppressive biomarkers in BC.
Collapse
Affiliation(s)
- Zheng Fang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yu-Xin Song
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Guan-Qun Wo
- The First Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hong-Lei Zhou
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Lei Li
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Si-Yuan Yang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiu Chen
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jian Zhang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jin-Hai Tang
- Department of General Surgery, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
11
|
Craniofacial Defects in Embryos with Homozygous Deletion of Eftud2 in Their Neural Crest Cells Are Not Rescued by Trp53 Deletion. Int J Mol Sci 2022; 23:ijms23169033. [PMID: 36012294 PMCID: PMC9409426 DOI: 10.3390/ijms23169033] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/27/2022] [Accepted: 07/29/2022] [Indexed: 12/12/2022] Open
Abstract
Embryos with homozygous mutation of Eftud2 in their neural crest cells (Eftud2ncc−/−) have brain and craniofacial malformations, hyperactivation of the P53-pathway and die before birth. Treatment of Eftud2ncc−/− embryos with pifithrin-α, a P53-inhibitor, partly improved brain and craniofacial development. To uncover if craniofacial malformations and death were indeed due to P53 hyperactivation we generated embryos with homozygous loss of function mutations in both Eftud2 and Trp53 in the neural crest cells. We evaluated the molecular mechanism underlying craniofacial development in pifithrin-α-treated embryos and in Eftud2; Trp53 double homozygous (Eftud2ncc−/−; Trp53ncc−/−) mutant embryos. Eftud2ncc−/− embryos that were treated with pifithrin-α or homozygous mutant for Trp53 in their neural crest cells showed reduced apoptosis in their neural tube and reduced P53-target activity. Furthermore, although the number of SOX10 positive cranial neural crest cells was increased in embryonic day (E) 9.0 Eftud2ncc−/−; Trp53ncc−/− embryos compared to Eftud2ncc−/− mutants, brain and craniofacial development, and survival were not improved in double mutant embryos. Furthermore, mis-splicing of both P53-regulated transcripts, Mdm2 and Foxm1, and a P53-independent transcript, Synj2bp, was increased in the head of Eftud2ncc−/−; Trp53ncc−/− embryos. While levels of Zmat3, a P53- regulated splicing factor, was similar to those of wild-type. Altogether, our data indicate that both P53-regulated and P53-independent pathways contribute to craniofacial malformations and death of Eftud2ncc−/− embryos.
Collapse
|
12
|
Zhang J, Qi Y. Depleting TMED3 alleviates the development of endometrial carcinoma. Cancer Cell Int 2022; 22:231. [PMID: 35854294 PMCID: PMC9295347 DOI: 10.1186/s12935-022-02649-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Accepted: 07/06/2022] [Indexed: 11/10/2022] Open
Abstract
Background As one of gynecologic tumors, endometrial carcinoma (EC) has been characterized by high incidence rate, but its molecular pathogenesis has remained unclear. TMED3 is a membrane protein and has been indicated to implicate several tumor-related diseases. In the current study, we aimed to explore the physiological function of TMED3 in EC progression. Methods Through bioinformatic analysis using The Cancer Genome Atlas database and immunohistochemistry assay on tissue microarray, we examined whether TMED3 was upregulated in EC tissues. After constructing TMED3-knockdown cell models via lentiviral transfection, qPCR and western blot were employed to determine the expression levels of TMED3 mRNA and protein. Then, Celigo cell counting assay, CCK8 assay, flow cytometry, wound-healing assay and Transwell assay were used to detect cell proliferation, cell cycle, cell apoptosis and cell migration, respectively. Results As a result, it was found that TMED3 was upregulated in EC cells, which was also verified in clinical samples. We then found that downregulation of TMED3 considerably restrained cell cycle, cell growth and migration but promoted apoptosis of EC cells. The following in-vivo experiments also verified that tumor growth was inhibited after TMED3 knockdown. The exploration in molecular mechanisms showed that TMED3 deletion may weaken cellular viability through upregulating pro-apoptotic proteins and targeting PI3K/AKT signaling pathways. Conclusions This study suggested that knocking down TMED3 affected the malignant phenotype of EC cells and thus limited tumor progression, which provided insights to the development of targeted drugs for EC treatment. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02649-0.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Shenyang, Liaoning, China
| | - Yue Qi
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, No.36 Sanhao Street, Shenyang, Liaoning, China.
| |
Collapse
|
13
|
Di Minin G, Holzner M, Grison A, Dumeau CE, Chan W, Monfort A, Jerome-Majewska LA, Roelink H, Wutz A. TMED2 binding restricts SMO to the ER and Golgi compartments. PLoS Biol 2022; 20:e3001596. [PMID: 35353806 PMCID: PMC9000059 DOI: 10.1371/journal.pbio.3001596] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 04/11/2022] [Accepted: 03/07/2022] [Indexed: 11/30/2022] Open
Abstract
Hedgehog (HH) signaling is important for embryonic pattering and stem cell differentiation. The G protein–coupled receptor (GPCR) Smoothened (SMO) is the key HH signal transducer modulating both transcription-dependent and transcription-independent responses. We show that SMO protects naive mouse embryonic stem cells (ESCs) from dissociation-induced cell death. We exploited this SMO dependency to perform a genetic screen in haploid ESCs where we identify the Golgi proteins TMED2 and TMED10 as factors for SMO regulation. Super-resolution microscopy shows that SMO is normally retained in the endoplasmic reticulum (ER) and Golgi compartments, and we demonstrate that TMED2 binds to SMO, preventing localization to the plasma membrane. Mutation of TMED2 allows SMO accumulation at the plasma membrane, recapitulating early events after HH stimulation. We demonstrate the physiologic relevance of this interaction in neural differentiation, where TMED2 functions to repress HH signal strength. Identification of TMED2 as a binder and upstream regulator of SMO opens the way for unraveling the events in the ER–Golgi leading to HH signaling activation. Hedgehog signals orchestrate tissue patterning by binding the receptor Patched and restricting the signal transducer Smoothened. A genetic screen reveals Tmed2 as a new interactor of Smoothened that is required for regulating Smoothened transport from the endoplasmic reticulum and Golgi to the plasma membrane and hence modulating the strength of Hedgehog signal transduction.
Collapse
Affiliation(s)
- Giulio Di Minin
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology ETH Hönggerberg, Zurich, Switzerland
- * E-mail: (GDM); (AW)
| | - Markus Holzner
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology ETH Hönggerberg, Zurich, Switzerland
| | - Alice Grison
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Charles E. Dumeau
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology ETH Hönggerberg, Zurich, Switzerland
| | - Wesley Chan
- Department Anatomy and Cell Biology, Human Genetics and McGill University, Montreal, Canada
- Department of Pediatrics, Human Genetics and McGill University, Montreal, Canada
| | - Asun Monfort
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology ETH Hönggerberg, Zurich, Switzerland
| | - Loydie A. Jerome-Majewska
- Department Anatomy and Cell Biology, Human Genetics and McGill University, Montreal, Canada
- Department of Pediatrics, Human Genetics and McGill University, Montreal, Canada
| | - Henk Roelink
- Department of Molecular and Cell Biology, University of California, Berkeley, California, United States of America
| | - Anton Wutz
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology ETH Hönggerberg, Zurich, Switzerland
- * E-mail: (GDM); (AW)
| |
Collapse
|
14
|
Fang T, Wu ZW, Wang Y, Wang F, Du ZQ, Yang CX. Comparative transcriptome analysis identifies important maternal molecules and associated biological pathways for pig and human mature oocytes. Reprod Domest Anim 2022; 57:643-652. [PMID: 35244301 DOI: 10.1111/rda.14105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 02/28/2022] [Indexed: 12/17/2022]
Abstract
Recent researches reveal that during oocyte maturation, species-specific molecular profile exists, and has important functional roles. However, molecular differences between pig (a larger animal model for human reproduction) and human mature oocytes remain unknown. Here, by comparative transcriptome analyses of single cell RNA-seq data, we aimed to identify the common and unique maternal factors and associated biological processes between in vivo and in vitro matured pig oocytes, and between in vitro matured human and pig oocytes. Annotated protein coding mRNAs were identified in pig in vivo (11147) and in vitro (11997), and human in vitro (14491) MII oocytes, respectively. For in vivo and in vitro derived pig MII oocytes, 10551 annotated maternal mRNAs were common, mainly enriched in signaling pathways such as cell cycle, oocyte meiosis, microtubule cytoskeleton, MAPK, RNA processing/binding. Besides, in vivo (596) and in vitro (1446) pig MII-specific mRNAs and their involved signaling pathways (in vivo: Bmp, calcium-mediated signaling, PI3K-Akt; in vitro: growth factor activity, JAK-STAT, cytokine-cytokine receptor interaction, calcium signaling pathway) were also found. As for in vitro derived human and pig MII oocytes, 10285 annotated mRNAs were common, enriched in a variety of signaling pathways (cell cycle, oocyte meiosis, microtubule, AMPK, RNA splicing, protein serine/threonine kinase activity, etc). In vitro MII-specific mRNAs were found for humans (4206) and pigs (1712), which were also enriched in species-specific signaling pathways (humans: golgi related terms, transcription repressor and hormone activity; pigs: ATP biosynthetic process, G protein-coupled peptide receptor activity, animoacyl-tRNA biosynthesis), respectively. These findings improve our understanding on oocyte maturation, and also the limitations of pig model for human oocyte maturation and fertilization.
Collapse
Affiliation(s)
- Ting Fang
- College of Animal Science, Yangtze University, JingZhou, 434025, Hubei, China
| | - Zi-Wei Wu
- College of Animal Science, Yangtze University, JingZhou, 434025, Hubei, China
| | - Yi Wang
- College of Animal Science, Yangtze University, JingZhou, 434025, Hubei, China
| | - Fang Wang
- College of Animal Science, Yangtze University, JingZhou, 434025, Hubei, China
| | - Zhi-Qiang Du
- College of Animal Science, Yangtze University, JingZhou, 434025, Hubei, China
| | - Cai-Xia Yang
- College of Animal Science, Yangtze University, JingZhou, 434025, Hubei, China
| |
Collapse
|
15
|
Yang Y, Liu S, Xie C, Li Q, Gao T, Liu M, Xi M, Zhao L. Trafficking Protein TMED3 Promotes Esophageal Squamous Cell Carcinoma. Biomed J 2022; 46:100528. [PMID: 35358714 DOI: 10.1016/j.bj.2022.03.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 03/02/2022] [Accepted: 03/21/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND The molecular mechanisms of esophageal squamous cell carcinoma (ESCC) remain poorly understood. Transmembrane emp24 trafficking protein 3 (TMED3) acts as an oncogene or tumor suppressor gene in different cancers. Our study was to explore the clinicopathological significance and functional roles of TMED3 in ESCC. METHODS Immunohistochemistry, qPCR, and western blotting were used to analyze the expression of TMED3 in ESCC tissues and cells. Statistical analysis was performed to analyze the relationship between TMED3 expression and tumor characteristics in patients with ESCC. The role of TMED3 in vitro and in vivo was investigated by performing functional verification experiments and using a xenograft mouse model. Proteins that are functionally related to TMED3 were recognized by Affymetrix microarray and Ingenuity Pathway Analysis (IPA). Functional verification experiments were performed to analyze the role of FAM60A (a protein functionally related to TMED3) in vitro. RESULTS We confirmed the overexpression of TMED3 was correlated with poor prognosis in ESCC patients. When TMED3 was knocked down, ESCC cell proliferation, migration, and invasion were inhibited whereas cell apoptosis was promoted in vitro, and tumorigenicity was inhibited in vivo. We further revealed significant changes in gene expression profile in TMED3 knockdown cells. Among these differentially expressed genes, FAM60A was overexpressed in ESCC tissues. Furthermore, knocking down FAM60A significantly weakened the proliferation ability of ESCC cells and reversed TMED3's tumorigenicity of ESCC cells. CONCLUSION Our study revealed an oncogenic role of TMED3 in ESCC.
