1
|
Panchal NK, Mohanty S, Prince SE. Computational insights into NIMA-related kinase 6: unraveling mutational effects on structure and function. Mol Cell Biochem 2024; 479:2989-3009. [PMID: 38117419 DOI: 10.1007/s11010-023-04910-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 11/25/2023] [Indexed: 12/21/2023]
Abstract
The NEK6 (NIMA-related kinase 6) serine/threonine kinase is a pivotal player in a multitude of cellular processes, including the regulation of the cell cycle and the response to DNA damage. Its significance extends to disease pathogenesis, as changes in NEK6 activity have been linked to the development of cancer. Non-synonymous single nucleotide polymorphisms (nsSNPs) in NEK6 have been linked to cancer as they alter the protein's native structure and function. The association between NEK6 activity and cancer development has prompted researchers to explore the effects of genetic variations within the NEK6 gene. Therefore, we utilized advanced computational tools to analyze 155 high-confidence nsSNPs in the NEK6 gene. From this analysis, 21 nsSNPs were identified as potentially harmful, raising concerns about their impact on NEK6 activity and cancer risk. These 21 mutations were then examined for structural alterations, and eight of nsSNPs (I51M, V76A, I134N, Y152D, R171Q, V186G, L237R, and C285S) were found to destabilize the protein. Among the destabilizing mutations screened, a specific mutation, R171Q, stood out due to its conserved nature. To understand its impact on the protein and conformation, all-atom molecular dynamics simulations (MDS) for 100 ns were performed for both Wildtype NEK6 (WT-NEK6) and R171Q. The simulations revealed that the R171Q variant was unstable and led to significant conformational changes in NEK6. This study provides valuable insights into NEK6 dysfunction caused by single amino acid alterations, offering a novel understanding of the molecular mechanisms underlying NEK6-related cancer progression.
Collapse
Affiliation(s)
- Nagesh Kishan Panchal
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, 632 014, India
| | - Shruti Mohanty
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, 632014, India
| | - Sabina Evan Prince
- Department of Biotechnology, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, 632 014, India.
| |
Collapse
|
2
|
Ullah MA, Garcillán B, Whitlock E, Figgett WA, Infantino S, Eslami M, Yang S, Rahman MA, Sheng YH, Weber N, Schneider P, Tam CS, Mackay F. An unappreciated cell survival-independent role for BAFF initiating chronic lymphocytic leukemia. Front Immunol 2024; 15:1345515. [PMID: 38469292 PMCID: PMC10927009 DOI: 10.3389/fimmu.2024.1345515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 01/25/2024] [Indexed: 03/13/2024] Open
Abstract
Background Chronic Lymphocytic Leukemia (CLL) is characterized by the expansion of CD19+ CD5+ B cells but its origin remains debated. Mutated CLL may originate from post-germinal center B cells and unmutated CLL from CD5+ mature B cell precursors. Irrespective of precursor types, events initiating CLL remain unknown. The cytokines BAFF and APRIL each play a significant role in CLL cell survival and accumulation, but their involvement in disease initiation remains unclear. Methods We generated novel CLL models lacking BAFF or APRIL. In vivo experiments were conducted to explore the impact of BAFF or APRIL loss on leukemia initiation, progression, and dissemination. Additionally, RNA-seq and quantitative real-time PCR were performed to unveil the transcriptomic signature influenced by BAFF in CLL. The direct role of BAFF in controlling the expression of tumor-promoting genes was further assessed in patient-derived primary CLL cells ex-vivo. Results Our findings demonstrate a crucial role for BAFF, but not APRIL, in the initiation and dissemination of CLL cells. In the absence of BAFF or its receptor BAFF-R, the TCL1 transgene only increases CLL cell numbers in the peritoneal cavity, without dissemination into the periphery. While BAFF binding to BAFF-R is dispensable for peritoneal CLL cell survival, it is necessary to activate a tumor-promoting gene program, potentially linked to CLL initiation and progression. This direct role of BAFF in controlling the expression of tumor-promoting genes was confirmed in patient-derived primary CLL cells ex-vivo. Conclusions Our study, involving both mouse and human CLL cells, suggests that BAFF might initiate CLL through mechanisms independent of cell survival. Combining current CLL therapies with BAFF inhibition could offer a dual benefit by reducing peripheral tumor burden and suppressing transformed CLL cell output.
Collapse
Affiliation(s)
- Md Ashik Ullah
- Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Cancer Program, Herston, QLD, Australia
| | - Beatriz Garcillán
- The Department of Microbiology and Immunology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Eden Whitlock
- Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Cancer Program, Herston, QLD, Australia
| | - William A. Figgett
- The Department of Microbiology and Immunology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
- Garvan Institute of Medical Research, Kinghorn Centre for Clinical Genomics, Darlinghurst, NSW, Australia
| | - Simona Infantino
- The Department of Microbiology and Immunology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
| | - Mahya Eslami
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
- Department of Oncology and Children’s Research Centre, University Children’s Hospital Zürich, Zürich, Switzerland
| | - SiLing Yang
- Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Cancer Program, Herston, QLD, Australia
| | - M. Arifur Rahman
- Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Cancer Program, Herston, QLD, Australia
| | - Yong H. Sheng
- Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Cancer Program, Herston, QLD, Australia
| | - Nicholas Weber
- Cancer Care Services, Royal Brisbane and Women’s Hospital, Herston, QLD, Australia
| | - Pascal Schneider
- Department of Immunobiology, University of Lausanne, Epalinges, Switzerland
| | - Constantine S. Tam
- Department of Haematology, Alfred Hospital, Melbourne, VIC, Australia
- Department of Haematology, Monash University, Melbourne, VIC, Australia
| | - Fabienne Mackay
- Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute, Cancer Program, Herston, QLD, Australia
- The Department of Microbiology and Immunology, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, VIC, Australia
- The Department of Immunology and Pathology, Monash University, VIC, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
3
|
Wu X, Deng K, Cai H, Zeng Z, Cao J, Zhang L, Lu Z, Cheng W. Nek6 knockdown polarized macrophages into a pro-inflammatory phenotype via inhibiting STAT3 expression. Int J Exp Pathol 2023; 104:237-246. [PMID: 37431082 PMCID: PMC10500168 DOI: 10.1111/iep.12489] [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: 01/05/2023] [Revised: 05/08/2023] [Accepted: 06/18/2023] [Indexed: 07/12/2023] Open
Abstract
Recently macrophage polarization has emerged as playing an essential role in the oathogenesis of atherosclerosis, which is the most important underlying process in many types of cardiovascular diseases. Although Nek6 has been reported to be involved in various cellular processes, the effect of Nek6 on macrophage polarization remains unknown. Macrophages exposed to lipopolysaccharide (LPS) or IL-4 were used to establish an in vitro model for the study of regulation of classically (M1) or alternatively (M2) activated macrophage. Bone marrow-derived macrophages (BMDMs) transfected with short hairpin RNA-targeting Nek6 were then in functional studies. We observed that Nek6 expression was decreased in both peritoneal macrophages (PMs) and BMDMs stimulated by LPS. This effect was seen at both mRNA and protein level. The opposite results were obtained after administration of IL-4. Macrophage-specific Nek6 knockdown significantly exacerbated pro-inflammatory M1 polarized macrophage gene expression in response to LPS challenge, but the anti-inflammatory response gene expression that is related to M2 macrophages was attenuated by Nek6 silencing followed by treatment with IL-4. Mechanistic studies exhibited that Nek6 knockdown inhibited the phosphorylated STAT3 expression that mediated the effect on macrophage polarization regulated by AdshNek6. Moreover, decreased Nek6 expression was also observed in atherosclerotic plaques. Collectively, these evidences suggested that Nek6 acts as a crucial site in macrophage polarization, and that this operates in a STAT3-dependent manner.
