1
|
Constanthin PE, Isidor N, De Seigneux S, Momjian S. Urinary oxytocin secretion after pituitary surgery, early arginine vasopressin deficiency and syndrome of inappropriate antidiuresis. Endocrine 2025; 88:262-272. [PMID: 39681826 PMCID: PMC11933140 DOI: 10.1007/s12020-024-04131-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024]
Abstract
PURPOSE Transient arginine vasopressin deficiency (AVP-D), previously called diabetes insipidus, is a well-known complication of transsphenoidal pituitary surgery (TPS) with no definite predictive biomarker to date making it difficult to anticipate. While oxytocin (OXT) was previously suggested as a possible biomarker to predict syndrome of inappropriate diuresis (SIAD)-related hyponatraemia after TPS, its secretion in patients presenting with AVP-D remains poorly understood. We therefore hypothesized that OXT might present a different secretion in the case of AVP-D which would support its potential as an early biomarker of AVP-D. Moreover, we hypothesized that abnormal secretion of OXT might occur later on, notably with SIAD. METHODS We measured the urinary output of OXT in 67 consecutive patients subjected to TPS and compared the values of oxytocin between time-points and OXT ratio between groups. The primary endpoint of our study was to identify a difference in urinary OXT excretion in patients suffering from AVP-D compared to patients remaining normonatraemic. As a secondary endpoint, we compared the evolution of OXT secretion after the diagnosis of AVP-D in both groups, comparing the patients that later developed SIAD with the ones that did not. RESULTS Patients developing AVP-D showed a delay in the increase of OXT secretion after TPS as shown by a significantly lower ratio of OXT between the first postoperative day and the day of surgery (0.88 VS 1.68, p = 0.0162, IC:0.2979-0.2642) but a significantly higher ratio of OXT between the fourth and the first postoperative days (1.17 VS 0.53, p = 0.0006, IC:-2.109-0.6092). Moreover, normonatraemic patients that did not show normalization of OXT levels at day 4 after surgery tended to develop SIAD later on. CONCLUSION Taken together, these results show for the first time that OXT release might help predict AVP-D after TPS and differentiate it from other pathologies of water-sodium balance.
Collapse
Affiliation(s)
- Paul E Constanthin
- Department of Neurosurgery, Hôpitaux Universitaires de Genève (HUG), Geneva, Switzerland
- Faculty of Medicine, Université de Genève (UNIGE), Geneva, Switzerland
| | - Nathalie Isidor
- NeuroCentre, University Hospitals of Geneva, Geneva, Switzerland
| | - Sophie De Seigneux
- Department of Nephrology, Hôpitaux Universitaires de Genève (HUG), Geneva, Switzerland
| | - Shahan Momjian
- Department of Neurosurgery, Hôpitaux Universitaires de Genève (HUG), Geneva, Switzerland.
- Faculty of Medicine, Université de Genève (UNIGE), Geneva, Switzerland.
| |
Collapse
|
2
|
Qi X, Zhou J, Wang X, Shen Y, Cao Y, Jiang L, Shen M, Zhang H, Wang T, Wei P, Xu R, Yang Y, Ding X, Wang C, Jia X, Yan Q, Li W, Lu C. HPV E6/E7-Induced Acetylation of a Peptide Encoded by a Long Non-Coding RNA Inhibits Ferroptosis to Promote the Malignancy of Cervical Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414018. [PMID: 39836502 PMCID: PMC11905060 DOI: 10.1002/advs.202414018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 01/05/2025] [Indexed: 01/23/2025]
Abstract
Although a fraction of functional peptides concealed within long non-coding RNAs (lncRNAs) is identified, it remains unclear whether lncRNA-encoded peptides are involved in the malignancy of cervical cancer (CC). Here, a 92-amino acid peptide is discovered, which is named TUBORF, encoded by lncRNA TUBA3FP and highly expressed in CC tissues. TUBORF inhibits ferroptosis to promote the malignant proliferation of CC cells. Mechanistically, human papillomavirus (HPV) oncogenes E6 and E7 upregulate TUBORF through CREB-binding protein (CBP)/E1A-binding protein p300 (p300)-mediated histone H3 lysine 27 acetylation (H3K27ac) of lncTUBA3FP enhancer. Furthermore, E6 and E7 elevate and recruit acetyltransferase establishment of sister chromatid cohesion N-acetyltransferase 1 (ESCO1) to bind to and acetylate TUBORF, which facilitates the degradation of immunity-related GTPase Q (IRGQ) via a ubiquitin-proteasome pathway, resulting in the inhibition of ferroptosis and promotion of the malignant proliferation of CC cells. Importantly, silencing ESCO1 or TURORF amplifies anticancer effects by paclitaxel both in CC cells and in vivo. These novel findings reveal oncopeptide TUBORF and its acetyltransferase ESCO1 as important regulators of ferroptosis and tumorigenesis during cervical cancer pathogenesis and establish the scientific basis for targeting these molecules for treating CC.
Collapse
Affiliation(s)
- Xiaoyu Qi
- Department of GynecologyWomen's Hospital of Nanjing Medical UniversityNanjing Women and Children's Healthcare HospitalNanjing Medical UniversityNanjing210004P. R. China
- Department of MicrobiologyNanjing Medical UniversityNanjing211166P. R. China
| | - Jing Zhou
- Department of MicrobiologyNanjing Medical UniversityNanjing211166P. R. China
| | - Xinyue Wang
- Department of MicrobiologyNanjing Medical UniversityNanjing211166P. R. China
| | - Yan Shen
- Department of MicrobiologyNanjing Medical UniversityNanjing211166P. R. China
| | - Yuxun Cao
- Department of MicrobiologyNanjing Medical UniversityNanjing211166P. R. China
| | - Liangzi Jiang
- Department of MicrobiologyNanjing Medical UniversityNanjing211166P. R. China
| | - Miaomiao Shen
- Department of Pathologythe First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjing210029P. R. China
| | - Haoran Zhang
- Department of MicrobiologyNanjing Medical UniversityNanjing211166P. R. China
| | - Tianjiao Wang
- Department of MicrobiologyNanjing Medical UniversityNanjing211166P. R. China
| | - Pengjun Wei
- Department of MicrobiologyNanjing Medical UniversityNanjing211166P. R. China
| | - Ruoqi Xu
- Department of MicrobiologyNanjing Medical UniversityNanjing211166P. R. China
| | - Yue Yang
- Department of MicrobiologyNanjing Medical UniversityNanjing211166P. R. China
| | - Xiangya Ding
- Department of GynecologyWomen's Hospital of Nanjing Medical UniversityNanjing Women and Children's Healthcare HospitalNanjing Medical UniversityNanjing210004P. R. China
- Department of MicrobiologyNanjing Medical UniversityNanjing211166P. R. China
| | - Cong Wang
- Department of Pathologythe First Affiliated Hospital of Nanjing Medical UniversityNanjing Medical UniversityNanjing210029P. R. China
| | - Xuemei Jia
- Department of GynecologyWomen's Hospital of Nanjing Medical UniversityNanjing Women and Children's Healthcare HospitalNanjing Medical UniversityNanjing210004P. R. China
| | - Qin Yan
- Department of MicrobiologyNanjing Medical UniversityNanjing211166P. R. China
- Key Laboratory of Pathogen Biology of Jiangsu ProvinceNanjing Medical UniversityNanjing211166P. R. China
| | - Wan Li
- Department of GynecologyWomen's Hospital of Nanjing Medical UniversityNanjing Women and Children's Healthcare HospitalNanjing Medical UniversityNanjing210004P. R. China
- Department of MicrobiologyNanjing Medical UniversityNanjing211166P. R. China
- Key Laboratory of Pathogen Biology of Jiangsu ProvinceNanjing Medical UniversityNanjing211166P. R. China
- Department of Infectious DiseasesChangzhou Third People's HospitalChangzhou Medical CenterNanjing Medical UniversityNanjing211166P. R. China
| | - Chun Lu
- Department of GynecologyWomen's Hospital of Nanjing Medical UniversityNanjing Women and Children's Healthcare HospitalNanjing Medical UniversityNanjing210004P. R. China
- Department of MicrobiologyNanjing Medical UniversityNanjing211166P. R. China
- Key Laboratory of Pathogen Biology of Jiangsu ProvinceNanjing Medical UniversityNanjing211166P. R. China
- Department of Infectious DiseasesChangzhou Third People's HospitalChangzhou Medical CenterNanjing Medical UniversityNanjing211166P. R. China
| |
Collapse
|
3
|
Güllülü Ö, Mayer BE, Toplek FB. Linking Gene Fusions to Bone Marrow Failure and Malignant Transformation in Dyskeratosis Congenita. Int J Mol Sci 2024; 25:1606. [PMID: 38338888 PMCID: PMC10855549 DOI: 10.3390/ijms25031606] [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: 12/14/2023] [Revised: 01/26/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
Dyskeratosis Congenita (DC) is a multisystem disorder intrinsically associated with telomere dysfunction, leading to bone marrow failure (BMF). Although the pathology of DC is largely driven by mutations in telomere-associated genes, the implications of gene fusions, which emerge due to telomere-induced genomic instability, remain unexplored. We meticulously analyzed gene fusions in RNA-Seq data from DC patients to provide deeper insights into DC's progression. The most significant DC-specific gene fusions were subsequently put through in silico assessments to ascertain biophysical and structural attributes, including charge patterning, inherent disorder, and propensity for self-association. Selected candidates were then analyzed using deep learning-powered structural predictions and molecular dynamics simulations to gauge their potential for forming higher-order oligomers. Our exploration revealed that genes participating in fusion events play crucial roles in upholding genomic stability, facilitating hematopoiesis, and suppressing tumors. Notably, our analysis spotlighted a particularly disordered polyampholyte fusion protein that exhibits robust higher-order oligomerization dynamics. To conclude, this research underscores the potential significance of several high-confidence gene fusions in the progression of BMF in DC, particularly through the dysregulation of genomic stability, hematopoiesis, and tumor suppression. Additionally, we propose that these fusion proteins might hold a detrimental role, specifically in inducing proteotoxicity-driven hematopoietic disruptions.
