1
|
Ximin Y, Hashimoto H, Wada I, Hosokawa N. Visualization of ER-to-Golgi trafficking of procollagen X. Cell Struct Funct 2024; 49:67-81. [PMID: 39245571 PMCID: PMC11930776 DOI: 10.1247/csf.24024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 08/31/2024] [Indexed: 09/10/2024] Open
Abstract
Collagen is the most abundant protein in the extracellular matrix of animals, and 28 types of collagen have been reported in humans. We previously analyzed the endoplasmic reticulum (ER)-to-Golgi transport of fibril-forming type III collagen (Hirata et al., 2022) and network-forming type IV collagen (Matsui et al., 2020), both of which have long collagenous triple-helical regions. To understand the ER-to-Golgi trafficking of various types of collagens, we analyzed the transport of short-chain type X collagen in this study. We fused cysteine-free GFP to the N-telopeptide region of procollagen X (GFP-COL10A1), as employed in our previous analysis of procollagens III and IV, and analyzed its transport by live-cell imaging. Procollagen X was transported to the Golgi apparatus via vesicular and tubular carriers containing ERGIC53 and RAB1B, similar to those used for procollagen III. Carriers containing procollagen X probably used the same transport processes as those containing conventional cargoes such as α1-antitrypsin. SAR1, TANGO1, SLY1/SCFD1, and BET3/TRAPPC3 were required for trafficking of procollagen X, which are different from the factors required for trafficking of procollagens III (SAR1, TANGO1, and CUL3) and IV (SAR1 and SLY1/SCFD1). These findings reveal that accommodation of various types of collagens with different shapes into carriers may require fine-tuning of the ER-to-Golgi transport machinery.Key words: collagen, GFP-procollagen X, ER-to-Golgi trafficking, export from ER, TANGO1.
Collapse
Affiliation(s)
- Yuan Ximin
- Laboratory of Molecular and Cellular Biology, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Hitoshi Hashimoto
- Department of Cell Science, Institute of Biomedical Sciences, Fukushima Medical University, School of Medicine, Fukushima 960-1295, Japan
| | - Ikuo Wada
- Department of Cell Science, Institute of Biomedical Sciences, Fukushima Medical University, School of Medicine, Fukushima 960-1295, Japan
| | - Nobuko Hosokawa
- Laboratory of Molecular and Cellular Biology, Institute for Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
2
|
Yang N, Shi L, Xu P, Ren F, Li C, Qi X. Identification of potential drug targets for amyotrophic lateral sclerosis by Mendelian randomization analysis based on brain and plasma proteomics. Exp Gerontol 2024; 195:112538. [PMID: 39116956 DOI: 10.1016/j.exger.2024.112538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 07/18/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
Amyotrophic lateral sclerosis as a fatal neurodegenerative disease currently lacks effective therapeutic agents. Thus, finding new therapeutic targets to drive disease treatment is necessary. In this study, we utilized brain and plasma proteins as genetic instruments obtained from genome-wide association studies to conduct a Mendelian randomization analysis to identify potential drug targets for amyotrophic lateral sclerosis. Additionally, we validated our results externally using other datasets. We also used Bayesian co-localization analysis and phenotype scanning. Furthermore, we constructed a protein-protein interaction network to elucidate potential correlations between the identified proteins and existing targets. Mendelian randomization analysis indicated that elevated levels of ANO5 (OR = 1.30; 95 % CI, 1.14-1.49; P = 1.52E-04), SCFD1 (OR = 3.82; 95 % CI, 2.39-6.10; P = 2.19E-08), and SIGLEC9 (OR = 1.05; 95% CI, 1.03-1.07; P = 4.71E-05) are associated with an increased risk of amyotrophic lateral sclerosis, with external validation supporting these findings. Co-localization analysis confirmed that ANO5, SCFD1, and SIGLEC9 (coloc.abf-PPH4 = 0.848, 0.984, and 0.945, respectively) shared the same variant with amyotrophic lateral sclerosis, further substantiating potential role of these proteins as a therapeutic target. There are interactive relationships between the potential proteins and existing targets of amyotrophic lateral sclerosis. Our findings suggested that elevated levels of ANO5, SCFD1, and SIGLEC9 are connected with an increased risk of amyotrophic lateral sclerosis and might be promising therapeutic targets. However, further exploration is necessary to fully understand the underlying mechanisms involved.
Collapse
Affiliation(s)
- Ni Yang
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Liangyuan Shi
- Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, China.
| | - Pengfei Xu
- Qingdao Hiser Hospital Affiliated of Qingdao University (Qingdao Traditional Chinese Medicine Hospital), Qingdao, China
| | - Fang Ren
- Department of Laboratory, Jimo District Qingdao Hospital of Traditional Chinese Medicine, Qingdao, China
| | - Chunlin Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xianghua Qi
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
3
|
Yang Y, Zhang X, Zhao Q, Zhang J, Lou X. Compromised COPII vesicle trafficking leads to glycogenic hepatopathy. Dis Model Mech 2024; 17:dmm050748. [PMID: 39139065 PMCID: PMC11463966 DOI: 10.1242/dmm.050748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 08/06/2024] [Indexed: 08/15/2024] Open
Abstract
Being a vital cellular process, coat protein complex II (COPII) vesicle trafficking has been found to play a crucial role in liver metabolism. However, its functions and the underlying mechanisms in systemic metabolic homeostasis have not been fully understood. Here, with a newly identified gene trap zebrafish line (sec31anju221), we show that compromised COPII vesicle trafficking leads to biphasic abnormal hepatic metabolism. During the larval stage, deficiency of COPII-mediated trafficking leads to activation of the unfolded protein response and the development of hepatic steatosis. By using epistasis analysis, we found that the eIF2α-ATF4 pathway serves as the primary effector for liver steatosis. In adult sec31anju221 fish, the hepatosteatosis was reversed and the phenotype switched to glycogenic hepatopathy. Proteomic profiling and biochemical assays indicate that sec31anju221 fish are in a state of hypothyroidism. Moreover, our study shows that thyroid hormone treatment alleviates the metabolic defects. This study provides insights into processes of liver diseases associated with vesicle trafficking impairments and expands our understanding of the pathological interplay between thyroid and liver.
Collapse
Affiliation(s)
- Yuxi Yang
- Medical School, Nanjing University, Nanjing 210093, China
| | - Xue Zhang
- Research Center for Life Sciences Computing, Zhejiang Laboratory, Hangzhou 311100, China
| | - Qingshun Zhao
- Medical School, Nanjing University, Nanjing 210093, China
| | - Jingzi Zhang
- Medical School, Nanjing University, Nanjing 210093, China
| | - Xin Lou
- Research Center for Life Sciences Computing, Zhejiang Laboratory, Hangzhou 311100, China
| |
Collapse
|
4
|
Wang L, Lin M, Hou L, Rikihisa Y. Anaplasma phagocytophilum effector EgeA facilitates infection by hijacking TANGO1 and SCFD1 from ER-Golgi exit sites to pathogen-occupied inclusions. Proc Natl Acad Sci U S A 2024; 121:e2405209121. [PMID: 39106308 PMCID: PMC11331065 DOI: 10.1073/pnas.2405209121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 07/01/2024] [Indexed: 08/09/2024] Open
Abstract
The obligatory intracellular bacterium Anaplasma phagocytophilum causes human granulocytic anaplasmosis, an emerging zoonosis. Anaplasma has limited biosynthetic and metabolic capacities, yet it effectively replicates inside of inclusions/vacuoles of eukaryotic host cells. Here, we describe a unique Type IV secretion system (T4SS) effector, ER-Golgi exit site protein of Anaplasma (EgeA). In cells infected by Anaplasma, secreted native EgeA, EgeA-GFP, and the C-terminal half of EgeA (EgeA-C)-GFP localized to Anaplasma-containing inclusions. In uninfected cells, EgeA-C-GFP localized to cis-Golgi, whereas the N-terminal half of EgeA-GFP localized to the ER. Pull-down assays identified EgeA-GFP binding to a transmembrane protein in the ER, Transport and Golgi organization protein 1 (TANGO1). By yeast two-hybrid analysis, EgeA-C directly bound Sec1 family domain-containing protein 1 (SCFD1), a host protein of the cis-Golgi network that binds TANGO1 at ER-Golgi exit sites (ERES). Both TANGO1 and SCFD1 localized to the Anaplasma inclusion surface. Furthermore, knockdown of Anaplasma EgeA or either host TANGO1 or SCFD1 significantly reduced Anaplasma infection. TANGO1 and SCFD1 prevent ER congestion and stress by facilitating transport of bulky or unfolded proteins at ERES. A bulky cargo collagen and the ER-resident chaperon BiP were transported into Anaplasma inclusions, and several ER stress marker genes were not up-regulated in Anaplasma-infected cells. Furthermore, EgeA transfection reduced collagen overexpression-induced BiP upregulation. These results suggest that by binding to the two ERES proteins, EgeA redirects the cargo-adapted ERES to pathogen-occupied inclusions and reduces ERES congestion, which facilitates Anaplasma nutrient acquisition and reduces ER stress for Anaplasma survival and proliferation.
