1
|
McLaurin DM, Tucker SK, Siddique SJ, Challagundla L, Gibert Y, Hebert MD. A Novel Role for Coilin in Vertebrate Innate Immunity. FASEB J 2025; 39:e70580. [PMID: 40277349 PMCID: PMC12023821 DOI: 10.1096/fj.202403276r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 04/04/2025] [Accepted: 04/18/2025] [Indexed: 04/26/2025]
Abstract
Coilin is a protein localized in the nucleus, where it plays a role in the assembly of the Cajal Body and is involved in ribonucleoprotein biogenesis. Our recent research has uncovered new roles for coilin, including its involvement in producing microRNAs and in modifying other proteins through phosphorylation and SUMOylation. We also proposed that coilin could respond to stress signals. In plants, coilin has been shown to help regulate immune genes and activate defense mechanisms, especially in response to stress. In this study, we used two vertebrate models to study coilin function: a human primary foreskin fibroblast cell line deficient in coilin through RNA interference and a newly created zebrafish line with a mutation in the coilin gene generated by CRISPR-Cas9. Transcriptomic analysis in these two models of coilin deficiency revealed dysregulation of immunity-related genes in both species. To phenotypically validate the transcriptomic results, we challenged zebrafish coilin mutants with lipopolysaccharide (LPS), which triggers an innate immune response, and identified an attenuated response to LPS in vivo in the zebrafish coilin mutants. Our results support a vital novel function for coilin in vertebrates in regulating the expression of immunity-related genes. Moreover, these findings could lead to more research on how coilin regulates innate immunity in animals and humans.
Collapse
Affiliation(s)
- Douglas M. McLaurin
- Department of Cell and Molecular BiologyThe University of Mississippi Medical CenterJacksonMississippiUSA
| | - Sara K. Tucker
- Department of Cell and Molecular BiologyThe University of Mississippi Medical CenterJacksonMississippiUSA
| | - Shanzida J. Siddique
- Department of Cell and Molecular BiologyThe University of Mississippi Medical CenterJacksonMississippiUSA
| | - Lavanya Challagundla
- Department of Cell and Molecular BiologyThe University of Mississippi Medical CenterJacksonMississippiUSA
| | - Yann Gibert
- Department of Cell and Molecular BiologyThe University of Mississippi Medical CenterJacksonMississippiUSA
| | - Michael D. Hebert
- Department of Cell and Molecular BiologyThe University of Mississippi Medical CenterJacksonMississippiUSA
| |
Collapse
|
2
|
Karpathiou G, Mobarki M, Musawi S, Papoudou-Bai A, Péoc'h M. Coilin Is Not Expressed by Immunohistochemistry in Hodgkin and Diffuse Large B-cell Lymphomas. Appl Immunohistochem Mol Morphol 2025:00129039-990000000-00223. [PMID: 40256852 DOI: 10.1097/pai.0000000000001259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 03/10/2025] [Indexed: 04/22/2025]
Abstract
Coilin is the signature protein of Cajal bodies (CBs), membrane-less organelles probably acting as sites for post-transcriptional RNA modification. Recent data suggest that coilin may be a regulator of the NF-kB activity, and Hodgkin lymphomas are hallmarks of neoplasms with NF-kB dysregulation. To the best of our knowledge, the immunohistochemical expression of coilin has been never investigated in Hodgkin lymphomas. We herein examined, by immunohistochemistry, full tissue sections of 58 classical Hodgkin lymphomas diagnosed in 31 male and 27 female patients and found that none of the cases expressed coilin. We compared these findings with Coilin expression in diffuse large B-cell lymphomas (DLBCL), where the marker was also negative. This finding represents the first data on coilin in lymphomas and prompts further studies to explore this downregulation.
