1
|
Zhai F, Li Y, Zheng J, Yan C, Wang S, Yang W, Jin J, Luo X, Zhan Z, Shi J, Wang S, Lin Y, Kong L, Ge Y, Wang H, Ye M, Jin X. SPOP/NOLC1/B4GALT1 signaling axis enhances paclitaxel resistance in endometrial cancer by inducing O-dysglycosylation. Oncogene 2025:10.1038/s41388-025-03347-7. [PMID: 40097806 DOI: 10.1038/s41388-025-03347-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/18/2025] [Accepted: 03/10/2025] [Indexed: 03/19/2025]
Abstract
The effective treatment of paclitaxel-resistant patients remains a major challenge. We found that nucleolar and coiled body phosphoprotein 1 (NOLC1) was highly expressed in the paclitaxel-resistant endometrial cancer (ECa) cells and pathological tissue of ECa patients, which could promote the occurrence and progression of ECa cells. Mechanistically, we confirmed that the E3 ubiquitin ligase substrate-binding adaptor SPOP mediates the ubiquitination and degradation of NOLC1, thereby maintaining normal protein levels. However, ECa-associated SPOP mutants abrogated the binding and ubiquitination of NOLC1, resulting in the accumulation of NOLC1, and ultimately promoting the proliferation, migration, and invasion of ECa cells. In addition, we demonstrated that NOLC1 could act as a transcriptional factor to activate the transcriptional expression of B4GALT1, ultimately leading to abnormal glycosylation metabolism. Moreover, knockdown of B4GALT1 can partly counteract the cancer-promoting effect caused by the overexpression of NOLC1 in vitro and in vivo. Based on these findings, an O-glycosylation inhibitor combined with paclitaxel could effectively improve the sensitivity of paclitaxel-resistant cells. In summary, we found that SPOP can negatively regulate the NOLC1-B4GALT1 signaling axis in ECa, whereas ECa-associated SPOP mutants lead to abnormal activation of this signaling axis, leading to glycosylation metabolism disorders. In addition, paclitaxel combined with B4GALT1-KD or glycosylation inhibitors can significantly inhibit the growth of paclitaxel-resistant endometrial cancer cells.
Collapse
Affiliation(s)
- Fengguang Zhai
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, China
- Department of Radiotherapy and Chemotherapy, The First Hospital of Ningbo University, Ningbo, China
| | - Yuxuan Li
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, China
| | - Jingfei Zheng
- Department of Obstetrics and Gynecology, Yinzhou Renmin Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, China
| | - Chunhong Yan
- Department of Obstetrics and Gynecology, Yinzhou Renmin Hospital Affiliated to Medical School of Ningbo University, Ningbo, Zhejiang, China
| | - Shuyan Wang
- Department of Histopathology, Ningbo Clinical Pathology Diagnosis Center, Ningbo, China
| | - Weili Yang
- Department of Gynecology, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| | - Jiabei Jin
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, China
| | - Xia Luo
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, China
| | - Ziqing Zhan
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, China
- Department of Radiotherapy and Chemotherapy, The First Hospital of Ningbo University, Ningbo, China
| | - Jiaxin Shi
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, China
| | - Siyuan Wang
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, China
| | - Yan Lin
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, China
| | - Lili Kong
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, China
| | - Yidong Ge
- Department of Gynecology, The Affiliated People's Hospital of Ningbo University, Ningbo, China
| | - Haoyun Wang
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, China
| | - Meng Ye
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, China.
- Department of Radiotherapy and Chemotherapy, The First Hospital of Ningbo University, Ningbo, China.
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology, Health Science Center, Ningbo University, Ningbo, China.
| |
Collapse
|
2
|
Zhai F, Wang J, Luo X, Ye M, Jin X. Roles of NOLC1 in cancers and viral infection. J Cancer Res Clin Oncol 2023; 149:10593-10608. [PMID: 37296317 DOI: 10.1007/s00432-023-04934-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023]
Abstract
BACKGROUND The nucleolus is considered the center of metabolic control and an important organelle for the biogenesis of ribosomal RNA (rRNA). Nucleolar and coiled-body phosphoprotein 1(NOLC1), which was originally identified as a nuclear localization signal-binding protein is a nucleolar protein responsible for nucleolus construction and rRNA synthesis, as well as chaperone shuttling between the nucleolus and cytoplasm. NOLC1 plays an important role in a variety of cellular life activities, including ribosome biosynthesis, DNA replication, transcription regulation, RNA processing, cell cycle regulation, apoptosis, and cell regeneration. PURPOSE In this review, we introduce the structure and function of NOLC1. Then we elaborate its upstream post-translational modification and downstream regulation. Meanwhile, we describe its role in cancer development and viral infection which provide a direction for future clinical applications. METHODS The relevant literatures from PubMed have been reviewed for this article. CONCLUSION NOLC1 plays an important role in the progression of multiple cancers and viral infection. In-depth study of NOLC1 provides a new perspective for accurate diagnosis of patients and selection of therapeutic targets.
Collapse
Affiliation(s)
- Fengguang Zhai
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
- The Affiliated First Hospital, Ningbo University, Ningbo, 315020, China
| | - Jie Wang
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
- The Affiliated First Hospital, Ningbo University, Ningbo, 315020, China
| | - Xia Luo
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Meng Ye
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China.
- The Affiliated First Hospital, Ningbo University, Ningbo, 315020, China.
| | - Xiaofeng Jin
- Department of Biochemistry and Molecular Biology, Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo, 315211, China.
