1
|
Saraiva HFRDA, Sangalli JR, Alves L, da Silveira JC, Meirelles FV, Perecin F. NPPC and AREG supplementation in IVM systems alter mRNA translation and decay programs-related gene expression in bovine COC. Anim Reprod 2024; 21:e20230101. [PMID: 39021501 PMCID: PMC11253787 DOI: 10.1590/1984-3143-ar2023-0101] [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: 06/20/2023] [Accepted: 04/29/2024] [Indexed: 07/20/2024] Open
Abstract
During oocyte meiosis resumption, a coordinated program of transcript translation and decay machinery promotes a remodeling of mRNA stores, which determines the success of the acquisition of competence and early embryo development. We investigated levels of two genes related to mRNA translation (CPEB1 and CPEB4) and two related to mRNA degradation (CNOT7 and ZFP36L2) machinery and found ZFP36L2 downregulated in in vitro-matured bovine oocytes compared to in vivo counterparts. Thereafter, we tested the effects of a pre-IVM step with NPPC and a modified IVM with AREG on the modulation of members of mRNA translation and degradation pathways in cumulus cells and oocytes. Our data showed a massive upregulation of genes associated with translational and decay processes in cumulus cells, promoted by NPPC and AREG supplementation, up to 9h of IVM. The oocytes were less affected by NPPC and AREG, and even though ZFP36L2 transcript and protein levels were downregulated at 9 and 19h of IVM, only one (KDM4C) from the ten target genes evaluated was differently expressed in these treatments. These data suggest that cumulus cells are more prone to respond to NPPC and AREG supplementation in vitro, regarding translational and mRNA decay programs. Given the important nursing role of these cells, further studies could contribute to a better understanding of the impact of these modulators in maternal mRNA modulation and improve IVM outcomes.
Collapse
Affiliation(s)
| | - Juliano Rodrigues Sangalli
- Faculdade de Zootecnia e Engenharia de Alimentos, Departamento de Medicina Veterinária, Universidade de São Paulo, Pirassununga, SP, Brasil
| | - Luana Alves
- Faculdade de Zootecnia e Engenharia de Alimentos, Departamento de Medicina Veterinária, Universidade de São Paulo, Pirassununga, SP, Brasil
| | - Juliano Coelho da Silveira
- Faculdade de Zootecnia e Engenharia de Alimentos, Departamento de Medicina Veterinária, Universidade de São Paulo, Pirassununga, SP, Brasil
| | - Flávio Vieira Meirelles
- Faculdade de Zootecnia e Engenharia de Alimentos, Departamento de Medicina Veterinária, Universidade de São Paulo, Pirassununga, SP, Brasil
| | - Felipe Perecin
- Faculdade de Zootecnia e Engenharia de Alimentos, Departamento de Medicina Veterinária, Universidade de São Paulo, Pirassununga, SP, Brasil
| |
Collapse
|
2
|
Nishisaka H, Tomohiro T, Fukuzumi K, Fukao A, Funakami Y, Fujiwara T. Deciphering the Akt1-HuD interaction in HuD-mediated neuronal differentiation. Biochimie 2024; 221:20-26. [PMID: 38244852 DOI: 10.1016/j.biochi.2024.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 01/17/2024] [Accepted: 01/17/2024] [Indexed: 01/22/2024]
Abstract
The RNA-binding protein HuD/ELAVL4 is essential for neuronal development and synaptic plasticity by governing various post-transcriptional processes of target mRNAs, including stability, translation, and localization. We previously showed that the linker region and poly(A)-binding domain of HuD play a pivotal role in promoting translation and inducing neurite outgrowth. In addition, we found that HuD interacts exclusively with the active form of Akt1, through the linker region. Although this interaction is essential for neurite outgrowth, HuD is not a substrate for Akt1, raising questions about the dynamics between HuD-mediated translational stimulation and its association with active Akt1. Here, we demonstrate that active Akt1 interacts with the cap-binding complex via HuD. We identify key amino acids in linker region of HuD responsible for Akt1 interaction, leading to the generation of two point-mutated HuD variants: one that is incapable of binding to Akt1 and another that can interact with Akt1 regardless of its phosphorylation status. In vitro translation assays using these mutants reveal that HuD-mediated translation stimulation is independent of its binding to Akt1. In addition, it is evident that the interaction between HuD and active Akt1 is essential for HuD-induced neurite outgrowth, whereas a HuD mutant capable of binding to any form of Akt1 leads to aberrant neurite development. Collectively, our results revisit the understanding of the HuD-Akt1 interaction in translation and suggest that this interaction contributes to HuD-mediated neurite outgrowth via a unique molecular mechanism distinct from translation regulation.
Collapse
Affiliation(s)
| | - Takumi Tomohiro
- Faculty of Pharmacy, Kindai University, Higashi-Osaka, Japan
| | - Kako Fukuzumi
- Faculty of Pharmacy, Kindai University, Higashi-Osaka, Japan
| | - Akira Fukao
- Faculty of Pharmacy, Kindai University, Higashi-Osaka, Japan
| | | | | |
Collapse
|
3
|
Magg V, Manetto A, Kopp K, Wu CC, Naghizadeh M, Lindner D, Eke L, Welsch J, Kallenberger SM, Schott J, Haucke V, Locker N, Stoecklin G, Ruggieri A. Turnover of PPP1R15A mRNA encoding GADD34 controls responsiveness and adaptation to cellular stress. Cell Rep 2024; 43:114069. [PMID: 38602876 DOI: 10.1016/j.celrep.2024.114069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 01/25/2024] [Accepted: 03/21/2024] [Indexed: 04/13/2024] Open
Abstract
The integrated stress response (ISR) is a key cellular signaling pathway activated by environmental alterations that represses protein synthesis to restore homeostasis. To prevent sustained damage, the ISR is counteracted by the upregulation of growth arrest and DNA damage-inducible 34 (GADD34), a stress-induced regulatory subunit of protein phosphatase 1 that mediates translation reactivation and stress recovery. Here, we uncover a novel ISR regulatory mechanism that post-transcriptionally controls the stability of PPP1R15A mRNA encoding GADD34. We establish that the 3' untranslated region of PPP1R15A mRNA contains an active AU-rich element (ARE) recognized by proteins of the ZFP36 family, promoting its rapid decay under normal conditions and stabilization for efficient expression of GADD34 in response to stress. We identify the tight temporal control of PPP1R15A mRNA turnover as a component of the transient ISR memory, which sets the threshold for cellular responsiveness and mediates adaptation to repeated stress conditions.
Collapse
Affiliation(s)
- Vera Magg
- Heidelberg University, Medical Faculty Heidelberg, Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research, 69120 Heidelberg, Germany
| | - Alessandro Manetto
- Heidelberg University, Medical Faculty Heidelberg, Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research, 69120 Heidelberg, Germany
| | - Katja Kopp
- Heidelberg University, Medical Faculty Heidelberg, Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research, 69120 Heidelberg, Germany
| | - Chia Ching Wu
- Heidelberg University, Medical Faculty Heidelberg, Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research, 69120 Heidelberg, Germany
| | - Mohsen Naghizadeh
- Heidelberg University, Medical Faculty Mannheim, Division of Biochemistry, Mannheim Institute for Innate Immunoscience (MI3) and Mannheim Cancer Center (MCC), 68167 Mannheim, Germany
| | - Doris Lindner
- Heidelberg University, Medical Faculty Mannheim, Division of Biochemistry, Mannheim Institute for Innate Immunoscience (MI3) and Mannheim Cancer Center (MCC), 68167 Mannheim, Germany
| | - Lucy Eke
- Faculty of Health and Medical Sciences, School of Biosciences and Medicine, University of Surrey, Guildford, UK
| | - Julia Welsch
- Heidelberg University, Medical Faculty Heidelberg, Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research, 69120 Heidelberg, Germany
| | - Stefan M Kallenberger
- Digital Health Center, Berlin Institute of Health (BIH) and Charité, 10178 Berlin, Germany; Medical Oncology, National Center for Tumor Diseases, Heidelberg University, 69120 Heidelberg, Germany
| | - Johanna Schott
- Heidelberg University, Medical Faculty Mannheim, Division of Biochemistry, Mannheim Institute for Innate Immunoscience (MI3) and Mannheim Cancer Center (MCC), 68167 Mannheim, Germany
| | - Volker Haucke
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, 13125 Berlin, Germany; Freie Universität Berlin, Faculty of Biology, Chemistry, and Pharmacy, 14195 Berlin, Germany
| | - Nicolas Locker
- Faculty of Health and Medical Sciences, School of Biosciences and Medicine, University of Surrey, Guildford, UK; The Pirbright Institute, GU24 0NF Pirbright, UK
| | - Georg Stoecklin
- Heidelberg University, Medical Faculty Mannheim, Division of Biochemistry, Mannheim Institute for Innate Immunoscience (MI3) and Mannheim Cancer Center (MCC), 68167 Mannheim, Germany; Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany.
| | - Alessia Ruggieri
- Heidelberg University, Medical Faculty Heidelberg, Department of Infectious Diseases, Molecular Virology, Center for Integrative Infectious Disease Research, 69120 Heidelberg, Germany.
| |
Collapse
|
4
|
Tran QTN, Gan PXL, Liao W, Mok YK, Chai CLL, Wong WSF. Degradation of MK2 with natural compound andrographolide: A new modality for anti-inflammatory therapy. Pharmacol Res 2023; 194:106861. [PMID: 37480973 DOI: 10.1016/j.phrs.2023.106861] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/26/2023] [Accepted: 07/18/2023] [Indexed: 07/24/2023]
Abstract
The p38MAPK-MK2 signaling axis functions as an initiator of inflammation. Targeting the p38MAPK-MK2 signaling axis represents a direct therapeutic intervention of inflammatory diseases. We described here a novel role of andrographolide (AG), a small-molecule ent-labdane natural compound, as an inhibitor of p38MAPK-MK2 axis via MK2 degradation. AG was found to bind to the activation loop of MK2, located at the interface of the p38MAPK-MK2 biomolecular complex. This interaction disrupted the complex formation and predisposed MK2 to proteasome-mediated degradation. We showed that AG induced MK2 degradation in a concentration- and time-dependent manner and exerted its anti-inflammatory effects by enhancing the mRNA-destabilizing activity of tristetraprolin, thereby inhibiting pro-inflammatory mediator production (e.g., TNF-α, MCP-1). Administration of AG via intratracheal (i.t.) route to mice induced MK2 downregulation in lung alveolar macrophages, but not lung tissues, and prevented macrophage activation. Our study also demonstrated that the anti-inflammatory effects achieved by AG via MK2 degradation were more durable and sustained than that achieved by the conventional MK2 kinase inhibitors (e.g., PF-3644022). Taken together, our findings illustrated a novel mode of action of AG by modulating the p38MAPK-MK2 signaling axis and would pave the way for the development of a novel class of anti-inflammatory agents targeting MK2 for degradation by harnessing the privileged scaffold of AG.
Collapse
Affiliation(s)
- Quy T N Tran
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, 117600, Singapore; Department of Pharmacy, Faculty of Science, National University of Singapore, 117543, Singapore; Drug Discovery and Optimization Platform (DDOP), Yong Loo Lin School of Medicine, National University Health System, 117600, Singapore
| | - Phyllis X L Gan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, 117600, Singapore
| | - Wupeng Liao
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, 117600, Singapore; Singapore-HUJ Alliance for Research and Enterprise (SHARE), National University of Singapore, Singapore
| | - Yu Keung Mok
- Department of Biological Sciences, Faculty of Science, National University of Singapore, 117543, Singapore
| | - Christina L L Chai
- Department of Pharmacy, Faculty of Science, National University of Singapore, 117543, Singapore; Drug Discovery and Optimization Platform (DDOP), Yong Loo Lin School of Medicine, National University Health System, 117600, Singapore.
| | - W S Fred Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, 117600, Singapore; Drug Discovery and Optimization Platform (DDOP), Yong Loo Lin School of Medicine, National University Health System, 117600, Singapore; Singapore-HUJ Alliance for Research and Enterprise (SHARE), National University of Singapore, Singapore.
| |
Collapse
|
5
|
Zhang J, Ge P, Liu J, Luo Y, Guo H, Zhang G, Xu C, Chen H. Glucocorticoid Treatment in Acute Respiratory Distress Syndrome: An Overview on Mechanistic Insights and Clinical Benefit. Int J Mol Sci 2023; 24:12138. [PMID: 37569514 PMCID: PMC10418884 DOI: 10.3390/ijms241512138] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/20/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Acute lung injury/acute respiratory distress syndrome (ALI/ARDS), triggered by various pathogenic factors inside and outside the lungs, leads to diffuse lung injury and can result in respiratory failure and death, which are typical clinical critical emergencies. Severe acute pancreatitis (SAP), which has a poor clinical prognosis, is one of the most common diseases that induces ARDS. When SAP causes the body to produce a storm of inflammatory factors and even causes sepsis, clinicians will face a two-way choice between anti-inflammatory and anti-infection objectives while considering the damaged intestinal barrier and respiratory failure, which undoubtedly increases the difficulty of the diagnosis and treatment of SAP-ALI/ARDS. For a long time, many studies have been devoted to applying glucocorticoids (GCs) to control the inflammatory response and prevent and treat sepsis and ALI/ARDS. However, the specific mechanism is not precise, the clinical efficacy is uneven, and the corresponding side effects are endless. This review discusses the mechanism of action, current clinical application status, effectiveness assessment, and side effects of GCs in the treatment of ALI/ARDS (especially the subtype caused by SAP).