Collapse
Affiliation(s)
- Lidan Wang
- Department of Veterinary Biosciences, College of Veterinary Medicine, Infectious Diseases Institute, The Ohio State University, Columbus, OH43210
| | - Mingqun Lin
- Department of Veterinary Biosciences, College of Veterinary Medicine, Infectious Diseases Institute, The Ohio State University, Columbus, OH43210
| | - Libo Hou
- Department of Veterinary Biosciences, College of Veterinary Medicine, Infectious Diseases Institute, The Ohio State University, Columbus, OH43210
| | - Yasuko Rikihisa
- Department of Veterinary Biosciences, College of Veterinary Medicine, Infectious Diseases Institute, The Ohio State University, Columbus, OH43210
| |
Collapse
|
5
|
Guillemyn B, De Saffel H, Bek JW, Tapaneeyaphan P, De Clercq A, Jarayseh T, Debaenst S, Willaert A, De Rycke R, Byers PH, Rosseel T, Coucke P, Blaumeiser B, Syx D, Malfait F, Symoens S. Syntaxin 18 Defects in Human and Zebrafish Unravel Key Roles in Early Cartilage and Bone Development. J Bone Miner Res 2023; 38:1718-1730. [PMID: 37718532 DOI: 10.1002/jbmr.4914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 08/08/2023] [Accepted: 09/09/2023] [Indexed: 09/19/2023]
Abstract
SNARE proteins comprise a conserved protein family responsible for catalyzing membrane fusion during vesicle traffic. Syntaxin18 (STX18) is a poorly characterized endoplasmic reticulum (ER)-resident t-SNARE. Recently, together with TANGO1 and SLY1, its involvement was shown in ER to Golgi transport of collagen II during chondrogenesis. We report a fetus with a severe osteochondrodysplasia in whom we identified a homozygous substitution of the highly conserved p.Arg10 to Pro of STX18. CRISPR/Cas9-mediated Stx18 deficiency in zebrafish reveals a crucial role for Stx18 in cartilage and bone development. Furthermore, increased expression of multiple components of the Stx18 SNARE complex and of COPI and COPII proteins suggests that Stx18 deficiency impairs antero- and retrograde vesicular transport in the crispant stx18 zebrafish. Taken together, our studies highlight a new candidate gene for a recessive form of osteochondrodysplasia, thereby possibly broadening the SNAREopathy phenotypic spectrum and opening new doors toward future research avenues. © 2023 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Brecht Guillemyn
- Department of Biomolecular Medicine, Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
| | - Hanna De Saffel
- Department of Biomolecular Medicine, Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
| | - Jan Willem Bek
- Department of Biomolecular Medicine, Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
| | - Piyanoot Tapaneeyaphan
- Department of Biomolecular Medicine, Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
| | - Adelbert De Clercq
- Department of Biomolecular Medicine, Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
| | - Tamara Jarayseh
- Department of Biomolecular Medicine, Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
| | - Sophie Debaenst
- Department of Biomolecular Medicine, Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
| | - Andy Willaert
- Department of Biomolecular Medicine, Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
| | - Riet De Rycke
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- VIB Center for Inflammation Research and Bioimaging Core, Ghent, Belgium
| | - Peter H Byers
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Department of Medicine (Medical Genetics), University of Washington, Seattle, WA, USA
| | - Toon Rosseel
- Department of Biomolecular Medicine, Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
| | - Paul Coucke
- Department of Biomolecular Medicine, Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
| | - Bettina Blaumeiser
- Department of Medical Genetics, University and University Hospital of Antwerp, Antwerp, Belgium
| | - Delfien Syx
- Department of Biomolecular Medicine, Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
| | - Fransiska Malfait
- Department of Biomolecular Medicine, Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
| | - Sofie Symoens
- Department of Biomolecular Medicine, Center for Medical Genetics Ghent, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
6
|
Seregin AA, Smirnova LP, Dmitrieva EM, Zavialova MG, Simutkin GG, Ivanova SA. Differential Expression of Proteins Associated with Bipolar Disorder as Identified Using the PeptideShaker Software. Int J Mol Sci 2023; 24:15250. [PMID: 37894929 PMCID: PMC10607299 DOI: 10.3390/ijms242015250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 09/29/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
The prevalence of bipolar disorder (BD) in modern society is growing rapidly, but due to the lack of paraclinical criteria, its differential diagnosis with other mental disorders is somewhat challenging. In this regard, the relevance of proteomic studies is increasing due to the development of methods for processing large data arrays; this contributes to the discovery of protein patterns of pathological processes and the creation of new methods of diagnosis and treatment. It seems promising to search for proteins involved in the pathogenesis of BD in an easily accessible material-blood serum. Sera from BD patients and healthy individuals were purified via affinity chromatography to isolate 14 major proteins and separated using 1D SDS-PAGE. After trypsinolysis, the proteins in the samples were identified via HPLC/mass spectrometry. Mass spectrometric data were processed using the OMSSA and X!Tandem search algorithms using the UniProtKB database, and the results were analyzed using PeptideShaker. Differences in proteomes were assessed via an unlabeled NSAF-based analysis using a two-tailed Bonferroni-adjusted t-test. When comparing the blood serum proteomes of BD patients and healthy individuals, 10 proteins showed significant differences in NSAF values. Of these, four proteins were predominantly present in BD patients with the maximum NSAF value: 14-3-3 protein zeta/delta; ectonucleoside triphosphate diphosphohydrolase 7; transforming growth factor-beta-induced protein ig-h3; and B-cell CLL/lymphoma 9 protein. Further exploration of the role of these proteins in BD is warranted; conducting such studies will help develop new paraclinical criteria and discover new targets for BD drug therapy.
Collapse
Affiliation(s)
- Alexander A. Seregin
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634014, Russia; (A.A.S.)
| | - Liudmila P. Smirnova
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634014, Russia; (A.A.S.)
| | - Elena M. Dmitrieva
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634014, Russia; (A.A.S.)
| | | | - German G. Simutkin
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634014, Russia; (A.A.S.)
| | - Svetlana A. Ivanova
- Mental Health Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk 634014, Russia; (A.A.S.)
| |
Collapse
|
7
|
Huttner IG, Santiago CF, Jacoby A, Cheng D, Trivedi G, Cull S, Cvetkovska J, Chand R, Berger J, Currie PD, Smith KA, Fatkin D. Loss of Sec-1 Family Domain-Containing 1 ( scfd1) Causes Severe Cardiac Defects and Endoplasmic Reticulum Stress in Zebrafish. J Cardiovasc Dev Dis 2023; 10:408. [PMID: 37887855 PMCID: PMC10607167 DOI: 10.3390/jcdd10100408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 09/15/2023] [Accepted: 09/20/2023] [Indexed: 10/28/2023] Open
Abstract
Dilated cardiomyopathy (DCM) is a common heart muscle disorder that frequently leads to heart failure, arrhythmias, and death. While DCM is often heritable, disease-causing mutations are identified in only ~30% of cases. In a forward genetic mutagenesis screen, we identified a novel zebrafish mutant, heart and head (hahvcc43), characterized by early-onset cardiomyopathy and craniofacial defects. Linkage analysis and next-generation sequencing identified a nonsense variant in the highly conserved scfd1 gene, also known as sly1, that encodes sec1 family domain-containing 1. Sec1/Munc18 proteins, such as Scfd1, are involved in membrane fusion regulating endoplasmic reticulum (ER)/Golgi transport. CRISPR/Cas9-engineered scfd1vcc44 null mutants showed severe cardiac and craniofacial defects and embryonic lethality that recapitulated the phenotype of hahvcc43 mutants. Electron micrographs of scfd1-depleted cardiomyocytes showed reduced myofibril width and sarcomere density, as well as reticular network disorganization and fragmentation of Golgi stacks. Furthermore, quantitative PCR analysis showed upregulation of ER stress response and apoptosis markers. Both heterozygous hahvcc43 mutants and scfd1vcc44 mutants survived to adulthood, showing chamber dilation and reduced ventricular contraction. Collectively, our data implicate scfd1 loss-of-function as the genetic defect at the hahvcc43 locus and provide new insights into the role of scfd1 in cardiac development and function.
Collapse
Affiliation(s)
- Inken G. Huttner
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (I.G.H.); (C.F.S.); (A.J.); (D.C.); (G.T.); (S.C.); (J.C.); (R.C.)
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Celine F. Santiago
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (I.G.H.); (C.F.S.); (A.J.); (D.C.); (G.T.); (S.C.); (J.C.); (R.C.)
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Arie Jacoby
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (I.G.H.); (C.F.S.); (A.J.); (D.C.); (G.T.); (S.C.); (J.C.); (R.C.)
| | - Delfine Cheng
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (I.G.H.); (C.F.S.); (A.J.); (D.C.); (G.T.); (S.C.); (J.C.); (R.C.)
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW 2052, Australia
| | - Gunjan Trivedi
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (I.G.H.); (C.F.S.); (A.J.); (D.C.); (G.T.); (S.C.); (J.C.); (R.C.)
| | - Stephen Cull
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (I.G.H.); (C.F.S.); (A.J.); (D.C.); (G.T.); (S.C.); (J.C.); (R.C.)
| | - Jasmina Cvetkovska
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (I.G.H.); (C.F.S.); (A.J.); (D.C.); (G.T.); (S.C.); (J.C.); (R.C.)
| | - Renee Chand
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (I.G.H.); (C.F.S.); (A.J.); (D.C.); (G.T.); (S.C.); (J.C.); (R.C.)
| | - Joachim Berger
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; (J.B.); (P.D.C.)
- European Molecular Biology Labs (EMBL) Australia, Victorian Node, Monash University, Clayton, VIC 3800, Australia
| | - Peter D. Currie
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3800, Australia; (J.B.); (P.D.C.)
- European Molecular Biology Labs (EMBL) Australia, Victorian Node, Monash University, Clayton, VIC 3800, Australia
| | - Kelly A. Smith
- Department of Anatomy & Physiology, The University of Melbourne, Parkville, VIC 3010, Australia;
| | - Diane Fatkin
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute, Darlinghurst, NSW 2010, Australia; (I.G.H.); (C.F.S.); (A.J.); (D.C.); (G.T.); (S.C.); (J.C.); (R.C.)
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Kensington, NSW 2052, Australia
- Cardiology Department, St Vincent’s Hospital, Darlinghurst, NSW 2010, Australia
| |
Collapse
|
8
|
Gu XJ, Su WM, Dou M, Jiang Z, Duan QQ, Wang H, Ren YL, Cao B, Wang Y, Chen YP. Identifying novel genes for amyotrophic lateral sclerosis by integrating human brain proteomes with genome-wide association data. J Neurol 2023:10.1007/s00415-023-11757-4. [PMID: 37148340 DOI: 10.1007/s00415-023-11757-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/27/2023] [Accepted: 04/29/2023] [Indexed: 05/08/2023]
Abstract
BACKGROUND Genome-Wide Association Studies (GWAS) have identified numerous risk genes for Amyotrophic Lateral Sclerosis (ALS); however, the mechanisms by which these loci confer ALS risk are uncertain. This study aims to identify novel causal proteins in the brains of patients with ALS using an integrative analytical pipeline. METHODS Using the datasets of Protein Quantitative Trait Loci (pQTL) (NpQTL1 = 376, NpQTL2 = 152), expression QTL (eQTL) (N = 452), and the largest ALS GWAS (NALS=27,205, NControls = 110,881), we performed a systematic analytical pipeline including Proteome-Wide Association Study (PWAS), Mendelian Randomization (MR), Bayesian colocalization, and Transcriptome-Wide Association Study (TWAS) to identify novel causal proteins for ALS in the brain. RESULTS Using PWAS, we found that the altered protein abundance of 12 genes in the brain was associated with ALS. Three genes (SCFD1, SARM1 and CAMLG) were identified as lead causal genes for ALS with solid evidence (False discovery rate < 0.05, in MR analysis; PPH4 > 80% for Bayesian colocalization). Specifically, an increased abundance of SCFD1 and CAMLG led to an increased risk of ALS, whereas a higher abundance of SARM1 led to a decreased risk of developing ALS. TWAS showed that SCFD1 and CAMLG were related to ALS at the transcriptional level. CONCLUSIONS SCFD1, CAMLG, and SARM1 exhibited robust associations and causality with ALS. The study findings provide novel clues for identifying potential therapeutic targets in ALS. Further studies are required to explore the mechanisms underlying the identified genes.