Collapse
Affiliation(s)
- Ömer Güllülü
- Department of Structural Biology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Benjamin E. Mayer
- Computational Biology & Simulation, Technische Universität Darmstadt, 64287 Darmstadt, Germany
| | - Fran Bačić Toplek
- Dipartimento di Bioscienze, Università degli Studi di Milano, 20133 Milano, Italy
| |
Collapse
|
4
|
Comprehensive Analysis of Transcriptomic Profiles Identified the Prediction of Prognosis and Drug Sensitivity of Aminopeptidase-Like 1 (NPEPL1) for Clear Cell Renal Cell Carcinoma. JOURNAL OF ONCOLOGY 2023; 2023:4732242. [PMID: 36816355 PMCID: PMC9931475 DOI: 10.1155/2023/4732242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 10/24/2022] [Accepted: 11/24/2022] [Indexed: 02/10/2023]
Abstract
Aminopeptidase-like 1 (NPEPL1) is a member of the aminopeptidase group that plays a role in the development and progression of various diseases. Expression of NPEPL1 has been reported to be involved in prostate, breast, and colorectal cancers. However, the role and mechanism of NPEPL1 in clear cell renal cell carcinoma (ccRCC) are unclear. The Cancer Genome Atlas (TCGA) and Human Protein Atlas (HPA) databases were used to predict the relationship between clinicopathological features and NPEPL1 expression. Changes in immune status and drug sensitivity with NPEPL1 expression were analyzed by the "CIBERSORT" function in R software. The results found that NPEPL1 expression was upregulated in ccRCC tissues, with expression progressively increasing with ccRCC stage and grade. Patients with high NPEPL1 expression presented with a poor prognosis across different clinicopathological features. Univariate and multivariate Cox regression analyses indicated that aberrant NPEPL1 expression was an independent risk factor for ccRCC. The nomogram showed that NPEPL1 expression improved the accuracy of predicting the prognosis of ccRCC patients. The Gene Ontology (GO) term enrichment analysis and the Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis revealed that NPEPL1 may be involved in the development of ccRCC through the voltage-gated calcium channel complex, channel activity, cAMP signaling pathway, and oxytocin signaling pathway. The coexpression analysis found that NPEPL1 altered tumor characteristics by interacting with related genes. The "CIBERSORT" analysis showed that elevated NPEPL1 expression was followed by an enrichment of regulatory T cells and follicular helper T cells in the microenvironment. The drug sensitivity analysis found patients with high NPEPL1 expression had a higher benefit from axitinib, cisplatin, and GSK429286A. In conclusion, upregulation of NPEPL1 expression was involved in ccRCC prognosis and treatment. NPEPL1 could be used as a therapeutic target to guide clinical dosing.
Collapse
|
5
|
Hou W, Li Y, Zhang J, Xia Y, Wang X, Chen H, Lou H. Cohesin in DNA damage response and double-strand break repair. Crit Rev Biochem Mol Biol 2022; 57:333-350. [PMID: 35112600 DOI: 10.1080/10409238.2022.2027336] [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: 08/05/2021] [Revised: 01/03/2022] [Accepted: 01/06/2022] [Indexed: 11/03/2022]
Abstract
Cohesin, a four-subunit ring comprising SMC1, SMC3, RAD21 and SA1/2, tethers sister chromatids by DNA replication-coupled cohesion (RC-cohesion) to guarantee correct chromosome segregation during cell proliferation. Postreplicative cohesion, also called damage-induced cohesion (DI-cohesion), is an emerging critical player in DNA damage response (DDR). In this review, we sum up recent progress on how cohesin regulates the DNA damage checkpoint activation and repair pathway choice, emphasizing postreplicative cohesin loading and DI-cohesion establishment in yeasts and mammals. DI-cohesion and RC-cohesion show distinct features in many aspects. DI-cohesion near or far from the break sites might undergo different regulations and execute different tasks in DDR and DSB repair. Furthermore, some open questions in this field and the significance of this new scenario to our understanding of genome stability maintenance and cohesinopathies are discussed.
Collapse
Affiliation(s)
- Wenya Hou
- Shenzhen University General Hospital, Guangdong Key Laboratory for Genome Stability & Disease Prevention, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - Yan Li
- Shenzhen University General Hospital, Guangdong Key Laboratory for Genome Stability & Disease Prevention, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - Jiaxin Zhang
- Shenzhen University General Hospital, Guangdong Key Laboratory for Genome Stability & Disease Prevention, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - Yisui Xia
- Shenzhen University General Hospital, Guangdong Key Laboratory for Genome Stability & Disease Prevention, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| | - Xueting Wang
- Shenzhen University General Hospital, Guangdong Key Laboratory for Genome Stability & Disease Prevention, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
- Union Shenzhen Hospital, Department of Dermatology, Huazhong University of Science and Technology (Nanshan Hospital), Shenzhen, Guangdong, China
| | - Hongxiang Chen
- Union Shenzhen Hospital, Department of Dermatology, Huazhong University of Science and Technology (Nanshan Hospital), Shenzhen, Guangdong, China
| | - Huiqiang Lou
- Shenzhen University General Hospital, Guangdong Key Laboratory for Genome Stability & Disease Prevention, Shenzhen University School of Medicine, Shenzhen, Guangdong, China
| |
Collapse
|
6
|
Kawasumi R, Abe T, Psakhye I, Miyata K, Hirota K, Branzei D. Vertebrate CTF18 and DDX11 essential function in cohesion is bypassed by preventing WAPL-mediated cohesin release. Genes Dev 2021; 35:1368-1382. [PMID: 34503989 PMCID: PMC8494208 DOI: 10.1101/gad.348581.121] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/16/2021] [Indexed: 01/26/2023]
Abstract
The alternative PCNA loader containing CTF18-DCC1-CTF8 facilitates sister chromatid cohesion (SCC) by poorly defined mechanisms. Here we found that in DT40 cells, CTF18 acts complementarily with the Warsaw breakage syndrome DDX11 helicase in mediating SCC and proliferation. We uncover that the lethality and cohesion defects of ctf18 ddx11 mutants are associated with reduced levels of chromatin-bound cohesin and rescued by depletion of WAPL, a cohesin-removal factor. On the contrary, high levels of ESCO1/2 acetyltransferases that acetylate cohesin to establish SCC do not rescue ctf18 ddx11 phenotypes. Notably, the tight proximity of sister centromeres and increased anaphase bridges characteristic of WAPL-depleted cells are abrogated by loss of both CTF18 and DDX11 The results reveal that vertebrate CTF18 and DDX11 collaborate to provide sufficient amounts of chromatin-loaded cohesin available for SCC generation in the presence of WAPL-mediated cohesin-unloading activity. This process modulates chromosome structure and is essential for cellular proliferation in vertebrates.