- The Affiliated First Hospital, Ningbo University, Ningbo, 315020, China.
| |
Collapse
|
3
|
Radhakrishnan S, Martin CA, Rammohan A, Vij M, Chandrasekar M, Rela M. Significance of nucleologenesis, ribogenesis, and nucleolar proteome in the pathogenesis and recurrence of hepatocellular carcinoma. Expert Rev Gastroenterol Hepatol 2023; 17:363-378. [PMID: 36919496 DOI: 10.1080/17474124.2023.2191189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Accepted: 03/11/2023] [Indexed: 03/16/2023]
Abstract
INTRODUCTION Emerging evidence suggests that enhanced ribosome biogenesis, increased size, and quantitative distribution of nucleoli are associated with dysregulated transcription, which in turn drives a cell into aberrant cellular proliferation and malignancy. Nucleolar alterations have been considered a prognostic histological marker for aggressive tumors. More recently, advancements in the understanding of chromatin network (nucleoplasm viscosity) regulated liquid-liquid phase separation mechanism of nucleolus formation and their multifunctional role shed light on other regulatory processes, apart from ribosomal biogenesis of the nucleolus. AREAS COVERED Using hepatocellular carcinoma as a model to study the role of nucleoli in tumor progression, we review the potential of nucleolus coalescence in the onset and development of tumors through non-ribosomal biogenesis pathways, thereby providing new avenues for early diagnosis and cancer therapy. EXPERT OPINION Molecular-based classifications have failed to identify the nucleolar-based molecular targets that facilitate cell-cycle progression. However, the algorithm-based tumor risk identification with high-resolution medical images suggests prominent nucleoli, karyotheca, and increased nucleus/cytoplasm ratio as largely associated with tumor recurrence. Nonetheless, the role of the non-ribosomal functions of nucleoli in tumorigenesis remains elusive. This clearly indicates the lacunae in the study of the nucleolar proteins pertaining to cancer. [Figure: see text].
Collapse
Affiliation(s)
| | | | - Ashwin Rammohan
- The Institute of Liver Disease & Transplantation, Dr. Rela Institute & Medical Centre, Chennai, India
| | - Mukul Vij
- Department of Pathology, Dr. Rela Institute & Medical Centre, Chennai, India
| | - Mani Chandrasekar
- Department of Oncology, Dr. Rela Institute & Medical Centre, Chennai, India
| | - Mohamed Rela
- Cell Laboratory, National Foundation for Liver Research, Chennai, India
- The Institute of Liver Disease & Transplantation, Dr. Rela Institute & Medical Centre, Chennai, India
| |
Collapse
|
4
|
Chen S, Li Y, Wu M, Xue L, Zhu J, Wu M, Zhang Q, He G, Li G, Fu S, Zheng C, Deng X. Nucleolar and Coiled-Body Phosphoprotein 1 Is Associated With Stemness and Represents a Potential Therapeutic Target in Triple-Negative Breast Cancer. Front Oncol 2022; 12:731528. [PMID: 35174077 PMCID: PMC8841672 DOI: 10.3389/fonc.2022.731528] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 01/10/2022] [Indexed: 11/29/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer and lacks approved specific targeted therapies. One of the major reasons why TNBC is difficult to treat is the high proportion of cancer stem cells within the tumor tissue. Nucleolus is the location of ribosome biogenesis which is frequently overactivated in cancer cells and overactivation of ribosome biogenesis frequently drives the malignant transformation of cancer. Nucleolar and coiled-body phosphoprotein 1 (NOLC1) is a nucleolar protein responsible for nucleolus organization and rRNA synthesis and plays an important role in ribosome biogenesis. However, the correlation of NOLC1 expression with patient prognosis and its value as a therapeutic target have not been evaluated in TNBC. In the current study, based on bioinformatics analysis of the online databases, we found that the expression of NOLC1 was higher in breast cancer tissues than normal tissues, and NOLC1 was expressed at a higher level in TNBC than other subtypes of breast cancer. GSEA analysis revealed that stemness-related pathways were significantly enriched in breast cancer with high NOLC1 gene expression. Further analyses using gene expression profiling interactive analysis 2 (GEPIA2), tumor immune estimation resource (TIMER) and search tool for retrieval of interacting genes/proteins (STRING) demonstrated that NOLC1 was significantly associated with stemness in both all breast cancer and basal-like breast cancer/TNBC patients at both gene and protein levels. Knockdown of NOLC1 by siRNA decreased the protein level of the key stemness regulators MYC and ALDH and inhibited the sphere-forming capacity in TNBC cell line MDA-MB-231. Univariate and multivariate Cox regression analyses demonstrated that NOLC1 was an independent risk factor for overall survival in breast cancer. PrognoScan and Kaplan-Meier plotter analyses revealed that high expression of NOLC1 was associated with poor prognosis in both all breast cancer and TNBC patients. Further immunohistochemical analysis of breast cancer patient samples revealed that TNBC cells had a lower level of NOLC1 in the nucleus compared with non-TNBC cells. These findings suggest that NOLC1 is closely associated with the stemness properties of TNBC and represents a potential therapeutic target for TNBC.