Collapse
Affiliation(s)
- Jinquan Zhang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
| | - Peng Ge
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Jie Liu
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yalan Luo
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Haoya Guo
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Guixin Zhang
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Caiming Xu
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
- Department of Molecular Diagnostics and Experimental Therapeutics, Beckman Research Institute of City of Hope, Biomedical Research Center, Comprehensive Cancer Center, Monrovia, CA 91016, USA
| | - Hailong Chen
- Department of General Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian 116044, China
- Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| |
Collapse
|
6
|
Li Y, Chen B, Jiang X, Li Y, Wang X, Huang S, Wu X, Xiao Y, Shi D, Huang X, He L, Chen X, Ouyang Y, Li J, Song L, Lin C. A Wnt-induced lncRNA-DGCR5 splicing switch drives tumor-promoting inflammation in esophageal squamous cell carcinoma. Cell Rep 2023; 42:112542. [PMID: 37210725 DOI: 10.1016/j.celrep.2023.112542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 04/04/2023] [Accepted: 05/03/2023] [Indexed: 05/23/2023] Open
Abstract
Alternative splicing (AS) is a critical mechanism for the aberrant biogenesis of long non-coding RNA (lncRNA). Although the role of Wnt signaling in AS has been implicated, it remains unclear how it mediates lncRNA splicing during cancer progression. Herein, we identify that Wnt3a induces a splicing switch of lncRNA-DGCR5 to generate a short variant (DGCR5-S) that correlates with poor prognosis in esophageal squamous cell carcinoma (ESCC). Upon Wnt3a stimulation, active nuclear β-catenin acts as a co-factor of FUS to facilitate the spliceosome assembly and the generation of DGCR5-S. DGCR5-S inhibits TTP's anti-inflammatory activity by protecting it from PP2A-mediated dephosphorylation, thus fostering tumor-promoting inflammation. Importantly, synthetic splice-switching oligonucleotides (SSOs) disrupt the splicing switch of DGCR5 and potently suppress ESCC tumor growth. These findings uncover the mechanism for Wnt signaling in lncRNA splicing and suggest that the DGCR5 splicing switch may be a targetable vulnerability in ESCC.
Collapse
Affiliation(s)
- Yue Li
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Boyu Chen
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xingyu Jiang
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yudong Li
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xin Wang
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Shumei Huang
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China
| | - Xuxia Wu
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yunyun Xiao
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Dongni Shi
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xinjian Huang
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Lixin He
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xiangfu Chen
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Ying Ouyang
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Jun Li
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510080, China.
| | - Libing Song
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China; Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Guangzhou Institute of Oncology, Tumor Hospital, Guangzhou Medical University, Guangzhou 511436, China.
| | - Chuyong Lin
- Department of Experimental Research, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou 510060, China.
| |
Collapse
|
7
|
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
|
8
|
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: 6] [Impact Index Per Article: 3.0] [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
|
9
|
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
|
10
|
Kwak Y, Daly CWP, Fogarty EA, Grimson A, Kwak H. Dynamic and widespread control of poly(A) tail length during macrophage activation. RNA (NEW YORK, N.Y.) 2022; 28:947-971. [PMID: 35512831 PMCID: PMC9202586 DOI: 10.1261/rna.078918.121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 03/21/2022] [Indexed: 06/14/2023]
Abstract
The poly(A) tail enhances translation and transcript stability, and tail length is under dynamic control during cell state transitions. Tail regulation plays essential roles in translational timing and fertilization in early development, but poly(A) tail dynamics have not been fully explored in post-embryonic systems. Here, we examined the landscape and impact of tail length control during macrophage activation. Upon activation, more than 1500 mRNAs, including proinflammatory genes, underwent distinctive changes in tail lengths. Increases in tail length correlated with mRNA levels regardless of transcriptional activity, and many mRNAs that underwent tail extension encode proteins necessary for immune function and post-transcriptional regulation. Strikingly, we found that ZFP36, whose protein product destabilizes target transcripts, undergoes tail extension. Our analyses indicate that many mRNAs undergoing tail lengthening are, in turn, degraded by elevated levels of ZFP36, constituting a post-transcriptional feedback loop that ensures transient regulation of transcripts integral to macrophage activation. Taken together, this study establishes the complexity, relevance, and widespread nature of poly(A) tail dynamics, and the resulting post-transcriptional regulation during macrophage activation.
Collapse
Affiliation(s)
- Yeonui Kwak
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853, USA
- Graduate Field of Genetics, Genomics, and Development, Cornell University, Ithaca, New York 14853, USA
| | - Ciarán W P Daly
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853, USA
- Graduate Field of Biochemistry, Molecular, and Cell Biology, Cornell University, Ithaca, New York 14853, USA
| | - Elizabeth A Fogarty
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853, USA
| | - Andrew Grimson
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853, USA
| | - Hojoong Kwak
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, New York 14853, USA
| |
Collapse
|
11
|
Solone XKV, Caldara AL, Wells B, Qiao H, Wade LR, Salerno JC, Helms KA, Smith KER, McMurry JL, Chrestensen CA. MAP kinases differentially bind and phosphorylate NOS3 via two unique NOS3 sites. FEBS Open Bio 2022; 12:1075-1086. [PMID: 35182051 PMCID: PMC9063426 DOI: 10.1002/2211-5463.13384] [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: 10/15/2021] [Revised: 01/12/2022] [Accepted: 02/17/2022] [Indexed: 11/25/2022] Open
Abstract
Nitric oxide synthase 3 (NOS3) is a major vasoprotective enzyme that catalyzes the conversion of l-arginine to nitric oxide (NO) in response to a significant number of signaling pathways. Here, we provide evidence that NOS3 interactions with MAP kinases have physiological relevance. Binding interactions of NOS3 with c-Jun N-terminal kinase (JNK1α1 ), p38α, and ERK2 were characterized using optical biosensing with full-length NOS3 and NOS3 specific peptides and phosphopeptides. Like p38α and ERK2, JNK1α1 exhibited high-affinity binding to full-length NOS3 (KD 15 nm). Rate constants exhibited fast-on, slow-off binding (kon = 4106 m-1 s-1 ; koff = 6.2 × 10-5 s-1 ). Further analysis using synthetic NOS3 peptides revealed two MAP kinase binding sites unique to NOS3. p38α evinced similar affinity with both NOS3 binding sites. For ERK2 and JNK1α1, the affinity at the two sites differed. However, NOS3 peptides with a phosphate at either S114 or S633 did not meaningfully interact with the kinases. Immunoblotting revealed that each kinase phosphorylated NOS3 with a unique pattern. JNK1α1 predominantly phosphorylated NOS3 at S114, ERK2 at S600, and p38α phosphorylated both residues. In vitro production of NO was unchanged by phosphorylation at these sites. In human microvascular endothelial cells, endogenous interactions of all the MAP kinases with NOS3 were captured using proximity ligation assay in resting cells. Our results underscore the importance of MAP kinase interactions, identifying two unique NOS3 interaction sites with potential for modulation by MAP kinase phosphorylation (S114) and other signaling inputs, like protein kinase A (S633).
Collapse
Affiliation(s)
- Xzaviar K. V. Solone
- Department of Molecular & Cellular BiologyKennesaw State UniversityGAUSA
- Present address:
Department of Molecular Genetics and MicrobiologyUniversity of FloridaGainesvilleFLUSA
| | - Amber L. Caldara
- Department of Molecular & Cellular BiologyKennesaw State UniversityGAUSA
| | - Brady Wells
- Department of Chemistry & BiochemistryKennesaw State UniversityGAUSA
| | - Hao Qiao
- Department of Chemistry & BiochemistryKennesaw State UniversityGAUSA
| | - Lydia R. Wade
- Department of Chemistry & BiochemistryKennesaw State UniversityGAUSA
| | - John C. Salerno
- Department of Molecular & Cellular BiologyKennesaw State UniversityGAUSA
| | - Katy A. Helms
- Department of Molecular & Cellular BiologyKennesaw State UniversityGAUSA
- Present address:
Wake Forest Medical CenterWinston‐SalemNCUSA
| | | | | | | |
Collapse
|
12
|
Abstract
Mitogen-activated protein kinase (MAPK)-activated protein kinases (MAPKAPKs) are defined by their exclusive activation by MAPKs. They can be activated by classical and atypical MAPKs that have been stimulated by mitogens and various stresses. Genetic deletions of MAPKAPKs and availability of highly specific small-molecule inhibitors have continuously increased our functional understanding of these kinases. MAPKAPKs cooperate in the regulation of gene expression at the level of transcription; RNA processing, export, and stability; and protein synthesis. The diversity of stimuli for MAPK activation, the cross talk between the different MAPKs and MAPKAPKs, and the specific substrate pattern of MAPKAPKs orchestrate immediate-early and inflammatory responses in space and time and ensure proper control of cell growth, differentiation, and cell behavior. Hence, MAPKAPKs are promising targets for cancer therapy and treatments for conditions of acute and chronic inflammation, such as cytokine storms and rheumatoid arthritis. Expected final online publication date for the Annual Review of Biochemistry, Volume 91 is June 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Natalia Ronkina
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany;
| | - Matthias Gaestel
- Institute of Cell Biochemistry, Hannover Medical School, Hannover, Germany;
| |
Collapse
|
13
|
Suzuki T, Hoshina M, Nishijima S, Hoshina N, Kikuguchi C, Tomohiro T, Fukao A, Fujiwara T, Yamamoto T. Regulation of CCR4-NOT complex deadenylase activity and cellular responses by MK2-dependent phosphorylation of CNOT2. RNA Biol 2022; 19:234-246. [PMID: 35129087 PMCID: PMC8820811 DOI: 10.1080/15476286.2021.2021676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
CCR4-NOT complex-mediated mRNA deadenylation serves critical functions in multiple biological processes, yet how this activity is regulated is not fully understood. Here, we show that osmotic stress induces MAPKAPK-2 (MK2)-mediated phosphorylation of CNOT2. Programmed cell death is greatly enhanced by osmotic stress in CNOT2-depleted cells, indicating that CNOT2 is responsible for stress resistance of cells. Although wild-type (WT) and non-phosphorylatable CNOT2 mutants reverse this sensitivity, a phosphomimetic form of CNOT2, in which serine at the phosphorylation site is replaced with glutamate, does not have this function. We also show that mRNAs have elongated poly(A) tails in CNOT2-depleted cells and that introduction of CNOT2 WT or a non-phosphorylatable mutant, but not phosphomimetic CNOT2, renders their poly(A) tail lengths comparable to those in control HeLa cells. Consistent with this, the CCR4-NOT complex containing phosphomimetic CNOT2 exhibits less deadenylase activity than that containing CNOT2 WT. These data suggest that CCR4-NOT complex deadenylase activity is regulated by post-translational modification, yielding dynamic control of mRNA deadenylation.
Collapse
Affiliation(s)
- Toru Suzuki
- Laboratory for Immunogenetics, Center for Integrative Medical Sciences, Riken, Yokohama, Japan
| | - Miyuki Hoshina
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Japan
| | - Saori Nishijima
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Japan
| | - Naosuke Hoshina
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Japan
| | - Chisato Kikuguchi
- Laboratory for Immunogenetics, Center for Integrative Medical Sciences, Riken, Yokohama, Japan
| | - Takumi Tomohiro
- Laboratory of Biochemistry, Kindai University, Higashi-Osaka, Japan
| | - Akira Fukao
- Laboratory of Biochemistry, Kindai University, Higashi-Osaka, Japan
| | | | - Tadashi Yamamoto
- Cell Signal Unit, Okinawa Institute of Science and Technology Graduate University, Onna, Japan
| |
Collapse
|
14
|
Morgan D, Berggren KL, Spiess CD, Smith HM, Tejwani A, Weir SJ, Lominska CE, Thomas SM, Gan GN. Mitogen-activated protein kinase-activated protein kinase-2 (MK2) and its role in cell survival, inflammatory signaling, and migration in promoting cancer. Mol Carcinog 2021; 61:173-199. [PMID: 34559922 DOI: 10.1002/mc.23348] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 08/25/2021] [Accepted: 08/27/2021] [Indexed: 12/19/2022]
Abstract
Cancer and the immune system share an intimate relationship. Chronic inflammation increases the risk of cancer occurrence and can also drive inflammatory mediators into the tumor microenvironment enhancing tumor growth and survival. The p38 MAPK pathway is activated both acutely and chronically by stress, inflammatory chemokines, chronic inflammatory conditions, and cancer. These properties have led to extensive efforts to find effective drugs targeting p38, which have been unsuccessful. The immediate downstream serine/threonine kinase and substrate of p38 MAPK, mitogen-activated-protein-kinase-activated-protein-kinase-2 (MK2) protects cells against stressors by regulating the DNA damage response, transcription, protein and messenger RNA stability, and motility. The phosphorylation of downstream substrates by MK2 increases inflammatory cytokine production, drives an immune response, and contributes to wound healing. By binding directly to p38 MAPK, MK2 is responsible for the export of p38 MAPK from the nucleus which gives MK2 properties that make it unique among the large number of p38 MAPK substrates. Many of the substrates of both p38 MAPK and MK2 are separated between the cytosol and nucleus and interfering with MK2 and altering this intracellular translocation has implications for the actions of both p38 MAPK and MK2. The inhibition of MK2 has shown promise in combination with both chemotherapy and radiotherapy as a method for controlling cancer growth and metastasis in a variety of cancers. Whereas the current data are encouraging the field requires the development of selective and well tolerated drugs to target MK2 and a better understanding of its effects for effective clinical use.