Collapse
Affiliation(s)
- Ryotaro Kawasumi
- International Foundation of Medicine (IFOM), the Fondazione Italiana per la Ricerca sul Cancro (FIRC) Institute for Molecular Oncology Foundation, Milan 20139, Italy
| | - Takuya Abe
- International Foundation of Medicine (IFOM), the Fondazione Italiana per la Ricerca sul Cancro (FIRC) Institute for Molecular Oncology Foundation, Milan 20139, Italy
- Department of Chemistry, Graduate School of Science and Engineering, Tokyo Metropolitan University, Hachioji-shi, Tokyo 192-0397, Japan
| | - Ivan Psakhye
- International Foundation of Medicine (IFOM), the Fondazione Italiana per la Ricerca sul Cancro (FIRC) Institute for Molecular Oncology Foundation, Milan 20139, Italy
| | - Keiji Miyata
- Department of Chemistry, Graduate School of Science and Engineering, Tokyo Metropolitan University, Hachioji-shi, Tokyo 192-0397, Japan
| | - Kouji Hirota
- Department of Chemistry, Graduate School of Science and Engineering, Tokyo Metropolitan University, Hachioji-shi, Tokyo 192-0397, Japan
| | - Dana Branzei
- International Foundation of Medicine (IFOM), the Fondazione Italiana per la Ricerca sul Cancro (FIRC) Institute for Molecular Oncology Foundation, Milan 20139, Italy
- Istituto di Genetica Molecolare, Consiglio Nazionale delle Ricerche (IGM-CNR), Pavia 27100, Italy
| |
Collapse
|
7
|
Ajam T, De I, Petkau N, Whelan G, Pena V, Eichele G. Alternative catalytic residues in the active site of Esco acetyltransferases. Sci Rep 2020; 10:9828. [PMID: 32555289 PMCID: PMC7300003 DOI: 10.1038/s41598-020-66795-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 05/13/2020] [Indexed: 11/20/2022] Open
Abstract
Cohesin is a protein complex whose core subunits, Smc1, Smc3, Scc1, and SA1/SA2 form a ring-like structure encircling the DNA. Cohesins play a key role in the expression, repair, and segregation of eukaryotic genomes. Following a catalytic mechanism that is insufficiently understood, Esco1 and Esco2 acetyltransferases acetylate the cohesin subunit Smc3, thereby inducing stabilization of cohesin on DNA. As a prerequisite for structure-guided investigation of enzymatic activity, we determine here the crystal structure of the mouse Esco2/CoA complex at 1.8 Å resolution. We reconstitute cohesin as tri- or tetrameric assemblies and use those as physiologically-relevant substrates for enzymatic assays in vitro. Furthermore, we employ cell-based complementation studies in mouse embryonic fibroblast deficient for Esco1 and Esco2, as a means to identify catalytically-important residues in vivo. These analyses demonstrate that D567/S566 and E491/S527, located on opposite sides of the murine Esco2 active site cleft, are critical for catalysis. Our experiments support a catalytic mechanism of acetylation where residues D567 and E491 are general bases that deprotonate the ε-amino group of lysine substrate, also involving two nearby serine residues - S566 and S527- that possess a proton relay function.
Collapse
Affiliation(s)
- Tahereh Ajam
- Genes and Behavior Department, Max Planck Institute for Biophysical Chemistry, 37077, Göttingen, Germany
| | - Inessa De
- Research Group Macromolecular Crystallography, Max Planck Institute for Biophysical Chemistry, 37077, Göttingen, Germany.,European Molecular Biology Laboratory (EMBL), Structural and Computational Biology Unit, Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Nikolai Petkau
- Genes and Behavior Department, Max Planck Institute for Biophysical Chemistry, 37077, Göttingen, Germany
| | - Gabriela Whelan
- Genes and Behavior Department, Max Planck Institute for Biophysical Chemistry, 37077, Göttingen, Germany
| | - Vladimir Pena
- Research Group Macromolecular Crystallography, Max Planck Institute for Biophysical Chemistry, 37077, Göttingen, Germany. .,Structural Biology Division, The Institute of Cancer Research, SW3 6JB, London, United Kingdom.
| | - Gregor Eichele
- Genes and Behavior Department, Max Planck Institute for Biophysical Chemistry, 37077, Göttingen, Germany.
| |
Collapse
|
8
|
Leylek TR, Jeusset LM, Lichtensztejn Z, McManus KJ. Reduced Expression of Genes Regulating Cohesion Induces Chromosome Instability that May Promote Cancer and Impact Patient Outcomes. Sci Rep 2020; 10:592. [PMID: 31953484 PMCID: PMC6969069 DOI: 10.1038/s41598-020-57530-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 01/03/2020] [Indexed: 12/15/2022] Open
Abstract
Chromosome instability (CIN), or continual changes in chromosome complements, is an enabling feature of cancer; however, the molecular determinants of CIN remain largely unknown. Emerging data now suggest that aberrant sister chromatid cohesion may induce CIN and contribute to cancer. To explore this possibility, we employed clinical and fundamental approaches to systematically assess the impact reduced cohesion gene expression has on CIN and cancer. Ten genes encoding critical functions in cohesion were evaluated and remarkably, each exhibits copy number losses in 12 common cancer types, and reduced expression is associated with worse patient survival. To gain mechanistic insight, we combined siRNA-based silencing with single cell quantitative imaging microscopy to comprehensively assess the impact reduced expression has on CIN in two karyotypically stable cell lines. We show that reduced expression induces CIN phenotypes, namely increases in micronucleus formation and nuclear areas. Subsequent direct tests involving a subset of prioritized genes also revealed significant changes in chromosome numbers with corresponding increases in moderate and severe cohesion defects within mitotic chromosome spreads. Collectively, our clinical and fundamental findings implicate reduced sister chromatid cohesion, resulting from gene copy number losses, as a key pathogenic event in the development and progression of many cancer types.
Collapse
Affiliation(s)
- Tarik R Leylek
- Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, Manitoba, R3E 0J9, Canada
| | - Lucile M Jeusset
- Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, Manitoba, R3E 0J9, Canada
- Research Institute in Oncology & Hematology, CancerCare Manitoba, Winnipeg, Manitoba, R3E 0V9, Canada
| | - Zelda Lichtensztejn
- Research Institute in Oncology & Hematology, CancerCare Manitoba, Winnipeg, Manitoba, R3E 0V9, Canada
| | - Kirk J McManus
- Department of Biochemistry & Medical Genetics, University of Manitoba, Winnipeg, Manitoba, R3E 0J9, Canada.
- Research Institute in Oncology & Hematology, CancerCare Manitoba, Winnipeg, Manitoba, R3E 0V9, Canada.
| |
Collapse
|
9
|
Kim JT, Cho HJ, Park SY, Oh BM, Hwang YS, Baek KE, Lee YH, Kim HC, Lee HG. DNA Replication and Sister Chromatid Cohesion 1 (DSCC1) of the Replication Factor Complex CTF18-RFC is Critical for Colon Cancer Cell Growth. J Cancer 2019; 10:6142-6153. [PMID: 31762824 PMCID: PMC6856584 DOI: 10.7150/jca.32339] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 08/26/2019] [Indexed: 01/01/2023] Open
Abstract
DNA replication and sister chromatid cohesion 1 (DSCC1) combines with chromosome transmission-fidelity protein 18 (CTF18) to form a CTF18-DSCC1-CTF8 (CTF18-1-8) module, which in combination with CTF18-replication factor C (RFC) acts as a proliferating cell nuclear antigen (PCNA) loader during DNA replication-associated processes. It was found that DSCC1 was overexpressed in tumor tissues from patients with colon cancer and that the survival probability of patients with colon cancer was lower when the expression of cytosolic DSCC1 was higher in tumor regions (P=0.047). By using DSCC1- or CTF18-knockdown cell lines (HCT116-shDSCC1 or HCT116-shCTF18, respectively), it was confirmed that DSCC1-knockdown inhibits cell proliferation and invasion, but that CTF18-knockdown does not. Tumors in mice xenografted with shDSCC1 cells were significantly smaller compared with those in mice in the mock group or those xenografted with shCTF18 cells. The shDSCC1 cells were highly sensitive to γ-irradiation and other DNA replication inhibitory treatments, resulting in low cell viability. The present results suggested that DSCC1 is the most important component in the CTF18-1-8 module for CTF18-RFC and is highly relevant to the growth and metastasis of colon cancer cells, and, therefore, it may be a potential therapeutic target for colon cancer treatment.
