1
|
Pankammoon P, Qing Y, Zhao H, Jiao D, Li H, Wang F, Wiriyahdamrong T, Guo J, Li W, Chuammitri P, Thitaram C, Wei H, Sathanowongs A. Transcriptomic insights into developmental arrest in fluorescent labeling transgenic Asian elephant ( Elephas maximus) embryos via inter-order cloning. Front Cell Dev Biol 2025; 13:1532962. [PMID: 40092629 PMCID: PMC11907086 DOI: 10.3389/fcell.2025.1532962] [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: 11/22/2024] [Accepted: 01/27/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction Asian elephants (Elephas maximus) provide a unique model for studying cloning in large mammals. As an endangered species with declining populations and limited oocyte availability, interspecies somatic cell nuclear transfer (iSCNT) combined with transcriptomic analysis holds promise for advancing iSCNT embryonic arrest development and further facilitating applications in conservation efforts, therapeutic cloning, and regenerative medicine. Methods This study conducted low-input RNA sequencing analyses on transgenic Asian elephant-pig (AE-P) inter-order cloned embryos expressing enhanced green fluorescent protein (EGFP) at the 2- and 4-cell stages. Differential gene expressions, pathway enrichment, and hub gene analyses were performed to identify the molecular mechanisms and core genes influencing normal and arrest development. Results and Discussion Approximately 25% of clean reads successfully aligned with the Asian elephant genome. The transcriptomic analysis revealed that inter-order cloned embryos with earlier cleavage at the 2- and 4-cell stages exhibited signs of residual transcriptomic memory and incomplete epigenetic reprogramming, while arrested embryos showed indications of nucleocytoplasmic incompatibility and nDNA-mtDNA mismatch. Hub gene analyses indicated core genes such as NDUFC2, NDUFS3, NDUFAB1, SDHC, SDHB, NUP54, NUP43, NUP37, NDC1, CDK1, and CCNB1 linked to energy production, nucleocytoplasmic transport, and cell cycle regulation highlighting the overall challenges in cloning Asian elephant inter-order embryos. Altogether, the analysis of high-throughput sequencing enhances the reliability of iSCNT production in this study, advancing our understanding of cellular reprogramming and molecular roadblocks in AE-P inter-order cloned embryos. Transcriptomic analyses have identified key factors contributing to developmental barriers in iSCNT, offering valuable insights into the complexities of these challenges.
Collapse
Affiliation(s)
| | - Yubo Qing
- Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
- Science and Technology Department of Yunnan Province, Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Kunming, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Heng Zhao
- Science and Technology Department of Yunnan Province, Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Kunming, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Deling Jiao
- Science and Technology Department of Yunnan Province, Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Kunming, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Honghui Li
- Science and Technology Department of Yunnan Province, Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Kunming, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Fengchong Wang
- Science and Technology Department of Yunnan Province, Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Kunming, China
- College of Animal Science, Yunnan Agricultural University, Kunming, China
| | - Thanapa Wiriyahdamrong
- Science and Technology Department of Yunnan Province, Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Kunming, China
- College of Animal Science, Yunnan Agricultural University, Kunming, China
| | - Jianxiong Guo
- Science and Technology Department of Yunnan Province, Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Kunming, China
| | - Wengui Li
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | | | - Chatchote Thitaram
- Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Hongjiang Wei
- Science and Technology Department of Yunnan Province, Yunnan Province Key Laboratory for Porcine Gene Editing and Xenotransplantation, Kunming, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | | |
Collapse
|
2
|
Mohamadynejad P, Moghanibashi M, Bagheri K. Identification of novel nuclear pore complex associated proteins in esophageal carcinoma by an integrated bioinformatics analysis. J Biomol Struct Dyn 2024; 42:7221-7232. [PMID: 37504972 DOI: 10.1080/07391102.2023.2240414] [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: 01/29/2023] [Accepted: 07/15/2023] [Indexed: 07/29/2023]
Abstract
Nucleoporins (NUPs) are components of the nuclear pore complex (NPC) that participate in the nucleocytoplasmic transport of macromolecules as well as in many essential processes that may be led to carcinogenesis. We selected three expression profile microarray datasets from GEO and as well as TCGA data to identify differentially expressed NUPs genes in esophageal carcinoma. Our findings indicated that NUP133, NUP37, NUP43, NUP50, GLE1 and NDC1 are overexpressed in esophageal carcinoma, among which NUP50 and GLE1genes are reported for the first time in esophageal carcinoma. All identified NUPs were also associated with distant metastasis.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Parisa Mohamadynejad
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Mehdi Moghanibashi
- Department of Genetics, Faculty of Medicine, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| | - Kambiz Bagheri
- Department of Immunology, Faculty of Medicine, Kazerun Branch, Islamic Azad University, Kazerun, Iran
| |
Collapse
|
3
|
Lv Y, Wang C, Liu R, Wu S, Chen J, Zheng X, Jiang T, Chen L. NUP37 promotes the proliferation and invasion of glioma cells through DNMT1-mediated methylation. Cell Death Discov 2024; 10:373. [PMID: 39174498 PMCID: PMC11341718 DOI: 10.1038/s41420-024-02138-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 07/30/2024] [Accepted: 08/07/2024] [Indexed: 08/24/2024] Open
Abstract
Nuclear regulation has potential in cancer therapy, with the nuclear pore complex (NPC) serving as a critical channel between the nucleus and cytoplasm, playing a role in regulating various biological processes and cancer. DNA methylation, an epigenetic modification mediated by DNA methyltransferases (DNMTs), influences gene expression and cell differentiation, and is crucial for the development and progression of tumor cells. Gliomas are the most common primary brain tumors, with glioblastoma being particularly aggressive, characterized by invasiveness, migration capability, and resistance to conventional treatments, resulting in poor prognosis. Our study revealed that the expression level of NUP37 affects the proliferation and invasion of glioma cells, and that the overexpression of DNMT1 can alleviate the adverse effects caused by NUP37 depletion. These findings suggest that NUP37 promotes the proliferation and invasion of glioma cells through its interaction with DNMT1.
Collapse
Affiliation(s)
- Yongqiang Lv
- Department of Neurosurgery, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
| | - Chaolian Wang
- Department of Neurosurgery, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
| | - Ruoyu Liu
- Department of Neurosurgery, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
| | - Shaoxian Wu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
| | - Junjun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
| | - Xiao Zheng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China
| | - Tianwei Jiang
- Department of Neurosurgery, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China.
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China.
| | - Lujun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China.
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China.
- Institute of Cell Therapy, The Third Affiliated Hospital of Suzhou University, Changzhou, Jiangsu, China.
| |
Collapse
|
4
|
Li Y, Zhu J, Zhai F, Kong L, Li H, Jin X. Advances in the understanding of nuclear pore complexes in human diseases. J Cancer Res Clin Oncol 2024; 150:374. [PMID: 39080077 PMCID: PMC11289042 DOI: 10.1007/s00432-024-05881-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 07/03/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND Nuclear pore complexes (NPCs) are sophisticated and dynamic protein structures that straddle the nuclear envelope and act as gatekeepers for transporting molecules between the nucleus and the cytoplasm. NPCs comprise up to 30 different proteins known as nucleoporins (NUPs). However, a growing body of research has suggested that NPCs play important roles in gene regulation, viral infections, cancer, mitosis, genetic diseases, kidney diseases, immune system diseases, and degenerative neurological and muscular pathologies. PURPOSE In this review, we introduce the structure and function of NPCs. Then We described the physiological and pathological effects of each component of NPCs which provide a direction for future clinical applications. METHODS The literatures from PubMed have been reviewed for this article. CONCLUSION This review summarizes current studies on the implications of NPCs in human physiology and pathology, highlighting the mechanistic underpinnings of NPC-associated diseases.