Collapse
Affiliation(s)
- Xiao-Jing Gu
- Mental Health Center, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Wei-Ming Su
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Institute of Brain Science and Brain-Inspired Technology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Centre for Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Meng Dou
- Chengdu Institute of Computer Application, Chinese Academy of Sciences, Chengdu, 610041, Sichuan, China
| | - Zheng Jiang
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Institute of Brain Science and Brain-Inspired Technology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Centre for Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Qing-Qing Duan
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Institute of Brain Science and Brain-Inspired Technology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Centre for Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Han Wang
- Department of Pathophysiology, West China College of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yan-Ling Ren
- Department of Pathophysiology, West China College of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Bei Cao
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Institute of Brain Science and Brain-Inspired Technology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
- Centre for Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yi Wang
- Department of Pathophysiology, West China College of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yong-Ping Chen
- Department of Neurology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
- Institute of Brain Science and Brain-Inspired Technology, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
- Centre for Rare Diseases, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
9
|
Iyer VJ, Osman MA. The Antipsychotic Drug Haldol Modulates IQGAP1-Signaling and Inhibits Cell Proliferation in Triple Negative Breast Cancer Cell Lines. MICROPUBLICATION BIOLOGY 2023; 2023:10.17912/micropub.biology.000823. [PMID: 37215640 PMCID: PMC10199339 DOI: 10.17912/micropub.biology.000823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/24/2023]
Abstract
The signaling scaffold oncoprotein IQGAP1 was identified as a classification and therapeutic biomarker in triple negative breast cancer (TNBC) cell lines. Here, we report that the antipsychotic drug Haldol induces novel protein-protein interactions with IQGAP1 and inhibits cell proliferation in TNBC cell lines. The identified proteins share known functions of IQGAP1 in secretion, transcription and apoptosis and provide further classification tools and potential precision therapeutic targets for Haldol in TNBC.
Collapse
Affiliation(s)
- Varun J. Iyer
- Department of Medicine, Division of Oncology, University of Toledo Medical Center, Toledo, Ohio, 43614 United States
| | - Mahasin A. Osman
- Department of Medicine, Division of Oncology, University of Toledo Medical Center, Toledo, Ohio, 43614 United States
| |
Collapse
|
10
|
Bartolec TK, Vázquez-Campos X, Norman A, Luong C, Johnson M, Payne RJ, Wilkins MR, Mackay JP, Low JKK. Cross-linking mass spectrometry discovers, evaluates, and corroborates structures and protein-protein interactions in the human cell. Proc Natl Acad Sci U S A 2023; 120:e2219418120. [PMID: 37071682 PMCID: PMC10151615 DOI: 10.1073/pnas.2219418120] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 03/16/2023] [Indexed: 04/19/2023] Open
Abstract
Significant recent advances in structural biology, particularly in the field of cryoelectron microscopy, have dramatically expanded our ability to create structural models of proteins and protein complexes. However, many proteins remain refractory to these approaches because of their low abundance, low stability, or-in the case of complexes-simply not having yet been analyzed. Here, we demonstrate the power of using cross-linking mass spectrometry (XL-MS) for the high-throughput experimental assessment of the structures of proteins and protein complexes. This included those produced by high-resolution but in vitro experimental data, as well as in silico predictions based on amino acid sequence alone. We present the largest XL-MS dataset to date, describing 28,910 unique residue pairs captured across 4,084 unique human proteins and 2,110 unique protein-protein interactions. We show that models of proteins and their complexes predicted by AlphaFold2, and inspired and corroborated by the XL-MS data, offer opportunities to deeply mine the structural proteome and interactome and reveal mechanisms underlying protein structure and function.
Collapse
Affiliation(s)
- Tara K. Bartolec
- Systems Biology Initiative, School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Randwick, NSW2052, Australia
| | - Xabier Vázquez-Campos
- Systems Biology Initiative, School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Randwick, NSW2052, Australia
| | - Alexander Norman
- School of Chemistry, University of Sydney, Sydney, NSW2006, Australia
| | - Clement Luong
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW2006, Australia
| | - Marcus Johnson
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW2006, Australia
| | - Richard J. Payne
- School of Chemistry, University of Sydney, Sydney, NSW2006, Australia
- Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Sydney, Sydney, NSW2006, Australia
| | - Marc R. Wilkins
- Systems Biology Initiative, School of Biotechnology and Biomolecular Sciences, The University of New South Wales, Randwick, NSW2052, Australia
| | - Joel P. Mackay
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW2006, Australia
| | - Jason K. K. Low
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW2006, Australia
| |
Collapse
|
11
|
Yildiz O, Schroth J, Tree T, Turner MR, Shaw PJ, Henson SM, Malaspina A. Senescent-like Blood Lymphocytes and Disease Progression in Amyotrophic Lateral Sclerosis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:e200042. [PMID: 36323511 PMCID: PMC9673751 DOI: 10.1212/nxi.0000000000200042] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 08/22/2022] [Indexed: 03/11/2023]
Abstract
BACKGROUND AND OBJECTIVES Aging is known to exacerbate neuroinflammation, and in the neurodegenerative disorder amyotrophic lateral sclerosis (ALS), an older age is associated with a worse prognosis. We have previously shown the activation of cell senescence pathways in the proteome of peripheral blood mononuclear cells and the increase of proinflammatory cytokines in blood from individuals living with ALS. In this single-center, retrospective study, we investigated the expression of senescent-like blood mononuclear cells in ALS. METHODS We first applied multidimensional cytometry by time-of-flight (CyTOF) to study the senescent immunophenotype of blood mononuclear cells from 21 patients with ALS and 10 healthy controls (HCs). We then used targeted flow cytometry (FC) to investigate frequencies of senescent blood lymphocytes in 40 patients with ALS and 20 HCs. Longitudinal analysis included 2 additional time points in 17 patients with ALS. Frequencies of senescent-like lymphocytes were analyzed in relation to survival. RESULTS Unsupervised clustering of CyTOF data showed higher frequencies of senescent CD4+CD27-CD57+ T cells in patients with ALS compared with those in HCs (p = 0.0017, false discovery (FDR)-adjusted p = 0.029). Moderate to strong negative correlations were identified between CD4 T central memory-cell frequencies and survival (R = -061, p = 0.01; FDR-adjusted p < 0.1) and between CD95 CD8 cells and ALS functional rating scale revised at baseline (R = -0.72, p = 0.001; FDR-adjusted p < 0.1).Targeted FC analysis showed higher memory T regulatory cells (p = 0.0052) and memory CD8+ T cell (M-Tc; p = 0.0006) in bulbar ALS (A-B) compared with those in limb ALS (A-L), while late memory B cells (LM-B) were also elevated in A-B and fast-progressing ALS (p = 0.0059). Higher M-Tc levels separated A-B from A-L (AUC: 0.887; p < 0.0001). A linear regression model with prespecified clinical independent variables and neurofilament light chain plasma concentration showed that higher frequencies of LM-B predicted a shorter survival (hazard ratio: 1.094, CI: 1.026-1.167; p = 0.006). DISCUSSION Our data suggest that a systemic elevation of senescent and late memory T and B lymphocytes is a feature of faster progressing ALS and of ALS individuals with bulbar involvement. Lymphocyte senescence and their memory state may be central to the immune dysregulation known to drive disease progression in ALS and a target for biomarkers and therapeutics discovery.
Collapse
Affiliation(s)
- Ozlem Yildiz
- From the Neuroscience and Trauma Centre (O.Y., A.M.), Blizard Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London; Queen Square Motor Neuron Disease Centre (A.M.), Neuromuscular Department, Institute of Neurology, University College London; Translational Medicine and Therapeutics (J.S., S.M.H.), William Harvey Research Institute, Barts and the London, Queen Mary University of London; Department of Immunobiology (T.T.), School of Immunology & Microbial Sciences, King's College London; Nuffield Department of Clinical Neurosciences (M.R.T.), University of Oxford; and Sheffield Institute for Translational Neuroscience (P.J.S.), University of Sheffield, UK
| | - Johannes Schroth
- From the Neuroscience and Trauma Centre (O.Y., A.M.), Blizard Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London; Queen Square Motor Neuron Disease Centre (A.M.), Neuromuscular Department, Institute of Neurology, University College London; Translational Medicine and Therapeutics (J.S., S.M.H.), William Harvey Research Institute, Barts and the London, Queen Mary University of London; Department of Immunobiology (T.T.), School of Immunology & Microbial Sciences, King's College London; Nuffield Department of Clinical Neurosciences (M.R.T.), University of Oxford; and Sheffield Institute for Translational Neuroscience (P.J.S.), University of Sheffield, UK
| | - Timothy Tree
- From the Neuroscience and Trauma Centre (O.Y., A.M.), Blizard Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London; Queen Square Motor Neuron Disease Centre (A.M.), Neuromuscular Department, Institute of Neurology, University College London; Translational Medicine and Therapeutics (J.S., S.M.H.), William Harvey Research Institute, Barts and the London, Queen Mary University of London; Department of Immunobiology (T.T.), School of Immunology & Microbial Sciences, King's College London; Nuffield Department of Clinical Neurosciences (M.R.T.), University of Oxford; and Sheffield Institute for Translational Neuroscience (P.J.S.), University of Sheffield, UK
| | - Martin R Turner
- From the Neuroscience and Trauma Centre (O.Y., A.M.), Blizard Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London; Queen Square Motor Neuron Disease Centre (A.M.), Neuromuscular Department, Institute of Neurology, University College London; Translational Medicine and Therapeutics (J.S., S.M.H.), William Harvey Research Institute, Barts and the London, Queen Mary University of London; Department of Immunobiology (T.T.), School of Immunology & Microbial Sciences, King's College London; Nuffield Department of Clinical Neurosciences (M.R.T.), University of Oxford; and Sheffield Institute for Translational Neuroscience (P.J.S.), University of Sheffield, UK
| | - Pamela J Shaw
- From the Neuroscience and Trauma Centre (O.Y., A.M.), Blizard Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London; Queen Square Motor Neuron Disease Centre (A.M.), Neuromuscular Department, Institute of Neurology, University College London; Translational Medicine and Therapeutics (J.S., S.M.H.), William Harvey Research Institute, Barts and the London, Queen Mary University of London; Department of Immunobiology (T.T.), School of Immunology & Microbial Sciences, King's College London; Nuffield Department of Clinical Neurosciences (M.R.T.), University of Oxford; and Sheffield Institute for Translational Neuroscience (P.J.S.), University of Sheffield, UK
| | - Sian M Henson
- From the Neuroscience and Trauma Centre (O.Y., A.M.), Blizard Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London; Queen Square Motor Neuron Disease Centre (A.M.), Neuromuscular Department, Institute of Neurology, University College London; Translational Medicine and Therapeutics (J.S., S.M.H.), William Harvey Research Institute, Barts and the London, Queen Mary University of London; Department of Immunobiology (T.T.), School of Immunology & Microbial Sciences, King's College London; Nuffield Department of Clinical Neurosciences (M.R.T.), University of Oxford; and Sheffield Institute for Translational Neuroscience (P.J.S.), University of Sheffield, UK
| | - Andrea Malaspina
- From the Neuroscience and Trauma Centre (O.Y., A.M.), Blizard Institute, Barts and The London School of Medicine & Dentistry, Queen Mary University of London; Queen Square Motor Neuron Disease Centre (A.M.), Neuromuscular Department, Institute of Neurology, University College London; Translational Medicine and Therapeutics (J.S., S.M.H.), William Harvey Research Institute, Barts and the London, Queen Mary University of London; Department of Immunobiology (T.T.), School of Immunology & Microbial Sciences, King's College London; Nuffield Department of Clinical Neurosciences (M.R.T.), University of Oxford; and Sheffield Institute for Translational Neuroscience (P.J.S.), University of Sheffield, UK.
