1
|
Akçeşme B, Hekimoğlu H, Chirasani VR, İş Ş, Atmaca HN, Waldern JM, Ramos SBV. Identification of deleterious non-synonymous single nucleotide polymorphisms in the mRNA decay activator ZFP36L2. RNA Biol 2025; 22:1-15. [PMID: 39668715 DOI: 10.1080/15476286.2024.2437590] [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] [Revised: 10/31/2024] [Accepted: 11/19/2024] [Indexed: 12/14/2024] Open
Abstract
More than 4,000 single nucleotide polymorphisms (SNP) variants have been identified in the human ZFP36L2 gene, however only a few have been studied in the context of protein function. The tandem zinc finger domain of ZFP36L2, an RNA binding protein, is the functional domain that binds to its target mRNAs. This protein/RNA interaction triggers mRNA degradation, controlling gene expression. We identified 32 non-synonymous SNPs (nsSNPs) in the tandem zinc finger domain of ZFP36L2 that could have possible deleterious impacts in humans. Using different bioinformatic strategies, we prioritized five among these 32 nsSNPs, namely rs375096815, rs1183688047, rs1214015428, rs1215671792 and rs920398592 to be validated. When we experimentally tested the functionality of these protein variants using gel shift assays, all five (Y154H, R160W, R184C, G204D, and C206F) resulted in a dramatic reduction in RNA binding compared to the WT protein. To understand the mechanistic effect of these variants on the protein/RNA interaction, we employed DUET, DynaMut and PyMOL to investigate structural changes in the protein. Additionally, we conducted Molecular Docking and Molecular Dynamics Simulations to fine tune the active behaviour of this biomolecular system at an atomic level. Our results propose atomic explanations for the impact of each of these five genetic variants identified.
Collapse
Affiliation(s)
- Betül Akçeşme
- Program of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Ilidža/Sarajevo, Bosnia and Herzegovina
- Hamidiye School of Medicine, Department of Basic Medical Sciences, Division of Medical Biology, University of Health Sciences, Üsküdar/İstanbul, Turkey
| | - Hilal Hekimoğlu
- Institute of Health Sciences, İstanbul University, Fatih/İstanbul, Turkey
| | - Venkat R Chirasani
- Biochemistry and Biophysics Department, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
- Biochemistry and Biophysics Department, R. L. Juliano Structural Bioinformatics Core, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - Şeyma İş
- Hamidiye School of Medicine, Department of Basic Medical Sciences, Division of Medical Biology, University of Health Sciences, Üsküdar/İstanbul, Turkey
- Department of Molecular Biotechnology, Division of Bioinformatics, Turkish-German University, Beykoz/İstanbul, Turkey
| | - Habibe Nur Atmaca
- Department of Medical Biology, Faculty of Medicine, Ondokuz Mayıs University, Atakum/Samsun, Turkey
| | - Justin M Waldern
- Biology Department, University of North Carolina, Chapel Hill, NC, USA
| | - Silvia B V Ramos
- Biochemistry and Biophysics Department, School of Medicine, University of North Carolina, Chapel Hill, NC, USA
| |
Collapse
|
2
|
Madarász K, Mótyán JA, Chang Chien YC, Bedekovics J, Csoma SL, Méhes G, Mokánszki A. BCOR-rearranged sarcomas: In silico insights into altered domains and BCOR interactions. Comput Biol Med 2025; 191:110144. [PMID: 40228447 DOI: 10.1016/j.compbiomed.2025.110144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 04/01/2025] [Accepted: 04/03/2025] [Indexed: 04/16/2025]
Abstract
BCOR (BCL-6 corepressor) rearranged small round cell sarcoma (BRS) represents an uncommon soft tissue malignancy, frequently characterized by the BCOR::CCNB3 fusion. Other noteworthy fusions include BCOR::MAML3, BCOR::CLGN, BCOR::MAML1, ZC3H7B::BCOR, KMT2D::BCOR, CIITA::BCOR, RTL9::BCOR, and AHR::BCOR. The BCOR gene plays a pivotal role in the Polycomb Repressive Complex 1 (PRC1), essential for histone modification and gene silencing. It interfaces with the Polycomb group RING finger homolog (PCGF1). This study employed comprehensive in silico methodologies to investigate the structural and functional effects of BCOR fusion events in BRS. The analysis revealed significant alterations in the domain architecture of BCOR, which resulted in the loss of BCL6-regulated transcriptional repression. Furthermore, IUPred3 prediction indicated a significant increase in disorder in the C-terminal regions of the BCOR in the fusion proteins. A detailed analysis of the physicochemical properties by ProtParam revealed a decrease in isoelectric point, stability, and hydrophobicity. The analysis of protein structures predicted by AlphaFold3 using the PRODIGY algorithm revealed statistically significant deviations in binding affinities for BCOR-PCGF1 dimers and a non-canonical PRC1 variant tetramer compared to the wild-type BCOR. The findings provide a comprehensive summary and elucidation of the fusion proteome associated with BRS, suggesting a substantial impact on the stability and functionality of the fusion proteins, thereby contributing to the oncogenic mechanisms underlying BRS. In this study, we provide the first compilation and comparative analysis of the known BCOR fusions of BRS and introduce a new in silico approach to enhance a better understanding of the molecular basis of BRS.
Collapse
Affiliation(s)
- Kristóf Madarász
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032, Debrecen, Hungary.
| | - János András Mótyán
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, 4032, Debrecen, Hungary.
| | - Yi-Che Chang Chien
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032, Debrecen, Hungary.
| | - Judit Bedekovics
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032, Debrecen, Hungary.
| | - Szilvia Lilla Csoma
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032, Debrecen, Hungary.
| | - Gábor Méhes
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032, Debrecen, Hungary.
| | - Attila Mokánszki
- Department of Pathology, Faculty of Medicine, University of Debrecen, 4032, Debrecen, Hungary.
| |
Collapse
|
3
|
Wang L, He LF, Xiong X, Wu ZN, Tian M, Cao GQ, Lu HX, Ji XP, Zhang YL, Kovarik P, Zhang W, Liu Y. Deletion of smooth muscle ZFP36 promotes neointimal hyperplasia in mice. Acta Pharmacol Sin 2025; 46:1317-1328. [PMID: 39890944 PMCID: PMC12032071 DOI: 10.1038/s41401-024-01473-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 12/29/2024] [Indexed: 02/03/2025]
Abstract
Platelet-derived growth factor (PDGF-BB) released from the injured intima induces the proliferation and migration of vascular smooth muscle cells (VSMCs), which is the key mechanism of neointimal hyperplasia. Zinc finger 36 (ZFP36), a widespread RNA-binding protein, is important for pathological processes in many diseases. In this study we investigated the role of ZFP36 in VSMCs proliferation, migration and neointimal hyperplasia in mice. We generated smooth muscle-specific Zfp36 knockout (Zfp36SMKO) mice, and established restenosis mouse models by ligation of left carotid artery in Zfp36SMKO mice. We showed that the expression levels of ZFP36 were significantly decreased in human atherosclerotic coronary arteries and murine injured carotid arteries compared with controls. Compared to control Zfp36fl/fl mice, Zfp36SMKO mice displayed accelerated neointimal hyperplasia. In cultured mouse VSMCs, PDGF-BB (20 ng/mL) significantly downregulated ZFP36 expression through KLF4 binding site in Zfp36 promoter. We revealed that ZFP36 could bind to the mRNA of cell migration-inducing protein (CEMIP) and promoted its degradation in VSMCs, thereby reducing the expression of CEMIP protein. Knockdown of Cemip inhibited VSMCs proliferation and migration induced by Zfp36 knockout, thereby suppressing neointimal hyperplasia in Zfp36SMKO mice. We conclude that vascular smooth muscle ZFP36 has a protective effect against neointimal hyperplasia by reducing CEMIP expression. ZFP36 is downregulated by vascular injury and PDGF-BB treatment, which promotes VSMCs proliferation and migration and neointima formation. The results suggest that targeting ZFP36 may represent a novel therapeutic strategy for preventing or treating neointimal hyperplasia and related cardiovascular diseases.
Collapse
Affiliation(s)
- Lei Wang
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, 250012, China
- Department of Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Li-Fan He
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, 250012, China
- Department of Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Xiao Xiong
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Zhi-Nan Wu
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Mi Tian
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, 250014, China
| | - Guang-Qing Cao
- Department of Cardiac Surgery, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Hui-Xia Lu
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Xiao-Ping Ji
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Yan-Ling Zhang
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Pavel Kovarik
- Max Perutz Labs, University of Vienna, Vienna Biocenter (VBC), Vienna, Austria
| | - Wencheng Zhang
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, 250012, China.
| | - Yan Liu
- State Key Laboratory for Innovation and Transformation of Luobing Theory; Key Laboratory of Cardiovascular Remodeling and Function Research of MOE, NHC, CAMS and Shandong Province; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, 250012, China.
| |
Collapse
|
4
|
Hubiernatorova A, Novak J, Vaskovicova M, Sekac D, Kropyvko S, Hodny Z. Tristetraprolin affects invasion-associated genes expression and cell motility in triple-negative breast cancer model. Cytoskeleton (Hoboken) 2025; 82:311-326. [PMID: 39319680 PMCID: PMC12063522 DOI: 10.1002/cm.21934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 09/05/2024] [Accepted: 09/09/2024] [Indexed: 09/26/2024]
Abstract
Tristetraprolin (TTP) is an RNA-binding protein that negatively regulates its target mRNAs and has been shown to inhibit tumor progression and invasion. Tumor invasion requires precise regulation of cytoskeletal components, and dysregulation of cytoskeleton-associated genes can significantly alter cell motility and invasive capability. Several genes, including SH3PXD2A, SH3PXD2B, CTTN, WIPF1, and WASL, are crucial components of the cytoskeleton reorganization machinery and are essential for adequate cell motility. These genes are also involved in invasion processes, with SH3PXD2A, SH3PXD2B, WIPF1, and CTTN being key components of invadopodia-specialized structures that facilitate invasion. However, the regulation of these genes is not well understood. This study demonstrates that ectopic expression of TTP in MDA-MB-231 cells leads to decreased mRNA levels of CTTN and SH3PXD2A, as well as defects in cell motility and actin filament organization. Additionally, doxorubicin significantly increases TTP expression and reduces the mRNA levels of cytoskeleton-associated genes, enhancing our understanding of how doxorubicin may affect the transcriptional profile of cells. However, doxorubicin affects target mRNAs differently than TTP ectopic expression, suggesting it may not be the primary mechanism of doxorubicin in breast cancer (BC) treatment. High TTP expression is considered as a positive prognostic marker in multiple cancers, including BC. Given that doxorubicin is a commonly used drug for treating triple-negative BC, using TTP as a prognostic marker in this cohort of patients might be limited since it might be challenging to understand if high TTP expression occurred due to the favorable physiological state of the patient or as a consequence of treatment.
Collapse
Affiliation(s)
- Anastasiia Hubiernatorova
- Department of Functional GenomicsInstitute of Molecular Biology and Genetics NAS of UkraineKyivUkraine
- Laboratory of Cell Regeneration and PlasticityInstitute of Animal Physiology and Genetics of the Czech Academy of SciencesLibechovCzech Republic
| | - Josef Novak
- Laboratory of Genome IntegrityInstitute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| | - Michaela Vaskovicova
- Department of Cell Biology, Faculty of ScienceCharles UniversityPragueCzech Republic
- Laboratory of DNA IntegrityInstitute of Animal Physiology and Genetics of the Czech Academy of SciencesLibechovCzech Republic
| | - David Sekac
- Laboratory of Cell Regeneration and PlasticityInstitute of Animal Physiology and Genetics of the Czech Academy of SciencesLibechovCzech Republic
- Department of Cell Biology, Faculty of ScienceCharles UniversityPragueCzech Republic
| | - Serhii Kropyvko
- Department of Functional GenomicsInstitute of Molecular Biology and Genetics NAS of UkraineKyivUkraine
| | - Zdenek Hodny
- Laboratory of Genome IntegrityInstitute of Molecular Genetics of the Czech Academy of SciencesPragueCzech Republic
| |
Collapse
|
5
|
Pommier A, Bleuse S, Deletang K, Varilh J, Nadaud M, Boisguerin P, Bourdin A, Taulan-Cadars M. The RNA-Binding Protein Tristetraprolin Contributes to CFTR mRNA Stability in Cystic Fibrosis. Am J Respir Cell Mol Biol 2025; 72:320-331. [PMID: 39417720 DOI: 10.1165/rcmb.2023-0209oc] [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: 06/08/2023] [Accepted: 10/17/2024] [Indexed: 10/19/2024] Open
Abstract
Cystic fibrosis (CF) is the most common inherited disorder and is characterized by an inflammatory phenotype. We found that in bronchial epithelium reconstituted form lung tissue biopsies from patients with CF, the RNA-binding protein tristetraprolin (TTP), a key regulator of inflammation, is dysregulated in cells that strongly express cytokines and ILs. TTP activity is regulated by extensive posttranslational modifications, particularly phosphorylation. We found that, in addition to mRNA downregulation, phosphorylated TTP (which cannot bind to mRNA) accumulated in CF cultures, suggesting that the imbalance in TTP phosphorylation status could contribute to the inflammatory phenotype in CF. We confirmed TTP's destabilizing role on IL8 mRNA through its 3' UTR sequence in CF cells. We next demonstrated that TTP phosphorylation is mainly regulated by MK2 through the activation of ERK, which also was hyperphosphorylated. TTP is considered a mRNA decay factor with some exception, and we present a new positive role of TTP in CF cultures. We determined that TTP binds to specific adenylate-uridylate-rich element motifs on the 3' UTR of mRNA sequences and also, for the first time to our knowledge, to the 3' UTR of the cystic fibrosis transmembrane conductance regulator (CFTR), where TTP binding stabilizes the mRNA level. This study identified new partners that can be targeted in CF and proposes a new way to control CFTR gene expression.
Collapse
Affiliation(s)
| | - Solenne Bleuse
- Université de Montpellier, and
- PhyMedExp, INSERM U1046, CNRS UMR 9214, Université de Montpellier, Montpellier, France; and
| | - Karine Deletang
- PhyMedExp, INSERM U1046, CNRS UMR 9214, Université de Montpellier, Montpellier, France; and
| | - Jessica Varilh
- PhyMedExp, INSERM U1046, CNRS UMR 9214, Université de Montpellier, Montpellier, France; and
| | - Marion Nadaud
- PhyMedExp, INSERM U1046, CNRS UMR 9214, Université de Montpellier, Montpellier, France; and
| | - Prisca Boisguerin
- PhyMedExp, INSERM U1046, CNRS UMR 9214, Université de Montpellier, Montpellier, France; and
| | - Arnaud Bourdin
- Université de Montpellier, and
- PhyMedExp, INSERM U1046, CNRS UMR 9214, Université de Montpellier, Montpellier, France; and
- Department of Respiratory Diseases, CHU Arnaud de Villeneuve, Montpellier, France
| | - Magali Taulan-Cadars
- Université de Montpellier, and
- PhyMedExp, INSERM U1046, CNRS UMR 9214, Université de Montpellier, Montpellier, France; and
| |
Collapse
|
6
|
Huffman JE, Nicholas J, Hahn J, Heath AS, Raffield LM, Yanek LR, Brody JA, Thibord F, Almasy L, Bartz TM, Bielak LF, Bowler RP, Carrasquilla GD, Chasman DI, Chen MH, Emmert DB, Ghanbari M, Haessler J, Hottenga JJ, Kleber ME, Le NQ, Lee J, Lewis JP, Li-Gao R, Luan J, Malmberg A, Mangino M, Marioni RE, Martinez-Perez A, Pankratz N, Polasek O, Richmond A, Rodriguez BAT, Rotter JI, Steri M, Suchon P, Trompet S, Weiss S, Zare M, Auer P, Cho MH, Christofidou P, Davies G, de Geus E, Deleuze JF, Delgado GE, Ekunwe L, Faraday N, Gögele M, Greinacher A, Gao H, Howard T, Joshi PK, Kilpeläinen TO, Lahti J, Linneberg A, Naitza S, Noordam R, Paüls-Vergés F, Rich SS, Rosendaal FR, Rudan I, Ryan KA, Souto JC, van Rooij FJA, Wang H, Zhao W, Becker LC, Beswick A, Brown MR, Cade BE, Campbell H, Cho K, Crapo JD, Curran JE, de Maat MPM, Doyle M, Elliott P, Floyd JS, Fuchsberger C, Grarup N, Guo X, Harris SE, Hou L, Kolcic I, Kooperberg C, Menni C, Nauck M, O'Connell JR, Orrù V, Psaty BM, Räikkönen K, Smith JA, Soria JM, Stott DJ, van Hylckama Vlieg A, Watkins H, Willemsen G, Wilson PWF, Ben-Shlomo Y, et alHuffman JE, Nicholas J, Hahn J, Heath AS, Raffield LM, Yanek LR, Brody JA, Thibord F, Almasy L, Bartz TM, Bielak LF, Bowler RP, Carrasquilla GD, Chasman DI, Chen MH, Emmert DB, Ghanbari M, Haessler J, Hottenga JJ, Kleber ME, Le NQ, Lee J, Lewis JP, Li-Gao R, Luan J, Malmberg A, Mangino M, Marioni RE, Martinez-Perez A, Pankratz N, Polasek O, Richmond A, Rodriguez BAT, Rotter JI, Steri M, Suchon P, Trompet S, Weiss S, Zare M, Auer P, Cho MH, Christofidou P, Davies G, de Geus E, Deleuze JF, Delgado GE, Ekunwe L, Faraday N, Gögele M, Greinacher A, Gao H, Howard T, Joshi PK, Kilpeläinen TO, Lahti J, Linneberg A, Naitza S, Noordam R, Paüls-Vergés F, Rich SS, Rosendaal FR, Rudan I, Ryan KA, Souto JC, van Rooij FJA, Wang H, Zhao W, Becker LC, Beswick A, Brown MR, Cade BE, Campbell H, Cho K, Crapo JD, Curran JE, de Maat MPM, Doyle M, Elliott P, Floyd JS, Fuchsberger C, Grarup N, Guo X, Harris SE, Hou L, Kolcic I, Kooperberg C, Menni C, Nauck M, O'Connell JR, Orrù V, Psaty BM, Räikkönen K, Smith JA, Soria JM, Stott DJ, van Hylckama Vlieg A, Watkins H, Willemsen G, Wilson PWF, Ben-Shlomo Y, Blangero J, Boomsma D, Cox SR, Dehghan A, Eriksson JG, Fiorillo E, Fornage M, Hansen T, Hayward C, Ikram MA, Jukema JW, Kardia SLR, Lange LA, März W, Mathias RA, Mitchell BD, Mook-Kanamori DO, Morange PE, Pedersen O, Pramstaller PP, Redline S, Reiner A, Ridker PM, Silverman EK, Spector TD, Völker U, Wareham NJ, Wilson JF, Yao J, Trégouët DA, Johnson AD, Wolberg AS, de Vries PS, Sabater-Lleal M, Morrison AC, Smith NL. Whole-genome analysis of plasma fibrinogen reveals population-differentiated genetic regulators with putative liver roles. Blood 2024; 144:2248-2265. [PMID: 39226462 PMCID: PMC11600029 DOI: 10.1182/blood.2023022596] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 09/05/2024] Open
Abstract
ABSTRACT Genetic studies have identified numerous regions associated with plasma fibrinogen levels in Europeans, yet missing heritability and limited inclusion of non-Europeans necessitates further studies with improved power and sensitivity. Compared with array-based genotyping, whole-genome sequencing (WGS) data provide better coverage of the genome and better representation of non-European variants. To better understand the genetic landscape regulating plasma fibrinogen levels, we meta-analyzed WGS data from the National Heart, Lung, and Blood Institute's Trans-Omics for Precision Medicine (TOPMed) program (n = 32 572), with array-based genotype data from the Cohorts for Heart and Aging Research in Genomic Epidemiology Consortium (n = 131 340) imputed to the TOPMed or Haplotype Reference Consortium panel. We identified 18 loci that have not been identified in prior genetic studies of fibrinogen. Of these, 4 are driven by common variants of small effect with reported minor allele frequency (MAF) at least 10 percentage points higher in African populations. Three signals (SERPINA1, ZFP36L2, and TLR10) contain predicted deleterious missense variants. Two loci, SOCS3 and HPN, each harbor 2 conditionally distinct, noncoding variants. The gene region encoding the fibrinogen protein chain subunits (FGG;FGB;FGA) contains 7 distinct signals, including 1 novel signal driven by rs28577061, a variant common in African ancestry populations but extremely rare in Europeans (MAFAFR = 0.180; MAFEUR = 0.008). Through phenome-wide association studies in the VA Million Veteran Program, we found associations between fibrinogen polygenic risk scores and thrombotic and inflammatory disease phenotypes, including an association with gout. Our findings demonstrate the utility of WGS to augment genetic discovery in diverse populations and offer new insights for putative mechanisms of fibrinogen regulation.