Collapse
Affiliation(s)
- Yuxuan Li
- The Affiliated Lihuili Hospital of Ningbo University, Ningbo, 315040, Zhejiang, China
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Nngbo University, Ningbo, 315211, Zhejiang, China
| | - Jie Zhu
- The Affiliated Lihuili Hospital of Ningbo University, Ningbo, 315040, Zhejiang, China
| | - Fengguang Zhai
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Nngbo University, Ningbo, 315211, Zhejiang, China
| | - Lili Kong
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Nngbo University, Ningbo, 315211, Zhejiang, China
| | - Hong Li
- The Affiliated Lihuili Hospital of Ningbo University, Ningbo, 315040, Zhejiang, China.
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Nngbo University, Ningbo, 315211, Zhejiang, China.
| | - Xiaofeng Jin
- The Affiliated Lihuili Hospital of Ningbo University, Ningbo, 315040, Zhejiang, China.
- Department of Biochemistry and Molecular Biology, and Zhejiang Key Laboratory of Pathophysiology, Health Science Center, Nngbo University, Ningbo, 315211, Zhejiang, China.
| |
Collapse
|
5
|
Ester L, Cabrita I, Ventzke M, Kieckhöfer E, Christodoulou M, Mandel AM, Diefenhardt P, Fabretti F, Benzing T, Habbig S, Schermer B. The role of the FSGS disease gene product and nuclear pore protein NUP205 in regulating nuclear localization and activity of transcriptional regulators YAP and TAZ. Hum Mol Genet 2023; 32:3153-3165. [PMID: 37565816 PMCID: PMC10630254 DOI: 10.1093/hmg/ddad135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 07/31/2023] [Accepted: 08/10/2023] [Indexed: 08/12/2023] Open
Abstract
Mutations in genes encoding nuclear pore proteins (NUPs) lead to the development of steroid-resistant nephrotic syndrome and focal segmental glomerulosclerosis (FSGS). However, the precise molecular mechanisms by which NUP dysfunction contributes to podocyte injury preceding FSGS remain unclear. The tightly regulated activity of Yes-associated protein (YAP) and WW-domain-containing transcription regulator 1 (TAZ), the transcriptional effectors of the Hippo pathway, is crucial for podocytes and the maintenance of the glomerular filter. In this study, we investigate the impact of NUPs on the regulation of YAP/TAZ nuclear import and activity in podocytes. In unbiased interactome studies using quantitative label-free mass spectrometry, we identify the FSGS disease gene products NUP107, NUP133, NUP205, and Exportin-5 (XPO5) as components of YAP and TAZ protein complexes in podocytes. Moreover, we demonstrate that NUP205 is essential for YAP/TAZ nuclear import. Consistently, both the nuclear interaction of YAP/TAZ with TEA domain transcription factor 1 and their transcriptional activity were dependent on NUP205 expression. Additionally, we elucidate a regulatory feedback mechanism whereby YAP activity is modulated in response to TAZ-mediated NUP205 expression. In conclusion, this study establishes a connection between the FSGS disease protein NUP205 and the activity of the transcriptional regulators and Hippo effectors YAP and TAZ and it proposes a potential pathological role of YAP/TAZ dysregulation in podocytes of patients with pathogenic NUP205 variants.
Collapse
Affiliation(s)
- Lioba Ester
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Inês Cabrita
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Michel Ventzke
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Emilia Kieckhöfer
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Marita Christodoulou
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Amrei M Mandel
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Paul Diefenhardt
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Francesca Fabretti
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Thomas Benzing
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Sandra Habbig
- Department of Pediatrics, University of Cologne, Faculty of Medicine and University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Bernhard Schermer
- Department II of Internal Medicine and Center for Molecular Medicine Cologne, University of Cologne, Faculty of Medicine and University Hospital Cologne, Kerpener Str. 62, 50937 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Faculty of Medicine and University Hospital Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| |
Collapse
|
6
|
Kofler M, Kapus A. Nuclear Import and Export of YAP and TAZ. Cancers (Basel) 2023; 15:4956. [PMID: 37894323 PMCID: PMC10605228 DOI: 10.3390/cancers15204956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/05/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
Yes-associated Protein (YAP) and its paralog Transcriptional Coactivator with PDZ-binding Motif (TAZ) are major regulators of gene transcription/expression, primarily controlled by the Hippo pathway and the cytoskeleton. Integrating an array of chemical and mechanical signals, they impact growth, differentiation, and regeneration. Accordingly, they also play key roles in tumorigenesis and metastasis formation. Their activity is primarily regulated by their localization, that is, Hippo pathway- and/or cytoskeleton-controlled cytosolic or nuclear sequestration. While many details of such prevailing retention models have been elucidated, much less is known about their actual nuclear traffic: import and export. Although their size is not far from the cutoff for passive diffusion through the nuclear pore complex (NPC), and they do not contain any classic nuclear localization (NLS) or nuclear export signal (NES), evidence has been accumulating that their shuttling involves mediated and thus regulatable/targetable processes. The aim of this review is to summarize emerging information/concepts about their nucleocytoplasmic shuttling, encompassing the relevant structural requirements (NLS, NES), nuclear transport receptors (NTRs, karyophererins), and NPC components, along with the potential transport mechanisms and their regulation. While dissecting retention vs. transport is often challenging, the emerging picture suggests that YAP/TAZ shuttles across the NPC via multiple, non-exclusive, mediated mechanisms, constituting a novel and intriguing facet of YAP/TAZ biology.
Collapse
Affiliation(s)
- Michael Kofler
- Keenan Research Centre for Biomedical Science of the St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada;
| | - András Kapus
- Keenan Research Centre for Biomedical Science of the St. Michael’s Hospital, Toronto, ON M5B 1W8, Canada;
- Department of Surgery, University of Toronto, Toronto, ON M5T 1P5, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5B 1T8, Canada
| |
Collapse
|
7
|
Liu G, Guo Z, Zhao X, Sun J, Yue S, Li M, Chen Z, Ma Z, Zhao H. Whole Genome Resequencing Identifies Single-Nucleotide Polymorphism Markers of Growth and Reproduction Traits in Zhedong and Zi Crossbred Geese. Genes (Basel) 2023; 14:487. [PMID: 36833414 PMCID: PMC9956059 DOI: 10.3390/genes14020487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/08/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
The broodiness traits of domestic geese are a bottleneck that prevents the rapid development of the goose industry. To reduce the broodiness of the Zhedong goose and thus improve it, this study hybridized it with the Zi goose, which has almost no broody behavior. Genome resequencing was performed for the purebred Zhedong goose, as well as the F2 and F3 hybrids. The results showed that the F1 hybrids displayed significant heterosis in growth traits, and their body weight was significantly greater than those of the other groups. The F2 hybrids showed significant heterosis in egg-laying traits, and the number of eggs laid was significantly greater than those of the other groups. A total of 7,979,421 single-nucleotide polymorphisms (SNPs) were obtained, and three SNPs were screened. Molecular docking results showed that SNP11 located in the gene NUDT9 altered the structure and affinity of the binding pocket. The results suggested that SNP11 is an SNP related to goose broodiness. In the future, we will use the cage breeding method to sample the same half-sib families to accurately identify SNP markers of growth and reproductive traits.