Collapse
Affiliation(s)
- Deri Morgan
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Kiersten L Berggren
- Department of Internal Medicine, Division of Medical Oncology, Section of Radiation Oncology, UNM School of Medicine, The University of New Mexico, Albuquerque, New Mexico, USA
| | - Colby D Spiess
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Hannah M Smith
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Ajay Tejwani
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Scott J Weir
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Christopher E Lominska
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Sufi M Thomas
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, USA.,Department of Otolaryngology, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Gregory N Gan
- Department of Radiation Oncology, University of Kansas Medical Center, Kansas City, Kansas, USA.,Department of Cancer Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
15
|
Al-Matouq J, Al-Haj L, Al-Saif M, Khabar KSA. Post-transcriptional screen of cancer amplified genes identifies ERBB2/Her2 signaling as AU-rich mRNA stability-promoting pathway. Oncogenesis 2021; 10:61. [PMID: 34535639 PMCID: PMC8448767 DOI: 10.1038/s41389-021-00351-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/19/2021] [Indexed: 02/08/2023] Open
Abstract
Amplification of specific cancer genes leads to their over-expression contributing to tumor growth, spread, and drug resistance. Little is known about the ability of these amplified oncogenes to augment the expression of cancer genes through post-transcriptional control. The AU-rich elements (ARE)-mediated mRNA decay is compromised for many key cancer genes leading to their increased abundance and effects. Here, we performed a post-transcriptional screen for frequently amplified cancer genes demonstrating that ERBB2/Her2 overexpression was able to augment the post-transcriptional effects. The ERBB1/2 inhibitor, lapatinib, led to the reversal of the aberrant ARE-mediated process in ERBB2-amplified breast cancer cells. The intersection of overexpressed genes associated with ERBB2 amplification in TCGA datasets with ARE database (ARED) identified ERBB2-associated gene cluster. Many of these genes were over-expressed in the ERBB2-positive SKBR3 cells compared to MCF10A normal-like cells, and were under-expressed due to ERBB2 siRNA treatment. Lapatinib accelerated the ARE-mRNA decay for several ERBB2-regulated genes. The ERBB2 inhibitor decreased both the abundance and stability of the phosphorylated inactive form of the mRNA decay-promoting protein, tristetraprolin (ZFP36/TTP). The ERBB2 siRNA was also able to reduce the phosphorylated ZFP36/TTP form. In contrast, ectopic expression of ERBB2 in MCF10A or HEK293 cells led to increased abundance of the phosphorylated ZFP36/TTP. The effect of ERBB2 on TTP phosphorylation appeared to be mediated via the MAPK-MK2 pathway. Screening for the impact of other amplified cancer genes in HEK293 cells also demonstrated that EGFR, AKT2, CCND1, CCNE1, SKP2, and FGFR3 caused both increased abundance of phosphorylated ZFP36/TTP and ARE-post-transcriptional reporter activity. Thus, specific amplified oncogenes dysregulate post-transcriptional ARE-mediated effects, and targeting the ARE-mediated pathway itself may provide alternative therapeutic approaches.
Collapse
Affiliation(s)
- Jenan Al-Matouq
- grid.415310.20000 0001 2191 4301Molecular BioMedicine Program, King Faisal Specialist Hospital and Research Centre, Riyadh, 11211 Saudi Arabia ,Present Address: Mohammed Al-Mana College for Medical Science, Dammam, Saudi Arabia
| | - Latifa Al-Haj
- grid.415310.20000 0001 2191 4301Molecular BioMedicine Program, King Faisal Specialist Hospital and Research Centre, Riyadh, 11211 Saudi Arabia
| | - Maher Al-Saif
- grid.415310.20000 0001 2191 4301Molecular BioMedicine Program, King Faisal Specialist Hospital and Research Centre, Riyadh, 11211 Saudi Arabia
| | - Khalid S. A. Khabar
- grid.415310.20000 0001 2191 4301Molecular BioMedicine Program, King Faisal Specialist Hospital and Research Centre, Riyadh, 11211 Saudi Arabia
| |
Collapse
|
16
|
Al‐Qahtani QH, Moghrabi WN, Al‐Yahya S, Al‐Haj L, Al‐Saif M, Mahmoud L, Al‐Mohanna F, Al‐Souhibani N, Alaiya A, Hitti E, Khabar KSA. Kinome inhibition reveals a role for polo-like kinase 1 in targeting post-transcriptional control in cancer. Mol Oncol 2021; 15:2120-2139. [PMID: 33411958 PMCID: PMC8334256 DOI: 10.1002/1878-0261.12897] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 12/09/2020] [Accepted: 01/05/2021] [Indexed: 12/15/2022] Open
Abstract
Dysfunctions in post-transcriptional control are observed in cancer and chronic inflammatory diseases. Here, we employed a kinome inhibitor library (n = 378) in a reporter system selective for 3'-untranslated region-AU-rich elements (ARE). Fifteen inhibitors reduced the ARE-reporter activity; among the targets is the polo-like kinase 1 (PLK1). RNA-seq experiments demonstrated that the PLK1 inhibitor, volasertib, reduces the expression of cytokine and cell growth ARE mRNAs. PLK1 inhibition caused accelerated mRNA decay in cancer cells and was associated with reduced phosphorylation and stability of the mRNA decay-promoting protein, tristetraprolin (ZFP36/TTP). Ectopic expression of PLK1 increased abundance and stability of high molecular weight of ZFP36/TTP likely of the phosphorylated form. PLK1 effect was associated with the MAPK-MK2 pathway, a major regulator of ARE-mRNA stability, as evident from MK2 inhibition, in vitro phosphorylation, and knockout experiments. Mutational analysis demonstrates that TTP serine 186 is a target for PLK1 effect. Treatment of mice with the PLK1 inhibitor reduced both ZFP36/TTP phosphorylation in xenograft tumor tissues, and the tumor size. In cancer patients' tissues, PLK1/ARE-regulated gene cluster was overexpressed in solid tumors and associated with poor survival. The data showed that PLK1-mediated post-transcriptional aberration could be a therapeutic target.
Collapse
Affiliation(s)
- Qamraa H. Al‐Qahtani
- Molecular BioMedicine ProgramFaisal Specialist Hospital and Research CentreRiyadhKingSaudi Arabia
- Present address:
Department of Pharmacology and ToxicologyCollege of PharmacyKing Saud UniversityRiyadh11495Saudi Arabia
| | - Walid N. Moghrabi
- Molecular BioMedicine ProgramFaisal Specialist Hospital and Research CentreRiyadhKingSaudi Arabia
| | - Suhad Al‐Yahya
- Molecular BioMedicine ProgramFaisal Specialist Hospital and Research CentreRiyadhKingSaudi Arabia
| | - Latifa Al‐Haj
- Molecular BioMedicine ProgramFaisal Specialist Hospital and Research CentreRiyadhKingSaudi Arabia
| | - Maher Al‐Saif
- Molecular BioMedicine ProgramFaisal Specialist Hospital and Research CentreRiyadhKingSaudi Arabia
| | - Linah Mahmoud
- Molecular BioMedicine ProgramFaisal Specialist Hospital and Research CentreRiyadhKingSaudi Arabia
| | - Falah Al‐Mohanna
- Department of Comparative MedicineKing Faisal Specialist Hospital and Research CentreRiyadhSaudi Arabia
| | - Norah Al‐Souhibani
- Molecular BioMedicine ProgramFaisal Specialist Hospital and Research CentreRiyadhKingSaudi Arabia
| | - Ayodele Alaiya
- Stem Cell and Tissue Engineering ProgramFaisal Specialist Hospital and Research CentreRiyadhKingSaudi Arabia
| | - Edward Hitti
- Molecular BioMedicine ProgramFaisal Specialist Hospital and Research CentreRiyadhKingSaudi Arabia
| | - Khalid S. A. Khabar
- Molecular BioMedicine ProgramFaisal Specialist Hospital and Research CentreRiyadhKingSaudi Arabia
| |
Collapse
|
17
|
Makita S, Takatori H, Nakajima H. Post-Transcriptional Regulation of Immune Responses and Inflammatory Diseases by RNA-Binding ZFP36 Family Proteins. Front Immunol 2021; 12:711633. [PMID: 34276705 PMCID: PMC8282349 DOI: 10.3389/fimmu.2021.711633] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 06/17/2021] [Indexed: 12/11/2022] Open
Abstract
Post-transcriptional regulation is involved in the regulation of many inflammatory genes. Zinc finger protein 36 (ZFP36) family proteins are RNA-binding proteins involved in messenger RNA (mRNA) metabolism pathways. The ZFP36 family is composed of ZFP36 (also known as tristetraprolin, TTP), ZFP36L1, ZFP36L2, and ZFP36L3 (only in rodents). The ZFP36 family proteins contain two tandemly repeated CCCH-type zinc-finger motifs, bind to adenine uridine-rich elements in the 3’-untranslated regions (3’ UTR) of specific mRNA, and lead to target mRNA decay. Although the ZFP36 family members are structurally similar, they are known to play distinct functions and regulate different target mRNAs, probably due to their cell-type-specific expression patterns. For instance, ZFP36 has been well-known to function as an anti-inflammatory modulator in murine models of systemic inflammatory diseases by down-regulating the production of various pro-inflammatory cytokines, including TNF-α. Meanwhile, ZFP36L1 is required for the maintenance of the marginal-zone B cell compartment. Recently, we found that ZFP36L2 reduces the expression of Ikzf2 (encoding HELIOS) and suppresses regulatory T cell function. This review summarizes the current understanding of the post-transcriptional regulation of immunological responses and inflammatory diseases by RNA-binding ZFP36 family proteins.
Collapse
Affiliation(s)
- Sohei Makita
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hiroaki Takatori
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan.,Department of Rheumatology, Hamamatsu Medical Center, Hamamatsu, Japan
| | - Hiroshi Nakajima
- Department of Allergy and Clinical Immunology, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
18
|
Gordon D, Hellriegel ET, Hope HR, Burt D, Monahan JB. Safety, Tolerability, Pharmacokinetics, and Pharmacodynamics of the MK2 Inhibitor ATI-450 in Healthy Subjects: A Placebo-Controlled, Randomized Phase 1 Study. Clin Pharmacol 2021; 13:123-134. [PMID: 34140814 PMCID: PMC8203602 DOI: 10.2147/cpaa.s305308] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 05/12/2021] [Indexed: 11/23/2022] Open
Abstract
Purpose ATI-450 is an oral, small-molecule inhibitor of the p38α mitogen-activated protein kinase (MAPK)/MAPK-activated protein kinase 2 (MK2) inflammatory signaling pathway. This phase 1, single and multiple ascending dose (SAD, MAD) study evaluated ATI-450 safety, tolerability, pharmacokinetics, and pharmacodynamics. Patients and Methods Healthy adults were randomly assigned to SAD (10, 30, 50, 100 mg; n=24) and MAD (10, 30, 50 mg twice daily [BID] for 7 days; n=24) cohorts of ATI-450 or placebo (n=14). Safety and tolerability were evaluated through clinical and laboratory assessments. Pharmacokinetic parameters were evaluated in plasma samples; pharmacodynamic assessments included quantification of cytokine levels (tumor necrosis factor α [TNF-α], interleukin [IL]-1β, IL-6, IL-8) and phosphorylation of the MK2 downstream substrate, heat shock protein 27 (p-HSP27). Results The most common adverse events were headache (10/48, 20.8%), dizziness (6/48, 12.5%), upper respiratory tract infection (3/48, 6.3%), and constipation (3/48, 6.3%). Pharmacokinetics were dose-proportional, with a terminal half-life of 9‒12 hours in the MAD cohorts on day 7. Dose- and concentration-dependent inhibition of ex vivo stimulated cytokines and target biomarker was observed. On day 7, patients in the 50 mg BID dose cohort recorded mean trough drug levels that were 1.4, 2.2, 2.3, and 2.4 times greater than the IC80 for TNF-α, IL-1β, IL-8, and p-HSP27, respectively. Mean Cmax was 3.5, 5.4, 5.6, and 6.0 times greater than the IC80 for TNF-α, IL-1β, IL-8, and p-HSP27, respectively. IL-6 inhibition >50% was noted for part of the dosing interval. Conclusion ATI-450 was well tolerated at the doses investigated, exhibited dose- and time-independent (ie, linear) pharmacokinetics, and dose-related pharmacodynamic effects. These results support further study of ATI-450 in immunoinflammatory diseases in phase 2 trials.
Collapse
Affiliation(s)
- David Gordon
- Research and Development, Aclaris Therapeutics, Inc., Wayne, PA, USA
| | | | - Heidi Rath Hope
- Research and Development, Aclaris Therapeutics, Inc., and Confluence Discovery Technologies, Inc., St. Louis, MO, USA
| | - David Burt
- Research and Development, Aclaris Therapeutics, Inc., Wayne, PA, USA
| | - Joseph B Monahan
- Research and Development, Aclaris Therapeutics, Inc., and Confluence Discovery Technologies, Inc., St. Louis, MO, USA
| |
Collapse
|
19
|
Hsieh HH, Chen YA, Chang YJ, Wang HH, Yu YH, Lin SW, Huang YJ, Lin S, Chang CJ. The functional characterization of phosphorylation of tristetraprolin at C-terminal NOT1-binding domain. JOURNAL OF INFLAMMATION-LONDON 2021; 18:22. [PMID: 34090459 PMCID: PMC8180021 DOI: 10.1186/s12950-021-00288-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 05/20/2021] [Indexed: 12/26/2022]
Abstract
Background Tristetraprolin (TTP) family proteins contain conserved tandem CCCH zinc-finger binding to AU-rich elements and C-terminal NOT1-binding domain. TTP is phosphorylated extensively in cells, and its mRNA destabilization activity is regulated by protein phosphorylation. Methods We generated an antibody against phospho-Serine316 located at the C-terminal NOT1-binding site and examined TTP phosphorylation in LPS-stimulated RAW264.7 cells. Knockout of TTP was created in RAW264.7 cells using CRISPR/Cas9 gene editing to explore TTP functions. Results We demonstrated that Ser316 was phosphorylated by p90 ribosomal S6 kinase 1 (RSK1) and p38-activated protein kinase (MK2) and dephosphorylated by Protein Phosphatase 2A (PP2A). A phosphorylation-mimic mutant of S316D resulted in dissociation with the CCR4-NOT deadenylase complex through weakening interaction with CNOT1. Furthermore, Ser316 and serines 52 and 178 were independently contributed to the CCR4-NOT complex recruitment in the immunoprecipitation assay using phosphor-mimic mutants. In RAW264.7 macrophages, TTP was induced, and Ser316 was phosphorylated through RSK1 and MK2 by LPS stimulation. Knockout of TTP resulted in TNFα mRNA increased due to mRNA stabilization. Overexpression of non-phosphorylated S316A TTP mutant can restore TTP activity and lead to TNFα mRNA decreased. GST pull-down and RNA pull-down analyses demonstrated that endogenous TTP with Ser316 phosphorylation decreased the interaction with CNOT1. Conclusions Our results suggest that the TTP-mediated mRNA stability is modulated by Ser316 phosphorylation via regulating the TTP interaction with the CCR4-NOT deadenylase complex. Supplementary Information The online version contains supplementary material available at 10.1186/s12950-021-00288-2.