| |
Collapse
|
12
|
Zhang X, Yang Y, Wei Y, Zhao Q, Lou X. blf and the drl cluster synergistically regulate cell fate commitment during zebrafish primitive hematopoiesis. Development 2022; 149:285945. [PMID: 36420817 DOI: 10.1242/dev.200919] [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: 05/06/2022] [Accepted: 11/14/2022] [Indexed: 11/25/2022]
Abstract
Hematopoiesis is a highly coordinated process that generates all the body's blood cells, and perturbations in embryonic hematopoiesis may result in illnesses ranging from fetal anemia to various leukemias. Correct establishment of hematopoietic progenitor cell fate is essential for the development of adequate blood cell subpopulations, although regulators of cell fate commitment have not been fully defined. Here, we show that primary erythropoiesis and myelopoiesis in zebrafish embryos are synergistically regulated by blf and the drl cluster, as simultaneous depletion led to severe erythrocyte aplasia and excessive macrophage formation at the expense of neutrophil development. Integrative analysis of transcriptome- and genome-wide binding data revealed that blf and drl cluster genes are responsible for constraining the expression of vasculogenesis-promoting genes in the intermediate cell mass and monocytopoiesis-promoting genes in the rostral blood island. This indicates that blf and drl cluster genes act as determinants of the fate commitment of erythroid and myeloid progenitor cells. Furthermore, a rescue screen demonstrated that Zfp932 is a potential mammalian functional equivalent to zebrafish blf and drl cluster genes. Our data provide insight into conserved cell fate commitment mechanisms of primitive hematopoiesis.
Collapse
Affiliation(s)
- Xue Zhang
- Medical School, Nanjing University, Nanjing, 210093, China
| | - Yuxi Yang
- Medical School, Nanjing University, Nanjing, 210093, China
| | - Yuxuan Wei
- Medical School, Nanjing University, Nanjing, 210093, China
| | - Qingshun Zhao
- Medical School, Nanjing University, Nanjing, 210093, China
| | - Xin Lou
- Research Institute of Intelligent Computing, Zhejiang Lab, Hangzhou, 311100, China
| |
Collapse
|
13
|
Park SY, Muschalik N, Chadwick J, Munro S. In vivo characterization of Drosophila golgins reveals redundancy and plasticity of vesicle capture at the Golgi apparatus. Curr Biol 2022; 32:4549-4564.e6. [PMID: 36103876 PMCID: PMC9849145 DOI: 10.1016/j.cub.2022.08.054] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 06/29/2022] [Accepted: 08/18/2022] [Indexed: 01/26/2023]
Abstract
The Golgi is the central sorting station in the secretory pathway and thus the destination of transport vesicles arriving from the endoplasmic reticulum and endosomes and from within the Golgi itself. Cell viability, therefore, requires that the Golgi accurately receives multiple classes of vesicle. One set of proteins proposed to direct vesicle arrival at the Golgi are the golgins, long coiled-coil proteins localized to specific parts of the Golgi stack. In mammalian cells, three of the golgins, TMF, golgin-84, and GMAP-210, can capture intra-Golgi transport vesicles when placed in an ectopic location. However, the individual golgins are not required for cell viability, and mouse knockout mutants only have defects in specific tissues. To further illuminate this system, we examine the Drosophila orthologs of these three intra-Golgi golgins. We show that ectopic forms can capture intra-Golgi transport vesicles, but strikingly, the cargo present in the vesicles captured by each golgin varies between tissues. Loss-of-function mutants show that the golgins are individually dispensable, although the loss of TMF recapitulates the male fertility defects observed in mice. However, the deletion of multiple golgins results in defects in glycosylation and loss of viability. Examining the vesicles captured by a particular golgin when another golgin is missing reveals that the vesicle content in one tissue changes to resemble that of a different tissue. This reveals a plasticity in Golgi organization between tissues, providing an explanation for why the Golgi is sufficiently robust to tolerate the loss of many of the individual components of its membrane traffic machinery.
Collapse
Affiliation(s)
- Sung Yun Park
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Nadine Muschalik
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Jessica Chadwick
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK
| | - Sean Munro
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge CB2 0QH, UK.
| |
Collapse
|
14
|
Pan J, Purev C, Zhao H, Zhang Z, Wang F, Wendoule N, Qi G, Liu Y, Zhou H. Discovery of exercise-related genes and pathway analysis based on comparative genomes of Mongolian originated Abaga and Wushen horse. Open Life Sci 2022; 17:1269-1281. [PMID: 36249530 PMCID: PMC9518662 DOI: 10.1515/biol-2022-0487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 07/21/2022] [Accepted: 07/28/2022] [Indexed: 11/17/2022] Open
Abstract
The Mongolian horses have excellent endurance and stress resistance to adapt to the cold and harsh plateau conditions. Intraspecific genetic diversity is mainly embodied in various genetic advantages of different branches of the Mongolian horse. Since people pay progressive attention to the athletic performance of horse, we expect to guide the exercise-oriented breeding of horses through genomics research. We obtained the clean data of 630,535,376,400 bp through the entire genome second-generation sequencing for the whole blood of four Abaga horses and ten Wushen horses. Based on the data analysis of single nucleotide polymorphism, we severally detected that 479 and 943 positively selected genes, particularly exercise related, were mainly enriched on equine chromosome 4 in Abaga horses and Wushen horses, which implied that chromosome 4 may be associated with the evolution of the Mongolian horse and athletic performance. Four hundred and forty genes of positive selection were enriched in 12 exercise-related pathways and narrowed in 21 exercise-related genes in Abaga horse, which were distinguished from Wushen horse. So, we speculated that the Abaga horse may have oriented genes for the motorial mechanism and 21 exercise-related genes also provided a molecular genetic basis for exercise-directed breeding of the Mongolian horse.
Collapse
Affiliation(s)
- Jing Pan
- Faculty of Life Sciences, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia Autonomous Region, People’s Republic of China
- Department of Reproductive Medicine, Inner Mongolia Maternal and Child Health Care Hospitaly, Hohhot, Inner Mongolia Autonomous Region, People’s Republic of China
| | - Chimge Purev
- Mongolia-China Joint Laboratory of Applied Molecular Biology, “Administration of the Science Park” CSTI, Ulaanbaatar, Mongolia
| | - Hongwei Zhao
- Beijing 8omics Gene Technology Co. Ltd, Beijing, People’s Republic of China
| | - Zhipeng Zhang
- Faculty of Life Sciences, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia Autonomous Region, People’s Republic of China
| | - Feng Wang
- Faculty of Life Sciences, Nankai University, Tianjin, People’s Republic of China
| | - Nashun Wendoule
- Animal Husbandry Workstation of Ewenki Autonomous County, Hulun Buir, Inner Mongolia Autonomous Region, People’s Republic of China
| | - Guichun Qi
- Bayanta Village of Animal Husbandry and Veterinary Station of Ewenki Autonomous County, Hulun Buir, Inner Mongolia Autonomous Region, People’s Republic of China
| | - Yongbin Liu
- Sheep Collaboration and Innovation Center, Inner Mongolia Universityy, Hohhot, Inner Mongolia Autonomous Region, People’s Republic of China
| | - Huanmin Zhou
- Faculty of Life Sciences, Inner Mongolia Agricultural University, Hohhot, Inner Mongolia Autonomous Region, People’s Republic of China
- Sheep Collaboration and Innovation Center, Inner Mongolia Universityy, Hohhot, Inner Mongolia Autonomous Region, People’s Republic of China
| |
Collapse
|
15
|
Pan S, Liu X, Liu T, Zhao Z, Dai Y, Wang YY, Jia P, Liu F. Causal Inference of Genetic Variants and Genes in Amyotrophic Lateral Sclerosis. Front Genet 2022; 13:917142. [PMID: 35812739 PMCID: PMC9257137 DOI: 10.3389/fgene.2022.917142] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Accepted: 05/16/2022] [Indexed: 11/13/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal progressive multisystem disorder with limited therapeutic options. Although genome-wide association studies (GWASs) have revealed multiple ALS susceptibility loci, the exact identities of causal variants, genes, cell types, tissues, and their functional roles in the development of ALS remain largely unknown. Here, we reported a comprehensive post-GWAS analysis of the recent large ALS GWAS (n = 80,610), including functional mapping and annotation (FUMA), transcriptome-wide association study (TWAS), colocalization (COLOC), and summary data-based Mendelian randomization analyses (SMR) in extensive multi-omics datasets. Gene property analysis highlighted inhibitory neuron 6, oligodendrocytes, and GABAergic neurons (Gad1/Gad2) as functional cell types of ALS and confirmed cerebellum and cerebellar hemisphere as functional tissues of ALS. Functional annotation detected the presence of multiple deleterious variants at three loci (9p21.2, 12q13.3, and 12q14.2) and highlighted a list of SNPs that are potentially functional. TWAS, COLOC, and SMR identified 43 genes at 24 loci, including 23 novel genes and 10 novel loci, showing significant evidence of causality. Integrating multiple lines of evidence, we further proposed that rs2453555 at 9p21.2 and rs229243 at 14q12 functionally contribute to the development of ALS by regulating the expression of C9orf72 in pituitary and SCFD1 in skeletal muscle, respectively. Together, these results advance our understanding of the biological etiology of ALS, feed into new therapies, and provide a guide for subsequent functional experiments.