Collapse
Affiliation(s)
- Guojun Liu
- Heilongjiang Academy of Agricultural Sciences, Animal Husbandry Research Institute, No. 368 Xuefu Road, Harbin 150086, China
| | - Zhenhua Guo
- Heilongjiang Academy of Agricultural Sciences, Animal Husbandry Research Institute, No. 368 Xuefu Road, Harbin 150086, China
| | - Xiuhua Zhao
- Heilongjiang Academy of Agricultural Sciences, Animal Husbandry Research Institute, No. 368 Xuefu Road, Harbin 150086, China
| | - Jinyan Sun
- Heilongjiang Academy of Agricultural Sciences, Animal Husbandry Research Institute, No. 368 Xuefu Road, Harbin 150086, China
| | - Shan Yue
- Heilongjiang Academy of Agricultural Sciences, Animal Husbandry Research Institute, No. 368 Xuefu Road, Harbin 150086, China
| | - Manyu Li
- Heilongjiang Academy of Agricultural Sciences, Animal Husbandry Research Institute, No. 368 Xuefu Road, Harbin 150086, China
| | - Zhifeng Chen
- Heilongjiang Academy of Agricultural Sciences, Qiqihare Branch Academy, No. 2 Heyi Road, Qiqihare 161005, China
| | - Zhigang Ma
- Heilongjiang Academy of Agricultural Sciences, Qiqihare Branch Academy, No. 2 Heyi Road, Qiqihare 161005, China
| | - Hui Zhao
- Liaoning Academy of Agricultural Sciences, No. 84 Dongling Road, Shenyang 110161, China
| |
Collapse
|
8
|
Fang Z, Wang F, Zhang M, Huang H, Lin Z. Identification of Co-Expression Modules and Genes Associated With Tumor Progression in Oral Squamous Cell Carcinoma. Pathol Oncol Res 2022; 28:1610481. [PMID: 36052378 PMCID: PMC9426548 DOI: 10.3389/pore.2022.1610481] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 06/28/2022] [Indexed: 12/24/2022]
Abstract
Oral squamous cell carcinoma (OSCC) is a common head-and-neck cancer with a deficiency of early diagnosis and poor prognosis. To identify potential diagnostic and prognostic markers of OSCC, we firstly used weighted gene co-expression network analysis (WGCNA) to build a co-expression module from GSE42743. Next, we performed Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses on specified units from selected modules utilizing Database for Annotation, Visualization, and Integrated Discovery (DAVID). Additionally, we identified and validate hub genes of these specified modules from multiple datasets like GEPIA and TCGA. In total 16 co-expression modules were built by 17,238 genes of 74 tumor samples utilizing WGCNA. Through pathway and functional enrichment analysis, the turquoise module was most firmly relevant to the cell cycle, oocyte meiosis, and p53 signaling pathway. Hub genes VRK1, NUP37, HMMR, SPC25, and RUVBL1 were identified to be related to oral cancer at both molecular level and clinical levels. The expressions of these genes differed in tumor tissues and normal tissues. Meanwhile, patients with high hub gene expression had a poor prognosis clinically. To conclude, five hub genes were identified to be relevant to oral cancer from the molecular level and the clinical level. Therefore, the detection of these genes was of great significance. They can be regarded as diagnostic and prognostic biomarkers for oral cancer. Also, they could shed light on the improvement of patients’ overall survival and prognosis, which needs further analysis in the future.
Collapse
Affiliation(s)
- Zhijie Fang
- Department of Otolaryngology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Feifei Wang
- Department of Nursing, Suzhou BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Suzhou, China
| | - Mengya Zhang
- Department of Otolaryngology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Hua Huang
- Department of Otolaryngology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Zhiqiang Lin
- Department of Otolaryngology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
- *Correspondence: Zhiqiang Lin,
| |
Collapse
|
9
|
Vrazas V, Moustafa S, Makridakis M, Karakasiliotis I, Vlahou A, Mavromara P, Katsani KR. A Proteomic Approach to Study the Biological Role of Hepatitis C Virus Protein Core+1/ARFP. Viruses 2022; 14:v14081694. [PMID: 36016316 PMCID: PMC9518822 DOI: 10.3390/v14081694] [Citation(s) in RCA: 3] [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: 07/02/2022] [Revised: 07/26/2022] [Accepted: 07/28/2022] [Indexed: 11/16/2022] Open
Abstract
Hepatitis C virus is the major cause of chronic liver diseases and the only cytoplasmic RNA virus known to be oncogenic in humans. The viral genome gives rise to ten mature proteins and to additional proteins, which are the products of alternative translation initiation mechanisms. A protein-known as ARFP (alternative reading frame protein) or Core+1 protein-is synthesized by an open reading frame overlapping the HCV Core coding region in the (+1) frame of genotype 1a. Almost 20 years after its discovery, we still know little of the biological role of the ARFP/Core+1 protein. Here, our differential proteomic analysis of stable hepatoma cell lines expressing the Core+1/Long isoform of HCV-1a relates the expression of the Core+1/Long isoform with the progression of the pathology of HCV liver disease to cancer.
Collapse
Affiliation(s)
- Vasileios Vrazas
- Laboratory of Biochemistry and Molecular Virology, Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (V.V.); (P.M.)
| | - Savvina Moustafa
- Clinical Immunology-Rheumatology Unit, 2nd Department of Medicine and Laboratory, Hippokration General Hospital of Athens, 11527 Athens, Greece;
| | - Manousos Makridakis
- Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece; (A.V.); (M.M.)
| | - Ioannis Karakasiliotis
- Laboratory of Biology, Department of Medicine, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| | - Antonia Vlahou
- Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece; (A.V.); (M.M.)
| | - Penelope Mavromara
- Laboratory of Biochemistry and Molecular Virology, Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (V.V.); (P.M.)
| | - Katerina R. Katsani
- Laboratory of Biochemistry and Molecular Virology, Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece; (V.V.); (P.M.)
- Correspondence:
| |
Collapse
|
10
|
Tang Z, Yang Y, Chen W, Li E, Liang T. Demethylation at enhancer upregulates MCM2 and NUP37 expression predicting poor survival in hepatocellular carcinoma patients. J Transl Med 2022; 20:49. [PMID: 35093119 PMCID: PMC8800332 DOI: 10.1186/s12967-022-03249-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 01/14/2022] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Identification of novel biomarker is important for development of molecular-targeted therapy agents for patients with hepatocellular carcinoma (HCC). This study aims to identify potential prognostic biomarkers and investigate epigenetic mechanism of HCC development. METHODS Public bulk-RNA seq datasets and proteomic dataset were screened for identification of potential prognostic biomarkers for HCC patients. Public methylomic datasets were analyzed for deciphering the epigenetic mechanism regulating HCC-associated gene expression. Immunoblotting, immunohistochemistry, real-time PCR, and pyrosequencing were used to validate the findings from bioinformatic analyses. RESULTS Minichromosome maintenance complex component 2 (MCM2) and nucleoporin 37 (NUP37) were overexpressed in human HCC tissues and hepatoma cell lines. MCM2 significantly positively correlated with NUP37 expression. Higher expression of MCM2 or NUP37 was significantly associated with advanced tumor stage and worse overall survival in 3 large independent HCC cohorts (n = 820). MCM2 and NUP37 overexpression are independent prognostic risk factors for HCC patients. Demethylation at an enhancer of MCM2 gene was a common event in patients with HCC, which significantly negatively correlated with MCM2 and NUP37 mRNA expression. CONCLUSIONS Demethylation at enhancer regulates MCM2 and NUP37 expression in HCC. MCM2 and NUP37 are promising prognostic biomarkers and potential targets for epigenetic therapy in HCC patients.