Collapse
Affiliation(s)
- Yuxian Yang
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Shiliang Liu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Chunxia Xie
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Qiaoqiao Li
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Tiantian Gao
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Mengzhong Liu
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Mian Xi
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| | - Lei Zhao
- Department of Radiation Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China.
| |
Collapse
|
16
|
Wang RF, Hong YG, Hao LQ, Yu HT. Expression of TMED3 is independently associated with colorectal cancer prognosis. Exp Ther Med 2022; 23:286. [PMID: 35317448 PMCID: PMC8908477 DOI: 10.3892/etm.2022.11215] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 03/24/2021] [Indexed: 12/24/2022] Open
Affiliation(s)
- Rong-Fei Wang
- Department of Tumor, Anus and Intestine, Jinhua People's Hospital, Jinhua, Zhejiang 321000, P.R. China
| | - Yong-Gang Hong
- Department of Colorectal Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R. China
| | - Li-Qiang Hao
- Department of Colorectal Surgery, Changhai Hospital, Second Military Medical University, Shanghai 200433, P.R. China
| | - Hai-Tao Yu
- Department of Oncology, Lanxi People's Hospital, Lanxi, Zhejiang 321100, P.R. China
| |
Collapse
|
17
|
Mendes LFS, Costa-Filho AJ. A gold revision of the Golgi Dynamics (GOLD) domain structure and associated cell functionalities. FEBS Lett 2022; 596:973-990. [PMID: 35099811 DOI: 10.1002/1873-3468.14300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/04/2022] [Accepted: 01/20/2022] [Indexed: 11/06/2022]
Abstract
The classical secretory pathway is the key membrane-based delivery system in eukaryotic cells. Several families of proteins involved in the secretory pathway, with functionalities going from cargo sorting receptors to the maintenance and dynamics of secretory organelles, share soluble globular domains predicted to mediate protein-protein interactions. One of them is "Golgi Dynamics" (GOLD) domain, named after its strong association with the Golgi apparatus. There are many GOLD-containing protein families, such as the Transmembrane emp24 domain-containing proteins (TMED/p24 family), animal SEC14-like proteins, Human Golgi resident protein ACBD3, a splice variant of TICAM2 called TRAM with GOLD domain and FYCO1. Here, we critically review the state-of-the-art knowledge of the structures and functions of the main representatives of GOLD-containing proteins in vertebrates. We provide the first unified description of the GOLD domain structure across different families since the first high-resolution structure was determined. With a brand-new update on the definition of the GOLD domain, we also discuss how its tertiary structure fits the β-sandwich-like fold map and give exciting new directions for forthcoming studies.
Collapse
Affiliation(s)
- Luis Felipe S Mendes
- Laboratório de Biofísica Molecular, Departamento de Física, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| | - Antonio J Costa-Filho
- Laboratório de Biofísica Molecular, Departamento de Física, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, SP, Brasil
| |
Collapse
|
18
|
Xie A, Xu X, Kuang P, Zhang L, Yu F. TMED3 promotes the progression and development of lung squamous cell carcinoma by regulating EZR. Cell Death Dis 2021; 12:804. [PMID: 34429402 PMCID: PMC8385054 DOI: 10.1038/s41419-021-04086-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 07/27/2021] [Accepted: 07/29/2021] [Indexed: 12/23/2022]
Abstract
Lung squamous cell carcinoma (LUSC) has a poor clinical prognosis and lacks effective targeted therapy. The transmembrane emp24 trafficking protein 3 (TMED3) belongs to the TMED family, which is responsible for the transport of intracellular proteins. This study was to explore the clinicopathological significance and biological effects of TMED3 in LUSC. Expression of TMED3 in LUSC was detected by immunohistochemical (IHC). The loss-of-function assays were used to investigate the effects of TMED3 on proliferation, apoptosis, cell cycle, and migration of LUSC cells. The influence of TMED3 knockdown on tumor growth in vivo was evaluated by mice xenograft models. In addition, the downstream target of TMED3 was recognized by RNA sequencing and Ingenuity Pathway Analysis (IPA). Moreover, TMED3 was upregulated in LUSC tissue, which was positively correlated with pathological grade. TMED3 knockdown was involved in the regulation of LUSC cell function, such as inhibition of proliferation, reduction of colony formation, induction of apoptosis, and reduction of migration. TMED3 knockdown induced abnormalities in apoptosis-related proteins in LUSC cells. In addition, the inhibition of cell migration by TMED3 knockdown was achieved by regulating EMT. Mechanically, EZR was considered as a potential target for TMED3 to regulate the progress of LUSC. Inhibition of EZR can inhibit the progression of LUSC, and even reduce the promoting effects of TMED3 overexpression on LUSC. In conclusion, TMED3 promoted the progression and development of LUSC by EZR, which may be a novel therapeutic target for LUSC.
Collapse
Affiliation(s)
- An Xie
- Jiangxi Institute of Urology, The First Affiliated Hospital of Nanchang University, 17 Yong Wai Zheng Street, Nanchang City, Jiangxi Province, China
| | - Xinping Xu
- Jiangxi Institute of Respiratory Disease, The First Affiliated Hospital of Nanchang University, 17 Yong Wai Zheng Street, Nanchang City, Jiangxi Province, China
| | - Peng Kuang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yong Wai Zheng Street, Nanchang City, Jiangxi Province, China
| | - Ling Zhang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yong Wai Zheng Street, Nanchang City, Jiangxi Province, China
| | - Feng Yu
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yong Wai Zheng Street, Nanchang City, Jiangxi Province, China.
| |
Collapse
|
19
|
Yang YC, Chien MH, Lai TC, Tung MC, Jan YH, Chang WM, Jung SM, Chen MH, Yeh CN, Hsiao M. Proteomics-based identification of TMED9 is linked to vascular invasion and poor prognoses in patients with hepatocellular carcinoma. J Biomed Sci 2021; 28:29. [PMID: 33888099 PMCID: PMC8063382 DOI: 10.1186/s12929-021-00727-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 04/14/2021] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Due to the difficulties in early diagnosing and treating hepatocellular carcinoma (HCC), prognoses for patients remained poor in the past decade. In this study, we established a screening model to discover novel prognostic biomarkers in HCC patients. METHODS Candidate biomarkers were screened by liquid chromatography with tandem mass spectrometry (LC-MS/MS) analyses of five HCC normal (N)/tumor (T) paired tissues and preliminarily verified them through several in silico database analyses. Expression levels and functional roles of candidate biomarkers were respectively evaluated by immunohistochemical staining in N/T paired tissue (n = 120) and MTS, colony formation, and transwell migration/invasion assays in HCC cell lines. Associations of clinicopathological features and prognoses with candidate biomarkers in HCC patients were analyzed from GEO and TCGA datasets and our recruited cohort. RESULTS We found that the transmembrane P24 trafficking protein 9 (TMED9) protein was elevated in HCC tissues according to a global proteomic analysis. Higher messenger (m)RNA and protein levels of TMED9 were observed in HCC tissues compared to normal liver tissues or pre-neoplastic lesions. The TMED9 mRNA expression level was significantly associated with an advanced stage and a poor prognosis of overall survival (OS, p = 0.00084) in HCC patients. Moreover, the TMED9 protein expression level was positively correlated with vascular invasion (p = 0.026), OS (p = 0.044), and disease-free survival (p = 0.015) in our recruited Taiwanese cohort. In vitro, manipulation of TMED9 expression in HCC cells significantly affected cell migratory, invasive, proliferative, and colony-forming abilities. CONCLUSIONS Ours is the first work to identify an oncogenic role of TMED9 in HCC cells and may provide insights into the application of TMED9 as a novel predictor of clinical outcomes and a potential therapeutic target in patients with HCC.
Collapse
Affiliation(s)
- Yi-Chieh Yang
- Department of Medical Research, Tungs' Taichung Metro Harbor Hospital, Taichung, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan.,Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan
| | - Ming-Hsien Chien
- Graduate Institute of Clinical Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan. .,Pulmonary Research Center, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan. .,Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, Taipei, Taiwan. .,TMU Research Center of Cancer Translational Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Tsung-Ching Lai
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan.,Division of Pulmonary Medicine, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Min-Che Tung
- Department of Medical Research, Tungs' Taichung Metro Harbor Hospital, Taichung, Taiwan
| | - Yi-Hua Jan
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan
| | - Wei-Ming Chang
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan.,School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei, Taiwan
| | - Shih-Ming Jung
- Department of Pathology, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan
| | - Ming-Huang Chen
- Department of Oncology, Taipei Veterans General Hospital and School of Medicine, National Yang-Ming University, Taipei, 112, Taiwan. .,School of Medicine, National Yang-Ming University, Taipei, Taiwan.
| | - Chun-Nan Yeh
- Department of General Surgery and Liver Research Center, Linkou Branch, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, 333, Taiwan.
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, 128 Academia Road, Section 2, Nankang, Taipei, 11529, Taiwan. .,Department of Biochemistry, Kaohsiung Medical University, Kaohsiung, Taiwan. .,Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
20
|
Guernsey MW, van Kruistum H, Reznick DN, Pollux BJA, Baker JC. Molecular Signatures of Placentation and Secretion Uncovered in Poeciliopsis Maternal Follicles. Mol Biol Evol 2021; 37:2679-2690. [PMID: 32421768 PMCID: PMC7475030 DOI: 10.1093/molbev/msaa121] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Placentation evolved many times independently in vertebrates. Although the core functions of all placentas are similar, we know less about how this similarity extends to the molecular level. Here, we study Poeciliopsis, a unique genus of live-bearing fish that have independently evolved complex placental structures at least three times. The maternal follicle is a key component of these structures. It envelops yolk-rich eggs and is morphologically simple in lecithotrophic species but has elaborate villous structures in matrotrophic species. Through sequencing, the follicle transcriptome of a matrotrophic, Poeciliopsis retropinna, and lecithotrophic, P. turrubarensis, species we found genes known to be critical for placenta function expressed in both species despite their difference in complexity. Additionally, when we compare the transcriptome of different river populations of P. retropinna, known to vary in maternal provisioning, we find differential expression of secretory genes expressed specifically in the top layer of villi cells in the maternal follicle. This provides some of the first evidence that the placental structures of Poeciliopsis function using a secretory mechanism rather than direct contact with maternal circulation. Finally, when we look at the expression of placenta proteins at the maternal–fetal interface of a larger sampling of Poeciliopsis species, we find expression of key maternal and fetal placenta proteins in their cognate tissue types of all species, but follicle expression of prolactin is restricted to only matrotrophic species. Taken together, we suggest that all Poeciliopsis follicles are poised for placenta function but require expression of key genes to form secretory villi.