Collapse
Affiliation(s)
- Xiaoyan Wu
- Department of Cardiology, Zhongnan HospitalWuhan UniversityWuhanChina
- Institute of Myocardial Injury and RepairWuhan UniversityWuhanChina
| | - Ke‐Qiong Deng
- Department of Cardiology, Zhongnan HospitalWuhan UniversityWuhanChina
- Institute of Myocardial Injury and RepairWuhan UniversityWuhanChina
| | - Huan‐Huan Cai
- Department of Cardiology, Zhongnan HospitalWuhan UniversityWuhanChina
- Institute of Myocardial Injury and RepairWuhan UniversityWuhanChina
| | - Ziyue Zeng
- Department of Cardiology, Zhongnan HospitalWuhan UniversityWuhanChina
- Institute of Myocardial Injury and RepairWuhan UniversityWuhanChina
| | - Jian‐Lei Cao
- Department of Cardiology, Zhongnan HospitalWuhan UniversityWuhanChina
- Institute of Myocardial Injury and RepairWuhan UniversityWuhanChina
| | - Lin Zhang
- Department of Cardiology, Zhongnan HospitalWuhan UniversityWuhanChina
- Institute of Myocardial Injury and RepairWuhan UniversityWuhanChina
| | - Zhibing Lu
- Department of Cardiology, Zhongnan HospitalWuhan UniversityWuhanChina
- Institute of Myocardial Injury and RepairWuhan UniversityWuhanChina
| | - Wen‐Lin Cheng
- Department of Cardiology, Zhongnan HospitalWuhan UniversityWuhanChina
- Institute of Myocardial Injury and RepairWuhan UniversityWuhanChina
| |
Collapse
|
4
|
Zhang H, Li B. NIMA-related kinase 6 as an effective target inhibits the hepatocarcinogenesis and progression of hepatocellular carcinoma. Heliyon 2023; 9:e15971. [PMID: 37260886 PMCID: PMC10227323 DOI: 10.1016/j.heliyon.2023.e15971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 06/02/2023] Open
Abstract
Background NIMA-related kinase 6 (NEK 6) is over-expressed in some tumor cell lines and tissues. However, its expression in hepatocellular carcinoma (HCC) and its correlation with clinical features remain unclear. Methods Total RNA from HCC liver tissues, other liver specimens, and hepatic cell lines was extracted and QPCR was adopted to detect NEK6 expression. The correlation between NEK6 expression and the clinical characteristics of HCC was analyzed. Scratch assay, Transwell assay, and tumor-formation assay were used to evaluate the effects of NEK6 on the HCC progression in vitro and in vivo. Results The expression of NEK6 was up-regulated in HCC tissues and HCC cell lines: Li-7 and HepG2. The overexpression of NEK6 was correlated with hepatitis B virus infection and tumor diameter (P = 0.045). When down-regulated the expression of NEK6, both the migration and invasion capabilities of Li-7 and HepG2 cells and the growth of xenograft tumors were suppressed. (P < 0.05). Conclusions NEK6 expression was up-regulated in HCC and correlated with the progression, suggesting it might be a valuable biomarker and a potential therapeutic target for HCC.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Hepatobiliary Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Bo Li
- Department of Hepatobiliary Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
5
|
Guo W, Wang H, Tharkeshwar AK, Couthouis J, Braems E, Masrori P, Van Schoor E, Fan Y, Ahuja K, Moisse M, Jacquemyn M, da Costa RFM, Gajjar M, Balusu S, Tricot T, Fumagalli L, Hersmus N, Janky R, Impens F, Berghe PV, Ho R, Thal DR, Vandenberghe R, Hegde ML, Chandran S, De Strooper B, Daelemans D, Van Damme P, Van Den Bosch L, Verfaillie C. CRISPR/Cas9 screen in human iPSC-derived cortical neurons identifies NEK6 as a novel disease modifier of C9orf72 poly(PR) toxicity. Alzheimers Dement 2023; 19:1245-1259. [PMID: 35993441 PMCID: PMC9943798 DOI: 10.1002/alz.12760] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 06/14/2022] [Accepted: 07/08/2022] [Indexed: 11/10/2022]
Abstract
INTRODUCTION The most common genetic cause of frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) are hexanucleotide repeats in chromosome 9 open reading frame 72 (C9orf72). These repeats produce dipeptide repeat proteins with poly(PR) being the most toxic one. METHODS We performed a kinome-wide CRISPR/Cas9 knock-out screen in human induced pluripotent stem cell (iPSC) -derived cortical neurons to identify modifiers of poly(PR) toxicity, and validated the role of candidate modifiers using in vitro, in vivo, and ex-vivo studies. RESULTS Knock-down of NIMA-related kinase 6 (NEK6) prevented neuronal toxicity caused by poly(PR). Knock-down of nek6 also ameliorated the poly(PR)-induced axonopathy in zebrafish and NEK6 was aberrantly expressed in C9orf72 patients. Suppression of NEK6 expression and NEK6 activity inhibition rescued axonal transport defects in cortical neurons from C9orf72 patient iPSCs, at least partially by reversing p53-related DNA damage. DISCUSSION We identified NEK6, which regulates poly(PR)-mediated p53-related DNA damage, as a novel therapeutic target for C9orf72 FTD/ALS.