Collapse
Affiliation(s)
- Jong-Tae Kim
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Hee Jun Cho
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Sang Yoon Park
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Byung Moo Oh
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Department of Biomolecular Science, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Yo Sep Hwang
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Department of Biomolecular Science, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Kyoung Eun Baek
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Young-Ha Lee
- Department of Infection Biology, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Hee Cheol Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hee Gu Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea.,Department of Biomolecular Science, University of Science and Technology (UST), Daejeon, Republic of Korea
| |
Collapse
|
10
|
Cui ZJ, Zhou XH, Zhang HY. DNA Methylation Module Network-Based Prognosis and Molecular Typing of Cancer. Genes (Basel) 2019; 10:genes10080571. [PMID: 31357729 PMCID: PMC6722866 DOI: 10.3390/genes10080571] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/11/2019] [Accepted: 07/26/2019] [Indexed: 12/25/2022] Open
Abstract
Achieving cancer prognosis and molecular typing is critical for cancer treatment. Previous studies have identified some gene signatures for the prognosis and typing of cancer based on gene expression data. Some studies have shown that DNA methylation is associated with cancer development, progression, and metastasis. In addition, DNA methylation data are more stable than gene expression data in cancer prognosis. Therefore, in this work, we focused on DNA methylation data. Some prior researches have shown that gene modules are more reliable in cancer prognosis than are gene signatures and that gene modules are not isolated. However, few studies have considered cross-talk among the gene modules, which may allow some important gene modules for cancer to be overlooked. Therefore, we constructed a gene co-methylation network based on the DNA methylation data of cancer patients, and detected the gene modules in the co-methylation network. Then, by permutation testing, cross-talk between every two modules was identified; thus, the module network was generated. Next, the core gene modules in the module network of cancer were identified using the K-shell method, and these core gene modules were used as features to study the prognosis and molecular typing of cancer. Our method was applied in three types of cancer (breast invasive carcinoma, skin cutaneous melanoma, and uterine corpus endometrial carcinoma). Based on the core gene modules identified by the constructed DNA methylation module networks, we can distinguish not only the prognosis of cancer patients but also use them for molecular typing of cancer. These results indicated that our method has important application value for the diagnosis of cancer and may reveal potential carcinogenic mechanisms.
Collapse
Affiliation(s)
- Ze-Jia Cui
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China
| | - Xiong-Hui Zhou
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China.
| | - Hong-Yu Zhang
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
11
|
Identification of important invasion and proliferation related genes in adrenocortical carcinoma. Med Oncol 2019; 36:73. [PMID: 31321566 DOI: 10.1007/s12032-019-1296-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 07/01/2019] [Indexed: 12/17/2022]
|
12
|
Wang H, Lv YJ, Xu WH, Pang WF, Zhao YY, Yang N, Wang ZP, Lu L, Liu Y, Zhang SY, Yuan XL. The correlation of ESCO1 expression with a prognosis of prostate cancer and anti-tumor effect of ESCO1 silencing. Transl Cancer Res 2019; 8:950-961. [PMID: 35116834 PMCID: PMC8798848 DOI: 10.21037/tcr.2019.05.34] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 05/30/2019] [Indexed: 11/30/2022]
Abstract
Background Recently, it has been reported that establishment of sister chromatid cohesion N-acetyltransferase 1 (ESCO1) is involved in tumorigenesis. However, its role in prostate cancer remains unclear. In the present study, the association between ESCO1 expression and the prognosis of prostate cancer was investigated, and the potential molecular mechanisms underlying its actions in tumor progression were also examined. Methods Immunohistochemical analysis was performed to detect the expression of ESCO1 in benign prostatic hyperplasia (BPH), human prostate cancer, and metastasis tissue samples, and the association between the establishment of ESCO1 expression and the prognosis of prostate cancer was investigated. The effect of ESCO1 expression on the viability, migration, and invasion of prostate cancer cells in vitro was analyzed, along with the effect of ESCO1 silencing on the growth of prostate tumors in vivo. Results The results demonstrated an increase in the expression of ESCO1 in prostate cancer tissue when compared with BPH, and it was significantly associated with tumor malignancy and poor patient survival. Additionally, knockdown of ESCO1 significantly inhibited the viability and migration of prostate cancer cell. Furthermore, we found that knockdown of ESCO1 significantly inhibited tumor growth in vivo. Pathway analysis identified that the silencing of ESCO1 significantly decreased the phosphorylation levels of protein kinase B. Conclusions The results of the present study indicate that ESCO1 plays a vital role in the progression of human prostate cancer; furthermore, ESCO1 may potentially serve as a prognostic marker and a novel therapeutic target for this disease.
Collapse
Affiliation(s)
- Hui Wang
- Department of Oncology and Cancer Biotherapy Center, The 4th Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yan-Ju Lv
- Department of Oncology and Cancer Biotherapy Center, The 4th Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Wan-Hai Xu
- Department of Urology, The 4th Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Wei-Feng Pang
- Department of Oncology and Cancer Biotherapy Center, The 4th Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Yu-Ying Zhao
- Department of Oncology and Cancer Biotherapy Center, The 4th Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Ning Yang
- Department of Oncology and Cancer Biotherapy Center, The 4th Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Zhi-Peng Wang
- Department of Oncology and Cancer Biotherapy Center, The 4th Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Lu Lu
- Department of Oncology and Cancer Biotherapy Center, The 4th Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Ying Liu
- Department of Oncology and Cancer Biotherapy Center, The 4th Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Shi-Ying Zhang
- Department of Urology, Air Force General Hospital, Beijing 100142, China
| | - Xue-Li Yuan
- Department of Oncology and Cancer Biotherapy Center, The 4th Affiliated Hospital of Harbin Medical University, Harbin 150001, China.,Department of Urology, The 4th Affiliated Hospital of Harbin Medical University, Harbin 150001, China.,Department of Urology, Air Force General Hospital, Beijing 100142, China.,Department of Urology, Peking University Shougang Hospital, Beijing 100144, China
| |
Collapse
|
13
|
Hnisz D, Schuijers J, Li CH, Young RA. Regulation and Dysregulation of Chromosome Structure in Cancer. ANNUAL REVIEW OF CANCER BIOLOGY-SERIES 2018. [DOI: 10.1146/annurev-cancerbio-030617-050134] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cancer arises from genetic alterations that produce dysregulated gene expression programs. Normal gene regulation occurs in the context of chromosome loop structures called insulated neighborhoods, and recent studies have shown that these structures are altered and can contribute to oncogene dysregulation in various cancer cells. We review the types of genetic and epigenetic alterations that influence neighborhood structures and contribute to gene dysregulation in cancer, present models for insulated neighborhoods associated with the most prominent human oncogenes, and discuss how such models may lead to further advances in cancer diagnosis and therapy.
Collapse
Affiliation(s)
- Denes Hnisz
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142, USA;,
| | - Jurian Schuijers
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142, USA;,
| | - Charles H. Li
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142, USA;,
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Richard A. Young
- Whitehead Institute for Biomedical Research, Cambridge, Massachusetts 02142, USA;,
- Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
14
|
Kawasumi R, Abe T, Arakawa H, Garre M, Hirota K, Branzei D. ESCO1/2's roles in chromosome structure and interphase chromatin organization. Genes Dev 2017; 31:2136-2150. [PMID: 29196537 PMCID: PMC5749162 DOI: 10.1101/gad.306084.117] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 11/09/2017] [Indexed: 11/24/2022]
Abstract
In this study, Kawasumi et al. researched how ESCO1/2 acetyltransferases mediating SMC3 acetylation and sister chromatid cohesion (SCC) interact and contribute to chromosome structure and proliferation. Using chicken DT40 cell lines with mutations in ESCO1/2, SMC3 acetylation, and the cohesin remover WAPL, they show that cohesion establishment by vertebrate ESCO1/2 is linked to interphase chromatin architecture formation. ESCO1/2 acetyltransferases mediating SMC3 acetylation and sister chromatid cohesion (SCC) are differentially required for genome integrity and development. Here we established chicken DT40 cell lines with mutations in ESCO1/2, SMC3 acetylation, and the cohesin remover WAPL. Both ESCO1 and ESCO2 promoted SCC, while ESCO2 was additionally and specifically required for proliferation and centromere integrity. ESCO1 overexpression fully suppressed the slow proliferation and centromeric separation phenotypes of esco2 cells but only partly suppressed its chromosome arm SCC defects. Concomitant inactivation of ESCO1 and ESCO2 caused lethality owing to compromised mitotic chromosome segregation. Neither wapl nor acetyl-mimicking smc3-QQ mutations rescued esco1 esco2 lethality. Notably, esco1 esco2 wapl conditional mutants showed very severe proliferation defects associated with catastrophic mitoses and also abnormal interphase chromatin organization patterns. The results indicate that cohesion establishment by vertebrate ESCO1/2 is linked to interphase chromatin architecture formation, a newly identified function of cohesin acetyltransferases that is both fundamentally and medically relevant.