Collapse
Affiliation(s)
- Hsin-Hui Hsieh
- Graduate Institute of Biochemical Sciences, College of Life Science, National Taiwan University, No. 1 Sec 4 Roosevelt Rd, Taipei, 106, Taiwan
| | - Yen-An Chen
- Graduate Institute of Biochemical Sciences, College of Life Science, National Taiwan University, No. 1 Sec 4 Roosevelt Rd, Taipei, 106, Taiwan
| | - Yao-Jen Chang
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Hsin-Hui Wang
- Department of Pediatrics, Division of Pediatric Immunology and Nephrology, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Pediatrics, Faculty of Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan.,Institute of Emergency and Critical Care Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ya-Han Yu
- Graduate Institute of Biochemical Sciences, College of Life Science, National Taiwan University, No. 1 Sec 4 Roosevelt Rd, Taipei, 106, Taiwan
| | - Sheng-Wei Lin
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Yin-Jung Huang
- Department of Pediatrics, Division of Pediatric Immunology and Nephrology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Steven Lin
- Graduate Institute of Biochemical Sciences, College of Life Science, National Taiwan University, No. 1 Sec 4 Roosevelt Rd, Taipei, 106, Taiwan.,Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Ching-Jin Chang
- Graduate Institute of Biochemical Sciences, College of Life Science, National Taiwan University, No. 1 Sec 4 Roosevelt Rd, Taipei, 106, Taiwan. .,Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
20
|
Rappl P, Brüne B, Schmid T. Role of Tristetraprolin in the Resolution of Inflammation. BIOLOGY 2021; 10:biology10010066. [PMID: 33477783 PMCID: PMC7832405 DOI: 10.3390/biology10010066] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/14/2021] [Accepted: 01/16/2021] [Indexed: 12/18/2022]
Abstract
Simple Summary Chronic inflammatory diseases account for up to 60% of deaths worldwide and, thus, are considered a great threat for human health by the World Health Organization. Nevertheless, acute inflammatory reactions are an integral part of the host defense against invading pathogens or injuries. To avoid excessive damage due to the persistence of a highly reactive environment, inflammations need to resolve in a coordinate and timely manner, ensuring for the immunological normalization of the affected tissues. Since post-transcriptional regulatory mechanisms are essential for effective resolution, the present review discusses the key role of the RNA-binding and post-transcriptional regulatory protein tristetraprolin in establishing resolution of inflammation. Abstract Inflammation is a crucial part of immune responses towards invading pathogens or tissue damage. While inflammatory reactions are aimed at removing the triggering stimulus, it is important that these processes are terminated in a coordinate manner to prevent excessive tissue damage due to the highly reactive inflammatory environment. Initiation of inflammatory responses was proposed to be regulated predominantly at a transcriptional level, whereas post-transcriptional modes of regulation appear to be crucial for resolution of inflammation. The RNA-binding protein tristetraprolin (TTP) interacts with AU-rich elements in the 3′ untranslated region of mRNAs, recruits deadenylase complexes and thereby facilitates degradation of its targets. As TTP regulates the mRNA stability of numerous inflammatory mediators, it was put forward as a crucial post-transcriptional regulator of inflammation. Here, we summarize the current understanding of the function of TTP with a specific focus on its role in adding to resolution of inflammation.
Collapse
Affiliation(s)
- Peter Rappl
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (P.R.); (B.B.)
| | - Bernhard Brüne
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (P.R.); (B.B.)
- German Cancer Consortium (DKTK), Partner Site Frankfurt, 60590 Frankfurt, Germany
- Frankfurt Cancer Institute, Goethe-University Frankfurt, 60596 Frankfurt, Germany
- Project Group Translational Medicine and Pharmacology TMP, Fraunhofer Institute for Molecular and Applied Ecology, 60596 Frankfurt, Germany
| | - Tobias Schmid
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (P.R.); (B.B.)
- Correspondence:
| |
Collapse
|
21
|
Beihammer G, Maresch D, Altmann F, Van Damme EJM, Strasser R. Lewis A Glycans Are Present on Proteins Involved in Cell Wall Biosynthesis and Appear Evolutionarily Conserved Among Natural Arabidopsis thaliana Accessions. FRONTIERS IN PLANT SCIENCE 2021; 12:630891. [PMID: 33777069 PMCID: PMC7991798 DOI: 10.3389/fpls.2021.630891] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 01/18/2021] [Indexed: 05/02/2023]
Abstract
N-glycosylation is a highly abundant protein modification present in all domains of life. Terminal sugar residues on complex-type N-glycans mediate various crucial biological processes in mammals such as cell-cell recognition or protein-ligand interactions. In plants, the Lewis A trisaccharide constitutes the only known outer-chain elongation of complex N-glycans. Lewis A containing complex N-glycans appear evolutionary conserved, having been identified in all plant species analyzed so far. Despite their ubiquitous occurrence, the biological function of this complex N-glycan modification is currently unknown. Here, we report the identification of Lewis A bearing glycoproteins from three different plant species: Arabidopsis thaliana, Nicotiana benthamiana, and Oryza sativa. Affinity purification via the JIM84 antibody, directed against Lewis A structures on complex plant N-glycans, was used to enrich Lewis A bearing glycoproteins, which were subsequently identified via nano-LC-MS. Selected identified proteins were recombinantly expressed and the presence of Lewis A confirmed via immunoblotting and site-specific N-glycan analysis. While the proteins identified in O. sativa are associated with diverse functions, proteins from A. thaliana and N. benthamiana are mainly involved in cell wall biosynthesis. However, a Lewis A-deficient mutant line of A. thaliana showed no change in abundance of cell wall constituents such as cellulose or lignin. Furthermore, we investigated the presence of Lewis A structures in selected accessions from the 1001 genome database containing amino acid variations in the enzymes required for Lewis A biosynthesis. Besides one relict line showing no detectable levels of Lewis A, the modification was present in all other tested accessions. The data provided here comprises the so far first attempt at identifying Lewis A bearing glycoproteins across different species and will help to shed more light on the role of Lewis A structures in plants.
Collapse
Affiliation(s)
- Gernot Beihammer
- Department of Applied Genetics and Cell Biology, Institute of Plant Biotechnology and Cell Biology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Daniel Maresch
- Division of Biochemistry, Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Friedrich Altmann
- Division of Biochemistry, Department of Chemistry, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Els J. M. Van Damme
- Laboratory of Biochemistry and Glycobiology, Department of Biotechnology, Ghent University, Ghent, Belgium
| | - Richard Strasser
- Department of Applied Genetics and Cell Biology, Institute of Plant Biotechnology and Cell Biology, University of Natural Resources and Life Sciences, Vienna, Austria
- *Correspondence: Richard Strasser
| |
Collapse
|
22
|
Rezcallah MC, Al-Mazi T, Ammit AJ. Cataloguing the phosphorylation sites of tristetraprolin (TTP): Functional implications for inflammatory diseases. Cell Signal 2020; 78:109868. [PMID: 33276085 DOI: 10.1016/j.cellsig.2020.109868] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/29/2020] [Accepted: 11/29/2020] [Indexed: 01/10/2023]
Abstract
Tristetraprolin (TTP) is a destabilizing mRNA binding protein known to regulate gene expression of a wide variety of targets, including those that control inflammation. TTP expression, regulation and function is controlled by phosphorylation. While the importance of key serine (S) sites (S52 and S178 in mice and S186 in humans) has been recognized, other sites on the hyperphosphorylated TTP protein have more recently emerged as playing an important role in regulating cellular signalling and downstream functions of TTP. In order to propel investigation of TTP and fully exploit its potential as a drug target in inflammatory disease, this review will catalogue TTP phosphorylation sites in both the murine and human TTP protein, the known and unknown roles and functions of these sites, the kinases and phosphatases that act upon TTP and overview methodological approaches to increase our knowledge of this important protein regulated by phosphorylation.
Collapse
Affiliation(s)
- Maria C Rezcallah
- Woolcock Emphysema Centre, Woolcock Institute of Medical Research, University of Sydney, NSW, Australia; School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Trisha Al-Mazi
- Woolcock Emphysema Centre, Woolcock Institute of Medical Research, University of Sydney, NSW, Australia; School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia
| | - Alaina J Ammit
- Woolcock Emphysema Centre, Woolcock Institute of Medical Research, University of Sydney, NSW, Australia; School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW, Australia.
| |
Collapse
|
23
|
Sun X, Zhang H, Xie L, Qian C, Ye Y, Mao H, Wang B, Zhang H, Zhang Y, He X, Zhang S. Tristetraprolin destabilizes NOX2 mRNA and protects dopaminergic neurons from oxidative damage in Parkinson's disease. FASEB J 2020; 34:15047-15061. [PMID: 32954540 DOI: 10.1096/fj.201902967r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 08/04/2020] [Accepted: 08/31/2020] [Indexed: 01/11/2023]
Abstract
Tristetraprolin (TTP), an RNA-binding protein encoded by the ZFP36 gene, is vital for neural differentiation; however, its involvement in neurodegenerative diseases such as Parkinson's disease (PD) remains unclear. To explore the role of TTP in PD, an in vitro 1-methyl-4-phenylpyridinium (MPP+ ) cell model and an in vivo 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) of PD were used. Transfection of small interfering (si)-TTP RNA upregulated pro-oxidative NOX2 expression and ROS formation, downregulated anti-oxidative GSH and SOD activity;si-TTP upregulated pro-apoptotic cleaved-caspase-3 expression, and downregulated antiapoptotic Bcl-2 expression; while overexpression (OE)-TTP lentivirus caused opposite effects. Through database prediction, luciferase experiment, RNA immunoprecipitation (RIP), and mRNA stability analysis, we evaluated the potential binding sites of TTP to 3'-untranslated regions (3'-UTR) of NOX2 mRNA. TTP affected the NOX2 luciferase activity by binding to two sites in the NOX2 3'-UTR. RIP-qPCR confirmed TTP binding to both sites, with a higher affinity for site-2. In addition, TTP reduced the NOX2 mRNA stability. si-NOX2 and antioxidant N-acetyl cysteine (NAC) reversed si-TTP-induced cell apoptosis. In MPTP-treated mice, TTP expression increased and was co-located with dopaminergic neurons. TTP also inhibited NOX2 and decreased the oxidative stress in vivo. In conclusion, TTP protects against dopaminergic oxidative injury by promoting NOX2 mRNA degradation in the MPP+ /MPTP model of PD, suggesting that TTP could be a potential therapeutic target for regulating the oxidative stress in PD.
Collapse
Affiliation(s)
- Xiang Sun
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Hongbo Zhang
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Linghai Xie
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Chen Qian
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yongyi Ye
- Department of Neurosurgery, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Hengxu Mao
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Baoyan Wang
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Huan Zhang
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yizhou Zhang
- Tarbut V'Torah Community Day School, Irvine, CA, USA
| | - Xiaozheng He
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shizhong Zhang
- The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
24
|
Choudhary I, Vo T, Bathula CS, Lamichhane R, Lewis BW, Looper J, Jeyaseelan S, Blackshear PJ, Saini Y, Patial S. Tristetraprolin Overexpression in Non-hematopoietic Cells Protects Against Acute Lung Injury in Mice. Front Immunol 2020; 11:2164. [PMID: 32983182 PMCID: PMC7493631 DOI: 10.3389/fimmu.2020.02164] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/10/2020] [Indexed: 12/26/2022] Open
Abstract
Tristetraprolin (TTP) is a mRNA binding protein that binds to adenylate-uridylate-rich elements within the 3′ untranslated regions of certain transcripts, such as tumor necrosis factor (Tnf) mRNA, and increases their rate of decay. Modulation of TTP expression is implicated in inflammation; however, its role in acute lung inflammation remains unknown. Accordingly, we tested the role of TTP in lipopolysaccharide (LPS)-induced acute lung injury (ALI) in mice. LPS-challenged TTP-knockout (TTPKO) mice, as well as myeloid cell-specific TTP-deficient (TTPmyeKO) mice, exhibited significant increases in lung injury, although these responses were more robust in the TTPKO. Mice with systemic overexpression of TTP (TTPΔARE) were protected from ALI, as indicated by significantly reduced neutrophilic infiltration, reduced levels of neutrophil chemoattractants, and histological parameters of ALI. Interestingly, while irradiated wild-type (WT) mice reconstituted with TTPKO hematopoietic progenitor cells (HPCs) showed exaggerated ALI, their reconstitution with the TTPΔARE HPCs mitigated ALI. The reconstitution of irradiated TTPΔARE mice with HPCs from either WT or TTPΔARE donors conferred significant protection against ALI. In contrast, irradiated TTPΔARE mice reconstituted with TTPKO HPCs had exaggerated ALI, but the response was milder as compared to WT recipients that received TTPKO HPCs. Finally, the reconstitution of irradiated TTPKO recipient mice with TTPΔARE HPCs did not confer any protection to the TTPKO mice. These data together suggest that non-HPCs-specific overexpression of TTP within the lungs protects against ALI via downregulation of neutrophil chemoattractants and reduction in neutrophilic infiltration.