Collapse
Affiliation(s)
- Siyu Pan
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xinxuan Liu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, China
| | - Tianzi Liu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
| | - Zhongming Zhao
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Yulin Dai
- Center for Precision Health, School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Yin-Ying Wang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
| | - Peilin Jia
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- *Correspondence: Fan Liu, ; Peilin Jia,
| | - Fan Liu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, China
- School of Future Technology, University of Chinese Academy of Sciences, Beijing, China
- *Correspondence: Fan Liu, ; Peilin Jia,
| |
Collapse
|
16
|
Siokas V, Aloizou AM, Liampas I, Bakirtzis C, Nasios G, Paterakis K, Sgantzos M, Bogdanos DP, Spandidos DA, Tsatsakis A, Mitsias PD, Dardiotis E. Lack of an association between SCFD1 rs10139154 polymorphism and amyotrophic lateral sclerosis. Mol Med Rep 2022; 25:146. [PMID: 35234271 PMCID: PMC8915390 DOI: 10.3892/mmr.2022.12662] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/15/2022] [Indexed: 11/05/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disease. Through a genome‑wide association study (GWAS), the Sec1 family domain‑containing protein 1 (SCFD1) rs10139154 variant at 14q12 has emerged as a risk factor gene for ALS. Moreover, it has been reported to influence the age at onset (AAO) of patients with ALS. The aim of the present study was to assess the association of the SCFD1 rs10139154 polymorphism with the risk of developing ALS. For this purpose, 155 patients with sporadic ALS and 155 healthy controls were genotyped for the SCFD1 rs10139154. The effect of the SCFD1 rs10139154 polymorphism was then examined on the following parameters: i) The risk of developing ALS; ii) the AAO of ALS; iii) the site of ALS onset (patients with bulbar onset ALS vs. healthy controls; and patients with limb onset ALS vs. healthy controls); and iv) the AAO of ALS onset with subgroup analyses based on the site of onset (bulbar and limb, crude and adjusted for sex). The analysis of all the outcomes was performed assuming five genetic models. Crude and adjusted analyses were applied. The threshold for statistical significance was set at 0.05. The results revealed no association between SCFD1 rs10139154 and any of the examined phenotypes in any of the models examined. On the whole, based on the findings of the present study, SCFD1 rs10139154 does not appear to play a determining role in the risk of developing ALS.
Collapse
Affiliation(s)
- Vasileios Siokas
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41100 Larissa, Greece
| | - Athina-Maria Aloizou
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41100 Larissa, Greece
| | - Ioannis Liampas
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41100 Larissa, Greece
| | - Christos Bakirtzis
- B' Department of Neurology, AHEPA University Hospital, Aristotle University of Thessaloniki, 54636 Thessaloniki, Greece
| | - Grigorios Nasios
- Department of Speech and Language Therapy, University of Ioannina, 45500 Ioannina, Greece
| | - Konstantinos Paterakis
- Department of Neurosurgery, University Hospital of Larissa, Medical School, University of Thessaly, 41100 Larissa, Greece
| | - Markos Sgantzos
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41100 Larissa, Greece
| | - Dimitrios P Bogdanos
- Department of Rheumatology and Clinical Immunology, University General Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41100 Larissa, Greece
| | - Demetrios A Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Aristidis Tsatsakis
- Laboratory of Toxicology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Panayiotis D Mitsias
- Department of Neurology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Efthimios Dardiotis
- Department of Neurology, Laboratory of Neurogenetics, University Hospital of Larissa, Faculty of Medicine, School of Health Sciences, University of Thessaly, 41100 Larissa, Greece
| |
Collapse
|
17
|
Hirata Y, Matsui Y, Wada I, Hosokawa N. ER-to-Golgi trafficking of procollagen III via conventional vesicular and tubular carriers. Mol Biol Cell 2022; 33:ar21. [PMID: 35044867 PMCID: PMC9250382 DOI: 10.1091/mbc.e21-07-0372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Collagen is the major protein component of the extracellular matrix. Synthesis of procollagens starts in the endoplasmic reticulum (ER), and three ⍺ chains form a rigid triple helix 300-400 nm in length. It remains unclear how such a large cargo is transported from the ER to the Golgi apparatus. In this study, to elucidate the intracellular transport of fibril-forming collagens, we fused cysteine-free GFP to the N-telopeptide region of procollagen III (GFP-COL3A1) and analyzed transport by live-cell imaging. We found that the maturation dynamics of procollagen III were largely different from those of network-forming procollagen IV (Matsui et al. 2020). Proline hydroxylation of procollagen III uniquely triggered the formation of intralumenal droplet-like structures similar to events caused by liquid-liquid phase separation, and ER exit sites surrounded large droplets containing chaperones. Procollagen III was transported to the Golgi apparatus via vesicular and tubular carriers containing ERGIC53 and RAB1B; this process required TANGO1 and CUL3, which we previously reported were dispensable for procollagen IV. GFP-COL3A1 and mCherry-⍺1AT were co-transported in the same vesicle. Based on these findings, we propose that shortly after ER exit, enlarged carriers containing procollagen III fuse to ERGIC for transport to the Golgi apparatus by conventional cargo carriers. [Media: see text] [Media: see text] [Media: see text] [Media: see text] [Media: see text] [Media: see text] [Media: see text] [Media: see text] [Media: see text] [Media: see text] [Media: see text] [Media: see text] [Media: see text].
Collapse
Affiliation(s)
- Yukihiro Hirata
- Laboratory of Molecular and Cellular Biology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Yuto Matsui
- Laboratory of Molecular and Cellular Biology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Ikuo Wada
- Department of Cell Science, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Nobuko Hosokawa
- Laboratory of Molecular and Cellular Biology, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
18
|
Huang H, Ouyang Q, Zhu M, Yu H, Mei K, Liu R. mTOR-mediated phosphorylation of VAMP8 and SCFD1 regulates autophagosome maturation. Nat Commun 2021; 12:6622. [PMID: 34785650 PMCID: PMC8595342 DOI: 10.1038/s41467-021-26824-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 10/20/2021] [Indexed: 02/06/2023] Open
Abstract
The mammalian target of rapamycin (mTORC1) has been shown to regulate autophagy at different steps. However, how mTORC1 regulates the N-ethylmaleimide-sensitive protein receptor (SNARE) complex remains elusive. Here we show that mTORC1 inhibits formation of the SNARE complex (STX17-SNAP29-VAMP8) by phosphorylating VAMP8, thereby blocking autophagosome-lysosome fusion. A VAMP8 phosphorylation mimic mutant is unable to promote autophagosome-lysosome fusion in vitro. Furthermore, we identify SCFD1, a Sec1/Munc18-like protein, that localizes to the autolysosome and is required for SNARE complex formation and autophagosome-lysosome fusion. VAMP8 promotes SCFD1 recruitment to autolysosomes when dephosphorylated. Consistently, phosphorylated VAMP8 or SCFD1 depletion inhibits autophagosome-lysosome fusion, and expression of phosphomimic VAMP8 leads to increased lipid droplet accumulation when expressed in mouse liver. Thus, our study supports that mTORC1-mediated phosphorylation of VAMP8 blocks SCFD1 recruitment, thereby inhibiting STX17-SNAP29-VAMP8 complex formation and autophagosome-lysosome fusion. Autophagy relies on coordinated fusion of organelle membranes, although the interplay between the regulatory machinery is not well studied. Here, the authors show that SNARE complex formation is inhibited by mTORC1 phosphorylation of VAMP8, which prevents autophagosome-lysosome fusion.
Collapse
Affiliation(s)
- Hong Huang
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China.,School of Life Sciences, Nanjing Agricultural University, Nanjing, China.,National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China.,Jiangsu Collaborative Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, China
| | - Qinqin Ouyang
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China.,School of Life Sciences, Nanjing Agricultural University, Nanjing, China.,National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China.,Jiangsu Collaborative Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, China
| | - Min Zhu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Haijia Yu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Kunrong Mei
- School of Pharmaceutical Science and Technology, Tianjin University, Tianjing, China
| | - Rong Liu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China. .,School of Life Sciences, Nanjing Agricultural University, Nanjing, China. .,National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China. .,Jiangsu Collaborative Innovation Center of Meat Production and Processing, Nanjing Agricultural University, Nanjing, China.