Collapse
Affiliation(s)
- Sisi Chen
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Departments of Pathology and Pathophysiology, Hunan Normal University School of Medicine, Changsha, China
- Key Laboratory of Translational Cancer Stem Cell Research, Hunan Normal University, Changsha, China
| | - Ying Li
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Departments of Pathology and Pathophysiology, Hunan Normal University School of Medicine, Changsha, China
- Key Laboratory of Translational Cancer Stem Cell Research, Hunan Normal University, Changsha, China
| | - Muyao Wu
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Departments of Pathology and Pathophysiology, Hunan Normal University School of Medicine, Changsha, China
- Key Laboratory of Translational Cancer Stem Cell Research, Hunan Normal University, Changsha, China
| | - Lian Xue
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Departments of Pathology and Pathophysiology, Hunan Normal University School of Medicine, Changsha, China
- Key Laboratory of Translational Cancer Stem Cell Research, Hunan Normal University, Changsha, China
| | - Jianyu Zhu
- Key Laboratory of Translational Cancer Stem Cell Research, Hunan Normal University, Changsha, China
- Department of Pathophysiology, Jishou University School of Medicine, Jishou, China
| | - Mi Wu
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Departments of Pathology and Pathophysiology, Hunan Normal University School of Medicine, Changsha, China
- Key Laboratory of Translational Cancer Stem Cell Research, Hunan Normal University, Changsha, China
| | - Qiuting Zhang
- Key Laboratory of Translational Cancer Stem Cell Research, Hunan Normal University, Changsha, China
| | - Guangchun He
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Departments of Pathology and Pathophysiology, Hunan Normal University School of Medicine, Changsha, China
- Key Laboratory of Translational Cancer Stem Cell Research, Hunan Normal University, Changsha, China
| | - Guifei Li
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Departments of Pathology and Pathophysiology, Hunan Normal University School of Medicine, Changsha, China
- Key Laboratory of Translational Cancer Stem Cell Research, Hunan Normal University, Changsha, China
| | - Shujun Fu
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Departments of Pathology and Pathophysiology, Hunan Normal University School of Medicine, Changsha, China
- Key Laboratory of Translational Cancer Stem Cell Research, Hunan Normal University, Changsha, China
| | - Chanjuan Zheng
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Departments of Pathology and Pathophysiology, Hunan Normal University School of Medicine, Changsha, China
- Key Laboratory of Translational Cancer Stem Cell Research, Hunan Normal University, Changsha, China
- Department of Preventive Medicine, Hunan Normal University School of Medicine, Changsha, China
- *Correspondence: Chanjuan Zheng, ; Xiyun Deng,
| | - Xiyun Deng
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, Departments of Pathology and Pathophysiology, Hunan Normal University School of Medicine, Changsha, China
- Key Laboratory of Translational Cancer Stem Cell Research, Hunan Normal University, Changsha, China
- *Correspondence: Chanjuan Zheng, ; Xiyun Deng,
| |
Collapse
|
5
|
Ruhs S, Griesler B, Huebschmann R, Stroedecke K, Straetz N, Ihling C, Sinz A, Masch A, Schutkowski M, Gekle M, Grossmann C. Modulation of transcriptional mineralocorticoid receptor activity by casein kinase 1. FASEB J 2021; 36:e22059. [PMID: 34847273 DOI: 10.1096/fj.202100977rr] [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: 06/14/2021] [Revised: 10/16/2021] [Accepted: 11/08/2021] [Indexed: 11/11/2022]
Abstract
The mineralocorticoid receptor (MR) with its ligand aldosterone (aldo) physiologically regulates electrolyte homeostasis and blood pressure but it can also lead to pathophysiological effects in the cardiovascular system. Previous results show that posttranslational modifications (PTM) can influence MR signaling and function. Based on in silico and in vitro data, casein kinase 1 (CK1) was predicted as a candidate for MR phosphorylation. To gain a deeper mechanistic insight into MR activation, we investigated the influence of CK1 on MR function in HEK cells. Co-immunoprecipitation experiments indicated that the MR is located in a protein-protein complex with CK1α and CK1ε. Reporter gene assays with pharmacological inhibitors and MR constructs demonstrated that especially CK1ε acts as a positive modulator of GRE activity via the C-terminal MR domains CDEF. CK1 enhanced the binding affinity of aldosterone to the MR, facilitated nuclear translocation and DNA interaction of the MR, and led to expression changes of pathophysiologically relevant genes like Per-1 and Phlda1. By peptide microarray and site-directed mutagenesis experiments, we identified the highly conserved T800 as a direct CK1 phosphorylation site of the MR, which modulates the nuclear import and genomic activity of the receptor. Direct phosphorylation of the MR was unable to fully account for all of the CK1 effects on MR signaling, suggesting additional phosphorylation of MR co-regulators. By LC/MS/MS, we identified the MR-associated proteins NOLC1 and TCOF1 as candidates for such CK1-regulated co-factors. Overall, we found that CK1 acts as a co-activator of MR GRE activity through direct and indirect phosphorylation, which accelerates cytosolic-nuclear trafficking, facilitates nuclear accumulation and DNA binding of the MR, and increases the expression of pathologically relevant MR-target genes.
Collapse
Affiliation(s)
- Stefanie Ruhs
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany.,Department of Anesthesiology and Surgical Intensive Care, University Hospital Halle (Saale), Halle (Saale), Germany
| | - Bruno Griesler
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Ralf Huebschmann
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Katharina Stroedecke
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Nicole Straetz
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Christian Ihling
- Department of Pharmaceutical Chemistry & Bioanalytics, Center for Structural Mass Spectrometry, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Andrea Sinz
- Department of Pharmaceutical Chemistry & Bioanalytics, Center for Structural Mass Spectrometry, Institute of Pharmacy, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Antonia Masch
- Department of Enzymology, Institute of Biochemistry and Biotechnology, Martin Luther University of Halle-Wittenberg, Halle (Saale), Germany
| | - Mike Schutkowski
- Department of Enzymology, Institute of Biochemistry and Biotechnology, Martin Luther University of Halle-Wittenberg, Halle (Saale), Germany
| | - Michael Gekle
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Claudia Grossmann
- Julius Bernstein Institute of Physiology, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
6
|
Knockdown of NOLC1 Inhibits PI3K-AKT Pathway to Improve the Poor Prognosis of Esophageal Carcinoma. JOURNAL OF ONCOLOGY 2021; 2021:9944132. [PMID: 34046062 PMCID: PMC8128555 DOI: 10.1155/2021/9944132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 04/30/2021] [Indexed: 11/29/2022]
Abstract
Objective Esophageal carcinoma (ESCA) is a common malignant gastrointestinal tumor. The abnormal expression of NOLC1 is involved in the tumorigenesis of various human tumors, whereas the function and mechanism of NOLC1 in ESCA remain unclear. In this study, we explored the relationship between NOLC1 and poor prognosis of ESCA, and its role and mechanism in the occurrence of ESCA. Methods The NOLC1 expression in ESCA tissues and cell lines was determined by qRT-PCR, immunohistochemistry, or western blot. The Kaplan–Meier method was conducted to estimate the overall survival. Cox regression analysis was carried out to examine the association between patient characteristics and prognosis. A recombined lentiviral vector containing NOLC1 was applied for transfecting ESCA cells (Eca109 and TE-13) and established a stable cell line with low NOLC1 expression or high NOLC1 expression, in the absence or presence of PI3K inhibitor (LY294002) treatment. Cell proliferation, apoptosis rate, invasion ability, migration ability, and PI3K/AKT pathway were detected by CCK8 assay, flow cytometry, Transwell assay, wound-healing assay, and western blot. Results NOLC1 overexpression was observed in ESCA tissues and ESCA cell lines (EC9706, Eca109, TE-13, Kyse170, T.TN) compared with adjacent normal tissues and normal esophageal cell line HEEC. NOLC1 overexpression was markedly associated with bigger tumor size, lymph node metastasis, and advanced TNM stage. Patients with NOLC1 overexpression have shorter overall survival than that of those with low NOLC1 expression. NOLC1 overexpression was considered to be an independent poor prognostic factor affecting overall survival. NOLC1 knockdown inhibited proliferation, migration, invasion, and cyclin B1 expression and promoted the apoptosis and cleaved-caspase-3 expression of Eca109 and TE-13 cells. NOLC1 overexpression accelerated proliferation, migration, invasion, and cyclin B1 expression and inhibited the apoptosis and cleaved-caspase-3 expression of ESCA cells via activating PI3K/AKT pathway. Rescue experiments showed that PI3K inhibitor (LY294002) could reverse the phenomenon caused by NOLC1 overexpression. Conclusion NOLC1 may be a marker for poor prognosis. It can participate in the occurrence and development of ESCA via the PI3K/AKT pathway.