Collapse
Affiliation(s)
- Wenting Guo
- Stem Cell Institute, Department of Devolpment and Regeneration, KU Leuven, Leuven, Belgium
- Department of Neurosciences, Experimental Neurology, Laboratory of Neurobiology, KU Leuven-University of Leuven, Leuven, Belgium
- VIB, Center for Brain & Disease Research, Leuven, Belgium and Leuven Brain Institute (LBI), Leuven, Belgium
| | - Haibo Wang
- Division of DNA Repair Research, Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, Houston, Texas, USA
- Department of Neuroscience Research at Neurological Surgery, Weill Medical College, New York, New York, USA
| | - Arun Kumar Tharkeshwar
- Department of Neurosciences, Experimental Neurology, Laboratory of Neurobiology, KU Leuven-University of Leuven, Leuven, Belgium
- VIB, Center for Brain & Disease Research, Leuven, Belgium and Leuven Brain Institute (LBI), Leuven, Belgium
| | - Julien Couthouis
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
| | - Elke Braems
- Department of Neurosciences, Experimental Neurology, Laboratory of Neurobiology, KU Leuven-University of Leuven, Leuven, Belgium
- VIB, Center for Brain & Disease Research, Leuven, Belgium and Leuven Brain Institute (LBI), Leuven, Belgium
| | - Pegah Masrori
- Department of Neurosciences, Experimental Neurology, Laboratory of Neurobiology, KU Leuven-University of Leuven, Leuven, Belgium
- VIB, Center for Brain & Disease Research, Leuven, Belgium and Leuven Brain Institute (LBI), Leuven, Belgium
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Evelien Van Schoor
- Department of Neurosciences, Experimental Neurology, Laboratory of Neurobiology, KU Leuven-University of Leuven, Leuven, Belgium
- VIB, Center for Brain & Disease Research, Leuven, Belgium and Leuven Brain Institute (LBI), Leuven, Belgium
- Laboratory of Neuropathology, Department of Imaging and Pathology, KU Leuven, and Leuven Brain Institute (LBI), Leuven, Belgium
| | - Yannan Fan
- Stem Cell Institute, Department of Devolpment and Regeneration, KU Leuven, Leuven, Belgium
| | - Karan Ahuja
- Stem Cell Institute, Department of Devolpment and Regeneration, KU Leuven, Leuven, Belgium
| | - Matthieu Moisse
- Department of Neurosciences, Experimental Neurology, Laboratory of Neurobiology, KU Leuven-University of Leuven, Leuven, Belgium
- VIB, Center for Brain & Disease Research, Leuven, Belgium and Leuven Brain Institute (LBI), Leuven, Belgium
| | - Maarten Jacquemyn
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, Leuven, Belgium
| | | | - Madhavsai Gajjar
- Stem Cell Institute, Department of Devolpment and Regeneration, KU Leuven, Leuven, Belgium
| | - Sriram Balusu
- VIB, Center for Brain & Disease Research, Leuven, Belgium and Leuven Brain Institute (LBI), Leuven, Belgium
| | - Tine Tricot
- Stem Cell Institute, Department of Devolpment and Regeneration, KU Leuven, Leuven, Belgium
| | - Laura Fumagalli
- Department of Neurosciences, Experimental Neurology, Laboratory of Neurobiology, KU Leuven-University of Leuven, Leuven, Belgium
- VIB, Center for Brain & Disease Research, Leuven, Belgium and Leuven Brain Institute (LBI), Leuven, Belgium
| | - Nicole Hersmus
- Department of Neurosciences, Experimental Neurology, Laboratory of Neurobiology, KU Leuven-University of Leuven, Leuven, Belgium
- VIB, Center for Brain & Disease Research, Leuven, Belgium and Leuven Brain Institute (LBI), Leuven, Belgium
| | | | - Francis Impens
- VIB-UGent Center for Medical Biotechnology, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
- VIB Proteomics Core, Ghent, Belgium
| | - Pieter Vanden Berghe
- Translational Research Centre for Gastrointestinal Disorders (TARGID), KU Leuven–University of Leuven, Leuven, Belgium
| | - Ritchie Ho
- Center for Neural Science and Medicine, Board of Governors Regenerative Medicine Institute, Departments of Biomedical Sciences and Neurology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Dietmar Rudolf Thal
- Laboratory of Neuropathology, Department of Imaging and Pathology, KU Leuven, and Leuven Brain Institute (LBI), Leuven, Belgium
| | - Rik Vandenberghe
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
- KU Leuven-Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Muralidhar L. Hegde
- Division of DNA Repair Research, Department of Neurosurgery, Center for Neuroregeneration, Houston Methodist Research Institute, Houston, Texas, USA
- Department of Neuroscience Research at Neurological Surgery, Weill Medical College, New York, New York, USA
| | - Siddharthan Chandran
- UK-Dementia Research Institute at University College London, London, UK
- Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Bart De Strooper
- VIB, Center for Brain & Disease Research, Leuven, Belgium and Leuven Brain Institute (LBI), Leuven, Belgium
- UK-Dementia Research Institute at University College London, London, UK
| | - Dirk Daelemans
- KU Leuven Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy, Rega Institute for Medical Research, Leuven, Belgium
| | - Philip Van Damme
- Department of Neurosciences, Experimental Neurology, Laboratory of Neurobiology, KU Leuven-University of Leuven, Leuven, Belgium
- VIB, Center for Brain & Disease Research, Leuven, Belgium and Leuven Brain Institute (LBI), Leuven, Belgium
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Ludo Van Den Bosch
- Department of Neurosciences, Experimental Neurology, Laboratory of Neurobiology, KU Leuven-University of Leuven, Leuven, Belgium
- VIB, Center for Brain & Disease Research, Leuven, Belgium and Leuven Brain Institute (LBI), Leuven, Belgium
| | - Catherine Verfaillie
- Stem Cell Institute, Department of Devolpment and Regeneration, KU Leuven, Leuven, Belgium
| |
Collapse
|
6
|
Panchal NK, Mohanty S, Prince SE. NIMA-related kinase-6 (NEK6) as an executable target in cancer. CLINICAL & TRANSLATIONAL ONCOLOGY : OFFICIAL PUBLICATION OF THE FEDERATION OF SPANISH ONCOLOGY SOCIETIES AND OF THE NATIONAL CANCER INSTITUTE OF MEXICO 2023; 25:66-77. [PMID: 36074296 DOI: 10.1007/s12094-022-02926-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/09/2022] [Indexed: 01/07/2023]
Abstract
Cancer is a disease that develops when cells begin to divide uncontrollably and spreads to other parts of the body. Proliferation and invasion of cancerous cells are generally known to be influenced by cell cycle-related proteins in human malignancies. Therefore, in this review, we have emphasized on the serine/threonine kinase named NEK6. NEK6 is been deliberated to play a critical role in mitosis progression that includes mitotic spindle formation, metaphase to anaphase transition, and centrosome separation. Moreover, it has a mechanistic role in DNA repair and can cause apoptosis when inhibited. Past studies have connected NEK6 protein expression to cancer cell senescence. Besides, there are reports relating NEK6 to a range of malignancies including breast, lung, ovarian, prostate, kidney, liver, and others. Given its significance, this review attempts to describe the structural and functional aspects of NEK6 in various cellular processes, as well as how it is linked to different forms of cancer. Lastly, we have accentuated, on some of the plausible inhibitors that have been explored against NEK6 overexpression.