Collapse
Affiliation(s)
- Jennifer E. Huffman
- Palo Alto VA Institute for Research, VA Palo Alto Heath Care System, Palo Alto, CA
- MAVERIC, VA Boston Healthcare System, Boston, MA
| | - Jayna Nicholas
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Julie Hahn
- Human Genetics Center, Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Adam S. Heath
- Human Genetics Center, Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Laura M. Raffield
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Lisa R. Yanek
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jennifer A. Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
| | - Florian Thibord
- National Heart, Lung, and Blood Institute, Division of Intramural Research, Population Sciences Branch, The Framingham Heart Study, Framingham, MA
| | - Laura Almasy
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Traci M. Bartz
- Department of Biostatistics, University of Washington, Seattle, WA
| | - Lawrence F. Bielak
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI
| | | | - Germán D. Carrasquilla
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Daniel I. Chasman
- Division of Preventive Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Ming-Huei Chen
- National Heart, Lung, and Blood Institute, Division of Intramural Research, Population Sciences Branch, The Framingham Heart Study, Framingham, MA
| | - David B. Emmert
- Institute for Biomedicine (affiliated to the University of Lübeck), Eurac Research, Bolzano, Italy
| | - Mohsen Ghanbari
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Jeffrey Haessler
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Jouke-Jan Hottenga
- Netherlands Twin Register, Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
| | - Marcus E. Kleber
- SYNLAB MVZ für Humangenetik Mannheim, Mannheim, Germany
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ngoc-Quynh Le
- Unit of Genomics of Complex Disease, Institut d’Investigació Biomèdica Sant Pau, Barcelona, Spain
| | - Jiwon Lee
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Joshua P. Lewis
- Department of Medicine, University of Maryland, Baltimore, MD
| | - Ruifang Li-Gao
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - Anni Malmberg
- Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, United Kingdom
- National Institute for Health and Care Research Biomedical Research Centre, Guy’s and St Thomas’ Foundation Trust, London, United Kingdom
| | - Riccardo E. Marioni
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Angel Martinez-Perez
- Unit of Genomics of Complex Disease, Institut de Recerca Sant Paul, Barcelona, Spain
- Centre for Biomedical Network Research on Rare Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - Nathan Pankratz
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN
| | - Ozren Polasek
- Faculty of Medicine, University of Split, Split, Croatia
| | - Anne Richmond
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Benjamin A. T. Rodriguez
- National Heart, Lung, and Blood Institute, Division of Intramural Research, Population Sciences Branch, The Framingham Heart Study, Framingham, MA
| | - Jerome I. Rotter
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Maristella Steri
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato-Cagliari, Italy
| | - Pierre Suchon
- Centre de Recherche en Cardiovascular et Nutrition, INSERM, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Aix Marseille University, Marseille, France
- Laboratory of Haematology, La Timone Hospital, Marseille, France
| | - Stella Trompet
- Section of Gerontology and Geriatrics, Department of Internal Medicin, Leiden University Medical Center, Leiden, The Netherlands
| | - Stefan Weiss
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, German Center for Cardiovascular Research, Partner Site Greifswald, University Medicine Greifswald, Greifswald, Germany
| | - Marjan Zare
- Maternal-Fetal Medicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
| | - Paul Auer
- Division of Biostatistics, Institute for Health and Equity, and Cancer Center, Medical College of Wisconsin, Milwaukee, WI
| | - Michael H. Cho
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Paraskevi Christofidou
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, United Kingdom
| | - Gail Davies
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Eco de Geus
- Netherlands Twin Register, Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
| | - Jean-François Deleuze
- Centre National de Recherche en Génomique Humaine, Commissariat a l'Energie Atomique et aux Energies Alternatives, Université Paris-Saclay, Evry, France
| | - Graciela E. Delgado
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Lynette Ekunwe
- Jackson Heart Study, University of Mississippi Medical Center, Jackson, MS
| | - Nauder Faraday
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Martin Gögele
- Institute for Biomedicine (affiliated to the University of Lübeck), Eurac Research, Bolzano, Italy
| | - Andreas Greinacher
- Department of Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
| | - He Gao
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- Medical Research Council-Public Health England Centre for Environment and Health, School of Public Health, Imperial College London, London, United Kingdom
| | - Tom Howard
- Department of Human Genetics, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX
| | - Peter K. Joshi
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland
| | - Tuomas O. Kilpeläinen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jari Lahti
- Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
| | - Allan Linneberg
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Silvia Naitza
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato-Cagliari, Italy
| | - Raymond Noordam
- Section of Gerontology and Geriatrics, Department of Internal Medicin, Leiden University Medical Center, Leiden, The Netherlands
| | - Ferran Paüls-Vergés
- Unit of Genomics of Complex Disease, Institut d’Investigació Biomèdica Sant Pau, Barcelona, Spain
| | - Stephen S. Rich
- Department of Public Health Sciences, Center for Public Health Genomics, University of Virginia, Charlottesville, VA
| | - Frits R. Rosendaal
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Igor Rudan
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland
| | | | - Juan Carlos Souto
- Unit of Genomics of Complex Disease, Institut de Recerca Sant Paul, Barcelona, Spain
- Centre for Biomedical Network Research on Rare Diseases, Instituto de Salud Carlos III, Madrid, Spain
- Unit of Thrombosis and Hemostasis, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Frank J. A. van Rooij
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Heming Wang
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Wei Zhao
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI
| | - Lewis C. Becker
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Andrew Beswick
- Translational Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Michael R. Brown
- Human Genetics Center, Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Brian E. Cade
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Harry Campbell
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland
| | - Kelly Cho
- MAVERIC, VA Boston Healthcare System, Boston, MA
- Department of Medicine, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA
| | | | - Joanne E. Curran
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX
| | - Moniek P. M. de Maat
- Department of Hematology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Margaret Doyle
- Department of Pathology and Laboratory Medicine, The University of Vermont Larner College of Medicine, Colchester, VT
| | - Paul Elliott
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- Medical Research Council-Public Health England Centre for Environment and Health, School of Public Health, Imperial College London, London, United Kingdom
- United Kingdom-Dementia Research Institute, Imperial College London, London, United Kingdom
| | - James S. Floyd
- Departments of Medicine and Epidemiology, University of Washington, Seattle, WA
| | - Christian Fuchsberger
- Institute for Biomedicine (affiliated to the University of Lübeck), Eurac Research, Bolzano, Italy
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Xiuqing Guo
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - Sarah E. Harris
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Lifang Hou
- Department of Preventive Medicine, Northwestern University, Chicago, IL
| | - Ivana Kolcic
- Faculty of Medicine, University of Split, Split, Croatia
| | - Charles Kooperberg
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, United Kingdom
| | - Matthias Nauck
- Institute of Clinical Chemistry and Laboratory Medicine, German Center for Cardiovascular Research, Partner Site Greifswald, Greifswald, Germany, University Medicine Greifswald, Greifswald, Germany
| | | | - Valeria Orrù
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Lanusei, Italy
| | - Bruce M. Psaty
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
- Department of Epidemiology, University of Washington, Seattle, WA
- Department of Health Systems and Population Health, University of Washington, Seattle, WA
| | - Katri Räikkönen
- Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
| | - Jennifer A. Smith
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI
| | - Jose Manuel Soria
- Unit of Genomics of Complex Disease, Institut de Recerca Sant Paul, Barcelona, Spain
- Centre for Biomedical Network Research on Rare Diseases, Instituto de Salud Carlos III, Madrid, Spain
| | - David J. Stott
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
| | | | - Hugh Watkins
- Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Gonneke Willemsen
- Netherlands Twin Register, Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
| | - Peter W. F. Wilson
- VA Atlanta Healthcare System, Decatur, GA
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
| | - Yoav Ben-Shlomo
- Poulation Health Sciences, University of Bristol, Bristol, United Kingdom
| | - John Blangero
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX
| | - Dorret Boomsma
- Netherlands Twin Register, Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
| | - Simon R. Cox
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Abbas Dehghan
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- Medical Research Council-Public Health England Centre for Environment and Health, School of Public Health, Imperial College London, London, United Kingdom
- United Kingdom-Dementia Research Institute, Imperial College London, London, United Kingdom
| | - Johan G. Eriksson
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Centre, Helsinki, Finland
- Department of Obstetrics and Gynecology, Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore
| | - Edoardo Fiorillo
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Lanusei, Italy
| | - Myriam Fornage
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Caroline Hayward
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - M. Arfan Ikram
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - J. Wouter Jukema
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Netherlands Heart Institute, Utrecht, The Netherlands
| | - Sharon L. R. Kardia
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI
| | - Leslie A. Lange
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Winfried März
- Synlab Academy, Synlab Holding Deutschland GmbH, and Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Rasika A. Mathias
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Braxton D. Mitchell
- Department of Medicine, University of Maryland, Baltimore, MD
- Geriatric Research and Education Clinical Center, Baltimore Veterans Administration Medical Center, Baltimore, MD
| | - Dennis O. Mook-Kanamori
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, The Netherlands
| | - Pierre-Emmanuel Morange
- Centre de Recherche en Cardiovascular et Nutrition, INSERM, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Aix Marseille University, Marseille, France
- Laboratory of Haematology, La Timone Hospital, Marseille, France
| | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Peter P. Pramstaller
- Institute for Biomedicine (affiliated to the University of Lübeck), Eurac Research, Bolzano, Italy
| | - Susan Redline
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Department of Medicine, Beth Israel Deaconness Medical Center, Boston, MA
| | - Alexander Reiner
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Paul M. Ridker
- Division of Preventive Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Edwin K. Silverman
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Tim D. Spector
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, United Kingdom
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, German Center for Cardiovascular Research, Partner Site Greifswald, University Medicine Greifswald, Greifswald, Germany
| | - Nicholas J. Wareham
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
| | - James F. Wilson
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland
| | - Jie Yao
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
| | - VA Million Veteran Program
- Palo Alto VA Institute for Research, VA Palo Alto Heath Care System, Palo Alto, CA
- MAVERIC, VA Boston Healthcare System, Boston, MA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Human Genetics Center, Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
- National Heart, Lung, and Blood Institute, Division of Intramural Research, Population Sciences Branch, The Framingham Heart Study, Framingham, MA
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA
- Department of Biostatistics, University of Washington, Seattle, WA
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI
- National Jewish Health, Denver, CO
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Division of Preventive Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Institute for Biomedicine (affiliated to the University of Lübeck), Eurac Research, Bolzano, Italy
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA
- Netherlands Twin Register, Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
- SYNLAB MVZ für Humangenetik Mannheim, Mannheim, Germany
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Unit of Genomics of Complex Disease, Institut d’Investigació Biomèdica Sant Pau, Barcelona, Spain
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Department of Medicine, University of Maryland, Baltimore, MD
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, United Kingdom
- National Institute for Health and Care Research Biomedical Research Centre, Guy’s and St Thomas’ Foundation Trust, London, United Kingdom
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Unit of Genomics of Complex Disease, Institut de Recerca Sant Paul, Barcelona, Spain
- Centre for Biomedical Network Research on Rare Diseases, Instituto de Salud Carlos III, Madrid, Spain
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN
- Faculty of Medicine, University of Split, Split, Croatia
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland, United Kingdom
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato-Cagliari, Italy
- Centre de Recherche en Cardiovascular et Nutrition, INSERM, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Aix Marseille University, Marseille, France
- Laboratory of Haematology, La Timone Hospital, Marseille, France
- Section of Gerontology and Geriatrics, Department of Internal Medicin, Leiden University Medical Center, Leiden, The Netherlands
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, German Center for Cardiovascular Research, Partner Site Greifswald, University Medicine Greifswald, Greifswald, Germany
- Maternal-Fetal Medicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- Division of Biostatistics, Institute for Health and Equity, and Cancer Center, Medical College of Wisconsin, Milwaukee, WI
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre National de Recherche en Génomique Humaine, Commissariat a l'Energie Atomique et aux Energies Alternatives, Université Paris-Saclay, Evry, France
- Jackson Heart Study, University of Mississippi Medical Center, Jackson, MS
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
- Medical Research Council-Public Health England Centre for Environment and Health, School of Public Health, Imperial College London, London, United Kingdom
- Department of Human Genetics, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
- Department of Public Health Sciences, Center for Public Health Genomics, University of Virginia, Charlottesville, VA
- Unit of Thrombosis and Hemostasis, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI
- Translational Health Sciences, University of Bristol, Bristol, United Kingdom
- Department of Medicine, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX
- Department of Hematology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Pathology and Laboratory Medicine, The University of Vermont Larner College of Medicine, Colchester, VT
- United Kingdom-Dementia Research Institute, Imperial College London, London, United Kingdom
- Departments of Medicine and Epidemiology, University of Washington, Seattle, WA
- Department of Preventive Medicine, Northwestern University, Chicago, IL
- Institute of Clinical Chemistry and Laboratory Medicine, German Center for Cardiovascular Research, Partner Site Greifswald, Greifswald, Germany, University Medicine Greifswald, Greifswald, Germany
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Lanusei, Italy
- Department of Epidemiology, University of Washington, Seattle, WA
- Department of Health Systems and Population Health, University of Washington, Seattle, WA
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
- Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- VA Atlanta Healthcare System, Decatur, GA
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
- Poulation Health Sciences, University of Bristol, Bristol, United Kingdom
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Centre, Helsinki, Finland
- Department of Obstetrics and Gynecology, Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Netherlands Heart Institute, Utrecht, The Netherlands
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
- Synlab Academy, Synlab Holding Deutschland GmbH, and Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Geriatric Research and Education Clinical Center, Baltimore Veterans Administration Medical Center, Baltimore, MD
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, The Netherlands
- Department of Medicine, Beth Israel Deaconness Medical Center, Boston, MA
- Bordeaux Population Health Research Center, INSERM UMR 1219, University of Bordeaux, Bordeaux, France
- Department of Pathology and Laboratory Medicine and UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Cardiovascular Medicine Unit, Department of Medicine, Karolinska Institutet, Center for Molecular Medicine, Stockholm, Sweden
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, WA
- Seattle Epidemiologic Research and Information Center, Department of Veterans Affairs Office of Research and Development, Seattle, WA
| | - NHLBI Trans-Omics for Precision Medicine (TOPMed) Consortium
- Palo Alto VA Institute for Research, VA Palo Alto Heath Care System, Palo Alto, CA
- MAVERIC, VA Boston Healthcare System, Boston, MA
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Human Genetics Center, Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA
- National Heart, Lung, and Blood Institute, Division of Intramural Research, Population Sciences Branch, The Framingham Heart Study, Framingham, MA
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA
- Department of Biostatistics, University of Washington, Seattle, WA
- Department of Epidemiology, School of Public Health, University of Michigan, Ann Arbor, MI
- National Jewish Health, Denver, CO
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
- Division of Preventive Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Institute for Biomedicine (affiliated to the University of Lübeck), Eurac Research, Bolzano, Italy
- Department of Epidemiology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA
- Netherlands Twin Register, Department of Biological Psychology, Vrije Universiteit, Amsterdam, The Netherlands
- SYNLAB MVZ für Humangenetik Mannheim, Mannheim, Germany
- Vth Department of Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Unit of Genomics of Complex Disease, Institut d’Investigació Biomèdica Sant Pau, Barcelona, Spain
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Department of Medicine, University of Maryland, Baltimore, MD
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
- MRC Epidemiology Unit, University of Cambridge School of Clinical Medicine, Cambridge, United Kingdom
- Department of Psychology and Logopedics, University of Helsinki, Helsinki, Finland
- Department of Twin Research and Genetic Epidemiology, Kings College London, London, United Kingdom
- National Institute for Health and Care Research Biomedical Research Centre, Guy’s and St Thomas’ Foundation Trust, London, United Kingdom
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Unit of Genomics of Complex Disease, Institut de Recerca Sant Paul, Barcelona, Spain
- Centre for Biomedical Network Research on Rare Diseases, Instituto de Salud Carlos III, Madrid, Spain
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN
- Faculty of Medicine, University of Split, Split, Croatia
- Medical Research Council Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, Scotland, United Kingdom
- The Institute for Translational Genomics and Population Sciences, Department of Pediatrics, The Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, CA
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Monserrato-Cagliari, Italy
- Centre de Recherche en Cardiovascular et Nutrition, INSERM, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, Aix Marseille University, Marseille, France
- Laboratory of Haematology, La Timone Hospital, Marseille, France
- Section of Gerontology and Geriatrics, Department of Internal Medicin, Leiden University Medical Center, Leiden, The Netherlands
- Interfaculty Institute for Genetics and Functional Genomics, Department of Functional Genomics, German Center for Cardiovascular Research, Partner Site Greifswald, University Medicine Greifswald, Greifswald, Germany
- Maternal-Fetal Medicine Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
- Division of Biostatistics, Institute for Health and Equity, and Cancer Center, Medical College of Wisconsin, Milwaukee, WI
- Channing Division of Network Medicine and Division of Pulmonary and Critical Care Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
- Lothian Birth Cohorts, Department of Psychology, University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre National de Recherche en Génomique Humaine, Commissariat a l'Energie Atomique et aux Energies Alternatives, Université Paris-Saclay, Evry, France
- Jackson Heart Study, University of Mississippi Medical Center, Jackson, MS
- Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Transfusion Medicine, University Medicine Greifswald, Greifswald, Germany
- Medical Research Council-Public Health England Centre for Environment and Health, School of Public Health, Imperial College London, London, United Kingdom
- Department of Human Genetics, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX
- Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, University of Edinburgh, Edinburgh, Scotland
- Center for Clinical Research and Prevention, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
- Department of Public Health Sciences, Center for Public Health Genomics, University of Virginia, Charlottesville, VA
- Unit of Thrombosis and Hemostasis, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Survey Research Center, Institute for Social Research, University of Michigan, Ann Arbor, MI
- Translational Health Sciences, University of Bristol, Bristol, United Kingdom
- Department of Medicine, Brigham & Women’s Hospital and Harvard Medical School, Boston, MA
- Department of Human Genetics and South Texas Diabetes and Obesity Institute, University of Texas Rio Grande Valley School of Medicine, Brownsville, TX
- Department of Hematology, Erasmus MC University Medical Center, Rotterdam, The Netherlands
- Department of Pathology and Laboratory Medicine, The University of Vermont Larner College of Medicine, Colchester, VT
- United Kingdom-Dementia Research Institute, Imperial College London, London, United Kingdom
- Departments of Medicine and Epidemiology, University of Washington, Seattle, WA
- Department of Preventive Medicine, Northwestern University, Chicago, IL
- Institute of Clinical Chemistry and Laboratory Medicine, German Center for Cardiovascular Research, Partner Site Greifswald, Greifswald, Germany, University Medicine Greifswald, Greifswald, Germany
- Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Lanusei, Italy
- Department of Epidemiology, University of Washington, Seattle, WA
- Department of Health Systems and Population Health, University of Washington, Seattle, WA
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom
- Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
- VA Atlanta Healthcare System, Decatur, GA
- Division of Cardiology, Emory University School of Medicine, Atlanta, GA
- Department of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, GA
- Poulation Health Sciences, University of Bristol, Bristol, United Kingdom
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
- Folkhälsan Research Centre, Helsinki, Finland
- Department of Obstetrics and Gynecology, Yong Loo Lin School of Medicine, National University Singapore, Singapore, Singapore
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
- Netherlands Heart Institute, Utrecht, The Netherlands
- Division of Biomedical Informatics and Personalized Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
- Synlab Academy, Synlab Holding Deutschland GmbH, and Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
- Geriatric Research and Education Clinical Center, Baltimore Veterans Administration Medical Center, Baltimore, MD
- Department of Public Health and Primary Care, Leiden University Medical Center, Leiden, The Netherlands
- Department of Medicine, Beth Israel Deaconness Medical Center, Boston, MA
- Bordeaux Population Health Research Center, INSERM UMR 1219, University of Bordeaux, Bordeaux, France
- Department of Pathology and Laboratory Medicine and UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Cardiovascular Medicine Unit, Department of Medicine, Karolinska Institutet, Center for Molecular Medicine, Stockholm, Sweden
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, WA
- Seattle Epidemiologic Research and Information Center, Department of Veterans Affairs Office of Research and Development, Seattle, WA
| | - David-Alexandre Trégouët
- Bordeaux Population Health Research Center, INSERM UMR 1219, University of Bordeaux, Bordeaux, France
| | - Andrew D. Johnson
- National Heart, Lung, and Blood Institute, Division of Intramural Research, Population Sciences Branch, The Framingham Heart Study, Framingham, MA
| | - Alisa S. Wolberg
- Department of Pathology and Laboratory Medicine and UNC Blood Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC
| | - Paul S. de Vries
- Human Genetics Center, Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Maria Sabater-Lleal
- Unit of Genomics of Complex Disease, Institut de Recerca Sant Paul, Barcelona, Spain
- Centre for Biomedical Network Research on Rare Diseases, Instituto de Salud Carlos III, Madrid, Spain
- Cardiovascular Medicine Unit, Department of Medicine, Karolinska Institutet, Center for Molecular Medicine, Stockholm, Sweden
| | - Alanna C. Morrison
- Human Genetics Center, Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX
| | - Nicholas L. Smith
- Department of Epidemiology, University of Washington, Seattle, WA
- Kaiser Permanente Washington Health Research Institute, Kaiser Permanente Washington, Seattle, WA
- Seattle Epidemiologic Research and Information Center, Department of Veterans Affairs Office of Research and Development, Seattle, WA
| |
Collapse
|
7
|
He SL, Wang X, Kim SI, Kong L, Liu A, Wang L, Wang Y, Shan L, He P, Jang JC. Modulation of stress granule dynamics by phosphorylation and ubiquitination in plants. iScience 2024; 27:111162. [PMID: 39569378 PMCID: PMC11576400 DOI: 10.1016/j.isci.2024.111162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 11/22/2024] Open
Abstract
The Arabidopsis tandem CCCH zinc finger 1 (TZF1) is an RNA-binding protein that plays a pivotal role in plant growth and stress response. In this report, we show that TZF1 contains two intrinsically disordered regions necessary for its localization to stress granules (SGs). TZF1 recruits mitogen-activated protein kinase (MAPK) signaling components and an E3 ubiquitin ligase KEEP-ON-GOING (KEG) to SGs. TZF1 is phosphorylated by MPKs and ubiquitinated by KEG. Using a high throughput Arabidopsis protoplasts transient expression system, mutant studies reveal that the phosphorylation of specific residues plays differential roles in enhancing or reducing TZF1 SG assembly and protein-protein interaction with mitogen-activated kinase kinase 5 in SGs. Ubiquitination appears to play a positive role in TZF1 SG assembly, because mutations cause a reduction of typical SGs, while enhancing the assembly of large SGs encompassing the nucleus. Together, our results demonstrate that plant SG assembly is distinctively regulated by phosphorylation and ubiquitination.