Collapse
|
7
|
A Zic2/Runx2/NOLC1 signaling axis mediates tumor growth and metastasis in clear cell renal cell carcinoma. Cell Death Dis 2021; 12:319. [PMID: 33767130 PMCID: PMC7994417 DOI: 10.1038/s41419-021-03617-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 12/26/2022]
Abstract
Clear cell renal cell carcinoma (ccRCC) is one of the most common malignancies with rapid growth and high metastasis, but lacks effective therapeutic targets. Here, using public sequencing data analyses, quantitative real-time PCR assay, western blotting, and IHC staining, we characterized that runt-related transcription factor 2 (Runx2) was significantly upregulated in ccRCC tissues than that in normal renal tissues, which was associated with the worse survival of ccRCC patients. Overexpression of Runx2 promoted malignant proliferation and migration of ccRCC cells, and inversely, interfering Runx2 with siRNA attenuates its oncogenic ability. RNA sequencing and functional studies revealed that Runx2 enhanced ccRCC cell growth and metastasis via downregulation of tumor suppressor nucleolar and coiled-body phosphoprotein 1 (NOLC1). Moreover, increased Zic family member 2 (Zic2) was responsible for the upregulation of Runx2 and its oncogenic functions in ccRCC. Kaplan-Meier survival analyses indicated that ccRCC patients with high Zic2/Runx2 and low NOLC1 had the worst outcome. Therefore, our study demonstrates that Zic2/Runx2/NOLC1 signaling axis promotes ccRCC progression, providing a set of potential targets and prognostic indicators for patients with ccRCC.
Collapse
|
8
|
Luo JH, Liu S, Tao J, Ren BG, Luo K, Chen ZH, Nalesnik M, Cieply K, Ma T, Cheng SY, Chen Q, Michalopoulos GK, Nelson JB, Bhargava R, Zhang J, Ma D, Jarrard D, Pennathur A, Luketich JD, DeFranco DB, Monga SP, Tseng G, Yu YP. Pten-NOLC1 fusion promotes cancers involving MET and EGFR signalings. Oncogene 2021; 40:1064-1076. [PMID: 33323972 PMCID: PMC7880894 DOI: 10.1038/s41388-020-01582-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 11/03/2020] [Accepted: 11/20/2020] [Indexed: 02/07/2023]
Abstract
Inactivation of Pten gene through deletions and mutations leading to excessive pro-growth signaling pathway activations frequently occurs in cancers. Here, we report a Pten derived pro-cancer growth gene fusion Pten-NOLC1 originated from a chr10 genome rearrangement and identified through a transcriptome sequencing analysis of human cancers. Pten-NOLC1 fusion is present in primary human cancer samples and cancer cell lines from different organs. The product of Pten-NOLC1 is a nuclear protein that interacts and activates promoters of EGFR, c-MET, and their signaling molecules. Pten-NOLC1 promotes cancer proliferation, growth, invasion, and metastasis, and reduces the survival of animals xenografted with Pten-NOLC1-expressing cancer cells. Genomic disruption of Pten-NOLC1 induces cancer cell death, while genomic integration of this fusion gene into the liver coupled with somatic Pten deletion produces spontaneous liver cancers in mice. Our studies indicate that Pten-NOLC1 gene fusion is a driver for human cancers.
Collapse
Affiliation(s)
- Jian-Hua Luo
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA.
