1
|
Pommier A, Bleuse S, Deletang K, Varilh J, Nadaud M, Boisguerin P, Bourdin A, Taulan-Cadars M. The RNA-Binding Protein Tristetraprolin Contributes to CFTR mRNA Stability in Cystic Fibrosis. Am J Respir Cell Mol Biol 2025; 72:320-331. [PMID: 39417720 DOI: 10.1165/rcmb.2023-0209oc] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 10/17/2024] [Indexed: 10/19/2024] Open
Abstract
Cystic fibrosis (CF) is the most common inherited disorder and is characterized by an inflammatory phenotype. We found that in bronchial epithelium reconstituted form lung tissue biopsies from patients with CF, the RNA-binding protein tristetraprolin (TTP), a key regulator of inflammation, is dysregulated in cells that strongly express cytokines and ILs. TTP activity is regulated by extensive posttranslational modifications, particularly phosphorylation. We found that, in addition to mRNA downregulation, phosphorylated TTP (which cannot bind to mRNA) accumulated in CF cultures, suggesting that the imbalance in TTP phosphorylation status could contribute to the inflammatory phenotype in CF. We confirmed TTP's destabilizing role on IL8 mRNA through its 3' UTR sequence in CF cells. We next demonstrated that TTP phosphorylation is mainly regulated by MK2 through the activation of ERK, which also was hyperphosphorylated. TTP is considered a mRNA decay factor with some exception, and we present a new positive role of TTP in CF cultures. We determined that TTP binds to specific adenylate-uridylate-rich element motifs on the 3' UTR of mRNA sequences and also, for the first time to our knowledge, to the 3' UTR of the cystic fibrosis transmembrane conductance regulator (CFTR), where TTP binding stabilizes the mRNA level. This study identified new partners that can be targeted in CF and proposes a new way to control CFTR gene expression.
Collapse
Affiliation(s)
| | - Solenne Bleuse
- Université de Montpellier, and
- PhyMedExp, INSERM U1046, CNRS UMR 9214, Université de Montpellier, Montpellier, France; and
| | - Karine Deletang
- PhyMedExp, INSERM U1046, CNRS UMR 9214, Université de Montpellier, Montpellier, France; and
| | - Jessica Varilh
- PhyMedExp, INSERM U1046, CNRS UMR 9214, Université de Montpellier, Montpellier, France; and
| | - Marion Nadaud
- PhyMedExp, INSERM U1046, CNRS UMR 9214, Université de Montpellier, Montpellier, France; and
| | - Prisca Boisguerin
- PhyMedExp, INSERM U1046, CNRS UMR 9214, Université de Montpellier, Montpellier, France; and
| | - Arnaud Bourdin
- Université de Montpellier, and
- PhyMedExp, INSERM U1046, CNRS UMR 9214, Université de Montpellier, Montpellier, France; and
- Department of Respiratory Diseases, CHU Arnaud de Villeneuve, Montpellier, France
| | - Magali Taulan-Cadars
- Université de Montpellier, and
- PhyMedExp, INSERM U1046, CNRS UMR 9214, Université de Montpellier, Montpellier, France; and
| |
Collapse
|
2
|
Singh K, Oladipupo SS. An overview of CCN4 (WISP1) role in human diseases. J Transl Med 2024; 22:601. [PMID: 38937782 PMCID: PMC11212430 DOI: 10.1186/s12967-024-05364-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 06/01/2024] [Indexed: 06/29/2024] Open
Abstract
CCN4 (cellular communication network factor 4), a highly conserved, secreted cysteine-rich matricellular protein is emerging as a key player in the development and progression of numerous disease pathologies, including cancer, fibrosis, metabolic and inflammatory disorders. Over the past two decades, extensive research on CCN4 and its family members uncovered their diverse cellular mechanisms and biological functions, including but not limited to cell proliferation, migration, invasion, angiogenesis, wound healing, repair, and apoptosis. Recent studies have demonstrated that aberrant CCN4 expression and/or associated downstream signaling is key to a vast array of pathophysiological etiology, suggesting that CCN4 could be utilized not only as a non-invasive diagnostic or prognostic marker, but also as a promising therapeutic target. The cognate receptor of CCN4 remains elusive till date, which limits understanding of the mechanistic insights on CCN4 driven disease pathologies. However, as therapeutic agents directed against CCN4 begin to make their way into the clinic, that may start to change. Also, the pathophysiological significance of CCN4 remains underexplored, hence further research is needed to shed more light on its disease and/or tissue specific functions to better understand its clinical translational benefit. This review highlights the compelling evidence of overlapping and/or diverse functional and mechanisms regulated by CCN4, in addition to addressing the challenges, study limitations and knowledge gaps on CCN4 biology and its therapeutic potential.