Collapse
Affiliation(s)
- Siou-Luan He
- Department of Horticulture and Crop Science, Center for Applied Plant Sciences, and Center for RNA Biology, The Ohio State University, Columbus, OH 43210, USA
| | - Xiling Wang
- Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, and University of Chinese Academy of Sciences, Beijing 10093, China
| | - Sung-Il Kim
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1085, USA
| | - Liang Kong
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1085, USA
| | - Ailing Liu
- Department of Horticulture and Crop Science, Center for Applied Plant Sciences, and Center for RNA Biology, The Ohio State University, Columbus, OH 43210, USA
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha 410128, Hunan, China
| | - Lei Wang
- Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, and University of Chinese Academy of Sciences, Beijing 10093, China
| | - Ying Wang
- Plant Pathology Department and Plant Molecular Cellular Biology Program, University of Florida, Gainesville, FL 32611, USA
| | - Libo Shan
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1085, USA
| | - Ping He
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1085, USA
| | - Jyan-Chyun Jang
- Department of Horticulture and Crop Science, Center for Applied Plant Sciences, and Center for RNA Biology, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
8
|
Zheng W, Guo J, Ma S, Sun R, Song Y, Chen Y, Mao R, Fan Y. The NEDD4-binding protein N4BP1 degrades mRNA substrates through the coding sequence independent of nonsense-mediated decay. J Biol Chem 2024; 300:107954. [PMID: 39491646 DOI: 10.1016/j.jbc.2024.107954] [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: 07/12/2024] [Revised: 10/17/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024] Open
Abstract
3'UTRs are recognized for their role in regulating mRNA turnover while the turnover of a specific group of mRNAs mediated by coding sequences (CDSs) remains poorly understood. N4BP1 is a critical inflammatory regulator in vivo with a molecular mechanism that is not yet clearly defined. Our study reveals that N4BP1 efficiently degrades its mRNA targets via CDS rather than the 3'-UTR. This CDS-dependent mRNA turnover mechanism appears to be a general feature of N4BP1, as evidenced by testing multiple mRNA substrates, such as Fos-C, Fos-B, Jun-B, and C-X-C motif chemokine ligand 1. Detailed mapping of the motif identified a crucial 33-nt (289-322) sequence near the 5'-end of Fos-C-CDS, where the presence of polyC is necessary for N4BP1-mediated degradation. Functional studies involving domain deletion and point mutations showed that both the K homology and N4BP1, YacP-like nuclease domains are essential for N4BP1 to restrict mRNA substrates. The function of N4BP1 in mRNA turnover is not dependent on nonsense-mediated decay as it efficiently restricts mRNA substrates even in cells deficient in UPF1, UPF3A, and UPF3B. Additionally, the function of N4BP1 is not reliant on LUC7L3 despite its known association with this protein. Our findings suggest that N4BP1 acts as an endoribonuclease to degrade mRNA substrates primarily through CDSs containing a C-rich motif.
Collapse
Affiliation(s)
- Wen Zheng
- Department of Pathogenic Biology, School of Medicine, Nantong University, Nantong, China; Laboratory of Medical Science, School of Medicine, Nantong University, Nantong, China
| | - Jinjing Guo
- Department of Pathogenic Biology, School of Medicine, Nantong University, Nantong, China; Laboratory of Medical Science, School of Medicine, Nantong University, Nantong, China
| | - Shuyan Ma
- Department of Pathogenic Biology, School of Medicine, Nantong University, Nantong, China; Laboratory of Medical Science, School of Medicine, Nantong University, Nantong, China
| | - Rong Sun
- Laboratory of Medical Science, School of Medicine, Nantong University, Nantong, China
| | - Yihua Song
- Department of Stomatology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Yuanmeng Chen
- Department of Pathogenic Biology, School of Medicine, Nantong University, Nantong, China; Laboratory of Medical Science, School of Medicine, Nantong University, Nantong, China
| | - Renfang Mao
- Department of Pathophysiology, School of Medicine, Nantong University, Nantong, China.
| | - Yihui Fan
- Department of Pathogenic Biology, School of Medicine, Nantong University, Nantong, China; Laboratory of Medical Science, School of Medicine, Nantong University, Nantong, China.
| |
Collapse
|
9
|
Riggs CL, Kedersha N, Amarsanaa M, Zubair SN, Ivanov P, Anderson P. UBAP2L contributes to formation of P-bodies and modulates their association with stress granules. J Cell Biol 2024; 223:e202307146. [PMID: 39007803 PMCID: PMC11248227 DOI: 10.1083/jcb.202307146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 03/06/2024] [Accepted: 06/10/2024] [Indexed: 07/16/2024] Open
Abstract
Stress triggers the formation of two distinct cytoplasmic biomolecular condensates: stress granules (SGs) and processing bodies (PBs), both of which may contribute to stress-responsive translation regulation. Though PBs can be present constitutively, stress can increase their number and size and lead to their interaction with stress-induced SGs. The mechanism of such interaction, however, is largely unknown. Formation of canonical SGs requires the RNA binding protein Ubiquitin-Associated Protein 2-Like (UBAP2L), which is a central SG node protein in the RNA-protein interaction network of SGs and PBs. UBAP2L binds to the essential SG and PB proteins G3BP and DDX6, respectively. Research on UBAP2L has mostly focused on its role in SGs, but not its connection to PBs. We find that UBAP2L is not solely an SG protein but also localizes to PBs in certain conditions, contributes to PB biogenesis and SG-PB interactions, and can nucleate hybrid granules containing SG and PB components in cells. These findings inform a new model for SG and PB formation in the context of UBAP2L's role.
Collapse
Affiliation(s)
- Claire L Riggs
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Nancy Kedersha
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Misheel Amarsanaa
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Department of Biological Sciences, Wellesley College, Wellesley, MA, USA
| | - Safiyah Noor Zubair
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Pavel Ivanov
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Paul Anderson
- Division of Rheumatology, Inflammation and Immunity, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
10
|
Gosztyla ML, Zhan L, Olson S, Wei X, Naritomi J, Nguyen G, Street L, Goda GA, Cavazos FF, Schmok JC, Jain M, Uddin Syed E, Kwon E, Jin W, Kofman E, Tankka AT, Li A, Gonzalez V, Lécuyer E, Dominguez D, Jovanovic M, Graveley BR, Yeo GW. Integrated multi-omics analysis of zinc-finger proteins uncovers roles in RNA regulation. Mol Cell 2024; 84:3826-3842.e8. [PMID: 39303722 PMCID: PMC11633308 DOI: 10.1016/j.molcel.2024.08.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 06/19/2024] [Accepted: 08/06/2024] [Indexed: 09/22/2024]
Abstract
RNA interactome studies have revealed that hundreds of zinc-finger proteins (ZFPs) are candidate RNA-binding proteins (RBPs), yet their RNA substrates and functional significance remain largely uncharacterized. Here, we present a systematic multi-omics analysis of the DNA- and RNA-binding targets and regulatory roles of more than 100 ZFPs representing 37 zinc-finger families. We show that multiple ZFPs are previously unknown regulators of RNA splicing, alternative polyadenylation, stability, or translation. The examined ZFPs show widespread sequence-specific RNA binding and preferentially bind proximal to transcription start sites. Additionally, several ZFPs associate with their targets at both the DNA and RNA levels. We highlight ZNF277, a C2H2 ZFP that binds thousands of RNA targets and acts as a multi-functional RBP. We also show that ZNF473 is a DNA/RNA-associated protein that regulates the expression and splicing of cell cycle genes. Our results reveal diverse roles for ZFPs in transcriptional and post-transcriptional gene regulation.
Collapse
Affiliation(s)
- Maya L Gosztyla
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92037, USA; Sanford Stem Cell Institute and UCSD Stem Cell Program, University of California San Diego, La Jolla, CA 92037, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Lijun Zhan
- Department of Genetics and Genome Sciences, Institute for Systems Genomics, UConn Health, Farmington, CT 06030, USA
| | - Sara Olson
- Department of Genetics and Genome Sciences, Institute for Systems Genomics, UConn Health, Farmington, CT 06030, USA
| | - Xintao Wei
- Department of Genetics and Genome Sciences, Institute for Systems Genomics, UConn Health, Farmington, CT 06030, USA
| | - Jack Naritomi
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92037, USA; Sanford Stem Cell Institute and UCSD Stem Cell Program, University of California San Diego, La Jolla, CA 92037, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Grady Nguyen
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92037, USA; Sanford Stem Cell Institute and UCSD Stem Cell Program, University of California San Diego, La Jolla, CA 92037, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Lena Street
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Grant A Goda
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA; Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Francisco F Cavazos
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Jonathan C Schmok
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92037, USA; Sanford Stem Cell Institute and UCSD Stem Cell Program, University of California San Diego, La Jolla, CA 92037, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Manya Jain
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92037, USA; Sanford Stem Cell Institute and UCSD Stem Cell Program, University of California San Diego, La Jolla, CA 92037, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Easin Uddin Syed
- Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada; Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada; School of Pharmacy, Brac University, Dhaka 1212, Bangladesh
| | - Eunjeong Kwon
- Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada
| | - Wenhao Jin
- Sanford Laboratories for Innovative Medicines, La Jolla, CA 92037, USA
| | - Eric Kofman
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92037, USA; Sanford Stem Cell Institute and UCSD Stem Cell Program, University of California San Diego, La Jolla, CA 92037, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Alexandra T Tankka
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92037, USA; Sanford Stem Cell Institute and UCSD Stem Cell Program, University of California San Diego, La Jolla, CA 92037, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Allison Li
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92037, USA; Sanford Stem Cell Institute and UCSD Stem Cell Program, University of California San Diego, La Jolla, CA 92037, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Valerie Gonzalez
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92037, USA; Sanford Stem Cell Institute and UCSD Stem Cell Program, University of California San Diego, La Jolla, CA 92037, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92037, USA
| | - Eric Lécuyer
- Institut de Recherches Cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada; Department of Biochemistry and Molecular Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada; Division of Experimental Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| | - Daniel Dominguez
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27514, USA
| | - Marko Jovanovic
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Brenton R Graveley
- Department of Genetics and Genome Sciences, Institute for Systems Genomics, UConn Health, Farmington, CT 06030, USA
| | - Gene W Yeo
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, CA 92037, USA; Sanford Stem Cell Institute and UCSD Stem Cell Program, University of California San Diego, La Jolla, CA 92037, USA; Institute for Genomic Medicine, University of California, San Diego, La Jolla, CA 92037, USA; Sanford Laboratories for Innovative Medicines, La Jolla, CA 92037, USA; Center for RNA Technologies and Therapeutics, University of California, San Diego, La Jolla, CA 92037, USA.
| |
Collapse
|
11
|
Li J, Yu J, Shen A, Lai S, Liu Z, He TS. The RNA-binding proteins regulate innate antiviral immune signaling by modulating pattern recognition receptors. Virol J 2024; 21:225. [PMID: 39304943 PMCID: PMC11414252 DOI: 10.1186/s12985-024-02503-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 09/12/2024] [Indexed: 09/22/2024] Open
Abstract
Viral infections pose significant threats to human health, leading to a diverse spectrum of infectious diseases. The innate immune system serves as the primary barrier against viruses and bacteria in the early stages of infection. A rapid and forceful antiviral innate immune response is triggered by distinguishing between self-nucleic acids and viral nucleic acids. RNA-binding proteins (RBPs) are a diverse group of proteins which contain specific structural motifs or domains for binding RNA molecules. In the last decade, numerous of studies have outlined that RBPs influence viral replication via diverse mechanisms, directly recognizing viral nucleic acids and modulating the activity of pattern recognition receptors (PRRs). In this review, we summarize the functions of RBPs in regulation of host-virus interplay by controlling the activation of PRRs, such as RIG-I, MDA5, cGAS and TLR3. RBPs are instrumental in facilitating the identification of viral RNA or DNA, as well as viral structural proteins within the cellular cytoplasm and nucleus, functioning as co-receptor elements. On the other hand, RBPs are capable of orchestrating the activation of PRRs and facilitating the transmission of antiviral signals to downstream adaptor proteins by post-translational modifications or aggregation. Gaining a deeper comprehension of the interaction between the host and viruses is crucial for the development of novel therapeutics targeting viral infections.
Collapse
Affiliation(s)
- Jianguo Li
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
- Center for Immunology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, China
- Graduate School, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Jingge Yu
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
- Department of Blood Transfusion, Jingmen Central Hospital, Jingmen, China
| | - Ao Shen
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
- Graduate School, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Suwen Lai
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China
| | - Zhiping Liu
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China.
- Center for Immunology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, China.
| | - Tian-Sheng He
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, 341000, China.
- Center for Immunology, Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, Jiangxi, China.
| |
Collapse
|
12
|
Tants JN, Oberstrass L, Weigand JE, Schlundt A. Structure and RNA-binding of the helically extended Roquin CCCH-type zinc finger. Nucleic Acids Res 2024; 52:9838-9853. [PMID: 38953172 PMCID: PMC11381341 DOI: 10.1093/nar/gkae555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 06/07/2024] [Accepted: 06/17/2024] [Indexed: 07/03/2024] Open
Abstract
Zinc finger (ZnF) domains appear in a pool of structural contexts and despite their small size achieve varying target specificities, covering single-stranded and double-stranded DNA and RNA as well as proteins. Combined with other RNA-binding domains, ZnFs enhance affinity and specificity of RNA-binding proteins (RBPs). The ZnF-containing immunoregulatory RBP Roquin initiates mRNA decay, thereby controlling the adaptive immune system. Its unique ROQ domain shape-specifically recognizes stem-looped cis-elements in mRNA 3'-untranslated regions (UTR). The N-terminus of Roquin contains a RING domain for protein-protein interactions and a ZnF, which was suggested to play an essential role in RNA decay by Roquin. The ZnF domain boundaries, its RNA motif preference and its interplay with the ROQ domain have remained elusive, also driven by the lack of high-resolution data of the challenging protein. We provide the solution structure of the Roquin-1 ZnF and use an RBNS-NMR pipeline to show that the ZnF recognizes AU-rich RNAs. We systematically refine the contributions of adenines in a poly(U)-background to specific complex formation. With the simultaneous binding of ROQ and ZnF to a natural target transcript of Roquin, our study for the first time suggests how Roquin integrates RNA shape and sequence features through the ROQ-ZnF tandem.
Collapse
Affiliation(s)
- Jan-Niklas Tants
- Institute for Molecular Biosciences and Biomolecular Resonance Center (BMRZ), Goethe University Frankfurt, Max-von-Laue-Str. 7-9, 60438 Frankfurt, Germany
| | - Lasse Oberstrass
- University of Marburg, Department of Pharmacy, Institute of Pharmaceutical Chemistry, Marbacher Weg 6, 35037 Marburg, Germany
| | - Julia E Weigand
- University of Marburg, Department of Pharmacy, Institute of Pharmaceutical Chemistry, Marbacher Weg 6, 35037 Marburg, Germany
| | - Andreas Schlundt
- Institute for Molecular Biosciences and Biomolecular Resonance Center (BMRZ), Goethe University Frankfurt, Max-von-Laue-Str. 7-9, 60438 Frankfurt, Germany
- University of Greifswald, Institute of Biochemistry, Felix-Hausdorff-Str. 4, 17489 Greifswald, Germany
| |
Collapse
|
13
|
Wei J, Ning H, Ramos‐Espinosa O, Eickhoff CS, Hou R, Wang Q, Fu M, Liu EY, Fan D, Hoft DF, Liu J. Tristetraprolin mediates immune evasion of mycobacterial infection in macrophages. FASEB Bioadv 2024; 6:249-262. [PMID: 39114448 PMCID: PMC11301268 DOI: 10.1096/fba.2024-00022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/15/2024] [Accepted: 05/30/2024] [Indexed: 08/10/2024] Open
Abstract
Immune evasion of Mycobacterium tuberculosis (Mtb) facilitates intracellular bacterial growth. The mechanisms of immune evasion, however, are still not fully understood. In this study, we reveal that tristetraprolin (TTP), one of the best characterized RNA-binding proteins controlling the stability of targeted mRNAs, mediates innate immune evasion of mycobacteria. We found that TTP knockout mice displayed reduced bacterial burden in the early stage after Mtb aerosol challenge. Macrophages deficient in TTP also showed an inhibition in intracellular mycobacterial growth. Live mycobacteria induced TTP protein expression in macrophages, which was blocked by the mTOR inhibitor rapamycin. Rapamycin and AZD8055 specifically blocked 4EBP1 phosphorylation in infected macrophages and suppressed intracellular BCG growth. Rapamycin promoted TTP protein degradation through the ubiquitination pathway, whereas the proteasome inhibitor MG-132 blocked rapamycin function and thus stabilized TTP protein. TTP induction suppressed the expression of iNOS/TNF-α/IL-12/IL-23, and weakened protective immune responses in macrophages, whereas rapamycin enhanced the bactericidal effects through TTP inhibition. Moreover, blocking TTP binding increased the expression of TNF-α and iNOS and suppressed intracellular mycobacterial growth. Overall, our study reveals a novel role for RNA-binding protein TTP in Mtb immune evasion mechanisms and provides a potential target for host-directed therapy against tuberculosis (TB).