| | - Silvia Liu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Junyan Tao
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Bao-Guo Ren
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Katherine Luo
- Department of Biology, Columbia University, 1212 Amsterdam Avenue, New York, NY, 10027, USA
| | - Zhang-Hui Chen
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Michael Nalesnik
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Kathleen Cieply
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Tianzhou Ma
- Department of Biostatistics, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
- Department of Epidemiology and Biostatistics, University of Maryland School of Public Health, College Park, MD, 20742, USA
| | - Shi-Yuan Cheng
- Department of Neurology, Northwestern University School of Medicine, Chicago, IL, 60611, USA
| | - Qi Chen
- Department of Pharmacology, Toxicology & Therapeutics, University of Kansas, Kansas City, KS, 66160, USA
| | - George K Michalopoulos
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Joel B Nelson
- Department of Urology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Rohit Bhargava
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Jun Zhang
- Department of Medicine, University of Iowa, Iowa City, IA, 52242, USA
- Department of Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Deqin Ma
- Department of Pathology, University of Iowa, Iowa City, IA, 52242, USA
| | - David Jarrard
- Department of Urology, University of Wisconsin Madison, Madison, WI, 53792, USA
| | - Arjun Pennathur
- Department of Thoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - James D Luketich
- Department of Thoracic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Donald B DeFranco
- Department of Pharmacology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - Satdarshan Paul Monga
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA
| | - George Tseng
- Department of Biology, Columbia University, 1212 Amsterdam Avenue, New York, NY, 10027, USA
| | - Yan-Ping Yu
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
9
|
Chen W, Cen S, Zhou X, Yang T, Wu K, Zou L, Luo J, Li C, Lv D, Mao X. Circular RNA CircNOLC1, Upregulated by NF-KappaB, Promotes the Progression of Prostate Cancer via miR-647/PAQR4 Axis. Front Cell Dev Biol 2021; 8:624764. [PMID: 33490086 PMCID: PMC7820754 DOI: 10.3389/fcell.2020.624764] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Accepted: 12/14/2020] [Indexed: 12/13/2022] Open
Abstract
Background CircRNAs recently have shown critical roles in tumor biology. However, their roles in prostate cancer (PCa) remains largely unclear. Methods CircRNA microarrays were performed in immortal prostate cell line RWPE1 and PCa cell lines as DU145, PC3, LNCaP, C4-2, and 22RV1. Combined with upregulated circRNAs in PCa tissues, circNOLC1 expression was validated in PCa cells and tissues via qRT-PCR and FISH. Sanger sequencing, actinomycin D, gDNA, and cDNA, RNase R assays were used to assess the circular characteristics of circNOLC1. CCK-8, colony formation, transwell migration assays, and mice xenograft models were conducted to evaluate the functions of PCa cells after circNOLC1 knockdown and overexpression. RNA pulldown, luciferase reporter assay, FISH (fluorescence in situ hybridization), and CHIP were utilized to illustrate the further mechanisms of circNOLC1. Results Our research indicated that circNOLC1 was overexpressed in PCa cells and tissues, and circNOLC1 was more stable than linear NOLC1 mRNA. CircNOLC1 promoted PCa cells proliferation and migration in vitro and vivo. Additionally, we found that circNOLC1 could upregulate PAQR4 expression by sponging miR-647, leading to the activation of PI3K/Akt pathway. Moreover, NF-kappaB was identified to bind to the NOLC1 promoter sites and upregulated both NOLC1 and circNOLC1 expression. Conclusion CircNOLC1, elevated by transcription factor NF-kappaB, promotes PCa progression via a miR-647/PAQR4 axis, and circNOLC1 is a potential biomarker and target for PCa treatment.
Collapse
Affiliation(s)
- Wenbin Chen
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shengren Cen
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xumin Zhou
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Taowei Yang
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Kaihui Wu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Libin Zou
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Junqi Luo
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Chuanyin Li
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Daojun Lv
- Guangdong Key Laboratory of Urology, Department of Urology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiangming Mao
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
10
|
Wang S, Shen HW, Chai H, Liang Y. Complex harmonic regularization with differential evolution in a memetic framework for biomarker selection. PLoS One 2019; 14:e0210786. [PMID: 30763332 PMCID: PMC6375558 DOI: 10.1371/journal.pone.0210786] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 01/02/2019] [Indexed: 01/23/2023] Open
Abstract
For studying cancer and genetic diseases, the issue of identifying high correlation genes from high-dimensional data is an important problem. It is a great challenge to select relevant biomarkers from gene expression data that contains some important correlation structures, and some of the genes can be divided into different groups with a common biological function, chromosomal location or regulation. In this paper, we propose a penalized accelerated failure time model CHR-DE using a non-convex regularization (local search) with differential evolution (global search) in a wrapper-embedded memetic framework. The complex harmonic regularization (CHR) can approximate to the combination ℓp(12≤p<1) and ℓq (1 ≤ q < 2) for selecting biomarkers in group. And differential evolution (DE) is utilized to globally optimize the CHR’s hyperparameters, which make CHR-DE achieve strong capability of selecting groups of genes in high-dimensional biological data. We also developed an efficient path seeking algorithm to optimize this penalized model. The proposed method is evaluated on synthetic and three gene expression datasets: breast cancer, hepatocellular carcinoma and colorectal cancer. The experimental results demonstrate that CHR-DE is a more effective tool for feature selection and learning prediction.
Collapse
Affiliation(s)
- Sai Wang
- Faculty of Information Technology, Macau University of Science and Technology, Taipa, Macau
| | - Hai-Wei Shen
- Faculty of Information Technology, Macau University of Science and Technology, Taipa, Macau
| | - Hua Chai
- Faculty of Information Technology, Macau University of Science and Technology, Taipa, Macau
| | - Yong Liang
- Faculty of Information Technology, Macau University of Science and Technology, Taipa, Macau
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Taipa, Macau
- * E-mail:
| |
Collapse
|
11
|
Sun HJ, Ren XS, Xiong XQ, Chen YZ, Zhao MX, Wang JJ, Zhou YB, Han Y, Chen Q, Li YH, Kang YM, Zhu GQ. NLRP3 inflammasome activation contributes to VSMC phenotypic transformation and proliferation in hypertension. Cell Death Dis 2017; 8:e3074. [PMID: 28981106 PMCID: PMC5680591 DOI: 10.1038/cddis.2017.470] [Citation(s) in RCA: 189] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 08/17/2017] [Accepted: 08/22/2017] [Indexed: 01/12/2023]
Abstract
Inflammation is involved in pathogenesis of hypertension. NLRP3 inflammasome activation is a powerful mediator of inflammatory response via caspase-1 activation. The present study was designed to determine the roles and mechanisms of NLRP3 inflammasome in phenotypic modulation and proliferation of vascular smooth muscle cells (VSMCs) in hypertension. Experiments were conducted in spontaneously hypertensive rats (SHR) and primary aortic VSMCs. NLRP3 inflammasome activation was observed in the media of aorta in SHR and in the VSMCs from SHR. Knockdown of NLRP3 inhibited inflammasome activation, VSMC phenotypic transformation and proliferation in SHR-derived VSMCs. Increased NFκB activation, histone acetylation and histone acetyltransferase expression were observed in SHR-derived VSMCs and in media of aorta in SHR. Chromatin immunoprecipitation analysis revealed the increased histone acetylation, p65-NFκB and Pol II occupancy at the NLRP3 promoter in vivo and in vitro. Inhibition of NFκB with BAY11-7082 or inhibition of histone acetyltransferase with curcumin prevented the NLRP3 inflammasome activation, VSMC phenotype switching and proliferation in VSMCs from SHR. Moreover, curcumin repressed NFκB activation. Silencing of NLRP3 gene ameliorated hypertension, vascular remodeling, NLRP3 inflammasome activation and phenotype switching in the aorta of SHR. These results indicate that NLRP3 inflammasome activation response to histone acetylation and NFκB activation contributes to VSMC phenotype switching and proliferation and vascular remodeling in hypertension.