Collapse
Affiliation(s)
- Georgia Karpathiou
- Pathology Department, University Hospital of Saint-Etienne, Saint-Etienne, France
| | | | - Shaqraa Musawi
- Department of Medical Laboratories Technology, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | | | - Michel Péoc'h
- Pathology Department, University Hospital of Saint-Etienne, Saint-Etienne, France
| |
Collapse
|
3
|
Lu L, Zhou X, Zheng J, Li D. Coilin Affects the Prognosis of Hepatocellular Carcinoma Through Cell Cycle and Apoptosis. J Hepatocell Carcinoma 2025; 12:367-382. [PMID: 40008396 PMCID: PMC11853879 DOI: 10.2147/jhc.s500119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Purpose Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality with a challenging prognosis. HCC lacks effective prognostic biomarkers. We investigated the diagnostic and prognostic value of COIL expression in HCC. Patients and Methods This study evaluated the expression and prognostic significance of COIL using data from the TCGA and local hospital samples, with 374 and 118 liver cancer patients in the TCGA database and local hospital, respectively. The techniques include bioinformatics analysis, qRT-PCR, immunohistochemistry (IHC), and in vitro cell experiments, which encompass CCK-8 assays, wound healing assays, and Transwell invasion assays. The relationship between COIL expression and clinical outcomes was assessed, and COIL's biological function in HCC was investigated through cellular assays. Results Analysis of cell lines and HCC tissue samples revealed that COIL mRNA or protein expression levels were significantly higher in HCC cell lines/tissues compared to normal liver cells/tissues. Univariate and multivariate analyses indicated that COIL is an independent prognostic factor for overall survival (OS) in HCC. Additionally, 14% of HCC patients had alterations in the COIL gene, and patients with COIL gene alterations had significantly lower OS (p<0.001) and disease-free survival (DFS) (p<0.001) compared to those without gene alterations. Knockdown of COIL expression inhibited the proliferation, migration, and invasion of Hep3B, HepG2, and Huh7. Compared to the control group, COIL knockdown cells showed a marked reduction in CDC25C and CCNB1 protein levels, suggesting that COIL knockdown leads to G2/M phase cell cycle arrest. After COIL knockdown, caspase-3 and BCL-2 protein levels were downregulated, while cleaved caspase and BAX protein levels were upregulated, indicating that COIL knockdown promotes apoptosis in liver cancer cells. Conclusion COIL is an independent predictor of prognosis. COIL's association with poor OS and its role in enhancing cancer cell proliferation and invasion highlight its potential as a therapeutic target.
Collapse
Affiliation(s)
- Lingling Lu
- Department of Infection Disease, Fujian Medical University Union Hospital, Fuzhou, People’s Republic of China
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, People’s Republic of China
| | - Xiaoling Zhou
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, People’s Republic of China
- Department of Hepatobiliary Disease, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, People’s Republic of China
| | - Jiaolong Zheng
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, People’s Republic of China
- Department of Hepatobiliary Disease, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, People’s Republic of China
| | - Dongliang Li
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, People’s Republic of China
- Department of Hepatobiliary Disease, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, People’s Republic of China
| |
Collapse
|
4
|
Nsengimana B, Khan FA, Awan UA, Wang D, Fang N, Wei W, Zhang W, Ji S. Pseudogenes and Liquid Phase Separation in Epigenetic Expression. Front Oncol 2022; 12:912282. [PMID: 35875144 PMCID: PMC9305658 DOI: 10.3389/fonc.2022.912282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/13/2022] [Indexed: 11/24/2022] Open
Abstract
Pseudogenes have been considered as non-functional genes. However, peptides and long non-coding RNAs produced by pseudogenes are expressed in different tumors. Moreover, the dysregulation of pseudogenes is associated with cancer, and their expressions are higher in tumors compared to normal tissues. Recent studies show that pseudogenes can influence the liquid phase condensates formation. Liquid phase separation involves regulating different epigenetic stages, including transcription, chromatin organization, 3D DNA structure, splicing, and post-transcription modifications like m6A. Several membrane-less organelles, formed through the liquid phase separate, are also involved in the epigenetic regulation, and their defects are associated with cancer development. However, the association between pseudogenes and liquid phase separation remains unrevealed. The current study sought to investigate the relationship between pseudogenes and liquid phase separation in cancer development, as well as their therapeutic implications.
Collapse
Affiliation(s)
- Bernard Nsengimana
- Laboratory of Cell Signal Transduction, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Faiz Ali Khan
- Laboratory of Cell Signal Transduction, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- School of Life Sciences, Henan University, Kaifeng, China
- Department of Basic Sciences Research, Shaukat Khanum Memorial Cancer Hospital and Research Centre (SKMCH&RC), Lahore, Pakistan
| | - Usman Ayub Awan
- Department of Medical Laboratory Technology, The University of Haripur, Haripur, Pakistan
| | - Dandan Wang
- Laboratory of Cell Signal Transduction, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Na Fang
- Laboratory of Cell Signal Transduction, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
| | - Wenqiang Wei
- Laboratory of Cell Signal Transduction, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- *Correspondence: Wenqiang Wei, ; Weijuan Zhang, ; Shaoping Ji,
| | - Weijuan Zhang
- Laboratory of Cell Signal Transduction, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- *Correspondence: Wenqiang Wei, ; Weijuan Zhang, ; Shaoping Ji,
| | - Shaoping Ji
- Laboratory of Cell Signal Transduction, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Henan University, Kaifeng, China
- *Correspondence: Wenqiang Wei, ; Weijuan Zhang, ; Shaoping Ji,
| |
Collapse
|
5
|
Del Moral-Morales A, González-Orozco JC, Hernández-Vega AM, Hernández-Ortega K, Peña-Gutiérrez KM, Camacho-Arroyo I. EZH2 Mediates Proliferation, Migration, and Invasion Promoted by Estradiol in Human Glioblastoma Cells. Front Endocrinol (Lausanne) 2022; 13:703733. [PMID: 35197928 PMCID: PMC8859835 DOI: 10.3389/fendo.2022.703733] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 01/11/2022] [Indexed: 12/12/2022] Open
Abstract
Glioblastomas (GBM) are the most frequent and aggressive brain tumors. 17β-estradiol (E2) increases proliferation, migration, and invasion of human GBM cells; however underlying mechanisms are no fully understood. Zeste 2 Enhancer Homologous enzyme (EZH2) is a methyltransferase part of Polycomb 2 repressor complex (PRC2). In GBM, EZH2 is overexpressed and involved in the cell cycle, migration, and invasion processes. We studied the role of EZH2 in the pro-oncogenic actions of E2 in human GBM cells. EZH2 gene silencing and pharmacological inhibition of EZH2 blocked proliferation, migration, and invasion of GBM cells induced by E2. We identified in silico additional putative estrogen response elements (EREs) at the EZH2 promoter, but E2 did not modify EZH2 expression. In silico analysis also revealed that among human GBM samples, EZH2 expression was homogeneous; in contrast, the heterogeneous expression of estrogen receptors (ERs) allowed the classification of the samples into groups. Even in the GBM cluster with high expression of ERs and those of their target genes, the expression of PCR2 target genes did not change. Overall, our data suggest that in GBM cells, pro-oncogenic actions of E2 are mediated by EZH2, without changes in EZH2 expression and by mechanisms that appear to be unrelated to the transcriptional activity of ERs.