Collapse
Affiliation(s)
- Ishita Choudhary
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Thao Vo
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Chandra S Bathula
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Richa Lamichhane
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Brandon W Lewis
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Jayme Looper
- Veterinary Clinical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Samithamby Jeyaseelan
- Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Perry J Blackshear
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, NC, United States
| | - Yogesh Saini
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| | - Sonika Patial
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, United States
| |
Collapse
|
25
|
Uehata T, Takeuchi O. RNA Recognition and Immunity-Innate Immune Sensing and Its Posttranscriptional Regulation Mechanisms. Cells 2020; 9:cells9071701. [PMID: 32708595 PMCID: PMC7407594 DOI: 10.3390/cells9071701] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 07/13/2020] [Accepted: 07/15/2020] [Indexed: 12/13/2022] Open
Abstract
RNA acts as an immunostimulatory molecule in the innate immune system to activate nucleic acid sensors. It functions as an intermediate, conveying genetic information to control inflammatory responses. A key mechanism for RNA sensing is discriminating self from non-self nucleic acids to initiate antiviral responses reliably, including the expression of type I interferon (IFN) and IFN-stimulated genes. Another important aspect of the RNA-mediated inflammatory response is posttranscriptional regulation of gene expression, where RNA-binding proteins (RBPs) have essential roles in various RNA metabolisms, including splicing, nuclear export, modification, and translation and mRNA degradation. Recent evidence suggests that the control of mRNA stability is closely involved in signal transduction and orchestrates immune responses. In this study, we review the current understanding of how RNA is sensed by host RNA sensing machinery and discuss self/non-self-discrimination in innate immunity focusing on mammalian species. Finally, we discuss how posttranscriptional regulation by RBPs shape immune reactions.
Collapse
|
26
|
Tabassum N, Eschen-Lippold L, Athmer B, Baruah M, Brode M, Maldonado-Bonilla LD, Hoehenwarter W, Hause G, Scheel D, Lee J. Phosphorylation-dependent control of an RNA granule-localized protein that fine-tunes defence gene expression at a post-transcriptional level. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2020; 101:1023-1039. [PMID: 31628867 DOI: 10.1111/tpj.14573] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 09/18/2019] [Accepted: 10/03/2019] [Indexed: 05/12/2023]
Abstract
Mitogen-activated protein kinase (MAPK) cascades are key signalling modules of plant defence responses to pathogen-associated molecular patterns [PAMPs; e.g. the bacterial peptide flagellin (flg22)]. Tandem zinc finger protein 9 (TZF9) is a RNA-binding protein that is phosphorylated by two PAMP-responsive MAPKs, MPK3 and MPK6. We mapped the major phosphosites in TZF9 and showed their importance for controlling in vitro RNA-binding activity, in vivo flg22-induced rapid disappearance of TZF9-labelled processing body-like structures and TZF9 protein turnover. Microarray analysis showed a strong discordance between transcriptome (total mRNA) and translatome (polysome-associated mRNA) in the tzf9 mutant, with more mRNAs associated with ribosomes in the absence of TZF9. This suggests that TZF9 may sequester and inhibit the translation of subsets of mRNAs. Fittingly, TZF9 physically interacts with poly(A)-binding protein 2 (PAB2), a hallmark constituent of stress granules - sites for stress-induced translational stalling/arrest. TZF9 even promotes the assembly of stress granules in the absence of stress. Hence, MAPKs may control defence gene expression post-transcriptionally through release from translation arrest within TZF9-PAB2-containing RNA granules or by perturbing the function of PAB2 in translation control (e.g. in the mRNA closed-loop model of translation).
Collapse
Affiliation(s)
- Naheed Tabassum
- Leibniz Institute of Plant Biochemistry, Weinberg 3, Halle/Saale, D-06120, Germany
| | | | - Benedikt Athmer
- Leibniz Institute of Plant Biochemistry, Weinberg 3, Halle/Saale, D-06120, Germany
| | - Manaswita Baruah
- Leibniz Institute of Plant Biochemistry, Weinberg 3, Halle/Saale, D-06120, Germany
| | - Martina Brode
- Leibniz Institute of Plant Biochemistry, Weinberg 3, Halle/Saale, D-06120, Germany
| | | | | | - Gerd Hause
- Biocenter, Martin Luther University Halle-Wittenberg, Weinbergweg 22, D-06120, Halle/Saale, Germany
| | - Dierk Scheel
- Leibniz Institute of Plant Biochemistry, Weinberg 3, Halle/Saale, D-06120, Germany
| | - Justin Lee
- Leibniz Institute of Plant Biochemistry, Weinberg 3, Halle/Saale, D-06120, Germany
| |
Collapse
|
27
|
Ingawale DK, Mandlik SK. New insights into the novel anti-inflammatory mode of action of glucocorticoids. Immunopharmacol Immunotoxicol 2020; 42:59-73. [PMID: 32070175 DOI: 10.1080/08923973.2020.1728765] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Inflammation is a physiological intrinsic host response to injury meant for removal of noxious stimuli and maintenance of homeostasis. It is a defensive body mechanism that involves immune cells, blood vessels and molecular mediators of inflammation. Glucocorticoids (GCs) are steroidal hormones responsible for regulation of homeostatic and metabolic functions of body. Synthetic GCs are the most useful anti-inflammatory drugs used for the treatment of chronic inflammatory diseases such as asthma, chronic obstructive pulmonary disease (COPD), allergies, multiple sclerosis, tendinitis, lupus, atopic dermatitis, ulcerative colitis, rheumatoid arthritis and osteoarthritis whereas, the long term use of GCs are associated with many side effects. The anti-inflammatory and immunosuppressive (desired) effects of GCs are usually mediated by transrepression mechanism whereas; the metabolic and toxic (undesired) effects are usually manifested by transactivation mechanism. Though GCs are most potent anti-inflammatory and immunosuppressive drugs, the common problem associated with their use is GC resistance. Several research studies are rising to comprehend these mechanisms, which would be helpful in improving the GC resistance in asthma and COPD patients. This review aims to focus on identification of new drug targets in inflammation which will be helpful in the resolution of inflammation. The ample understanding of GC mechanisms of action helps in the development of novel anti-inflammatory drugs for the treatment of inflammatory and autoimmune disease with reduced side effects and minimal toxicity.
Collapse
Affiliation(s)
- Deepa K Ingawale
- Department of Pharmacology, Poona College of Pharmacy, Bharati Vidyapeeth Deemed University, Pune, India
| | - Satish K Mandlik
- Department of Pharmacology, Sinhgad College of Pharmacy, Pune, India
| |
Collapse
|
28
|
Lee S, Micalizzi D, Truesdell SS, Bukhari SIA, Boukhali M, Lombardi-Story J, Kato Y, Choo MK, Dey-Guha I, Ji F, Nicholson BT, Myers DT, Lee D, Mazzola MA, Raheja R, Langenbucher A, Haradhvala NJ, Lawrence MS, Gandhi R, Tiedje C, Diaz-Muñoz MD, Sweetser DA, Sadreyev R, Sykes D, Haas W, Haber DA, Maheswaran S, Vasudevan S. A post-transcriptional program of chemoresistance by AU-rich elements and TTP in quiescent leukemic cells. Genome Biol 2020; 21:33. [PMID: 32039742 PMCID: PMC7011231 DOI: 10.1186/s13059-020-1936-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 01/15/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Quiescence (G0) is a transient, cell cycle-arrested state. By entering G0, cancer cells survive unfavorable conditions such as chemotherapy and cause relapse. While G0 cells have been studied at the transcriptome level, how post-transcriptional regulation contributes to their chemoresistance remains unknown. RESULTS We induce chemoresistant and G0 leukemic cells by serum starvation or chemotherapy treatment. To study post-transcriptional regulation in G0 leukemic cells, we systematically analyzed their transcriptome, translatome, and proteome. We find that our resistant G0 cells recapitulate gene expression profiles of in vivo chemoresistant leukemic and G0 models. In G0 cells, canonical translation initiation is inhibited; yet we find that inflammatory genes are highly translated, indicating alternative post-transcriptional regulation. Importantly, AU-rich elements (AREs) are significantly enriched in the upregulated G0 translatome and transcriptome. Mechanistically, we find the stress-responsive p38 MAPK-MK2 signaling pathway stabilizes ARE mRNAs by phosphorylation and inactivation of mRNA decay factor, Tristetraprolin (TTP) in G0. This permits expression of ARE mRNAs that promote chemoresistance. Conversely, inhibition of TTP phosphorylation by p38 MAPK inhibitors and non-phosphorylatable TTP mutant decreases ARE-bearing TNFα and DUSP1 mRNAs and sensitizes leukemic cells to chemotherapy. Furthermore, co-inhibiting p38 MAPK and TNFα prior to or along with chemotherapy substantially reduces chemoresistance in primary leukemic cells ex vivo and in vivo. CONCLUSIONS These studies uncover post-transcriptional regulation underlying chemoresistance in leukemia. Our data reveal the p38 MAPK-MK2-TTP axis as a key regulator of expression of ARE-bearing mRNAs that promote chemoresistance. By disrupting this pathway, we develop an effective combination therapy against chemosurvival.
Collapse
Affiliation(s)
- Sooncheol Lee
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 185 Cambridge St, CPZN4202, Boston, MA, 02114, USA
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, Massachusetts, USA
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Douglas Micalizzi
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 185 Cambridge St, CPZN4202, Boston, MA, 02114, USA
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, Massachusetts, USA
| | - Samuel S Truesdell
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 185 Cambridge St, CPZN4202, Boston, MA, 02114, USA
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Syed I A Bukhari
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 185 Cambridge St, CPZN4202, Boston, MA, 02114, USA
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, Massachusetts, USA
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Myriam Boukhali
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 185 Cambridge St, CPZN4202, Boston, MA, 02114, USA
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, Massachusetts, USA
| | - Jennifer Lombardi-Story
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 185 Cambridge St, CPZN4202, Boston, MA, 02114, USA
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, Massachusetts, USA
| | - Yasutaka Kato
- Laboratory of Oncology, Hokuto Hospital, Obihiro, Japan
| | - Min-Kyung Choo
- Cutaneous Biology Research Center, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Ipsita Dey-Guha
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 185 Cambridge St, CPZN4202, Boston, MA, 02114, USA
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, Massachusetts, USA
| | - Fei Ji
- Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Benjamin T Nicholson
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 185 Cambridge St, CPZN4202, Boston, MA, 02114, USA
| | - David T Myers
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 185 Cambridge St, CPZN4202, Boston, MA, 02114, USA
| | - Dongjun Lee
- Department of Convergence Medical Science, Pusan National University School of Medicine, Yangsan, 50612, 1257-1258, South Korea
| | - Maria A Mazzola
- Center for Neurological Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Radhika Raheja
- Center for Neurological Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Adam Langenbucher
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 185 Cambridge St, CPZN4202, Boston, MA, 02114, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Nicholas J Haradhvala
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 185 Cambridge St, CPZN4202, Boston, MA, 02114, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
- Broad Institute of Harvard & MIT, Cambridge, MA, 02142, USA
| | - Michael S Lawrence
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 185 Cambridge St, CPZN4202, Boston, MA, 02114, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
- Broad Institute of Harvard & MIT, Cambridge, MA, 02142, USA
| | - Roopali Gandhi
- Center for Neurological Diseases, Brigham & Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Christopher Tiedje
- Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, Blegdamsvej 3B, 2200, Copenhagen, Denmark
| | - Manuel D Diaz-Muñoz
- Centre de Physiopathologie Toulouse-Purpan, INSERM UMR1043/CNRS U5282, Toulouse, France
| | - David A Sweetser
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 185 Cambridge St, CPZN4202, Boston, MA, 02114, USA
- Department of Pediatrics, Divisions of Pediatric Hematology/Oncology and Medical Genetics, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Ruslan Sadreyev
- Department of Molecular Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - David Sykes
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, Massachusetts, USA
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA
| | - Wilhelm Haas
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 185 Cambridge St, CPZN4202, Boston, MA, 02114, USA
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, Massachusetts, USA
| | - Daniel A Haber
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 185 Cambridge St, CPZN4202, Boston, MA, 02114, USA
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, Massachusetts, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, 20815, USA
| | - Shyamala Maheswaran
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 185 Cambridge St, CPZN4202, Boston, MA, 02114, USA
- Department of Surgery, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Shobha Vasudevan
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 185 Cambridge St, CPZN4202, Boston, MA, 02114, USA.
- Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, 02114, Massachusetts, USA.
- Center for Regenerative Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA.
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, 02138, USA.
| |
Collapse
|
29
|
Mahmoud L, Abdulkarim AS, Kutbi S, Moghrabi W, Altwijri S, Khabar KSA, Hitti EG. Post-Transcriptional Inflammatory Response to Intracellular Bacterial c-di-AMP. Front Immunol 2020; 10:3050. [PMID: 32010134 PMCID: PMC6979040 DOI: 10.3389/fimmu.2019.03050] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 12/12/2019] [Indexed: 12/31/2022] Open
Abstract
Cyclic-di-AMP (c-di-AMP) is a bacterial second messenger that is produced by intracellular bacterial pathogens in mammalian host macrophages. Previous reports have shown that c-di-AMP is recognized by intracellular pattern recognition receptors of the innate immune system and stimulate type I interferon response. Here we report that the response to c-di-AMP includes a post-transcriptional component that is involved in the induction of additional inflammatory cytokines including IL-6, CXCL2, CCL3, and CCL4. Their mRNAs contain AU-rich elements (AREs) in their 3' UTR that promote decay and repress translation. We show that c-di-AMP leads to the phosphorylation of p38 MAPK as well as the induction of the ARE-binding protein TTP, both of which are components of a signaling pathway that modulate the expression of ARE-containing mRNAs at the post-transcriptional level. Pharmacological inhibition of p38 reduces the c-di-AMP-dependent release of induced cytokines, while TTP knockdown increases their release and mRNA stability. C-di-AMP can specifically increase the expression of a nano-Luciferase reporter that contains AREs. We propose a non-canonical intracellular mode of activation of the p38 MAPK pathway with the subsequent enhancement in the expression of inflammatory cytokines. C-di-AMP is widely distributed in bacteria, including infectious intracellular pathogens; hence, understanding of its post-transcriptional gene regulatory effect on the host response may provide novel approaches for therapy.