| |
Collapse
|
19
|
Iacoangeli A, Fogh I, Selvackadunco S, Topp SD, Shatunov A, van Rheenen W, Al-Khleifat A, Opie-Martin S, Ratti A, Calvo A, Van Damme P, Robberecht W, Chio A, Dobson RJ, Hardiman O, Shaw CE, van den Berg LH, Andersen PM, Smith BN, Silani V, Veldink JH, Breen G, Troakes C, Al-Chalabi A, Jones AR. SCFD1 expression quantitative trait loci in amyotrophic lateral sclerosis are differentially expressed. Brain Commun 2021; 3:fcab236. [PMID: 34708205 PMCID: PMC8545614 DOI: 10.1093/braincomms/fcab236] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 08/05/2021] [Accepted: 08/12/2021] [Indexed: 11/14/2022] Open
Abstract
Evidence indicates that common variants found in genome-wide association studies increase risk of disease through gene regulation via expression Quantitative Trait Loci. Using multiple genome-wide methods, we examined if Single Nucleotide Polymorphisms increase risk of Amyotrophic Lateral Sclerosis through expression Quantitative Trait Loci, and whether expression Quantitative Trait Loci expression is consistent across people who had Amyotrophic Lateral Sclerosis and those who did not. In combining public expression Quantitative Trait Loci data with Amyotrophic Lateral Sclerosis genome-wide association studies, we used Summary-data-based Mendelian Randomization to confirm that SCFD1 was the only gene that was genome-wide significant in mediating Amyotrophic Lateral Sclerosis risk via expression Quantitative Trait Loci (Summary-data-based Mendelian Randomization beta = 0.20, standard error = 0.04, P-value = 4.29 × 10-6). Using post-mortem motor cortex, we tested whether expression Quantitative Trait Loci showed significant differences in expression between Amyotrophic Lateral Sclerosis (n = 76) and controls (n = 25), genome-wide. Of 20 757 genes analysed, the two most significant expression Quantitative Trait Loci to show differential in expression between Amyotrophic Lateral Sclerosis and controls involve two known Amyotrophic Lateral Sclerosis genes (SCFD1 and VCP). Cis-acting SCFD1 expression Quantitative Trait Loci downstream of the gene showed significant differences in expression between Amyotrophic Lateral Sclerosis and controls (top expression Quantitative Trait Loci beta = 0.34, standard error = 0.063, P-value = 4.54 × 10-7). These SCFD1 expression Quantitative Trait Loci also significantly modified Amyotrophic Lateral Sclerosis survival (number of samples = 4265, hazard ratio = 1.11, 95% confidence interval = 1.05-1.17, P-value = 2.06 × 10-4) and act as an Amyotrophic Lateral Sclerosis trans-expression Quantitative Trait Loci hotspot for a wider network of genes enriched for SCFD1 function and Amyotrophic Lateral Sclerosis pathways. Using gene-set analyses, we found the genes that correlate with this trans-expression Quantitative Trait Loci hotspot significantly increase risk of Amyotrophic Lateral Sclerosis (beta = 0.247, standard deviation = 0.017, P = 0.001) and schizophrenia (beta = 0.263, standard deviation = 0.008, P-value = 1.18 × 10-5), a disease that genetically correlates with Amyotrophic Lateral Sclerosis. In summary, SCFD1 expression Quantitative Trait Loci are a major factor in Amyotrophic Lateral Sclerosis, not only influencing disease risk but are differentially expressed in post-mortem Amyotrophic Lateral Sclerosis. SCFD1 expression Quantitative Trait Loci show distinct expression profiles in Amyotrophic Lateral Sclerosis that correlate with a wider network of genes that also confer risk of the disease and modify the disease's duration.
Collapse
Affiliation(s)
- Alfredo Iacoangeli
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience King's College London, 5 Cutcombe Road, London SE5 9RT, UK
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Isabella Fogh
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience King's College London, 5 Cutcombe Road, London SE5 9RT, UK
| | - Sashika Selvackadunco
- MRC London Neurodegenerative Diseases Brain Bank, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Simon D Topp
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience King's College London, 5 Cutcombe Road, London SE5 9RT, UK
| | - Aleksey Shatunov
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience King's College London, 5 Cutcombe Road, London SE5 9RT, UK
| | - Wouter van Rheenen
- Department of Neurology, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Ahmad Al-Khleifat
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience King's College London, 5 Cutcombe Road, London SE5 9RT, UK
| | - Sarah Opie-Martin
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience King's College London, 5 Cutcombe Road, London SE5 9RT, UK
| | - Antonia Ratti
- Department of Neurology-Stroke Unit and Laboratory of Neuroscience, Istituto Auxologico Italiano, IRCCS, Milan, Italy
| | - Andrea Calvo
- Department of Neuroscience 'Rita Levi Montalcini', ALS Centre, University of Turin, Torino, Italy
- Neuroscience Institute of Torino (NIT), University of Torino, Torino, Piemonte, Italy
| | - Philip Van Damme
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
- Department of Neurosciences, Laboratory of Neurobiology, VIB Center for Brain and Disease Research, Leuven, Belgium
| | - Wim Robberecht
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Adriano Chio
- Department of Neuroscience 'Rita Levi Montalcini', ALS Centre, University of Turin, Torino, Italy
- Neuroscience Institute of Torino (NIT), University of Torino, Torino, Piemonte, Italy
| | - Richard J Dobson
- Department of Biostatistics and Health Informatics, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Orla Hardiman
- Academic Unit of Neurology, Trinity Biomedical Sciences Institute, University of Dublin Trinity College, Dublin, Ireland
- Department of Neurology, Beaumont Hospital, Dublin 9, Ireland
| | - Christopher E Shaw
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience King's College London, 5 Cutcombe Road, London SE5 9RT, UK
| | - Leonard H van den Berg
- Department of Neurology, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Peter M Andersen
- Department of Clinical Sciences, Neurosciences, Umeå University, Umeå, Sweden
| | - Bradley N Smith
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience King's College London, 5 Cutcombe Road, London SE5 9RT, UK
| | - Vincenzo Silani
- Department of Neurology-Stroke Unit and Laboratory of Neuroscience, Istituto Auxologico Italiano, IRCCS, Milan, Italy
- Department of Pathophysiology and Transplantation, "Dino Ferrari" Center, Università degli Studi di Milano, Milan, Italy
- Aldo Ravelli Center for Neurotechnology and Experimental Brain Therapeutics, Università degli Studi di Milano, Milan, Italy
| | - Jan H Veldink
- Department of Neurology, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Gerome Breen
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Claire Troakes
- MRC London Neurodegenerative Diseases Brain Bank, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Ammar Al-Chalabi
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience King's College London, 5 Cutcombe Road, London SE5 9RT, UK
- Department of Neurology, King's College Hospital, London, UK
| | - Ashley R Jones
- Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience King's College London, 5 Cutcombe Road, London SE5 9RT, UK
| |
Collapse
|
20
|
MicroRNA expression profile in Lampetra morii upon Vibrio anguillarum infection and miR-4561 characterization targeting lip. Commun Biol 2021; 4:995. [PMID: 34417547 PMCID: PMC8379177 DOI: 10.1038/s42003-021-02525-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 08/06/2021] [Indexed: 12/12/2022] Open
Abstract
As a critical evolutionary pivot between invertebrates and vertebrates, lampreys provide rich genetic information. Lamprey immune protein (LIP) is a key immune regulator. MicroRNAs, well-conserved in the response to immunological stress, remain understudied in lamprey immunity. We generated a lamprey microRNA expression atlas, using deep sequencing, upon Vibrio anguillarum infection. Using comparative methods, we found that miR-4561 potentially regulates innate immunity via interaction with lip. We found a sequence in the 3′-UTR region of LIP mRNA complementary to the miR-4561 seed region; miR-4561 expression was negatively correlated with LIP. During V. anguillarum infection, miR-4561 inhibited LIP expression and bacterial clearance. Notably, LIP expression in supraneural body cells was necessary for the Gram-negative immune response. Additionally, we observed that overexpression of miR-4561 induced apoptosis in embryonic cells, suggesting a role in embryonic development. Collectively, we show lamprey microRNAs may significantly affect gene regulation and provide new insights on LIP-mediated immune regulation. Ma et al characterise differentially expressed miRNAs in Lamprey upon bacterial infection. They identify miR-4561 as being decreased after infection and show that it targets the 3′UTR of the mRNA encoding lamprey immune protein (LIP) involved in lamprey innate immunity and regulates survival in lamprey embryos.
Collapse
|
21
|
Iacoangeli A, Lin T, Al Khleifat A, Jones AR, Opie-Martin S, Coleman JRI, Shatunov A, Sproviero W, Williams KL, Garton F, Restuadi R, Henders AK, Mather KA, Needham M, Mathers S, Nicholson GA, Rowe DB, Henderson R, McCombe PA, Pamphlett R, Blair IP, Schultz D, Sachdev PS, Newhouse SJ, Proitsi P, Fogh I, Ngo ST, Dobson RJB, Wray NR, Steyn FJ, Al-Chalabi A. Genome-wide Meta-analysis Finds the ACSL5-ZDHHC6 Locus Is Associated with ALS and Links Weight Loss to the Disease Genetics. Cell Rep 2020; 33:108323. [PMID: 33113361 PMCID: PMC7610013 DOI: 10.1016/j.celrep.2020.108323] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 07/28/2020] [Accepted: 10/07/2020] [Indexed: 12/12/2022] Open
Abstract
We meta-analyze amyotrophic lateral sclerosis (ALS) genome-wide association study (GWAS) data of European and Chinese populations (84,694 individuals). We find an additional significant association between rs58854276 spanning ACSL5-ZDHHC6 with ALS (p = 8.3 × 10-9), with replication in an independent Australian cohort (1,502 individuals; p = 0.037). Moreover, B4GALNT1, G2E3-SCFD1, and TRIP11-ATXN3 are identified using a gene-based analysis. ACSL5 has been associated with rapid weight loss, as has another ALS-associated gene, GPX3. Weight loss is frequent in ALS patients and is associated with shorter survival. We investigate the effect of the ACSL5 and GPX3 single-nucleotide polymorphisms (SNPs), using longitudinal body composition and weight data of 77 patients and 77 controls. In patients' fat-free mass, although not significant, we observe an effect in the expected direction (rs58854276: -2.1 ± 1.3 kg/A allele, p = 0.053; rs3828599: -1.0 ± 1.3 kg/A allele, p = 0.22). No effect was observed in controls. Our findings support the increasing interest in lipid metabolism in ALS and link the disease genetics to weight loss in patients.