Collapse
Affiliation(s)
- Zengwei Tang
- grid.13402.340000 0004 1759 700XDepartment of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003 Zhejiang China ,grid.13402.340000 0004 1759 700XZhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003 Zhejiang China ,Zhejiang Provincial Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, Hangzhou, 310003 Zhejiang China ,grid.13402.340000 0004 1759 700XCancer Center, Zhejiang University, Hangzhou, 310058 Zhejiang China
| | - Yuan Yang
- grid.506261.60000 0001 0706 7839Department of Hematology, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730 China
| | - Wen Chen
- grid.13402.340000 0004 1759 700XDepartment of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003 Zhejiang China ,grid.13402.340000 0004 1759 700XZhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003 Zhejiang China ,Zhejiang Provincial Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, Hangzhou, 310003 Zhejiang China ,grid.13402.340000 0004 1759 700XCancer Center, Zhejiang University, Hangzhou, 310058 Zhejiang China
| | - Enliang Li
- grid.13402.340000 0004 1759 700XDepartment of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003 Zhejiang China ,grid.13402.340000 0004 1759 700XZhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003 Zhejiang China ,Zhejiang Provincial Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, Hangzhou, 310003 Zhejiang China ,grid.13402.340000 0004 1759 700XCancer Center, Zhejiang University, Hangzhou, 310058 Zhejiang China
| | - Tingbo Liang
- grid.13402.340000 0004 1759 700XDepartment of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, 310003 Zhejiang China ,grid.13402.340000 0004 1759 700XZhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003 Zhejiang China ,Zhejiang Provincial Innovation Center for the Study of Pancreatic Diseases, Zhejiang Province, Hangzhou, 310003 Zhejiang China ,grid.13402.340000 0004 1759 700XCancer Center, Zhejiang University, Hangzhou, 310058 Zhejiang China ,grid.510538.a0000 0004 8156 0818Research Center for Healthcare Data Science, Zhejiang Lab, Hangzhou, 310003 Zhejiang China
| |
Collapse
|
11
|
Guo Q, Liu Q, Wang N, Wang J, Sun A, Qiao J, Yan L. The function of Nucleoporin 37 on mouse oocyte maturation and preimplantation embryo development. J Assist Reprod Genet 2022; 39:107-116. [PMID: 35022896 PMCID: PMC8866631 DOI: 10.1007/s10815-021-02330-x] [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: 05/19/2021] [Accepted: 09/20/2021] [Indexed: 01/03/2023] Open
Abstract
PURPOSE Nucleoporin 37 (NUP37) has been reported to activate the YAP-TEAD signaling, which is crucial for early embryo development. However, whether NUP37 is involved in oocyte meiosis and embryo development remains largely unknown. The study aimed to clarify the function of Nup37 in oocyte maturation and early embryo development, and to explore the mechanism. METHODS The expression level and subcellular localization of NUP37 were explored. After knocking down of Nup37 by microinjecting interfering RNA (siRNA), the oocyte maturation rate, aberrant PB1 extrusion rate, and blastocyst formation rate were evaluated. In addition, the effect of the downregulation of Nup37 on YAP-TEAD signaling was confirmed by immunofluorescence staining and real-time quantitative PCR. RESULTS NUP37 was highly expressed in oocytes and early embryos; it mainly localized to the nuclear periphery at mice GV stage oocytes and early embryos. Nup37 depletion led to aberrant PB1 extrusion at the MII stage oocyte and a decreased blastocyst formation rate. The reduction of NUP37 caused YAP1 mislocalization and decreased the expression of Tead1, Tead2, and Tead4 during mice embryo development, thus affecting the YAP-TEAD activity and embryo developmental competence. CONCLUSIONS In summary, NUP37 played an important role in mice oocyte maturation and preimplantation embryo development.
Collapse
Affiliation(s)
- Qianying Guo
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191 China ,National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191 China ,Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education Beijing Key, Beijing, 100191 China ,Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191 China ,Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences, Beijing, China
| | - Qiang Liu
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191 China ,National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191 China ,Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education Beijing Key, Beijing, 100191 China ,Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191 China ,Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences, Beijing, China
| | - Nan Wang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191 China ,National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191 China ,Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education Beijing Key, Beijing, 100191 China ,Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191 China ,Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences, Beijing, China
| | - Jing Wang
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191 China ,National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191 China ,Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education Beijing Key, Beijing, 100191 China ,Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191 China ,Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences, Beijing, China
| | - Andi Sun
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191 China ,National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191 China ,Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education Beijing Key, Beijing, 100191 China ,Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191 China ,Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences, Beijing, China
| | - Jie Qiao
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191 China ,National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191 China ,Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education Beijing Key, Beijing, 100191 China ,Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191 China ,Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences, Beijing, China
| | - Liying Yan
- Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191 China ,National Clinical Research Center for Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191 China ,Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education Beijing Key, Beijing, 100191 China ,Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology, Beijing, 100191 China ,Research Units of Comprehensive Diagnosis and Treatment of Oocyte Maturation Arrest, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
12
|
Zhang J, Lv W, Liu Y, Fu W, Chen B, Ma Q, Gao X, Cui X. Nucleoporin 37 promotes the cell proliferation, migration, and invasion of gastric cancer through activating the PI3K/AKT/mTOR signaling pathway. In Vitro Cell Dev Biol Anim 2021; 57:987-997. [PMID: 34888748 DOI: 10.1007/s11626-021-00627-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 10/03/2021] [Indexed: 02/07/2023]
Abstract
Gastric cancer is a kind of malignant tumor in the world. Emerging studies have proved the regulatory role of nucleoporin 37 in the development of several malignant tumors. However, the potential effect of NUP37 in gastric cancer is still unclear. In this study, we searched for the Cancer Genome Atlas analysis to explore the potential correlation between NUP37 and gastric cancer. Then, we analyzed NUP37 expression in gastric cancer tissues and cell lines. After constructing a NUP37-silenced model in NCI-N87 cells and a NUP37-overexpressed model in MKN45 cells, we evaluated the role of NUP37 in cell proliferation, migration, and invasion as well as its underlying mechanism. TCGA analysis showed that NUP37 expression was highly expressed in stomach adenocarcinoma, which showed a lower survival rate than normal samples. Moreover, NUP37 was found to be highly expressed in gastric cancer tissues and cell lines. Functionally, NUP37 deficiency promoted gastric cancer cell apoptosis and inhibited cell proliferation, migration, and invasion, whereas NUP37 overexpression exhibited the opposite results. Mechanically, upregulation of NUP37 activated the PI3K/AKT/mTOR signaling pathway. Furthermore, the rescue assay exhibited that the mTOR inhibitor rapamycin significantly reversed the promoting effect of NUP37 in cell proliferation, migration, and invasion. In conclusion, our study identified that NUP37 promoted malignant behavior of gastric cancer cells including invasion, proliferation, and migration through activating the PI3K and its downregulated signaling pathway, indicating that NUP37 might become a novel prognostic target for further gastric cancer therapy.
Collapse
Affiliation(s)
- Jishui Zhang
- The Second Department of General Surgery, Cangzhou Central Hospital, No. 16 Xinhua West Road, Cangzhou, 061001, Hebei, China.
| | - Wenhao Lv
- The Second Department of General Surgery, Cangzhou Central Hospital, No. 16 Xinhua West Road, Cangzhou, 061001, Hebei, China
| | - Yagang Liu
- The Second Department of General Surgery, Cangzhou Central Hospital, No. 16 Xinhua West Road, Cangzhou, 061001, Hebei, China
| | - Weihua Fu
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, 300052, China
| | - Baosheng Chen
- The Second Department of General Surgery, Cangzhou Central Hospital, No. 16 Xinhua West Road, Cangzhou, 061001, Hebei, China
| | - Qiutong Ma
- The Second Department of General Surgery, Cangzhou Central Hospital, No. 16 Xinhua West Road, Cangzhou, 061001, Hebei, China
| | - Xin Gao
- Department of Radiotherapy, Cangzhou Central Hospital, Cangzhou, 061001, Hebei, China
| | - Xiuxia Cui
- Department of Nuclear Medicine, Cangzhou Central Hospital, Cangzhou, 061001, Hebei, China
| |
Collapse
|
13
|
Sosnowska M, Kutwin M, Strojny B, Wierzbicki M, Cysewski D, Szczepaniak J, Ficek M, Koczoń P, Jaworski S, Chwalibog A, Sawosz E. Diamond Nanofilm Normalizes Proliferation and Metabolism in Liver Cancer Cells. Nanotechnol Sci Appl 2021; 14:115-137. [PMID: 34511890 PMCID: PMC8420805 DOI: 10.2147/nsa.s322766] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 07/28/2021] [Indexed: 01/10/2023] Open
Abstract
Purpose Surgical resection of hepatocellular carcinoma can be associated with recurrence resulting from the degeneration of residual volume of the liver. The objective was to assess the possibility of using a biocompatible nanofilm, made of a colloid of diamond nanoparticles (nfND), to fill the side after tumour resection and optimize its contact with proliferating liver cells, minimizing their cancerous transformation. Methods HepG2 and C3A liver cancer cells and HS-5 non-cancer cells were used. An aqueous colloid of diamond nanoparticles, which covered the cell culture plate, was used to create the nanofilm. The roughness of the resulting nanofilm was measured by atomic force microscopy. Mitochondrial activity and cell proliferation were measured by XTT and BrdU assays. Cell morphology and a scratch test were used to evaluate the invasiveness of cells. Flow cytometry determined the number of cells within the cell cycle. Protein expression in was measured by mass spectrometry. Results The nfND created a surface with increased roughness and exposed oxygen groups compared with a standard plate. All cell lines were prone to settling on the nanofilm, but cancer cells formed more relaxed clusters. The surface compatibility was dependent on the cell type and decreased in the order C3A >HepG2 >HS-5. The invasion was reduced in cancer lines with the greatest effect on the C3A line, reducing proliferation and increasing the G2/M cell population. Among the proteins with altered expression, membrane and nuclear proteins dominated. Conclusion In vitro studies demonstrated the antiproliferative properties of nfND against C3A liver cancer cells. At the same time, the need to personalize potential therapy was indicated due to the differential protein synthetic responses in C3A vs HepG2 cells. We documented that nfND is a source of signals capable of normalizing the expression of many intracellular proteins involved in the transformation to non-cancerous cells.