Collapse
Affiliation(s)
- Nagesh Kishan Panchal
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, 632014, India
| | - Shruti Mohanty
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, 632014, India
| | - Sabina Evan Prince
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore, 632014, India.
| |
Collapse
|
7
|
Li SC, Jia ZK, Yang JJ, Ning XH. Telomere-related gene risk model for prognosis and drug treatment efficiency prediction in kidney cancer. Front Immunol 2022; 13:975057. [PMID: 36189312 PMCID: PMC9523360 DOI: 10.3389/fimmu.2022.975057] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Kidney cancer is one of the most common urological cancers worldwide, and kidney renal clear cell cancer (KIRC) is the major histologic subtype. Our previous study found that von-Hippel Lindau (VHL) gene mutation, the dominant reason for sporadic KIRC and hereditary kidney cancer-VHL syndrome, could affect VHL disease-related cancers development by inducing telomere shortening. However, the prognosis role of telomere-related genes in kidney cancer has not been well discussed. In this study, we obtained the telomere-related genes (TRGs) from TelNet. We obtained the clinical information and TRGs expression status of kidney cancer patients in The Cancer Genome Atlas (TCGA) database, The International Cancer Genome Consortium (ICGC) database, and the Clinical Proteomic Tumor Analysis Consortium (CPTAC) database. Totally 353 TRGs were differential between tumor and normal tissues in the TCGA-KIRC dataset. The total TCGA cohort was divided into discovery and validation TCGA cohorts and then using univariate cox regression, lasso regression, and multivariate cox regression method to conduct data analysis sequentially, ten TRGs (ISG15, RFC2, TRIM15, NEK6, PRKCQ, ATP1A1, ELOVL3, TUBB2B, PLCL1, NR1H3) risk model had been constructed finally. The kidney patients in the high TRGs risk group represented a worse outcome in the discovery TCGA cohort (p<0.001), and the result was validated by these four cohorts (validation TCGA cohort, total TCGA cohort, ICGC cohort, and CPTAC cohort). In addition, the TRGs risk score is an independent risk factor for kidney cancer in all these five cohorts. And the high TRGs risk group correlated with worse immune subtypes and higher tumor mutation burden in cancer tissues. In addition, the high TRGs risk group might benefit from receiving immune checkpoint inhibitors and targeted therapy agents. Moreover, the proteins NEK6, RF2, and ISG15 were upregulated in tumors both at the RNA and protein levels, while PLCL1 and PRKCQ were downregulated. The other five genes may display the contrary expression status at the RNA and protein levels. In conclusion, we have constructed a telomere-related genes risk model for predicting the outcomes of kidney cancer patients, and the model may be helpful in selecting treatment agents for kidney cancer patients.
Collapse
|
8
|
Joseph BB, Edeen PT, Meadows S, Binti S, Fay DS. An unexpected role for the conserved ADAM-family metalloprotease ADM-2 in Caenorhabditis elegans molting. PLoS Genet 2022; 18:e1010249. [PMID: 35639786 PMCID: PMC9187072 DOI: 10.1371/journal.pgen.1010249] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 06/10/2022] [Accepted: 05/11/2022] [Indexed: 11/18/2022] Open
Abstract
Molting is a widespread developmental process in which the external extracellular matrix (ECM), the cuticle, is remodeled to allow for organismal growth and environmental adaptation. Studies in the nematode Caenorhabditis elegans have identified a diverse set of molting-associated factors including signaling molecules, intracellular trafficking regulators, ECM components, and ECM-modifying enzymes such as matrix metalloproteases. C. elegans NEKL-2 and NEKL-3, two conserved members of the NEK family of protein kinases, are essential for molting and promote the endocytosis of environmental steroid-hormone precursors by the epidermis. Steroids in turn drive the cyclic induction of many genes required for molting. Here we report a role for the sole C. elegans ADAM–meltrin metalloprotease family member, ADM-2, as a mediator of molting. Loss of adm-2, including mutations that disrupt the metalloprotease domain, led to the strong suppression of molting defects in partial loss-of-function nekl mutants. ADM-2 is expressed in the epidermis, and its trafficking through the endo-lysosomal network was disrupted after NEKL depletion. We identified the epidermally expressed low-density lipoprotein receptor–related protein, LRP-1, as a candidate target of ADM-2 regulation. Whereas loss of ADM-2 activity led to the upregulation of apical epidermal LRP-1, ADM-2 overexpression caused a reduction in LRP-1 levels. Consistent with this, several mammalian ADAMs, including the meltrin ADAM12, have been shown to regulate mammalian LRP1 via proteolysis. In contrast to mammalian homologs, however, the regulation of LRP-1 by ADM-2 does not appear to involve the metalloprotease function of ADM-2, nor is proteolytic processing of LRP-1 strongly affected in adm-2 mutants. Our findings suggest a noncanonical role for an ADAM family member in the regulation of a lipoprotein-like receptor and lead us to propose that endocytic trafficking may be important for both the internalization of factors that promote molting as well as the removal of proteins that can inhibit the process. The molecular and cellular features of molting in nematodes share many similarities with cellular and developmental processes that occur in mammals. This includes the degradation and reorganization of extracellular matrix materials that surround cells, as well as the intracellular machineries that allow cells to sample and modify their environments. In the current study, we found an unexpected function for a conserved protein that cleaves other proteins on the external surface of cells. Rather than promoting molting through extracellular matrix reorganization, however, the ADM-2 protease appears to function as a negative regulator of molting. This observation can be explained in part by data showing that ADM-2 negatively regulates a cell surface receptor required for molting. Surprisingly, it appears to do so through a mechanism that does not involve proteolysis. Our data provide insights into the mechanisms controlling molting and link several conserved proteins to show how they function together during development.