Collapse
Affiliation(s)
- Ryotaro Kawasumi
- The FIRC (Italian Foundation for Cancer Research) Institute of Molecular Oncology (IFOM), 20139 Milan, Italy
| | - Takuya Abe
- The FIRC (Italian Foundation for Cancer Research) Institute of Molecular Oncology (IFOM), 20139 Milan, Italy.,Department of Chemistry, Graduate School of Science and Engineering, Tokyo Metropolitan University, Hachioji-shi, Tokyo 192-0397, Japan
| | - Hiroshi Arakawa
- The FIRC (Italian Foundation for Cancer Research) Institute of Molecular Oncology (IFOM), 20139 Milan, Italy
| | - Massimiliano Garre
- The FIRC (Italian Foundation for Cancer Research) Institute of Molecular Oncology (IFOM), 20139 Milan, Italy
| | - Kouji Hirota
- Department of Chemistry, Graduate School of Science and Engineering, Tokyo Metropolitan University, Hachioji-shi, Tokyo 192-0397, Japan
| | - Dana Branzei
- The FIRC (Italian Foundation for Cancer Research) Institute of Molecular Oncology (IFOM), 20139 Milan, Italy.,Istituto di Genetica Molecolare, Consiglio Nazionale delle Ricerche (IGM-CNR), 27100 Pavia, Italy
| |
Collapse
|
15
|
Le Gallo M, Rudd ML, Urick ME, Hansen NF, Zhang S, Lozy F, Sgroi DC, Vidal Bel A, Matias-Guiu X, Broaddus RR, Lu KH, Levine DA, Mutch DG, Goodfellow PJ, Salvesen HB, Mullikin JC, Bell DW. Somatic mutation profiles of clear cell endometrial tumors revealed by whole exome and targeted gene sequencing. Cancer 2017; 123:3261-3268. [PMID: 28485815 DOI: 10.1002/cncr.30745] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 02/08/2017] [Accepted: 02/14/2017] [Indexed: 01/10/2023]
Abstract
BACKGROUND The molecular pathogenesis of clear cell endometrial cancer (CCEC), a tumor type with a relatively unfavorable prognosis, is not well defined. We searched exome-wide for novel somatically mutated genes in CCEC and assessed the mutational spectrum of known and candidate driver genes in a large cohort of cases. METHODS We conducted whole exome sequencing of paired tumor-normal DNAs from 16 cases of CCEC (12 CCECs and the CCEC components of 4 mixed histology tumors). Twenty-two genes-of-interest were Sanger-sequenced from another 47 cases of CCEC. Microsatellite instability (MSI) and microsatellite stability (MSS) were determined by genotyping 5 mononucleotide repeats. RESULTS Two tumor exomes had relatively high mutational loads and MSI. The other 14 tumor exomes were MSS and had 236 validated nonsynonymous or splice junction somatic mutations among 222 protein-encoding genes. Among the 63 cases of CCEC in this study, we identified frequent somatic mutations in TP53 (39.7%), PIK3CA (23.8%), PIK3R1 (15.9%), ARID1A (15.9%), PPP2R1A (15.9%), SPOP (14.3%), and TAF1 (9.5%), as well as MSI (11.3%). Five of 8 mutations in TAF1, a gene with no known role in CCEC, localized to the putative histone acetyltransferase domain and included 2 recurrently mutated residues. Based on patterns of MSI and mutations in 7 genes, CCEC subsets molecularly resembled serous endometrial cancer (SEC) or endometrioid endometrial cancer (EEC). CONCLUSION Our findings demonstrate molecular similarities between CCEC and SEC and EEC and implicate TAF1 as a novel candidate CCEC driver gene. Cancer 2017;123:3261-8. © 2017 American Cancer Society.
Collapse
Affiliation(s)
- Matthieu Le Gallo
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Meghan L Rudd
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Mary Ellen Urick
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Nancy F Hansen
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Suiyuan Zhang
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | -
- National Institutes of Health Intramural Sequencing Center, National Institutes of Health, Bethesda, Maryland
| | - Fred Lozy
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| | - Dennis C Sgroi
- Molecular Pathology Unit, Massachusetts General Hospital, Charlestown, Massachusetts.,Center for Cancer Research, Massachusetts General Hospital, Charlestown, Massachusetts.,Department of Pathology, Harvard Medical School, Boston, Massachusetts
| | - August Vidal Bel
- Department of Pathology and Experimental Therapeutics, University of Barcelona, Barcelona, Spain.,Department of Pathology, Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Xavier Matias-Guiu
- Department of Pathology, Hospital Universitari de Bellvitge, Barcelona, Spain.,Department of Pathology and Molecular Genetics/Oncological Pathology Group, Hospital Universitari Arnau de Vilanova, Universitat de Lleida, IRB Lleida, Lleida, Spain
| | - Russell R Broaddus
- Division of Surgery, Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Karen H Lu
- Division of Surgery, Department of Gynecologic Oncology and Reproductive Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Douglas A Levine
- Gynecologic Oncology, Perlmutter Cancer Center, New York University Langone Medical Center, New York, New York
| | - David G Mutch
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri
| | - Paul J Goodfellow
- Department of Obstetrics and Gynecology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Helga B Salvesen
- Department of Clinical Science, Center for Cancer Biomarkers, University of Bergen, Bergen, Norway.,Department of Gynecology and Obstetrics, Haukeland University Hospital, Bergen, Norway
| | - James C Mullikin
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland.,National Institutes of Health Intramural Sequencing Center, National Institutes of Health, Bethesda, Maryland
| | - Daphne W Bell
- Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
16
|
Sudha D, Kohansal-Nodehi M, Kovuri P, Manda SS, Neriyanuri S, Gopal L, Bhende P, Chidambaram S, Arunachalam JP. Proteomic profiling of human intraschisis cavity fluid. Clin Proteomics 2017; 14:13. [PMID: 28450823 PMCID: PMC5404285 DOI: 10.1186/s12014-017-9148-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 04/07/2017] [Indexed: 12/24/2022] Open
Abstract
Background X-linked retinoschisis (XLRS) is a vitreoretinal degenerative disorder causing vision deterioration, due to structural defects in retina. The hallmark of this disease includes radial streaks arising from the fovea and splitting of inner retinal layers (schisis). Although these retinal changes are attributed to mutations in the retinoschisin gene, schisis is also observed in patients who do not carry mutations. In addition, the origin of intraschisis fluid, the triggering point of schisis formation and its progression are largely unknown still. So far, there is no report on the complete proteomic analysis of this fluid. Schisis fluid proteome could reflect biochemical changes in the disease condition, helping in better understanding and management of retinoschisis. Therefore it was of interest to investigate the intraschisis fluid proteome using high-resolution mass spectrometry. Methods Two male XLRS patients (aged 4 and 40 years) underwent clinical and genetic evaluation followed by surgical extraction of intraschisis fluids. The two fluid samples were resolved on a SDS-PAGE and the processed peptides were analyzed by Q-Exactive plus hybrid quadrupole-Orbitrap mass spectrometry. Functional annotation of the identified proteins was performed using Ingenuity pathway analysis software. Results Mass spectrometry analysis detected 770 nonredundant proteins in the intraschisis fluid. Retinol dehydrogenase 14 was found to be abundant in the schisis fluid. Gene ontology based analysis indicated that 19% of the intraschisis fluid proteins were localized to the extracellular matrix and 15% of the proteins were involved in signal transduction. Functional annotation identified three primary canonical pathways to be associated with the schisis fluid proteome viz., LXR/RXR activation, complement system and acute phase response signalling, which are involved in immune and inflammatory responses. Collectively, our results show that intraschisis fluid comprises specific inflammatory proteins which highly reflect the disease environment. Conclusion Based on our study, it is suggested that inflammation might play a key role in the pathogenesis of XLRS. To our knowledge, this is the first report describing the complete proteome of intraschisis fluid, which could serve as a template for future research and facilitate the development of therapeutic modalities for XLRS. Electronic supplementary material The online version of this article (doi:10.1186/s12014-017-9148-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Dhandayuthapani Sudha
- SN ONGC Department of Genetics and Molecular Biology, Vision Research Foundation, Chennai, India.,School of Biotechnology, SASTRA University, Thanjavur, India
| | | | - Purnima Kovuri
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry, India
| | | | - Srividya Neriyanuri
- Elite School of Optometry, Unit of Medical Research Foundation, Chennai, India
| | - Lingam Gopal
- Shri Bhagwan Mahavir Vitreo-Retinal Services, Medical Research Foundation, Chennai, India
| | - Pramod Bhende
- Shri Bhagwan Mahavir Vitreo-Retinal Services, Medical Research Foundation, Chennai, India
| | | | - Jayamuruga Pandian Arunachalam
- SN ONGC Department of Genetics and Molecular Biology, Vision Research Foundation, Chennai, India.,Central Inter-Disciplinary Research Facility (CIDRF), Sri Balaji Vidyapeeth Medical University, Mahatma Gandhi Medical College and Research Institute Campus, Puducherry, India
| |
Collapse
|
17
|
Sharma Bhai P, Sharma D, Saxena R, Verma IC. Next-Generation Sequencing Reveals a Nonsense Mutation (p.Arg364Ter) in MRE11A Gene in an Indian Patient with Familial Breast Cancer. Breast Care (Basel) 2017; 12:114-116. [PMID: 28559769 DOI: 10.1159/000457786] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND The MRN complex consisting of MRE11A-RAD50-NBS1 proteins is involved in the repair of double-strand breaks, and mutations in genes coding for the MRN complex have been identified in families with breast and ovarian cancer. CASE REPORT In a BRCA-negative family with positive history of breast and endometrial cancer, next-generation sequencing-based panel testing identified a mutation in the MRE11A gene (NM_005590 c.1090C>T: p.Arg364Ter). This mutation results in a shorter mutated protein lacking 2 DNA binding domains (the GAR domain and the RAD50 binding site), abolishing the function of protein. CONCLUSION This case provides insight into the role of the MRE11A gene in causing breast cancer susceptibility in families, and supports the use of multigene panel testing in cases with hereditary predisposition to breast cancer.