Collapse
Affiliation(s)
- Michael W Guernsey
- Department of Genetics, Stanford University School of Medicine, Stanford, CA
| | - Henri van Kruistum
- Department of Animal Sciences, Wageningen University, Wageningen, The Netherlands
| | - David N Reznick
- Department of Biology, University of California Riverside, Riverside, CA
| | - Bart J A Pollux
- Department of Animal Sciences, Wageningen University, Wageningen, The Netherlands
| | - Julie C Baker
- Department of Genetics, Stanford University School of Medicine, Stanford, CA
| |
Collapse
|
21
|
Beauchamp MC, Djedid A, Bareke E, Merkuri F, Aber R, Tam AS, Lines MA, Boycott KM, Stirling PC, Fish JL, Majewski J, Jerome-Majewska LA. Mutation in Eftud2 causes craniofacial defects in mice via mis-splicing of Mdm2 and increased P53. Hum Mol Genet 2021; 30:739-757. [PMID: 33601405 DOI: 10.1093/hmg/ddab051] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 02/06/2021] [Accepted: 02/11/2021] [Indexed: 01/19/2023] Open
Abstract
EFTUD2 is mutated in patients with mandibulofacial dysostosis with microcephaly (MFDM). We generated a mutant mouse line with conditional mutation in Eftud2 and used Wnt1-Cre2 to delete it in neural crest cells. Homozygous deletion of Eftud2 causes brain and craniofacial malformations, affecting the same precursors as in MFDM patients. RNAseq analysis of embryonic heads revealed a significant increase in exon skipping and increased levels of an alternatively spliced Mdm2 transcript lacking exon 3. Exon skipping in Mdm2 was also increased in O9-1 mouse neural crest cells after siRNA knock-down of Eftud2 and in MFDM patient cells. Moreover, we found increased nuclear P53, higher expression of P53-target genes and increased cell death. Finally, overactivation of the P53 pathway in Eftud2 knockdown cells was attenuated by overexpression of non-spliced Mdm2, and craniofacial development was improved when Eftud2-mutant embryos were treated with Pifithrin-α, an inhibitor of P53. Thus, our work indicates that the P53-pathway can be targeted to prevent craniofacial abnormalities and shows a previously unknown role for alternative splicing of Mdm2 in the etiology of MFDM.
Collapse
Affiliation(s)
- Marie-Claude Beauchamp
- Research Institute of the McGill University Health Centre at Glen Site, Montreal, QC H4A 3J1, Canada
| | - Anissa Djedid
- Department of Human Genetics, McGill University, Montreal, QC H3A 0G1, Canada
| | - Eric Bareke
- Department of Human Genetics, McGill University, Montreal, QC H3A 0G1, Canada
| | - Fjodor Merkuri
- Department of Biological Sciences, University of Massachusetts Lowell, Lowell, MA 01854, USA
| | - Rachel Aber
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 2B2, Canada
| | - Annie S Tam
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC V5Z 1L3, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| | - Matthew A Lines
- CHEO Research Institute, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - Kym M Boycott
- CHEO Research Institute, University of Ottawa, Ottawa, ON K1H 8L1, Canada
| | - Peter C Stirling
- Terry Fox Laboratory, British Columbia Cancer Agency, Vancouver, BC V5Z 1L3, Canada.,Department of Medical Genetics, University of British Columbia, Vancouver, BC V6H 3N1, Canada
| | - Jennifer L Fish
- Department of Biological Sciences, University of Massachusetts Lowell, Lowell, MA 01854, USA
| | - Jacek Majewski
- Department of Human Genetics, McGill University, Montreal, QC H3A 0G1, Canada
| | - Loydie A Jerome-Majewska
- Research Institute of the McGill University Health Centre at Glen Site, Montreal, QC H4A 3J1, Canada.,Department of Human Genetics, McGill University, Montreal, QC H3A 0G1, Canada.,Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 2B2, Canada.,Department of Pediatrics, McGill University, Montreal, QC H4A 3J1, Canada
| |
Collapse
|
22
|
Singh S, Nguyen HC, Ehsan M, Michels DCR, Singh P, Qadura M, Singh KK. Pravastatin-induced changes in expression of long non-coding and coding RNAs in endothelial cells. Physiol Rep 2021; 9:e14661. [PMID: 33369888 PMCID: PMC7769171 DOI: 10.14814/phy2.14661] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 11/02/2020] [Accepted: 11/06/2020] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVE Atherosclerosis is the main cause of the cardiovascular disease (CVD). Elevated blood cholesterol and inflammation of the endothelium are two major mechanisms contributing to the establishment of atherosclerotic plaques. Statins, such as pravastatin, are blood-cholesterol lowering drugs commonly prescribed for patients with or at risk for CVDs. In addition to lowering blood cholesterols, statins have recently been shown to improve endothelial function in both hyper- and normocholesterolemic patients with atherosclerosis. To understand the molecular mechanisms underlying the endothelial function improvement by statins, we assessed the RNA profile of pravastatin-treated endothelial cells, particularly their mRNAs and long non-coding RNAs (lncRNAs). METHODS Human umbilical vein endothelial cells (HUVECs) treated with pravastatin (10 µM) for 24 hr were profiled for lncRNAs and mRNAs using the Arraystar Human lncRNA Expression Microarray V3.0. RESULTS Of the 30,584 different lncRNAs screened, 95 were significantly upregulated, while 86 were downregulated in HUVECs responding to pravastatin. LINC00281 and BC045663 were the most upregulated (~8-fold) and downregulated (~3.5-fold) lncRNAs, respectively. Of the 26,106 different mRNAs screened in the pravastatin-treated HUVEC samples, 190 were significantly upregulated, while 90 were downregulated. Assigning the differentially expressed genes by bioinformatics into functional groups revealed their molecular signaling involvement in the following physiological processes: osteoclast differentiation, Rap1 signaling pathway, hematopoiesis, immunity, and neurotrophin signaling pathway. CONCLUSIONS This is the first lncRNA and mRNA expression profiling of pravastatin-mediated changes in human endothelial cells. Our results reveal potential novel targets and mechanisms for pravastatin-mediated vascular protection in atherosclerosis.
Collapse
Affiliation(s)
- Shweta Singh
- Department of Chemical and Biochemical EngineeringSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
| | - Hien C. Nguyen
- Department of Medical BiophysicsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
- Department of Anatomy and Cell BiologySchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
| | - Mehroz Ehsan
- Department of Medical BiophysicsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
- Schulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
| | - David C. R. Michels
- Department of Medical BiophysicsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
| | - Priyanka Singh
- Schulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
| | - Mohammad Qadura
- Vascular SurgeryKeenan Research Centre for Biomedical Science and Li Ka Shing Knowledge Institute of St. Michael’s HospitalTorontoONCanada
- Institute of Medical ScienceUniversity of TorontoTorontoONCanada
| | - Krishna K. Singh
- Department of Medical BiophysicsSchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
- Department of Anatomy and Cell BiologySchulich School of Medicine and DentistryUniversity of Western OntarioLondonONCanada
- Institute of Medical ScienceUniversity of TorontoTorontoONCanada
- Pharmacology and ToxicologyUniversity of TorontoTorontoONCanada
| |
Collapse
|
23
|
Ge X, Jiang W, Jiang Y, Lv X, Liu X, Wang X. Expression and Importance of TMED2 in Multiple Myeloma Cells. Cancer Manag Res 2020; 12:12895-12903. [PMID: 33364837 PMCID: PMC7751311 DOI: 10.2147/cmar.s278570] [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: 08/24/2020] [Accepted: 11/20/2020] [Indexed: 11/23/2022] Open
Abstract
Objective TMED2 is a member of the transmembrane emp24 domain (Tmed)/p24 protein family, which is significantly upregulated in breast cancer, ovarian cancer and other tumour tissues. The purpose of this study was to investigate the expression of TMED2 in MM cell lines and its effect on the biological behaviour of MM cell lines. Methods Real-time quantitative PCR (RT-qPCR) was used to detect the expression of TMED2 in MM cell lines, including MM.1S and RPMI 8226 cells, and lentivirus vector-mediated TMED2 gene silencing was used to further study the effect of the downregulation of TMED2 expression on cell viability, the cell cycle, and apoptosis. Results Based on the RT-qPCR results, the expression of the TMED2 mRNA was increased in the MM cell lines MM.1S and RPMI 8226 compared with endogenous control GAPDH. The expression of the TMED2 mRNA was substantially reduced after transfection of the shRNA targeting TMED2 (shTMED2) in both MM cell lines. The CCK-8 assay showed significant decreases in the viability of MM.1S and RPMI 8226 cells, suggesting that the TMED2 gene plays an important role in the proliferation of these two cell lines. The cell cycle of MM.1S and RPMI 8226 cells was substantially altered by shTMED2, as evidenced by the increased number of cells in G1 phase and decreased number of cells in S and G2/M phases. The FACS analysis revealed a significant increase in the apoptosis of MM.1S and RPMI 8226 cells due to the increased activity of Caspase 3/7, suggesting that the TMED2 gene is significantly related to the apoptosis of these two cell lines. Conclusion Based on these results, TMED2 may play an important role in the pathogenesis of MM. This novel study may contribute to further investigations of useful biomarkers and potential therapeutic targets in patients with MM.
Collapse
Affiliation(s)
- Xueling Ge
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, People's Republic of China
| | - Wei Jiang
- Information Center, Shandong Mental Health Center, Jinan, Shandong 250014, People's Republic of China
| | - Yujie Jiang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, People's Republic of China
| | - Xiao Lv
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, People's Republic of China
| | - Xin Liu
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, People's Republic of China
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, People's Republic of China
| |
Collapse
|
24
|
Dvela-Levitt M, Shaw JL, Greka A. A Rare Kidney Disease To Cure Them All? Towards Mechanism-Based Therapies for Proteinopathies. Trends Mol Med 2020; 27:394-409. [PMID: 33341352 DOI: 10.1016/j.molmed.2020.11.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/20/2020] [Accepted: 11/20/2020] [Indexed: 12/30/2022]
Abstract
Autosomal dominant tubulointerstitial kidney diseases (ADTKDs) are a group of rare genetic diseases that lead to kidney failure. Mutations in the MUC1 gene cause ADTKD-MUC1 (MUC1 kidney disease, MKD), a disorder with no available therapies. Recent studies have identified the molecular and cellular mechanisms that drive MKD disease pathogenesis. Armed with patient-derived cell lines and pluripotent stem cell (iPSC)-derived kidney organoids, it was found that MKD is a toxic proteinopathy caused by the intracellular accumulation of misfolded MUC1 protein in the early secretory pathway. We discuss the advantages of studying rare monogenic kidney diseases, describe effective patient-derived model systems, and highlight recent mechanistic insights into protein quality control that have implications for additional proteinopathies beyond rare kidney diseases.
Collapse
Affiliation(s)
- Moran Dvela-Levitt
- The Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA; Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Jillian L Shaw
- The Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA
| | - Anna Greka
- The Broad Institute of Massachusetts Institute of Technology (MIT) and Harvard, Cambridge, MA, USA; Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
25
|
Zhang X, Hao H, Zhuang H, Wang J, Sheng Y, Xu F, Dou J, Chen C, Shen Y. Circular RNA circ_0008305 aggravates hepatocellular carcinoma growth through binding to miR‐186 and inducing TMED2. J Cell Mol Med 2020; 26:1742-1753. [PMID: 33210454 PMCID: PMC8918415 DOI: 10.1111/jcmm.15945] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 09/01/2020] [Accepted: 09/14/2020] [Indexed: 12/17/2022] Open
Abstract
Dysregulation of circRNAs is reported to exert crucial roles in cancers, including hepatocellular carcinoma (HCC). So far, the function of circRNAs in HCC development remains poorly known. Currently, our data showed that circ_0008305 was highly elevated in HCC cell lines and 30 paired tissue samples of HCC. As evidenced, suppression of circ_0008305 repressed HCC cell growth significantly. Meanwhile, up‐regulation of circ_0008305 significantly reduced HCC cell growth. Mechanistically, we displayed that circ_0008305 could bind with miR‐186 by using bioinformatics analysis. miR‐186 has been reported to be a crucial tumour oncogene in many cancers. In addition, we proved miR‐186 was greatly decreased in HCC. The direct correlation between miR‐186 and circ_0008305 was confirmed in our work. In addition, up‐regulation of miR‐186 obviously restrained HCC progression. Increased expression of transmembrane p24 trafficking protein 2 (TMED2) is significantly related to the unfavourable outcomes in cancer patients. At our present work, we proved that TMED2 could act as a direct target of miR‐186. Mechanistically, we demonstrated that circ_0008305 up‐regulated TMED2 expression by sponging miR‐186, which resulted in significantly induced HCC progression in vitro and in vivo. These revealed the significant role of circ_0008305 in HCC progression, which might indicate a new perspective on circRNAs in HCC development.