Collapse
Affiliation(s)
- Jiawei Wei
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal MedicineSaint Louis University School of Medicine, Saint Louis UniversitySt. LouisMissouriUSA
| | - Huan Ning
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal MedicineSaint Louis University School of Medicine, Saint Louis UniversitySt. LouisMissouriUSA
| | - Octavio Ramos‐Espinosa
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal MedicineSaint Louis University School of Medicine, Saint Louis UniversitySt. LouisMissouriUSA
| | - Christopher S. Eickhoff
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal MedicineSaint Louis University School of Medicine, Saint Louis UniversitySt. LouisMissouriUSA
| | - Rong Hou
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal MedicineSaint Louis University School of Medicine, Saint Louis UniversitySt. LouisMissouriUSA
| | - Qinghong Wang
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal MedicineSaint Louis University School of Medicine, Saint Louis UniversitySt. LouisMissouriUSA
| | - Mingui Fu
- Shock/Trauma Research Center, Department of Basic Medical Science, School of MedicineUniversity of Missouri‐Kansas CityKansas CityMissouriUSA
| | - Ethan Y. Liu
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal MedicineSaint Louis University School of Medicine, Saint Louis UniversitySt. LouisMissouriUSA
| | - Daping Fan
- Department of Cell Biology and AnatomyUniversity of South Carolina School of MedicineColumbiaSouth CarolinaUSA
| | - Daniel F. Hoft
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal MedicineSaint Louis University School of Medicine, Saint Louis UniversitySt. LouisMissouriUSA
| | - Jianguo Liu
- Division of Infectious Diseases, Allergy and Immunology, Department of Internal MedicineSaint Louis University School of Medicine, Saint Louis UniversitySt. LouisMissouriUSA
| |
Collapse
|
14
|
He SL, Li B, Zahurancik WJ, Arthur HC, Sidharthan V, Gopalan V, Wang L, Jang JC. Overexpression of stress granule protein TZF1 enhances salt stress tolerance by targeting ACA11 mRNA for degradation in Arabidopsis. FRONTIERS IN PLANT SCIENCE 2024; 15:1375478. [PMID: 38799098 PMCID: PMC11122021 DOI: 10.3389/fpls.2024.1375478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 04/03/2024] [Indexed: 05/29/2024]
Abstract
Tandem CCCH zinc finger (TZF) proteins play diverse roles in plant growth and stress response. Although as many as 11 TZF proteins have been identified in Arabidopsis, little is known about the mechanism by which TZF proteins select and regulate the target mRNAs. Here, we report that Arabidopsis TZF1 is a bona-fide stress granule protein. Ectopic expression of TZF1 (TZF1 OE), but not an mRNA binding-defective mutant (TZF1H186Y OE), enhances salt stress tolerance in Arabidopsis. RNA-seq analyses of NaCl-treated plants revealed that the down-regulated genes in TZF1 OE plants are enriched for functions in salt and oxidative stress responses. Because many of these down-regulated mRNAs contain AU- and/or U-rich elements (AREs and/or UREs) in their 3'-UTRs, we hypothesized that TZF1-ARE/URE interaction might contribute to the observed gene expression changes. Results from RNA immunoprecipitation-quantitative PCR analysis, gel-shift, and mRNA half-life assays indicate that TZF1 binds and triggers degradation of the autoinhibited Ca2+-ATPase 11 (ACA11) mRNA, which encodes a tonoplast-localized calcium pump that extrudes calcium and dampens signal transduction pathways necessary for salt stress tolerance. Furthermore, this salt stress-tolerance phenotype was recapitulated in aca11 null mutants. Collectively, our findings demonstrate that TZF1 binds and initiates degradation of specific mRNAs to enhance salt stress tolerance.
Collapse
Affiliation(s)
- Siou-Luan He
- Department of Horticulture and Crop Science, The Ohio State University, Columbus, OH, United States
- Center for Applied Plant Sciences, The Ohio State University, Columbus, OH, United States
- Center for RNA Biology, The Ohio State University, Columbus, OH, United States
| | - Bin Li
- Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, and University of Chinese Academy of Sciences, Beijing, China
- Academician Workstation of Agricultural High-Tech Industrial Area of the Yellow River Delta, National Center of Technology Innovation for Comprehensive Utilization of Saline-Alkali Land, Shandong, China
| | - Walter J. Zahurancik
- Center for RNA Biology, The Ohio State University, Columbus, OH, United States
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, United States
| | - Henry C. Arthur
- Center for RNA Biology, The Ohio State University, Columbus, OH, United States
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, United States
| | - Vaishnavi Sidharthan
- Center for RNA Biology, The Ohio State University, Columbus, OH, United States
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, United States
| | - Venkat Gopalan
- Center for RNA Biology, The Ohio State University, Columbus, OH, United States
- Department of Chemistry and Biochemistry, The Ohio State University, Columbus, OH, United States
| | - Lei Wang
- Key Laboratory of Plant Molecular Physiology, Institute of Botany, Chinese Academy of Sciences, and University of Chinese Academy of Sciences, Beijing, China
- Academician Workstation of Agricultural High-Tech Industrial Area of the Yellow River Delta, National Center of Technology Innovation for Comprehensive Utilization of Saline-Alkali Land, Shandong, China
| | - Jyan-Chyun Jang
- Department of Horticulture and Crop Science, The Ohio State University, Columbus, OH, United States
- Center for Applied Plant Sciences, The Ohio State University, Columbus, OH, United States
- Center for RNA Biology, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
15
|
Kawai T, Ikegawa M, Ori D, Akira S. Decoding Toll-like receptors: Recent insights and perspectives in innate immunity. Immunity 2024; 57:649-673. [PMID: 38599164 DOI: 10.1016/j.immuni.2024.03.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/18/2024] [Accepted: 03/05/2024] [Indexed: 04/12/2024]
Abstract
Toll-like receptors (TLRs) are an evolutionarily conserved family in the innate immune system and are the first line of host defense against microbial pathogens by recognizing pathogen-associated molecular patterns (PAMPs). TLRs, categorized into cell surface and endosomal subfamilies, recognize diverse PAMPs, and structural elucidation of TLRs and PAMP complexes has revealed their intricate mechanisms. TLRs activate common and specific signaling pathways to shape immune responses. Recent studies have shown the importance of post-transcriptional regulation in TLR-mediated inflammatory responses. Despite their protective functions, aberrant responses of TLRs contribute to inflammatory and autoimmune disorders. Understanding the delicate balance between TLR activation and regulatory mechanisms is crucial for deciphering their dual role in immune defense and disease pathogenesis. This review provides an overview of recent insights into the history of TLR discovery, elucidation of TLR ligands and signaling pathways, and their relevance to various diseases.
Collapse
Affiliation(s)
- Taro Kawai
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Nara 630-0192, Japan; Life Science Collaboration Center (LiSCo), Nara Institute of Science and Technology (NAIST), Nara 630-0192, Japan.
| | - Moe Ikegawa
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Nara 630-0192, Japan
| | - Daisuke Ori
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Nara 630-0192, Japan
| | - Shizuo Akira
- Center for Advanced Modalities and DSS (CAMaD), Osaka University, Osaka 565-0871, Japan; Laboratory of Host Defense, Immunology Frontier Research Center (IFReC), Osaka University, Osaka 565-0871, Japan; Department of Host Defense, Research Institute for Microbial Diseases (RIMD), Osaka University, Osaka 565-0871, Japan.
| |
Collapse
|
16
|
Tanaka-Yano M, Zong L, Park B, Yanai H, Tekin-Turhan F, Blackshear PJ, Beerman I. Tristetraprolin overexpression drives hematopoietic changes in young and middle-aged mice generating dominant mitigating effects on induced inflammation in murine models. GeroScience 2024; 46:1271-1284. [PMID: 37535204 PMCID: PMC10828162 DOI: 10.1007/s11357-023-00879-2] [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: 05/12/2023] [Accepted: 07/17/2023] [Indexed: 08/04/2023] Open
Abstract
Tristetraprolin (TTP), encoded by Zfp36 in mice, is one of the best-characterized tandem zinc-finger mRNA binding proteins involved in mRNA deadenylation and decay. TTPΔARE mice lack an AU-rich motif in the 3'-untranslated regions of TTP mRNA, leading to increased TTP mRNA stability and more TTP protein, resulting in elevated mRNA decay rates of TTP targets. We examined the effect of TTP overexpression on the hematopoietic system in both young and middle-aged mice using TTPΔARE mice and found alterations in blood cell frequencies, with loss of platelets and B220 cells and gains of eosinophils and T cells. TTPΔARE mice also have skewed primitive populations in the bone marrow, with increases in myeloid-biased hematopoietic stem cells (HSCs) but decreases in granulocyte/macrophage-biased multipotent progenitors (MPP3) in both young and middle-aged mice. Changes in the primitive cells' frequencies were associated with transcriptional alterations in the TTP overexpression cells specific to age as well as cell type. Regardless of age, there was a consistent elevation of transcripts regulated by TNFα and TGFβ signaling pathways in both the stem and multipotent progenitor populations. HSCs with TTP overexpression had decreased reconstitution potential in murine transplants but generated hematopoietic environments that mitigated the inflammatory response to the collagen antibody-induced arthritis (CAIA) challenge, which models rheumatoid arthritis and other autoimmune disorders. This dampening of the inflammatory response was even present when there was only a small frequency of TTP overexpressing cells present in the middle-aged mice. We provide an analysis of the early hematopoietic compartments with elevated TTP expression in both young and middle-aged mice which inhibits the reconstitution potential of the HSCs but generates a hematopoietic system that provides dominant repression of induced inflammation.
Collapse
Affiliation(s)
- Mayuri Tanaka-Yano
- Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD, 21224, USA
| | - Le Zong
- Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD, 21224, USA
| | - Bongsoo Park
- Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD, 21224, USA
| | - Hagai Yanai
- Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD, 21224, USA
| | - Ferda Tekin-Turhan
- Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD, 21224, USA
| | - Perry J Blackshear
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, 27709, USA
| | - Isabel Beerman
- Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute On Aging, National Institutes of Health, 251 Bayview Boulevard, Baltimore, MD, 21224, USA.
| |
Collapse
|
17
|
Zhang L, Kwack KH, Thiyagarajan R, Mullaney KK, Lamb NA, Bard JE, Sohn J, Seldeen KL, Arao Y, Blackshear PJ, Abrams SI, Troen BR, Kirkwood KL. Tristetraprolin regulates the skeletal phenotype and osteoclastogenic potential through monocytic myeloid-derived suppressor cells. FASEB J 2024; 38:e23338. [PMID: 38038723 PMCID: PMC11128769 DOI: 10.1096/fj.202301703r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/07/2023] [Accepted: 11/14/2023] [Indexed: 12/02/2023]
Abstract
Tristetraprolin (TTP; also known as NUP475, GOS24, or TIS11), encoded by Zfp36, is an RNA-binding protein that regulates target gene expression by promoting mRNA decay and preventing translation. Although previous studies have indicated that TTP deficiency is associated with systemic inflammation and a catabolic-like skeletal phenotype, the mechanistic underpinnings remain unclear. Here, using both TTP-deficient (TTPKO) and myeloid-specific TTPKO (cTTPKO) mice, we reveal that global absence or loss of TTP in the myeloid compartment results in a reduced bone microarchitecture, whereas gain-of-function TTP knock-in (TTPKI) mice exhibit no significant loss of bone microarchitecture. Flow cytometry analysis revealed a significant immunosuppressive immune cell phenotype with increased monocytic myeloid-derived suppressor cells (M-MDSCs) in TTPKO and cTTPKO mice, whereas no significant changes were observed in TTPKI mice. Single-cell transcriptomic analyses of bone marrow myeloid progenitor cell populations indicated a dramatic increase in early MDSC marker genes for both cTTPKO and TTPKO bone marrow populations. Consistent with these phenotypic and transcriptomic data, in vitro osteoclastogenesis analysis of bone marrow M-MDSCs from cTTPKO and TTPKO displayed enhanced osteoclast differentiation and functional capacity. Focused transcriptomic analyses of differentiated M-MDSCs showed increased osteoclast-specific transcription factors and cell fusion gene expression. Finally, functional data showed that M-MDSCs from TTP loss-of-function mice were capable of osteoclastogenesis and bone resorption in a context-dependent manner. Collectively, these findings indicate that TTP plays a central role in regulating osteoclastogenesis through multiple mechanisms, including induction of M-MDSCs that appear to regulate skeletal phenotype.
Collapse
Affiliation(s)
| | - Kyu Hwan Kwack
- Department of Oral Microbiology, College of Dentistry, Kyung Hee University, Seoul, Republic of Korea
| | - Ramkumar Thiyagarajan
- Departments of Medicine, University at Buffalo, Buffalo, NY, USA
- Division of Geriatrics and Palliative Medicine, University at Buffalo, Buffalo, NY, USA
- Research Service, Veterans Affairs Western New York Healthcare Service, Buffalo, NY, USA
| | - Kylie K. Mullaney
- Departments of Oral Biology, University at Buffalo, Buffalo, NY, USA
| | - Natalie A. Lamb
- Departments of Biochemistry, University at Buffalo, Buffalo, NY, USA
- Genomics and Bioinformatics Core, New York State Center of Excellence for Bioinformatics and Life Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Jonathan E. Bard
- Departments of Biochemistry, University at Buffalo, Buffalo, NY, USA
- Genomics and Bioinformatics Core, New York State Center of Excellence for Bioinformatics and Life Sciences, University at Buffalo, The State University of New York, Buffalo, NY, USA
| | - Jiho Sohn
- Departments of Oral Biology, University at Buffalo, Buffalo, NY, USA
- Departments of Medicine, University at Buffalo, Buffalo, NY, USA
| | - Kenneth L. Seldeen
- Departments of Medicine, University at Buffalo, Buffalo, NY, USA
- Division of Geriatrics and Palliative Medicine, University at Buffalo, Buffalo, NY, USA
- Research Service, Veterans Affairs Western New York Healthcare Service, Buffalo, NY, USA
| | - Yukitomo Arao
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
| | - Perry J. Blackshear
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, USA
- Departments of Biochemistry & Medicine, Duke University Medical Center, Durham, NC, USA
| | - Scott I. Abrams
- Departments of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Bruce R. Troen
- Departments of Medicine, University at Buffalo, Buffalo, NY, USA
- Departments of Biochemistry, University at Buffalo, Buffalo, NY, USA
- Research Service, Veterans Affairs Western New York Healthcare Service, Buffalo, NY, USA
| | - Keith L. Kirkwood
- Departments of Oral Biology, University at Buffalo, Buffalo, NY, USA
- Head & Neck/Plastic & Reconstructive Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| |
Collapse
|
18
|
Snyder BL, Huang R, Burkholder AB, Donahue DR, Mahler BW, Bortner CD, Lai WS, Blackshear PJ. Synergistic roles of tristetraprolin family members in myeloid cells in the control of inflammation. Life Sci Alliance 2024; 7:e202302222. [PMID: 37903626 PMCID: PMC10616675 DOI: 10.26508/lsa.202302222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 10/12/2023] [Accepted: 10/13/2023] [Indexed: 11/01/2023] Open
Abstract
Members of the tristetraprolin (TTP) family of RNA-binding proteins can bind to and promote the decay of specific transcripts containing AU-rich motifs. ZFP36 (TTP) is best known for regulating pro-inflammatory cytokine expression in myeloid cells; however, its mammalian paralogues ZFP36L1 and ZFP36L2 have not been viewed as important in controlling inflammation. We knocked out these genes in myeloid cells in mice, singly and together. Single-gene myeloid-specific knockouts resulted in almost no spontaneous phenotypes. In contrast, mice with myeloid cell deficiency of all three genes developed severe inflammation, with a median survival of 8 wk. Macrophages from these mice expressed many more stabilized transcripts than cells from myeloid-specific TTP knockout mice; many of these encoded pro-inflammatory cytokines and chemokines. The failure of weight gain, arthritis, and early death could be prevented completely by two normal alleles of any of the three paralogues, and even one normal allele of Zfp36 or Zfp36l2 was enough to prevent the inflammatory phenotype. Our findings emphasize the importance of all three family members, acting in concert, in myeloid cell function.
Collapse
Affiliation(s)
- Brittany L Snyder
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, Durham, NC, USA
- Department of Biochemistry, Duke University Medical Center, Durham, NC, USA
| | - Rui Huang
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, Durham, NC, USA
- Department of Biochemistry, Duke University Medical Center, Durham, NC, USA
| | - Adam B Burkholder
- Bioinformatics Support Group, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, Durham, NC, USA
| | - Danielle R Donahue
- NIH Mouse Imaging Facility, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Beth W Mahler
- Experimental Pathology Laboratories, Inc., Research Triangle Park, Durham, NC, USA
| | - Carl D Bortner
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, Durham, NC, USA
| | - Wi S Lai
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, Durham, NC, USA
| | - Perry J Blackshear
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, NIH, Research Triangle Park, Durham, NC, USA
- Department of Biochemistry, Duke University Medical Center, Durham, NC, USA
- Department of Medicine, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
19
|
Al-Yahya S, Al-Saif M, Al-Ghamdi M, Moghrabi W, Khabar KS, Al-Souhibani N. Post-transcriptional regulation of BIRC5/survivin expression and induction of apoptosis in breast cancer cells by tristetraprolin. RNA Biol 2024; 21:1-15. [PMID: 38111129 PMCID: PMC10761079 DOI: 10.1080/15476286.2023.2286101] [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] [Accepted: 11/14/2023] [Indexed: 12/20/2023] Open
Abstract
Inhibition of apoptosis is one of the hallmarks of cancer and is a target of various therapeutic interventions. BIRC5 is an inhibitor of apoptosis that is aberrantly expressed in cancer leading to sustained growth of tumours. Post-transcriptional control mechanisms involving RNA-binding proteins and AU-rich elements (AREs) are fundamental to many cellular processes and changes in the expression or function of these proteins can promote an aberrant and pathological phenotype. BIRC5 mRNA has an ARE in its 3' UTR making it a candidate for regulation by the RNA binding proteins tristetraprolin (TTP) and HuR (ELAVL1). In this study, we investigated the binding of TTP and HuR by RNA-immunoprecipitation assays and found that these proteins were associated with BIRC5 mRNA to varying extents. Consequently, BIRC5 expression decreased when TTP was overexpressed and apoptosis was induced. In the absence of TTP, BIRC5 mRNA was stabilized, protein expression increased and the number of apoptotic cells declined. As an ARE-mRNA stabilizing protein, recombinant HuR led to upregulation of BIRC5 expression, whereas HuR silencing was concomitant with downregulation of BIRC5 mRNA and protein and increased cell death. Survival analyses demonstrated that increased TTP and low BIRC5 expression predicted an overall better prognosis compared to dysregulated TTP and high BIRC5. Thus, the results present a novel target of ARE-mediated post-transcriptional regulation.
Collapse
Affiliation(s)
- Suhad Al-Yahya
- Molecular Biomedicine Department, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Maher Al-Saif
- Molecular Biomedicine Department, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Maha Al-Ghamdi
- Biomedical Physics Department, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Walid Moghrabi
- Molecular Biomedicine Department, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Khalid S.A. Khabar
- Molecular Biomedicine Department, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Norah Al-Souhibani
- Molecular Biomedicine Department, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| |
Collapse
|
20
|
Nelson EV, Ross SJ, Olejnik J, Hume AJ, Deeney DJ, King E, Grimins AO, Lyons SM, Cifuentes D, Mühlberger E. The 3' Untranslated Regions of Ebola Virus mRNAs Contain AU-Rich Elements Involved in Posttranscriptional Stabilization and Decay. J Infect Dis 2023; 228:S488-S497. [PMID: 37551415 PMCID: PMC10651315 DOI: 10.1093/infdis/jiad312] [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: 04/18/2023] [Revised: 07/29/2023] [Accepted: 08/01/2023] [Indexed: 08/09/2023] Open
Abstract
The 3' untranslated regions (UTRs) of Ebola virus (EBOV) mRNAs are enriched in their AU content and therefore represent potential targets for RNA binding proteins targeting AU-rich elements (ARE-BPs). ARE-BPs are known to fine-tune RNA turnover and translational activity. We identified putative AREs within EBOV mRNA 3' UTRs and assessed whether they might modulate mRNA stability. Using mammalian and zebrafish embryo reporter assays, we show a conserved, ARE-BP-mediated stabilizing effect and increased reporter activity with the tested EBOV 3' UTRs. When coexpressed with the prototypic ARE-BP tristetraprolin (TTP, ZFP36) that mainly destabilizes its target mRNAs, the EBOV nucleoprotein (NP) 3' UTR resulted in decreased reporter gene activity. Coexpression of NP with TTP led to reduced NP protein expression and diminished EBOV minigenome activity. In conclusion, the enrichment of AU residues in EBOV 3' UTRs makes them possible targets for cellular ARE-BPs, leading to modulation of RNA stability and translational activity.