Collapse
Affiliation(s)
- Aylin Del Moral-Morales
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - Juan Carlos González-Orozco
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - Ana María Hernández-Vega
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - Karina Hernández-Ortega
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - Karla Mariana Peña-Gutiérrez
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| | - Ignacio Camacho-Arroyo
- Unidad de Investigación en Reproducción Humana, Instituto Nacional de Perinatología-Facultad de Química, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, Mexico
| |
Collapse
|
6
|
Liang ZL, Tan HW, Wu JY, Chen XL, Wang XY, Xu YM, Lau ATY. The Impact of ZIP8 Disease-Associated Variants G38R, C113S, G204C, and S335T on Selenium and Cadmium Accumulations: The First Characterization. Int J Mol Sci 2021; 22:ijms222111399. [PMID: 34768831 PMCID: PMC8583799 DOI: 10.3390/ijms222111399] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/14/2021] [Accepted: 10/14/2021] [Indexed: 02/05/2023] Open
Abstract
The metal cation symporter ZIP8 (SLC39A8) is a transmembrane protein that imports the essential micronutrients iron, manganese, and zinc, as well as heavy toxic metal cadmium (Cd). It has been recently suggested that selenium (Se), another essential micronutrient that has long been known for its role in human health and cancer risk, may also be transported by the ZIP8 protein. Several mutations in the ZIP8 gene are associated with the aberrant ion homeostasis of cells and can lead to human diseases. However, the intricate relationships between ZIP8 mutations, cellular Se homeostasis, and human diseases (including cancers and illnesses associated with Cd exposure) have not been explored. To further verify if ZIP8 is involved in cellular Se transportation, we first knockout (KO) the endogenous expression of ZIP8 in the HeLa cells using the CRISPR/Cas9 system. The elimination of ZIP8 expression was examined by PCR, DNA sequencing, immunoblot, and immunofluorescence analyses. Inductively coupled plasma mass spectrometry indicated that reduced uptake of Se, along with other micronutrients and Cd, was observed in the ZIP8-KO cells. In contrast, when ZIP8 was overexpressed, increased Se uptake could be detected in the ZIP8-overexpressing cells. Additionally, we found that ZIP8 with disease-associated single-point mutations G38R, G204C, and S335T, but not C113S, showed reduced Se transport ability. We then evaluated the potential of Se on Cd cytotoxicity prevention and therapy of cancers. Results indicated that Se could suppress Cd-induced cytotoxicity via decreasing the intracellular Cd transported by ZIP8, and Se exhibited excellent anticancer activity against not all but only selected cancer cell lines, under restricted experimental conditions. Moreover, clinical-based bioinformatic analyses revealed that up-regulated ZIP8 gene expression was common across multiple cancer types, and selenoproteins that were significantly co-expressed with ZIP8 in these cancers had been identified. Taken together, this study concludes that ZIP8 is an important protein in modulating cellular Se levels and provides insights into the roles of ZIP8 and Se in disease prevention and therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Yan-Ming Xu
- Correspondence: (Y.-M.X.); (A.T.Y.L.); Tel.: +86-754-8890-0437 (Y.-M.X.); +86-754-8853-0052 (A.T.Y.L.)
| | - Andy T. Y. Lau
- Correspondence: (Y.-M.X.); (A.T.Y.L.); Tel.: +86-754-8890-0437 (Y.-M.X.); +86-754-8853-0052 (A.T.Y.L.)
| |
Collapse
|