Collapse
Affiliation(s)
- Kirti Singh
- Biotherapeutic Enabling Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46225, USA
| | - Sunday S Oladipupo
- Biotherapeutic Enabling Biology, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, IN, 46225, USA.
| |
Collapse
|
3
|
Soffritti I, D’Accolti M, Maccari C, Bini F, Mazziga E, Arcangeletti MC, Caselli E. Coinfection of Dermal Fibroblasts by Human Cytomegalovirus and Human Herpesvirus 6 Can Boost the Expression of Fibrosis-Associated MicroRNAs. Microorganisms 2023; 11:412. [PMID: 36838377 PMCID: PMC9958881 DOI: 10.3390/microorganisms11020412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/01/2023] [Accepted: 02/04/2023] [Indexed: 02/10/2023] Open
Abstract
Tissue fibrosis can affect every type of tissue or organ, often leading to organ malfunction; however, the mechanisms involved in this process are not yet clarified. A role has been hypothesized for Human Cytomegalovirus (HCMV) and Human Herpesvirus 6 (HHV-6) infections as triggers of systemic sclerosis (SSc), a severe autoimmune disease causing progressive tissue fibrosis, since both viruses and antiviral immune responses toward them have been detected in patients. Moreover, HCMV or HHV-6A infection was reported to increase the expression of fibrosis-associated transcriptional factors and miRNAs in human dermal fibroblasts. However, it is unlikely that they have separate effects in the infected host, as both viruses are highly prevalent in the human population. Thus, our study aimed to investigate, by quantitative real-time PCR microarray, the impact of HCMV/HHV-6A coinfection on the expression of pro-fibrotic miRNAs in coinfected cells, compared to the effect of single viruses. The results showed a possible synergistic effect of the two viruses on pro-fibrotic miRNA expression, thus suggesting that HCMV and HHV-6 may enhance each other and cooperate at inducing enhanced miRNA-driven fibrosis. These data may also suggest a possible use of virus-induced miRNAs as novel diagnostic or prognostic biomarkers for SSc and its clinical treatment.
Collapse
Affiliation(s)
- Irene Soffritti
- Section of Microbiology, Department of Chemical, Pharmaceutical and Agricultural Sciences and LTTA, University of Ferrara, 44121 Ferrara, Italy
| | - Maria D’Accolti
- Section of Microbiology, Department of Chemical, Pharmaceutical and Agricultural Sciences and LTTA, University of Ferrara, 44121 Ferrara, Italy
| | - Clara Maccari
- Laboratory of Microbiology and Virology, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Francesca Bini
- Section of Microbiology, Department of Chemical, Pharmaceutical and Agricultural Sciences and LTTA, University of Ferrara, 44121 Ferrara, Italy
| | - Eleonora Mazziga
- Section of Microbiology, Department of Chemical, Pharmaceutical and Agricultural Sciences and LTTA, University of Ferrara, 44121 Ferrara, Italy
| | - Maria-Cristina Arcangeletti
- Laboratory of Microbiology and Virology, Department of Medicine and Surgery, University of Parma, 43126 Parma, Italy
| | - Elisabetta Caselli
- Section of Microbiology, Department of Chemical, Pharmaceutical and Agricultural Sciences and LTTA, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
4
|
Time and phenotype-dependent transcriptome analysis in AAV-TGFβ1 and Bleomycin-induced lung fibrosis models. Sci Rep 2022; 12:12190. [PMID: 35842487 PMCID: PMC9288451 DOI: 10.1038/s41598-022-16344-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 07/08/2022] [Indexed: 11/19/2022] Open
Abstract
We have previously established a novel mouse model of lung fibrosis based on Adeno-associated virus (AAV)-mediated pulmonary overexpression of TGFβ1. Here, we provide an in-depth characterization of phenotypic and transcriptomic changes (mRNA and miRNA) in a head-to-head comparison with Bleomycin-induced lung injury over a 4-week disease course. The analyses delineate the temporal state of model-specific and commonly altered pathways, thereby providing detailed insights into the processes underlying disease development. They further guide appropriate model selection as well as interventional study design. Overall, Bleomycin-induced fibrosis resembles a biphasic process of acute inflammation and subsequent transition into fibrosis (with partial resolution), whereas the TGFβ1-driven model is characterized by pronounced and persistent fibrosis with concomitant inflammation and an equally complex disease phenotype as observed upon Bleomycin instillation. Finally, based on an integrative approach combining lung function data, mRNA/miRNA profiles, their correlation and miRNA target predictions, we identify putative drug targets and miRNAs to be explored as therapeutic candidates for fibrotic diseases. Taken together, we provide a comprehensive analysis and rich data resource based on RNA-sequencing, along with a strategy for transcriptome-phenotype coupling. The results will be of value for TGFβ research, drug discovery and biomarker identification in progressive fibrosing interstitial lung diseases.