Collapse
Affiliation(s)
- Emily V Nelson
- Department of Virology, Immunology, and Microbiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts, USA
| | - Stephen J Ross
- Department of Virology, Immunology, and Microbiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, USA
- Department of Biochemistry and Cell Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts, USA
| | - Judith Olejnik
- Department of Virology, Immunology, and Microbiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, USA
| | - Adam J Hume
- Department of Virology, Immunology, and Microbiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, USA
| | - Dylan J Deeney
- Department of Virology, Immunology, and Microbiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, USA
| | - Emily King
- Department of Biochemistry and Cell Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts, USA
| | - Autumn O Grimins
- Department of Biochemistry and Cell Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts, USA
| | - Shawn M Lyons
- Department of Biochemistry and Cell Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts, USA
| | - Daniel Cifuentes
- Department of Virology, Immunology, and Microbiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts, USA
- Department of Biochemistry and Cell Biology, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts, USA
| | - Elke Mühlberger
- Department of Virology, Immunology, and Microbiology, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, USA
| |
Collapse
|
21
|
Šimková K, Naraine R, Vintr J, Soukup V, Šindelka R. RNA localization during early development of the axolotl. Front Cell Dev Biol 2023; 11:1260795. [PMID: 37928901 PMCID: PMC10620976 DOI: 10.3389/fcell.2023.1260795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/26/2023] [Indexed: 11/07/2023] Open
Abstract
The asymmetric localization of biomolecules is critical for body plan development. One of the most popular model organisms for early embryogenesis studies is Xenopus laevis but there is a lack of information in other animal species. Here, we compared the early development of two amphibian species-the frog X. laevis and the axolotl Ambystoma mexicanum. This study aimed to identify asymmetrically localized RNAs along the animal-vegetal axis during the early development of A. mexicanum. For that purpose, we performed spatial transcriptome-wide analysis at low resolution, which revealed dynamic changes along the animal-vegetal axis classified into the following categories: profile alteration, de novo synthesis and degradation. Surprisingly, our results showed that many of the vegetally localized genes, which are important for germ cell development, are degraded during early development. Furthermore, we assessed the motif presence in UTRs of degraded mRNAs and revealed the enrichment of several motifs in RNAs of germ cell markers. Our results suggest novel reorganization of the transcriptome during embryogenesis of A. mexicanum to converge to the similar developmental pattern as the X. laevis.
Collapse
Affiliation(s)
- Kateřina Šimková
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czechia
| | - Ravindra Naraine
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czechia
| | - Jan Vintr
- Department of Zoology, Faculty of Science, Charles University, Prague, Czechia
| | - Vladimír Soukup
- Department of Zoology, Faculty of Science, Charles University, Prague, Czechia
| | - Radek Šindelka
- Laboratory of Gene Expression, Institute of Biotechnology of the Czech Academy of Sciences, Vestec, Czechia
| |
Collapse
|
22
|
Jiang X, Xiao Y, Hou W, Yu J, He TS, Xu LG. The RNA-binding protein ZFP36 strengthens innate antiviral signaling by targeting RIG-I for K63-linked ubiquitination. J Cell Physiol 2023; 238:2348-2360. [PMID: 37565597 DOI: 10.1002/jcp.31088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 06/28/2023] [Accepted: 07/14/2023] [Indexed: 08/12/2023]
Abstract
Innate immunity is the first line of defense against infections, which functions as a significant role in resisting pathogen invasion. Rapid immune response is initiated by pattern recognition receptors (PRRs) quickly distinguishing "self" and "non-self." Upon evolutionarily conserved pathogen-associated molecular pattern (PAMP) is recognized by PRRs, innate immune response against infection is triggered via an orchestration of molecular interaction, cytokines cascades, and immune cells. RIG-I plays a critical role in type I interferon (IFN-I) production by direct recognition of cytoplasmic double-stranded viral RNA. However, the activation mechanism of RIG-I is incompletely understood. In this study, we reported RNA-binding protein ZFP36 as a positive regulator of RIG-I-mediated IFN-I production. ZFP36 is a member of Zinc finger proteins (ZFPs) characterized by the zinc finger (ZnF) motif, which broadly involved gene transcription and signal transduction. However, its role in regulating antiviral innate immune signaling is still unclear. We found that ZFP36 associates with RIG-I and potentiates the FN-β production induced by SeV. Mechanistically, ZFP36 promotes K63-linked polyubiquitination of RIG-I, mostly at K154/K164/K172, thereby facilitating the activation of RIG-I during infection. While the mutant ZFP36 (C118S/C162S) failed to increase polyubiquitination of RIG-I and SeV induced FN-β. Our findings collectively demonstrated that ZFP36 acts as a positive regulator in antiviral innate immunity by targeting RIG-I for K63-linked ubiquitination, thus improving our understanding of the activation mechanism of RIG-I.
Collapse
Affiliation(s)
- Xue Jiang
- College of Life Science, Jiangxi Normal University, Nanchang, Jiangxi, China
| | - Yanping Xiao
- College of Life Science, Jiangxi Normal University, Nanchang, Jiangxi, China
| | - Wen Hou
- College of Life Science, Jiangxi Normal University, Nanchang, Jiangxi, China
| | - Jingge Yu
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Tian-Sheng He
- College of Life Science, Jiangxi Normal University, Nanchang, Jiangxi, China
- School of Basic Medicine, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Liang-Guo Xu
- College of Life Science, Jiangxi Normal University, Nanchang, Jiangxi, China
| |
Collapse
|
23
|
Kolb KL, Mira ALS, Auer ED, Bucco ID, de Lima e Silva CE, dos Santos PI, Hoch VBB, Oliveira LC, Hauser AB, Hundt JE, Shuldiner AR, Lopes FL, Boysen TJ, Franke A, Pinto LFR, Soares-Lima SC, Kretzschmar GC, Boldt ABW. Glucocorticoid Receptor Gene ( NR3C1) Polymorphisms and Metabolic Syndrome: Insights from the Mennonite Population. Genes (Basel) 2023; 14:1805. [PMID: 37761945 PMCID: PMC10530687 DOI: 10.3390/genes14091805] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 09/06/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
The regulation of the hypothalamic-pituitary-adrenal (HPA) axis is associated with polymorphisms and the methylation degree of the glucocorticoid receptor gene (NR3C1) and is potentially involved in the development of metabolic syndrome (MetS). In order to evaluate the association between MetS with the polymorphisms, methylation, and gene expression of the NR3C1 in the genetically isolated Brazilian Mennonite population, we genotyped 20 NR3C1 polymorphisms in 74 affected (MetS) and 138 unaffected individuals without affected first-degree relatives (Co), using exome sequencing, as well as five variants from non-exonic regions, in 70 MetS and 166 Co, using mass spectrometry. The methylation levels of 11 1F CpG sites were quantified using pyrosequencing (66 MetS and 141 Co), and the NR3C1 expression was evaluated via RT-qPCR (14 MetS and 25 Co). Age, physical activity, and family environment during childhood were associated with MetS. Susceptibility to MetS, independent of these factors, was associated with homozygosity for rs10482605*C (OR = 4.74, pcorr = 0.024) and the haplotype containing TTCGTTGATT (rs3806855*T_ rs3806854*T_rs10482605*C_rs10482614*G_rs6188*T_rs258813*T_rs33944801*G_rs34176759*A_rs17209258*T_rs6196*T, OR = 4.74, pcorr = 0.048), as well as for the CCT haplotype (rs41423247*C_ rs6877893*C_rs258763*T), OR = 6.02, pcorr = 0.030), but not to the differences in methylation or gene expression. Thus, NR3C1 polymorphisms seem to modulate the susceptibility to MetS in Mennonites, independently of lifestyle and early childhood events, and their role seems to be unrelated to DNA methylation and gene expression.
Collapse
Affiliation(s)
- Kathleen Liedtke Kolb
- Laboratory of Human Molecular Genetics, Department of Genetics, Federal University of Paraná (UFPR), Centro Politécnico, Jardim das Américas, Curitiba 81531-990, PR, Brazil; (K.L.K.); (A.L.S.M.); (E.D.A.); (I.D.B.); (C.E.d.L.e.S.); (P.I.d.S.); (V.B.-B.H.); (L.C.O.); (G.C.K.)
- Postgraduate Program in Genetics, Department of Genetics, Federal University of Paraná (UFPR), Centro Politécnico, Jardim das Américas, Curitiba 81531-990, PR, Brazil
| | - Ana Luiza Sprotte Mira
- Laboratory of Human Molecular Genetics, Department of Genetics, Federal University of Paraná (UFPR), Centro Politécnico, Jardim das Américas, Curitiba 81531-990, PR, Brazil; (K.L.K.); (A.L.S.M.); (E.D.A.); (I.D.B.); (C.E.d.L.e.S.); (P.I.d.S.); (V.B.-B.H.); (L.C.O.); (G.C.K.)
- Postgraduate Program in Genetics, Department of Genetics, Federal University of Paraná (UFPR), Centro Politécnico, Jardim das Américas, Curitiba 81531-990, PR, Brazil
| | - Eduardo Delabio Auer
- Laboratory of Human Molecular Genetics, Department of Genetics, Federal University of Paraná (UFPR), Centro Politécnico, Jardim das Américas, Curitiba 81531-990, PR, Brazil; (K.L.K.); (A.L.S.M.); (E.D.A.); (I.D.B.); (C.E.d.L.e.S.); (P.I.d.S.); (V.B.-B.H.); (L.C.O.); (G.C.K.)
- Postgraduate Program in Genetics, Department of Genetics, Federal University of Paraná (UFPR), Centro Politécnico, Jardim das Américas, Curitiba 81531-990, PR, Brazil
| | - Isabela Dall’Oglio Bucco
- Laboratory of Human Molecular Genetics, Department of Genetics, Federal University of Paraná (UFPR), Centro Politécnico, Jardim das Américas, Curitiba 81531-990, PR, Brazil; (K.L.K.); (A.L.S.M.); (E.D.A.); (I.D.B.); (C.E.d.L.e.S.); (P.I.d.S.); (V.B.-B.H.); (L.C.O.); (G.C.K.)
| | - Carla Eduarda de Lima e Silva
- Laboratory of Human Molecular Genetics, Department of Genetics, Federal University of Paraná (UFPR), Centro Politécnico, Jardim das Américas, Curitiba 81531-990, PR, Brazil; (K.L.K.); (A.L.S.M.); (E.D.A.); (I.D.B.); (C.E.d.L.e.S.); (P.I.d.S.); (V.B.-B.H.); (L.C.O.); (G.C.K.)
| | - Priscila Ianzen dos Santos
- Laboratory of Human Molecular Genetics, Department of Genetics, Federal University of Paraná (UFPR), Centro Politécnico, Jardim das Américas, Curitiba 81531-990, PR, Brazil; (K.L.K.); (A.L.S.M.); (E.D.A.); (I.D.B.); (C.E.d.L.e.S.); (P.I.d.S.); (V.B.-B.H.); (L.C.O.); (G.C.K.)
- Postgraduate Program in Internal Medicine, Medical Clinic Department, UFPR, Rua General Carneiro, 181, 11th Floor, Alto da Glória, Curitiba 80210-170, PR, Brazil
| | - Valéria Bumiller-Bini Hoch
- Laboratory of Human Molecular Genetics, Department of Genetics, Federal University of Paraná (UFPR), Centro Politécnico, Jardim das Américas, Curitiba 81531-990, PR, Brazil; (K.L.K.); (A.L.S.M.); (E.D.A.); (I.D.B.); (C.E.d.L.e.S.); (P.I.d.S.); (V.B.-B.H.); (L.C.O.); (G.C.K.)
| | - Luana Caroline Oliveira
- Laboratory of Human Molecular Genetics, Department of Genetics, Federal University of Paraná (UFPR), Centro Politécnico, Jardim das Américas, Curitiba 81531-990, PR, Brazil; (K.L.K.); (A.L.S.M.); (E.D.A.); (I.D.B.); (C.E.d.L.e.S.); (P.I.d.S.); (V.B.-B.H.); (L.C.O.); (G.C.K.)
| | - Aline Borsato Hauser
- Laboratory School of Clinical Analysis, Department of Pharmacy, Federal University of Paraná (UFPR), Av. Pref. Lothário Meissner, 632, Jardim Botânico, Curitiba 80210-170, PR, Brazil;
| | - Jennifer Elisabeth Hundt
- Lübeck Institute of Experimental Dermatology, University of Lübeck, Ratzeburger Allee, 160, Haus 32, 23562 Lübeck, Germany;
| | - Alan R. Shuldiner
- Regeneron Genetics Center, 777 Old Saw Mill River Road, Tarrytown, NY 10591, USA;
| | - Fabiana Leão Lopes
- Human Genetics Branch, National Institute of Mental Health, 35 Convent Drive, Bethesda, MD 20892, USA;
- Institute of Psychiatry, Federal University Rio de Janeiro, Av. Venceslau Brás, 71, Rio de Janeiro 22290-140, RJ, Brazil
| | - Teide-Jens Boysen
- Institute of Clinical Molecular Biology (IKMB), Christian-Albrechts-University of Kiel, 24105 Kiel, Germany; (T.-J.B.); (A.F.)
| | - Andre Franke
- Institute of Clinical Molecular Biology (IKMB), Christian-Albrechts-University of Kiel, 24105 Kiel, Germany; (T.-J.B.); (A.F.)
| | - Luis Felipe Ribeiro Pinto
- Brazilian National Cancer Institute, Rua André Cavalcanti, 37, Rio de Janeiro 20231-050, RJ, Brazil; (L.F.R.P.); (S.C.S.-L.)
| | - Sheila Coelho Soares-Lima
- Brazilian National Cancer Institute, Rua André Cavalcanti, 37, Rio de Janeiro 20231-050, RJ, Brazil; (L.F.R.P.); (S.C.S.-L.)
| | - Gabriela Canalli Kretzschmar
- Laboratory of Human Molecular Genetics, Department of Genetics, Federal University of Paraná (UFPR), Centro Politécnico, Jardim das Américas, Curitiba 81531-990, PR, Brazil; (K.L.K.); (A.L.S.M.); (E.D.A.); (I.D.B.); (C.E.d.L.e.S.); (P.I.d.S.); (V.B.-B.H.); (L.C.O.); (G.C.K.)
- Postgraduate Program in Genetics, Department of Genetics, Federal University of Paraná (UFPR), Centro Politécnico, Jardim das Américas, Curitiba 81531-990, PR, Brazil
- Faculdades Pequeno Príncipe, Av. Iguaçu, 333, Curitiba 80230-020, PR, Brazil
- Instituto de Pesquisa Pelé Pequeno Príncipe, Av. Silva Jardim, 1632, Curitiba 80250-060, PR, Brazil
| | - Angelica Beate Winter Boldt
- Laboratory of Human Molecular Genetics, Department of Genetics, Federal University of Paraná (UFPR), Centro Politécnico, Jardim das Américas, Curitiba 81531-990, PR, Brazil; (K.L.K.); (A.L.S.M.); (E.D.A.); (I.D.B.); (C.E.d.L.e.S.); (P.I.d.S.); (V.B.-B.H.); (L.C.O.); (G.C.K.)
- Postgraduate Program in Genetics, Department of Genetics, Federal University of Paraná (UFPR), Centro Politécnico, Jardim das Américas, Curitiba 81531-990, PR, Brazil
| |
Collapse
|
24
|
Huffman JE, Nicolas J, Hahn J, Heath AS, Raffield LM, Yanek LR, Brody JA, Thibord F, Almasy L, Bartz TM, Bielak LF, Bowler RP, Carrasquilla GD, Chasman DI, Chen MH, Emmert DB, Ghanbari M, Haessle J, Hottenga JJ, Kleber ME, Le NQ, Lee J, Lewis JP, Li-Gao R, Luan J, Malmberg A, Mangino M, Marioni RE, Martinez-Perez A, Pankratz N, Polasek O, Richmond A, Rodriguez BA, Rotter JI, Steri M, Suchon P, Trompet S, Weiss S, Zare M, Auer P, Cho MH, Christofidou P, Davies G, de Geus E, Deleuze JF, Delgado GE, Ekunwe L, Faraday N, Gögele M, Greinacher A, He G, Howard T, Joshi PK, Kilpeläinen TO, Lahti J, Linneberg A, Naitza S, Noordam R, Paüls-Vergés F, Rich SS, Rosendaal FR, Rudan I, Ryan KA, Souto JC, van Rooij FJ, Wang H, Zhao W, Becker LC, Beswick A, Brown MR, Cade BE, Campbell H, Cho K, Crapo JD, Curran JE, de Maat MP, Doyle M, Elliott P, Floyd JS, Fuchsberger C, Grarup N, Guo X, Harris SE, Hou L, Kolcic I, Kooperberg C, Menni C, Nauck M, O'Connell JR, Orrù V, Psaty BM, Räikkönen K, Smith JA, Soria JM, Stott DJ, van Hylckama Vlieg A, Watkins H, Willemsen G, Wilson P, Ben-Shlomo Y, et alHuffman JE, Nicolas J, Hahn J, Heath AS, Raffield LM, Yanek LR, Brody JA, Thibord F, Almasy L, Bartz TM, Bielak LF, Bowler RP, Carrasquilla GD, Chasman DI, Chen MH, Emmert DB, Ghanbari M, Haessle J, Hottenga JJ, Kleber ME, Le NQ, Lee J, Lewis JP, Li-Gao R, Luan J, Malmberg A, Mangino M, Marioni RE, Martinez-Perez A, Pankratz N, Polasek O, Richmond A, Rodriguez BA, Rotter JI, Steri M, Suchon P, Trompet S, Weiss S, Zare M, Auer P, Cho MH, Christofidou P, Davies G, de Geus E, Deleuze JF, Delgado GE, Ekunwe L, Faraday N, Gögele M, Greinacher A, He G, Howard T, Joshi PK, Kilpeläinen TO, Lahti J, Linneberg A, Naitza S, Noordam R, Paüls-Vergés F, Rich SS, Rosendaal FR, Rudan I, Ryan KA, Souto JC, van Rooij FJ, Wang H, Zhao W, Becker LC, Beswick A, Brown MR, Cade BE, Campbell H, Cho K, Crapo JD, Curran JE, de Maat MP, Doyle M, Elliott P, Floyd JS, Fuchsberger C, Grarup N, Guo X, Harris SE, Hou L, Kolcic I, Kooperberg C, Menni C, Nauck M, O'Connell JR, Orrù V, Psaty BM, Räikkönen K, Smith JA, Soria JM, Stott DJ, van Hylckama Vlieg A, Watkins H, Willemsen G, Wilson P, Ben-Shlomo Y, Blangero J, Boomsma D, Cox SR, Dehghan A, Eriksson JG, Fiorillo E, Fornage M, Hansen T, Hayward C, Ikram MA, Jukema JW, Kardia SL, Lange LA, März W, Mathias RA, Mitchell BD, Mook-Kanamori DO, Morange PE, Pedersen O, Pramstaller PP, Redline S, Reiner A, Ridker PM, Silverman EK, Spector TD, Völker U, Wareham N, Wilson JF, Yao J, Trégouët DA, Johnson AD, Wolberg AS, de Vries PS, Sabater-Lleal M, Morrison AC, Smith NL. Whole genome analysis of plasma fibrinogen reveals population-differentiated genetic regulators with putative liver roles. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.07.23291095. [PMID: 37398003 PMCID: PMC10312878 DOI: 10.1101/2023.06.07.23291095] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Genetic studies have identified numerous regions associated with plasma fibrinogen levels in Europeans, yet missing heritability and limited inclusion of non-Europeans necessitates further studies with improved power and sensitivity. Compared with array-based genotyping, whole genome sequencing (WGS) data provides better coverage of the genome and better representation of non-European variants. To better understand the genetic landscape regulating plasma fibrinogen levels, we meta-analyzed WGS data from the NHLBI's Trans-Omics for Precision Medicine (TOPMed) program (n=32,572), with array-based genotype data from the Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) Consortium (n=131,340) imputed to the TOPMed or Haplotype Reference Consortium panel. We identified 18 loci that have not been identified in prior genetic studies of fibrinogen. Of these, four are driven by common variants of small effect with reported MAF at least 10% higher in African populations. Three ( SERPINA1, ZFP36L2 , and TLR10) signals contain predicted deleterious missense variants. Two loci, SOCS3 and HPN , each harbor two conditionally distinct, non-coding variants. The gene region encoding the protein chain subunits ( FGG;FGB;FGA ), contains 7 distinct signals, including one novel signal driven by rs28577061, a variant common (MAF=0.180) in African reference panels but extremely rare (MAF=0.008) in Europeans. Through phenome-wide association studies in the VA Million Veteran Program, we found associations between fibrinogen polygenic risk scores and thrombotic and inflammatory disease phenotypes, including an association with gout. Our findings demonstrate the utility of WGS to augment genetic discovery in diverse populations and offer new insights for putative mechanisms of fibrinogen regulation. Key Points Largest and most diverse genetic study of plasma fibrinogen identifies 54 regions (18 novel), housing 69 conditionally distinct variants (20 novel).Sufficient power achieved to identify signal driven by African population variant.Links to (1) liver enzyme, blood cell and lipid genetic signals, (2) liver regulatory elements, and (3) thrombotic and inflammatory disease.