Collapse
Affiliation(s)
- Pratibha Sharma Bhai
- Institute of Medical Genetics and Genomics, Sir Ganga Ram Hospital, Rajinder Nagar, New Delhi, India
| | - Deepak Sharma
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, India
| | - Renu Saxena
- Institute of Medical Genetics and Genomics, Sir Ganga Ram Hospital, Rajinder Nagar, New Delhi, India
| | - Ishwar C Verma
- Institute of Medical Genetics and Genomics, Sir Ganga Ram Hospital, Rajinder Nagar, New Delhi, India
| |
Collapse
|
18
|
Rivera-Colón Y, Maguire A, Liszczak GP, Olia AS, Marmorstein R. Molecular Basis for Cohesin Acetylation by Establishment of Sister Chromatid Cohesion N-Acetyltransferase ESCO1. J Biol Chem 2016; 291:26468-26477. [PMID: 27803161 PMCID: PMC5159507 DOI: 10.1074/jbc.m116.752220] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 10/21/2016] [Indexed: 11/06/2022] Open
Abstract
Protein acetylation is a prevalent posttranslational modification that is regulated by diverse acetyltransferase enzymes. Although histone acetyltransferases (HATs) have been well characterized both structurally and mechanistically, far less is known about non-histone acetyltransferase enzymes. The human ESCO1 and ESCO2 paralogs acetylate the cohesin complex subunit SMC3 to regulate the separation of sister chromatids during mitosis and meiosis. Missense mutations within the acetyltransferase domain of these proteins correlate with diseases, including endometrial cancers and Roberts syndrome. Despite their biological importance, the mechanisms underlying acetylation by the ESCO proteins are not understood. Here, we report the X-ray crystal structure of the highly conserved zinc finger-acetyltransferase moiety of ESCO1 with accompanying structure-based mutagenesis and biochemical characterization. We find that the ESCO1 acetyltransferase core is structurally homologous to the Gcn5 HAT, but contains unique additional features including a zinc finger and an ∼40-residue loop region that appear to play roles in protein stability and SMC3 substrate binding. We identify key residues that play roles in substrate binding and catalysis, and rationalize the functional consequences of disease-associated mutations. Together, these studies reveal the molecular basis for SMC3 acetylation by ESCO1 and have broader implications for understanding the structure/function of non-histone acetyltransferases.
Collapse
Affiliation(s)
- Yadilette Rivera-Colón
- From the Department of Biochemistry and Biophysics, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104 and
| | - Andrew Maguire
- From the Department of Biochemistry and Biophysics, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104 and
- the Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Glen P Liszczak
- From the Department of Biochemistry and Biophysics, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104 and
- the Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Adam S Olia
- From the Department of Biochemistry and Biophysics, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104 and
| | - Ronen Marmorstein
- From the Department of Biochemistry and Biophysics, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104 and
- the Department of Chemistry, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
19
|
Lin SJ, Tapia-Alveal C, Jabado OJ, Germain D, O'Connell MJ. An acetyltransferase-independent function of Eso1 regulates centromere cohesion. Mol Biol Cell 2016; 27:4002-4010. [PMID: 27798241 PMCID: PMC5156541 DOI: 10.1091/mbc.e16-08-0596] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 10/11/2016] [Accepted: 10/12/2016] [Indexed: 12/21/2022] Open
Abstract
Eukaryotes contain three essential Structural Maintenance of Chromosomes (SMC) complexes: cohesin, condensin, and Smc5/6. Cohesin forms a ring-shaped structure that embraces sister chromatids to promote their cohesion. The cohesiveness of cohesin is promoted by acetylation of N-terminal lysines of the Smc3 subunit by the acetyltransferases Eco1 in Saccharomyces cerevisiae and the homologue, Eso1, in Schizosaccharomyces pombe. In both yeasts, these acetyltransferases are essential for cell viability. However, whereas nonacetylatable Smc3 mutants are lethal in S. cerevisiae, they are not in S. pombe We show that the lethality of a temperature-sensitive allele of eso1 (eso1-H17) is due to activation of the spindle assembly checkpoint (SAC) and is associated with premature centromere separation. The lack of cohesion at the centromeres does not correlate with Psm3 acetylation or cohesin levels at the centromeres, but is associated ith significantly reduced recruitment of the cohesin regulator Pds5. The SAC activation in this context is dependent on Smc5/6 function, which is required to remove cohesin from chromosome arms but not centromeres. The mitotic defects caused by Smc5/6 and Eso1 dysfunction are cosuppressed in double mutants. This identifies a novel function (or functions) for Eso1 and Smc5/6 at centromeres and extends the functional relationships between these SMC complexes.
Collapse
Affiliation(s)
- Su-Jiun Lin
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Claudia Tapia-Alveal
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Omar J Jabado
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Doris Germain
- Department of Hematology and Oncology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Matthew J O'Connell
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| |
Collapse
|
20
|
Cisyk AL, Penner-Goeke S, Lichtensztejn Z, Nugent Z, Wightman RH, Singh H, McManus KJ. Characterizing the prevalence of chromosome instability in interval colorectal cancer. Neoplasia 2015; 17:306-16. [PMID: 25810015 PMCID: PMC4372653 DOI: 10.1016/j.neo.2015.02.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Revised: 01/29/2015] [Accepted: 02/04/2015] [Indexed: 12/11/2022] Open
Abstract
A substantial proportion of colorectal cancers (CRCs) are interval CRCs (I-CRCs; i.e., CRCs diagnosed soon after a colonoscopy). Chromosomal instability (CIN) is defined as an increase in the rate of which whole chromosomes/large chromosomal fragments are gained or lost and is observed in 85% of non-hereditary CRCs. The contribution of CIN to the etiology of I-CRCs remains unknown. We established a fluorescence in situ hybridization (FISH) approach to characterize CIN by enumerating specific chromosomes and determined the prevalence of numerical CIN in a population-based cohort of I-CRCs and control (sporadic) CRCs. Using the population-based Manitoba Health administrative databases and Manitoba Cancer Registry, we identified an age, sex, and colonic site of CRC matched cohort of I-CRCs and controls and retrieved their archived paraffin-embedded tumor samples. FISH chromosome enumeration probes specifically recognizing the pericentric regions of chromosomes 8, 11, and 17 were first used on cell lines and then CRC tissue microarrays to detect aneusomy, which was then used to calculate a CIN score (CS). The 15th percentile CS for control CRC was used to define CIN phenotype. Mean CSs were similar in the control CRCs and I-CRCs; 82% of I-CRCs exhibited a CIN phenotype, which was similar to that in the control CRCs. This study suggests that CIN is the most prevalent contributor to genomic instability in I-CRCs. Further studies should evaluate CIN and microsatellite instability (MSI) in the same cohort of I-CRCs to corroborate our findings and to further assess concomitant contribution of CIN and MSI to I-CRCs.