Collapse
Affiliation(s)
- Braveen B. Joseph
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, Wyoming, United States of America
| | - Phillip T. Edeen
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, Wyoming, United States of America
| | - Sarina Meadows
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, Wyoming, United States of America
| | - Shaonil Binti
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, Wyoming, United States of America
| | - David S. Fay
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, Wyoming, United States of America
- * E-mail:
| |
Collapse
|
9
|
In Mitosis You Are Not: The NIMA Family of Kinases in Aspergillus, Yeast, and Mammals. Int J Mol Sci 2022; 23:ijms23074041. [PMID: 35409400 PMCID: PMC8999480 DOI: 10.3390/ijms23074041] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 11/17/2022] Open
Abstract
The Never in mitosis gene A (NIMA) family of serine/threonine kinases is a diverse group of protein kinases implicated in a wide variety of cellular processes, including cilia regulation, microtubule dynamics, mitotic processes, cell growth, and DNA damage response. The founding member of this family was initially identified in Aspergillus and was found to play important roles in mitosis and cell division. The yeast family has one member each, Fin1p in fission yeast and Kin3p in budding yeast, also with functions in mitotic processes, but, overall, these are poorly studied kinases. The mammalian family, the main focus of this review, consists of 11 members named Nek1 to Nek11. With the exception of a few members, the functions of the mammalian Neks are poorly understood but appear to be quite diverse. Like the prototypical NIMA, many members appear to play important roles in mitosis and meiosis, but their functions in the cell go well beyond these well-established activities. In this review, we explore the roles of fungal and mammalian NIMA kinases and highlight the most recent findings in the field.
Collapse
|
10
|
Gao WL, Niu L, Chen WL, Zhang YQ, Huang WH. Integrative Analysis of the Expression Levels and Prognostic Values for NEK Family Members in Breast Cancer. Front Genet 2022; 13:798170. [PMID: 35368696 PMCID: PMC8967485 DOI: 10.3389/fgene.2022.798170] [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: 11/01/2021] [Accepted: 02/07/2022] [Indexed: 12/29/2022] Open
Abstract
Background: In the latest rankings, breast cancer ranks first in incidence and fifth in mortality among female malignancies worldwide. Early diagnosis and treatment can improve the prognosis and prolong the survival of breast cancer (BC) patients. The NIMA-related kinase (NEK), a group of serine/threonine kinase, is a large and conserved gene family that includes NEK1–NEK11. The NEK plays a pivotal role in the cell cycle and microtubule formation. However, an integrative analysis of the effect and prognosis value of NEK family members on BC patients is still lacking. Methods: In this study, the expression profiles of NEK family members in BC and its subgroups were analyzed using UALCAN, GEPIA2, and Human Protein Atlas datasets. The prognostic values of NEK family members in BC were evaluated using the Kaplan–Meier plotter. Co-expression profiles and genetic alterations of NEK family members were analyzed using the cBioPortal database. The function and pathway enrichment analysis of the NEK family were performed using the WebGestalt database. The correlation analysis of the NEK family and immune cell infiltration in BC was conducted using the TIMER 2.0 database. Results: In this study, we compared and analyzed the prognosis values of the NEKs. We found that NEK9 was highly expressed in normal breast tissues than BC, and NEK2, NEK6, and NEK11 were significantly highly expressed in BC than adjacent normal tissues. Interestingly, the expression levels of NEK2, NEK6, and NEK10 were not only remarkably correlated with the tumor stage but also with the molecular subtype. Through multilevel research, we found that high expression levels of NEK1, NEK3, NEK8, NEK9, NEK10, and NEK11 suggested a better prognosis value in BC, while high expression levels of NEK2 and NEK6 suggested a poor prognosis value in BC. Conclusion: Our studies show the prognosis values of the NEKs in BC. Thus, we suggest that NEKs may be regarded as novel biomarkers for predicting potential prognosis values and potential therapeutic targets of BC patients.
Collapse
Affiliation(s)
- Wen-Liang Gao
- Department of Breast-Thyroid-Surgery and Cancer Research Center, Xiang’an Hospital of Xiamen University, Xiamen, China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid Cancers, Xiamen, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiamen, China
| | - Lei Niu
- Department of Breast-Thyroid-Surgery and Cancer Research Center, Xiang’an Hospital of Xiamen University, Xiamen, China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid Cancers, Xiamen, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiamen, China
| | - Wei-Ling Chen
- Department of Breast-Thyroid-Surgery and Cancer Research Center, Xiang’an Hospital of Xiamen University, Xiamen, China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid Cancers, Xiamen, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiamen, China
| | - Yong-Qu Zhang
- Department of Breast-Thyroid-Surgery and Cancer Research Center, Xiang’an Hospital of Xiamen University, Xiamen, China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid Cancers, Xiamen, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiamen, China
- *Correspondence: Yong-Qu Zhang, ; Wen-He Huang,
| | - Wen-He Huang
- Department of Breast-Thyroid-Surgery and Cancer Research Center, Xiang’an Hospital of Xiamen University, Xiamen, China
- Xiamen Research Center of Clinical Medicine in Breast and Thyroid Cancers, Xiamen, China
- Xiamen Key Laboratory of Endocrine-Related Cancer Precision Medicine, Xiamen, China
- *Correspondence: Yong-Qu Zhang, ; Wen-He Huang,
| |
Collapse
|
11
|
Yin S, Yang S, Luo Y, Lu J, Hu G, Wang K, Shao Y, Zhou S, Koo S, Qiu Y, Wang T, Yu H. Cyclin-dependent kinase 1 as a potential target for lycorine against hepatocellular carcinoma. Biochem Pharmacol 2021; 193:114806. [PMID: 34673013 DOI: 10.1016/j.bcp.2021.114806] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 10/03/2021] [Accepted: 10/04/2021] [Indexed: 12/15/2022]
Abstract
The pathological changes and possible underlying molecular mechanisms of hepatocellular carcinoma (HCC) are currently unclear. Effective treatment of this pathological state remains a challenge. The purpose of this study is to obtain some key genes with diagnostic and prognostic meaning and to identify potential therapeutic agents for HCC treatment. Here, CDK1, CCNB1 and CCNB2 were found to be highly expressed in HCC patients and accompanied by poor prognosis, and knockdown of them by siRNA drastically induced autophagy and senescence in hepatoma cells. Simultaneously, the anti-HCC effect of lycorine was comparable to that of interfering with these three genes, and lycorine significantly promoted the decrease both in protein and mRNA expression of CDK1. Molecular validation mechanistically demonstrated that lycorine might attenuate the degradation rate of CDK1 via interaction with it, which had been confirmed by cellular thermal shift assay and drug affinity responsive targets stability assay. Taken together, these findings suggested that CDK1, CCNB1 and CCNB2 could be regarded as potential diagnostic and prognostic biomarkers for HCC, and CDK1 might serve as a promising therapeutic target for lycorine against HCC.