Collapse
Affiliation(s)
- Xiaoyu Zhang
- Division of Gastrointestinal Surgery Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University Huai'an China
| | - Hui‐Hui Hao
- Department of Pharmacology Jiangsu College of Nursing Huai'an China
| | - Hai‐Wen Zhuang
- Division of Gastrointestinal Surgery Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University Huai'an China
| | - Jian Wang
- Division of Gastrointestinal Surgery Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University Huai'an China
| | - Yu Sheng
- Division of Gastrointestinal Surgery Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University Huai'an China
| | - Fang Xu
- Division of Gastrointestinal Surgery Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University Huai'an China
| | - Jin Dou
- Division of Gastrointestinal Surgery Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University Huai'an China
| | - Chuang Chen
- Division of Hepatobiliary Surgery Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University Huai'an China
| | - Yang Shen
- Operating Room Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University Huai'an China
| |
Collapse
|
26
|
El-Darzi N, Mast N, Petrov AM, Pikuleva IA. 2-Hydroxypropyl-β-cyclodextrin reduces retinal cholesterol in wild-type and Cyp27a1 -/- Cyp46a1 -/- mice with deficiency in the oxysterol production. Br J Pharmacol 2020; 178:3220-3234. [PMID: 32698250 DOI: 10.1111/bph.15209] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 07/14/2020] [Accepted: 07/16/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE 2-Hydroxypropyl-β-cyclodextrin (HPCD) is an FDA approved vehicle for drug delivery and an efficient cholesterol-lowering agent. HPCD was proposed to lower tissue cholesterol via multiple mechanisms including those mediated by oxysterols. CYP27A1 and CYP46A1 are the major oxysterol-producing enzymes in the retina that convert cholesterol to 27- and 24-hydroxycholesterol, respectively. We investigated whether HPCD treatments affected the retina of wild-type and Cyp27a1-/- Cyp46a1-/- mice that do not produce the major retinal oxysterols. EXPERIMENTAL APPROACH HPCD administration was either by i.p., p.o. or s.c. Delivery to the retina was confirmed by angiography using the fluorescently labelled HPCD. Effects on the levels of retinal sterols, mRNA and proteins were evaluated by GC-MS, qRT-PCR and label-free approach, respectively. KEY RESULTS In both wild-type and Cyp27a1-/- Cyp46a1-/- mice, HPCD crossed the blood-retinal barrier when delivered i.p. and lowered the retinal cholesterol content when administered p.o. and s.c. In both genotypes, oral HPCD treatment affected the expression of cholesterol-related genes as well as the proteins involved in endocytosis, lysosomal function and lipid homeostasis. Mechanistically, liver X receptors and the altered expression of Lipe (hormone-sensitive lipase), Nceh1 (neutral cholesterol ester hydrolase 1) and NLTP (non-specific lipid-transfer protein) could mediate some of the HPCD effects. CONCLUSIONS AND IMPLICATIONS HPCD treatment altered retinal cholesterol homeostasis and is a potential therapeutic approach for the reduction of drusen and subretinal drusenoid deposits, cholesterol-rich lesions and hallmarks of age-related macular degeneration. LINKED ARTICLES This article is part of a themed issue on Oxysterols, Lifelong Health and Therapeutics. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.16/issuetoc.
Collapse
Affiliation(s)
- Nicole El-Darzi
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Natalia Mast
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Alexey M Petrov
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Irina A Pikuleva
- Department of Ophthalmology and Visual Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
27
|
Pu Y, Zhang Y, Zhang T, Han J, Ma Y, Liu X. Identification of Novel lncRNAs Differentially Expressed in Placentas of Chinese Ningqiang Pony and Yili Horse Breeds. Animals (Basel) 2020; 10:E119. [PMID: 31940795 PMCID: PMC7022612 DOI: 10.3390/ani10010119] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/30/2019] [Accepted: 01/09/2020] [Indexed: 12/24/2022] Open
Abstract
As a nutrient sensor, the placenta plays a key role in regulating fetus growth and development. Long non-coding RNAs (lncRNAs) have been shown to regulate growth-related traits. However, the biological function of lncRNAs in horse placentas remains unclear. To compare the expression patterns of lncRNAs in the placentas of the Chinese Ningqiang (NQ) and Yili (YL) breeds, we performed a transcriptome analysis using RNA sequencing (RNA-seq) technology. NQ is a pony breed with an average adult height at the withers of less than 106 cm, whereas that of YL is around 148 cm. Based on 813 million high-quality reads and stringent quality control procedures, 3011 transcripts coding for 1464 placental lncRNAs were identified and mapped to the horse reference genome. We found 107 differentially expressed lncRNAs (DELs) between NQ and YL, including 68 up-regulated and 39 down-regulated DELs in YL. Six (TBX3, CACNA1F, EDN3, KAT5, ZNF281, TMED2, and TGFB1) out of the 233 genes targeted by DELs were identified as being involved in limb development, skeletal myoblast differentiation, and embryo development. Two DELs were predicted to target the TBX3 gene, which was found to be under strong selection and associated with small body size in the Chinese Debao pony breed. This finding suggests the potential functional significance of placental lncRNAs in regulating horse body size.
Collapse
Affiliation(s)
- Yabin Pu
- Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China; (Y.P.); (Y.Z.)
- CAAS-ILRI Joint Laboratory on Livestock and Forage Genetic Resources, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China;
| | - Yanli Zhang
- Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China; (Y.P.); (Y.Z.)
- CAAS-ILRI Joint Laboratory on Livestock and Forage Genetic Resources, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China;
| | - Tian Zhang
- State Key Laboratory of Dao-di Herbs, National Resource Center for Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China;
| | - Jianlin Han
- CAAS-ILRI Joint Laboratory on Livestock and Forage Genetic Resources, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China;
- International Livestock Research Institute (ILRI), Nairobi 00100, Kenya
| | - Yuehui Ma
- Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China; (Y.P.); (Y.Z.)
- CAAS-ILRI Joint Laboratory on Livestock and Forage Genetic Resources, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China;
| | - Xuexue Liu
- Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China; (Y.P.); (Y.Z.)
- CAAS-ILRI Joint Laboratory on Livestock and Forage Genetic Resources, Institute of Animal Science, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China;
| |
Collapse
|
28
|
Abstract
Regulated transport through the secretory pathway is essential for embryonic development and homeostasis. Disruptions in this process impact cell fate, differentiation and survival, often resulting in abnormalities in morphogenesis and in disease. Several congenital malformations are caused by mutations in genes coding for proteins that regulate cargo protein transport in the secretory pathway. The severity of mutant phenotypes and the unclear aetiology of transport protein-associated pathologies have motivated research on the regulation and mechanisms through which these proteins contribute to morphogenesis. This review focuses on the role of the p24/transmembrane emp24 domain (TMED) family of cargo receptors in development and disease.
Collapse
|
29
|
Sun MS, Zhang J, Jiang LQ, Pan YX, Tan JY, Yu F, Guo L, Yin L, Shen C, Shu HB, Liu Y. TMED2 Potentiates Cellular IFN Responses to DNA Viruses by Reinforcing MITA Dimerization and Facilitating Its Trafficking. Cell Rep 2019; 25:3086-3098.e3. [PMID: 30540941 DOI: 10.1016/j.celrep.2018.11.048] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 10/22/2018] [Accepted: 11/12/2018] [Indexed: 02/06/2023] Open
Abstract
Mediator of IRF3 activation (MITA), also known as stimulator of interferon genes (STING), plays a vital role in the innate immune responses to cytosolic dsDNA. The trafficking of MITA from the ER to perinuclear vesicles is necessary for its activation of the downstream molecules, which lead to the production of interferons and pro-inflammatory cytokines. However, the exact mechanism of MITA activation remains elusive. Here, we report that transmembrane emp24 protein transport domain containing 2 (TMED2) potentiates DNA virus-induced MITA signaling. The suppression or deletion of TMED2 markedly impairs the production of type I IFNs upon HSV-1 infection. TMED2-deficient cells harbor greater HSV-1 load than the control cells. Mechanistically, TMED2 associates with MITA only upon viral stimulation, and this process potentiates MITA activation by reinforcing its dimerization and facilitating its trafficking. These findings suggest an essential role of TMED2 in cellular IFN responses to DNA viruses.
Collapse
Affiliation(s)
- Ming-Shun Sun
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Jie Zhang
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Li-Qun Jiang
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yi-Xi Pan
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Jiao-Yi Tan
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Fang Yu
- Department of Pathology, Zhongnan Hospital of Wuhan University, Wuhan 430072, China
| | - Lin Guo
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Lei Yin
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Chao Shen
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Hong-Bing Shu
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China; Medical Research Institute of Wuhan University, Wuhan University, Wuhan 430072, China
| | - Yu Liu
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| |
Collapse
|
30
|
Mishra S, Bernal C, Silvano M, Anand S, Ruiz i Altaba A. The protein secretion modulator TMED9 drives CNIH4/TGFα/GLI signaling opposing TMED3-WNT-TCF to promote colon cancer metastases. Oncogene 2019; 38:5817-5837. [PMID: 31253868 PMCID: PMC6755966 DOI: 10.1038/s41388-019-0845-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 03/13/2019] [Accepted: 05/14/2019] [Indexed: 12/13/2022]
Abstract
How cells in primary tumors initially become pro-metastatic is not understood. A previous genome-wide RNAi screen uncovered colon cancer metastatic suppressor and WNT promoting functions of TMED3, a member of the p24 ER-to-Golgi protein secretion family. Repression of canonical WNT signaling upon knockdown (kd) of TMED3 might thus be sufficient to drive metastases. However, searching for transcriptional influences on other family members here we find that TMED3 kd leads to enhanced TMED9, that TMED9 acts downstream of TMED3 and that TMED9 kd compromises metastasis. Importantly, TMED9 pro-metastatic function is linked to but distinct from the repression of TMED3-WNT-TCF signaling. Functional rescue of the migratory deficiency of TMED9 kd cells identifies TGFα as a mediator of TMED9 pro-metastatic activity. Moreover, TMED9 kd compromises the biogenesis, and thus function, of TGFα. Analyses in three colon cancer cell types highlight a TMED9-dependent gene set that includes CNIH4, a member of the CORNICHON family of TGFα exporters. Our data indicate that TGFA and CNIH4, which display predictive value for disease-free survival, promote colon cancer cell metastatic behavior, and suggest that TMED9 pro-metastatic function involves the modulation of the secretion of TGFα ligand. Finally, TMED9/TMED3 antagonism impacts WNT-TCF and GLI signaling, where TMED9 primacy over TMED3 leads to the establishment of a positive feedback loop together with CNIH4, TGFα, and GLI1 that enhances metastases. We propose that primary colon cancer cells can transition between two states characterized by secretion-transcription regulatory loops gated by TMED3 and TMED9 that modulate their metastatic proclivities.