Collapse
Affiliation(s)
- A L Cisyk
- University of Manitoba, Winnipeg, Canada; Department of Biochemistry and Medical Genetics, Winnipeg, Canada; Research Institute of Oncology and Hematology, Winnipeg, Canada
| | - S Penner-Goeke
- University of Manitoba, Winnipeg, Canada; Department of Biochemistry and Medical Genetics, Winnipeg, Canada; Research Institute of Oncology and Hematology, Winnipeg, Canada
| | - Z Lichtensztejn
- University of Manitoba, Winnipeg, Canada; Department of Biochemistry and Medical Genetics, Winnipeg, Canada; Research Institute of Oncology and Hematology, Winnipeg, Canada
| | - Z Nugent
- CancerCare Manitoba, Winnipeg, Canada
| | - R H Wightman
- University of Manitoba, Winnipeg, Canada; Department of Pathology, Winnipeg, Canada; Grace Hospital, Winnipeg, Canada
| | - H Singh
- University of Manitoba, Winnipeg, Canada; Department of Internal Medicine, Health Sciences Centre, Winnipeg, Canada
| | - K J McManus
- University of Manitoba, Winnipeg, Canada; Department of Biochemistry and Medical Genetics, Winnipeg, Canada; Research Institute of Oncology and Hematology, Winnipeg, Canada.
| |
Collapse
|
21
|
Increased expression of ESCO1 is correlated with poor patient survival and its role in human bladder cancer. Tumour Biol 2015; 37:5165-70. [PMID: 26547586 DOI: 10.1007/s13277-015-4375-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 11/03/2015] [Indexed: 12/19/2022] Open
Abstract
There is increasing evidence suggesting that establishment of sister chromatid cohesion N-acetyltransferase 1 (ESCO1) was involved in tumorigenesis. However, its role in bladder cancer remains unclear. In this study, we aimed to study the clinical correlation and biological significance of ESCO1 in bladder cancer. Our results showed that ESCO1 was significantly over-expressed in bladder cancer tissues compared with that in adjacent normal tissues. And, increased ESCO1 expression was significantly associated with higher grade (P < 0.001), higher tumor stage (P = 0.014), and multifocality (P = 0.042). Kaplan-Meier analysis and Cox proportional hazards model were performed to determine the prognostic significance of ESCO1, and the results showed that ESCO1 is a useful prognostic marker for bladder cancer patients. Moreover, we found that ESCO1 knockdown inhibited the growth, migration, and invasion of bladder cancer cells. In conclusion, our findings indicated that ESCO1 may play an important role in human bladder cancer, and ESCO1 might serve as a novel target and prognosis factor for human bladder cancer.
Collapse
|
22
|
Talukder AK, Ravishankar S, Sasmal K, Gandham S, Prabhukumar J, Achutharao PH, Barh D, Blasi F. XomAnnotate: Analysis of Heterogeneous and Complex Exome- A Step towards Translational Medicine. PLoS One 2015; 10:e0123569. [PMID: 25905921 PMCID: PMC4408095 DOI: 10.1371/journal.pone.0123569] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 02/20/2015] [Indexed: 12/14/2022] Open
Abstract
In translational cancer medicine, implicated pathways and the relevant master genes are of focus. Exome's specificity, processing-time, and cost advantage makes it a compelling tool for this purpose. However, analysis of exome lacks reliable combinatory analysis tools and techniques. In this paper we present XomAnnotate – a meta- and functional-analysis software for exome. We compared UnifiedGenotyper, Freebayes, Delly, and Lumpy algorithms that were designed for whole-genome and combined their strengths in XomAnnotate for exome data through meta-analysis to identify comprehensive mutation profile (SNPs/SNVs, short inserts/deletes, and SVs) of patients. The mutation profile is annotated followed by functional analysis through pathway enrichment and network analysis to identify most critical genes and pathways implicated in the disease genesis. The efficacy of the software is verified through MDS and clustering and tested with available 11 familial non-BRCA1/BRCA2 breast cancer exome data. The results showed that the most significantly affected pathways across all samples are cell communication and antigen processing and presentation. ESCO1, HYAL1, RAF1 and PRKCA emerged as the key genes. Network analysis further showed the purine and propanotate metabolism pathways along with RAF1 and PRKCA genes to be master regulators in these patients. Therefore, XomAnnotate is able to use exome data to identify entire mutation landscape, pathways, and the master genes accurately with wide concordance from earlier microarray and whole-genome studies -- making it a suitable biomedical software for using exome in next-generation translational medicine.
Collapse
Affiliation(s)
- Asoke K. Talukder
- InterpretOmics India Pvt Ltd, #329, 7 Main, HAL 2 Stage, Indiranagar, Bangalore, 560 008, Karnataka, India
- * E-mail:
| | - Shashidhar Ravishankar
- InterpretOmics India Pvt Ltd, #329, 7 Main, HAL 2 Stage, Indiranagar, Bangalore, 560 008, Karnataka, India
| | - Krittika Sasmal
- InterpretOmics India Pvt Ltd, #329, 7 Main, HAL 2 Stage, Indiranagar, Bangalore, 560 008, Karnataka, India
| | - Santhosh Gandham
- InterpretOmics India Pvt Ltd, #329, 7 Main, HAL 2 Stage, Indiranagar, Bangalore, 560 008, Karnataka, India
| | - Jyothsna Prabhukumar
- InterpretOmics India Pvt Ltd, #329, 7 Main, HAL 2 Stage, Indiranagar, Bangalore, 560 008, Karnataka, India
| | - Prahalad H. Achutharao
- InterpretOmics India Pvt Ltd, #329, 7 Main, HAL 2 Stage, Indiranagar, Bangalore, 560 008, Karnataka, India
| | - Debmalya Barh
- InterpretOmics India Pvt Ltd, #329, 7 Main, HAL 2 Stage, Indiranagar, Bangalore, 560 008, Karnataka, India
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur, West Bengal, 721172, India
| | - Francesco Blasi
- Laboratory of Transcriptional Regulation in Development and Cancer, IFOM (Fondazione Istituto FIRC di Oncologia Molecolare), Milano, Italy
| |
Collapse
|
23
|
Xue LC, Xu ZW, Wang KH, Wang N, Zhang XX, Wang S. Regulatory network of microRNAs, target genes, transcription factors and host genes in endometrial cancer. Asian Pac J Cancer Prev 2015; 16:475-83. [PMID: 25684474 DOI: 10.7314/apjcp.2015.16.2.475] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Genes and microRNAs (miRNAs) have important roles in human oncology. However, most of the biological factors are reported in disperse form which makes it hard to discover the pathology. In this study, genes and miRNAs involved in human endometrial cancer(EC) were collected and formed into regulatory networks following their interactive relations, including miRNAs targeting genes, transcription factors (TFs) regulating miRNAs and miRNAs included in their host genes. Networks are constructed hierarchically at three levels: differentially expressed, related and global. Among the three, the differentially expressed network is the most important and fundamental network that contains the key genes and miRNAs in EC. The target genes, TFs and miRNAs are differentially expressed in EC so that any mutation in them may impact on EC development. Some key pathways in networks were highlighted to analyze how they interactively influence other factors and carcinogenesis. Upstream and downstream pathways of the differentially expressed genes and miRNAs were compared and analyzed. The purpose of this study was to partially reveal the deep regulatory mechanisms in EC using a new method that combines comprehensive genes and miRNAs together with their relationships. It may contribute to cancer prevention and gene therapy of EC.
Collapse
Affiliation(s)
- Lu-Chen Xue
- Department of Software Engineering, Jilin University, Changchun, China E-mail :
| | | | | | | | | | | |
Collapse
|
24
|
Rudd ML, Mohamed H, Price JC, O'Hara AJ, Le Gallo M, Urick ME, Cruz P, Zhang S, Hansen NF, Godwin AK, Sgroi DC, Wolfsberg TG, Mullikin JC, Merino MJ, Bell DW. Mutational analysis of the tyrosine kinome in serous and clear cell endometrial cancer uncovers rare somatic mutations in TNK2 and DDR1. BMC Cancer 2014; 14:884. [PMID: 25427824 PMCID: PMC4258955 DOI: 10.1186/1471-2407-14-884] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 11/13/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Endometrial cancer (EC) is the 8th leading cause of cancer death amongst American women. Most ECs are endometrioid, serous, or clear cell carcinomas, or an admixture of histologies. Serous and clear ECs are clinically aggressive tumors for which alternative therapeutic approaches are needed. The purpose of this study was to search for somatic mutations in the tyrosine kinome of serous and clear cell ECs, because mutated kinases can point to potential therapeutic targets. METHODS In a mutation discovery screen, we PCR amplified and Sanger sequenced the exons encoding the catalytic domains of 86 tyrosine kinases from 24 serous, 11 clear cell, and 5 mixed histology ECs. For somatically mutated genes, we next sequenced the remaining coding exons from the 40 discovery screen tumors and sequenced all coding exons from another 72 ECs (10 clear cell, 21 serous, 41 endometrioid). We assessed the copy number of mutated kinases in this cohort of 112 tumors using quantitative real time PCR, and we used immunoblotting to measure expression of these kinases in endometrial cancer cell lines. RESULTS Overall, we identified somatic mutations in TNK2 (tyrosine kinase non-receptor, 2) and DDR1 (discoidin domain receptor tyrosine kinase 1) in 5.3% (6 of 112) and 2.7% (3 of 112) of ECs. Copy number gains of TNK2 and DDR1 were identified in another 4.5% and 0.9% of 112 cases respectively. Immunoblotting confirmed TNK2 and DDR1 expression in endometrial cancer cell lines. Three of five missense mutations in TNK2 and one of two missense mutations in DDR1 are predicted to impact protein function by two or more in silico algorithms. The TNK2(P761Rfs*72) frameshift mutation was recurrent in EC, and the DDR1(R570Q) missense mutation was recurrent across tumor types. CONCLUSIONS This is the first study to systematically search for mutations in the tyrosine kinome in clear cell endometrial tumors. Our findings indicate that high-frequency somatic mutations in the catalytic domains of the tyrosine kinome are rare in clear cell ECs. We uncovered ten new mutations in TNK2 and DDR1 within serous and endometrioid ECs, thus providing novel insights into the mutation spectrum of each gene in EC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Daphne W Bell
- Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda MD 20892, USA.