Collapse
Affiliation(s)
- Malwina Sosnowska
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Marta Kutwin
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Barbara Strojny
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Mateusz Wierzbicki
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Dominik Cysewski
- Spectrometry Laboratory, Institute of Biochemistry and Biophysics, Polish Academy of Science, Warsaw, Poland
| | - Jarosław Szczepaniak
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - Mateusz Ficek
- Department of Metrology and Optoelectronics, Gdansk University of Technology, Gdansk, Poland
| | - Piotr Koczoń
- Department of Chemistry, Institute of Food Sciences, Warsaw University of Life Sciences, Warsaw, Poland
| | - Sławomir Jaworski
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| | - André Chwalibog
- Department of Veterinary and Animal, Sciences, University of Copenhagen, Frederiksberg, Denmark
| | - Ewa Sawosz
- Department of Nanobiotechnology, Institute of Biology, Warsaw University of Life Sciences, Warsaw, Poland
| |
Collapse
|
14
|
Li K, Liu T. Evaluation of Oncogene NUP37 as a Potential Novel Biomarker in Breast Cancer. Front Oncol 2021; 11:669655. [PMID: 34386417 PMCID: PMC8353244 DOI: 10.3389/fonc.2021.669655] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 07/12/2021] [Indexed: 11/26/2022] Open
Abstract
Purpose There is an urgent need to identify oncogenes that may be beneficial to diagnose and develop target therapy for breast cancer. Methods Based on the GEO database, DECenter was used to screen the differentially overexpressed genes in breast cancer samples. Search Tool for the Retrieval of Interacting Genes and Cytoscape were performed to construct the PPI network to predict the hub gene. Functional and pathway enrichment were performed based on GO analysis. GEO2R, Oncomine, human tissue microarray staining, and western blot were applied to confirm the expression of NUP37. The association between NUP37 expression and prognosis in patients with breast cancer were assessed using the Kaplan–Meier plotter online tool and OncoLnc. siRNAs were used to knock down NUP37 and evaluate proliferation, migration, and stemness in breast cancer cells. Results We found that 138 genes were differentially upregulated in breast cancer samples, mainly comprising components of the nucleus and involved in the cell cycle process. NUP37 was identified as a hub gene that is upregulated in breast cancer patients related to a significantly worse survival rate. Furthermore, we confirmed that the downregulation of NUP37 in breast cancer cells results in the inhibition of cell growth, migration, and stemness. Conclusions High expression of NUP37 in breast cancer patients is associated with a poorer prognosis and promotion of cell growth, migration, and stemness. The multiple bioinformatics and experimental analysis help provide a comprehensive understanding of the roles of NUP37 as a potential marker for diagnosis and prognosis and as a novel therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Kangdi Li
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Liu
- The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
15
|
Quan Y, Li Z, Zhu K, Liang J. Transcatheter arterial chemoembolization combined with Hippo/YAP inhibition significantly improve the survival of rats with transplanted hepatocellular carcinoma. Lipids Health Dis 2021; 20:74. [PMID: 34304741 PMCID: PMC8310593 DOI: 10.1186/s12944-021-01486-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 05/28/2021] [Indexed: 01/28/2023] Open
Abstract
Background This study aimed to explore the effect of inhibiting the Hippo/Yes-associated protein (YAP) signaling pathway on the outcomes of transcatheter arterial chemoembolization (TACE) in treating transplanted hepatocellular carcinoma (HCC). Methods A transplanted HCC rat model was established. Then, rats were randomly divided into four groups: Sham, TACE, verteporfin (inhibitor of Hippo/YAP), and TACE+verteporfin. Lent-OE-YAP was transfected into rats to overexpress YAP in vivo. After treatments, morphological changes, tumor weight, and the overall survival of rats in different groups were analyzed. Real-time PCR, immunohistochemistry staining, and Western blotting were used to determine the expression of factors related to the Hippo/YAP signaling pathway. Results Tumor weight and tissue lesions in the TACE and verteporfin groups were significantly reduced compared with the Sham group. Verteporfin significantly decreased tumor weight after TACE treatment. In addition, verteporfin significantly improved the overall survival of rats with transplanted HCC after TACE treatment. Compared with the Sham group, both TACE and verteporfin groups exhibited significantly decreased expression of macrophage-stimulating (MST)1, MST2, long-acting thyroid stimulator 1, transcriptional co-activator with PDZ-binding motif (TAZ), Yes-associated protein (YAP), TEA domain transcription factor (TEAD)1, TEAD2, TEAD3, and TEAD4. TACE plus verteporfin significantly enhanced the downregulation of effectors in the Hippo/YAP signaling pathway and decreased tumor size, while the overexpression of YAP exerted opposite effects. Conclusion The inhibition of the Hippo/YAP signaling pathway via verteporfin significantly improved the outcomes of TACE in treating transplanted HCC. Supplementary Information The online version contains supplementary material available at 10.1186/s12944-021-01486-w.
Collapse
Affiliation(s)
- Yi Quan
- Department of Oncology Medilcal Center, The First People's Hospital of Zhaoqing, Zhaoqing, Guangdong, 526000, China.
| | - Zhi Li
- Department of Interventional, First Affiliated Hospital of Suzhou University, Suzhou, Jiangsu, 215006, China
| | - Kangshun Zhu
- Department of Minimally Invasive Medicine, Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510000, China
| | - Jundi Liang
- Department of Oncology Medilcal Center, The First People's Hospital of Zhaoqing, Zhaoqing, Guangdong, 526000, China
| |
Collapse
|
16
|
Liu Z, Wang H, Jia Y, Wang J, Wang Y, Bian L, Liu B, Lian X, Zhang B, Ren Z, Zhang W, Dai W, Gao Y. Significantly high expression of NUP37 leads to poor prognosis of glioma patients by promoting the proliferation of glioma cells. Cancer Med 2021; 10:5218-5234. [PMID: 34264013 PMCID: PMC8335818 DOI: 10.1002/cam4.3954] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 04/11/2021] [Accepted: 04/12/2021] [Indexed: 01/02/2023] Open
Abstract
Background The carcinogenic effect of NUP37 has been reported recently in a variety of tumors, but its research in the field of glioma has not been paid attention. The main purpose of this study is to reveal the relationship between NUP37 and prognosis or clinical characteristics of glioma patients. Methods First, as a retrospective study, this study included thousands of tissue samples based on a variety of public databases and clinicopathological tissues. Second, a series of bioinformatics analysis methods were used to analyze the NUP37 and glioma samples from multiple databases such as the CGGA, TCGA, GEO, HPA, and GEPIA. Third, to analyze the relationship between the expression level of NUP37 in tumor tissues and cells and a variety of clinical prognostic molecular characteristics, whether it can be an independent risk factor leading to poor prognosis in glioma and whether it has clinical diagnostic value; GSEA was used to analyze the cancer‐related signaling pathways that may be activated by high expression of NUP37. Fifth, CMap was used to analyze small molecule drugs that may inhibit NUP37 expression. Finally, the meta‐analysis of thousands of tissue samples from seven datasets and cell proliferation and migration experiments confirmed that NUP37 has a malignant effect on glioma. Results NUP37 is highly expressed in glioma patient tissues and glioma cells, significantly correlates with reduced overall survival, and may serve as an independent prognostic factor with some diagnostic value. Silencing NUP37 suppresses malignant biological behaviors of glioma cells. 4 small molecule drugs that had potential targeting inhibitory effects on NUP37 overexpression. Conclusions This study demonstrates for the first time a malignant role of NUP37 in glioma and provides a vision to unravel the complex pathological mechanisms of glioma and a potentially valuable biomarker for implementing individualized diagnosis and treatment of glioma.