Collapse
Affiliation(s)
- Linah Mahmoud
- Molecular BioMedicine Program, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Alaa S Abdulkarim
- Molecular BioMedicine Program, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Shaima Kutbi
- Molecular BioMedicine Program, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Walid Moghrabi
- Molecular BioMedicine Program, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Sulaiman Altwijri
- Molecular BioMedicine Program, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Khalid S A Khabar
- Molecular BioMedicine Program, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| | - Edward G Hitti
- Molecular BioMedicine Program, King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia
| |
Collapse
|
30
|
Nair PM, Starkey MR, Haw TJ, Liu G, Collison AM, Mattes J, Wark PA, Morris JC, Verrills NM, Clark AR, Ammit AJ, Hansbro PM. Enhancing tristetraprolin activity reduces the severity of cigarette smoke-induced experimental chronic obstructive pulmonary disease. Clin Transl Immunology 2019; 8:e01084. [PMID: 31921419 PMCID: PMC6946917 DOI: 10.1002/cti2.1084] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 09/29/2019] [Accepted: 09/29/2019] [Indexed: 12/18/2022] Open
Abstract
OBJECTIVE Chronic obstructive pulmonary disease (COPD) is a progressive disease that causes significant mortality and morbidity worldwide and is primarily caused by the inhalation of cigarette smoke (CS). Lack of effective treatments for COPD means there is an urgent need to identify new therapeutic strategies for the underlying mechanisms of pathogenesis. Tristetraprolin (TTP) encoded by the Zfp36 gene is an anti-inflammatory protein that induces mRNA decay, especially of transcripts encoding inflammatory cytokines, including those implicated in COPD. METHODS Here, we identify a novel protective role for TTP in CS-induced experimental COPD using Zfp36aa/aa mice, a genetically modified mouse strain in which endogenous TTP cannot be phosphorylated, rendering it constitutively active as an mRNA-destabilising factor. TTP wild-type (Zfp36 +/+) and Zfp36aa/aa active C57BL/6J mice were exposed to CS for four days or eight weeks, and the impact on acute inflammatory responses or chronic features of COPD, respectively, was assessed. RESULTS After four days of CS exposure, Zfp36aa/aa mice had reduced numbers of airway neutrophils and lymphocytes and mRNA expression levels of cytokines compared to wild-type controls. After eight weeks, Zfp36aa/aa mice had reduced pulmonary inflammation, airway remodelling and emphysema-like alveolar enlargement, and lung function was improved. We then used pharmacological treatments in vivo (protein phosphatase 2A activator, AAL(S), and the proteasome inhibitor, bortezomib) to promote the activation and stabilisation of TTP and show that hallmark features of CS-induced experimental COPD were ameliorated. CONCLUSION Collectively, our study provides the first evidence for the therapeutic potential of inducing TTP as a treatment for COPD.
Collapse
Affiliation(s)
- Prema M Nair
- Priority Research Centres for Healthy Lungs, Grow Up Well and Cancer Research, Innovation and TranslationHunter Medical Research InstituteUniversity of NewcastleNSWAustralia
- School of Biomedical Sciences and PharmacyFaculty of Health and MedicineUniversity of NewcastleCallaghanNSWAustralia
| | - Malcolm R Starkey
- Priority Research Centres for Healthy Lungs, Grow Up Well and Cancer Research, Innovation and TranslationHunter Medical Research InstituteUniversity of NewcastleNSWAustralia
- School of Biomedical Sciences and PharmacyFaculty of Health and MedicineUniversity of NewcastleCallaghanNSWAustralia
| | - Tatt Jhong Haw
- Priority Research Centres for Healthy Lungs, Grow Up Well and Cancer Research, Innovation and TranslationHunter Medical Research InstituteUniversity of NewcastleNSWAustralia
- School of Biomedical Sciences and PharmacyFaculty of Health and MedicineUniversity of NewcastleCallaghanNSWAustralia
| | - Gang Liu
- Priority Research Centres for Healthy Lungs, Grow Up Well and Cancer Research, Innovation and TranslationHunter Medical Research InstituteUniversity of NewcastleNSWAustralia
- School of Biomedical Sciences and PharmacyFaculty of Health and MedicineUniversity of NewcastleCallaghanNSWAustralia
| | - Adam M Collison
- Priority Research Centres for Healthy Lungs, Grow Up Well and Cancer Research, Innovation and TranslationHunter Medical Research InstituteUniversity of NewcastleNSWAustralia
| | - Joerg Mattes
- Priority Research Centres for Healthy Lungs, Grow Up Well and Cancer Research, Innovation and TranslationHunter Medical Research InstituteUniversity of NewcastleNSWAustralia
| | - Peter A. Wark
- Priority Research Centres for Healthy Lungs, Grow Up Well and Cancer Research, Innovation and TranslationHunter Medical Research InstituteUniversity of NewcastleNSWAustralia
| | | | - Nikki M Verrills
- Priority Research Centres for Healthy Lungs, Grow Up Well and Cancer Research, Innovation and TranslationHunter Medical Research InstituteUniversity of NewcastleNSWAustralia
- School of Biomedical Sciences and PharmacyFaculty of Health and MedicineUniversity of NewcastleCallaghanNSWAustralia
| | - Andrew R Clark
- Institute of Inflammation and AgeingCollege of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| | - Alaina J Ammit
- Woolcock Emphysema CentreWoolcock Institute of Medical ResearchUniversity of SydneyNSWAustralia
- School of Life SciencesFaculty of ScienceUniversity of Technology SydneySydneyNSWAustralia
| | - Philip M Hansbro
- Priority Research Centres for Healthy Lungs, Grow Up Well and Cancer Research, Innovation and TranslationHunter Medical Research InstituteUniversity of NewcastleNSWAustralia
- School of Biomedical Sciences and PharmacyFaculty of Health and MedicineUniversity of NewcastleCallaghanNSWAustralia
- School of Life SciencesFaculty of ScienceUniversity of Technology SydneySydneyNSWAustralia
- Centenary InstituteCentre for InflammationUniversity of Technology SydneySydneyNSWAustralia
| |
Collapse
|
31
|
CXCL4 is a driver of cytokine mRNA stability in monocyte-derived dendritic cells. Mol Immunol 2019; 114:524-534. [DOI: 10.1016/j.molimm.2019.09.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2019] [Revised: 08/16/2019] [Accepted: 09/04/2019] [Indexed: 12/12/2022]
|
32
|
Clark AR, Ohlmeyer M. Protein phosphatase 2A as a therapeutic target in inflammation and neurodegeneration. Pharmacol Ther 2019; 201:181-201. [PMID: 31158394 PMCID: PMC6700395 DOI: 10.1016/j.pharmthera.2019.05.016] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 05/29/2019] [Indexed: 12/11/2022]
Abstract
Protein phosphatase 2A (PP2A) is a highly complex heterotrimeric enzyme that catalyzes the selective removal of phosphate groups from protein serine and threonine residues. Emerging evidence suggests that it functions as a tumor suppressor by constraining phosphorylation-dependent signalling pathways that regulate cellular transformation and metastasis. Therefore, PP2A-activating drugs (PADs) are being actively sought and investigated as potential novel anti-cancer treatments. Here we explore the concept that PP2A also constrains inflammatory responses through its inhibitory effects on various signalling pathways, suggesting that PADs may be effective in the treatment of inflammation-mediated pathologies.
Collapse
Affiliation(s)
- Andrew R Clark
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, B15 2TT, United Kingdom.
| | | |
Collapse
|
33
|
Yoshinaga M, Takeuchi O. RNA binding proteins in the control of autoimmune diseases. Immunol Med 2019; 42:53-64. [DOI: 10.1080/25785826.2019.1655192] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Affiliation(s)
- Masanori Yoshinaga
- Department of Medical Chemistry, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Osamu Takeuchi
- Department of Medical Chemistry, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
34
|
Normal Saline solutions cause endothelial dysfunction through loss of membrane integrity, ATP release, and inflammatory responses mediated by P2X7R/p38 MAPK/MK2 signaling pathways. PLoS One 2019; 14:e0220893. [PMID: 31412063 PMCID: PMC6693757 DOI: 10.1371/journal.pone.0220893] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 07/25/2019] [Indexed: 12/12/2022] Open
Abstract
Resuscitation with 0.9% Normal Saline (NS), a non-buffered acidic solution, leads to increased morbidity and mortality in the critically ill. The goal of this study was to determine the molecular mechanisms of endothelial injury after exposure to NS. The hypothesis of this investigation is that exposure of endothelium to NS would lead to loss of cell membrane integrity, resulting in release of ATP, activation of the purinergic receptor (P2X7R), and subsequent activation of stress activated signaling pathways and inflammation. Human saphenous vein endothelial cells (HSVEC) incubated in NS, but not buffered electrolyte solution (Plasma-Lyte, PL), exhibited abnormal morphology and increased release of lactate dehydrogenase (LDH), adenosine triphosphate (ATP), and decreased transendothelial resistance (TEER), suggesting loss of membrane integrity. Incubation of intact rat aorta (RA) or human saphenous vein in NS but not PL led to impaired endothelial-dependent relaxation which was ameliorated by apyrase (hydrolyzes ATP) or SB203580 (p38 MAPK inhibitor). Exposure of HSVEC to NS but not PL led to activation of p38 MAPK and its downstream substrate, MAPKAP kinase 2 (MK2). Treatment of HSVEC with exogenous ATP led to interleukin 1β (IL-1β) release and increased vascular cell adhesion molecule (VCAM) expression. Treatment of RA with IL-1β led to impaired endothelial relaxation. IL-1β treatment of HSVEC led to increases in p38 MAPK and MK2 phosphorylation, and increased levels of arginase II. Incubation of porcine saphenous vein (PSV) in PL with pH adjusted to 6.0 or less also led to impaired endothelial function, suggesting that the acidic nature of NS is what contributes to endothelial dysfunction. Volume overload resuscitation in a porcine model after hemorrhage with NS, but not PL, led to acidosis and impaired endothelial function. These data suggest that endothelial dysfunction caused by exposure to acidic, non-buffered NS is associated with loss of membrane integrity, release of ATP, and is modulated by P2X7R-mediated inflammatory responses.
Collapse
|
35
|
Lai WS, Stumpo DJ, Wells ML, Gruzdev A, Hicks SN, Nicholson CO, Yang Z, Faccio R, Webster MW, Passmore LA, Blackshear PJ. Importance of the Conserved Carboxyl-Terminal CNOT1 Binding Domain to Tristetraprolin Activity In Vivo. Mol Cell Biol 2019; 39:e00029-19. [PMID: 31036567 PMCID: PMC6580703 DOI: 10.1128/mcb.00029-19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 02/12/2019] [Accepted: 04/19/2019] [Indexed: 01/19/2023] Open
Abstract
Tristetraprolin (TTP) is an anti-inflammatory protein that modulates the stability of certain cytokine/chemokine mRNAs. After initial high-affinity binding to AU-rich elements in 3' untranslated regions of target mRNAs, mediated through its tandem zinc finger (TZF) domain, TTP promotes the deadenylation and ultimate decay of target transcripts. These transcripts and their encoded proteins accumulate abnormally in TTP knockout (KO) mice, leading to a severe inflammatory syndrome. To assess the importance of the highly conserved C-terminal CNOT1 binding domain (CNBD) of TTP to the TTP deficiency phenotype in mice, we created a mouse model in which TTP lacked its CNBD. CNBD deletion mice exhibited a less severe phenotype than the complete TTP KO mice. In macrophages, the stabilization of target transcripts seen in KO mice was partially normalized in the CNBD deletion mice. In cell-free experiments, recombinant TTP lacking its CNBD could still activate target mRNA deadenylation by purified recombinant Schizosaccharomyces pombe CCR4/NOT complexes, although to a lesser extent than full-length TTP. Thus, TTP lacking its CNBD can still act to promote target mRNA instability in vitro and in vivo These data have implications for TTP family members throughout the eukarya, since species from all four kingdoms contain proteins with linked TZF and CNOT1 binding domains.
Collapse
Affiliation(s)
- Wi S Lai
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Deborah J Stumpo
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Melissa L Wells
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Artiom Gruzdev
- Reproductive & Developmental Biology Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Stephanie N Hicks
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Cindo O Nicholson
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Zhengfeng Yang
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- Shriners Hospitals for Children, St. Louis, Missouri, USA
| | - Roberta Faccio
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, Missouri, USA
- Shriners Hospitals for Children, St. Louis, Missouri, USA
| | | | - Lori A Passmore
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - Perry J Blackshear
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
- Departments of Medicine and Biochemistry, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
36
|
Li Y, Wang W, Chao Y, Zhang F, Wang C. CTRP13 attenuates vascular calcification by regulating Runx2. FASEB J 2019; 33:9627-9637. [PMID: 31145871 DOI: 10.1096/fj.201900293rrr] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Vascular calcification is strongly associated with increased cardiovascular mortality and morbidity. C1q/TNF-related protein-13 (CTRP13) is a secreted adipokine that plays important roles in the cardiovascular system. However, the functional role of CTRP13 in the development of vascular calcification has yet to be explored. In this study, we collected blood samples from patients with chronic renal failure (CRF) and from rats with adenine-induced CRF. We found that the serum CTRP13 levels were decreased in patients and rats with CRF and were negatively associated with calcium deposition in the abdominal aorta. Compared to those of the controls, ectopic CTRP13 treatment significantly attenuated the calcium accumulation and alkaline phosphatase activity in the abdominal aorta of CRF rats, and β-glycerophosphate induced the formation of arterial rings and of vascular smooth muscle cells (VSMCs) and decreased the number of VSMCs that transitioned from a contractile to an osteogenic phenotype. The overexpression of Runx2 blocked CTRP13-reduced VSMC calcification. Mechanistically, CTRP13 repressed the phosphorylation of tristetraprolin (TTP), thereby activating TTP and increasing the TTP binding to the 3'untranslated region of the Runx2 mRNA, accelerating the Runx2 mRNA destabilization and degradation. In summary, these findings reveal that CTRP13 regulation is a novel method for the prevention of vascular calcification, representing a novel mechanism of the regulation of Runx2 expression in VSMCs.-Li, Y., Wang, W., Chao, Y., Zhang, F., Wang, C. CTRP13 attenuates vascular calcification by regulating Runx2.