| |
Collapse
|
25
|
Koppensteiner R, Samartzis EP, Noske A, von Teichman A, Dedes I, Gwerder M, Imesch P, Ikenberg K, Moch H, Fink D, Stucki M, Dedes KJ. Effect of MRE11 loss on PARP-inhibitor sensitivity in endometrial cancer in vitro. PLoS One 2014; 9:e100041. [PMID: 24927325 PMCID: PMC4057395 DOI: 10.1371/journal.pone.0100041] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Accepted: 05/21/2014] [Indexed: 01/19/2023] Open
Abstract
AIM OF THE STUDY To evaluate the frequency of MRE11/RAD50/NBS1 (MRN)-complex loss of protein expression in endometrial cancers (EC) and to determine whether loss of MRE11 renders the cancer cells sensitive to Poly(ADP-ribose) polymerase (PARP)-inhibitory treatment. METHODS MRN expression was examined in 521 samples of endometrial carcinomas and in 10 cancer cell lines. A putative mutation hotspot in the form of an intronic poly(T) allele in MRE11 was sequenced in selected cases (n = 26). Sensitivity to the PARP-inhibitor, BMN673 was tested in colony formation assays before and after MRE11 silencing using siRNA. Homologous recombination (HR) DNA repair was evaluated by RAD51-foci formation assay upon irradiation and drug treatment. RESULTS Loss of MRE11 protein was found in 30.7% of EC tumours and significantly associated with loss of RAD50, NBS1 and mismatch repair protein expression. One endometrial cell line showed a markedly reduced MRE11 expression due to a homozygous poly(T) mutation of MRE11, thereby exhibiting an increased sensitivity to BMN673. MRE11 depletion sensitizes MRE11 expressing EC cell lines to the treatment with BMN673. The increased sensitivity to PARP-inhibition correlates with reduced RAD51 foci formation upon ionizing radiation in MRE11-depleted cells. CONCLUSION Loss of the MRE11 protein predicts sensitivity to PARP-inhibitor sensitivity in vitro, defining it as an additional synthetic lethal gene with PARP. The high incidence of MRE11 loss in ECs can be potentially exploited for PARP-inhibitor therapy. Furthermore, MRE11 protein expression using immunohistochemistry could be investigated as a predictive biomarker for PARP-inhibitor treatment.
Collapse
Affiliation(s)
| | | | - Aurelia Noske
- Institute of Surgical Pathology, University Hospital of Zurich, Zurich, Switzerland
| | - Adriana von Teichman
- Institute of Surgical Pathology, University Hospital of Zurich, Zurich, Switzerland
| | - Ioannis Dedes
- Division of Gynaecology, University Hospital of Zurich, Zurich, Switzerland
| | - Myriam Gwerder
- Division of Gynaecology, University Hospital of Zurich, Zurich, Switzerland
| | - Patrick Imesch
- Division of Gynaecology, University Hospital of Zurich, Zurich, Switzerland
| | - Kristian Ikenberg
- Institute of Surgical Pathology, University Hospital of Zurich, Zurich, Switzerland
| | - Holger Moch
- Institute of Surgical Pathology, University Hospital of Zurich, Zurich, Switzerland
| | - Daniel Fink
- Division of Gynaecology, University Hospital of Zurich, Zurich, Switzerland
| | - Manuel Stucki
- Division of Gynaecology, University Hospital of Zurich, Zurich, Switzerland
| | - Konstantin J. Dedes
- Division of Gynaecology, University Hospital of Zurich, Zurich, Switzerland
- * E-mail:
| |
Collapse
|
26
|
Henninger EE, Pursell ZF. DNA polymerase ε and its roles in genome stability. IUBMB Life 2014; 66:339-51. [PMID: 24861832 DOI: 10.1002/iub.1276] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Accepted: 05/02/2014] [Indexed: 12/14/2022]
Abstract
DNA Polymerase Epsilon (Pol ε) is one of three DNA Polymerases (along with Pol δ and Pol α) required for nuclear DNA replication in eukaryotes. Pol ε is comprised of four subunits, the largest of which is encoded by the POLE gene and contains the catalytic polymerase and exonuclease activities. The 3'-5' exonuclease proofreading activity is able to correct DNA synthesis errors and helps protect against genome instability. Recent cancer genome sequencing efforts have shown that 3% of colorectal and 7% of endometrial cancers contain mutations within the exonuclease domain of POLE and are associated with significantly elevated levels of single nucleotide substitutions (15-500 per Mb) and microsatellite stability. POLE mutations have also been found in other tumor types, though at lower frequency, suggesting roles in tumorigenesis more broadly in different tissue types. In addition to its proofreading activity, Pol ε contributes to genome stability through multiple mechanisms that are discussed in this review.
Collapse
Affiliation(s)
- Erin E Henninger
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, USA
| | | |
Collapse
|
27
|
Sajesh BV, Lichtensztejn Z, McManus KJ. Sister chromatid cohesion defects are associated with chromosome instability in Hodgkin lymphoma cells. BMC Cancer 2013; 13:391. [PMID: 23962039 PMCID: PMC3751861 DOI: 10.1186/1471-2407-13-391] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Accepted: 08/19/2013] [Indexed: 12/25/2022] Open
Abstract
Background Chromosome instability manifests as an abnormal chromosome complement and is a pathogenic event in cancer. Although a correlation between abnormal chromosome numbers and cancer exist, the underlying mechanisms that cause chromosome instability are poorly understood. Recent data suggests that aberrant sister chromatid cohesion causes chromosome instability and thus contributes to the development of cancer. Cohesion normally functions by tethering nascently synthesized chromatids together to prevent premature segregation and thus chromosome instability. Although the prevalence of aberrant cohesion has been reported for some solid tumors, its prevalence within liquid tumors is unknown. Consequently, the current study was undertaken to evaluate aberrant cohesion within Hodgkin lymphoma, a lymphoid malignancy that frequently exhibits chromosome instability. Methods Using established cytogenetic techniques, the prevalence of chromosome instability and aberrant cohesion was examined within mitotic spreads generated from five commonly employed Hodgkin lymphoma cell lines (L-1236, KM-H2, L-428, L-540 and HDLM-2) and a lymphocyte control. Indirect immunofluorescence and Western blot analyses were performed to evaluate the localization and expression of six critical proteins involved in the regulation of sister chromatid cohesion. Results We first confirmed that all five Hodgkin lymphoma cell lines exhibited chromosome instability relative to the lymphocyte control. We then determined that each Hodgkin lymphoma cell line exhibited cohesion defects that were subsequently classified into mild, moderate or severe categories. Surprisingly, ~50% of the mitotic spreads generated from L-540 and HDLM-2 harbored cohesion defects. To gain mechanistic insight into the underlying cause of the aberrant cohesion we examined the localization and expression of six critical proteins involved in cohesion. Although all proteins produced the expected nuclear localization pattern, striking differences in RAD21 expression was observed: RAD21 expression was lowest in L-540 and highest within HDLM-2. Conclusion We conclude that aberrant cohesion is a common feature of all five Hodgkin lymphoma cell lines evaluated. We further conclude that aberrant RAD21 expression is a strong candidate to underlie aberrant cohesion, chromosome instability and contribute to the development of the disease. Our findings support a growing body of evidence suggesting that cohesion defects and aberrant RAD21 expression are pathogenic events that contribute to tumor development.
Collapse
Affiliation(s)
- Babu V Sajesh
- Manitoba Institute of Cell Biology and the Department of Biochemistry & Medical Genetics, University of Manitoba, ON6010 - 675 McDermot Avenue, Winnipeg, Manitoba MB R3E 0V9, Canada
| | | | | |
Collapse
|