Collapse
Affiliation(s)
- Shuangshuang Yin
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Shenshen Yang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Yanming Luo
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Jia Lu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Gaoyong Hu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Kailong Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Yingying Shao
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Shiyue Zhou
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China
| | - Sangho Koo
- Department of Chemistry, Myongji University, Yongin, Gyeonggi-Do 17058, South Korea
| | - Yuling Qiu
- School of Pharmacy, Tianjin Medical University, Tianjin 300070, China.
| | - Tao Wang
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
| | - Haiyang Yu
- State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 300193, China.
| |
Collapse
|
12
|
Hong J, Won M, Ro H. The Molecular and Pathophysiological Functions of Members of the LNX/PDZRN E3 Ubiquitin Ligase Family. Molecules 2020; 25:E5938. [PMID: 33333989 PMCID: PMC7765395 DOI: 10.3390/molecules25245938] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/10/2020] [Accepted: 12/10/2020] [Indexed: 12/27/2022] Open
Abstract
The ligand of Numb protein-X (LNX) family, also known as the PDZRN family, is composed of four discrete RING-type E3 ubiquitin ligases (LNX1, LNX2, LNX3, and LNX4), and LNX5 which may not act as an E3 ubiquitin ligase owing to the lack of the RING domain. As the name implies, LNX1 and LNX2 were initially studied for exerting E3 ubiquitin ligase activity on their substrate Numb protein, whose stability was negatively regulated by LNX1 and LNX2 via the ubiquitin-proteasome pathway. LNX proteins may have versatile molecular, cellular, and developmental functions, considering the fact that besides these proteins, none of the E3 ubiquitin ligases have multiple PDZ (PSD95, DLGA, ZO-1) domains, which are regarded as important protein-interacting modules. Thus far, various proteins have been isolated as LNX-interacting proteins. Evidence from studies performed over the last two decades have suggested that members of the LNX family play various pathophysiological roles primarily by modulating the function of substrate proteins involved in several different intracellular or intercellular signaling cascades. As the binding partners of RING-type E3s, a large number of substrates of LNX proteins undergo degradation through ubiquitin-proteasome system (UPS) dependent or lysosomal pathways, potentially altering key signaling pathways. In this review, we highlight recent and relevant findings on the molecular and cellular functions of the members of the LNX family and discuss the role of the erroneous regulation of these proteins in disease progression.
Collapse
Affiliation(s)
- Jeongkwan Hong
- Department of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University, Daejeon 305-764, Korea;
| | - Minho Won
- Biotechnology Process Engineering Center, Korea Research Institute of Bioscience & Biotechnology (KRIBB), 30 Yeongudanji-ro, Cheongwon-gu, Cheongju 28116, Korea
| | - Hyunju Ro
- Department of Biological Sciences, College of Bioscience and Biotechnology, Chungnam National University, Daejeon 305-764, Korea;
| |
Collapse
|
13
|
MicroRNA-325-3p Facilitates Immune Escape of Mycobacterium tuberculosis through Targeting LNX1 via NEK6 Accumulation to Promote Anti-Apoptotic STAT3 Signaling. mBio 2020; 11:mBio.00557-20. [PMID: 32487755 PMCID: PMC7267881 DOI: 10.1128/mbio.00557-20] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Intracellular survival of Mycobacterium tuberculosis results in bacterial proliferation and the spread of infection in lungs, consequently deteriorating the conditions of tuberculosis (TB) patients. This research discovers a new immune escape pathway of M. tuberculosis by modulating host miR-325-3p expression, thus leading to the intracellular survival of M. tuberculosis. These findings make a contribution to the understanding of the immune escape of M. tuberculosis, and they provide a theoretical basis for the development of therapeutic approaches for drug-resistant TB. Tuberculosis (TB) is an infectious disease caused by Mycobacterium tuberculosis that poses threats to the public. M. tuberculosis survives in macrophages by escaping from immune surveillance and clearance, which exacerbates the bacterial proliferation. However, the molecular mechanisms of this immune escape have not yet been fully understood. Using multiple cell and mouse models, we found that microRNA-325-3p (miR-325-3p) is upregulated after M. tuberculosis infection and Mir325-deficient mice show resistance to M. tuberculosis. We demonstrated that miR-325-3p directly targets LNX1, an E3 ubiquitin ligase of NEK6, and that this hampers the proteasomal degradation of NEK6 in macrophages. The abnormal accumulation of NEK6 leads to the activation of STAT3 signaling, thus inhibiting the process of apoptosis and promoting the intracellular survival of M. tuberculosis. Our findings not only reveal a new immune escape pathway of M. tuberculosis but also may provide new insights into the development of therapeutic approaches for drug-resistant TB.
Collapse
|
14
|
Melo-Hanchuk TD, Martins MB, Cunha LL, Soares FA, Ward LS, Vassallo J, Kobarg J. Expression of the NEK family in normal and cancer tissue: an immunohistochemical study. BMC Cancer 2020; 20:23. [PMID: 31906878 PMCID: PMC6945616 DOI: 10.1186/s12885-019-6408-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 11/28/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The NEK serine/threonine protein kinases are involved in cell cycle checkpoints, DNA damage repair, and apoptosis. Alterations in these pathways are frequently associated with cell malignant cellular transformations. Thyroid cancer is the most common malignant tumour in the endocrine system. Despite good treatment methods, the number of cases has increased significantly in recent years. Here, we studied the expression of NEK1, NEK2, NEK3, and NEK5 in different types of normal and malignant tissues, using tissue microarray analysis, and identified NEKs as potential markers in thyroid malignancy. METHODS The studied cases comprised multiple cancer tissue microarrays, including breast, colon, esophagus, kidney, lung, pancreas, prostate, stomach, thyroid and uterine cervix, as well as 281 patients who underwent thyroid resection for thyroid cancer or thyroid nodules. The expression of NEK1, NEK2, NEK3, and NEK5 was analyzed by immunohistochemistry. The expression pattern was evaluated in terms of intensity by two methods, semiquantitative and quantitative, and was compared between normal and cancer tissue. RESULTS We analysed the expression of each member of the NEK family in a tissue-dependent manner. Compared to normal tissue, most of the evaluated proteins showed lower expression in lung tumour. However, in the thyroid, the expression was higher in malignant tissue, especially for NEK 1, NEK3 and NEK5. Concerning characteristics of the thyroid tumour, such as aggressiveness, NEK1 expression was higher in tumours with multifocality and in patients with lymph node metastasis. NEK3 expression was stronger in patients with stage II, that involved metastasis. NEK5, on the other hand, showed high expression in patients with invasion and metastasis and in patients with tumour size > 4 cm. Furthermore, this work, demonstrated for the first time a high specificity and sensitivity of over-expression of NEK1 in classical and follicular variants of papillary thyroid cancer and NEK3 in tall-cell papillary thyroid cancer. CONCLUSION Taken together, the NEK protein kinases emerge as important proteins in thyroid cancer development and may help to identify malignancy and aggressiveness features during diagnosis. TRIAL REGISTRATION This study was retrospectively registered. www.accamargo.org.br/cientistas-pesquisadores/comite-de-etica-em-pequisa-cep.