Collapse
Affiliation(s)
- Sonakshi Mishra
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva Medical School, 1 rue Michel Servet, CH1211, Geneva, Switzerland
| | - Carolina Bernal
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva Medical School, 1 rue Michel Servet, CH1211, Geneva, Switzerland
| | - Marianna Silvano
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva Medical School, 1 rue Michel Servet, CH1211, Geneva, Switzerland
| | - Santosh Anand
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva Medical School, 1 rue Michel Servet, CH1211, Geneva, Switzerland
| | - Ariel Ruiz i Altaba
- Department of Genetic Medicine and Development, Faculty of Medicine, University of Geneva Medical School, 1 rue Michel Servet, CH1211, Geneva, Switzerland.
| |
Collapse
|
31
|
Ha M, Moon H, Choi D, Kang W, Kim JH, Lee KJ, Park D, Kang CD, Oh SO, Han ME, Kim YH, Lee D. Prognostic Role of TMED3 in Clear Cell Renal Cell Carcinoma: A Retrospective Multi-Cohort Analysis. Front Genet 2019; 10:355. [PMID: 31057605 PMCID: PMC6478656 DOI: 10.3389/fgene.2019.00355] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 04/02/2019] [Indexed: 12/02/2022] Open
Abstract
Transmembrane p24 trafficking protein 3 (TMED3) is a metastatic suppressor in colon cancer and hepatocellular carcinoma. However, its function in the progression of clear cell renal cell carcinoma (ccRCC) is unknown. Here, we report that TMED3 could be a new prognostic marker for ccRCC. Patient data were extracted from cohorts in the Cancer Genome Atlas (TCGA) and the International Cancer Genome Consortium (ICGC). Differential expression of TMED3 was observed between the low stage (Stage I and II) and high stage (Stage III and IV) patients in the TCGA and ICGC cohorts and between the low grade (Grade I and II) and high grade (Grade III and IV) patients in the TCGA cohort. Further, we evaluated TMED3 expression as a prognostic gene using Kaplan-Meier survival analysis, multivariate analysis, the time-dependent area under the curve (AUC) of Uno’s C-index, and the AUC of the receiver operating characteristics at 5 years. The Kaplan-Meier analysis revealed that TMED3 overexpression was associated with poor prognosis for ccRCC patients. Analysis of the C-indices and area under the receiver operating characteristic curve further supported this. Multivariate analysis confirmed the prognostic significance of TMED3 expression levels (P = 0.005 and 0.006 for TCGA and ICGC, respectively). Taken together, these findings demonstrate that TMED3 is a potential prognostic factor for ccRCC.
Collapse
Affiliation(s)
- Mihyang Ha
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, South Korea
| | - Hwan Moon
- Department of Premedicine, School of Medicine, Pusan National University, Yangsan, South Korea
| | - Dongwook Choi
- Division of Drug Process Development, New Drug Development Center, Osong Medical Innovation Foundation, Cheongju, South Korea
| | - Wonmo Kang
- Division of Drug Process Development, New Drug Development Center, Osong Medical Innovation Foundation, Cheongju, South Korea
| | - Ji-Hong Kim
- Division of Drug Process Development, New Drug Development Center, Osong Medical Innovation Foundation, Cheongju, South Korea
| | - Keon Jin Lee
- Division of Drug Process Development, New Drug Development Center, Osong Medical Innovation Foundation, Cheongju, South Korea
| | - Dongsu Park
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States.,Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, United States.,Center for Skeletal Medicine and Biology, Baylor College of Medicine, Houston, TX, United States
| | - Chi-Dug Kang
- Department of Biochemistry, Pusan National University School of Medicine, Yangsan, South Korea.,Department of Convergence Medical Science, Pusan National University School of Medicine, Yangsan, South Korea
| | - Sae-Ock Oh
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, South Korea
| | - Myoung-Eun Han
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan, South Korea
| | - Yun Hak Kim
- Department of Anatomy, Biomedical Research Institute, School of Medicine, Pusan National University, Yangsan, South Korea.,Department of Biomedical Informatics, Biomedical Research Institute, School of Medicine, Pusan National University, Yangsan, South Korea
| | - Dongjun Lee
- Department of Convergence Medical Science, Pusan National University School of Medicine, Yangsan, South Korea
| |
Collapse
|
32
|
Hou W, Jerome-Majewska LA. TMED2/emp24 is required in both the chorion and the allantois for placental labyrinth layer development. Dev Biol 2018; 444:20-32. [PMID: 30236446 DOI: 10.1016/j.ydbio.2018.09.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 09/10/2018] [Accepted: 09/10/2018] [Indexed: 01/08/2023]
Abstract
TMED2, a member of the transmembrane emp24 domain (TMED) family, is required for transport of cargo proteins between the ER and Golgi. TMED2 is also important for normal morphogenesis of mouse embryos and their associated placenta, and in fact Tmed2 homozygous mutant embryos arrest at mid-gestation due to a failure of placental labyrinth layer formation. Differentiation of the placental labyrinth layer depends on chorioallantoic attachment (contact between the chorion and allantois), and branching morphogenesis (mingling of cells from these two tissues). Since Tmed2 mRNA was found in both the chorion and allantois, and 50% of Tmed2 homozygous mutant embryos failed to undergo chorioallantoic attachment, the tissue-specific requirement of Tmed2 during placental labyrinth layer formation remained a mystery. Herein, we report differential localization of TMED2 protein in the chorion and allantois, abnormal ER retention of Fibronectin in Tmed2 homozygous mutant allantoises and cell-autonomous requirement for Tmed2 in the chorion for chorioallantoic attachment and fusion. Using an ex vivo model of explanted chorions and allantoises, we showed that chorioallantoic attachment failed to occur in 50% of samples when homozygous mutant chorions were recombined with wild type allantoises. Furthermore, though expression of genes associated with trophoblast differentiation was maintained in Tmed2 mutant chorions with chorioallantoic attachment, expression of these genes was attenuated. In addition, Tmed2 homozygous mutant allantoises could undergo branching morphogenesis, however the region of mixing between mutant and wild type cells was reduced, and expression of genes associated with trophoblast differentiation was also attenuated. Our data also suggest that Fibronectin is a cargo protein of TMED2 and indicates that Tmed2 is required cell-autonomously and non-autonomously in the chorion and the allantois for placental labyrinth layer formation.
Collapse
Affiliation(s)
- Wenyang Hou
- Department of Human Genetics, McGill University, 1205 Avenue Docteur Penfield, N5/13, Montreal, QC, Canada
| | - Loydie A Jerome-Majewska
- Department of Human Genetics, McGill University, 1205 Avenue Docteur Penfield, N5/13, Montreal, QC, Canada; Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada; Department of Pediatrics, McGill University, 1001 Decarie Blvd, EM02210, Montreal, QC, Canada H4A 3J1; McGill University Health Centre Glen Site, 1001 Decarie Blvd, EM0.2210, Montreal, QC, Canada H4A 3J1.
| |
Collapse
|
33
|
Hall E, Dekker Nitert M, Volkov P, Malmgren S, Mulder H, Bacos K, Ling C. The effects of high glucose exposure on global gene expression and DNA methylation in human pancreatic islets. Mol Cell Endocrinol 2018; 472:57-67. [PMID: 29183809 DOI: 10.1016/j.mce.2017.11.019] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 10/20/2017] [Accepted: 11/23/2017] [Indexed: 01/02/2023]
Abstract
BACKGROUND Type 2 diabetes (T2D) is a complex disease characterised by chronic hyperglycaemia. The effects of elevated glucose on global gene expression in combination with DNA methylation patterns have not yet been studied in human pancreatic islets. Our aim was to study the impact of 48 h exposure to high (19 mM) versus control (5.6 mM) glucose levels on glucose-stimulated insulin secretion, gene expression and DNA methylation in human pancreatic islets. RESULTS While islets kept at 5.6 mM glucose secreted significantly more insulin in response to short term glucose-stimulation (p = 0.0067), islets exposed to high glucose for 48 h were desensitised and unresponsive to short term glucose-stimulation with respect to insulin secretion (p = 0.32). Moreover, the exposure of human islets to 19 mM glucose resulted in significantly altered expression of eight genes (FDR<5%), with five of these (GLRA1, RASD1, VAC14, SLCO5A1, CHRNA5) also exhibiting changes in DNA methylation (p < 0.05). A gene set enrichment analysis of the expression data showed significant enrichment of e.g. TGF-beta signalling pathway, Notch signalling pathway and SNARE interactions in vesicular transport; these pathways are of relevance for islet function and possibly also diabetes. We also found increased DNA methylation of CpG sites annotated to PDX1 in human islets exposed to 19 mM glucose for 48 h. Finally, we could functionally validate a role for Glra1 in insulin secretion. CONCLUSION Our data demonstrate that high glucose levels affect human pancreatic islet gene expression and several of these genes also exhibit epigenetic changes. This might contribute to the impaired insulin secretion seen in T2D.
Collapse
Affiliation(s)
- Elin Hall
- Epigenetics and Diabetes, Lund University Diabetes Centre (LUDC), Box 50332, 20213 Malmö, Sweden
| | - Marloes Dekker Nitert
- School of Medicine, Royal Brisbane Clinical School, The University of Queensland, Herston Qld 4029, Australia
| | - Petr Volkov
- Epigenetics and Diabetes, Lund University Diabetes Centre (LUDC), Box 50332, 20213 Malmö, Sweden
| | - Siri Malmgren
- Epigenetics and Diabetes, Lund University Diabetes Centre (LUDC), Box 50332, 20213 Malmö, Sweden; Molecular Metabolism, Lund University Diabetes Centre (LUDC), Box 50332, 20213 Malmö, Sweden
| | - Hindrik Mulder
- Molecular Metabolism, Lund University Diabetes Centre (LUDC), Box 50332, 20213 Malmö, Sweden
| | - Karl Bacos
- Epigenetics and Diabetes, Lund University Diabetes Centre (LUDC), Box 50332, 20213 Malmö, Sweden
| | - Charlotte Ling
- Epigenetics and Diabetes, Lund University Diabetes Centre (LUDC), Box 50332, 20213 Malmö, Sweden.
| |
Collapse
|
34
|
Pastor-Cantizano N, Bernat-Silvestre C, Marcote MJ, Aniento F. Loss of Arabidopsis p24 function affects ERD2 trafficking and Golgi structure, and activates the unfolded protein response. J Cell Sci 2018; 131:jcs.203802. [PMID: 28871045 DOI: 10.1242/jcs.203802] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 08/30/2017] [Indexed: 01/22/2023] Open
Abstract
The p24 family of proteins (also known as the TMED family) are key regulators of protein trafficking along the secretory pathway, but very little is known about their functions in plants. A quadruple loss-of-function mutant affecting the p24 genes from the δ-1 subclass of the p24δ subfamily (p24δ3δ4δ5δ6) showed alterations in the Golgi, suggesting that these p24 proteins play a role in the organization of the compartments of the early secretory pathway in Arabidopsis Loss of p24δ-1 proteins also induced the accumulation of the K/HDEL receptor ERD2a (ER lumen protein-retaining receptor A) at the Golgi and increased secretion of BiP family proteins, ER chaperones containing an HDEL signal, probably due to an inhibition of COPI-dependent Golgi-to-ER transport of ERD2a and thus retrieval of K/HDEL ligands. Although the p24δ3δ4δ5δ6 mutant showed enhanced sensitivity to salt stress, it did not show obvious phenotypic alterations under standard growth conditions. Interestingly, this mutant showed a constitutive activation of the unfolded protein response (UPR) and the transcriptional upregulation of the COPII subunit gene SEC31A, which may help the plant to cope with the transport defects seen in the absence of p24 proteins.