Collapse
Affiliation(s)
- Hai-Jian Sun
- Department of Physiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 210029, China.,Department of Basic Medicine, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Xing-Sheng Ren
- Department of Physiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Xiao-Qing Xiong
- Department of Physiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yun-Zhi Chen
- Department of Physiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Ming-Xia Zhao
- Department of Physiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Jue-Jin Wang
- Department of Physiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Ye-Bo Zhou
- Department of Physiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Ying Han
- Department of Physiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Qi Chen
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yue-Hua Li
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| | - Yu-Ming Kang
- Department of Physiology and Pathophysiology, Cardiovascular Research Center, Xi'an Jiaotong University School of Medicine, Xi'an, Shanxi 710061, China
| | - Guo-Qing Zhu
- Department of Physiology, Key Laboratory of Cardiovascular Disease and Molecular Intervention, Nanjing Medical University, Nanjing, Jiangsu 210029, China.,Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 210029, China
| |
Collapse
|
12
|
Yuan F, Zhang Y, Ma L, Cheng Q, Li G, Tong T. Enhanced NOLC1 promotes cell senescence and represses hepatocellular carcinoma cell proliferation by disturbing the organization of nucleolus. Aging Cell 2017; 16:726-737. [PMID: 28493459 PMCID: PMC5506443 DOI: 10.1111/acel.12602] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2017] [Indexed: 01/11/2023] Open
Abstract
The nucleolus is a key organelle that is responsible for the synthesis of rRNA and assembly of ribosomal subunits, which is also the center of metabolic control because of the critical role of ribosomes in protein synthesis. Perturbations of rRNA biogenesis are closely related to cell senescence and tumor progression; however, the underlying molecular mechanisms are not well understood. Here, we report that cellular senescence‐inhibited gene (CSIG) knockdown up‐regulated NOLC1 by stabilizing the 5′UTR of NOLC1 mRNA, and elevated NOLC1 induced the retention of NOG1 in the nucleolus, which is responsible for rRNA processing. Besides, the expression of NOLC1 was negatively correlated with CSIG in the aged mouse tissue and replicative senescent 2BS cells, and the down‐regulation of NOLC1 could rescue CSIG knockdown‐induced 2BS senescence. Additionally, NOLC1 expression was decreased in human hepatocellular carcinoma (HCC) tissue, and the ectopic expression of NOLC1 repressed the proliferation of HCC cells and tumor growth in a HCC xenograft model.
Collapse
Affiliation(s)
- Fuwen Yuan
- Peking University Research Center on Aging; Department of Biochemistry and Molecular Biology; Peking University Health Science Center, Beijing, Key Laboratory of Protein Posttranslational Modifications and Cell Function; Beijing 100191 China
| | - Yu Zhang
- Peking University Research Center on Aging; Department of Biochemistry and Molecular Biology; Peking University Health Science Center, Beijing, Key Laboratory of Protein Posttranslational Modifications and Cell Function; Beijing 100191 China
| | - Liwei Ma
- Peking University Research Center on Aging; Department of Biochemistry and Molecular Biology; Peking University Health Science Center, Beijing, Key Laboratory of Protein Posttranslational Modifications and Cell Function; Beijing 100191 China
| | - Qian Cheng
- Peking University Research Center on Aging; Department of Biochemistry and Molecular Biology; Peking University Health Science Center, Beijing, Key Laboratory of Protein Posttranslational Modifications and Cell Function; Beijing 100191 China
- Department of Hepatobilliary Surgery; Beijing, Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver Cancer; Peking University People's Hospital; Beijing 100044 China
| | - Guodong Li
- Peking University Research Center on Aging; Department of Biochemistry and Molecular Biology; Peking University Health Science Center, Beijing, Key Laboratory of Protein Posttranslational Modifications and Cell Function; Beijing 100191 China
| | - Tanjun Tong
- Peking University Research Center on Aging; Department of Biochemistry and Molecular Biology; Peking University Health Science Center, Beijing, Key Laboratory of Protein Posttranslational Modifications and Cell Function; Beijing 100191 China
| |
Collapse
|
13
|
Li Y, Liu S, Han M, Lu H, Wang Q, Zhang Y, Tursun K, Li Z, Feng S, Cheng J. NS5ATP13 Promotes Liver Fibrogenesis Via Activation of Hepatic Stellate Cells. J Cell Biochem 2017; 118:2463-2473. [PMID: 28133777 DOI: 10.1002/jcb.25913] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 01/25/2017] [Indexed: 12/28/2022]
Abstract
Liver fibrosis is a reversible wound-healing response to any etiology of chronic hepatic injuries. Activation of hepatic stellate cells (HSCs) is the key event in liver fibrogenesis. Generally, persistent activation and proliferation of HSCs results in liver fibrosis progression, while primary mechanisms of liver fibrosis resolution are apoptosis and reversion to a quiescent phenotype of activated HSCs. NS5ATP13 (HCV NS5A-transactivated protein 13) is involved in nucleologenesis and tumorigenesis, but its role in liver fibrosis and HSC activation remains unclear. This study found that NS5ATP13 was upregulated in both fibrotic liver tissues and activated human HSCs induced by TGF-β1. Moreover, NS5ATP13 enhanced extracellular matrix (ECM) production and HSC activation, with or without TGF-β1 treatment, likely involving the TGF-β1/Smad3 signaling pathway. Additionally, NS5ATP13 boosted HSC proliferation by inhibiting cell apoptosis. Furthermore, HCV NS5A promoted the profibrogenic effect of NS5ATP13 partly through TGF-β1 and NF-κB p65 (RelA) upregulation. Meanwhile, NS5ATP13 was required for the pro-fibrogenic effect of NF-κB. Moreover, NS5ATP13 and NF-κB phosphorylation as well as HSC activation were reduced by CX-4945, a CK2 specific inhibitor. These findings indicated that NS5ATP13 acts as a profibrogenic factor, providing a potential target for antifibrotic therapies. J. Cell. Biochem. 118: 2463-2473, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yaru Li