Collapse
|
5
|
Splicing mutations in the CFTR gene as therapeutic targets. Gene Ther 2022; 29:399-406. [PMID: 35650428 PMCID: PMC9385490 DOI: 10.1038/s41434-022-00347-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 05/07/2022] [Accepted: 05/12/2022] [Indexed: 11/08/2022]
Abstract
The marketing approval, about ten years ago, of the first disease modulator for patients with cystic fibrosis harboring specific CFTR genotypes (~5% of all patients) brought new hope for their treatment. To date, several therapeutic strategies have been approved and the number of CFTR mutations targeted by therapeutic agents is increasing. Although these drugs do not reverse the existing disease, they help to increase the median life expectancy. However, on the basis of their CFTR genotype, ~10% of patients presently do not qualify for any of the currently available CFTR modulator therapies, particularly patients with splicing mutations (~12% of the reported CFTR mutations). Efforts are currently made to develop therapeutic agents that target disease-causing CFTR variants that affect splicing. This highlights the need to fully identify them by scanning non-coding regions and systematically determine their functional consequences. In this review, we present some examples of CFTR alterations that affect splicing events and the different therapeutic options that are currently developed and tested for splice switching.
Collapse
|
6
|
Chen Y, Li X, Li Y, Wu Y, Huang G, Wang X, Guo S. Downregulation of microRNA‑423‑5p suppresses TGF‑β1‑induced EMT by targeting FOXP4 in airway fibrosis. Mol Med Rep 2022; 26:242. [PMID: 35642665 PMCID: PMC9185700 DOI: 10.3892/mmr.2022.12758] [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: 10/08/2021] [Accepted: 03/30/2022] [Indexed: 11/10/2022] Open
Abstract
Airway fibrosis (AF) is a common disease that can severely affect patient prognosis. Epithelial-mesenchymal transition (EMT) participates in the pathophysiological development of AF and several studies have demonstrated that some microRNAs (miRNAs) contribute to the development of EMT. The aim of this study was to investigate the function of miR-423-5p in the EMT process and its possible underlying mechanism in BEAS-2B cells. The present study utilized the BEAS-2B cell line to model EMT in AF. Online tools, fluorescence in situ hybridization analysis and an RNA pull-down assay were used to identify potential target genes of miR-423-5p. In addition, immunohistochemistry, wound healing assays, Transwell migration assays, flow cytometry, enzyme-linked immunosorbent assay, reverse transcription-quantitative PCR, western blot analysis and immunofluorescence staining were used to determine the function of miR-423-5p and its target gene in the EMT process in AF. The results indicated that the miR-423-5p expression in AF tissues and BEAS-2B cells stimulated with 10 ng/ml TGF-β1 for 24 h was significantly increased compared with that in the control group. Overexpression of miR-423-5p facilitated TGF-β1-induced EMT in BEAS-2B cells; by contrast, downregulation of miR-423-5p suppressed TGF-β1-induced EMT in BEAS-2B cells. Furthermore, forkhead box p4 (FOXP4) was identified as a potential target gene of miR-423-5p and changes in the miR-423-5p and FOXP4 expression were shown to significantly affect the expression of PI3K/AKT/mTOR pathway members. In summary, overexpression of miR-423-5P promoted the EMT process in AF by downregulating FOXP4 expression and the underlying mechanism may partly involve activation of the PI3K/AKT/mTOR pathway.