Collapse
Affiliation(s)
- Yongxia Li
- Department of Nephrology, The Central Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan, China
| | - Wenzhe Wang
- Department of Nephrology, The Central Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan, China
| | - Yuelin Chao
- Department of Cardiology, Nanjing First Hospital, Nanjing Medical University, Nanjing, China
| | - Fengxao Zhang
- Department of Nephrology, The Central Hospital of Wuhan, Huazhong University of Science and Technology, Wuhan, China
| | - Cheng Wang
- Department of Rheumatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
37
|
Otsuka H, Fukao A, Funakami Y, Duncan KE, Fujiwara T. Emerging Evidence of Translational Control by AU-Rich Element-Binding Proteins. Front Genet 2019; 10:332. [PMID: 31118942 PMCID: PMC6507484 DOI: 10.3389/fgene.2019.00332] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Accepted: 03/28/2019] [Indexed: 12/27/2022] Open
Abstract
RNA-binding proteins (RBPs) are key regulators of posttranscriptional gene expression and control many important biological processes including cell proliferation, development, and differentiation. RBPs bind specific motifs in their target mRNAs and regulate mRNA fate at many steps. The AU-rich element (ARE) is one of the major cis-regulatory elements in the 3′ untranslated region (UTR) of labile mRNAs. Many of these encode factors requiring very tight regulation, such as inflammatory cytokines and growth factors. Disruption in the control of these factors’ expression can cause autoimmune diseases, developmental disorders, or cancers. Therefore, these mRNAs are strictly regulated by various RBPs, particularly ARE-binding proteins (ARE-BPs). To regulate mRNA metabolism, ARE-BPs bind target mRNAs and affect some factors on mRNAs directly, or recruit effectors, such as mRNA decay machinery and protein kinases to target mRNAs. Importantly, some ARE-BPs have stabilizing roles, whereas others are destabilizing, and ARE-BPs appear to compete with each other when binding to target mRNAs. The function of specific ARE-BPs is modulated by posttranslational modifications (PTMs) including methylation and phosphorylation, thereby providing a means for cellular signaling pathways to regulate stability of specific target mRNAs. In this review, we summarize recent studies which have revealed detailed molecular mechanisms of ARE-BP-mediated regulation of gene expression and also report on the importance of ARE-BP function in specific physiological contexts and how this relates to disease. We also propose an mRNP regulatory network based on competition between stabilizing ARE-BPs and destabilizing ARE-BPs.
Collapse
Affiliation(s)
- Hiroshi Otsuka
- Graduate School of Frontier Sciences, University of Tokyo, Kashiwa, Japan
| | | | | | - Kent E Duncan
- University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | |
Collapse
|
38
|
Villaseñor T, Madrid-Paulino E, Maldonado-Bravo R, Pérez-Martínez L, Pedraza-Alva G. Mycobacterium bovis BCG promotes IL-10 expression by establishing a SYK/PKCα/β positive autoregulatory loop that sustains STAT3 activation. Pathog Dis 2019; 77:5512589. [PMID: 31175361 DOI: 10.1093/femspd/ftz032] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 06/05/2019] [Indexed: 01/02/2023] Open
Abstract
Mycobacterium ensures its survival inside macrophages and long-term infection by subverting the innate and adaptive immune response through the modulation of cytokine gene expression profiles. Different Mycobacterium species promote the expression of TGFβ and IL-10, which, at the early stages of infection, block the formation of the phagolysosome, thereby securing mycobacterial survival upon phagocytosis, and at later stages, antagonize IFNγ production and functions. Despite the key role of IL-10 in mycobacterium infection, the signal transduction pathways leading to IL-10 expression in infected macrophages are poorly understood. Here, we report that Mycobacterium bovis BCG promotes IL-10 expression and cytokine production by establishing a SYK/PKCα/β positive feedback loop that leads to STAT3 activation.
Collapse
Affiliation(s)
- Tomás Villaseñor
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos 62271, México
| | - Edgardo Madrid-Paulino
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos 62271, México
| | - Rafael Maldonado-Bravo
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos 62271, México
| | - Leonor Pérez-Martínez
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos 62271, México
| | - Gustavo Pedraza-Alva
- Laboratorio de Neuroinmunobiología, Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México (UNAM), Cuernavaca, Morelos 62271, México
| |
Collapse
|
39
|
Mansueto MS, Reens A, Rakhilina L, Chi A, Pan BS, Miller JR. A reevaluation of the spleen tyrosine kinase (SYK) activation mechanism. J Biol Chem 2019; 294:7658-7668. [PMID: 30923129 DOI: 10.1074/jbc.ra119.008045] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 03/25/2019] [Indexed: 11/06/2022] Open
Abstract
Spleen tyrosine kinase (SYK) is a signaling node in many immune pathways and comprises two tandem Src homology (SH) 2 domains, an SH2-kinase linker, and a C-terminal tyrosine kinase domain. Two prevalent models of SYK activation exist. The "OR-gate" model contends that SYK can be fully activated by phosphorylation or binding of its SH2 domains to a dual-phosphorylated immune-receptor tyrosine-based activation motif (ppITAM). An alternative model proposes that SYK activation requires ppITAM binding and phosphorylation of the SH2-kinase linker by a SRC family kinase such as LYN proto-oncogene, SRC family tyrosine kinase (LYN). To evaluate these two models, we generated directly comparable unphosphorylated (upSYK) and phosphorylated (pSYK) proteins with or without an N-terminal glutathione S-transferase (GST) tag, resulting in monomeric or obligatory dimeric SYK, respectively. We assessed the ability of a ppITAM peptide and LYN to activate these SYK proteins. The ppITAM peptide strongly activated GST-SYK but was less effective in activating upSYK untagged with GST. LYN alone activated untagged upSYK to a greater extent than did ppITAM, and inclusion of both proteins rapidly and fully activated upSYK. Using immunoblot and phosphoproteomic approaches, we correlated the kinetics and order of site-specific SYK phosphorylation. Our results are consistent with the alternative model, indicating that ppITAM binding primes SYK for rapid LYN-mediated phosphorylation of Tyr-352 and then Tyr-348 of the SH2-kinase linker, which facilitates activation loop phosphorylation and full SYK activation. This gradual activation mechanism may also explain how SYK maintains ligand-independent tonic signaling, important for B-cell development and survival.
Collapse
Affiliation(s)
| | | | | | - An Chi
- Chemical Biology, Merck & Co., Inc., Boston, Massachusetts 02115
| | | | | |
Collapse
|
40
|
Cardiovascular inflammation: RNA takes the lead. J Mol Cell Cardiol 2019; 129:247-256. [PMID: 30880251 DOI: 10.1016/j.yjmcc.2019.03.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 12/19/2022]
Abstract
Inflammation has recently gained tremendous attention as a key contributor in several chronic diseases. While physiological inflammation is essential to counter a wide variety of damaging stimuli and to improve wound healing, dysregulated inflammation such as in the myocardium and vasculature can promote cardiovascular diseases. Given the high severity, prevalence, and economic burden of these diseases, understanding the factors involved in the regulation of physiological inflammation is essential. Like other complex biological phenomena, RNA-based processes are emerging as major regulators of inflammatory responses. Among such processes are cis-regulatory elements in the mRNA of inflammatory genes, noncoding RNAs directing the production or localization of inflammatory cytokines/chemokines, or pathogenic RNA driving inflammatory responses. In this review, we describe several specific RNA-based molecular mechanisms by which physiological inflammation pertaining to cardiovascular diseases is regulated. These include the role of AU-rich element-containing mRNAs, long non-coding RNAs, microRNAs, and viral RNAs.
Collapse
|
41
|
Soni S, Anand P, Padwad YS. MAPKAPK2: the master regulator of RNA-binding proteins modulates transcript stability and tumor progression. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:121. [PMID: 30850014 PMCID: PMC6408796 DOI: 10.1186/s13046-019-1115-1] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 02/21/2019] [Indexed: 01/09/2023]
Abstract
The p38 mitogen-activated protein kinase (p38MAPK) pathway has been implicated in a variety of pathological conditions including inflammation and metastasis. Post-transcriptional regulation of genes harboring adenine/uridine-rich elements (AREs) in their 3'-untranslated region (3'-UTR) is controlled by MAPK-activated protein kinase 2 (MAPKAPK2 or MK2), a downstream substrate of the p38MAPK. In response to diverse extracellular stimuli, MK2 influences crucial signaling events, regulates inflammatory cytokines, transcript stability and critical cellular processes. Expression of genes involved in these vital cellular cascades is controlled by subtle interactions in underlying molecular networks and post-transcriptional gene regulation that determines transcript fate in association with RNA-binding proteins (RBPs). Several RBPs associate with the 3'-UTRs of the target transcripts and regulate their expression via modulation of transcript stability. Although MK2 regulates important cellular phenomenon, yet its biological significance in tumor progression has not been well elucidated till date. In this review, we have highlighted in detail the importance of MK2 as the master regulator of RBPs and its role in the regulation of transcript stability, tumor progression, as well as the possibility of use of MK2 as a therapeutic target in tumor management.
Collapse
Affiliation(s)
- Sourabh Soni
- Pharmacology and Toxicology Laboratory, Food and Nutraceuticals Division, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, Himachal Pradesh, India.,Academy of Scientific and Innovative Research, Chennai, Tamil Nadu, India
| | - Prince Anand
- Pharmacology and Toxicology Laboratory, Food and Nutraceuticals Division, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, Himachal Pradesh, India.,Academy of Scientific and Innovative Research, Chennai, Tamil Nadu, India
| | - Yogendra S Padwad
- Pharmacology and Toxicology Laboratory, Food and Nutraceuticals Division, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur, Himachal Pradesh, India. .,Academy of Scientific and Innovative Research, Chennai, Tamil Nadu, India.
| |
Collapse
|
42
|
Ostareck DH, Ostareck-Lederer A. RNA-Binding Proteins in the Control of LPS-Induced Macrophage Response. Front Genet 2019; 10:31. [PMID: 30778370 PMCID: PMC6369361 DOI: 10.3389/fgene.2019.00031] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Accepted: 01/17/2019] [Indexed: 12/18/2022] Open
Abstract
Innate immune response is triggered by pathogen components, like lipopolysaccharides (LPS) of gram-negative bacteria. LPS initiates Toll-like receptor 4 (TLR4) signaling, which involves mitogen activated protein kinases (MAPK) and nuclear factor kappa B (NFκB) in different pathway branches and ultimately induces inflammatory cytokine and chemokine expression, macrophage migration and phagocytosis. Timely gene transcription and post-transcriptional control of gene expression confer the adequate synthesis of signaling molecules. As trans-acting factors RNA binding proteins (RBPs) contribute significantly to the surveillance of gene expression. RBPs are involved in the regulation of mRNA processing, localization, stability and translation. Thereby they enable rapid cellular responses to inflammatory mediators and facilitate a coordinated systemic immune response. Specific RBP binding to conserved sequence motifs in their target mRNAs is mediated by RNA binding domains, like Zink-finger domains, RNA recognition motifs (RRM), and hnRNP K homology domains (KH), often arranged in modular arrays. In this review, we focus on RBPs Tristetraprolin (TTP), human antigen R (HUR), T-cell intracellular antigen 1 related protein (TIAR), and heterogeneous ribonuclear protein K (hnRNP K) in LPS induced macrophages as primary responding immune cells. We discuss recent experiments employing RNA immunoprecipitation and microarray analysis (RIP-Chip) and newly developed individual-nucleotide resolution crosslinking and immunoprecipitation (iCLIP), photoactivatable ribonucleoside-enhanced crosslinking (PAR-iCLIP) and RNA sequencing techniques (RNA-Seq). The global mRNA interaction profile analysis of TTP, HUR, TIAR, and hnRNP K exhibited valuable information about the post-transcriptional control of inflammation related gene expression with a broad impact on intracellular signaling and temporal cytokine expression.
Collapse
Affiliation(s)
- Dirk H Ostareck
- Department of Intensive Care Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | | |
Collapse
|
43
|
Mahmoud L, Moghrabi W, Khabar KSA, Hitti EG. Bi-phased regulation of the post-transcriptional inflammatory response by Tristetraprolin levels. RNA Biol 2019; 16:309-319. [PMID: 30664390 PMCID: PMC6380337 DOI: 10.1080/15476286.2019.1572437] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
AU-rich elements (AREs) are cis-acting instability and translation inhibition elements that are present in the 3ʹUTR of most inducible inflammatory mRNAs such as TNF and Cxcl2. mRNAs that contain AREs are, by default, repressed and only transiently expressed in response to stimuli. They are targeted by the inducible RNA-binding protein Tristetraprolin (TTP) which blocks their translation and facilitates their decay, thereby contributing to the quick termination of their expression. The exogenous over-expression of TTP in HEK293 cells can unexpectedly lead to the upregulation and extended expression of a nanoLuciferase reporter that contains the ARE of TNF. Here we show that, a moderate downregulation of the highly expressed endogenous TTP after LPS induction by siRNA in macrophages can lead to a reduction in the release of TNF and Cxcl2. We propose that, in contrast to their canonical function, very high levels of induced TTP at the onset of the inflammatory response can enhance the expression of ARE-mRNAs at the post-transcriptional level, independently of phosphorylation status. As the inflammatory response progresses, TTP levels diminish but they continuously regain their ability to reduce the expression of ARE-mRNAs to reach a turning point of ‘optimal TTP level’ with a maximum ability to repress ARE-mRNA expression. Below this level, a further reduction in TTP levels now leads to the loss of canonical-TTP function resulting in increased ARE-mRNA expression. These novel findings should contribute to the understanding of feedback loops that control the kinetics of the inflammatory response.