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100015, China
| | - Shunai Liu
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100015, China
| | - Ming Han
- Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100015, China.,Beijing Ditan Hospital, Teaching Hospital of Peking University, Beijing, 100015, China
| | - Hongping Lu
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100015, China
| | - Qi Wang
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100015, China
| | - Yu Zhang
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100015, China
| | - Kelbinur Tursun
- Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100015, China.,The First Affiliated Hospital of Xinjiang Medical University, Urumqi, 830054, China
| | - Zhongshu Li
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100015, China
| | - Shenghu Feng
- Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100015, China.,Beijing Ditan Hospital, Teaching Hospital of Peking University, Beijing, 100015, China
| | - Jun Cheng
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, 100015, China.,Beijing Key Laboratory of Emerging Infectious Diseases, Beijing, 100015, China
| |
Collapse
|
14
|
Kalashnikova AA, Rogge RA, Hansen JC. Linker histone H1 and protein-protein interactions. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1859:455-61. [PMID: 26455956 PMCID: PMC4775371 DOI: 10.1016/j.bbagrm.2015.10.004] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 09/21/2015] [Accepted: 10/05/2015] [Indexed: 01/11/2023]
Abstract
Linker histones H1 are ubiquitous chromatin proteins that play important roles in chromatin compaction, transcription regulation, nucleosome spacing and chromosome spacing. H1 function in DNA and chromatin structure stabilization is well studied and established. The current paradigm of linker histone mode of function considers all other cellular roles of linker histones to be a consequence from H1 chromatin compaction and repression. Here we review the multiple processes regulated by linker histones and the emerging importance of protein interactions in H1 functioning. We propose a new paradigm which explains the multi functionality of linker histones through linker histones protein interactions as a way to directly regulate recruitment of proteins to chromatin.
Collapse
Affiliation(s)
- Anna A Kalashnikova
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523-1870, USA
| | - Ryan A Rogge
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523-1870, USA
| | - Jeffrey C Hansen
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523-1870, USA.
| |
Collapse
|
15
|
Qian K, Zhang J, Lu J, Liu W, Yao X, Chen Q, Lu S, Xiang G, Liu H. FAM172A modulates apoptosis and proliferation of colon cancer cells via STAT1 binding to its promoter. Oncol Rep 2015; 35:1273-80. [PMID: 26676844 PMCID: PMC4750756 DOI: 10.3892/or.2015.4485] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Accepted: 11/12/2015] [Indexed: 12/19/2022] Open
Abstract
In our previous study, low expression of FAM172A protein was found in colon cancer tissues. This research was planned to explore the functions of FAM172A gene and examine the mechanisms of its transcriptional regulation. Firstly, flow cytometry showed that FAM172A inhibited proliferation and promoted apoptosis and differentiation of colon cancer cells. Then through continuous truncation, we identified the minimal functional promoter region of FAM172A. Subsequently, we found that STAT1, as a transcription factor, could bind to the minimal FAM172A promoter, as evaluated using Chromatin immunoprecipitation (ChIP) and Electrophoreticmobility shift assay (EMSA). The results of Western blot analysis and qRT-PCR indicated that STAT1 was able to upregulate the expression of FAM172A. Our results showed that FAM172A could suppress proliferation of colon cancer cells, and STAT1 could bind to the minimum promoter region of FAM172A and upregulated the expression of FAM172A. These results may provide advanced insights into the functions of FAM172A and its regulatory mechanisms.
Collapse
Affiliation(s)
- Kai Qian
- Department of Vascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Jinqian Zhang
- Department of Laboratory Medicine, The Second People's Hospital of Guangdong Province, Southern Medical University, Guangzhou, Guangdong 510317, P.R. China
| | - Jingbo Lu
- Department of Vascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Wenjun Liu
- Department of Vascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Xingxing Yao
- Department of Vascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Qing Chen
- Department of Vascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Shun Lu
- Department of Vascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| | - Guoan Xiang
- Department of General Surgery, The Second People's Hospital of Guangdong Province, Southern Medical University, Guangzhou, Guangdong 510317, P.R. China
| | - Hao Liu
- Department of Vascular Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong 510515, P.R. China
| |
Collapse
|
16
|
Lee HY, Choi K, Oh H, Park YK, Park H. HIF-1-dependent induction of Jumonji domain-containing protein (JMJD) 3 under hypoxic conditions. Mol Cells 2014; 37:43-50. [PMID: 24552709 PMCID: PMC3907005 DOI: 10.14348/molcells.2014.2250] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 11/17/2013] [Accepted: 11/18/2013] [Indexed: 01/20/2023] Open
Abstract
Jumonji domain-containing proteins (JMJD) catalyze the oxidative demethylation of a methylated lysine residue of histones by using O2, α-ketoglutarate, vitamin C, and Fe(II). Several JMJDs are induced by hypoxic stress to compensate their presumed reduction in catalytic activity under hypoxia. In this study, we showed that an H3K27me3 specific histone demethylase, JMJD3 was induced by hypoxia-inducible factor (HIF)-1α/β under hypoxia and that treatment with Clioquinol, a HIF-1α activator, increased JMJD3 expression even under normoxia. Chromatin immunoprecipitation (ChIP) analyses showed that both HIF-1α and its dimerization partner HIF-1β/Arnt occupied the first intron region of the mouse JMJD3 gene, whereas the HIF-1α/β heterodimer bound to the upstream region of the human JMJD3, indicating that human and mouse JMJD3 have hypoxia-responsive regulatory regions in different locations. This study shows that both mouse and human JMJD3 are induced by HIF-1.