Collapse
|
25
|
Xiang M, Liu L, Wu T, Wei B, Liu H. RNA-binding proteins in degenerative joint diseases: A systematic review. Ageing Res Rev 2023; 86:101870. [PMID: 36746279 DOI: 10.1016/j.arr.2023.101870] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 01/12/2023] [Accepted: 01/27/2023] [Indexed: 02/07/2023]
Abstract
RNA-binding proteins (RBPs), which are conserved proteins comprising multiple intermediate sequences, can interact with proteins, messenger RNA (mRNA) of coding genes, and non-coding RNAs to perform different biological functions, such as the regulation of mRNA stability, selective polyadenylation, and the management of non-coding microRNA (miRNA) synthesis to affect downstream targets. This article will highlight the functions of RBPs, in degenerative joint diseases (intervertebral disc degeneration [IVDD] and osteoarthritis [OA]). It will reviews the latest advancements on the regulatory mechanism of RBPs in degenerative joint diseases, in order to understand the pathophysiology, early diagnosis and treatment of OA and IVDD from a new perspective.
Collapse
Affiliation(s)
- Min Xiang
- Department of Orthopedics, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Ling Liu
- Department of Pediatrics, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Tingrui Wu
- Department of Orthopedics, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Bo Wei
- Department of Orthopedics, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China.
| | - Huan Liu
- Department of Orthopedics, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
26
|
Park J, Rah SY, An HS, Lee JY, Roh GS, Ryter SW, Park JW, Yang CH, Surh YJ, Kim UH, Chung HT, Joe Y. Metformin-induced TTP mediates communication between Kupffer cells and hepatocytes to alleviate hepatic steatosis by regulating lipophagy and necroptosis. Metabolism 2023; 141:155516. [PMID: 36773805 DOI: 10.1016/j.metabol.2023.155516] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/24/2023] [Accepted: 02/05/2023] [Indexed: 02/11/2023]
Abstract
OBJECTIVE Emerging evidence suggests that crosstalk between Kupffer cells (KCs) and hepatocytes protects against non-alcoholic fatty liver disease (NAFLD). However, the underlying mechanisms that lead to the reduction of steatosis in NAFLD remain obscure. METHODS Ttp+/+ and Ttp-/- mice were fed with a high-fat diet. Hepatic steatosis was analyzed by Nile Red staining and measurement of inflammatory cytokines. Lipid accumulation and cell death were evaluated in co-culture systems with primary hepatocytes and KCs derived from either Ttp+/+ or Ttp-/- mice. RESULTS Tristetraprolin (TTP), an mRNA binding protein, was essential for the protective effects of metformin in NAFLD. Metformin activated TTP via the AMPK-Sirt1 pathway in hepatocytes and KCs. TTP inhibited TNF-α production in KCs, which in turn decreased hepatocyte necroptosis. Downregulation of Rheb expression by TTP promoted hepatocyte lipophagy via mTORC1 inhibition and increased nuclear translocation of transcription factor-EB (TFEB). Consistently, TTP-deficient NAFLD mice failed to respond to metformin with respect to alleviation of hepatic steatosis, protection of hepatocyte necroptosis, or induction of lipophagy. CONCLUSIONS TTP, which is essential for the protective effects of metformin, may represent a novel primary therapeutic target in NAFLD.
Collapse
Affiliation(s)
- Jeongmin Park
- School of Biological Sciences, University of Ulsan, Ulsan 44610, Republic of Korea
| | - So-Young Rah
- National Creative Research Laboratory for Ca(2+) signaling Network, Chonbuk National University Medical School, Jeonju 54907, Republic of Korea
| | - Hyeong Seok An
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Jong Youl Lee
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Gu Seob Roh
- Department of Anatomy and Convergence Medical Science, Institute of Health Sciences, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | | | - Jeong Woo Park
- School of Biological Sciences, University of Ulsan, Ulsan 44610, Republic of Korea
| | - Chae Ha Yang
- College of Korean Medicine, Daegu Haany University, Daegu 42158, Republic of Korea
| | - Young-Joon Surh
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, Seoul 08826, Republic of Korea; Cancer Research Institute, Seoul National University, Seoul 03080, Republic of Korea
| | - Uh-Hyun Kim
- Department of Biochemistry, School of Medicine, Wonkwang University, Iksan 54538, Republic of Korea
| | - Hun Taeg Chung
- School of Biological Sciences, University of Ulsan, Ulsan 44610, Republic of Korea.
| | - Yeonsoo Joe
- School of Biological Sciences, University of Ulsan, Ulsan 44610, Republic of Korea.
| |
Collapse
|
27
|
Scinicariello S, Soderholm A, Schäfer M, Shulkina A, Schwartz I, Hacker K, Gogova R, Kalis R, Froussios K, Budroni V, Bestehorn A, Clausen T, Kovarik P, Zuber J, Versteeg GA. HUWE1 controls tristetraprolin proteasomal degradation by regulating its phosphorylation. eLife 2023; 12:e83159. [PMID: 36961408 PMCID: PMC10038661 DOI: 10.7554/elife.83159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 02/26/2023] [Indexed: 03/25/2023] Open
Abstract
Tristetraprolin (TTP) is a critical negative immune regulator. It binds AU-rich elements in the untranslated-regions of many mRNAs encoding pro-inflammatory mediators, thereby accelerating their decay. A key but poorly understood mechanism of TTP regulation is its timely proteolytic removal: TTP is degraded by the proteasome through yet unidentified phosphorylation-controlled drivers. In this study, we set out to identify factors controlling TTP stability. Cellular assays showed that TTP is strongly lysine-ubiquitinated, which is required for its turnover. A genetic screen identified the ubiquitin E3 ligase HUWE1 as a strong regulator of TTP proteasomal degradation, which we found to control TTP stability indirectly by regulating its phosphorylation. Pharmacological assessment of multiple kinases revealed that HUWE1-regulated TTP phosphorylation and stability was independent of the previously characterized effects of MAPK-mediated S52/S178 phosphorylation. HUWE1 function was dependent on phosphatase and E3 ligase binding sites identified in the TTP C-terminus. Our findings indicate that while phosphorylation of S52/S178 is critical for TTP stabilization at earlier times after pro-inflammatory stimulation, phosphorylation of the TTP C-terminus controls its stability at later stages.
Collapse
Affiliation(s)
- Sara Scinicariello
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of Vienna, Vienna BioCenter (VBC)ViennaAustria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna BioCenter (VBC)ViennaAustria
| | - Adrian Soderholm
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of Vienna, Vienna BioCenter (VBC)ViennaAustria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna BioCenter (VBC)ViennaAustria
| | - Markus Schäfer
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC)ViennaAustria
| | - Alexandra Shulkina
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of Vienna, Vienna BioCenter (VBC)ViennaAustria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna BioCenter (VBC)ViennaAustria
| | - Irene Schwartz
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of Vienna, Vienna BioCenter (VBC)ViennaAustria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna BioCenter (VBC)ViennaAustria
| | - Kathrin Hacker
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of Vienna, Vienna BioCenter (VBC)ViennaAustria
| | - Rebeca Gogova
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna BioCenter (VBC)ViennaAustria
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC)ViennaAustria
| | - Robert Kalis
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna BioCenter (VBC)ViennaAustria
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC)ViennaAustria
| | - Kimon Froussios
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC)ViennaAustria
| | - Valentina Budroni
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of Vienna, Vienna BioCenter (VBC)ViennaAustria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna BioCenter (VBC)ViennaAustria
| | - Annika Bestehorn
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of Vienna, Vienna BioCenter (VBC)ViennaAustria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna BioCenter (VBC)ViennaAustria
| | - Tim Clausen
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC)ViennaAustria
- Medical University of Vienna, Vienna BioCenter (VBC)ViennaAustria
| | - Pavel Kovarik
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of Vienna, Vienna BioCenter (VBC)ViennaAustria
| | - Johannes Zuber
- Research Institute of Molecular Pathology (IMP), Vienna BioCenter (VBC)ViennaAustria
- Medical University of Vienna, Vienna BioCenter (VBC)ViennaAustria
| | - Gijs A Versteeg
- Department of Microbiology, Immunobiology and Genetics, Max Perutz Labs, University of Vienna, Vienna BioCenter (VBC)ViennaAustria
| |
Collapse
|
28
|
Sowinska W, Wawro M, Biswas DD, Kochan J, Pustelny K, Solecka A, Gupta AS, Mockenhaupt K, Polak J, Kwinta B, Kordula T, Kasza A. The homeostatic function of Regnase-2 restricts neuroinflammation. FASEB J 2023; 37:e22798. [PMID: 36753401 PMCID: PMC9983307 DOI: 10.1096/fj.202201978r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 12/22/2022] [Accepted: 01/20/2023] [Indexed: 02/09/2023]
Abstract
The precise physiological functions and mechanisms regulating RNase Regnase-2 (Reg-2/ZC3H12B/MCPIP2) activity remain enigmatic. We found that Reg-2 actively modulates neuroinflammation in nontransformed cells, including primary astrocytes. Downregulation of Reg-2 in these cells results in increased mRNA levels of proinflammatory cytokines IL-1β and IL-6. In primary astrocytes, Reg-2 also regulates the mRNA level of Regnase-1 (Reg-1/ZC3H12A/MCPIP1). Reg-2 is expressed at high levels in the healthy brain, but its expression is reduced during neuroinflammation as well as glioblastoma progression. This process is associated with the upregulation of Reg-1. Conversely, overexpression of Reg-2 is accompanied by the downregulation of Reg-1 in glioma cells in a nucleolytic NYN/PIN domain-dependent manner. Interestingly, low levels of Reg-2 and high levels of Reg-1 correlate with poor-glioblastoma patients' prognoses. While Reg-2 restricts the basal levels of proinflammatory cytokines in resting astrocytes, its expression is reduced in IL-1β-activated astrocytes. Following IL-1β exposure, Reg-2 is phosphorylated, ubiquitinated, and degraded by proteasomes. Simultaneously, the Reg-2 transcript is destabilized by tristetraprolin (TTP) and Reg-1 through the AREs elements and conservative stem-loop structure present in its 3'UTR. Thus, the peer-control loop, of Reg-1 and Reg-2 opposing each other, exists. The involvement of TTP in Reg-2 mRNA turnover is confirmed by the observation that high TTP levels correlate with the downregulation of the Reg-2 expression in high-grade human gliomas. Additionally, obtained results reveal the importance of Reg-2 in inhibiting human and mouse glioma cell proliferation. Our current studies identify Reg-2 as a critical regulator of homeostasis in the brain.
Collapse
Affiliation(s)
- Weronika Sowinska
- Department of Cell Biochemistry, Faculty of Biotechnology, Biochemistry and Biophysics, Jagiellonian University, Krakow, Poland
| | - Mateusz Wawro
- Department of Cell Biochemistry, Faculty of Biotechnology, Biochemistry and Biophysics, Jagiellonian University, Krakow, Poland
| | - Debolina D. Biswas
- Department of Biochemistry and Molecular Biology, School of Medicine and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VI 23298, USA
| | - Jakub Kochan
- Department of Cell Biochemistry, Faculty of Biotechnology, Biochemistry and Biophysics, Jagiellonian University, Krakow, Poland
| | - Katarzyna Pustelny
- Department of Cell Biochemistry, Faculty of Biotechnology, Biochemistry and Biophysics, Jagiellonian University, Krakow, Poland
| | - Aleksandra Solecka
- Department of Cell Biochemistry, Faculty of Biotechnology, Biochemistry and Biophysics, Jagiellonian University, Krakow, Poland
| | - Angela S. Gupta
- Department of Biochemistry and Molecular Biology, School of Medicine and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VI 23298, USA
| | - Karli Mockenhaupt
- Department of Biochemistry and Molecular Biology, School of Medicine and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VI 23298, USA
| | - Jarosław Polak
- Department of Neurosurgery and Neurotraumatology, Jagiellonian University Medical College, Kraków, Poland
| | - Borys Kwinta
- Department of Neurosurgery and Neurotraumatology, Jagiellonian University Medical College, Kraków, Poland
| | - Tomasz Kordula
- Department of Biochemistry and Molecular Biology, School of Medicine and the Massey Cancer Center, Virginia Commonwealth University, Richmond, VI 23298, USA.,To whom correspondence should be addressed: Aneta Kasza, , Tel. (+48)126646521 and Tomasz Kordula, , Tel. (+1)804-828-0771
| | - Aneta Kasza
- Department of Cell Biochemistry, Faculty of Biotechnology, Biochemistry and Biophysics, Jagiellonian University, Krakow, Poland,To whom correspondence should be addressed: Aneta Kasza, , Tel. (+48)126646521 and Tomasz Kordula, , Tel. (+1)804-828-0771
| |
Collapse
|
29
|
RBP-RNA interactions in the control of autoimmunity and autoinflammation. Cell Res 2023; 33:97-115. [PMID: 36599968 PMCID: PMC9892603 DOI: 10.1038/s41422-022-00752-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Accepted: 11/07/2022] [Indexed: 01/06/2023] Open
Abstract
Autoimmunity and autoinflammation arise from aberrant immunological and inflammatory responses toward self-components, contributing to various autoimmune diseases and autoinflammatory diseases. RNA-binding proteins (RBPs) are essential for immune cell development and function, mainly via exerting post-transcriptional regulation of RNA metabolism and function. Functional dysregulation of RBPs and abnormities in RNA metabolism are closely associated with multiple autoimmune or autoinflammatory disorders. Distinct RBPs play critical roles in aberrant autoreactive inflammatory responses via orchestrating a complex regulatory network consisting of DNAs, RNAs and proteins within immune cells. In-depth characterizations of RBP-RNA interactomes during autoimmunity and autoinflammation will lead to a better understanding of autoimmune pathogenesis and facilitate the development of effective therapeutic strategies. In this review, we summarize and discuss the functions of RBP-RNA interactions in controlling aberrant autoimmune inflammation and their potential as biomarkers and therapeutic targets.
Collapse
|
30
|
Hong D, Jeong S. 3'UTR Diversity: Expanding Repertoire of RNA Alterations in Human mRNAs. Mol Cells 2023; 46:48-56. [PMID: 36697237 PMCID: PMC9880603 DOI: 10.14348/molcells.2023.0003] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 01/05/2023] [Accepted: 01/08/2023] [Indexed: 01/27/2023] Open
Abstract
Genomic information stored in the DNA is transcribed to the mRNA and translated to proteins. The 3' untranslated regions (3'UTRs) of the mRNA serve pivotal roles in posttranscriptional gene expression, regulating mRNA stability, translation, and localization. Similar to DNA mutations producing aberrant proteins, RNA alterations expand the transcriptome landscape and change the cellular proteome. Recent global analyses reveal that many genes express various forms of altered RNAs, including 3'UTR length variants. Alternative polyadenylation and alternative splicing are involved in diversifying 3'UTRs, which could act as a hidden layer of eukaryotic gene expression control. In this review, we summarize the functions and regulations of 3'UTRs and elaborate on the generation and functional consequences of 3'UTR diversity. Given that dynamic 3'UTR length control contributes to phenotypic complexity, dysregulated 3'UTR diversity might be relevant to disease development, including cancers. Thus, 3'UTR diversity in cancer could open exciting new research areas and provide avenues for novel cancer theragnostics.
Collapse
Affiliation(s)
- Dawon Hong
- Laboratory of RNA Cell Biology, Department of Bioconvergence Engineering, Dankook University Graduate School, Yongin 16892, Korea
| | - Sunjoo Jeong
- Laboratory of RNA Cell Biology, Department of Bioconvergence Engineering, Dankook University Graduate School, Yongin 16892, Korea
| |
Collapse
|
31
|
Kaposi's sarcoma-associated herpesvirus (KSHV) utilizes the NDP52/CALCOCO2 selective autophagy receptor to disassemble processing bodies. PLoS Pathog 2023; 19:e1011080. [PMID: 36634147 PMCID: PMC9876383 DOI: 10.1371/journal.ppat.1011080] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 01/25/2023] [Accepted: 12/22/2022] [Indexed: 01/13/2023] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) causes the inflammatory and angiogenic endothelial cell neoplasm, Kaposi's sarcoma (KS). We previously demonstrated that the KSHV Kaposin B (KapB) protein promotes inflammation via the disassembly of cytoplasmic ribonucleoprotein granules called processing bodies (PBs). PBs modify gene expression by silencing or degrading labile messenger RNAs (mRNAs), including many transcripts that encode inflammatory or angiogenic proteins associated with KS disease. Although our work implicated PB disassembly as one of the causes of inflammation during KSHV infection, the precise mechanism used by KapB to elicit PB disassembly was unclear. Here we reveal a new connection between the degradative process of autophagy and PB disassembly. We show that both latent KSHV infection and KapB expression enhanced autophagic flux via phosphorylation of the autophagy regulatory protein, Beclin. KapB was necessary for this effect, as infection with a recombinant virus that does not express the KapB protein did not induce Beclin phosphorylation or autophagic flux. Moreover, we showed that PB disassembly mediated by KSHV or KapB, depended on autophagy genes and the selective autophagy receptor NDP52/CALCOCO2 and that the PB scaffolding protein, Pat1b, co-immunoprecipitated with NDP52. These studies reveal a new role for autophagy and the selective autophagy receptor NDP52 in promoting PB turnover and the concomitant synthesis of inflammatory molecules during KSHV infection.
Collapse
|
32
|
Innate immune sensing of pathogens and its post-transcriptional regulations by RNA-binding proteins. Arch Pharm Res 2023; 46:65-77. [PMID: 36725818 PMCID: PMC9891759 DOI: 10.1007/s12272-023-01429-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 01/25/2023] [Indexed: 02/03/2023]
Abstract
Innate immunity is one of the most ancient and conserved aspect of the immune system. It is responsible for an anti-infective response and has been intrinsically linked to the generation of inflammation. While the inflammatory response entails signaling to the adaptive immune system, it can be self-perpetuating and over-exaggerated, resulting in deleterious consequences, including cytokine storm, sepsis, and the development of inflammatory and autoimmune diseases. Cytokines are the defining features of the immune system. They are critical to mediation of inflammation and host immune defense, and are tightly regulated at several levels, including transcriptional and post-transcriptional levels. Recently, the role of post-transcriptional regulation in fine-tuning cytokine expression has become more appreciated. This interest has advanced our understanding of how various mechanisms are integrated and regulated to determine the amount of cytokine production in cells during inflammatory responses. Here, we would like to review how innate immunity recognizes and responds to pathogens by pattern-recognition receptors, and the molecular mechanisms regulating inflammatory responses, with a focus on the post-transcriptional regulations of inflammatory mediators by RNA-binding proteins, especially Regnase-1. Finally, we will discuss the regulatory mechanisms of Regnase-1 and highlight therapeutic strategies based on targeting Regnase-1 activity and its turnover as potential treatment options for chronic and autoimmune diseases.