Collapse
Affiliation(s)
- Talita Diniz Melo-Hanchuk
- Departamento de Bioquímica e de Biologia Tecidual, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - Mariana Bonjiorno Martins
- Departamento de Bioquímica e de Biologia Tecidual, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - Lucas Leite Cunha
- Laboratório de Genética Molecular do Câncer, Faculdade de Ciências Médicas Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | | | - Laura Sterian Ward
- Laboratório de Genética Molecular do Câncer, Faculdade de Ciências Médicas Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - José Vassallo
- Departamento de Anatomia Patológica, Faculdade de Ciências Médicas, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil
| | - Jörg Kobarg
- Departamento de Bioquímica e de Biologia Tecidual, Instituto de Biologia, Universidade Estadual de Campinas, Campinas, São Paulo, Brazil. .,Faculdade de Ciências Farmacêuticas-UNICAMP, Universidade Estadual de Campinas, Campinas, Inst. de Biologia, Dep. Bioquímica e Biologia Tecidual, Rua Monteiro Lobato 255, CEP 13083-862, Campinas-SP, Brazil.
| |
Collapse
|
15
|
He Z, Ni X, Xia L, Shao Z. Overexpression of NIMA-related kinase 6 (NEK6) contributes to malignant growth and dismal prognosis in Human Breast Cancer. Pathol Res Pract 2018; 214:1648-1654. [DOI: 10.1016/j.prp.2018.07.030] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 07/16/2018] [Accepted: 07/25/2018] [Indexed: 12/16/2022]
|
16
|
Hirai Y, Tamura M, Otani J, Ishikawa F. NEK6-mediated phosphorylation of human TPP1 regulates telomere length through telomerase recruitment. Genes Cells 2016; 21:874-89. [DOI: 10.1111/gtc.12391] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 05/31/2016] [Indexed: 12/29/2022]
Affiliation(s)
- Yugo Hirai
- Department of Gene Mechanisms; Graduate School of Biostudies; Kyoto University; Yoshida-Konoe-cho Sakyo-ku Kyoto 606-8501 Japan
| | - Miki Tamura
- Department of Gene Mechanisms; Graduate School of Biostudies; Kyoto University; Yoshida-Konoe-cho Sakyo-ku Kyoto 606-8501 Japan
| | - Junji Otani
- Department of Gene Mechanisms; Graduate School of Biostudies; Kyoto University; Yoshida-Konoe-cho Sakyo-ku Kyoto 606-8501 Japan
| | - Fuyuki Ishikawa
- Department of Gene Mechanisms; Graduate School of Biostudies; Kyoto University; Yoshida-Konoe-cho Sakyo-ku Kyoto 606-8501 Japan
| |
Collapse
|
17
|
Sand M, Bechara FG, Gambichler T, Sand D, Bromba M, Hahn SA, Stockfleth E, Hessam S. Circular RNA expression in cutaneous squamous cell carcinoma. J Dermatol Sci 2016; 83:210-8. [PMID: 27298156 DOI: 10.1016/j.jdermsci.2016.05.012] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Revised: 03/12/2016] [Accepted: 05/23/2016] [Indexed: 12/24/2022]
Abstract
BACKGROUND CircularRNAs (circRNAs) are a reinvented class of abundant, stable, and evolutionary conserved non-coding RNAs with pivotal impacts on the cellular regulatory network and epigenetics by sequestering microRNAs (miRNAs) like a sponge. OBJECTIVE Purpose of the present study was to investigate circRNA expression in cutaneous squamous cell carcinoma (cSCC). METHODS A total of six cSCC and six non-lesional skin (control) biopsies were harvested. Microarray based circRNA expression was determined in the cSCC (n=3) and compared with the non-lesional skin (n=3) from a group of 13,617 distinct human circRNAs found in the Arraystar circRNA Array V2.0 (Arraystar, Rockville, USA). Microarray data were validated by quantitative real-time reverse transcription polymerase chain reaction in a separate group (cSCC, n=3 and non-lesional skin, n=3). miRNA binding to miRNA response elements (MREs) sequence data were acquired bioinformatically. Further data mining was performed to identify circRNAs containing MRE sequences that interacted with previously described miRNAs playing a role in cSCC formation. RESULTS A total of 322 circRNAs (143 up- and 179 down-regulated; fold change ≥2 and p<0.05) were identified as differentially expressed in cSCC. Furthermore, we identified a total of 1603 MREs that were part of the differentially expressed circRNAs. Among those circRNAs, a complementary MRE sequence was identified in 23 miRNAs previously known to be cSCC relevant. CONCLUSION This study showed that circRNAs are differentially expressed in cSCC and play an important role in tumor formation by interfering with cSCC relevant miRNAs through miRNA sequence complementary MREs participating in epigenetic control.
Collapse
Affiliation(s)
- Michael Sand
- Dermatologic Surgery Unit, Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum 44791 Bochum, Germany; Department of Plastic Surgery, St. Josef Hospital, Catholic Clinics of the Ruhr Peninsula, 45257 Essen, Germany.