Collapse
Affiliation(s)
- Alfredo Iacoangeli
- Department of Biostatistics and Health Informatics, King's College London, London, UK; Maurice Wohl Clinical Neuroscience Institute, King's College London, Department of Basic and Clinical Neuroscience, London, UK; National Institute for Health Research Biomedical Research Centre and Dementia Unit at South London and Maudsley NHS Foundation Trust and King's College London, London, UK.
| | - Tian Lin
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Brisbane QLD 4072, Australia
| | - Ahmad Al Khleifat
- Maurice Wohl Clinical Neuroscience Institute, King's College London, Department of Basic and Clinical Neuroscience, London, UK
| | - Ashley R Jones
- Maurice Wohl Clinical Neuroscience Institute, King's College London, Department of Basic and Clinical Neuroscience, London, UK
| | - Sarah Opie-Martin
- Maurice Wohl Clinical Neuroscience Institute, King's College London, Department of Basic and Clinical Neuroscience, London, UK
| | - Jonathan R I Coleman
- National Institute for Health Research Biomedical Research Centre and Dementia Unit at South London and Maudsley NHS Foundation Trust and King's College London, London, UK; Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Aleksey Shatunov
- Maurice Wohl Clinical Neuroscience Institute, King's College London, Department of Basic and Clinical Neuroscience, London, UK
| | - William Sproviero
- Maurice Wohl Clinical Neuroscience Institute, King's College London, Department of Basic and Clinical Neuroscience, London, UK
| | - Kelly L Williams
- Centre for Motor Neuron Disease Research, Macquarie University, Sidney NSW 2109, Australia
| | - Fleur Garton
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Brisbane QLD 4072, Australia
| | - Restuadi Restuadi
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Brisbane QLD 4072, Australia
| | - Anjali K Henders
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Brisbane QLD 4072, Australia
| | - Karen A Mather
- Centre for Healthy Brain Ageing, School of Psychiatry, UNSW Medicine, University of New South Wales, Sydney NSW, Australia; Neuroscience Research Australia, Randwick NSW, Australia
| | - Merilee Needham
- Fiona Stanley Hospital, 11 Robin Warren Drive, Murdoch Perth WA 6150, Australia; Notre Dame University, 32 Mouat Street, Fremantle WA 6160, Australia; Murdoch University, 90 South Street, Murdoch WA 6150, Australia
| | - Susan Mathers
- Calvary Health Care Bethlehem, Parkdale VIC 3195, Australia
| | - Garth A Nicholson
- ANZAC Research Institute, Concord Repatriation General Hospital, Sydney NSW 2139, Australia
| | - Dominic B Rowe
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Robert Henderson
- Centre for Clinical Research, The University of Queensland, Brisbane QLD, Australia; Queensland Brain Institute, The University of Queensland, Brisbane QLD, Australia
| | - Pamela A McCombe
- Centre for Clinical Research, The University of Queensland, Brisbane QLD, Australia; Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane QLD, Australia
| | - Roger Pamphlett
- Brain and Mind Centre, The University of Sydney, Sydney NSW, Australia
| | - Ian P Blair
- Social, Genetic and Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - David Schultz
- Flinders Medical Centre, Bedford Park SA 5042, Australia
| | - Perminder S Sachdev
- Centre for Healthy Brain Ageing, School of Psychiatry, UNSW Medicine, University of New South Wales, Sydney NSW, Australia; Neuropsychiatric Institute, Prince of Wales Hospital, Sydney NSW Australia
| | - Stephen J Newhouse
- Department of Biostatistics and Health Informatics, King's College London, London, UK; National Institute for Health Research Biomedical Research Centre and Dementia Unit at South London and Maudsley NHS Foundation Trust and King's College London, London, UK; Institute of Health Informatics, University College London, London, UK
| | - Petroula Proitsi
- Maurice Wohl Clinical Neuroscience Institute, King's College London, Department of Basic and Clinical Neuroscience, London, UK
| | - Isabella Fogh
- Maurice Wohl Clinical Neuroscience Institute, King's College London, Department of Basic and Clinical Neuroscience, London, UK; Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Shyuan T Ngo
- Centre for Clinical Research, The University of Queensland, Brisbane QLD, Australia; Queensland Brain Institute, The University of Queensland, Brisbane QLD, Australia; Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane QLD, Australia; Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane QLD, Australia
| | - Richard J B Dobson
- Department of Biostatistics and Health Informatics, King's College London, London, UK; National Institute for Health Research Biomedical Research Centre and Dementia Unit at South London and Maudsley NHS Foundation Trust and King's College London, London, UK; Institute of Health Informatics, University College London, London, UK
| | - Naomi R Wray
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Brisbane QLD 4072, Australia; Queensland Brain Institute, The University of Queensland, Brisbane QLD, Australia
| | - Frederik J Steyn
- Centre for Clinical Research, The University of Queensland, Brisbane QLD, Australia; Department of Neurology, Royal Brisbane and Women's Hospital, Brisbane QLD, Australia; School of Biomedical Sciences, The University of Queensland, Brisbane QLD, Australia
| | - Ammar Al-Chalabi
- Maurice Wohl Clinical Neuroscience Institute, King's College London, Department of Basic and Clinical Neuroscience, London, UK; King's College Hospital, Bessemer Road, London SE5 9RS, UK
| |
Collapse
|
22
|
Hannigan MM, Hoffman AM, Thompson JW, Zheng T, Nicchitta CV. Quantitative Proteomics Links the LRRC59 Interactome to mRNA Translation on the ER Membrane. Mol Cell Proteomics 2020; 19:1826-1849. [PMID: 32788342 DOI: 10.1074/mcp.ra120.002228] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 08/04/2020] [Indexed: 12/22/2022] Open
Abstract
Protein synthesis on the endoplasmic reticulum (ER) requires the dynamic coordination of numerous cellular components. Together, resident ER membrane proteins, cytoplasmic translation factors, and both integral membrane and cytosolic RNA-binding proteins operate in concert with membrane-associated ribosomes to facilitate ER-localized translation. Little is known, however, regarding the spatial organization of ER-localized translation. This question is of growing significance as it is now known that ER-bound ribosomes contribute to secretory, integral membrane, and cytosolic protein synthesis alike. To explore this question, we utilized quantitative proximity proteomics to identify neighboring protein networks for the candidate ribosome interactors SEC61β (subunit of the protein translocase), RPN1 (oligosaccharyltransferase subunit), SEC62 (translocation integral membrane protein), and LRRC59 (ribosome binding integral membrane protein). Biotin labeling time course studies of the four BioID reporters revealed distinct labeling patterns that intensified but only modestly diversified as a function of labeling time, suggesting that the ER membrane is organized into discrete protein interaction domains. Whereas SEC61β and RPN1 reporters identified translocon-associated networks, SEC62 and LRRC59 reporters revealed divergent protein interactomes. Notably, the SEC62 interactome is enriched in redox-linked proteins and ER luminal chaperones, with the latter likely representing proximity to an ER luminal chaperone reflux pathway. In contrast, the LRRC59 interactome is highly enriched in SRP pathway components, translation factors, and ER-localized RNA-binding proteins, uncovering a functional link between LRRC59 and mRNA translation regulation. Importantly, analysis of the LRRC59 interactome by native immunoprecipitation identified similar protein and functional enrichments. Moreover, [35S]-methionine incorporation assays revealed that siRNA silencing of LRRC59 expression reduced steady state translation levels on the ER by ca. 50%, and also impacted steady state translation levels in the cytosol compartment. Collectively, these data reveal a functional domain organization for the ER and identify a key role for LRRC59 in the organization and regulation of local translation.
Collapse
Affiliation(s)
- Molly M Hannigan
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Alyson M Hoffman
- Department of Biochemistry, Duke University School of Medicine, Durham, North Carolina, USA
| | - J Will Thompson
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine, Durham, North Carolina, USA; Department of Duke Proteomics and Metabolomics Shared Resource, Duke University School of Medicine, Durham, North Carolina, USA
| | - Tianli Zheng
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Christopher V Nicchitta
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina, USA; Department of Biochemistry, Duke University School of Medicine, Durham, North Carolina, USA.
| |
Collapse
|
23
|
Mitobe Y, Iino K, Takayama KI, Ikeda K, Suzuki T, Aogi K, Kawabata H, Suzuki Y, Horie-Inoue K, Inoue S. PSF Promotes ER-Positive Breast Cancer Progression via Posttranscriptional Regulation of ESR1 and SCFD2. Cancer Res 2020; 80:2230-2242. [PMID: 32213542 DOI: 10.1158/0008-5472.can-19-3095] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Revised: 01/15/2020] [Accepted: 03/18/2020] [Indexed: 11/16/2022]
Abstract
Endocrine therapy is standard treatment for estrogen receptor (ER)-positive breast cancer, yet long-term treatment often causes acquired resistance, which results in recurrence and metastasis. Recent studies have revealed that RNA-binding proteins (RBP) are involved in tumorigenesis. Here, we demonstrate that PSF/SFPQ is an RBP that potentially predicts poor prognosis of patients with ER-positive breast cancer by posttranscriptionally regulating ERα (ESR1) mRNA expression. Strong PSF immunoreactivity correlated with shorter overall survival in patients with ER-positive breast cancer. PSF was predominantly expressed in a model of tamoxifen-resistant breast cancer cells, and depletion of PSF attenuated proliferation of cultured cells and xenografted tumors. PSF expression was significantly associated with estrogen signaling. PSF siRNA downregulated ESR1 mRNA by inhibiting nuclear export of the RNA. Integrative analyses of microarray and RNA immunoprecipitation sequencing also identified SCFD2, TRA2B, and ASPM as targets of PSF. Among the PSF targets, SCFD2 was a poor prognostic indicator of breast cancer and SCFD2 knockdown significantly suppressed breast cancer cell proliferation. Collectively, this study shows that PSF plays a pathophysiologic role in ER-positive breast cancer by posttranscriptionally regulating expression of its target genes such as ESR1 and SCFD2. Overall, PSF and SCFD2 could be potential diagnostic and therapeutic targets for primary and hormone-refractory breast cancers. SIGNIFICANCE: This study defines oncogenic roles of RNA-binding protein PSF, which exhibits posttranscriptional regulation in ER-positive breast cancer.