Collapse
Affiliation(s)
- Zhendong Liu
- Department of Orthopaedic, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China
| | - Hongbo Wang
- Department of Orthopaedic, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China.,Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Yulong Jia
- Henan Provincial People's Hospital, Cerebrovascular Disease Hospital, People's Hospital of Zhengzhou University, People's Hospital of Henan University, Zhengzhou, China
| | - Jialin Wang
- Department of Orthopaedic, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China.,Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Yanbiao Wang
- Department of Orthopaedic, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China.,Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Lu Bian
- Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Binfeng Liu
- Department of Orthopaedic, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China.,Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Xiaoyu Lian
- Department of Orthopaedic, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China.,Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Bo Zhang
- Department of Orthopaedic, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China.,Henan University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Zhishuai Ren
- Department of Orthopaedic, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China.,Zhengzhou University People's Hospital, Henan Provincial People's Hospital, Zhengzhou, China
| | - Wang Zhang
- Department of Neurosurgery of the First Affiliate Hospital of Harbin Medical University, Harbin, China
| | - Weiwei Dai
- Xiangya Hospital Central South University, Changsha, China
| | - Yanzheng Gao
- Department of Orthopaedic, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, School of Clinical Medicine, Henan University, Zhengzhou, China
| |
Collapse
|
17
|
Zhou X, Zhi Y, Yu J, Xu D. The Yin and Yang of Autosomal Recessive Primary Microcephaly Genes: Insights from Neurogenesis and Carcinogenesis. Int J Mol Sci 2020; 21:ijms21051691. [PMID: 32121580 PMCID: PMC7084222 DOI: 10.3390/ijms21051691] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 02/23/2020] [Accepted: 02/26/2020] [Indexed: 12/26/2022] Open
Abstract
The stem cells of neurogenesis and carcinogenesis share many properties, including proliferative rate, an extensive replicative potential, the potential to generate different cell types of a given tissue, and an ability to independently migrate to a damaged area. This is also evidenced by the common molecular principles regulating key processes associated with cell division and apoptosis. Autosomal recessive primary microcephaly (MCPH) is a neurogenic mitotic disorder that is characterized by decreased brain size and mental retardation. Until now, a total of 25 genes have been identified that are known to be associated with MCPH. The inactivation (yin) of most MCPH genes leads to neurogenesis defects, while the upregulation (yang) of some MCPH genes is associated with different kinds of carcinogenesis. Here, we try to summarize the roles of MCPH genes in these two diseases and explore the underlying mechanisms, which will help us to explore new, attractive approaches to targeting tumor cells that are resistant to the current therapies.
Collapse
Affiliation(s)
- Xiaokun Zhou
- College of Biological Science and Engineering, Institute of Life Sciences, Fuzhou University, Fuzhou 350108, China; (X.Z.); (Y.Z.); (J.Y.)
| | - Yiqiang Zhi
- College of Biological Science and Engineering, Institute of Life Sciences, Fuzhou University, Fuzhou 350108, China; (X.Z.); (Y.Z.); (J.Y.)
| | - Jurui Yu
- College of Biological Science and Engineering, Institute of Life Sciences, Fuzhou University, Fuzhou 350108, China; (X.Z.); (Y.Z.); (J.Y.)
| | - Dan Xu
- College of Biological Science and Engineering, Institute of Life Sciences, Fuzhou University, Fuzhou 350108, China; (X.Z.); (Y.Z.); (J.Y.)
- Fujian Key Laboratory of Molecular Neurology, Institute of Neuroscience, Fujian Medical University, Fuzhou 350005, China
- Correspondence: ; Tel.: +86-17085937559
| |
Collapse
|
18
|
The HIPPO Transducer YAP and Its Targets CTGF and Cyr61 Drive a Paracrine Signalling in Cold Atmospheric Plasma-Mediated Wound Healing. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4910280. [PMID: 32104533 PMCID: PMC7040405 DOI: 10.1155/2020/4910280] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 01/27/2020] [Accepted: 01/29/2020] [Indexed: 12/15/2022]
Abstract
Reactive species play a pivotal role in orchestrating wound healing responses. They act as secondary messengers and drive redox-signalling pathways that are involved in the homeostatic, inflammatory, proliferative, and remodelling phases of wound healing. The application of Cold Atmospheric Plasma (CAP) to the wound site produces a profusion of short- and long-lived reactive species that have been demonstrated to be effective in promoting wound healing; however, knowledge of the mechanisms underlying CAP-mediated wound healing remains scarce. To address this, an in vitro coculture model was used to study the effects of CAP on wound healing and on paracrine crosstalk between dermal keratinocytes and fibroblasts. Using this coculture model, we observed a stimulatory effect on the migration ability of HaCaT cells that were cocultured with dermal fibroblasts. Additionally, CAP treatment resulted in an upregulation of the HIPPO transcription factor YAP in HaCaTs and fibroblasts. Downstream effectors of the HIPPO signalling pathway (CTGF and Cyr61) were also upregulated in dermal fibroblasts, and the administration of antioxidants could inhibit CAP-mediated wound healing and abrogate the gene expression of the HIPPO downstream effectors. Interestingly, we observed that HaCaT cells exhibited an improved cell migration rate when incubated with CAP-treated fibroblast-conditioned media compared to that observed after incubation with untreated media. An induction of CTGF and Cyr61 secretion was also observed upon CAP treatment in the fibroblast-conditioned media. Finally, exposure to recombinant CTGF and Cyr61 could also significantly improve HaCaT cell migration. In summary, our results validated that CAP activates a regenerative signalling pathway at the onset of wound healing. Additionally, CAP also stimulated a reciprocal communication between dermal fibroblasts and keratinocytes, resulting in improved keratinocyte wound healing in coculture.
Collapse
|
19
|
Huang L, Wang T, Wang F, Hu X, Zhan G, Jin X, Zhang L, Li Y. NUP37 silencing induces inhibition of cell proliferation, G1 phase cell cycle arrest and apoptosis in non-small cell lung cancer cells. Pathol Res Pract 2020; 216:152836. [PMID: 32014308 DOI: 10.1016/j.prp.2020.152836] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 01/19/2020] [Indexed: 01/24/2023]
Abstract
NUP37 has been reported as a component of the nuclear pore complex, which may be involved in tumorigenesis. Previous reports have shown that NUP37 acts as an oncogene in the development of hepatocellular carcinoma. However, its role in lung cancer remains unknown. The present study demonstrated for the first time that NUP37 expression was overexpressed in non-small cell lung cancer (NSCLC) samples compared with the corresponding expression noted in normal tissues. The results were derived by analyzing public datasets. Moreover, it was shown that NUP37 was overexpressed in advanced stage NSCLC samples compared with the corresponding expression of this protein in early stage NSCLC samples. Higher expression levels of NUP37 correlated with lower overall survival (OS) in NSCLC samples. Bioinformatic analysis indicated that NUP37 was involved in regulating cell cycle progression in NSCLC. Furthermore, knockdown of NUP37 suppressed cell growth and proliferation in A549and H1299 cells as demonstrated with the Celigo Cell Counting method and the MTT assay. Flow cytometry analysis indicated that knockdown of NUP37 induced significant S phage cell arrest and apoptosis in A549 and H1299 cells. The results showed that knockdown of NUP37 remarkably induced the protein levels of cleaved PARP, P53 and BCL2 in A549 cells. Therefore, it was concluded that NUP37 serves a distinguished role in the growth of lung cancer cells and may be considered as a potential biomarker and therapeutic target for lung cancer.