Collapse
Affiliation(s)
- Ho-Youl Lee
- Department of Life Science, University of Seoul, Seoul 130-743,
Korea
| | - Kang Choi
- Department of Life Science, University of Seoul, Seoul 130-743,
Korea
| | - Hookeun Oh
- Department of Life Science, University of Seoul, Seoul 130-743,
Korea
| | - Young-Kwon Park
- Department of Life Science, University of Seoul, Seoul 130-743,
Korea
| | - Hyunsung Park
- Department of Life Science, University of Seoul, Seoul 130-743,
Korea
| |
Collapse
|
17
|
Duan X, Zhang J, Liu S, Zhang M, Wang Q, Cheng J. Methylation of nucleolar and coiled-body phosphoprotein 1 is associated with the mechanism of tumorigenesis in hepatocellular carcinoma. Oncol Rep 2013; 30:2220-8. [PMID: 23970161 DOI: 10.3892/or.2013.2676] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 06/25/2013] [Indexed: 11/06/2022] Open
Abstract
Nucleolar and coiled-body phosphoprotein 1 (NOLC1) plays an essential role in the synthesis of rRNA and the biosynthesis of ribosomes. Previous studies suggest that NOLC1 is crucial for normal cell growth, and plays a role in the regulation of tumorigenesis of nasopharyngeal carcinoma (NPC) and demonstrate that both NOLC1 and tumor protein 53 work synergistically to activate the MDM2 promoter in NPC cells. Yet, the functioning of NOLC1 in liver cancer remains unknown. The aim of the present study was to understand how the NOLC1 gene is regulated in liver carcinogenesis. In this study, we showed that NOLC1 was silenced or downregulated in liver tumor tissues when compared with that in the matched non-cancer tissues. In addition, human hepatoma cells weakly expressed NOLC1, whereas cultured human normal liver cell lines expressed abundant levels. The hypermethylation status in the promoter CpG1 start region appeared to be correlated with the NOLC1 expression levels in liver cell lines or liver normal and tissue specimens. We found that four CpG dinucleotides were located at the CpG1 start region. Further molecular analysis of mutagenesis indicated that the four CpG dinucleotides play a role in the promoter activity of the NOLC1 gene. The expression of NOLC1 and DNA methylation of its promoter affected cell proliferation and apoptosis. The expression of NOLC1 in hepatoma cell lines was restored following exposure to the demethylation agent, 5-azacytidine. Low expression of NOLC1 in hepatoma cell lines and liver cancer tissues was associated with cyclin D3. In conclusion, our study demonstrated that DNA methylation is a key mechanism of silenced NOLC1 expression in human hepatocellular carcinoma cells, and NOLC1 gene hypermethylation of the four CpG dinucleotides is a potential biomarker for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Xuefei Duan
- Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, P.R. China
| | | | | | | | | | | |
Collapse
|
18
|
Yun SP, Park SS, Ryu JM, Park JH, Kim MO, Lee JH, Han HJ. Mechanism of PKA-dependent and lipid-raft independent stimulation of Connexin43 expression by oxytoxin in mouse embryonic stem cells. Mol Endocrinol 2012; 26:1144-57. [PMID: 22564436 DOI: 10.1210/me.2011-1343] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Previous studies shows that connexins appear very early during murine embryo development, the gap junctional intercellular communication found in the inner cell mass of early embryo is also maintained in embryonic stem cells (ESC), and expression of oxytocin receptor (OTR) is developmentally regulated at early embryonic development. However, effect of oxytocin (OT) on the regulation of the connexin43 (Cx43) and maintenance of undifferentiation is not fully understood in stem cells. Therefore, we investigated the effect of OT on Cx43 expression and related signaling cascades in mouse ESC. OT increased Cx43 expression that was inhibited by the OTR inhibitor atosiban. In experiments to examine whether the effect of OT depends on lipid rafts, caveolin-1 (cav-1), cav-2, and flotillin-2, but not OTR, were detected in lipid raft fractions. Also, colocalization of OTR, cav-1, and cav-2 was not detected. Moreover, the lipid raft disruptor methyl-β-cyclodextrin did not attenuate OT-induced Cx43 expression. In experiments to examine related signaling pathways, OT activated cAMP/protein kinase A (PKA) which was inhibited by adenylyl cyclase inhibitor SQ 22536 and PKA inhibitor PKI. OT increased nuclear factor κ-light-chain-enhancer of activated B cells (NF-κB) phosphorylation which was inhibited by PKI. OT also increased cAMP response element-binding (CREB)/CREB-binding protein (CBP) expression in the nucleus and induced the formation of CREB1/NF-κB/CBP complexes, which was blocked by the NF-κB-specific small interfering RNA, NF-κB inhibitors, SN50, and bay11-7082. Complex disruption by NF-κB inhibitors decreased OT-induced Cx43 expression. In conclusion, OT stimulates Cx43 expression through the NF-κB/CREB/CBP complex via the lipid raft-independent OTR-mediated cAMP/PKA in mouse ESC.
Collapse
Affiliation(s)
- Seung Pil Yun
- Department of Veterinary Physiology, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|