Collapse
Affiliation(s)
- Noelia Pastor-Cantizano
- Departamento de Bioquímica y Biología Molecular, Estructura de Recerca Interdisciplinar en Biotecnología i Biomedicina (ERI BIOTECMED), Facultat de Farmacia, Universitat de València, E-46100 Burjassot (Valencia), Spain
| | - Cesar Bernat-Silvestre
- Departamento de Bioquímica y Biología Molecular, Estructura de Recerca Interdisciplinar en Biotecnología i Biomedicina (ERI BIOTECMED), Facultat de Farmacia, Universitat de València, E-46100 Burjassot (Valencia), Spain
| | - María Jesús Marcote
- Departamento de Bioquímica y Biología Molecular, Estructura de Recerca Interdisciplinar en Biotecnología i Biomedicina (ERI BIOTECMED), Facultat de Farmacia, Universitat de València, E-46100 Burjassot (Valencia), Spain
| | - Fernando Aniento
- Departamento de Bioquímica y Biología Molecular, Estructura de Recerca Interdisciplinar en Biotecnología i Biomedicina (ERI BIOTECMED), Facultat de Farmacia, Universitat de València, E-46100 Burjassot (Valencia), Spain
| |
Collapse
|
35
|
Giudice J, Loehr JA, Rodney GG, Cooper TA. Alternative Splicing of Four Trafficking Genes Regulates Myofiber Structure and Skeletal Muscle Physiology. Cell Rep 2017; 17:1923-1933. [PMID: 27851958 DOI: 10.1016/j.celrep.2016.10.072] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2016] [Revised: 09/02/2016] [Accepted: 10/20/2016] [Indexed: 11/16/2022] Open
Abstract
During development, transcriptional and post-transcriptional networks are coordinately regulated to drive organ maturation. Alternative splicing contributes by producing temporal-specific protein isoforms. We previously found that genes undergoing splicing transitions during mouse postnatal heart development are enriched for vesicular trafficking and membrane dynamics functions. Here, we show that adult trafficking isoforms are also expressed in adult skeletal muscle and hypothesize that striated muscle utilizes alternative splicing to generate specific isoforms required for function of adult tissue. We deliver morpholinos into flexor digitorum brevis muscles in adult mice to redirect splicing of four trafficking genes to the fetal isoforms. The splicing switch results in multiple structural and functional defects, including transverse tubule (T-tubule) disruption and dihydropyridine receptor alpha (DHPR) and Ryr1 mislocalization, impairing excitation-contraction coupling, calcium handling, and force generation. The results demonstrate a previously unrecognized role for trafficking functions in adult muscle tissue homeostasis and a specific requirement for the adult splice variants.
Collapse
Affiliation(s)
- Jimena Giudice
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Cell Biology and Physiology, School of Medicine, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; McAllister Heart Institute, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; Curriculum in Genetics and Molecular Biology, The University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.
| | - James A Loehr
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - George G Rodney
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Thomas A Cooper
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular & Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
36
|
Novel endogenous retrovirus-derived transcript expressed in the bovine placenta is regulated by WNT signaling. Biochem J 2017; 474:3499-3512. [PMID: 28899944 PMCID: PMC5633919 DOI: 10.1042/bcj20170531] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 08/22/2017] [Accepted: 09/07/2017] [Indexed: 01/11/2023]
Abstract
Endogenous retroviruses (ERVs) are involved in placentation; perhaps, the most well-known ERVs are the syncytins, actively transcribed env genes involved in cell–cell fusion and possible morphological variations. However, ERVs other than syncytins that play an important role in placental development have not been well characterized. To identify ERV genes expressed during the onset of placentation in the bovine species, we characterized the expression profiles of bovine conceptus transcripts during the peri-attachment period using RNA-seq analysis, and confirming some candidates through real-time PCR. Using in silico and PCR analyses, we identified a novel ERV proviral sequence derived from a gag region, designated bovine endogenous retroviruses (BERV)-K3, containing Gag_p10 and Gag_p24, zinc finger domain. Initial expression of this ERV in bovine conceptuses was on day 20 (day 0 = day of estrus), soon after conceptus attachment to the endometrial epithelium, and its high placental expression was maintained up to the middle of pregnancy. The BERV-K3 transcript was also found in the uterine luminal and glandular epithelia, liver, kidney, intestine, and skin. BERV-K3 is located on chromosome 7 and integrated within LOC100848658, from which noncoding RNA could be transcribed. Furthermore, the expression of endogenous BERV-K3 in bovine trophoblast cell lines was induced by a WNT agonist, a signaling system common to genes expressed in placentas. These data support the argument that during the evolutionary process, mammals incorporated not only similar ERV sequences, but also ERVs unique to individual species. BERV-K3 is in the latter case, likely providing functions unique to ruminant gestation.
Collapse
|
37
|
Abstract
TMED2 is involved in morphogenesis of the mouse embryo and placenta. We found that expression of TMED2 was higher in epithelial ovarian cancer tissues than normal ovarian tissues. Silencing TMED2 decreased cell proliferation, migration, and invasion. Ectopic expression of TMED2 increased cell proliferation, migration and invasion. Silencing TMED2 inhibited ovarian cancer growth in mice. Silencing TMED2 inhibited IGF2/IGF1R/PI3K/Akt pathway. In agreement, ectopically expressed TMED2 activated IGF2/IGF1R/PI3K/Akt pathway. Mechanistic study revealed that TMED2 directly binds to AKT2, thereby facilitating its phosphorylation. We also found that TMED2 increased IGF1R expression by competing for miR-30a. Thus, TMED2 is oncogenic and a potential target for epithelial ovarian cancer therapy.
Collapse
|
38
|
Hou W, Gupta S, Beauchamp MC, Yuan L, Jerome-Majewska LA. Non-alcoholic fatty liver disease in mice with heterozygous mutation in TMED2. PLoS One 2017; 12:e0182995. [PMID: 28797121 PMCID: PMC5552249 DOI: 10.1371/journal.pone.0182995] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Accepted: 07/27/2017] [Indexed: 12/30/2022] Open
Abstract
The transmembrane emp24 domain/p24 (TMED) family are essential components of the vesicular transport machinery. Members of the TMED family serve as cargo receptors implicated in selection and packaging of endoplasmic reticulum (ER) luminal proteins into coatomer (COP) II coated vesicles for anterograde transport to the Golgi. Deletion or mutations of Tmed genes in yeast and Drosophila results in ER-stress and activation of the unfolded protein response (UPR). The UPR leads to expression of genes and proteins important for expanding the folding capacity of the ER, degrading misfolded proteins, and reducing the load of new proteins entering the ER. The UPR is activated in non-alcoholic fatty liver disease (NAFLD) in human and mouse and may contribute to the development and the progression of NAFLD. Tmed2, the sole member of the vertebrate Tmed β subfamily, exhibits tissue and temporal specific patterns of expression in embryos and developing placenta but is ubiquitously expressed in all adult organs. We previously identified a single point mutation, the 99J mutation, in the signal sequence of Tmed2 in an N-ethyl-N-nitrosourea (ENU) mutagenesis screen. Histological and molecular analysis of livers from heterozygous mice carrying the 99J mutation, Tmed299J/+, revealed a requirement for TMED2 in liver health. We show that Tmed299J/+ mice had decreased levels of TMED2 and TMED10, dilated endoplasmic reticulum membrane, and increased phosphorylation of eIF2α, indicating ER-stress and activation of the UPR. Increased expression of Srebp1a and 2 at the newborn stage and increased incidence of NAFLD were also found in Tmed299J/+ mice. Our data establishes Tmed299J/+ mice as a novel mouse model for NAFLD and supports a role for TMED2 in liver health.
Collapse
Affiliation(s)
- Wenyang Hou
- Department of Human Genetics, McGill University, Montreal, Québec, Canada
| | - Swati Gupta
- Department of Human Genetics, McGill University, Montreal, Québec, Canada
| | | | - Libin Yuan
- Department of Human Genetics, McGill University, Montreal, Québec, Canada
| | - Loydie A. Jerome-Majewska
- Department of Human Genetics, McGill University, Montreal, Québec, Canada
- Department of Pediatrics, McGill University Health Centre Glen Site, Montreal, Québec, Canada
- Department of Anatomy and Cell Biology, McGill University, Strathcona Anatomy and Dentistry Building, Québec, Canada
- * E-mail:
| |
Collapse
|
39
|
Zheng H, Yang Y, Han J, Jiang WH, Chen C, Wang MC, Gao R, Li S, Tian T, Wang J, Ma LJ, Ren H, Zhou WP. TMED3 promotes hepatocellular carcinoma progression via IL-11/STAT3 signaling. Sci Rep 2016; 6:37070. [PMID: 27901021 PMCID: PMC5128793 DOI: 10.1038/srep37070] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Accepted: 10/24/2016] [Indexed: 02/06/2023] Open
Abstract
Transmembrane p24 trafficking protein 3(TMED3) is a metastatic suppressor in colon cancer, but its function in the progression of hepatocellular carcinoma (HCC) is unknown. Here, we report that TMED3 was up-regulated in HCC and portal vein tumor thrombus. TMED3 up-regulation in HCC was significantly correlated with aggressive characteristics and predicted poor prognosis in HCC patients. TMED3 overexpression in HCC cell lines promoted cell migration and invasion. In contrast, TMED3 knockdown suppressed HCC metastasis both in vitro and in vivo. Gene microarray analysis revealed decreased IL-11 expression in TMED3-knockdown cells. We propose that TMED3 promotes HCC metastasis through IL-11/STAT3 signaling. Taken together, these findings demonstrate that TMED3 promotes HCC metastasis and is a potential prognostic biomarker in HCC.