| | - Falk G Bechara
- Dermatologic Surgery Unit, Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum 44791 Bochum, Germany
| | - Thilo Gambichler
- Dermatologic Surgery Unit, Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum 44791 Bochum, Germany
| | - Daniel Sand
- University of Michigan Kellogg Eye Center, Ann Arbor, MI 48105, USA
| | - Michael Bromba
- Department of Plastic Surgery, St. Josef Hospital, Catholic Clinics of the Ruhr Peninsula, 45257 Essen, Germany
| | - Stephan A Hahn
- Department of Internal Medicine, Knappschaftskrankenhaus University of Bochum, Zentrum für Klinische Forschung, Labor für Molekulare Gastroenterologische Onkologie, 44780 Bochum, Germany, Germany
| | - Eggert Stockfleth
- Dermatologic Surgery Unit, Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum 44791 Bochum, Germany
| | - Schapoor Hessam
- Dermatologic Surgery Unit, Department of Dermatology, Venereology and Allergology, Ruhr-University Bochum 44791 Bochum, Germany
| |
Collapse
|
18
|
Yochem J, Lažetić V, Bell L, Chen L, Fay D. C. elegans NIMA-related kinases NEKL-2 and NEKL-3 are required for the completion of molting. Dev Biol 2015; 398:255-66. [PMID: 25523392 PMCID: PMC4314388 DOI: 10.1016/j.ydbio.2014.12.008] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 11/29/2014] [Accepted: 12/04/2014] [Indexed: 12/20/2022]
Abstract
Caenorhabditis elegans molting is a process during which the apical extracellular matrix of the epidermis, the cuticle, is remodeled through a process of degradation and re-synthesis. Using a genetic approach, we identified nekl-3 as essential for the completion of molting. NEKL-3 is highly similar to the mammalian NEK kinase family members NEK6 and NEK7. Animals homozygous for a hypomorphic mutation in nekl-3, sv3, had a novel molting defect in which the central body region, but not the head or tail, was unable to shed the old cuticle. In contrast, a null mutation in nekl-3, gk506, led to complete enclosure within the old cuticle. nekl-2, which is most similar to mammalian NEK8, was also essential for molting. Mosaic analyses demonstrated that NEKL-2 and NEKL-3 were specifically required within the large epidermal syncytium, hyp7, to facilitate molting. Consistent with this, NEKL-2 and NEKL-3 were expressed at the apical surface of hyp7 where they localized to small spheres or tubular structures. Inhibition of nekl-2, but not nekl-3, led to the mislocalization of LRP-1/megalin, a cell surface receptor for low-density lipoprotein (LDL)-binding proteins. In addition, nekl-2 inhibition led to the mislocalization of several other endosome-associated proteins. Notably, LRP-1 acts within hyp7 to facilitate completion of molting, suggesting at least one mechanism by which NEKL-2 may influence molting. Notably, our studies failed to reveal a requirement for NEKL-2 or NEKL-3 in cell division, a function reported for several mammalian NEKs including NEK6 and NEK7. Our findings provide the first genetic and in vivo evidence for a role of NEK family members in endocytosis, which may be evolutionarily conserved.
Collapse
Affiliation(s)
- John Yochem
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, WY 82071, United States; Department of Genetics, Cell Biology, and Development and the Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, United States
| | - Vladimir Lažetić
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, WY 82071, United States
| | - Leslie Bell
- Department of Genetics, Cell Biology, and Development and the Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, United States
| | - Lihsia Chen
- Department of Genetics, Cell Biology, and Development and the Developmental Biology Center, University of Minnesota, Minneapolis, MN 55455, United States
| | - David Fay
- Department of Molecular Biology, College of Agriculture and Natural Resources, University of Wyoming, Laramie, WY 82071, United States.
| |
Collapse
|
19
|
Lee T, Di Paola D, Malina A, Mills JR, Kreps A, Grosse F, Tang H, Zannis-Hadjopoulos M, Larsson O, Pelletier J. Suppression of the DHX9 helicase induces premature senescence in human diploid fibroblasts in a p53-dependent manner. J Biol Chem 2014; 289:22798-22814. [PMID: 24990949 PMCID: PMC4132785 DOI: 10.1074/jbc.m114.568535] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 07/02/2014] [Indexed: 12/28/2022] Open
Abstract
DHX9 is an ATP-dependent DEXH box helicase with a multitude of cellular functions. Its ability to unwind both DNA and RNA, as well as aberrant, noncanonical polynucleotide structures, has implicated it in transcriptional and translational regulation, DNA replication and repair, and maintenance of genome stability. We report that loss of DHX9 in primary human fibroblasts results in premature senescence, a state of irreversible growth arrest. This is accompanied by morphological defects, elevation of senescence-associated β-galactosidase levels, and changes in gene expression closely resembling those encountered during replicative (telomere-dependent) senescence. Activation of the p53 signaling pathway was found to be essential to this process. ChIP analysis and investigation of nascent DNA levels revealed that DHX9 is associated with origins of replication and that its suppression leads to a reduction of DNA replication. Our results demonstrate an essential role of DHX9 in DNA replication and normal cell cycle progression.
Collapse
Affiliation(s)
- Teresa Lee
- Department of Biochemistry, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Domenic Di Paola
- Department of Biochemistry, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Abba Malina
- Department of Biochemistry, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - John R Mills
- Department of Biochemistry, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Amina Kreps
- Department of Biochemistry, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Frank Grosse
- Leibniz Institute for Age Research, Fritz Lipmann Institute, Jena D-07745, Germany
| | - Hengli Tang
- Department of Biological Science, Florida State University, Tallahassee, Florida 32306
| | - Maria Zannis-Hadjopoulos
- Department of Biochemistry, McGill University, Montreal, Quebec H3G 1Y6, Canada,; Department of Oncology, McGill University, Montreal, Quebec H3A 1A3, Canada; The Rosalind and Morris Goodman Cancer Research Center, McGill University, Montreal, Quebec H3A 1A3, Canada, and
| | - Ola Larsson
- Department of Oncology-Pathology, Karolinska Institute, Stockholm 171 77, Sweden
| | - Jerry Pelletier
- Department of Biochemistry, McGill University, Montreal, Quebec H3G 1Y6, Canada,; Department of Oncology, McGill University, Montreal, Quebec H3A 1A3, Canada; The Rosalind and Morris Goodman Cancer Research Center, McGill University, Montreal, Quebec H3A 1A3, Canada, and.
| |
Collapse
|
20
|
Meirelles GV, Perez AM, de Souza EE, Basei FL, Papa PF, Melo Hanchuk TD, Cardoso VB, Kobarg J. “Stop Ne(c)king around”: How interactomics contributes to functionally characterize Nek family kinases. World J Biol Chem 2014; 5:141-160. [PMID: 24921005 PMCID: PMC4050109 DOI: 10.4331/wjbc.v5.i2.141] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Revised: 01/07/2014] [Accepted: 02/18/2014] [Indexed: 02/05/2023] Open
Abstract
Aside from Polo and Aurora, a third but less studied kinase family involved in mitosis regulation is the never in mitosis-gene A (NIMA)-related kinases (Neks). The founding member of this family is the sole member NIMA of Aspergillus nidulans, which is crucial for the initiation of mitosis in that organism. All 11 human Neks have been functionally assigned to one of the three core functions established for this family in mammals: (1) centrioles/mitosis; (2) primary ciliary function/ciliopathies; and (3) DNA damage response (DDR). Recent findings, especially on Nek 1 and 8, showed however, that several Neks participate in parallel in at least two of these contexts: primary ciliary function and DDR. In the core section of this in-depth review, we report the current detailed functional knowledge on each of the 11 Neks. In the discussion, we return to the cross-connections among Neks and point out how our and other groups’ functional and interactomics studies revealed that most Neks interact with protein partners associated with two if not all three of the functional contexts. We then raise the hypothesis that Neks may be the connecting regulatory elements that allow the cell to fine tune and synchronize the cellular events associated with these three core functions. The new and exciting findings on the Nek family open new perspectives and should allow the Neks to finally claim the attention they deserve in the field of kinases and cell cycle biology.
Collapse
|