Collapse
|
33
|
Li X, An Y, Wang Q, Han X. The new ceRNA crosstalk between mRNAs and miRNAs in intervertebral disc degeneration. Front Cell Dev Biol 2022; 10:1083983. [PMID: 36531954 PMCID: PMC9755594 DOI: 10.3389/fcell.2022.1083983] [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: 10/29/2022] [Accepted: 11/18/2022] [Indexed: 01/28/2025] Open
Abstract
Degeneration of the intervertebral disc has been linked to lower back pain. To date, pathophysiological mechanisms of intervertebral disc degeneration (IDD) remain unclear; it is meaningful to find effective diagnostic biomarkers and new therapeutic strategies for IDD. This study aimed to reveal the molecular mechanism of IDD pathogenesis from the multidimensional transcriptomics perspective. Here, we acquired IDD bulk omics datasets (GSE67567 and GSE167199) including mRNA, microRNA expression profiles, and single-cell RNA sequencing (GSE199866) from the public Gene Expression Omnibus (GEO) database. Through principal component analysis and Venn analysis, we found different expression patterns in the IDD transcription level and identified 156 common DEGs in both bulk datasets. GO and KEGG functional analyses showed these dysregulators were mostly enriched in the collagen-containing extracellular matrix, cartilage development, chondrocyte differentiation, and immune response pathways. We also constructed a potentially dysregulated competing endogenous RNA (ceRNA) network between mRNAs and miRNAs related to IDD based on microRNA target information and co-expression analysis of RNA profiles and identified 36 ceRNA axes including ZFP36/miR-155-5p/FOS, BTG2/hsa-miR-185-5p/SOCS3, and COL9A2/hsa-miR-664a-5p/IBA57. Finally, in integrating bulk and single-cell transcriptome data analyses, a total of three marker genes, COL2A1, PAX1, and ZFP36L2, were identified. In conclusion, the key genes and the new ceRNA crosstalk we identified in intervertebral disc degeneration may provide new targets for the treatment of IDD.
Collapse
Affiliation(s)
- Xingye Li
- Department of Spine Surgery, Beijing Jishuitan Hospital, Fourth Clinical College of Peking University, Beijing, China
| | | | | | | |
Collapse
|
34
|
Qi Y, Wang M, Jiang Q. PABPC1--mRNA stability, protein translation and tumorigenesis. Front Oncol 2022; 12:1025291. [PMID: 36531055 PMCID: PMC9753129 DOI: 10.3389/fonc.2022.1025291] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/08/2022] [Indexed: 09/29/2023] Open
Abstract
Mammalian poly A-binding proteins (PABPs) are highly conserved multifunctional RNA-binding proteins primarily involved in the regulation of mRNA translation and stability, of which PABPC1 is considered a central regulator of cytoplasmic mRNA homing and is involved in a wide range of physiological and pathological processes by regulating almost every aspect of RNA metabolism. Alterations in its expression and function disrupt intra-tissue homeostasis and contribute to the development of various tumors. There is increasing evidence that PABPC1 is aberrantly expressed in a variety of tumor tissues and cancers such as lung, gastric, breast, liver, and esophageal cancers, and PABPC1 might be used as a potential biomarker for tumor diagnosis, treatment, and clinical application in the future. In this paper, we review the abnormal expression, functional role, and molecular mechanism of PABPC1 in tumorigenesis and provide directions for further understanding the regulatory role of PABPC1 in tumor cells.
Collapse
Affiliation(s)
- Ya Qi
- Department of Gynecology and Obstetrics, Shengjing Hospital Affiliated of China Medical University, Shenyang, Liaoning, China
| | - Min Wang
- Department of Gynecology and Obstetrics, Shengjing Hospital Affiliated of China Medical University, Shenyang, Liaoning, China
| | - Qi Jiang
- Second Department of Clinical Medicine, China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
35
|
Snyder BL, Blackshear PJ. Clinical implications of tristetraprolin (TTP) modulation in the treatment of inflammatory diseases. Pharmacol Ther 2022; 239:108198. [PMID: 35525391 PMCID: PMC9636069 DOI: 10.1016/j.pharmthera.2022.108198] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/19/2022] [Accepted: 04/25/2022] [Indexed: 11/24/2022]
Abstract
Abnormal regulation of pro-inflammatory cytokine and chemokine mediators can contribute to the excess inflammation characteristic of many autoimmune diseases, such as rheumatoid arthritis, psoriasis, Crohn's disease, type 1 diabetes, and many others. The tristetraprolin (TTP) family consists of a small group of related RNA-binding proteins that bind to preferred AU-rich binding sites within the 3'-untranslated regions of specific mRNAs to promote mRNA deadenylation and decay. TTP deficient mice develop a severe systemic inflammatory syndrome consisting of arthritis, myeloid hyperplasia, dermatitis, autoimmunity and cachexia, due at least in part to the excess accumulation of proinflammatory chemokine and cytokine mRNAs and their encoded proteins. To investigate the possibility that increased TTP expression or activity might have a beneficial effect on inflammatory diseases, at least two mouse models have been developed that provide proof of principle that increasing TTP activity can promote the decay of pro-inflammatory and other relevant transcripts, and decrease the severity of mouse models of inflammatory disease. Animal studies of this type are summarized here, and we briefly review the prospects for harnessing these insights for the development of TTP-based anti-inflammatory treatments in humans.
Collapse
Affiliation(s)
- Brittany L Snyder
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, United States of America; Department of Biochemistry, Duke University Medical Center, Durham, NC 27710, United States of America
| | - Perry J Blackshear
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, NC 27709, United States of America; Department of Medicine, Duke University Medical Center, Durham, NC 27710, United States of America; Department of Biochemistry, Duke University Medical Center, Durham, NC 27710, United States of America.
| |
Collapse
|
36
|
Serhii K, Anastasiia H, Oksana M, Kyrylo L, Liubov S, Nataliia V, Iryna I, Rostyslav S, Alla R. Tristetraprolin expression levels and methylation status in breast cancer. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
37
|
Cook ME, Bradstreet TR, Webber AM, Kim J, Santeford A, Harris KM, Murphy MK, Tran J, Abdalla NM, Schwarzkopf EA, Greco SC, Halabi CM, Apte RS, Blackshear PJ, Edelson BT. The ZFP36 family of RNA binding proteins regulates homeostatic and autoreactive T cell responses. Sci Immunol 2022; 7:eabo0981. [PMID: 36269839 PMCID: PMC9832469 DOI: 10.1126/sciimmunol.abo0981] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
RNA binding proteins are important regulators of T cell activation, proliferation, and cytokine production. The zinc finger protein 36 (ZFP36) family genes (Zfp36, Zfp36l1, and Zfp36l2) encode RNA binding proteins that promote the degradation of transcripts containing AU-rich elements. Numerous studies have demonstrated both individual and shared functions of the ZFP36 family in immune cells, but their collective function in T cells remains unclear. Here, we found a redundant and critical role for the ZFP36 proteins in regulating T cell quiescence. T cell-specific deletion of all three ZFP36 family members in mice resulted in early lethality, immune cell activation, and multiorgan pathology characterized by inflammation of the eyes, central nervous system, kidneys, and liver. Mice with T cell-specific deletion of any two Zfp36 genes were protected from this spontaneous syndrome. Triply deficient T cells overproduced proinflammatory cytokines, including IFN-γ, TNF, and GM-CSF, due to increased mRNA stability of these transcripts. Unexpectedly, T cell-specific deletion of both Zfp36l1 and Zfp36l2 rendered mice resistant to experimental autoimmune encephalomyelitits due to failed priming of antigen-specific CD4+ T cells. ZFP36L1 and ZFP36L2 double-deficient CD4+ T cells had poor proliferation during in vitro T helper cell polarization. Thus, the ZFP36 family redundantly regulates T cell quiescence at homeostasis, but ZFP36L1 and ZFP36L2 are specifically required for antigen-specific T cell clonal expansion.
Collapse
Affiliation(s)
- Melissa E. Cook
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Tara R. Bradstreet
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Ashlee M. Webber
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Jongshin Kim
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine; St. Louis, MO, USA
- Current address: Medical Science and Engineering Program, School of Convergence Science and Technology, Pohang University of Science and Technology; Pohang, Korea
| | - Andrea Santeford
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine; St. Louis, MO, USA
| | - Kevin M. Harris
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Maegan K. Murphy
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Jennifer Tran
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO, USA
- Department of Medicine, Washington University School of Medicine; St. Louis, MO, USA
| | - Nada M. Abdalla
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO, USA
| | - Elizabeth A. Schwarzkopf
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO, USA
- Current address: Wugen, Inc.; St. Louis, MO, USA
| | - Suellen C. Greco
- Division of Comparative Medicine, Washington University School of Medicine; St. Louis, MO, USA
| | - Carmen M. Halabi
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA
| | - Rajendra S. Apte
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine; St. Louis, MO, USA
- Department of Medicine, Washington University School of Medicine; St. Louis, MO, USA
- Department of Developmental Biology, Washington University School of Medicine; St. Louis, MO, USA
| | - Perry J. Blackshear
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health; Research Triangle Park, NC, USA
- Departments of Medicine and Biochemistry, Duke University Medical Center; Durham, NC, USA
| | - Brian T. Edelson
- Department of Pathology and Immunology, Washington University School of Medicine; St. Louis, MO, USA
| |
Collapse
|
38
|
Kwack KH, Zhang L, Kramer ED, Thiyagarajan R, Lamb NA, Arao Y, Bard JE, Seldeen KL, Troen BR, Blackshear PJ, Abrams SI, Kirkwood KL. Tristetraprolin limits age-related expansion of myeloid-derived suppressor cells. Front Immunol 2022; 13:1002163. [PMID: 36263047 PMCID: PMC9573970 DOI: 10.3389/fimmu.2022.1002163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Accepted: 09/16/2022] [Indexed: 11/13/2022] Open
Abstract
Aging results in enhanced myelopoiesis, which is associated with an increased prevalence of myeloid leukemias and the production of myeloid-derived suppressor cells (MDSCs). Tristetraprolin (TTP) is an RNA binding protein that regulates immune-related cytokines and chemokines by destabilizing target mRNAs. As TTP expression is known to decrease with age in myeloid cells, we used TTP-deficient (TTPKO) mice to model aged mice to study TTP regulation in age-related myelopoiesis. Both TTPKO and myeloid-specific TTPKO (cTTPKO) mice had significant increases in both MDSC subpopulations M-MDSCs (CD11b+Ly6ChiLy6G-) and PMN-MDSCs (CD11b+Ly6CloLy6G+), as well as macrophages (CD11b+F4/80+) in the spleen and mesenteric lymph nodes; however, no quantitative changes in MDSCs were observed in the bone marrow. In contrast, gain-of-function TTP knock-in (TTPKI) mice had no change in MDSCs compared with control mice. Within the bone marrow, total granulocyte-monocyte progenitors (GMPs) and monocyte progenitors (MPs), direct antecedents of M-MDSCs, were significantly increased in both cTTPKO and TTPKO mice, but granulocyte progenitors (GPs) were significantly increased only in TTPKO mice. Transcriptomic analysis of the bone marrow myeloid cell populations revealed that the expression of CC chemokine receptor 2 (CCR2), which plays a key role in monocyte mobilization to inflammatory sites, was dramatically increased in both cTTPKO and TTPKO mice. Concurrently, the concentration of CC chemokine ligand 2 (CCL2), a major ligand of CCR2, was high in the serum of cTTPKO and TTPKO mice, suggesting that TTP impacts the mobilization of M-MDSCs from the bone marrow to inflammatory sites during aging via regulation of the CCR2-CCL2 axis. Collectively, these studies demonstrate a previously unrecognized role for TTP in regulating age-associated myelopoiesis through the expansion of specific myeloid progenitors and M-MDSCs and their recruitment to sites of injury, inflammation, or other pathologic perturbations.
Collapse
Affiliation(s)
- Kyu Hwan Kwack
- Department of Oral Biology, University at Buffalo, Buffalo, NY, United States
- Department of Oral Microbiology, College of Dentistry, Kyung Hee University, Seoul, South Korea
| | - Lixia Zhang
- Department of Oral Biology, University at Buffalo, Buffalo, NY, United States
| | - Elliot D. Kramer
- Department of Medicine, University at Buffalo, Buffalo, NY, United States
- Departments of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Ramkumar Thiyagarajan
- Department of Medicine, University at Buffalo, Buffalo, NY, United States
- Division of Geriatrics and Palliative Medicine, University at Buffalo, Buffalo, NY, United States
- Research Service, Veterans Affairs Western New York Healthcare Service, Buffalo, NY, United States
| | - Natalie A. Lamb
- Department of Biochemistry, University at Buffalo, Buffalo, NY, United States
- Genomics and Bioinformatics Core, New York State Center of Excellence for Bioinformatics and Life Sciences, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Yukitomo Arao
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| | - Jonathan E. Bard
- Department of Biochemistry, University at Buffalo, Buffalo, NY, United States
- Genomics and Bioinformatics Core, New York State Center of Excellence for Bioinformatics and Life Sciences, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Kenneth L. Seldeen
- Department of Medicine, University at Buffalo, Buffalo, NY, United States
- Division of Geriatrics and Palliative Medicine, University at Buffalo, Buffalo, NY, United States
- Research Service, Veterans Affairs Western New York Healthcare Service, Buffalo, NY, United States
| | - Bruce R. Troen
- Department of Medicine, University at Buffalo, Buffalo, NY, United States
- Division of Geriatrics and Palliative Medicine, University at Buffalo, Buffalo, NY, United States
- Research Service, Veterans Affairs Western New York Healthcare Service, Buffalo, NY, United States
| | - Perry J. Blackshear
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
- Departments of Biochemistry & Medicine, Duke University Medical Center, Durham, NC, United States
| | - Scott I. Abrams
- Departments of Immunology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Keith L. Kirkwood
- Department of Oral Biology, University at Buffalo, Buffalo, NY, United States
- Head & Neck/Plastic & Reconstructive Surgery, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| |
Collapse
|
39
|
Carreño A, Lykke-Andersen J. The Conserved CNOT1 Interaction Motif of Tristetraprolin Regulates ARE-mRNA Decay Independently of the p38 MAPK-MK2 Kinase Pathway. Mol Cell Biol 2022; 42:e0005522. [PMID: 35920669 PMCID: PMC9476947 DOI: 10.1128/mcb.00055-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The regulation of the mRNA decay activator Tristetraprolin (TTP) by the p38 mitogen-activated protein kinase (MAPK) pathway during the mammalian inflammatory response represents a paradigm for the control of mRNA turnover by signaling. TTP activity is regulated through multiple phosphorylation sites, including an evolutionary conserved serine in its CNOT1 Interacting Motif (CIM) whose phosphorylation disrupts an interaction with CNOT1 of the CCR4-NOT deadenylase complex. Here we present evidence that the TTP CIM recruits the CCR4-NOT deadenylase complex and activates mRNA degradation cooperatively with the conserved tryptophan residues of TTP, previously identified to interact with CNOT9. Surprisingly, the TTP CIM remains unphosphorylated and capable of promoting association with the CCR4-NOT complex and mRNA decay upon activation of p38-MAPK-activated kinase MK2, a well-established regulator of TTP activity. The CIM is instead targeted by other kinases including PKCα. These observations suggest that signaling pathways regulate TTP activity in a cooperative manner and that the p38 MAPK-MK2 kinase pathway relies on the activation of additional kinase pathway(s) to fully control TTP function.
Collapse
Affiliation(s)
- Alberto Carreño
- Department of Molecular Biology, School of Biological Sciences, University of California San Diego, La Jolla, California, USA
| | - Jens Lykke-Andersen
- Department of Molecular Biology, School of Biological Sciences, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
40
|
Lu F, Hu F, Qiu B, Zou H, Xu J. Identification of novel biomarkers in septic cardiomyopathy via integrated bioinformatics analysis and experimental validation. Front Genet 2022; 13:929293. [PMID: 35957694 PMCID: PMC9358039 DOI: 10.3389/fgene.2022.929293] [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: 04/26/2022] [Accepted: 06/27/2022] [Indexed: 11/21/2022] Open
Abstract
Purpose: Septic cardiomyopathy (SCM) is an important world public health problem with high morbidity and mortality. It is necessary to identify SCM biomarkers at the genetic level to identify new therapeutic targets and strategies. Method: DEGs in SCM were identified by comprehensive bioinformatics analysis of microarray datasets (GSE53007 and GSE79962) downloaded from the GEO database. Subsequently, bioinformatics analysis was used to conduct an in-depth exploration of DEGs, including GO and KEGG pathway enrichment analysis, PPI network construction, and key gene identification. The top ten Hub genes were identified, and then the SCM model was constructed by treating HL-1 cells and AC16 cells with LPS, and these top ten Hub genes were examined using qPCR. Result: STAT3, SOCS3, CCL2, IL1R2, JUNB, S100A9, OSMR, ZFP36, and HAMP were significantly elevated in the established SCM cells model. Conclusion: After bioinformatics analysis and experimental verification, it was demonstrated that STAT3, SOCS3, CCL2, IL1R2, JUNB, S100A9, OSMR, ZFP36, and HAMP might play important roles in SCM.
Collapse
Affiliation(s)
- Feng Lu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Feng Hu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Baiquan Qiu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Hongpeng Zou
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jianjun Xu
- Department of Cardiothoracic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- *Correspondence: Jianjun Xu,
| |
Collapse
|
41
|
Chai G, Qi G, Wang D, Zhuang Y, Xu H, Bai Z, Bai MY, Hu R, Wang ZY, Zhou G, Kong Y. The CCCH zinc finger protein C3H15 negatively regulates cell elongation by inhibiting brassinosteroid signaling. PLANT PHYSIOLOGY 2022; 189:285-300. [PMID: 35139225 PMCID: PMC9070797 DOI: 10.1093/plphys/kiac046] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 01/10/2022] [Indexed: 05/20/2023]
Abstract
Plant CCCH proteins participate in the control of multiple developmental and adaptive processes, but the regulatory mechanisms underlying these processes are not well known. In this study, we showed that the Arabidopsis (Arabidopsis thaliana) CCCH protein C3H15 negatively regulates cell elongation by inhibiting brassinosteroid (BR) signaling. Genetic and biochemical evidence showed that C3H15 functions downstream of the receptor BR INSENSITIVE 1 (BRI1) as a negative regulator in the BR pathway. C3H15 is phosphorylated by the GLYCOGEN SYNTHASE KINASE 3 -like kinase BR-INSENSITIVE 2 (BIN2) at Ser111 in the cytoplasm in the absence of BRs. Upon BR perception, C3H15 transcription is enhanced, and the phosphorylation of C3H15 by BIN2 is reduced. The dephosphorylated C3H15 protein accumulates in the nucleus, where C3H15 regulates transcription via G-rich elements (typically GGGAGA). C3H15 and BRASSINAZOLE RESISTANT 1 (BZR1)/BRI1-EMS-SUPPRESSOR 1 (BES1), two central transcriptional regulators of BR signaling, directly suppress each other and share a number of BR-responsive target genes. Moreover, C3H15 antagonizes BZR1 and BES1 to regulate the expression of their shared cell elongation-associated target gene, SMALL AUXIN-UP RNA 15 (SAUR15). This study demonstrates that C3H15-mediated BR signaling may be parallel to, or even attenuate, the dominant BZR1 and BES1 signaling pathways to control cell elongation. This finding expands our understanding of the regulatory mechanisms underlying BR-induced cell elongation in plants.