Collapse
Affiliation(s)
- Yi Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xuan Li
- Department of Clinical Nutrition, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yishi Li
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yongchang Wu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Guichuan Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Xin Wang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| | - Shuliang Guo
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
7
|
Liu Z, Liu H, Yu D, Gao J, Ruan B, Long R. Downregulation of miR‑29b‑3p promotes α‑tubulin deacetylation by targeting the interaction of matrix metalloproteinase‑9 with integrin β1 in nasal polyps. Int J Mol Med 2021; 48:126. [PMID: 33982786 PMCID: PMC8128418 DOI: 10.3892/ijmm.2021.4959] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 04/12/2021] [Indexed: 11/06/2022] Open
Abstract
Matrix metalloproteinase (MMP)‑9 is a key enzyme responsible for extracellular matrix degradation and contributes to the progressive histological changes observed in lower respiratory tract infections. Integrin β1 and α‑tubulin are potential MMP‑9‑interacting proteins, and microRNA (miR)‑29b‑3p can regulate MMP‑9 expression. MMP‑9 is highly expressed in chronic rhinosinusitis with nasal polyps (CRSwNPs), regardless of its effects on miR‑29b‑3p, integrin β1 and α‑tubulin expression. In the present study, samples from 100 patients with CRSwNPs were examined via reverse transcription‑quantitative PCR to assess the mRNA expression of miR‑29b‑3p, and western blotting was performed to assess the protein expression of MMP‑2, MMP‑9, acetyl‑α‑tubulin, integrin β1 and tissue inhibitor of metalloproteinase 1 (TIMP‑1). A dual‑luciferase reporter assay was used to verify the direct binding of miR‑29b‑3p and MMP‑2/MMP‑9. Co‑immunoprecipitation (Co‑IP) and GST pull‑down assays showed that integrin β1 and α‑tubulin were MMP‑9‑interacting proteins. Cell viability, apoptosis and inflammatory cytokine levels were determined via a Cell Counting Kit‑8 assay, flow cytometry and ELISA, respectively. miR‑29b‑3p expression was found to be positively correlated with MMP‑2 and MMP‑9 expression. Whereas, TIMP‑1 expression was negatively correlated with MMP‑2 and MMP‑9 expression. The dual‑luciferase assay revealed that miR‑29b‑3p targeted the 3' untranslated region of MMP‑2/MMP‑9. The Co‑IP and GST pull‑down assays showed that MMP‑9 could directly bind to integrin β1 and indirectly bind to α‑tubulin. Finally, the overexpression of miR‑29b‑3p decreased the expression of MMP‑9 and increased the levels of acetyl‑α‑tubulin. By contrast, the knockdown of miR‑29b‑3p increased the expression of MMP‑9 and decreased the levels of acetyl‑α‑tubulin. Additionally, MMP‑9 expression was found to be negatively correlated with acetyl‑α‑tubulin expression. Of note, the expression of integrin β1 did not change following the overexpression and knockdown of MMP‑9. Finally, the overexpression of miR‑29b‑3p not only decreased MMP‑9 expression, but also alleviated lipopolysaccharide‑induced inflammation in NP69 cells. The results showed that the downregulation of miR‑29b‑3p promoted α‑tubulin deacetylation by increasing the number of MMP‑9‑integrin β1 complexes in CRSwNPs, thus targeting miR‑29b‑3p/MMP‑9 may be a potential novel strategy for the clinical treatment of CRSwNPs.
Collapse
Affiliation(s)
- Zhuohui Liu
- Department of Otolaryngology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Haoyu Liu
- Department of Otolaryngology, The First People's Hospital of Qujing, Qujing, Yunnan 655000, P.R. China
| | - Deshun Yu
- Department of Otolaryngology, Affiliated Hospital of Dali University, Dali, Yunnan 671000, P.R. China
| | - Jingyu Gao
- Department of Otolaryngology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Biao Ruan
- Department of Otolaryngology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| | - Ruiqing Long
- Department of Otolaryngology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, P.R. China
| |
Collapse
|