Collapse
Affiliation(s)
- Hao Zheng
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai 200438, China
| | - Yuan Yang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai 200438, China.,Department of Health Statistics, Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, China
| | - Jun Han
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai 200438, China.,Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, China
| | - Wei-Hua Jiang
- Department of Oncology, Shanghai Tongren Hospital, Shanghai Jiaotong University, 1111 Xianxia Road, Shanghai 200336, China
| | - Cheng Chen
- Department of Medical Oncology, Jinling Hospital, 305 Zhongshan East Road, Nanjing, Jiangsu 210000, China
| | - Meng-Chao Wang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai 200438, China
| | - Rong Gao
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, China
| | - Shuai Li
- Department of Computer Science, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY, 12180, United States
| | - Tao Tian
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai 200438, China
| | - Jian Wang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai 200438, China
| | - Li-Jun Ma
- Department of Oncology, Shanghai Tongren Hospital, Shanghai Jiaotong University, 1111 Xianxia Road, Shanghai 200336, China
| | - Hao Ren
- Department of Microbiology, Shanghai Key Laboratory of Medical Biodefense, Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, China
| | - Wei-Ping Zhou
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, 225 Changhai Road, Shanghai 200438, China
| |
Collapse
|
40
|
Hou W, Sarikaya DP, Jerome-Majewska LA. Ex vivo culture of pre-placental tissues reveals that the allantois is required for maintained expression of Gcm1 and Tpbpα. Placenta 2016; 47:12-23. [DOI: 10.1016/j.placenta.2016.08.091] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 08/05/2016] [Accepted: 08/29/2016] [Indexed: 11/17/2022]
|
41
|
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
|
42
|
Gupta S, Fahiminiya S, Wang T, Dempsey Nunez L, Rosenblatt DS, Gibson WT, Gilfix B, Bergeron JJM, Jerome-Majewska LA. Somatic overgrowth associated with homozygous mutations in both MAN1B1 and SEC23A. Cold Spring Harb Mol Case Stud 2016; 2:a000737. [PMID: 27148587 PMCID: PMC4853519 DOI: 10.1101/mcs.a000737] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Using whole-exome sequencing, we identified homozygous mutations in two unlinked genes, SEC23A c.1200G>C (p.M400I) and MAN1B1 c.1000C>T (p.R334C), associated with congenital birth defects in two patients from a consanguineous family. Patients presented with carbohydrate-deficient transferrin, tall stature, obesity, macrocephaly, and maloccluded teeth. The parents were healthy heterozygous carriers for both mutations and an unaffected sibling with tall stature carried the heterozygous mutation in SEC23A only. Mutations in SEC23A are responsible for craniolenticosultura dysplasia (CLSD). CLSD patients are short, have late-closing fontanels, and have reduced procollagen (pro-COL1A1) secretion because of abnormal pro-COL1A1 retention in the endoplasmic reticulum (ER). The mutation we identified in MAN1B1 was previously associated with reduced MAN1B1 protein and congenital disorders of glycosylation (CDG). CDG patients are also short, are obese, and have abnormal glycan remodeling. Molecular analysis of fibroblasts from the family revealed normal levels of SEC23A in all cells and reduced levels of MAN1B1 in cells with heterozygous or homozygous mutations in SEC23A and MAN1B1. Secretion of pro-COL1A1 was increased in fibroblasts from the siblings and patients, and pro-COL1A1 was retained in Golgi of heterozygous and homozygous mutant cells, although intracellular pro-COL1A1 was increased in patient fibroblasts only. We postulate that increased pro-COL1A1 secretion is responsible for tall stature in these patients and an unaffected sibling, and not previously discovered in patients with mutations in either SEC23A or MAN1B1. The patients in this study share biochemical and cellular characteristics consistent with mutations in MAN1B1 and SEC23A, indicating a digenic disease.
Collapse
Affiliation(s)
- Swati Gupta
- Department of Human Genetics, McGill University, Montreal, Quebec H3A 1B1, Canada
| | - Somayyeh Fahiminiya
- Department of Human Genetics, McGill University, Montreal, Quebec H3A 1B1, Canada
| | - Tracy Wang
- Department of Human Genetics, McGill University, Montreal, Quebec H3A 1B1, Canada
| | - Laura Dempsey Nunez
- Department of Human Genetics, McGill University, Montreal, Quebec H3A 1B1, Canada
| | - David S Rosenblatt
- Department of Human Genetics, McGill University, Montreal, Quebec H3A 1B1, Canada;; Department of Pediatrics, McGill University, Montreal, Quebec H4A 3J1, Canada
| | - William T Gibson
- Department of Medical Genetics, Child and Family Research Institute, Vancouver, British Columbia V6H 3V4, Canada
| | - Brian Gilfix
- Department of Medicine, McGill University, Montreal, Quebec H4A 3J1, Canada
| | - John J M Bergeron
- Department of Medicine, McGill University, Montreal, Quebec H4A 3J1, Canada
| | - Loydie A Jerome-Majewska
- Department of Human Genetics, McGill University, Montreal, Quebec H3A 1B1, Canada;; Department of Pediatrics, McGill University, Montreal, Quebec H4A 3J1, Canada;; Department of Anatomy and Cell Biology, McGill University, Montreal, Quebec H3A 0C7, Canada
| |
Collapse
|
43
|
Bickhart DM, Xu L, Hutchison JL, Cole JB, Null DJ, Schroeder SG, Song J, Garcia JF, Sonstegard TS, Van Tassell CP, Schnabel RD, Taylor JF, Lewin HA, Liu GE. Diversity and population-genetic properties of copy number variations and multicopy genes in cattle. DNA Res 2016; 23:253-62. [PMID: 27085184 PMCID: PMC4909312 DOI: 10.1093/dnares/dsw013] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 02/29/2016] [Indexed: 11/14/2022] Open
Abstract
The diversity and population genetics of copy number variation (CNV) in domesticated animals are not well understood. In this study, we analysed 75 genomes of major taurine and indicine cattle breeds (including Angus, Brahman, Gir, Holstein, Jersey, Limousin, Nelore, and Romagnola), sequenced to 11-fold coverage to identify 1,853 non-redundant CNV regions. Supported by high validation rates in array comparative genomic hybridization (CGH) and qPCR experiments, these CNV regions accounted for 3.1% (87.5 Mb) of the cattle reference genome, representing a significant increase over previous estimates of the area of the genome that is copy number variable (∼2%). Further population genetics and evolutionary genomics analyses based on these CNVs revealed the population structures of the cattle taurine and indicine breeds and uncovered potential diversely selected CNVs near important functional genes, including AOX1, ASZ1, GAT, GLYAT, and KRTAP9-1. Additionally, 121 CNV gene regions were found to be either breed specific or differentially variable across breeds, such as RICTOR in dairy breeds and PNPLA3 in beef breeds. In contrast, clusters of the PRP and PAG genes were found to be duplicated in all sequenced animals, suggesting that subfunctionalization, neofunctionalization, or overdominance play roles in diversifying those fertility-related genes. These CNV results provide a new glimpse into the diverse selection histories of cattle breeds and a basis for correlating structural variation with complex traits in the future.
Collapse
Affiliation(s)
- Derek M Bickhart
- USDA-ARS, Animal Genomics and Improvement Laboratory, Beltsville, MD 20705, USA
| | - Lingyang Xu
- USDA-ARS, Animal Genomics and Improvement Laboratory, Beltsville, MD 20705, USA Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| | - Jana L Hutchison
- USDA-ARS, Animal Genomics and Improvement Laboratory, Beltsville, MD 20705, USA
| | - John B Cole
- USDA-ARS, Animal Genomics and Improvement Laboratory, Beltsville, MD 20705, USA
| | - Daniel J Null
- USDA-ARS, Animal Genomics and Improvement Laboratory, Beltsville, MD 20705, USA
| | - Steven G Schroeder
- USDA-ARS, Animal Genomics and Improvement Laboratory, Beltsville, MD 20705, USA
| | - Jiuzhou Song
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD 20742, USA
| | | | - Tad S Sonstegard
- USDA-ARS, Animal Genomics and Improvement Laboratory, Beltsville, MD 20705, USA
| | | | - Robert D Schnabel
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA Informatics Institute, University of Missouri, Columbia, MO, USA
| | - Jeremy F Taylor
- Division of Animal Sciences, University of Missouri, Columbia, MO 65211, USA
| | - Harris A Lewin
- Department of Evolution and Ecology, University of California, Davis, CA 95616, USA
| | - George E Liu
- USDA-ARS, Animal Genomics and Improvement Laboratory, Beltsville, MD 20705, USA
| |
Collapse
|
44
|
Au CE, Hermo L, Byrne E, Smirle J, Fazel A, Simon PHG, Kearney RE, Cameron PH, Smith CE, Vali H, Fernandez-Rodriguez J, Ma K, Nilsson T, Bergeron JJM. Expression, sorting, and segregation of Golgi proteins during germ cell differentiation in the testis. Mol Biol Cell 2015; 26:4015-32. [PMID: 25808494 PMCID: PMC4710233 DOI: 10.1091/mbc.e14-12-1632] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 03/19/2015] [Indexed: 12/14/2022] Open
Abstract
A total of 1318 proteins characterized in the male germ cell Golgi apparatus reveal a new germ cell–specific Golgi marker and a new pan-Golgi marker for all cells. The localization of these and other Golgi proteins reveals differential expression linked to mitosis, meiosis, acrosome formation, and postacrosome Golgi migration and destination in the late spermatid. The molecular basis of changes in structure, cellular location, and function of the Golgi apparatus during male germ cell differentiation is unknown. To deduce cognate Golgi proteins, we isolated germ cell Golgi fractions, and 1318 proteins were characterized, with 20 localized in situ. The most abundant protein, GL54D of unknown function, is characterized as a germ cell–specific Golgi-localized type II integral membrane glycoprotein. TM9SF3, also of unknown function, was revealed to be a universal Golgi marker for both somatic and germ cells. During acrosome formation, several Golgi proteins (GBF1, GPP34, GRASP55) localize to both the acrosome and Golgi, while GL54D, TM9SF3, and the Golgi trafficking protein TMED7/p27 are segregated from the acrosome. After acrosome formation, GL54D, TM9SF3, TMED4/p25, and TMED7/p27 continue to mark Golgi identity as it migrates away from the acrosome, while the others (GBF1, GPP34, GRASP55) remain in the acrosome and are progressively lost in later steps of differentiation. Cytoplasmic HSP70.2 and the endoplasmic reticulum luminal protein-folding enzyme PDILT are also Golgi recruited but only during acrosome formation. This resource identifies abundant Golgi proteins that are expressed differentially during mitosis, meiosis, and postacrosome Golgi migration, including the last step of differentiation.
Collapse
Affiliation(s)
- Catherine E Au
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 0C7, Canada Division of Endocrinology and Metabolism, McGill University Health Centre Research Institute, Montreal, QC H3A 1A1, Canada
| | - Louis Hermo
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 0C7, Canada
| | - Elliot Byrne
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 0C7, Canada Division of Endocrinology and Metabolism, McGill University Health Centre Research Institute, Montreal, QC H3A 1A1, Canada
| | - Jeffrey Smirle
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 0C7, Canada Division of Endocrinology and Metabolism, McGill University Health Centre Research Institute, Montreal, QC H3A 1A1, Canada
| | - Ali Fazel
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 0C7, Canada Division of Endocrinology and Metabolism, McGill University Health Centre Research Institute, Montreal, QC H3A 1A1, Canada
| | - Paul H G Simon
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 0C7, Canada Division of Endocrinology and Metabolism, McGill University Health Centre Research Institute, Montreal, QC H3A 1A1, Canada
| | - Robert E Kearney
- Department of Biomedical Engineering Department, McGill University, Montreal, QC H3A 2B4, Canada
| | - Pamela H Cameron
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 0C7, Canada Division of Endocrinology and Metabolism, McGill University Health Centre Research Institute, Montreal, QC H3A 1A1, Canada
| | - Charles E Smith
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 0C7, Canada
| | - Hojatollah Vali
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 0C7, Canada
| | - Julia Fernandez-Rodriguez
- Centre for Cellular Imaging, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Kewei Ma
- Division of Endocrinology and Metabolism, McGill University Health Centre Research Institute, Montreal, QC H3A 1A1, Canada
| | - Tommy Nilsson
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 0C7, Canada Division of Endocrinology and Metabolism, McGill University Health Centre Research Institute, Montreal, QC H3A 1A1, Canada
| | - John J M Bergeron
- Department of Anatomy and Cell Biology, McGill University, Montreal, QC H3A 0C7, Canada Division of Endocrinology and Metabolism, McGill University Health Centre Research Institute, Montreal, QC H3A 1A1, Canada
| |
Collapse
|
45
|
Duquet A, Melotti A, Mishra S, Malerba M, Seth C, Conod A, Ruiz i Altaba A. A novel genome-wide in vivo screen for metastatic suppressors in human colon cancer identifies the positive WNT-TCF pathway modulators TMED3 and SOX12. EMBO Mol Med 2015; 6:882-901. [PMID: 24920608 PMCID: PMC4119353 DOI: 10.15252/emmm.201303799] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The progression of tumors to the metastatic state involves the loss of metastatic suppressor functions. Finding these, however, is difficult as in vitro assays do not fully predict metastatic behavior, and the majority of studies have used cloned cell lines, which do not reflect primary tumor heterogeneity. Here, we have designed a novel genome-wide screen to identify metastatic suppressors using primary human tumor cells in mice, which allows saturation screens. Using this unbiased approach, we have tested the hypothesis that endogenous colon cancer metastatic suppressors affect WNT-TCF signaling. Our screen has identified two novel metastatic suppressors: TMED3 and SOX12, the knockdown of which increases metastatic growth after direct seeding. Moreover, both modify the type of self-renewing spheroids, but only knockdown of TMED3 also induces spheroid cell spreading and lung metastases from a subcutaneous xenograft. Importantly, whereas TMED3 and SOX12 belong to different families involved in protein secretion and transcriptional regulation, both promote endogenous WNT-TCF activity. Treatments for advanced or metastatic colon cancer may thus not benefit from WNT blockers, and these may promote a worse outcome.