Collapse
Affiliation(s)
| | | | | | | | - Hua Xu
- Qingdao Institute of BioEnergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao 266101, China
| | - Zetao Bai
- Key Laboratory of Biology and Genetic Improvement of Oil Crops, Ministry of Agriculture and Rural Affairs, Oil Crops Research Institute of the Chinese Academy of Agricultural Sciences, Wuhan 430062, China
| | - Ming-Yi Bai
- Key Laboratory of Plant Cell Engineering and Germplasm Innovation, Ministry of Education, School of Life Sciences, Shandong University, Jinan 250100, China
| | - Ruibo Hu
- Qingdao Institute of BioEnergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao 266101, China
| | - Zeng-yu Wang
- College of Grassland Science, Qingdao Agricultural University, Qingdao 266109, China
| | | | | |
Collapse
|
42
|
Hicks SN, Venters RA, Blackshear PJ. Backbone and sidechain 1H, 15N and 13C resonance assignments of the free and RNA-bound tandem zinc finger domain of the tristetraprolin family member from Selaginella moellendorffii. BIOMOLECULAR NMR ASSIGNMENTS 2022; 16:153-158. [PMID: 35279790 PMCID: PMC9196822 DOI: 10.1007/s12104-022-10073-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 02/24/2022] [Indexed: 06/14/2023]
Abstract
Members of the tristetraprolin (TTP) family of RNA binding proteins (RBPs) regulate the metabolism of a variety of mRNA targets. In mammals, these proteins modulate many physiological processes, including immune cell activation, hematopoiesis, and embryonic development. Regulation of mRNA stability by these proteins requires that the tandem zinc finger (TZF) domain binds initially and directly to target mRNAs, ultimately leading to their deadenylation and decay. Proteins of this type throughout eukarya possess a highly conserved TZF domain, suggesting that they are all capable of high-affinity RNA binding. However, the mechanism of TTP-mediated mRNA decay is largely undefined. Given the vital role that these TTP family proteins play in maintaining RNA homeostasis throughout eukaryotes, we focused here on the first, key step in this process: recognition and binding of the TZF domain to target RNA. For these studies, we chose a primitive plant, the spikemoss Selaginella moellendorffii, which last shared a common ancestor with humans more than a billion years ago. Here we report the near complete backbone and side chain resonance assignments of the spikemoss TZF domain, including: (1) the assignment of the RNA-TZF domain complex, representing one of only two data sets currently available for the entire TTP family of proteins; and (2) the first NMR resonance assignments of the entire TZF domain, in the RNA-free form. This work will serve as the basis for further NMR structural investigations aimed at gaining insights into the process of RNA recognition and the mechanisms of TTP-mediated mRNA decay.
Collapse
Affiliation(s)
- Stephanie N Hicks
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA.
| | - Ronald A Venters
- Duke University NMR Center, Duke University, Durham, NC, 27710, USA
- Department of Radiology, Duke University, Durham, NC, 27710, USA
| | - Perry J Blackshear
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, 27709, USA
- Department of Medicine, Duke University Medical Center, Durham, NC, 27710, USA
- Department of Biochemistry, Duke University Medical Center, Durham, NC, 27710, USA
| |
Collapse
|
43
|
Activation of the MKK3-p38-MK2-ZFP36 Axis by Coronavirus Infection Restricts the Upregulation of AU-Rich Element-Containing Transcripts in Proinflammatory Responses. J Virol 2022; 96:e0208621. [PMID: 34985993 DOI: 10.1128/jvi.02086-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Coronavirus infections induce the expression of multiple proinflammatory cytokines and chemokines. We have previously shown that in cells infected with gammacoronavirus infectious bronchitis virus (IBV), interleukin 6 (IL-6), and IL-8 were drastically upregulated, and the MAP kinase p38 and the integrated stress response pathways were implicated in this process. In this study, we report that coronavirus infection activates a negative regulatory loop that restricts the upregulation of a number of proinflammatory genes. As revealed by the initial transcriptomic and subsequent validation analyses, the anti-inflammatory adenine-uridine (AU)-rich element (ARE)-binding protein, zinc finger protein 36 (ZFP36), and its related family members were upregulated in cells infected with IBV and three other coronaviruses, alphacoronaviruses porcine epidemic diarrhea virus (PEDV), human coronavirus 229E (HCoV-229E), and betacoronavirus HCoV-OC43, respectively. Characterization of the functional roles of ZFP36 during IBV infection demonstrated that ZFP36 promoted the degradation of transcripts coding for IL-6, IL-8, dual-specificity phosphatase 1 (DUSP1), prostaglandin-endoperoxide synthase 2 (PTGS2) and TNF-α-induced protein 3 (TNFAIP3), through binding to AREs in these transcripts. Consistently, knockdown and inhibition of JNK and p38 kinase activities reduced the expression of ZFP36, as well as the expression of IL-6 and IL-8. On the contrary, overexpression of mitogen-activated protein kinase kinase 3 (MKK3) and MAPKAP kinase-2 (MK2), the upstream and downstream kinases of p38, respectively, increased the expression of ZFP36 and decreased the expression of IL-8. Taken together, this study reveals an important regulatory role of the MKK3-p38-MK2-ZFP36 axis in coronavirus infection-induced proinflammatory response. IMPORTANCE Excessive and uncontrolled induction and release of proinflammatory cytokines and chemokines, the so-called cytokine release syndrome (CRS), would cause life-threatening complications and multiple organ failure in severe coronavirus infections, including severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS) and COVID-19. This study reveals that coronavirus infection also induces the expression of ZFP36, an anti-inflammatory ARE-binding protein, promoting the degradation of ARE-containing transcripts coding for IL-6 and IL-8 as well as a number of other proteins related to inflammatory response. Furthermore, the p38 MAP kinase, its upstream kinase MKK3 and downstream kinase MK2 were shown to play a regulatory role in upregulation of ZFP36 during coronavirus infection cycles. This MKK3-p38-MK2-ZFP36 axis would constitute a potential therapeutic target for severe coronavirus infections.
Collapse
|
44
|
Clayer E, Frank D, Anderton H, Zhang S, Kueh A, Heim V, Nutt SL, Chopin M, Bouillet P. ZC3H12C expression in dendritic cells is necessary to prevent lymphadenopathy of skin‐draining lymph nodes. Immunol Cell Biol 2022; 100:160-173. [PMID: 35048402 PMCID: PMC9303644 DOI: 10.1111/imcb.12521] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 11/23/2021] [Accepted: 01/17/2022] [Indexed: 11/28/2022]
Abstract
The role of RNA‐binding proteins of the CCCH‐containing family in regulating proinflammatory cytokine production and inflammation is increasingly recognized. We have identified ZC3H12C (Regnase‐3) as a potential post‐transcriptional regulator of tumor necrosis factor expression and have investigated its role in vivo by generating Zc3h12c‐deficient mice that express green fluorescent protein instead of ZC3H12C. Zc3h12c‐deficient mice develop hypertrophic lymph nodes. In the immune system, ZC3H12C expression is mostly restricted to the dendritic cell (DC) populations, and we show that DC‐restricted ZC3H12C depletion is sufficient to cause lymphadenopathy. ZC3H12C can regulate Tnf messenger RNA stability via its RNase activity in vitro, and we confirmed the role of Tnf in the development of lymphadenopathy. Finally, we found that loss of ZC3H12C did not impact the outcome of skin inflammation in the imiquimod‐induced murine model of psoriasis, despite Zc3h12c being identified as a risk factor for psoriasis susceptibility in several genome‐wide association studies. Our data suggest a role for ZC3H12C in DC‐driven skin homeostasis.
Collapse
Affiliation(s)
- Elise Clayer
- The Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia
- Department of Medical Biology The University of Melbourne Melbourne VIC Australia
- ZAUM – Centre of Allergy and Environment Helmholtz Centre and Technical University of Munich Munich Germany
| | - Daniel Frank
- The Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia
- Department of Medical Biology The University of Melbourne Melbourne VIC Australia
| | - Holly Anderton
- The Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia
- Department of Medical Biology The University of Melbourne Melbourne VIC Australia
| | - Shengbo Zhang
- The Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia
- Department of Medical Biology The University of Melbourne Melbourne VIC Australia
| | - Andrew Kueh
- The Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia
- Department of Medical Biology The University of Melbourne Melbourne VIC Australia
| | - Valentin Heim
- The Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia
- Department of Medical Biology The University of Melbourne Melbourne VIC Australia
- Immatics Biotechnologies GmbH Munich Germany
| | - Stephen L Nutt
- The Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia
- Department of Medical Biology The University of Melbourne Melbourne VIC Australia
| | - Michaël Chopin
- The Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia
- Department of Medical Biology The University of Melbourne Melbourne VIC Australia
| | - Philippe Bouillet
- The Walter and Eliza Hall Institute of Medical Research Parkville VIC Australia
- Department of Medical Biology The University of Melbourne Melbourne VIC Australia
| |
Collapse
|
45
|
Uppala JK, Ghosh C, Sabat G, Dey M. Pull-down of Biotinylated RNA and Associated Proteins. Bio Protoc 2022; 12:e4331. [PMID: 35340298 PMCID: PMC8899547 DOI: 10.21769/bioprotoc.4331] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 10/28/2021] [Accepted: 01/05/2022] [Indexed: 09/23/2023] Open
Abstract
Mapping networks of RNA-protein interactions in cells is essential for understanding the inner workings of many biological processes, including RNA processing, trafficking, and translation. Current in vivo methods for studying protein-RNA interactions rely mostly on purification of poly(A) transcripts, which represent only ~2-3% of total RNAs (Figure 1). Alternate robust methods for tagging RNA molecules with an RNA aptamer (e.g., MS2-, U1A- and biotin-RNA aptamer) and capturing the RNA-protein complex by the respective aptamer-specific partner are not extensively studied. Here, we describe a protocol (Figure 2) in which a biotin-RNA aptamer, referred to as the RNA mimic of biotin (RMB), was conjugated separately to two small RNA secondary structures that contribute to trafficking and translating HAC1 mRNA in the budding yeast Saccharomyces cerevisiae. The RMB-tagged RNA was expressed in yeast cells from a constitutive promoter. The biotinylated RNA bound to proteins was pulled down from the cell lysate by streptavidin agarose beads. RNA was detected by RT-PCR (Figure 3) and associated proteins by mass spectrometry (Figure 4). Our findings show that an RNA aptamer tag to RNA molecule is an effective method to explore the functional roles of RNA-protein networks in vivo.
Collapse
Affiliation(s)
- Jagadeesh K. Uppala
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, 3209 N Maryland Ave, WI-53211, USA
| | - Chandrima Ghosh
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, 3209 N Maryland Ave, WI-53211, USA
| | - Grzegorz Sabat
- Biotechnology Center, University of Wisconsin-Madison, Madison, Wisconsin, 53706, USA
| | - Madhusudan Dey
- Department of Biological Sciences, University of Wisconsin-Milwaukee, Milwaukee, 3209 N Maryland Ave, WI-53211, USA
| |
Collapse
|
46
|
Martens CR, Dorn LE, Kenney AD, Bansal SS, Yount JS, Accornero F. BEX1 is a critical determinant of viral myocarditis. PLoS Pathog 2022; 18:e1010342. [PMID: 35192678 PMCID: PMC8896894 DOI: 10.1371/journal.ppat.1010342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 03/04/2022] [Accepted: 02/04/2022] [Indexed: 11/18/2022] Open
Abstract
Viral infection of the heart is a common but underappreciated cause of heart failure. Viruses can cause direct cardiac damage by lysing infected cardiomyocytes. Inflammatory immune responses that limit viral replication can also indirectly cause damage during infection, making regulatory factors that fine-tune these responses particularly important. Identifying and understanding these factors that regulate cardiac immune responses during infection will be essential for developing targeted treatments for virus-associated heart failure. Our laboratory has discovered Brain Expressed X-linked protein 1 (BEX1) as a novel stress-regulated pro-inflammatory factor in the heart. Here we report that BEX1 plays a cardioprotective role in the heart during viral infection. Specifically, we adopted genetic gain- and loss-of-function strategies to modulate BEX1 expression in the heart in the context of coxsackievirus B3 (CVB3)-induced cardiomyopathy and found that BEX1 limits viral replication in cardiomyocytes. Interestingly, despite the greater viral load observed in mice lacking BEX1, inflammatory immune cell recruitment in the mouse heart was profoundly impaired in the absence of BEX1. Overall, the absence of BEX1 accelerated CVB3-driven heart failure and pathologic heart remodeling. This result suggests that limiting inflammatory cell recruitment has detrimental consequences for the heart during viral infections. Conversely, transgenic mice overexpressing BEX1 in cardiomyocytes revealed the efficacy of BEX1 for counteracting viral replication in the heart in vivo. We also found that BEX1 retains its antiviral role in isolated cells. Indeed, BEX1 was necessary and sufficient to counteract viral replication in both isolated primary cardiomyocytes and mouse embryonic fibroblasts suggesting a broader applicability of BEX1 as antiviral agent that extended to viruses other than CVB3, including Influenza A and Sendai virus. Mechanistically, BEX1 regulated interferon beta (IFN-β) expression in infected cells. Overall, our study suggests a multifaceted role of BEX1 in the cardiac antiviral immune response.
Collapse
Affiliation(s)
- Colton R. Martens
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Lisa E. Dorn
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Adam D. Kenney
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, United States of America
- Infectious Diseases Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Shyam S. Bansal
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Jacob S. Yount
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, United States of America
- Infectious Diseases Institute, The Ohio State University, Columbus, Ohio, United States of America
| | - Federica Accornero
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States of America
- Infectious Diseases Institute, The Ohio State University, Columbus, Ohio, United States of America
| |
Collapse
|
47
|
Cottonseed extracts regulate gene expression in human colon cancer cells. Sci Rep 2022; 12:1039. [PMID: 35058516 PMCID: PMC8776848 DOI: 10.1038/s41598-022-05030-3] [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: 06/01/2021] [Accepted: 12/16/2021] [Indexed: 11/18/2022] Open
Abstract
Cotton plant provides economically important fiber and cottonseed, but cottonseed contributes 20% of the crop value. Cottonseed value could be increased by providing high value bioactive compounds and polyphenolic extracts aimed at improving nutrition and preventing diseases because plant polyphenol extracts have been used as medicinal remedy for various diseases. The objective of this study was to investigate the effects of cottonseed extracts on cell viability and gene expression in human colon cancer cells. COLO 225 cells were treated with ethanol extracts from glanded and glandless cottonseed followed by MTT and qPCR assays. Cottonseed extracts showed minor effects on cell viability. qPCR assay analyzed 55 mRNAs involved in several pathways including DGAT, GLUT, TTP, IL, gossypol-regulated and TTP-mediated pathways. Using BCL2 mRNA as the internal reference, qPCR analysis showed minor effects of ethanol extracts from glanded seed coat and kernel and glandless seed coat on mRNA levels in the cells. However, glandless seed kernel extract significantly reduced mRNA levels of many genes involved in glucose transport, lipid biosynthesis and inflammation. The inhibitory effects of glandless kernel extract on gene expression may provide a useful opportunity for improving nutrition and healthcare associated with colon cancer. This in turn may provide the potential of increasing cottonseed value by using ethanol extract as a nutrition/health intervention agent.
Collapse
|
48
|
Natalia P, Zwirchmayr J, Rudžionytė I, Pulsinger A, Breuss JM, Uhrin P, Rollinger JM, de Martin R. Pterocarpus santalinus Selectively Inhibits a Subset of Pro-Inflammatory Genes in Interleukin-1 Stimulated Endothelial Cells. Front Pharmacol 2022; 12:802153. [PMID: 35115943 PMCID: PMC8804362 DOI: 10.3389/fphar.2021.802153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 12/22/2021] [Indexed: 11/13/2022] Open
Abstract
Based on the traditional use and scientific reports on the anti-inflammatory potential of red sandalwood, i.e., the heartwood of Pterocarpus santalinus L., we investigated its activity in a model of IL-1 stimulated endothelial cells. Endothelial cells were stimulated with IL-1 with or without prior incubation with a defined sandalwoodextract (PS), and analyzed for the expression of selected pro-inflammatory genes. The activity of NF-κB, a transcription factor of central importance for inflammatory gene expression was assessed by reporter gene analysis, Western blotting of IκBα, and nuclear translocation studies. In addition, microarray studies were performed followed by verification of selected genes by qPCR and supplemented by bioinformatics analysis. Our results show that PS is able to suppress the induction of E-selectin and VCAM-1, molecules that mediate key steps in the adhesion of leukocytes to the endothelium. It also suppressed the activity of an NF-κB reporter, IκBα phosphorylation and degradation, and the nuclear translocation of NF-κB RelA. In contrast, it stimulated JNK phosphorylation indicating the activation of the JNK signaling pathway. Gene expression profiling revealed that PS inhibits only a specific subset of IL-1 induced genes, while others remain unaffected. Most strongly suppressed genes were the signal transducer TRAF1 and the chemokine CX3CL1, whereas IL-8 was an example of a non-affected gene. Notably, PS also stimulated the expression of certain genes, including ones with negative regulatory function, e.g., members of the NR4A family, the mRNA destabilizing protein TTP as well as the transcription factors ATF3 and BHLHB40. These results provide mechanistic insight into the anti-inflammatory activity of PS, and suggest that it acts through the interplay of negative and positive regulators to achieve a differential inhibition of inflammatory gene expression.
Collapse
Affiliation(s)
- Priscilla Natalia
- Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Julia Zwirchmayr
- Department of Pharmaceutical Sciences, Division of Pharmacognosy, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Ieva Rudžionytė
- Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Alexandra Pulsinger
- Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Johannes M. Breuss
- Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Pavel Uhrin
- Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Judith M. Rollinger
- Department of Pharmaceutical Sciences, Division of Pharmacognosy, Faculty of Life Sciences, University of Vienna, Vienna, Austria
| | - Rainer de Martin
- Department of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
49
|
Aloufi N, Alluli A, Eidelman DH, Baglole CJ. Aberrant Post-Transcriptional Regulation of Protein Expression in the Development of Chronic Obstructive Pulmonary Disease. Int J Mol Sci 2021; 22:ijms222111963. [PMID: 34769392 PMCID: PMC8584689 DOI: 10.3390/ijms222111963] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 02/07/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is an incurable and prevalent respiratory disorder that is characterized by chronic inflammation and emphysema. COPD is primarily caused by cigarette smoke (CS). CS alters numerous cellular processes, including the post-transcriptional regulation of mRNAs. The identification of RNA-binding proteins (RBPs), microRNAs (miRNAs), and long non-coding RNAs (lncRNAs) as main factors engaged in the regulation of RNA biology opens the door to understanding their role in coordinating physiological cellular processes. Dysregulation of post-transcriptional regulation by foreign particles in CS may lead to the development of diseases such as COPD. Here we review current knowledge about post-transcriptional events that may be involved in the pathogenesis of COPD.
Collapse
Affiliation(s)
- Noof Aloufi
- Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada; (N.A.); (A.A.)
- Department of Medical Laboratory Technology, Applied Medical Science, Taibah University, Universities Road, Medina P.O. Box 344, Saudi Arabia
| | - Aeshah Alluli
- Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada; (N.A.); (A.A.)
| | - David H. Eidelman
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada;
| | - Carolyn J. Baglole
- Department of Pathology, McGill University, Montreal, QC H3A 2B4, Canada; (N.A.); (A.A.)
- Department of Medicine, McGill University, Montreal, QC H4A 3J1, Canada;
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC H3G 1Y6, Canada
- Correspondence:
| |
Collapse
|
50
|
Ok K, Filipovic MR, Michel SLJ. Targeting Zinc Finger Proteins with Exogenous Metals and Molecules: Lessons learned from Tristetraprolin, a CCCH type Zinc Finger. Eur J Inorg Chem 2021; 2021:3795-3805. [PMID: 34867080 PMCID: PMC8635303 DOI: 10.1002/ejic.202100402] [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: 05/10/2021] [Indexed: 11/09/2022]
Abstract
ZF proteins are ubiquitous eukaryotic proteins that play important roles in gene regulation. ZFs contain small domains made up of a combination of four cysteine and histidine residues, and are classified based up on the identity of these residues and their spacing. One emerging class of ZFs are the Cys3His (or CCCH) class of ZFs. These ZFs play key roles in regulating RNA. In this minireview, an overview of the CCCH class of ZFs, with a focus on tristetraprolin (TTP) is provided. TTP regulates inflammation by controlling cytokine mRNAs, and there is an interest in modulating TTP activity to control inflammation. Two methods to control TTP activity are to target with exogenous metals (a 'metals in medicine' approach) or to target with endogenous signaling molecules. Work that has been done to target TTP with Fe, Cu, Cd and Au as well as with H2S is reviewed. This includes attention to new methods that have been developed to monitor metal exchange with the spectroscopically silent ZnII including native electro-spray ionization mass spectrometry (ESI-MS), spin-filter inductively coupled plasma mass spectrometry (ICP-MS) and cryo-electro-spray mass spectrometry (CSI-MS); along with fluorescence anisotropy (FA) to follow RNA binding.
Collapse
Affiliation(s)
- Kiwon Ok
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| | - Milos R Filipovic
- Leibniz-Institut für Analytische, Wissenschaften-ISAS-e.V., 44227 Dortmund, Germany
| | - Sarah L J Michel
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, MD 21201, USA
| |
Collapse
|