Collapse
Affiliation(s)
- Lianglong Huang
- Respiratory Medicine, Zhejiang Provincial Armed Police Corps Hospital, 304052, Jiaxing, Zhejiang, PR China
| | - Tao Wang
- Respiratory Medicine, Zhejiang Provincial Armed Police Corps Hospital, 304052, Jiaxing, Zhejiang, PR China
| | - Fumu Wang
- Wenzhou Medical University, 325035, Wenzhou, Zhejiang, PR China
| | - Xiaoyi Hu
- Respiratory Medicine, Zhejiang Provincial Armed Police Corps Hospital, 304052, Jiaxing, Zhejiang, PR China
| | - Guoxiang Zhan
- Respiratory Medicine, Zhejiang Provincial Armed Police Corps Hospital, 304052, Jiaxing, Zhejiang, PR China
| | - Xiaohui Jin
- Respiratory Medicine, Zhejiang Provincial Armed Police Corps Hospital, 304052, Jiaxing, Zhejiang, PR China
| | - Licheng Zhang
- Respiratory Medicine, Zhejiang Provincial Armed Police Corps Hospital, 304052, Jiaxing, Zhejiang, PR China.
| | - Yuping Li
- Wenzhou Medical University, 325035, Wenzhou, Zhejiang, PR China.
| |
Collapse
|
20
|
Chen J, Wo D, Ma E, Yan H, Peng J, Zhu W, Fang Y, Ren DN. Deletion of low-density lipoprotein-related receptor 5 inhibits liver Cancer cell proliferation via destabilizing Nucleoporin 37. Cell Commun Signal 2019; 17:174. [PMID: 31881970 PMCID: PMC6935199 DOI: 10.1186/s12964-019-0495-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 12/05/2019] [Indexed: 11/24/2022] Open
Abstract
Background LRP5/6 are co-receptors in Wnt/β-catenin pathway. Recently, we discovered multiple β-catenin independent functions of LRP5/6 in tumor cells and in the diseased heart. Nucleoporin 37 (NUP37) is an important component of the nuclear pore complex (NPC), whose elevated expression is associated with worsened prognosis in liver cancer. Previous studies have shown that NUP37 interacted with YAP and activated YAP/TEAD signaling in liver cancer. Our preliminary findings showed a nuclear location of LRP5. We thus tested the hypothesis that LRP5 may act as a genuine regulator of YAP/TEAD signaling via modulating NUP37 in a β-catenin-independent way. Methods We performed siRNA knockdown of LRP5, LRP6, or β-catenin in liver cancer HepG2 cells to determine the effect on tumor cell proliferation. Protein expressions and interaction between LRP5 and NUP37 were determined using immunoprecipitation and western blot analyses. Results HepG2 cell proliferation was markedly inhibited by knockdown of LRP5 but not LRP6 or β-catenin, suggesting that LRP5 has a specific, β-catenin-independent role in inhibiting HepG2 cell proliferation. Knockdown of NUP37 by siRNA inhibited the proliferation of HepG2 cells, whereas overexpression of NUP37 reversed the decrease in cell proliferation induced by LRP5 knockdown. Immunoprecipitation assays confirmed that LRP5 bound to NUP37. Furthermore, LRP5 overexpression restored NUP37 knockdown-induced downregulation of YAP/TEAD pathway. Conclusions LRP5 deletion attenuates cell proliferation via destabilization of NUP37, in a β-catenin-independent manner. LRP5 therefore acts as a genuine regulator of YAP/TEAD signaling via maintaining the integrity of the NPC, and implicates a therapeutic strategy in targeting LRP5 for inhibiting liver cancer cell proliferation.
Collapse
Affiliation(s)
- Jinxiao Chen
- Department of Plastic and Burn Surgery, Ninth People's Hospital, Shanghai Jiaotong University, School of Medicine, 639 Zhi Zao Ju Road, Shanghai, 200011, People's Republic of China
| | - Da Wo
- Department of Plastic and Burn Surgery, Ninth People's Hospital, Shanghai Jiaotong University, School of Medicine, 639 Zhi Zao Ju Road, Shanghai, 200011, People's Republic of China
| | - En Ma
- Clinical and Translational Research Center, Research Institute of Heart Failure Shanghai East Hospital, Key Laboratory of Arrhythmias of Ministry of Education, Tongji University School of Medicine, Shanghai, China
| | - Hongwei Yan
- Clinical and Translational Research Center, Research Institute of Heart Failure Shanghai East Hospital, Key Laboratory of Arrhythmias of Ministry of Education, Tongji University School of Medicine, Shanghai, China
| | - Jun Peng
- Fujian Key Laboratory of Integrative Medicine on Geriatric, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, 350122, Fujian, China
| | - Weidong Zhu
- Clinical and Translational Research Center, Research Institute of Heart Failure Shanghai East Hospital, Key Laboratory of Arrhythmias of Ministry of Education, Tongji University School of Medicine, Shanghai, China
| | - Yong Fang
- Department of Plastic and Burn Surgery, Ninth People's Hospital, Shanghai Jiaotong University, School of Medicine, 639 Zhi Zao Ju Road, Shanghai, 200011, People's Republic of China.
| | - Dan-Ni Ren
- Fujian Key Laboratory of Integrative Medicine on Geriatric, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, 1 Qiuyang Road, Minhou Shangjie, Fuzhou, 350122, Fujian, China.
| |
Collapse
|
21
|
Donnaloja F, Jacchetti E, Soncini M, Raimondi MT. Mechanosensing at the Nuclear Envelope by Nuclear Pore Complex Stretch Activation and Its Effect in Physiology and Pathology. Front Physiol 2019; 10:896. [PMID: 31354529 PMCID: PMC6640030 DOI: 10.3389/fphys.2019.00896] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 06/27/2019] [Indexed: 02/03/2023] Open
Abstract
Cell fate is correlated to mechanotransduction, in which forces transmitted by the cytoskeleton filaments alter the nuclear shape, affecting transcription factor import/export, cells transcription activity and chromatin distribution. There is in fact evidence that stem cells cultured in 3D environments mimicking the native niche are able to maintain their stemness or modulate their cellular function. However, the molecular and biophysical mechanisms underlying cellular mechanosensing are still largely unclear. The propagation of mechanical stimuli via a direct pathway from cell membrane integrins to SUN proteins residing in the nuclear envelop has been demonstrated, but we suggest that the cells’ fate is mainly affected by the force distribution at the nuclear envelope level, where the SUN protein transmits the stimuli via its mechanical connection to several cell structures such as chromatin, lamina and the nuclear pore complex (NPC). In this review, we analyze the NPC structure and organization, which have not as yet been fully investigated, and its plausible involvement in cell fate. NPC is a multiprotein complex that spans the nuclear envelope, and is involved in several key cellular processes such as bidirectional nucleocytoplasmic exchange, cell cycle regulation, kinetochore organization, and regulation of gene expression. As several connections between the NPC and the nuclear envelope, chromatin and other transmembrane proteins have been identified, it is reasonable to suppose that nuclear deformations can alter the NPC structure. We provide evidence that the transmission of mechanical forces may significantly affects the basket conformation via the Nup153-SUN1 connection, both altering the passage of molecules through it and influencing the state of chromatin packing. Finally, we review the known correlations between a pathological NPC structure and diseases such as cancer, autoimmune disease, aging and laminopathies.