Collapse
Affiliation(s)
- Arnaud Duquet
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Alice Melotti
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Sonakshi Mishra
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Monica Malerba
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Chandan Seth
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Arwen Conod
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| | - Ariel Ruiz i Altaba
- Department of Genetic Medicine and Development, University of Geneva Medical School, Geneva, Switzerland
| |
Collapse
|
46
|
Liaunardy-Jopeace A, Gay NJ. Molecular and cellular regulation of toll-like receptor-4 activity induced by lipopolysaccharide ligands. Front Immunol 2014; 5:473. [PMID: 25339952 PMCID: PMC4186342 DOI: 10.3389/fimmu.2014.00473] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Accepted: 09/15/2014] [Indexed: 12/22/2022] Open
Abstract
As well as being the primary signaling receptor for bacterial endotoxin or lipopolysaccharide Toll-like receptor-4 function is modulated by numerous factors not only in the context of microbial pathogenesis but also autoimmune and allergic diseases. TLR4 is subject to multiple levels of endogenous control and regulation from biosynthesis and trafficking to signal transduction and degradation. On the other hand regulation of TLR4 activity breaks down during Gram −ve sepsis leading to systemic damage, multi organ failure, and death. In this article, we review how TLR4 traffics from the early secretory pathway, the cis/trans Golgi to the cell surface and endolysosomal compartments. We will present evidence about how these processes influence signaling and can potentially lead to increased sensitivity to ligand-dependent activation as well as ligand-independent constitutive activation that may contribute to pathogenesis in sepsis. We will also discuss how sustained signaling may be coupled to endocytosis and consider the potential molecular mechanisms of immuno-modulators that modify TLR4 signaling function including the cat allergen FelD1 and endogenous protein ligands such as the extracellular matrix protein tenascin C and calprotectin (MRP8/14).
Collapse
Affiliation(s)
| | - Nicholas J Gay
- Department of Biochemistry, University of Cambridge , Cambridge , UK
| |
Collapse
|
47
|
Liaunardy-Jopeace A, Bryant CE, Gay NJ. The COP II adaptor protein TMED7 is required to initiate and mediate the delivery of TLR4 to the plasma membrane. Sci Signal 2014; 7:ra70. [PMID: 25074978 PMCID: PMC4685749 DOI: 10.1126/scisignal.2005275] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Toll-like receptor 4 (TLR4), the receptor for the bacterial product endotoxin, is subject to multiple points of regulation at the levels of signaling, biogenesis, and trafficking. Dysregulation of TLR4 signaling can cause serious inflammatory diseases, such as sepsis. We found that the p24 family protein TMED7 (transmembrane emp24 protein transport domain containing 7) is required for the trafficking of TLR4 from the endoplasmic reticulum to the cell surface through the Golgi. TMED7 formed a stable complex with the ectodomain of TLR4, an interaction that required the coiled-coil and Golgi dynamics (GOLD) domains, but not the cytosolic, coat protein complex II (COP II) sorting motif, of TMED7. Depletion of TMED7 reduced TLR4 signaling mediated by the adaptor protein MyD88 (myeloid differentiation marker 88), but not that mediated by the adaptor proteins TRIF [Toll-interleukin-1 receptor (TIR) domain-containing adaptor protein inducing interferon-β] and TRAM (TRIF-related adaptor molecule). Truncated forms of TMED7 lacking the COP II sorting motif or the transmembrane domain were mislocalized and resulted in ligand-independent signaling that probably arises from receptors accumulated intracellularly. Together, these results support the hypothesis that p24 proteins perform a quality control step by recognizing correctly folded anterograde cargo, such as TLR4, in early secretory compartments and facilitating the translocation of this cargo to the cell surface.
Collapse
Affiliation(s)
| | - Clare E Bryant
- Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, UK
| | - Nicholas J Gay
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, UK.
| |
Collapse
|
48
|
Microarray analysis of perinatal-estrogen-induced changes in gene expression related to brain sexual differentiation in mice. PLoS One 2013; 8:e79437. [PMID: 24223949 PMCID: PMC3817063 DOI: 10.1371/journal.pone.0079437] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Accepted: 09/23/2013] [Indexed: 11/19/2022] Open
Abstract
Sexual dimorphism of the behaviors or physiological functions in mammals is mainly due to the sex difference of the brain. A number of studies have suggested that the brain is masculinized or defeminized by estradiol converted from testicular androgens in perinatal period in rodents. However, the mechanisms of estrogen action resulting in masculinization/defeminization of the brain have not been clarified yet. The large-scale analysis with microarray in the present study is an attempt to obtain the candidate gene(s) mediating the perinatal estrogen effect causing the brain sexual differentiation. Female mice were injected with estradiol benzoate (EB) or vehicle on the day of birth, and the hypothalamus was collected at either 1, 3, 6, 12, or 24 h after the EB injection. More than one hundred genes down-regulated by the EB treatment in a biphasic manner peaked at 3 h and 12-24 h after the EB treatment, while forty to seventy genes were constantly up-regulated after it. Twelve genes, including Ptgds, Hcrt, Tmed2, Klc1, and Nedd4, whose mRNA expressions were down-regulated by the neonatal EB treatment, were chosen for further examination by semiquantitative RT-PCR in the hypothalamus of perinatal intact male and female mice. We selected the genes based on the known profiles of their potential roles in brain development. mRNA expression levels of Ptgds, Hcrt, Tmed2, and Nedd4 were significantly lower in male mice than females at the day of birth, suggesting that the genes are down-regulated by estrogen converted from testicular androgen in perinatal male mice. Some genes, such as Ptgds encoding prostaglandin D2 production enzyme and Hcrt encording orexin, have been reported to have a role in neuroprotection. Thus, Ptgds and Hcrt could be possible candidate genes, which may mediate the effect of perinatal estrogen responsible for brain sexual differentiation.
Collapse
|
49
|
Hu CW, Tseng CW, Chien CW, Huang HC, Ku WC, Lee SJ, Chen YJ, Juan HF. Quantitative proteomics reveals diverse roles of miR-148a from gastric cancer progression to neurological development. J Proteome Res 2013; 12:3993-4004. [PMID: 23869555 DOI: 10.1021/pr400302w] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
MicroRNAs (miRNAs) are noncoding RNAs that control gene expression either by degradation of mRNAs or inhibition of protein translation. miR-148a has been reported to have the impacts on tumor progression. Here, a quantitative proteomics combined with stable isotope labeling was applied to identify the global profile of miR-148a-regulated downstream proteins. The data have been deposited to the ProteomeXchange with identifier PXD000190. A total of 2938 proteins were quantified, and 55 proteins were considered to be regulated by miR-148a. We found that not only proteins associated with cancer progression but also molecules involved in neural development were regulated by miR-148a. This study is the first to identify the function of miR-148a in neural development by using a proteomic approach. Analysis of a public clinical database also showed that the patients with neural diseases could display abnormal expression of miR-148a. Moreover, silencing of miR-148a led to the abnormal morphology and decreased expression of neuron-related markers in the developing brain of zebrafish. These results provided important insight into the regulation of neurological development elicited by miR-148a.
Collapse
Affiliation(s)
- Chia-Wei Hu
- Institute of Molecular and Cellular Biology and Department of Life Science, National Taiwan University, Taipei 106, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Montesinos JC, Langhans M, Sturm S, Hillmer S, Aniento F, Robinson DG, Marcote MJ. Putative p24 complexes in Arabidopsis contain members of the delta and beta subfamilies and cycle in the early secretory pathway. JOURNAL OF EXPERIMENTAL BOTANY 2013; 64:3147-67. [PMID: 23918961 PMCID: PMC3733144 DOI: 10.1093/jxb/ert157] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
p24 proteins are a family of type I membrane proteins localized to compartments of the early secretory pathway and to coat protein I (COPI)- and COPII-coated vesicles. They can be classified, by sequence homology, into four subfamilies, named p24α, p24β, p24γ, and p24δ. In contrast to animals and fungi, plants contain only members of the p24β and p24δ subfamilies, the latter probably including two different subclasses. It has previously been shown that transiently expressed red fluorescent protein (RFP)-p24δ5 (p24δ1 subclass) localizes to the endoplasmic reticulum (ER) at steady state as a consequence of highly efficient COPI-based recycling from the Golgi apparatus. It is now shown that transiently expressed RFP-p24δ9 (p24δ2 subclass) also localizes to the ER. In contrast, transiently expressed green fluorescent protein (GFP)-p24β3 mainly localizes to the Golgi apparatus (as p24β2) and exits the ER in a COPII-dependent manner. Immunogold electron microscopy in Arabidopsis root tip cells using specific antibodies shows that endogenous p24δ9 localizes mainly to the ER but also partially to the cis-Golgi. In contrast, endogenous p24β3 mainly localizes to the Golgi apparatus. By a combination of experiments using transient expression, knock-out mutants, and co-immunoprecipitation, it is proposed that Arabidopsis p24 proteins form different heteromeric complexes (including members of the β and δ subfamilies) which are important for their stability and their coupled trafficking at the ER-Golgi interface. Evidence is also provided for a role for p24δ5 in retrograde Golgi-ER transport of the KDEL-receptor ERD2.
Collapse
Affiliation(s)
- Juan Carlos Montesinos
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universitat de Valencia, Spain
- *These authors contributed equally to this work
| | - Markus Langhans
- Department of Plant Cell Biology, Centre for Organismal Studies, University of Heidelberg, Germany
- *These authors contributed equally to this work
| | - Silke Sturm
- Department of Plant Cell Biology, Centre for Organismal Studies, University of Heidelberg, Germany
- *These authors contributed equally to this work
| | - Stefan Hillmer
- Department of Plant Cell Biology, Centre for Organismal Studies, University of Heidelberg, Germany
| | - Fernando Aniento
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universitat de Valencia, Spain
| | - David G. Robinson
- Department of Plant Cell Biology, Centre for Organismal Studies, University of Heidelberg, Germany
| | - María Jesús Marcote
- Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universitat de Valencia, Spain
- To whom correspondence should be addressed. E-mail:
| |
Collapse
|