Collapse
Affiliation(s)
- Linah Mahmoud
- a Molecular BioMedicine Program , King Faisal Specialist Hospital & Research Centre , Riyadh , Saudi Arabia
| | - Walid Moghrabi
- a Molecular BioMedicine Program , King Faisal Specialist Hospital & Research Centre , Riyadh , Saudi Arabia
| | - Khalid S A Khabar
- a Molecular BioMedicine Program , King Faisal Specialist Hospital & Research Centre , Riyadh , Saudi Arabia
| | - Edward G Hitti
- a Molecular BioMedicine Program , King Faisal Specialist Hospital & Research Centre , Riyadh , Saudi Arabia
| |
Collapse
|
44
|
Liu Y, Wei W, Hong C, Wang Y, Sun X, Ma J, Zheng F. Calreticulin induced endothelial ICAM-1 up-regulation associated with tristetraprolin expression alteration through PI3K/Akt/eNOS/p38 MAPK signaling pathway in rheumatoid arthritis. Mol Immunol 2019; 107:10-20. [PMID: 30639474 DOI: 10.1016/j.molimm.2019.01.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Revised: 01/02/2019] [Accepted: 01/04/2019] [Indexed: 12/17/2022]
Abstract
The present study was undertaken to determine whether extracellular calreticulin (CRT) participates in the regulation of ICAM-1in rheumatoid arthritis (RA) and further explore the potential mechanism. Our results showed that ICAM-1 and VCAM-1 levels were positively correlated with CRT levels in RA serum and synovial fluid, respectively. In RA synovial tissue, increased co-expressions of CRT and ICAM-1 in vascular endothelium and perivascular areas and elevated co-location of CRT and VCAM-1 localized predominantly to lining layer were observed compared to those in OA. In in vitro HUVECs model, enhanced ICAM-1expression and increased phosphorylation levels of Akt and eNOS were detected in the presence of CRT. Increased phosphorylated eNOS was significantly inhibited by a PI3K inhibitor LY294002 and elevated ICAM-1expression was partially blocked by the inhibitors of both PI3K and eNOS (L-NAME). It has been certified that the RNA-binding protein TTP targets AU-rich elements in the ICAM-1 3'-UTR and suppresses ICAM-1 expression. Knocking down TTP in HUVECs led to an increased induction of ICAM-1 by CRT. We have currently known that activation of p38 downstream kinase MK-2 leads to phosphorylation and inactivation of human TTP. The block of p38 MAPK/MK-2 signaling led to decreased protein expression and mRNA stability of TTP and ICAM-1. Furthermore, L-NAME and/or LY294002 pre-treated HUVECs manifested decreased p38 and MK-2 phosphorylation, which was accompanied by reduced TTP and ICAM-1 protein expression as well as decreased mRNA stability. Our results suggested that CRT could promote ICAM-1 expression in endothelial cells through PI3K/Akt/eNOS/p38 MAPK signaling mediated TTP accumulation, probably in an inactive form, which may provide a possible proinflammatory mechanism of CRT in RA.
Collapse
Affiliation(s)
- Yixin Liu
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin 300203, China
| | - Wei Wei
- Department of Rheumatology, General Hospital, Tianjin Medical University, Tianjin 300052, China
| | - Chengcheng Hong
- Department of Laboratory Medicine, Children's Hospital of Tianjin, Tianjin 300203, China
| | - Yang Wang
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin 300203, China
| | - Xuguo Sun
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin 300203, China
| | - Jun Ma
- Department of Health Statistics, College of Public Health, Tianjin Medical University, Tianjin 300070, China.
| | - Fang Zheng
- Department of Clinical Immunology, School of Medical Laboratory, Tianjin Medical University, Tianjin 300203, China.
| |
Collapse
|
45
|
Yamasaki S. Recent advances in the role of RNA-binding protein, tristetraprolin, in arthritis. Immunol Med 2018; 41:98-102. [PMID: 30938272 DOI: 10.1080/25785826.2018.1531187] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
The expression levels of cytokines and chemokines are strictly regulated at transcriptional and post-transcriptional levels. These small proteins are closely related to inflammatory diseases such as rheumatoid arthritis (RA). The purpose of this review is to highlight the potential utilization of tristetraprolin (TTP) as a therapeutic target in treating RA. TTP is the most notable and well-characterized RNA-binding protein that destabilizes mRNA of pro-inflammatory cytokines. TTP is thought to play an important role in RA because its target mRNA includes a lot of inflammatory cytokines such as TNFα. Post-translational modifications, especially phosphorylation, seem to be critical for the anti-inflammatory effects of TTP. Importantly, various mouse models, many of which are consistent with in vitro studies, are now available to elicit a more detailed understanding of the pathogenic role of TTP. The results of these multidisciplinary studies indicate that it is possible to improve inflammation by controlling TTP activity. Through this review, I propose that the use of recently developed mouse models and establishment of clever designs to target TTP will greatly contribute to future drug development to treat RA.
Collapse
Affiliation(s)
- Satoshi Yamasaki
- a Division of Rheumatology, Kurume University Medical Center , Fukuoka , Japan
| |
Collapse
|
46
|
Kondo M, Noguchi A, Matsuura Y, Shimada M, Yokota N, Kawahara H. Novel phosphorelay-dependent control of ZFP36L1 protein during the cell cycle. Biochem Biophys Res Commun 2018; 501:387-393. [DOI: 10.1016/j.bbrc.2018.04.212] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 04/26/2018] [Indexed: 10/16/2022]
|
47
|
Qi MY, Song JW, Zhang Z, Huang S, Jing Q. P38 activation induces the dissociation of tristetraprolin from Argonaute 2 to increase ARE-mRNA stabilization. Mol Biol Cell 2018; 29:988-1002. [PMID: 29444957 PMCID: PMC5896936 DOI: 10.1091/mbc.e17-02-0105] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
ARE-mRNAs are actively degraded with tristetraprolin (TTP) in resting cells while they turn into stable messengers in activated cells. P38 plays a crucial role in stabilizing ARE-mRNA. Here we reveal that P38 activation represses the interaction between TTP and Ago2, thus restraining TTP from being targeted into processing bodies and stabilizing ARE-mRNA.
Collapse
Affiliation(s)
- Mei-Yan Qi
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jing-Wen Song
- Department of Cardiology, Changhai Hospital, Shanghai 200433, China
| | - Zhuo Zhang
- Department of Cardiology, Changhai Hospital, Shanghai 200433, China.,Synthetic Biology and Biotechnology Laboratory, State Key Laboratory of Bioreactor Engineering, Shanghai Collaborative Innovation Center for Biomanufacturing Technology, East China University of Science and Technology, Shanghai 200237, China
| | - Shuang Huang
- Department of Cardiology, Changhai Hospital, Shanghai 200433, China
| | - Qing Jing
- Key Laboratory of Stem Cell Biology, Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China.,Department of Cardiology, Changhai Hospital, Shanghai 200433, China
| |
Collapse
|
48
|
Steinkamp HM, Hathaway-Schrader JD, Chavez MB, Aartun JD, Zhang L, Jensen T, Shojaee Bakhtiari A, Helke KL, Stumpo DJ, Alekseyenko AV, Novince CM, Blackshear PJ, Kirkwood KL. Tristetraprolin Is Required for Alveolar Bone Homeostasis. J Dent Res 2018. [PMID: 29514008 DOI: 10.1177/0022034518756889] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Tristetraprolin (TTP) is an RNA-binding protein that targets numerous immunomodulatory mRNA transcripts for degradation. Many TTP targets are key players in the pathogenesis of periodontal bone loss, including tumor necrosis factor-α. To better understand the extent that host immune factors play during periodontal bone loss, we assessed alveolar bone levels, inflammation and osteoclast activity in periodontal tissues, and immune response in draining cervical lymph nodes in TTP-deficient and wild-type (WT) mice in an aging study. WT and TTP-deficient (knockout [KO]) mice were used for all studies under specific pathogen-free conditions. Data were collected on mice aged 3, 6, and 9 mo. Microcomputed tomography (µCT) was performed on maxillae where 3-dimensional images were generated and bone loss was assessed. Decalcified sections of specimens were scored for inflammation and stained with tartrate-resistant acid phosphate (TRAP) to visualize osteoclasts. Immunophenotyping was performed on single-cell suspensions isolated from primary and peripheral lymphoid tissues using flow cytometry. Results presented indicate that TTP KO mice had significantly more alveolar bone loss over time compared with WT controls. Bone loss was associated with significant increases in inflammatory cell infiltration and an increased percentage of alveolar bone surfaces apposed with TRAP+ cells. Furthermore, it was found that the draining cervical lymph nodes were significantly enlarged in TTP-deficient animals and contained a distinct pathological immune profile compared with WT controls. Finally, the oral microbiome in the TTP KO mice was significantly different with age from WT cohoused mice. The severe bone loss, inflammation, and increased osteoclast activity observed in these mice support the concept that TTP plays a critical role in the maintenance of alveolar bone homeostasis in the presence of oral commensal flora. This study suggests that TTP is required to inhibit excessive inflammatory host responses that contribute to periodontal bone loss, even in the absence of specific periodontal pathogens.
Collapse
Affiliation(s)
- H M Steinkamp
- 1 Department of Oral Health Sciences and Center for Oral Health Research, Medical University of South Carolina, Charleston, SC, USA
| | - J D Hathaway-Schrader
- 1 Department of Oral Health Sciences and Center for Oral Health Research, Medical University of South Carolina, Charleston, SC, USA
| | - M B Chavez
- 1 Department of Oral Health Sciences and Center for Oral Health Research, Medical University of South Carolina, Charleston, SC, USA
| | - J D Aartun
- 1 Department of Oral Health Sciences and Center for Oral Health Research, Medical University of South Carolina, Charleston, SC, USA
| | - L Zhang
- 1 Department of Oral Health Sciences and Center for Oral Health Research, Medical University of South Carolina, Charleston, SC, USA.,2 Department of Oral Biology, University at Buffalo, State University of New York, Buffalo, NY, USA
| | - T Jensen
- 1 Department of Oral Health Sciences and Center for Oral Health Research, Medical University of South Carolina, Charleston, SC, USA
| | - A Shojaee Bakhtiari
- 3 Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - K L Helke
- 4 Department of Comparative Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - D J Stumpo
- 3 Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - A V Alekseyenko
- 1 Department of Oral Health Sciences and Center for Oral Health Research, Medical University of South Carolina, Charleston, SC, USA.,3 Department of Public Health Sciences, Medical University of South Carolina, Charleston, SC, USA
| | - C M Novince
- 1 Department of Oral Health Sciences and Center for Oral Health Research, Medical University of South Carolina, Charleston, SC, USA
| | - P J Blackshear
- 5 Signal Transduction Laboratory, National Institute of Environmental Health Sciences, Research Triangle Park, Departments of Biochemistry & Medicine, Duke University Medical Center, Durham, NC, USA
| | - K L Kirkwood
- 2 Department of Oral Biology, University at Buffalo, State University of New York, Buffalo, NY, USA
| |
Collapse
|
49
|
MK2–TNF–Signaling Comes Full Circle. Trends Biochem Sci 2018; 43:170-179. [DOI: 10.1016/j.tibs.2017.12.002] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 12/01/2017] [Accepted: 12/04/2017] [Indexed: 12/27/2022]
|
50
|
Zhang X, Xiao S, Rameau RD, Devany E, Nadeem Z, Caglar E, Ng K, Kleiman FE, Saxena A. Nucleolin phosphorylation regulates PARN deadenylase activity during cellular stress response. RNA Biol 2018; 15:251-260. [PMID: 29168431 PMCID: PMC5798948 DOI: 10.1080/15476286.2017.1408764] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 09/11/2017] [Accepted: 11/16/2017] [Indexed: 01/28/2023] Open
Abstract
Nucleolin (NCL) is an abundant stress-responsive, RNA-binding phosphoprotein that controls gene expression by regulating either mRNA stability and/or translation. NCL binds to the AU-rich element (ARE) in the 3'UTR of target mRNAs, mediates miRNA functions in the nearby target sequences, and regulates mRNA deadenylation. However, the mechanism by which NCL phosphorylation affects these functions and the identity of the deadenylase involved, remain largely unexplored. Earlier we demonstrated that NCL phosphorylation is vital for cell cycle progression and proliferation, whereas phosphorylation-deficient NCL at six consensus CK2 sites confers dominant-negative effect on proliferation by increasing p53 expression, possibly mimicking cellular DNA damage conditions. In this study, we show that NCL phosphorylation at those CK2 consensus sites in the N-terminus is necessary to induce deadenylation upon oncogenic stimuli and UV stress. NCL-WT, but not hypophosphorylated NCL-6/S*A, activates poly (A)-specific ribonuclease (PARN) deadenylase activity. We further demonstrate that NCL interacts directly with PARN, and under non-stress conditions also forms (a) complex (es) with factors that regulate deadenylation, such as p53 and the ARE-binding protein HuR. Upon UV stress, the interaction of hypophosphorylated NCL-6/S*A with these proteins is favored. As an RNA-binding protein, NCL interacts with PARN deadenylase substrates such as TP53 and BCL2 mRNAs, playing a role in their downregulation under non-stress conditions. For the first time, we show that NCL phosphorylation offers specificity to its protein-protein, protein-RNA interactions, resulting in the PARN deadenylase regulation, and hence gene expression, during cellular stress responses.
Collapse
Affiliation(s)
- Xiaokan Zhang
- Chemistry Department, Hunter College, New York, NY, USA
| | - Shu Xiao
- Biology Department, Brooklyn College, Brooklyn, NY, USA
| | | | - Emral Devany
- Chemistry Department, Hunter College, New York, NY, USA
| | - Zaineb Nadeem
- Biology Department, Brooklyn College, Brooklyn, NY, USA
| | - Elif Caglar
- Biology Department, Brooklyn College, Brooklyn, NY, USA
| | - Kenneth Ng
- Biology Department, Brooklyn College, Brooklyn, NY, USA
| | | | - Anjana Saxena
- Biology Department, Brooklyn College, Brooklyn, NY, USA
| |
Collapse
|