Collapse
Affiliation(s)
- Yuichi Mitobe
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, Japan
| | - Kaori Iino
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, Japan
| | - Ken-Ichi Takayama
- Department of Systems Aging Science and Medicine, Tokyo Metropolitan Institute of Gerontology, Itabashi-ku, Tokyo, Japan
| | - Kazuhiro Ikeda
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, Japan
| | - Takashi Suzuki
- Department of Pathology and Histotechnology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Kenjiro Aogi
- Department of Surgery, National Shikoku Cancer Center, Matsuyama, Ehime, Japan
| | - Hidetaka Kawabata
- Department of Breast and Endocrine Surgery, Toranomon Hospital, Minato-ku, Tokyo, Japan
| | - Yutaka Suzuki
- Department of Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo, Kashiwa, Chiba, Japan
| | - Kuniko Horie-Inoue
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, Japan
| | - Satoshi Inoue
- Division of Gene Regulation and Signal Transduction, Research Center for Genomic Medicine, Saitama Medical University, Hidaka, Saitama, Japan. .,Department of Systems Aging Science and Medicine, Tokyo Metropolitan Institute of Gerontology, Itabashi-ku, Tokyo, Japan
| |
Collapse
|
24
|
In-depth proteome analysis of more than 12,500 proteins in buffalo mammary epithelial cell line identifies protein signatures for active proliferation and lactation. Sci Rep 2020; 10:4834. [PMID: 32179766 PMCID: PMC7075962 DOI: 10.1038/s41598-020-61521-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 02/25/2020] [Indexed: 12/14/2022] Open
Abstract
The mature mammary gland is made up of a network of ducts that terminates in alveoli. The innermost layer of alveoli is surrounded by the differentiated mammary epithelial cells (MECs), which are responsible for milk synthesis and secretion during lactation. However, the MECs are in a state of active proliferation during pregnancy, when they give rise to network like structures in the mammary gland. Buffalo (Bubalus bubalis) constitute a major source of milk for human consumption, and the MECs are the major precursor cells which are mainly responsible for their lactation potential. The proteome of MECs defines their functional state and suggests their role in various cellular activities such as proliferation and lactation. To date, the proteome profile of MECs from buffalo origin is not available. In the present study, we have profiled in-depth proteome of in vitro cultured buffalo MECs (BuMECs) during active proliferation using high throughput tandem mass spectrometry (MS). MS analysis identified a total of 8330, 5970, 5289, 4818 proteins in four sub-cellular fractions (SCFs) that included cytosolic (SCF-I), membranous and membranous organelle’s (SCF-II), nuclear (SCF-III), and cytoskeletal (SCF-IV). However, 792 proteins were identified in the conditioned media, which represented the secretome. Altogether, combined analysis of all the five fractions (SCFs- I to IV, and secretome) revealed a total of 12,609 non-redundant proteins. The KEGG analysis suggested that these proteins were associated with 325 molecular pathways. Some of the highly enriched molecular pathways observed were metabolic, MAPK, PI3-AKT, insulin, estrogen, and cGMP-PKG signalling pathway. The newly identified proteins in this study are reported to be involved in NOTCH signalling, transport and secretion processes.
Collapse
|
25
|
Chen Y, Lee K, Yang Y, Kawazoe N, Chen G. PLGA-collagen-ECM hybrid meshes mimicking stepwise osteogenesis and their influence on the osteogenic differentiation of hMSCs. Biofabrication 2020; 12:025027. [DOI: 10.1088/1758-5090/ab782b] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
26
|
Linders PT, Horst CVD, Beest MT, van den Bogaart G. Stx5-Mediated ER-Golgi Transport in Mammals and Yeast. Cells 2019; 8:cells8080780. [PMID: 31357511 PMCID: PMC6721632 DOI: 10.3390/cells8080780] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 07/25/2019] [Accepted: 07/25/2019] [Indexed: 01/12/2023] Open
Abstract
The soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) syntaxin 5 (Stx5) in mammals and its ortholog Sed5p in Saccharomyces cerevisiae mediate anterograde and retrograde endoplasmic reticulum (ER)-Golgi trafficking. Stx5 and Sed5p are structurally highly conserved and are both regulated by interactions with other ER-Golgi SNARE proteins, the Sec1/Munc18-like protein Scfd1/Sly1p and the membrane tethering complexes COG, p115, and GM130. Despite these similarities, yeast Sed5p and mammalian Stx5 are differently recruited to COPII-coated vesicles, and Stx5 interacts with the microtubular cytoskeleton, whereas Sed5p does not. In this review, we argue that these different Stx5 interactions contribute to structural differences in ER-Golgi transport between mammalian and yeast cells. Insight into the function of Stx5 is important given its essential role in the secretory pathway of eukaryotic cells and its involvement in infections and neurodegenerative diseases.
Collapse
Affiliation(s)
- Peter Ta Linders
- Tumor Immunology Lab, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Chiel van der Horst
- Tumor Immunology Lab, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Martin Ter Beest
- Tumor Immunology Lab, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Geert van den Bogaart
- Tumor Immunology Lab, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands.
- Department of Molecular Immunology, Groningen Biomolecular Sciences and Biotechnology Institute, University of Groningen, Nijenborgh 7, 9747 AG Groningen, The Netherlands.
| |
Collapse
|
27
|
Does SCFD1 rs10139154 Polymorphism Decrease Alzheimer’s Disease Risk? J Mol Neurosci 2019; 69:343-350. [DOI: 10.1007/s12031-019-01363-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 06/25/2019] [Indexed: 12/14/2022]
|
28
|
Yang Y, Li B, Zhang X, Zhao Q, Lou X. The zinc finger protein Zfpm1 modulates ventricular trabeculation through Neuregulin-ErbB signalling. Dev Biol 2019; 446:142-150. [DOI: 10.1016/j.ydbio.2019.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 12/26/2018] [Accepted: 01/01/2019] [Indexed: 01/22/2023]
|
29
|
Zhang L, Yang Y, Li B, Scott IC, Lou X. The DEAD box RNA helicase Ddx39ab is essential for myocyte and lens development in zebrafish. Development 2018; 145:dev.161018. [DOI: 10.1242/dev.161018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 03/13/2018] [Indexed: 12/20/2022]
Abstract
RNA helicases from the DEAD-box family are found in almost all organisms and have important roles in RNA metabolism including RNA synthesis, processing and degradation. The function and mechanism of action of most of these helicases in animal development and human disease are largely unexplored. In a zebrafish mutagenesis screen to identify genes essential for heart development we identified a mutant which disrupts the gene encoding the RNA helicase DEAD-box 39ab (ddx39ab). Homozygous ddx39ab mutant embryos exhibit profound cardiac and trunk muscle dystrophy, along with lens abnormalities, caused by abrupt terminal differentiation of cardiomyocyte, myoblast and lens fiber cells. Further investigation indicated that loss of ddx39ab hindered mRNA splicing of members of the kmt2 gene family, leading to mis-regulation of structural gene expression in cardiomyocyte, myoblast and lens fiber cells. Taken together, these results show that Ddx39ab plays an essential role in establishment of proper epigenetic status during differentiation of multiple cell lineages.
Collapse
Affiliation(s)
- Linlin Zhang
- Model Animal Research Center, Nanjing University, China
| | - Yuxi Yang
- Model Animal Research Center, Nanjing University, China
| | - Beibei Li
- Model Animal Research Center, Nanjing University, China
| | - Ian C. Scott
- Program in Developmental and Stem Cell Biology, The Hospital for Sick Children, Canada
- Department of Molecular Genetics, University of Toronto, Canada
| | - Xin Lou
- Model Animal Research Center, Nanjing University, China
| |
Collapse
|
30
|
Cutrona MB, Morgan NE, Simpson JC. Heritable Skeletal Disorders Arising from Defects in Processing and Transport of Type I Procollagen from the ER: Perspectives on Possible Therapeutic Approaches. Handb Exp Pharmacol 2018; 245:191-225. [PMID: 29071510 DOI: 10.1007/164_2017_67] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Rare bone disorders are a heterogeneous group of diseases, initially associated with mutations in type I procollagen (PC) genes. Recent developments from dissection at the molecular and cellular level have expanded the list of disease-causing proteins, revealing that disruption of the machinery that handles protein secretion can lead to failure in PC secretion and in several cases result in skeletal dysplasia. In parallel, cell-based in vitro studies of PC trafficking pathways offer clues to the identification of new disease candidate genes. Together, this raises the prospect of heritable bone disorders as a paradigm for biosynthetic protein traffic-related diseases, and an avenue through which therapeutic strategies can be explored.Here, we focus on human syndromes linked to defects in type I PC secretion with respect to the landscape of biosynthetic and protein transport steps within the early secretory pathway. We provide a perspective on possible therapeutic interventions for associated heritable craniofacial and skeletal disorders, considering different orders of complexity, from the cellular level by manipulation of proteostasis pathways to higher levels involving cell-based therapies for bone repair and regeneration.
Collapse
Affiliation(s)
- Meritxell B Cutrona
- School of Biology and Environmental Science, Conway Institute of Biomolecular and Biomedical Research, University College Dublin (UCD), Dublin, Ireland
| | - Niamh E Morgan
- School of Biology and Environmental Science, Conway Institute of Biomolecular and Biomedical Research, University College Dublin (UCD), Dublin, Ireland
| | - Jeremy C Simpson
- School of Biology and Environmental Science, Conway Institute of Biomolecular and Biomedical Research, University College Dublin (UCD), Dublin, Ireland.
| |
Collapse
|
31
|
Chen Y, Zhou Q, Gu X, Wei Q, Cao B, Liu H, Hou Y, Shang H. An association study between SCFD1 rs10139154 variant and amyotrophic lateral sclerosis in a Chinese cohort. Amyotroph Lateral Scler Frontotemporal Degener 2017; 19:413-418. [PMID: 29260601 DOI: 10.1080/21678421.2017.1418006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND A recent genome-wide association study (GWAS) demonstrated that the Sec1 family domain containing 1 (SCFD1) gene is associated with amyotrophic lateral sclerosis (ALS). The objective of our study was to investigate the association between the single nucleotide polymorphism (SNP) rs10139154 in the SCFD1 gene and ALS in a Chinese cohort. METHODS A cohort of 1074 sporadic ALS (SALS) patients from the Department of Neurology at the West China Hospital of Sichuan University were genotyped for rs10139154 using a polymerase chain reaction restriction fragment length polymorphism (PCR-RFLP) analysis. In addition, 927 unrelated healthy controls (HCs) from the same region were included. RESULTS After adjusting for age and sex, no significant differences in the genotype distributions and allele frequencies in the allelic, additive, dominant or recessive genetic models were found between SALS and HCs and between patients with spinal onset and bulbar onset. Remarkably, rs10139154 was shown to be associated with the age at onset (AAO) of ALS patients. Consistently, ALS patients with the "CC" genotype have an earlier mean AAO than that of patients with a "CG" and "CG + GG" genotype (p = 0.002 and 0.001, respectively). CONCLUSION Our results suggest that there is a lack of association of SCFD1 rs10139154 with the risk for ALS in a large Chinese population, but this variant may modulate the age of onset of ALS. These findings add further evidence to the suspected implication of the SCFD1 gene in the pathogenesis of disease in our ALS population.
Collapse
Affiliation(s)
- Yongping Chen
- a Department of Neurology , West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Qingqing Zhou
- a Department of Neurology , West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Xiaojing Gu
- a Department of Neurology , West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Qianqian Wei
- a Department of Neurology , West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Bei Cao
- a Department of Neurology , West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Hui Liu
- a Department of Neurology , West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Yanbing Hou
- a Department of Neurology , West China Hospital, Sichuan University , Chengdu , Sichuan , China
| | - Huifang Shang
- a Department of Neurology , West China Hospital, Sichuan University , Chengdu , Sichuan , China
| |
Collapse
|