Collapse
Affiliation(s)
- F Donnaloja
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta," Politecnico di Milano, Milan, Italy
| | - E Jacchetti
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta," Politecnico di Milano, Milan, Italy
| | - M Soncini
- Department of Electronics, Information and Bioengineering, Politecnico di Milano, Milan, Italy
| | - M T Raimondi
- Department of Chemistry, Materials and Chemical Engineering "Giulio Natta," Politecnico di Milano, Milan, Italy
| |
Collapse
|
22
|
Cianciolo Cosentino C, Berto A, Pelletier S, Hari M, Loffing J, Neuhauss SCF, Doye V. Moderate Nucleoporin 133 deficiency leads to glomerular damage in zebrafish. Sci Rep 2019; 9:4750. [PMID: 30894603 PMCID: PMC6426968 DOI: 10.1038/s41598-019-41202-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 02/26/2019] [Indexed: 01/13/2023] Open
Abstract
Although structural nuclear pore proteins (nucleoporins) are seemingly required in every cell type to assemble a functional nuclear transport machinery, mutations or deregulation of a subset of them have been associated with specific human hereditary diseases. In particular, previous genetic studies of patients with nephrotic syndrome identified mutations in Nup107 that impaired the expression or the localization of its direct partner at nuclear pores, Nup133. In the present study, we characterized the zebrafish nup133 orthologous gene and its expression pattern during larval development. Using a morpholino-mediated gene knockdown, we show that partial depletion of Nup133 in zebrafish larvae leads to the formation of kidney cysts, a phenotype that can be rescued by co-injection of wild type mRNA. Analysis of different markers for tubular and glomerular development shows that the overall kidney development is not affected by nup133 knockdown. Likewise, no gross defect in nuclear pore complex assembly was observed in these nup133 morphants. On the other hand, nup133 downregulation results in proteinuria and moderate foot process effacement, mimicking some of the abnormalities typically featured by patients with nephrotic syndrome. These data indicate that nup133 is a new gene required for proper glomerular structure and function in zebrafish.
Collapse
Affiliation(s)
- Chiara Cianciolo Cosentino
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland.,Institute of Anatomy, University of Zurich, Zurich, Switzerland.,Fondazione RiMED, Palermo, Italy
| | - Alessandro Berto
- Institut Jacques Monod, UMR7592 CNRS-Université Paris Diderot, Sorbonne Paris Cité, F-75205, Paris, France.,Ecole Doctorale SDSV, Université Paris Sud, F-91405, Orsay, France
| | - Stéphane Pelletier
- Institut Jacques Monod, UMR7592 CNRS-Université Paris Diderot, Sorbonne Paris Cité, F-75205, Paris, France
| | - Michelle Hari
- Institute of Molecular Life Sciences, University of Zurich, Zurich, Switzerland
| | | | | | - Valérie Doye
- Institut Jacques Monod, UMR7592 CNRS-Université Paris Diderot, Sorbonne Paris Cité, F-75205, Paris, France.
| |
Collapse
|
23
|
Abstract
Pancreatic cancer is an aggressive and intractable malignancy with high mortality. This is due in part to a high resistance to chemotherapeutics and radiation treatment conferred by diverse regulatory mechanisms. Among these, constituents of the nuclear envelope play a significant role in regulating oncogenesis and pancreatic tumor biology, and this review focuses on three specific components and their roles in cancer. The LINC complex is a nuclear envelope component formed by proteins with SUN and KASH domains that interact in the periplasmic space of the nuclear envelope. These interactions functionally and structurally couple the cytoskeleton to chromatin and facilitates gene regulation informed by cytoplasmic activity. Furthermore, cancer cell invasiveness is impacted by LINC complex biology. The nuclear lamina is adjacent to the inner nuclear membrane of the nuclear envelope and can actively regulate chromatin in addition to providing structural integrity to the nucleus. A disrupted lamina can impart biophysical compromise to nuclear structure and function, as well as form dysfunctional micronuclei that may lead to genomic instability and chromothripsis. In close relationship to the nuclear lamina is the nuclear pore complex, a large megadalton structure that spans both outer and inner membranes of the nuclear envelope. The nuclear pore complex mediates bidirectional nucleocytoplasmic transport and is comprised of specialized proteins called nucleoporins that are overexpressed in many cancers and are diagnostic markers for oncogenesis. Furthermore, recent demonstration of gene regulatory functions for discrete nucleoporins independent of their nuclear trafficking function suggests that these proteins may contribute more to malignant phenotypes beyond serving as biomarkers. The nuclear envelope is thus a complex, intricate regulator of cell signaling, with roles in pancreatic tumorigenesis and general oncogenic transformation.
Collapse
Affiliation(s)
| | - Randolph S. Faustino
- Genetics and Genomics, Sanford Research, Sioux Falls, SD 57104, USA
- Department of Pediatrics, Sanford School of Medicine of the University of South Dakota, Sioux Falls, SD 57105, USA
| |
Collapse
|
24
|
Guo Q, Wang J, Cao Z, Tang Y, Feng C, Huang F. Interaction of S100A1 with LATS1 promotes cell growth through regulation of the Hippo pathway in hepatocellular carcinoma. Int J Oncol 2018; 53:592-602. [PMID: 29901195 PMCID: PMC6017223 DOI: 10.3892/ijo.2018.4431] [Citation(s) in RCA: 7] [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: 02/09/2018] [Accepted: 04/16/2018] [Indexed: 02/07/2023] Open
Abstract
Despite advances in surgery and chemotherapy, the prognosis of patients with hepatocellular carcinoma (HCC) remains poor. In the present study, the role of S100A1 in the progression of HCC was investigated. Immunohistochemical staining was used to measure the expression of S100A1 in HCC tissues. S100A1 was knocked down by siRNA. A battery of experiments was used to evaluate the biology functions of S100A1. It was found that S100A1 was upregulated in HCC tissues, and its upregulation was associated with a large tumor size, low differentiation and shorter survival time. The biological experiments demonstrated that S100A1 functions as an oncogene in HCC. It was also found that S100A1 knockdown enhanced the inhibitory effects of cisplatin on HCC cells. The results showed that the downregulation of S100A1 induced the phosphorylation of yes‑associated protein (YAP), and treatment with CHX demonstrated that the downregulation of S100A1 accelerated YAP protein degradation. The downregulation of S100A1 did not alter the expression of mammalian sterile 20‑like kinase (MST)1/2 or phosphorylated MST1/2, but upregulated the phosphorylation of large tumor suppressor kinase 1 (LATS1). It was further confirmed that S100A1 interacted with LATS1. LATS1 depletion significantly reduced the effects of S100A1 on cell growth rate and apoptosis, and there was a positive correlation between phosphorylated LATS1 and S100A1 in clinical samples, indicating that LATS1 was responsible for the S100A1-induced changes in cancer cell growth and Hippo signaling. In conclusion, the results of the present study indicated that S100A1 functions as an oncogene and may be a biomarker for the prognosis of patients with HCC. S100A1 exerted its oncogenic function by interacting with LATS1 and activating YAP. S100A1 may serve as a target for novel therapies in HCC.
Collapse
Affiliation(s)
| | | | - Zeyu Cao
- Department of Hepatobiliary Surgery, The Third Xiang-Ya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Yongchang Tang
- Department of Hepatobiliary Surgery, The Third Xiang-Ya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Chao Feng
- Department of Hepatobiliary Surgery, The Third Xiang-Ya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| | - Feizhou Huang
- Department of Hepatobiliary Surgery, The Third Xiang-Ya Hospital, Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|